1
|
Domingues N, Catarino S, Cristóvão B, Rodrigues L, Carvalho FA, Sarmento MJ, Zuzarte M, Almeida J, Ribeiro-Rodrigues T, Correia-Rodrigues Â, Fernandes F, Rodrigues-Santos P, Aasen T, Santos NC, Korolchuk VI, Gonçalves T, Milosevic I, Raimundo N, Girão H. Connexin43 promotes exocytosis of damaged lysosomes through actin remodelling. EMBO J 2024; 43:3627-3649. [PMID: 39044100 PMCID: PMC11377567 DOI: 10.1038/s44318-024-00177-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 07/04/2024] [Accepted: 07/09/2024] [Indexed: 07/25/2024] Open
Abstract
A robust and efficient cellular response to lysosomal membrane damage prevents leakage from the lysosome lumen into the cytoplasm. This response is understood to happen through either lysosomal membrane repair or lysophagy. Here we report exocytosis as a third response mechanism to lysosomal damage, which is further potentiated when membrane repair or lysosomal degradation mechanisms are impaired. We show that Connexin43 (Cx43), a protein canonically associated with gap junctions, is recruited from the plasma membrane to damaged lysosomes, promoting their secretion and accelerating cell recovery. The effects of Cx43 on lysosome exocytosis are mediated by a reorganization of the actin cytoskeleton that increases plasma membrane fluidity and decreases cell stiffness. Furthermore, we demonstrate that Cx43 interacts with the actin nucleator Arp2, the activity of which was shown to be necessary for Cx43-mediated actin rearrangement and lysosomal exocytosis following damage. These results define a novel mechanism of lysosomal quality control whereby Cx43-mediated actin remodelling potentiates the secretion of damaged lysosomes.
Collapse
Affiliation(s)
- Neuza Domingues
- Univ Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Centre for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
- Clinical and Academic Centre of Coimbra, Coimbra, Portugal
- Multidisciplinary Institute of Ageing, University of Coimbra, Coimbra, Portugal
| | - Steve Catarino
- Univ Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Centre for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
- Clinical and Academic Centre of Coimbra, Coimbra, Portugal
| | - Beatriz Cristóvão
- Univ Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Centre for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
- Clinical and Academic Centre of Coimbra, Coimbra, Portugal
| | - Lisa Rodrigues
- Univ Coimbra, Center for Neurosciences and Cell Biology (CNC), Coimbra, Portugal
| | - Filomena A Carvalho
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Maria João Sarmento
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Mónica Zuzarte
- Univ Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Centre for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
- Clinical and Academic Centre of Coimbra, Coimbra, Portugal
| | - Jani Almeida
- Univ Coimbra, Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Centre for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
- Clinical and Academic Centre of Coimbra, Coimbra, Portugal
- Univ Coimbra, Center for Neurosciences and Cell Biology (CNC), Coimbra, Portugal
| | - Teresa Ribeiro-Rodrigues
- Univ Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Centre for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
- Clinical and Academic Centre of Coimbra, Coimbra, Portugal
| | - Ânia Correia-Rodrigues
- Univ Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Centre for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
- Clinical and Academic Centre of Coimbra, Coimbra, Portugal
| | - Fábio Fernandes
- Institute for Bioengineering and Biosciences (IBB) and Associate Laboratory i4HB-Institute for Health and Bioeconomy, Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Paulo Rodrigues-Santos
- Univ Coimbra, Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Centre for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
- Clinical and Academic Centre of Coimbra, Coimbra, Portugal
- Univ Coimbra, Center for Neurosciences and Cell Biology (CNC), Coimbra, Portugal
| | - Trond Aasen
- Vall d'Hebron Research Institute (VHIR), Barcelona, Spain
| | - Nuno C Santos
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Viktor I Korolchuk
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle, UK
| | - Teresa Gonçalves
- Univ Coimbra, Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Centre for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
- Univ Coimbra, Center for Neurosciences and Cell Biology (CNC), Coimbra, Portugal
| | - Ira Milosevic
- Multidisciplinary Institute of Ageing, University of Coimbra, Coimbra, Portugal
- University of Oxford, Centre for Human Genetics, Nuffield Department of Medicine, Oxford, UK
| | - Nuno Raimundo
- Multidisciplinary Institute of Ageing, University of Coimbra, Coimbra, Portugal
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA, USA
| | - Henrique Girão
- Univ Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal.
- Univ Coimbra, Faculty of Medicine, Coimbra, Portugal.
- Univ Coimbra, Centre for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal.
- Clinical and Academic Centre of Coimbra, Coimbra, Portugal.
| |
Collapse
|
2
|
Shebanova A, Perrin QM, Zhu K, Gudlur S, Chen Z, Sun Y, Huang C, Lim ZW, Mondarte EA, Sun R, Lim S, Yu J, Miao Y, Parikh AN, Ludwig A, Miserez A. Cellular Uptake of Phase-Separating Peptide Coacervates. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024:e2402652. [PMID: 39214144 DOI: 10.1002/advs.202402652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 06/07/2024] [Indexed: 09/04/2024]
Abstract
Peptide coacervates self-assembling via liquid-liquid phase separation are appealing intracellular delivery vehicles of macromolecular therapeutics (proteins, DNA, mRNA) owing to their non-cytotoxicity, high encapsulation capacity, and efficient cellular uptake. However, the mechanisms by which these viscoelastic droplets cross the cellular membranes remain unknown. Here, using multimodal imaging, data analytics, and biochemical inhibition assays, identify the key steps by which droplets enter the cell. find that the uptake follows a non-canonical pathway and instead integrates essential features of macropinocytosis and phagocytosis, namely active remodeling of the actin cytoskeleton and appearance of filopodia-like protrusions. Experiments using giant unilamellar vesicles show that the coacervates attach to the bounding membrane in a charge- and cholesterol-dependent manner but do not breach the lipid bilayer barrier. Cell uptake in the presence of small molecule inhibitors - interfering with actin and tubulin polymerization - confirm the active role of cytoskeleton remodeling, most prominently evident in electron microscopy imaging. These findings suggest a peculiar internalization mechanism for viscoelastic, glassy coacervate droplets combining features of non-specific uptake of fluids by macropinocytosis and particulate uptake of phagocytosis. The broad implications of this study will enable to enhance the efficacy and utility of coacervate-based strategies for intracellular delivery of macromolecular therapeutics.
Collapse
Affiliation(s)
- Anastasia Shebanova
- Centre for Sustainable Materials, School of Materials Science and Engineering, Nanyang Technological University (NTU), 50 Nanyang Avenue, Singapore, 637553, Singapore
| | - Quentin Moana Perrin
- Centre for Sustainable Materials, School of Materials Science and Engineering, Nanyang Technological University (NTU), 50 Nanyang Avenue, Singapore, 637553, Singapore
| | - Kexin Zhu
- School of Biological Sciences, NTU, 60 Nanyang Drive, Singapore, 637551, Singapore
| | - Sushanth Gudlur
- Centre for Sustainable Materials, School of Materials Science and Engineering, Nanyang Technological University (NTU), 50 Nanyang Avenue, Singapore, 637553, Singapore
| | - Zilin Chen
- Centre for Sustainable Materials, School of Materials Science and Engineering, Nanyang Technological University (NTU), 50 Nanyang Avenue, Singapore, 637553, Singapore
| | - Yue Sun
- Centre for Sustainable Materials, School of Materials Science and Engineering, Nanyang Technological University (NTU), 50 Nanyang Avenue, Singapore, 637553, Singapore
| | - Congxi Huang
- Centre for Sustainable Materials, School of Materials Science and Engineering, Nanyang Technological University (NTU), 50 Nanyang Avenue, Singapore, 637553, Singapore
| | - Zhi Wei Lim
- Centre for Sustainable Materials, School of Materials Science and Engineering, Nanyang Technological University (NTU), 50 Nanyang Avenue, Singapore, 637553, Singapore
| | - Evan Angelo Mondarte
- Centre for Sustainable Materials, School of Materials Science and Engineering, Nanyang Technological University (NTU), 50 Nanyang Avenue, Singapore, 637553, Singapore
| | - Ruoxuan Sun
- School of Chemistry, Chemical Engineering and Biotechnology, NTU, 70 Nanyang Drive, Singapore, 637457, Singapore
| | - Sierin Lim
- School of Chemistry, Chemical Engineering and Biotechnology, NTU, 70 Nanyang Drive, Singapore, 637457, Singapore
- Institute for Digital Molecular Analytics and Science (IDMxS), NTU, 59 Nanyang Drive, Singapore, 636921, Singapore
| | - Jing Yu
- Centre for Sustainable Materials, School of Materials Science and Engineering, Nanyang Technological University (NTU), 50 Nanyang Avenue, Singapore, 637553, Singapore
- Institute for Digital Molecular Analytics and Science (IDMxS), NTU, 59 Nanyang Drive, Singapore, 636921, Singapore
| | - Yansong Miao
- School of Biological Sciences, NTU, 60 Nanyang Drive, Singapore, 637551, Singapore
- Institute for Digital Molecular Analytics and Science (IDMxS), NTU, 59 Nanyang Drive, Singapore, 636921, Singapore
| | - Atul N Parikh
- Centre for Sustainable Materials, School of Materials Science and Engineering, Nanyang Technological University (NTU), 50 Nanyang Avenue, Singapore, 637553, Singapore
- Institute for Digital Molecular Analytics and Science (IDMxS), NTU, 59 Nanyang Drive, Singapore, 636921, Singapore
- Departments of Biomedical Engineering and Materials Science & Engineering, University of California, Davis, CA, 95616, USA
| | - Alexander Ludwig
- School of Biological Sciences, NTU, 60 Nanyang Drive, Singapore, 637551, Singapore
- NTU Institute of Structural Biology, NTU, 59 Nanyang Drive, Singapore, 636921, Singapore
| | - Ali Miserez
- Centre for Sustainable Materials, School of Materials Science and Engineering, Nanyang Technological University (NTU), 50 Nanyang Avenue, Singapore, 637553, Singapore
- School of Biological Sciences, NTU, 60 Nanyang Drive, Singapore, 637551, Singapore
| |
Collapse
|
3
|
Bouti P, Klein BJAM, Verkuijlen PJH, Schornagel K, van Alphen FPJ, Taris KKH, van den Biggelaar M, Hoogendijk AJ, van Bruggen R, Kuijpers TW, Matlung HL. SKAP2 acts downstream of CD11b/CD18 and regulates neutrophil effector function. Front Immunol 2024; 15:1344761. [PMID: 38487529 PMCID: PMC10937362 DOI: 10.3389/fimmu.2024.1344761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Accepted: 02/01/2024] [Indexed: 03/17/2024] Open
Abstract
Background The importance of CD11b/CD18 expression in neutrophil effector functions is well known. Beyond KINDLIN3 and TALIN1, which are involved in the induction of the high-affinity binding CD11b/CD18 conformation, the signaling pathways that orchestrate this response remain incompletely understood. Method We performed an unbiased screening method for protein selection by biotin identification (BioID) and investigated the KINDLIN3 interactome. We used liquid chromatography with tandem mass spectrometry as a powerful analytical tool. Generation of NB4 CD18, KINDLIN3, or SKAP2 knockout neutrophils was achieved using CRISPR-Cas9 technology, and the cells were examined for their effector function using flow cytometry, live cell imaging, microscopy, adhesion, or antibody-dependent cellular cytotoxicity (ADCC). Results Among the 325 proteins significantly enriched, we identified Src kinase-associated phosphoprotein 2 (SKAP2), a protein involved in actin polymerization and integrin-mediated outside-in signaling. CD18 immunoprecipitation in primary or NB4 neutrophils demonstrated the presence of SKAP2 in the CD11b/CD18 complex at a steady state. Under this condition, adhesion to plastic, ICAM-1, or fibronectin was observed in the absence of SKAP2, which could be abrogated by blocking the actin rearrangements with latrunculin B. Upon stimulation of NB4 SKAP2-deficient neutrophils, adhesion to fibronectin was enhanced whereas CD18 clustering was strongly reduced. This response corresponded with significantly impaired CD11b/CD18-dependent NADPH oxidase activity, phagocytosis, and cytotoxicity against tumor cells. Conclusion Our results suggest that SKAP2 has a dual role. It may restrict CD11b/CD18-mediated adhesion only under resting conditions, but its major contribution lies in the regulation of dynamic CD11b/CD18-mediated actin rearrangements and clustering as required for cellular effector functions of human neutrophils.
Collapse
Affiliation(s)
- Panagiota Bouti
- Department of Molecular Hematology Sanquin Research and Landsteiner Laboratory, Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam, Netherlands
| | - Bart J. A. M. Klein
- Department of Molecular Hematology Sanquin Research and Landsteiner Laboratory, Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam, Netherlands
| | - Paul J. H. Verkuijlen
- Department of Molecular Hematology Sanquin Research and Landsteiner Laboratory, Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam, Netherlands
| | - Karin Schornagel
- Department of Molecular Hematology Sanquin Research and Landsteiner Laboratory, Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam, Netherlands
| | - Floris P. J. van Alphen
- Department of Molecular Hematology Sanquin Research and Landsteiner Laboratory, Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam, Netherlands
| | - Kees-Karel H. Taris
- Department of Physics and Astronomy, Vrije Universiteit, Amsterdam, Netherlands
- LaserLaB Amsterdam, Vrije Universiteit, Amsterdam, Netherlands
| | - Maartje van den Biggelaar
- Department of Molecular Hematology Sanquin Research and Landsteiner Laboratory, Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam, Netherlands
| | - Arie J. Hoogendijk
- Department of Molecular Hematology Sanquin Research and Landsteiner Laboratory, Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam, Netherlands
| | - Robin van Bruggen
- Department of Molecular Hematology Sanquin Research and Landsteiner Laboratory, Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam, Netherlands
| | - Taco W. Kuijpers
- Department of Molecular Hematology Sanquin Research and Landsteiner Laboratory, Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam, Netherlands
- Department of Pediatric Immunology and Infectious Diseases, Emma Children’s Hospital, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Hanke L. Matlung
- Department of Molecular Hematology Sanquin Research and Landsteiner Laboratory, Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
4
|
Witt H, Vache M, Cordes A, Janshoff A. Detachment of giant liposomes - coupling of receptor mobility and membrane shape. SOFT MATTER 2020; 16:6424-6433. [PMID: 32588015 DOI: 10.1039/d0sm00863j] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Cellular adhesion is an intricate physical process controlled by ligand-receptor affinity, density, mobility, and external forces transmitted through the elastic properties of the cell. As a model for cellular adhesion we study the detachment of cell-sized liposomes and membrane-coated silica beads from supported bilayers using atomic force microscopy. Adhesion between the two surfaces is mediated by the interaction between the adhesive lipid anchored saccharides lactosylceramide and the ganglioside GM3. We found that force-distance curves of liposome detachment have a very peculiar, partially concave shape, reminiscent of the nonlinear extension of polymers. By contrast, detachment of membrane coated beads led to force-distance curves similar to the detachment of living cells. Theoretical modelling of the enforced detachment suggests that the non-convex force curve shape arises from the mobility of ligands provoking a switch of shapes from spherical to unduloidal during detachment.
Collapse
Affiliation(s)
- Hannes Witt
- Max Planck Institute for Dynamics and Self-Organization, Am Faßberg, 37077 Göttingen, Germany
| | | | | | | |
Collapse
|
5
|
MacKay L, Khadra A. The bioenergetics of integrin-based adhesion, from single molecule dynamics to stability of macromolecular complexes. Comput Struct Biotechnol J 2020; 18:393-416. [PMID: 32128069 PMCID: PMC7044673 DOI: 10.1016/j.csbj.2020.02.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 02/03/2020] [Accepted: 02/04/2020] [Indexed: 12/22/2022] Open
Abstract
The forces actively generated by motile cells must be transmitted to their environment in a spatiotemporally regulated manner, in order to produce directional cellular motion. This task is accomplished through integrin-based adhesions, large macromolecular complexes that link the actin-cytoskelton inside the cell to its external environment. Despite their relatively large size, adhesions exhibit rapid dynamics, switching between assembly and disassembly in response to chemical and mechanical cues exerted by cytoplasmic biochemical signals, and intracellular/extracellular forces, respectively. While in material science, force typically disrupts adhesive contact, in this biological system, force has a more nuanced effect, capable of causing assembly or disassembly. This initially puzzled experimentalists and theorists alike, but investigation into the mechanisms regulating adhesion dynamics have progressively elucidated the origin of these phenomena. This review provides an overview of recent studies focused on the theoretical understanding of adhesion assembly and disassembly as well as the experimental studies that motivated them. We first concentrate on the kinetics of integrin receptors, which exhibit a complex response to force, and then investigate how this response manifests itself in macromolecular adhesion complexes. We then turn our attention to studies of adhesion plaque dynamics that link integrins to the actin-cytoskeleton, and explain how force can influence the assembly/disassembly of these macromolecular structure. Subsequently, we analyze the effect of force on integrins populations across lengthscales larger than single adhesions. Finally, we cover some theoretical studies that have considered both integrins and the adhesion plaque and discuss some potential future avenues of research.
Collapse
Affiliation(s)
- Laurent MacKay
- Department of Physiology, McGill University, 3655 Promenade Sir William Osler, Montreal, Quebec, Canada
| | - Anmar Khadra
- Department of Physiology, McGill University, 3655 Promenade Sir William Osler, Montreal, Quebec, Canada
| |
Collapse
|
6
|
Intermittent rolling is a defect of the extravasation cascade caused by Myosin1e-deficiency in neutrophils. Proc Natl Acad Sci U S A 2019; 116:26752-26758. [PMID: 31811025 DOI: 10.1073/pnas.1902502116] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Neutrophil extravasation is a migratory event in response to inflammation that depends on cytoskeletal dynamics regulated by myosins. Myosin-1e (Myo1e) is a long-tailed class-I myosin that has not yet been studied in the context of neutrophil-endothelial interactions and neutrophil extravasation. Intravital microscopy of TNFα-inflamed cremaster muscles in Myo1e-deficient mice revealed that Myo1e is required for efficient neutrophil extravasation. Specifically, Myo1e deficiency caused increased rolling velocity, decreased firm adhesion, aberrant crawling, and strongly reduced transmigration. Interestingly, we observed a striking discontinuous rolling behavior termed "intermittent rolling," during which Myo1e-deficient neutrophils showed alternating rolling and jumping movements. Surprisingly, chimeric mice revealed that these effects were due to Myo1e deficiency in leukocytes. Vascular permeability was not significantly altered in Myo1e KO mice. Myo1e-deficient neutrophils showed diminished arrest, spreading, uropod formation, and chemotaxis due to defective actin polymerization and integrin activation. In conclusion, Myo1e critically regulates adhesive interactions of neutrophils with the vascular endothelium and neutrophil extravasation. Myo1e may therefore be an interesting target in chronic inflammatory diseases characterized by excessive neutrophil recruitment.
Collapse
|
7
|
MacKay L, Khadra A. Dynamics of Mechanosensitive Nascent Adhesion Formation. Biophys J 2019; 117:1057-1073. [PMID: 31493858 PMCID: PMC6818182 DOI: 10.1016/j.bpj.2019.08.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 07/31/2019] [Accepted: 08/02/2019] [Indexed: 01/09/2023] Open
Abstract
Cellular migration is a tightly regulated process that involves actin cytoskeleton, adaptor proteins, and integrin receptors. Forces are transmitted extracellularly through protein complexes of these molecules, called adhesions. Adhesions anchor the cell to its substrate, allowing it to migrate. In Chinese hamster ovary cells, three classes of adhesion can be identified: nascent adhesions (NAs), focal complexes, and focal adhesions, ranked here ascendingly based on size and stability. To understand the dynamics and mechanosensitive properties of NAs, a biophysical model of these NAs as colocalized clusters of integrins and adaptor proteins is developed. The model is then analyzed to characterize the dependence of NA area on biophysical parameters that regulate the number of integrins and adaptor proteins within NAs through a mechanosensitive coaggregation mechanism. Our results reveal that NA formation is triggered beyond a threshold of adaptor protein, integrin, or extracellular ligand densities, with these three factors listed in descending order of their relative influence on NA area. Further analysis of the model also reveals that an increase in coaggregation or reductions in integrin mobility inside the adhesion potentiate NA formation. By extending the model to consider the mechanosensitivity of the integrin bond, we identify mechanical stress, rather than mechanical load, as a permissive mechanical parameter that allows for noise-dependent and independent NA assembly, despite both parameters producing a bistable switch possessing a hysteresis. Stochastic simulations of the model confirm these results computationally. This study thus provides insight into the mechanical conditions defining NA dynamics.
Collapse
Affiliation(s)
- Laurent MacKay
- Department of Physiology, McGill University, Montreal, Quebec, Canada
| | - Anmar Khadra
- Department of Physiology, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
8
|
Vega BA, Belinka BA, Kachlany SC. Aggregatibacter actinomycetemcomitans Leukotoxin (LtxA; Leukothera ®): Mechanisms of Action and Therapeutic Applications. Toxins (Basel) 2019; 11:toxins11090489. [PMID: 31454891 PMCID: PMC6784247 DOI: 10.3390/toxins11090489] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 08/21/2019] [Accepted: 08/22/2019] [Indexed: 12/18/2022] Open
Abstract
Aggregatibacter actinomycetemcomitans is an oral pathogen that produces the RTX toxin, leukotoxin (LtxA; Leukothera®). A. actinomycetemcomitans is strongly associated with the development of localized aggressive periodontitis. LtxA acts as a virulence factor for A. actinomycetemcomitans to subvert the host immune response by binding to the β2 integrin lymphocyte function-associated antigen-1 (LFA-1; CD11a/CD18) on white blood cells (WBCs), causing cell death. In this paper, we reviewed the state of knowledge on LtxA interaction with WBCs and the subsequent mechanisms of induced cell death. Finally, we touched on the potential therapeutic applications of LtxA (trade name Leukothera®) toxin therapy for the treatment of hematological malignancies and immune-mediated diseases.
Collapse
Affiliation(s)
- Brian A Vega
- Department of Oral Biology, Rutgers School of Dental Medicine, Newark, NJ 07103, USA
- Actinobac Biomed, Inc., Princeton, NJ 08540, USA
| | | | - Scott C Kachlany
- Department of Oral Biology, Rutgers School of Dental Medicine, Newark, NJ 07103, USA.
- Actinobac Biomed, Inc., Princeton, NJ 08540, USA.
| |
Collapse
|
9
|
Gaylo-Moynihan A, Prizant H, Popović M, Fernandes NRJ, Anderson CS, Chiou KK, Bell H, Schrock DC, Schumacher J, Capece T, Walling BL, Topham DJ, Miller J, Smrcka AV, Kim M, Hughson A, Fowell DJ. Programming of Distinct Chemokine-Dependent and -Independent Search Strategies for Th1 and Th2 Cells Optimizes Function at Inflamed Sites. Immunity 2019; 51:298-309.e6. [PMID: 31399281 PMCID: PMC6904228 DOI: 10.1016/j.immuni.2019.06.026] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 06/18/2019] [Accepted: 06/26/2019] [Indexed: 02/08/2023]
Abstract
T-helper (Th) cell differentiation drives specialized gene programs that dictate effector T cell function at sites of infection. Here, we have shown Th cell differentiation also imposes discrete motility gene programs that shape Th1 and Th2 cell navigation of the inflamed dermis. Th1 cells scanned a smaller tissue area in a G protein-coupled receptor (GPCR) and chemokine-dependent fashion, while Th2 cells scanned a larger tissue area independent of GPCR signals. Differential chemokine reliance for interstitial migration was linked to STAT6 transcription-factor-dependent programming of integrin αVβ3 expression: Th2 cell differentiation led to high αVβ3 expression relative to Th1 cells. Th1 and Th2 cell modes of motility could be switched simply by manipulating the amount of αVβ3 on the cell surface. Deviating motility modes from those established during differentiation impaired effector function. Thus, programmed expression of αVβ3 tunes effector T cell reliance on environmental cues for optimal exploration of inflamed tissues.
Collapse
Affiliation(s)
- Alison Gaylo-Moynihan
- David H. Smith Center for Vaccine Biology and Immunology, Aab Institute of Biomedical Sciences, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Hen Prizant
- David H. Smith Center for Vaccine Biology and Immunology, Aab Institute of Biomedical Sciences, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Milan Popović
- David H. Smith Center for Vaccine Biology and Immunology, Aab Institute of Biomedical Sciences, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Ninoshka R J Fernandes
- David H. Smith Center for Vaccine Biology and Immunology, Aab Institute of Biomedical Sciences, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA; Department of Biomedical Engineering, University of Rochester, Rochester, NY 14642, USA
| | - Christopher S Anderson
- David H. Smith Center for Vaccine Biology and Immunology, Aab Institute of Biomedical Sciences, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Kevin K Chiou
- Department of Physics and Astronomy, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hannah Bell
- David H. Smith Center for Vaccine Biology and Immunology, Aab Institute of Biomedical Sciences, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Dillon C Schrock
- David H. Smith Center for Vaccine Biology and Immunology, Aab Institute of Biomedical Sciences, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Justin Schumacher
- Department of Biomedical Engineering, University of Rochester, Rochester, NY 14642, USA
| | - Tara Capece
- David H. Smith Center for Vaccine Biology and Immunology, Aab Institute of Biomedical Sciences, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Brandon L Walling
- David H. Smith Center for Vaccine Biology and Immunology, Aab Institute of Biomedical Sciences, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - David J Topham
- David H. Smith Center for Vaccine Biology and Immunology, Aab Institute of Biomedical Sciences, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Jim Miller
- David H. Smith Center for Vaccine Biology and Immunology, Aab Institute of Biomedical Sciences, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Alan V Smrcka
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Minsoo Kim
- David H. Smith Center for Vaccine Biology and Immunology, Aab Institute of Biomedical Sciences, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Angela Hughson
- David H. Smith Center for Vaccine Biology and Immunology, Aab Institute of Biomedical Sciences, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Deborah J Fowell
- David H. Smith Center for Vaccine Biology and Immunology, Aab Institute of Biomedical Sciences, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA.
| |
Collapse
|
10
|
Harrison DL, Fang Y, Huang J. T-Cell Mechanobiology: Force Sensation, Potentiation, and Translation. FRONTIERS IN PHYSICS 2019; 7:45. [PMID: 32601597 PMCID: PMC7323161 DOI: 10.3389/fphy.2019.00045] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
A T cell is a sensitive self-referential mechanical sensor. Mechanical forces influence the recognition, activation, differentiation, and function throughout the lifetime of a T cell. T cells constantly perceive and respond to physical stimuli through their surface receptors, cytoskeleton, and subcellular structures. Surface receptors receive physical cues in the form of forces generated through receptor-ligand binding events, which are dynamically regulated by contact tension, shear stress, and substrate rigidity. The resulting mechanotransduction not only influences T-cell recognition and signaling but also possibly modulates cell metabolism and gene expression. Moreover, forces also dynamically regulate the deformation, organization, and translocation of cytoskeleton and subcellular structures, leading to changes in T-cell mobility, migration, and infiltration. However, the roles and mechanisms of how mechanical forces modulate T-cell recognition, signaling, metabolism, and gene expression, are largely unknown and underappreciated. Here, we review recent technological and scientific advances in T-cell mechanobiology, discuss possible roles and mechanisms of T-cell mechanotransduction, and propose new research directions of this emerging field in health and disease.
Collapse
Affiliation(s)
- Devin L. Harrison
- The Graduate Program in Biophysical Sciences, The University of Chicago, Chicago, IL, United States
| | - Yun Fang
- The Graduate Program in Biophysical Sciences, The University of Chicago, Chicago, IL, United States
- Section of Pulmonary and Critical Care, Department of Medicine, The University of Chicago, Chicago, IL, United States
| | - Jun Huang
- The Graduate Program in Biophysical Sciences, The University of Chicago, Chicago, IL, United States
- Institute for Molecular Engineering, The University of Chicago, Chicago, IL, United States
| |
Collapse
|
11
|
Ščigalková I, Bystroňová J, Kovářová L, Pravda M, Velebný V, Riabov V, Klüter H, Kzhyshkowska J, Vrana NE. The effect of healing phenotype-inducing cytokine formulations within soft hydrogels on encapsulated monocytes and incoming immune cells. RSC Adv 2019; 9:21396-21404. [PMID: 35521319 PMCID: PMC9066154 DOI: 10.1039/c9ra02878a] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 06/17/2019] [Indexed: 11/21/2022] Open
Abstract
Hydrogels made from the derivatives of gelatin and hyaluronic acid were used as coatings to control the immune responses.
Collapse
Affiliation(s)
| | | | - Lenka Kovářová
- Contipro a.s
- 561 02 Dolni Dobrouc
- Czech Republic
- Institute of Physical Chemistry
- Faculty of Chemistry
| | | | | | - Vladimir Riabov
- Institute for Transfusion Medicine and Immunology
- Medical Faculty Mannheim
- University of Heidelberg
- 68167 Mannheim
- Germany
| | - Harald Klüter
- Institute for Transfusion Medicine and Immunology
- Medical Faculty Mannheim
- University of Heidelberg
- 68167 Mannheim
- Germany
| | - Julia Kzhyshkowska
- Institute for Transfusion Medicine and Immunology
- Medical Faculty Mannheim
- University of Heidelberg
- 68167 Mannheim
- Germany
| | - Nihal Engin Vrana
- Protip Medical
- 67000 Strasbourg
- France
- Inserm UMR 1121, Biomaterials and Bioengineering
- 67085 Strasbourg
| |
Collapse
|
12
|
Mechanics of antigen extraction in the B cell synapse. Mol Immunol 2018; 101:319-328. [PMID: 30036798 DOI: 10.1016/j.molimm.2018.07.018] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 07/11/2018] [Indexed: 12/16/2022]
Abstract
B cell encounter with antigen displayed on antigen-presenting cells leads to B cell immune synapse formation, internalisation of the antigen, and stimulation of antibody responses. The sensitivity with which B cells detect antigen, and the quality and quantity of antigen that B cells acquire, depend upon mechanical properties of the immune synapse including interfacial tension, the strength of intermolecular bonds, and the compliance of the molecules and membranes that participate in antigen presentation. In this review, we discuss our current understanding of how these various physical parameters influence B cell antigen extraction in the immune synapse and how a more comprehensive understanding of B cell mechanics may promote the development of new approaches to stimulate the production of desired antibodies.
Collapse
|
13
|
Plotnikov EY, Silachev DN, Popkov VA, Zorova LD, Pevzner IB, Zorov SD, Jankauskas SS, Babenko VA, Sukhikh GT, Zorov DB. Intercellular Signalling Cross-Talk: To Kill, To Heal and To Rejuvenate. Heart Lung Circ 2017; 26:648-659. [DOI: 10.1016/j.hlc.2016.12.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2016] [Revised: 11/22/2016] [Accepted: 12/06/2016] [Indexed: 12/16/2022]
|
14
|
Mattila PK, Batista FD, Treanor B. Dynamics of the actin cytoskeleton mediates receptor cross talk: An emerging concept in tuning receptor signaling. J Cell Biol 2016; 212:267-80. [PMID: 26833785 PMCID: PMC4748574 DOI: 10.1083/jcb.201504137] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Recent evidence implicates the actin cytoskeleton in the control of receptor signaling. This may be of particular importance in the context of immune receptors, such as the B cell receptor, where dysregulated signaling can result in autoimmunity and malignancy. Here, we discuss the role of the actin cytoskeleton in controlling receptor compartmentalization, dynamics, and clustering as a means to regulate receptor signaling through controlling the interactions with protein partners. We propose that the actin cytoskeleton is a point of integration for receptor cross talk through modulation of protein dynamics and clustering. We discuss the implication of this cross talk via the cytoskeleton for both ligand-induced and low-level constitutive (tonic) signaling necessary for immune cell survival.
Collapse
Affiliation(s)
- Pieta K Mattila
- Institute of Biomedicine, MediCity, University of Turku, 20520 Turku, Finland
| | - Facundo D Batista
- Lymphocyte Interaction Laboratory, The Francis Crick Institute, Cancer Research UK, London WC2A 3LY, England, UK
| | - Bebhinn Treanor
- Department of Biological Sciences, University of Toronto Scarborough, Toronto, Ontario M5T 1C6, Canada
| |
Collapse
|
15
|
Comrie WA, Burkhardt JK. Action and Traction: Cytoskeletal Control of Receptor Triggering at the Immunological Synapse. Front Immunol 2016; 7:68. [PMID: 27014258 PMCID: PMC4779853 DOI: 10.3389/fimmu.2016.00068] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2015] [Accepted: 02/12/2016] [Indexed: 01/03/2023] Open
Abstract
It is well known that F-actin dynamics drive the micron-scale cell shape changes required for migration and immunological synapse (IS) formation. In addition, recent evidence points to a more intimate role for the actin cytoskeleton in promoting T cell activation. Mechanotransduction, the conversion of mechanical input into intracellular biochemical changes, is thought to play a critical role in several aspects of immunoreceptor triggering and downstream signal transduction. Multiple molecules associated with signaling events at the IS have been shown to respond to physical force, including the TCR, costimulatory molecules, adhesion molecules, and several downstream adapters. In at least some cases, it is clear that the relevant forces are exerted by dynamics of the T cell actomyosin cytoskeleton. Interestingly, there is evidence that the cytoskeleton of the antigen-presenting cell also plays an active role in T cell activation, by countering the molecular forces exerted by the T cell at the IS. Since actin polymerization is itself driven by TCR and costimulatory signaling pathways, a complex relationship exists between actin dynamics and receptor activation. This review will focus on recent advances in our understanding of the mechanosensitive aspects of T cell activation, paying specific attention to how F-actin-directed forces applied from both sides of the IS fit into current models of receptor triggering and activation.
Collapse
Affiliation(s)
- William A Comrie
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania , Philadelphia, PA , USA
| | - Janis K Burkhardt
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania , Philadelphia, PA , USA
| |
Collapse
|
16
|
Slator PJ, Cairo CW, Burroughs NJ. Detection of Diffusion Heterogeneity in Single Particle Tracking Trajectories Using a Hidden Markov Model with Measurement Noise Propagation. PLoS One 2015; 10:e0140759. [PMID: 26473352 PMCID: PMC4608688 DOI: 10.1371/journal.pone.0140759] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 09/30/2015] [Indexed: 11/25/2022] Open
Abstract
We develop a Bayesian analysis framework to detect heterogeneity in the diffusive behaviour of single particle trajectories on cells, implementing model selection to classify trajectories as either consistent with Brownian motion or with a two-state (diffusion coefficient) switching model. The incorporation of localisation accuracy is essential, as otherwise false detection of switching within a trajectory was observed and diffusion coefficient estimates were inflated. Since our analysis is on a single trajectory basis, we are able to examine heterogeneity between trajectories in a quantitative manner. Applying our method to the lymphocyte function-associated antigen 1 (LFA-1) receptor tagged with latex beads (4 s trajectories at 1000 frames s−1), both intra- and inter-trajectory heterogeneity were detected; 12–26% of trajectories display clear switching between diffusive states dependent on condition, whilst the inter-trajectory variability is highly structured with the diffusion coefficients being related by D1 = 0.68D0 − 1.5 × 104 nm2 s−1, suggestive that on these time scales we are detecting switching due to a single process. Further, the inter-trajectory variability of the diffusion coefficient estimates (1.6 × 102 − 2.6 × 105 nm2 s−1) is very much larger than the measurement uncertainty within trajectories, suggesting that LFA-1 aggregation and cytoskeletal interactions are significantly affecting mobility, whilst the timescales of these processes are distinctly different giving rise to inter- and intra-trajectory variability. There is also an ‘immobile’ state (defined as D < 3.0 × 103 nm2 s−1) that is rarely involved in switching, immobility occurring with the highest frequency (47%) under T cell activation (phorbol-12-myristate-13-acetate (PMA) treatment) with enhanced cytoskeletal attachment (calpain inhibition). Such ‘immobile’ states frequently display slow linear drift, potentially reflecting binding to a dynamic actin cortex. Our methods allow significantly more information to be extracted from individual trajectories (ultimately limited by time resolution and time-series length), and allow statistical comparisons between trajectories thereby quantifying inter-trajectory heterogeneity. Such methods will be highly informative for the construction and fitting of molecule mobility models within membranes incorporating aggregation, binding to the cytoskeleton, or traversing membrane microdomains.
Collapse
Affiliation(s)
- Paddy J. Slator
- Systems Biology Centre, University of Warwick, Coventry, United Kingdom
- Systems Biology Doctoral Training Centre, University of Warwick, Coventry, United Kingdom
| | | | - Nigel J. Burroughs
- Systems Biology Centre, University of Warwick, Coventry, United Kingdom
- * E-mail:
| |
Collapse
|
17
|
Reyes R, Monjas A, Yánez-Mó M, Cardeñes B, Morlino G, Gilsanz A, Machado-Pineda Y, Lafuente E, Monk P, Sánchez-Madrid F, Cabañas C. Different states of integrin LFA-1 aggregation are controlled through its association with tetraspanin CD9. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1853:2464-80. [PMID: 26003300 DOI: 10.1016/j.bbamcr.2015.05.018] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Revised: 05/11/2015] [Accepted: 05/14/2015] [Indexed: 12/19/2022]
Abstract
The tetraspanin CD9 has been shown to interact with different members of the β1 and β3 subfamilies of integrins, regulating through these interactions cell adhesion, migration and signaling. Based on confocal microscopy co-localization and on co-immunoprecipitation results, we report here that CD9 associates with the β2 integrin LFA-1 in different types of leukocytes including T, B and monocytic cells. This association is resistant to stringent solubilization conditions which, together with data from chemical crosslinking, in situ Proximity Ligation Assays and pull-down experiments, suggest a primary/direct type of interaction mediated by the Large Extracellular Loop of the tetraspanin. CD9 exerts inhibitory effects on the adhesive function of LFA-1 and on LFA-1-dependent leukocyte cytotoxic activity. The mechanism responsible for this negative regulation exerted by CD9 on LFA-1 adhesion does not involve changes in the affinity state of this integrin but seems to be related to alterations in its state of aggregation.
Collapse
Affiliation(s)
- Raquel Reyes
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), 28049 Madrid, Spain; Departamento de Biología, Facultad de Ciencias, Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Alicia Monjas
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), 28049 Madrid, Spain
| | - María Yánez-Mó
- Unidad de Investigación, Hospital Santa Cristina, Instituto de Investigación Sanitaria La Princesa (IIS-IP), 28006 Madrid, Spain; Departamento de Biología Molecular, Facultad de Ciencias, Universidad Autonoma de Madrid, 28049 Madrid, Spain
| | - Beatriz Cardeñes
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), 28049 Madrid, Spain
| | - Giulia Morlino
- Departamento de Biología Vascular e Inflamación, Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain
| | - Alvaro Gilsanz
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), 28049 Madrid, Spain
| | | | - Esther Lafuente
- Departamento de Microbiología I, Area de Inmunología, Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Peter Monk
- University of Sheffield Medical School, Sheffield S10 2RX, UK
| | - Francisco Sánchez-Madrid
- Departamento de Biología Vascular e Inflamación, Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain; Servicio de Inmunología, Hospital de la Princesa, Instituto de Investigación Sanitaria La Princesa (IIS-IP), 28006 Madrid, Spain
| | - Carlos Cabañas
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), 28049 Madrid, Spain; Departamento de Microbiología I, Area de Inmunología, Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain.
| |
Collapse
|
18
|
Ostuni MA, Guellec J, Hermand P, Durand P, Combadière C, Pincet F, Deterre P. CX3CL1, a chemokine finely tuned to adhesion: critical roles of the stalk glycosylation and the membrane domain. Biol Open 2014; 3:1173-82. [PMID: 25395671 PMCID: PMC4265755 DOI: 10.1242/bio.20149845] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The multi-domain CX3CL1 transmembrane chemokine triggers leukocyte adherence without rolling and migration by presenting its chemokine domain (CD) to its receptor CX3CR1. Through the combination of functional adhesion assays with structural analysis using FRAP, we investigated the functional role of the other domains of CX3CL1, i.e., its mucin stalk, transmembrane domain, and cytosolic domain. Our results indicate that the CX3CL1 molecular structure is finely adapted to capture CX3CR1 in circulating cells and that each domain has a specific purpose: the mucin stalk is stiffened by its high glycosylation to present the CD away from the membrane, the transmembrane domain generates the permanent aggregation of an adequate amount of monomers to guarantee adhesion and prevent rolling, and the cytosolic domain ensures adhesive robustness by interacting with the cytoskeleton. We propose a model in which quasi-immobile CX3CL1 bundles are organized to quickly generate adhesive patches with sufficiently high strength to capture CX3CR1+ leukocytes but with sufficiently low strength to allow their patrolling behavior.
Collapse
Affiliation(s)
- Mariano A Ostuni
- INSERM, U 1135, Centre d'Immunologie et des Maladies Infectieuses, F-75013, Paris, France Sorbonne Universités, UPMC Université Paris 06, UMRS CR7, Centre d'Immunologie et des Maladies Infectieuses, F-75013, Paris, France CNRS, ERL 8255, Centre d'Immunologie et des Maladies Infectieuses, F-75013, Paris, France Present address: INSERM, U 1134, Biologie Intégrée du Globule Rouge; Université Paris Diderot; Institut National de la Transfusion Sanguine, 6 rue Alexandre Cabanel, 75015, Paris, France
| | - Julie Guellec
- INSERM, U 1135, Centre d'Immunologie et des Maladies Infectieuses, F-75013, Paris, France Sorbonne Universités, UPMC Université Paris 06, UMRS CR7, Centre d'Immunologie et des Maladies Infectieuses, F-75013, Paris, France CNRS, ERL 8255, Centre d'Immunologie et des Maladies Infectieuses, F-75013, Paris, France
| | - Patricia Hermand
- INSERM, U 1135, Centre d'Immunologie et des Maladies Infectieuses, F-75013, Paris, France Sorbonne Universités, UPMC Université Paris 06, UMRS CR7, Centre d'Immunologie et des Maladies Infectieuses, F-75013, Paris, France CNRS, ERL 8255, Centre d'Immunologie et des Maladies Infectieuses, F-75013, Paris, France
| | - Pauline Durand
- Sorbonne Universités, UPMC Université Paris 06, UMR 94550 ENS Laboratoire de Physique Statistique, F-75005, Paris, France
| | - Christophe Combadière
- INSERM, U 1135, Centre d'Immunologie et des Maladies Infectieuses, F-75013, Paris, France Sorbonne Universités, UPMC Université Paris 06, UMRS CR7, Centre d'Immunologie et des Maladies Infectieuses, F-75013, Paris, France CNRS, ERL 8255, Centre d'Immunologie et des Maladies Infectieuses, F-75013, Paris, France
| | - Frédéric Pincet
- Sorbonne Universités, UPMC Université Paris 06, UMR 94550 ENS Laboratoire de Physique Statistique, F-75005, Paris, France
| | - Philippe Deterre
- INSERM, U 1135, Centre d'Immunologie et des Maladies Infectieuses, F-75013, Paris, France Sorbonne Universités, UPMC Université Paris 06, UMRS CR7, Centre d'Immunologie et des Maladies Infectieuses, F-75013, Paris, France CNRS, ERL 8255, Centre d'Immunologie et des Maladies Infectieuses, F-75013, Paris, France
| |
Collapse
|
19
|
Minner DE, Rauch P, Käs J, Naumann CA. Polymer-tethered lipid multi-bilayers: a biomembrane-mimicking cell substrate to probe cellular mechano-sensing. SOFT MATTER 2014; 10:1189-1198. [PMID: 24652490 DOI: 10.1039/c3sm52298a] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Cells tiptoe through their environment forming highly localized and dynamic focal contacts. Experiments on polymeric gels of adjustable elasticity have shown that cells probe the viscoelasticity of their environment through an adaptive process of focal contact assembly/disassembly that critically affects cell adhesion, morphology, and motility. However, the specific mechanisms of this process have not yet been fully revealed. Here we report, for the first time, that fibroblast adhesion, morphology, and migration can also be controlled by altering the number of bilayers in a stack of multiple polymer-tethered lipid bilayers stabilized via maleimide-sulfhydral coupling chemistry. The observed changes in cell morphology, migration, and cytoskeletal organization in response to bilayer stacking correspond well with those previously observed on polymeric substrates of different polymer crosslinking density suggesting that variations in bilayer stacking are associated with changes in substrate viscoelasticity. This is in conceptual agreement with the existing knowledge about the structural, dynamic, and mechanical properties of polymer-lipid composite materials. Several distinct features, such as the lateral mobility of individual cell linkers and the immobilization of linker clusters, make the described substrates highly attractive tools for the study of dynamic, mechano-regulated cell linkages and cellular mechano-sensing.
Collapse
Affiliation(s)
- Daniel E Minner
- Department of Chemistry and Chemical Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202-3274, USA.
| | | | | | | |
Collapse
|
20
|
Luo J, Xu T, Li C, Ba X, Wang X, Jiang Y, Zeng X. p85-RhoGDI2, a novel complex, is required for PSGL-1-induced β1 integrin-mediated lymphocyte adhesion to VCAM-1. Int J Biochem Cell Biol 2013; 45:2764-73. [PMID: 24055812 DOI: 10.1016/j.biocel.2013.09.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Revised: 08/30/2013] [Accepted: 09/10/2013] [Indexed: 01/04/2023]
Abstract
P-selectin glycoprotein ligand-1 and β1 integrin play essential roles in T cell trafficking during inflammation. E-selectin and vascular cell adhesion molecule-1 are their ligands expressed on inflammation-activated endothelium. During the tethering and rolling of lymphocytes on endothelium, P-selectin glycoprotein ligand-1 binds E-selectin and induces signals. Subsequently, β1 integrin is activated and mediates stable adhesion. However, the intracellular signal pathways from PSGL-1 to β1 integrin have not yet been fully understood. Here, we find that p85, a regulatory subunit of phosphoinositide 3-kinase, forms a novel complex with Rho-GDP dissociation inhibitor-2, a lymphocyte-specific RhoGTPases dissociation inhibitor. Phosporylations of the p85-bound Rho-GDP dissociation inhibitor-2 on 130 and 153 tyrosine residues by c-Abl and Src were required for the complex to be recruited to P-selectin glycoprotein ligand-1 and thereby regulate β1 integrin-mediated T cell adhesion to vascular cell adhesion molecule-1. Both shRNAs to Rho-GDP dissociation inhibitor-2 and p85 and over-expression of Rho-GDP dissociation inhibitor-2 Y130F and Y153F significantly reduced the above-mentioned adhesion. Although Rho-GDP dissociation inhibitor-2 in the p85-Rho-GDP dissociation inhibitor-2 complex was also phosphorylated on 24 tyrosine residue by Syk, the phosphorylation is not required for the adhesion. Taken together, we find that specific phosphorylations on 130 and 153 tyrosine residues of p85-bound Rho-GDP dissociation inhibitor-2 are pivotal for P-selectin glycoprotein ligand-1-induced β1 integrin-mediated lymphocyte adhesion to vascular cell adhesion molecule-1. This will shed new light on the mechanisms that connect leukocyte initial rolling with subsequent adhesion.
Collapse
Affiliation(s)
- Jixian Luo
- Institute of Genetics and Cytology, Northeast Normal University, Changchun, China; Department of Bioscience, Shanxi University, Taiyuan, China
| | | | | | | | | | | | | |
Collapse
|
21
|
CD36 recruits α₅β₁ integrin to promote cytoadherence of P. falciparum-infected erythrocytes. PLoS Pathog 2013; 9:e1003590. [PMID: 24009511 PMCID: PMC3757042 DOI: 10.1371/journal.ppat.1003590] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2013] [Accepted: 07/15/2013] [Indexed: 12/15/2022] Open
Abstract
The adhesion of Plasmodium falciparum-infected erythrocytes (IRBC) to receptors on different host cells plays a divergent yet critical role in determining the progression and outcome of the infection. Based on our ex vivo studies with clinical parasite isolates from adult Thai patients, we have previously proposed a paradigm for IRBC cytoadherence under physiological shear stress that consists of a recruitment cascade mediated largely by P-selectin, ICAM-1 and CD36 on primary human dermal microvascular endothelium (HDMEC). In addition, we detected post-adhesion signaling events involving Src family kinases and the adaptor protein p130CAS in endothelial cells that lead to CD36 clustering and cytoskeletal rearrangement which enhance the magnitude of the adhesive strength, allowing adherent IRBC to withstand shear stress of up to 20 dynes/cm2. In this study, we addressed whether CD36 supports IRBC adhesion as part of an assembly of membrane receptors. Using a combination of flow chamber assay, atomic force and confocal microscopy, we showed for the first time by loss- and gain-of function assays that in the resting state, the integrin α5β1 does not support adhesive interactions between IRBC and HDMEC. Upon IRBC adhesion to CD36, the integrin is recruited either passively as part of a molecular complex with CD36, or actively to the site of IRBC attachment through phosphorylation of Src family kinases, a process that is Ca2+-dependent. Clustering of β1 integrin is associated with an increase in IRBC recruitment as well as in adhesive strength after attachment (∼40% in both cases). The adhesion of IRBC to a multimolecular complex on the surface of endothelial cells could be of critical importance in enabling adherent IRBC to withstand the high shear stress in the microcirculations. Targeting integrins may provide a novel approach to decrease IRBC cytoadherence to microvascular endothelium. Of the several species of malaria parasites that infect humans, disease caused by Plasmodium falciparum is responsible for most of the deaths. The unique pathological finding of this infection is the intense adhesion of infected red blood cells (IRBC) in the microcirculation, resulting in obstruction to blood flow and organ dysfunction. The focus of our research is to identify the molecules on host endothelial cells that support the adhesion as potential therapeutic targets. In this report, we showed for the first time a functional association between CD36, a well studied adhesion molecule for parasite adhesion, and α5β1, a member of the integrin family of adhesion molecules that are important for adhesion of leukocytes to blood vessels and cell adhesion to the extracellular matrix. We found that by itself, α5β1 integrin does not support IRBC adhesion. When IRBC adhere to CD36, the integrin is recruited to the site of adhesion through activation of the Src family kinase signaling pathway. As a result, both the number of adherent IRBC and the strength with which they adhere is greatly increased. These results demonstrate that IRBC adhesion is a dynamic and complex process. We need to identify each of the functional components to design anti-adhesive therapy.
Collapse
|
22
|
Robert P, Touchard D, Bongrand P, Pierres A. Biophysical description of multiple events contributing blood leukocyte arrest on endothelium. Front Immunol 2013; 4:108. [PMID: 23750158 PMCID: PMC3654224 DOI: 10.3389/fimmu.2013.00108] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Accepted: 04/26/2013] [Indexed: 12/16/2022] Open
Abstract
Blood leukocytes have a remarkable capacity to bind to and stop on specific blood vessel areas. Many studies have disclosed a key role of integrin structural changes following the interaction of rolling leukocytes with surface-bound chemoattractants. However, the functional significance of structural data and mechanisms of cell arrest are incompletely understood. Recent experiments revealed the unexpected complexity of several key steps of cell-surface interaction: (i) ligand-receptor binding requires a minimum amount of time to proceed and this is influenced by forces. (ii) Also, molecular interactions at interfaces are not fully accounted for by the interaction properties of soluble molecules. (iii) Cell arrest depends on nanoscale topography and mechanical properties of the cell membrane, and these properties are highly dynamic. Here, we summarize these results and we discuss their relevance to recent functional studies of integrin-receptor association in cells from a patient with type III leukocyte adhesion deficiency. It is concluded that an accurate understanding of all physical events listed in this review is needed to unravel the precise role of the multiple molecules and biochemical pathway involved in arrest triggering.
Collapse
Affiliation(s)
- Philippe Robert
- Laboratoire Adhésion and Inflammation, Aix-Marseille Université Marseille, France ; Institut National de la Santé et de la Recherche Médicale Marseille, France ; Centre National de la Recherche Scientifique Marseille, France ; Laboratoire d'Immunologie, Hôpitaux de Marseille, Hôpital de la Conception Marseille, France
| | | | | | | |
Collapse
|
23
|
Alenghat FJ, Golan DE. Membrane protein dynamics and functional implications in mammalian cells. CURRENT TOPICS IN MEMBRANES 2013; 72:89-120. [PMID: 24210428 PMCID: PMC4193470 DOI: 10.1016/b978-0-12-417027-8.00003-9] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The organization of the plasma membrane is both highly complex and highly dynamic. One manifestation of this dynamic complexity is the lateral mobility of proteins within the plane of the membrane, which is often an important determinant of intermolecular protein-binding interactions, downstream signal transduction, and local membrane mechanics. The mode of membrane protein mobility can range from random Brownian motion to immobility and from confined or restricted motion to actively directed motion. Several methods can be used to distinguish among the various modes of protein mobility, including fluorescence recovery after photobleaching, single-particle tracking, fluorescence correlation spectroscopy, and variations of these techniques. Here, we present both a brief overview of these methods and examples of their use to elucidate the dynamics of membrane proteins in mammalian cells-first in erythrocytes, then in erythroblasts and other cells in the hematopoietic lineage, and finally in non-hematopoietic cells. This multisystem analysis shows that the cytoskeleton frequently governs modes of membrane protein motion by stably anchoring the proteins through direct-binding interactions, by restricting protein diffusion through steric interactions, or by facilitating directed protein motion. Together, these studies have begun to delineate mechanisms by which membrane protein dynamics influence signaling sequelae and membrane mechanical properties, which, in turn, govern cell function.
Collapse
Affiliation(s)
- Francis J. Alenghat
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts, USA
| | - David E. Golan
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts, USA
- Hematology Division, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| |
Collapse
|
24
|
Wu X, Yu T, Bullard DC, Kucik DF. SDF-1α (CXCL12) regulation of lateral mobility contributes to activation of LFA-1 adhesion. Am J Physiol Cell Physiol 2012; 303:C666-72. [PMID: 22875786 DOI: 10.1152/ajpcell.00190.2012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Regulation of integrin activity enables leukocytes to circulate freely, avoiding inappropriate adhesion while maintaining the ability to adhere quickly at sites of infection or inflammation. This regulation involves at least two components: affinity for ligand and affinity-independent avidity effects such as lateral mobility. Using lymphocyte function associated antigen-1 (LFA-1) as a model, we investigated the role of integrin release from cytoskeletal motion constraints in response to the chemokine stromal cell-derived factor-1 (SDF-1α) in this process. All experiments were done in primary T cells to avoid nonphysiological activation processes often seen with the use of cell lines. We found that SDF-1α releases LFA-1 from cytoskeletal constraints as effectively as does cytochalasin D. The resultant increased diffusion is correlated with a robust increase in LFA-1-mediated adhesion under physiological shear stress. We further investigated the role of the highly conserved GFFKR sequence in the LFA-1 cytoplasmic domain. We report that the GFFKR sequence is both necessary and sufficient for regulation of the SDF-1α-triggered proadhesive release from cytoskeleton interactions. While this does not address the role of transient SDF-1α-induced conformational changes in the activation process, these results strongly suggest that any model of chemokine-induced LFA-1 activation must take into account chemokine-induced integrin lateral mobility. In addition, these results have ramifications for models of differential binding of LFA-1 to surface-bound vs. soluble intercellular adhesion molecule-1.
Collapse
Affiliation(s)
- Xing Wu
- Univ. of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | | | | | |
Collapse
|
25
|
Rullo J, Becker H, Hyduk SJ, Wong JC, Digby G, Arora PD, Cano AP, Hartwig J, McCulloch CA, Cybulsky MI. Actin polymerization stabilizes α4β1 integrin anchors that mediate monocyte adhesion. ACTA ACUST UNITED AC 2012; 197:115-29. [PMID: 22472442 PMCID: PMC3317807 DOI: 10.1083/jcb.201107140] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Rapid signaling and structural adaptations to the actin cytoskeleton enable leukocytes to stabilize α4 integrin–mediated adhesion and resist detachment from inflamed endothelium. Leukocytes arrested on inflamed endothelium via integrins are subjected to force imparted by flowing blood. How leukocytes respond to this force and resist detachment is poorly understood. Live-cell imaging with Lifeact-transfected U937 cells revealed that force triggers actin polymerization at upstream α4β1 integrin adhesion sites and the adjacent cortical cytoskeleton. Scanning electron microscopy revealed that this culminates in the formation of structures that anchor monocyte adhesion. Inhibition of actin polymerization resulted in cell deformation, displacement, and detachment. Transfection of dominant-negative constructs and inhibition of function or expression revealed key signaling steps required for upstream actin polymerization and adhesion stabilization. These included activation of Rap1, phosphoinositide 3-kinase γ isoform, and Rac but not Cdc42. Thus, rapid signaling and structural adaptations enable leukocytes to stabilize adhesion and resist detachment forces.
Collapse
Affiliation(s)
- Jacob Rullo
- Toronto General Research Institute, University Health Network, Toronto, M5G 2C4 Ontario, Canada.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Lateral mobility of individual integrin nanoclusters orchestrates the onset for leukocyte adhesion. Proc Natl Acad Sci U S A 2012; 109:4869-74. [PMID: 22411821 DOI: 10.1073/pnas.1116425109] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Integrins are cell membrane adhesion receptors involved in morphogenesis, immunity, tissue healing, and metastasis. A central, yet unresolved question regarding the function of integrins is how these receptors regulate both their conformation and dynamic nanoscale organization on the membrane to generate adhesion-competent microclusters upon ligand binding. Here we exploit the high spatial (nanometer) accuracy and temporal resolution of single-dye tracking to dissect the relationship between conformational state, lateral mobility, and microclustering of the integrin receptor lymphocyte function-associated antigen 1 (LFA-1) expressed on immune cells. We recently showed that in quiescent monocytes, LFA-1 preorganizes in nanoclusters proximal to nanoscale raft components. We now show that these nanoclusters are primarily mobile on the cell surface with a small (ca. 5%) subset of conformational-active LFA-1 nanoclusters preanchored to the cytoskeleton. Lateral mobility resulted crucial for the formation of microclusters upon ligand binding and for stable adhesion under shear flow. Activation of high-affinity LFA-1 by extracellular Ca(2+) resulted in an eightfold increase on the percentage of immobile nanoclusters and cytoskeleton anchorage. Although having the ability to bind to their ligands, these active nanoclusters failed to support firm adhesion in static and low shear-flow conditions because mobility and clustering capacity were highly compromised. Altogether, our work demonstrates an intricate coupling between conformation and lateral diffusion of LFA-1 and further underscores the crucial role of mobility for the onset of LFA-1 mediated leukocyte adhesion.
Collapse
|
27
|
Yang XH, Mirchev R, Deng X, Yacono P, Yang HL, Golan DE, Hemler ME. CD151 restricts the α6 integrin diffusion mode. J Cell Sci 2012; 125:1478-87. [PMID: 22328509 DOI: 10.1242/jcs.093963] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Laminin-binding integrins (α3β1, α6β1, α6β4, α7β1) are almost always expressed together with tetraspanin CD151. In every coexpressing cell analyzed to date, CD151 makes a fundamental contribution to integrin-dependent motility, invasion, morphology, adhesion and/or signaling. However, there has been minimal mechanistic insight into how CD151 affects integrin functions. In MDA-MB-231 mammary cells, tetraspanin CD151 knockdown impairs α6 integrin clustering and functions without decreasing α6 integrin expression or activation. Furthermore, CD151 knockdown minimally affects the magnitude of α6 integrin diffusion, as measured using single particle tracking. Instead, CD151 knockdown has a novel and unexpected dysregulating effect on the mode of α6 integrin diffusion. In control cells α6 integrin shows mostly random-confined diffusion (RCD) and some directed motion (DMO). In sharp contrast, in CD151-knockdown cells α6 integrin shows mostly DMO. In control cells α6 diffusion mode is sensitive to actin disruption, talin knockdown and phorbol ester stimulation. By contrast, CD151 knockdown cell α6 integrin is sensitive to actin disruption but desensitized to talin knockdown or phorbol ester stimulation, indicating dysregulation. Both phorbol ester and EGF stimulate cell spreading and promote α6 RCD in control cells. By contrast, CD151-ablated cells retain EGF effects but lose phorbol-ester-stimulated spreading and α6 RCD. For α6 integrins, physical association with CD151 promotes α6 RCD, in support of α6-mediated cable formation and adhesion. By comparison, for integrins not associated with CD151 (e.g. αv integrins), CD151 affects neither diffusion mode nor αv function. Hence, CD151 support of α6 RCD is specific and functionally relevant, and probably underlies diverse CD151 functions in skin, kidney and cancer cells.
Collapse
Affiliation(s)
- Xiuwei H Yang
- Dana-Farber Cancer Institute, 44 Binney Street, Boston, MA 02115, USA.
| | | | | | | | | | | | | |
Collapse
|
28
|
Abstract
Integrins play critical adhesion and signaling roles during development, wound healing, immunity, and cancer. Central to their function is a unique ability to dynamically modulate their adhesiveness and signaling properties through changes in conformation, both homo- and heterotypic protein-protein interactions and cellular distribution. Genetic, biochemical and structural studies have been instrumental in uncovering overall functions, describing ligand and regulatory protein interactions and elucidating the molecular architecture of integrins. However, such approaches alone are inadequate to describe how dynamic integrin behaviors are orchestrated in intact cells. To fill this void, a wide array of distinct light microscopy (largely fluorescence-based) imaging approaches have been developed and employed. Various microscopy technologies, including wide-field, optical sectioning (laser-scanning confocal, spinning-disk confocal, and multiphoton), TIRF and range of novel "Super-Resolution" techniques have been used in combination with diverse imaging modalities (such as IRM, FRET, FRAP, CALI, and fluorescence speckle imaging) to address distinct aspects of integrin function and regulation. This chapter provides an overview of these imaging approaches and how they have advanced our understanding of integrins.
Collapse
Affiliation(s)
- Christopher V Carman
- Center for Vascular Biology Research, Division of Molecular and Vascular Medicine, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
29
|
Schürpf T, Springer TA. Regulation of integrin affinity on cell surfaces. EMBO J 2011; 30:4712-27. [PMID: 21946563 DOI: 10.1038/emboj.2011.333] [Citation(s) in RCA: 139] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2011] [Accepted: 08/22/2011] [Indexed: 11/09/2022] Open
Abstract
Lymphocyte activation triggers adhesiveness of lymphocyte function-associated antigen-1 (LFA-1; integrin α(L)β(2)) for intercellular adhesion molecules (ICAMs) on endothelia or antigen-presenting cells. Whether the activation signal, after transmission through multiple domains to the ligand-binding αI domain, results in affinity changes for ligand has been hotly debated. Here, we present the first comprehensive measurements of LFA-1 affinities on T lymphocytes for ICAM-1 under a broad array of activating conditions. Only a modest increase in affinity for soluble ligand was detected after activation by chemokine or T-cell receptor ligation, conditions that primed LFA-1 and robustly induced lymphocyte adhesion to ICAM-1 substrates. By stabilizing well-defined LFA-1 conformations by Fab, we demonstrate the absolute requirement of the open LFA-1 headpiece for adhesiveness and high affinity. Interaction of primed LFA-1 with immobilized but not soluble ICAM-1 triggers energy-dependent affinity maturation of LFA-1 to an adhesive, high affinity state. Our results lend support to the traction or translational motion dependence of integrin activation.
Collapse
Affiliation(s)
- Thomas Schürpf
- Department of Pathology, Harvard Medical School, Immune Disease Institute and Children's Hospital, Boston, MA, USA
| | | |
Collapse
|
30
|
Pazdrak K, Young TW, Straub C, Stafford S, Kurosky A. Priming of eosinophils by GM-CSF is mediated by protein kinase CbetaII-phosphorylated L-plastin. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2011; 186:6485-96. [PMID: 21525390 PMCID: PMC3100773 DOI: 10.4049/jimmunol.1001868] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The priming of eosinophils by cytokines leading to augmented response to chemoattractants and degranulating stimuli is a characteristic feature of eosinophils in the course of allergic inflammation and asthma. Actin reorganization and integrin activation are implicated in eosinophil priming by GM-CSF, but their molecular mechanism of action is unknown. In this regard, we investigated the role of L-plastin, an eosinophil phosphoprotein that we identified from eosinophil proteome analysis. Phosphoproteomic analysis demonstrated the upregulation of phosphorylated L-plastin after eosinophil stimulation with GM-CSF. Additionally, coimmunoprecipitation studies demonstrated a complex formation of phosphorylated L-plastin with protein kinase CβII (PKCβII), GM-CSF receptor α-chain, and two actin-associated proteins, paxilin and cofilin. Inhibition of PKCβII with 4,5-bis(4-fluoroanilino)phtalimide or PKCβII-specific small interfering RNA blocked GM-CSF-induced phosphorylation of L-plastin. Furthermore, flow cytometric analysis also showed an upregulation of α(M)β(2) integrin, which was sensitive to PKCβII inhibition. In chemotaxis assay, GM-CSF treatment allowed eosinophils to respond to lower concentrations of eotaxin, which was abrogated by the above-mentioned PKCβII inhibitors. Similarly, inhibition of PKCβII blocked GM-CSF induced priming for degranulation as assessed by release of eosinophil cationic protein and eosinophil peroxidase in response to eotaxin. Importantly, eosinophil stimulation with a synthetic L-plastin peptide (residues 2-19) phosphorylated on Ser(5) upregulated α(M)β(2) integrin expression and increased eosinophil migration in response to eotaxin independent of GM-CSF stimulation. Our results establish a causative role for PKCβII and L-plastin in linking GM-CSF-induced eosinophil priming for chemotaxis and degranulation to signaling events associated with integrin activation via induction of PKCβII-mediated L-plastin phosphorylation.
Collapse
Affiliation(s)
- Konrad Pazdrak
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555
- UTMB National Heart, Lung, and Blood Institute Proteomics Center, University of Texas Medical Branch, Galveston, TX 77555
| | - Travis W. Young
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555
- UTMB National Heart, Lung, and Blood Institute Proteomics Center, University of Texas Medical Branch, Galveston, TX 77555
| | - Christof Straub
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555
- UTMB National Heart, Lung, and Blood Institute Proteomics Center, University of Texas Medical Branch, Galveston, TX 77555
| | - Susan Stafford
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555
- UTMB National Heart, Lung, and Blood Institute Proteomics Center, University of Texas Medical Branch, Galveston, TX 77555
| | - Alexander Kurosky
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555
- UTMB National Heart, Lung, and Blood Institute Proteomics Center, University of Texas Medical Branch, Galveston, TX 77555
| |
Collapse
|
31
|
Robert P, Canault M, Farnarier C, Nurden A, Grosdidier C, Barlogis V, Bongrand P, Pierres A, Chambost H, Alessi MC. A novel leukocyte adhesion deficiency III variant: kindlin-3 deficiency results in integrin- and nonintegrin-related defects in different steps of leukocyte adhesion. THE JOURNAL OF IMMUNOLOGY 2011; 186:5273-83. [PMID: 21441448 DOI: 10.4049/jimmunol.1003141] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Leukocyte adhesion deficiency type III is a recently described condition involving a Glanzmann-type bleeding syndrome and leukocyte adhesion deficiency. This was ascribed to a defect of the FERMT3 gene resulting in abnormal expression of kindlin-3, a protein expressed in hematopoietic cells with a major role in the regulation of integrin activation. In this article, we describe a patient with a new mutation of FERMT3 and lack of kindlin-3 expression in platelets and leukocytes. We assayed quantitatively the first steps of kindlin-3-defective leukocyte adhesion, namely, initial bond formation, bond strengthening, and early spreading. Initial bond formation was readily stimulated with neutrophils stimulated by fMLF, and neutrophils and lymphocytes stimulated by a phorbol ester or Mn(2+). In contrast, attachment strengthening was defective in the patient's lymphocytes treated with PMA or Mn(2+), or fMLF-stimulated neutrophils. However, attachment strengthening was normal in patient's neutrophils treated with phorbol ester or Mn(2+). In addition, the patient's T lymphocytes displayed defective integrin-mediated spreading and a moderate but significant decrease of spreading on anti-CD3-coated surfaces. Patient's neutrophils displayed a drastic alteration of integrin-mediated spreading after fMLF or PMA stimulation, whereas signaling-independent Mn(2+) allowed significant spreading. In conclusion, the consequences of kindlin-3 deficiency on β(2) integrin function depend on both cell type and the stimulus used for integrin activation. Our results suggest looking for a possible kindlin-3 involvement in membrane dynamical event independent of integrin-mediated adhesion.
Collapse
Affiliation(s)
- Philippe Robert
- Laboratoire d'Immunologie, Hôpital de la Conception, Assistance Publique-Hôpitaux de Marseille, 13385 Marseille Cedex 05, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Calabia-Linares C, Robles-Valero J, de la Fuente H, Perez-Martinez M, Martín-Cofreces N, Alfonso-Pérez M, Gutierrez-Vázquez C, Mittelbrunn M, Ibiza S, Urbano-Olmos FR, Aguado-Ballano C, Sánchez-Sorzano CO, Sanchez-Madrid F, Veiga E. Endosomal clathrin drives actin accumulation at the immunological synapse. J Cell Sci 2011; 124:820-30. [DOI: 10.1242/jcs.078832] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Antigen-specific cognate interaction of T lymphocytes with antigen-presenting cells (APCs) drives major morphological and functional changes in T cells, including actin rearrangements at the immune synapse (IS) formed at the cell–cell contact area. Here we show, using cell lines as well as primary cells, that clathrin, a protein involved in endocytic processes, drives actin accumulation at the IS. Clathrin is recruited towards the IS with parallel kinetics to that of actin. Knockdown of clathrin prevents accumulation of actin and proteins involved in actin polymerization, such as dynamin-2, the Arp2/3 complex and CD2AP at the IS. The clathrin pool involved in actin accumulation at the IS is linked to multivesicular bodies that polarize to the cell–cell contact zone, but not to plasma membrane or Golgi complex. These data underscore the role of clathrin as a platform for the recruitment of proteins that promote actin polymerization at the interface of T cells and APCs.
Collapse
Affiliation(s)
- Carmen Calabia-Linares
- Servicio de Inmunología, Hospital Universitario de la Princesa, Instituto de Investigación Sanitaria Hospital de la Princesa (IP), Diego de León, 62, 28006 Madrid, Spain
| | - Javier Robles-Valero
- Servicio de Inmunología, Hospital Universitario de la Princesa, Instituto de Investigación Sanitaria Hospital de la Princesa (IP), Diego de León, 62, 28006 Madrid, Spain
| | - Hortensia de la Fuente
- Servicio de Inmunología, Hospital Universitario de la Princesa, Instituto de Investigación Sanitaria Hospital de la Princesa (IP), Diego de León, 62, 28006 Madrid, Spain
| | - Manuel Perez-Martinez
- Servicio de Inmunología, Hospital Universitario de la Princesa, Instituto de Investigación Sanitaria Hospital de la Princesa (IP), Diego de León, 62, 28006 Madrid, Spain
| | - Noa Martín-Cofreces
- Servicio de Inmunología, Hospital Universitario de la Princesa, Instituto de Investigación Sanitaria Hospital de la Princesa (IP), Diego de León, 62, 28006 Madrid, Spain
- Department Biología Vascular e Inflamación, Centro Nacional de Investigaciones Cardiovasculares, Melchor Fernández Almagro s/n, 28029 Madrid, Spain
| | - Manuel Alfonso-Pérez
- Servicio de Inmunología, Hospital Universitario de la Princesa, Instituto de Investigación Sanitaria Hospital de la Princesa (IP), Diego de León, 62, 28006 Madrid, Spain
| | - Cristina Gutierrez-Vázquez
- Department Biología Vascular e Inflamación, Centro Nacional de Investigaciones Cardiovasculares, Melchor Fernández Almagro s/n, 28029 Madrid, Spain
| | - María Mittelbrunn
- Department Biología Vascular e Inflamación, Centro Nacional de Investigaciones Cardiovasculares, Melchor Fernández Almagro s/n, 28029 Madrid, Spain
| | - Sales Ibiza
- Department Biología Vascular e Inflamación, Centro Nacional de Investigaciones Cardiovasculares, Melchor Fernández Almagro s/n, 28029 Madrid, Spain
| | - Francisco R. Urbano-Olmos
- Laboratorio de Microscopía Electrónica de Transmisión, Facultad de Medicina, Universidad Autónoma de Madrid, Arzobispo Morcillo s/n, 28029 Madrid, Spain
| | - Covadonga Aguado-Ballano
- Laboratorio de Microscopía Electrónica de Transmisión, Facultad de Medicina, Universidad Autónoma de Madrid, Arzobispo Morcillo s/n, 28029 Madrid, Spain
| | - Carlos Oscar Sánchez-Sorzano
- Unidad de Biocomputación, Centro Nacional de Biotecnología (CSIC), Campus Universidad Autónoma s/n, 28049 Cantoblanco, Madrid
| | - Francisco Sanchez-Madrid
- Servicio de Inmunología, Hospital Universitario de la Princesa, Instituto de Investigación Sanitaria Hospital de la Princesa (IP), Diego de León, 62, 28006 Madrid, Spain
- Facultad de Medicina, Universidad Autónoma de Madrid, 28029 Madrid, Spain
| | - Esteban Veiga
- Servicio de Inmunología, Hospital Universitario de la Princesa, Instituto de Investigación Sanitaria Hospital de la Princesa (IP), Diego de León, 62, 28006 Madrid, Spain
- Facultad de Medicina, Universidad Autónoma de Madrid, 28029 Madrid, Spain
- Centro Nacional de Biotecnología (CSIC), Campus Universidad Autónoma s/n, 28049 Cantoblanco, Madrid
| |
Collapse
|
33
|
Hart R, Greaves DR. Chemerin contributes to inflammation by promoting macrophage adhesion to VCAM-1 and fibronectin through clustering of VLA-4 and VLA-5. THE JOURNAL OF IMMUNOLOGY 2010; 185:3728-39. [PMID: 20720202 DOI: 10.4049/jimmunol.0902154] [Citation(s) in RCA: 125] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Chemerin is a potent macrophage chemoattractant protein. We used murine peritoneal exudate cells (PECs) in adhesion, flow cytometry, and confocal microscopy assays to test the hypothesis that chemerin can also contribute to inflammation by promoting macrophage adhesion. Chemerin stimulated the adhesion of PECs to the extracellular matrix protein fibronectin and to the adhesion molecule VCAM-1 within a minute, with an EC(50) of 322 and 196 pM, respectively. Experiments using pertussis toxin and PECs from ChemR23(-/-) mice demonstrated that chemerin stimulated the adhesion of macrophages via the Gi protein-coupled receptor ChemR23. Blocking Abs against integrin subunits revealed that 89% of chemerin-stimulated adhesion to fibronectin was dependent on increased avidity of the integrin VLA-5 (alpha(5)beta(1)) and that 88% of adhesion to VCAM-1 was dependent on increased avidity of VLA-4 (alpha(4)beta(1)). Although chemerin was unable to induce an increase in integrin affinity as judged by the binding of soluble ligand, experiments using confocal microscopy revealed an increase in valency resulting from integrin clustering as the mechanism responsible for chemerin-stimulated macrophage adhesion. PI3K, Akt, and p38 were identified as key signaling mediators in chemerin-stimulated adhesion. The finding that chemerin can rapidly stimulate macrophage adhesion to extracellular matrix proteins and adhesion molecules, taken together with its ability to promote chemotaxis, suggests a novel role for chemerin in the recruitment and retention of macrophages at sites of inflammation.
Collapse
Affiliation(s)
- Rosie Hart
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | | |
Collapse
|
34
|
Alexandropoulos K, Regelmann AG. Regulation of T-lymphocyte physiology by the Chat-H/CasL adapter complex. Immunol Rev 2010; 232:160-74. [PMID: 19909363 DOI: 10.1111/j.1600-065x.2009.00831.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
The Cas family of proteins consists of at least four members implicated in the regulation of diverse cellular processes such as cell proliferation, adhesion, motility, and cancer cell metastasis. Cas family members have conserved C-termini that mediate constitutive heterotypic interactions with members of a different group of proteins, the NSP family. Both the Cas and NSP proteins have conserved domains that mediate protein-protein interactions with other cytoplasmic intermediates. Signaling modules assembled by these proteins in turn regulate signal transduction downstream of a variety of receptors including integrin, chemokine, and antigen receptors. T lymphocytes express the NSP protein NSP3/Chat-H and the Cas protein Hef1/CasL, which are found in a constitutive complex in naive T cells. We recently showed that Chat-H and Hef1/CasL regulate integrin-mediated adhesion and promote T-cell migration and trafficking downstream of activated chemokine receptors. It is currently unclear if the Chat-H/CasL module also plays a role in antigen receptor signaling. Here we review our current knowledge of how Chat-H and Hef1/CasL regulate T-cell physiology and whether this protein complex plays a functional role downstream of T-cell receptor activation.
Collapse
Affiliation(s)
- Konstantina Alexandropoulos
- Department of Medicine, Division of Clinical Immunology, The Immunology Institute, Mount Sinai School of Medicine, New York, NY 10029, USA.
| | | |
Collapse
|
35
|
Yu T, Wu X, Gupta KB, Kucik DF. Affinity, lateral mobility, and clustering contribute independently to beta 2-integrin-mediated adhesion. Am J Physiol Cell Physiol 2010; 299:C399-410. [PMID: 20445173 DOI: 10.1152/ajpcell.00039.2009] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Affinity changes and avidity modulation both contribute to activation of beta(2)-integrin-mediated adhesion, an essential, early step in inflammation. Avidity modulation, defined as an increase in adhesiveness independent of integrin conformational changes, might be due to integrin clustering, motion, or both. Increased integrin diffusion upon leukocyte activation has been demonstrated, but whether it is proadhesive in itself, or just constitutes a mechanism for integrin clustering, remains unclear. To understand the proadhesive effects of integrin affinity changes, clustering, and motion, an experimental system was devised to separate them. Clustering and integrin motion together were induced by cytochalasin D (CD) without inducing high-affinity; integrin motion could then be frozen by fixation; and high affinity was induced independently by Mn(2+). Adhesion was equivalent for fixed and unfixed cells except following pretreatment with CD or Mn(2+), which increased adhesion for both. However, fixed cells were less adhesive than unfixed cells after CD, even though integrin clustering was similar. A simple explanation is that CD induces both clustering and integrin motion, fixation then stops motion on fixed cells, but integrins continue to diffuse on unfixed cells, increasing the kinetics of integrin/ICAM-1 interactions to enhance adhesion. Affinity changes are then independent of, and additive to, avidity effects.
Collapse
Affiliation(s)
- Tao Yu
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama 35294, USA
| | | | | | | |
Collapse
|
36
|
Eckert RE, Neuder LE, Park J, Adler KB, Jones SL. Myristoylated alanine-rich C-kinase substrate (MARCKS) protein regulation of human neutrophil migration. Am J Respir Cell Mol Biol 2010; 42:586-94. [PMID: 19574534 PMCID: PMC2874444 DOI: 10.1165/rcmb.2008-0394oc] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2008] [Accepted: 05/29/2009] [Indexed: 01/01/2023] Open
Abstract
Neutrophil migration into infected tissues is essential for host defense, but products of activated neutrophils can be quite damaging to host cells. Neutrophil influx into the lung and airways and resultant inflammation characterizes diseases such as chronic obstructive pulmonary disease, bronchiectasis, and cystic fibrosis. To migrate, neutrophils must reorganize the actin cytoskeleton to establish a leading edge pseudopod and a trailing edge uropod. The actin-binding protein myristoylated alanine-rich C-kinase substrate (MARCKS) has been shown to bind and cross-link actin in a variety of cell types and to co-localize with F-actin in the leading edge lamellipodium of migrating fibroblasts. The hypothesis that MARCKS has a role in the regulation of neutrophil migration was tested using a cell-permeant peptide derived from the MARCKS myristoylated aminoterminus (MANS peptide). Treatment of isolated human neutrophils with MANS significantly inhibited both their migration and beta2 integrin-dependent adhesion in response to N-formyl-methionyl-leucyl-phenylalanine (fMLF), IL-8, or leukotriene (LT)B(4). The IC(50) for fMLF-induced migration and adhesion was 17.1 microM and 12.5 microM, respectively. MANS significantly reduced the F-actin content in neutrophils 30 seconds after fMLF stimulation, although the peptide did not alter the ability of cells to polarize or spread. MANS did not alter fMLF-induced increases in surface beta2 integrin expression. These results suggest that MARCKS, via its myristoylated aminoterminus, is a key regulator of neutrophil migration and adhesion.
Collapse
Affiliation(s)
- Rachael E. Eckert
- Department of Clinical Sciences, Department of Molecular Biomedical Sciences, and Center for Comparative Medicine and Translational Research, College of Veterinary Medicine, North Caroline State University, Raleigh, North Carolina
| | - Laura E. Neuder
- Department of Clinical Sciences, Department of Molecular Biomedical Sciences, and Center for Comparative Medicine and Translational Research, College of Veterinary Medicine, North Caroline State University, Raleigh, North Carolina
| | - Joungjoa Park
- Department of Clinical Sciences, Department of Molecular Biomedical Sciences, and Center for Comparative Medicine and Translational Research, College of Veterinary Medicine, North Caroline State University, Raleigh, North Carolina
| | - Kenneth B. Adler
- Department of Clinical Sciences, Department of Molecular Biomedical Sciences, and Center for Comparative Medicine and Translational Research, College of Veterinary Medicine, North Caroline State University, Raleigh, North Carolina
| | - Samuel L. Jones
- Department of Clinical Sciences, Department of Molecular Biomedical Sciences, and Center for Comparative Medicine and Translational Research, College of Veterinary Medicine, North Caroline State University, Raleigh, North Carolina
| |
Collapse
|
37
|
Nagarajan S. Mechanisms of anti-atherosclerotic functions of soy-based diets. J Nutr Biochem 2010; 21:255-60. [PMID: 19954957 DOI: 10.1016/j.jnutbio.2009.09.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2009] [Revised: 07/17/2009] [Accepted: 09/03/2009] [Indexed: 01/22/2023]
Abstract
Soy-based diets have been reported to protect against the development of atherosclerosis. However, the underlying mechanism(s) for this protection remains unknown. Although atherosclerosis was traditionally considered a disease associated with impaired lipid metabolism, in recent years the inflammatory components of atherosclerosis have been explored. Recent studies have convincingly delineated that uncontrolled chronic inflammation is the principal contributing factor for the initiation and progression of atherosclerosis. Interaction between activated monocytes and vascular endothelial cells is an early event in atherogenesis. The adhesion of leukocytes, including monocytes, to the inflamed-vascular endothelium and their transmigration into intima initiate the inflammatory processes. Following transmigration, monocytes in the intima are transformed to macrophages, which take up oxidized-LDL (oxLDL) to generate lipid-laden macrophages, also known as foam cells. Hence, in this review article the inflammatory processes associated with atherosclerosis and possible anti-inflammatory functions of soy-based diets contributing to the prevention of atherosclerosis are presented.
Collapse
Affiliation(s)
- Shanmugam Nagarajan
- Department of Microbiology and Immunology, Arkansas Children's Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, AR 72202, USA.
| |
Collapse
|
38
|
Gaborski TR, Sealander MN, Ehrenberg M, Waugh RE, McGrath JL. Image correlation microscopy for uniform illumination. J Microsc 2010; 237:39-50. [PMID: 20055917 DOI: 10.1111/j.1365-2818.2009.03300.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Image cross-correlation microscopy is a technique that quantifies the motion of fluorescent features in an image by measuring the temporal autocorrelation function decay in a time-lapse image sequence. Image cross-correlation microscopy has traditionally employed laser-scanning microscopes because the technique emerged as an extension of laser-based fluorescence correlation spectroscopy. In this work, we show that image correlation can also be used to measure fluorescence dynamics in uniform illumination or wide-field imaging systems and we call our new approach uniform illumination image correlation microscopy. Wide-field microscopy is not only a simpler, less expensive imaging modality, but it offers the capability of greater temporal resolution over laser-scanning systems. In traditional laser-scanning image cross-correlation microscopy, lateral mobility is calculated from the temporal de-correlation of an image, where the characteristic length is the illuminating laser beam width. In wide-field microscopy, the diffusion length is defined by the feature size using the spatial autocorrelation function. Correlation function decay in time occurs as an object diffuses from its original position. We show that theoretical and simulated comparisons between Gaussian and uniform features indicate the temporal autocorrelation function depends strongly on particle size and not particle shape. In this report, we establish the relationships between the spatial autocorrelation function feature size, temporal autocorrelation function characteristic time and the diffusion coefficient for uniform illumination image correlation microscopy using analytical, Monte Carlo and experimental validation with particle tracking algorithms. Additionally, we demonstrate uniform illumination image correlation microscopy analysis of adhesion molecule domain aggregation and diffusion on the surface of human neutrophils.
Collapse
Affiliation(s)
- T R Gaborski
- Department of Biomedical Engineering, School of Engineering Applied Science, University of Rochester, Rochester, NY 14627, USA
| | | | | | | | | |
Collapse
|
39
|
Weigel-Van Aken KAK. Pharmacological activation of guanine nucleotide exchange factors for the small GTPase Rap1 recruits high-affinity beta1 integrins as coreceptors for parvovirus B19: improved ex vivo gene transfer to human erythroid progenitor cells. Hum Gene Ther 2010; 20:1665-78. [PMID: 19702438 DOI: 10.1089/hum.2009.123] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Parvovirus B19 has potential as a gene therapy vector because of its restricted tropism for human erythroid progenitor cells in the bone marrow. B19 binds to the cell surface through P antigen and we identified activated beta(1) integrins as coreceptors for internalization. Because differentiation with phorbol ester induces beta(1) integrin coreceptor activity, but cell differentiation is not desirable in gene transfer to human progenitor cells and one of the downstream effectors of phorbol esters is the small GTPase Rap1, the role of Rap1 in the recruitment of beta(1) integrins on hematopoietic cells was examined. Expression of a constitutively active Rap1 (63E) was sufficient to recruit beta(1) integrin coreceptors in erythroleukemic K562 cells by inducing high-affinity integrin conformation. A crucial role of actin polymerization in Rap1-mediated beta(1) integrin recruitment was documented by complete inhibition of the 63E Rap1 effect with low-dose cytochalasin D and by the ability of a constitutively active mutant of the actin cytoskeleton regulator Rac1 to sensitize K562 cells to the pharmacological activation of endogenous Rap1, using the Rap1 exchange factor-specific 8-pCPT-2'-O-Me-cAMP [8-(4-chlorophenylthio)-2'-O-methyladenosine-3',5'-cyclic monophosphate]. Interestingly, in primary human erythroid progenitor cells, 8-pCPT-2'-O-Me-cAMP was sufficient to significantly increase B19-mediated gene transfer, suggesting that these cells possess the cytoskeleton organization capacity required for efficient recruitment of beta(1) integrins by brief pharmacological stimulation of Rap1 GTP loading. Because 8-pCPT-2'-O-Me-cAMP has been implicated in enhanced homing of progenitor cells, these results identify a novel tool with which to optimize ex vivo B19-mediated gene transfer and potentially improve homing of transduced cells by Rap1-beta(1) integrin activation with 8-pCPT-2'-O-Me-cAMP.
Collapse
Affiliation(s)
- Kirsten A K Weigel-Van Aken
- Division of Cellular and Molecular Therapy, Department of Pediatrics, Powell Gene Therapy Center, University of Florida College of Medicine, Gainesville, FL 32610, USA.
| |
Collapse
|
40
|
Das R, Cairo CW, Coombs D. A hidden Markov model for single particle tracks quantifies dynamic interactions between LFA-1 and the actin cytoskeleton. PLoS Comput Biol 2009; 5:e1000556. [PMID: 19893741 PMCID: PMC2768823 DOI: 10.1371/journal.pcbi.1000556] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2009] [Accepted: 10/06/2009] [Indexed: 12/17/2022] Open
Abstract
The extraction of hidden information from complex trajectories is a continuing problem in single-particle and single-molecule experiments. Particle trajectories are the result of multiple phenomena, and new methods for revealing changes in molecular processes are needed. We have developed a practical technique that is capable of identifying multiple states of diffusion within experimental trajectories. We model single particle tracks for a membrane-associated protein interacting with a homogeneously distributed binding partner and show that, with certain simplifying assumptions, particle trajectories can be regarded as the outcome of a two-state hidden Markov model. Using simulated trajectories, we demonstrate that this model can be used to identify the key biophysical parameters for such a system, namely the diffusion coefficients of the underlying states, and the rates of transition between them. We use a stochastic optimization scheme to compute maximum likelihood estimates of these parameters. We have applied this analysis to single-particle trajectories of the integrin receptor lymphocyte function-associated antigen-1 (LFA-1) on live T cells. Our analysis reveals that the diffusion of LFA-1 is indeed approximately two-state, and is characterized by large changes in cytoskeletal interactions upon cellular activation.
Collapse
Affiliation(s)
- Raibatak Das
- Department of Mathematics and Institute of Applied Mathematics, University of British Columbia, Vancouver, British Columbia, Canada.
| | | | | |
Collapse
|
41
|
Diez-Ahedo R, Normanno D, Esteban O, Bakker GJ, Figdor CG, Cambi A, Garcia-Parajo MF. Dynamic re-organization of individual adhesion nanoclusters in living cells by ligand-patterned surfaces. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2009; 5:1258-1263. [PMID: 19367602 DOI: 10.1002/smll.200801699] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Affiliation(s)
- Ruth Diez-Ahedo
- CIBER-Bioengineering Biomateriales and Nanomedicine and BioNanoPhotonics Group IBEC-Institut de Bioenginyeria de Catalunya Baldiri Reixac 15-21, 08028 Barcelona, Spain
| | | | | | | | | | | | | |
Collapse
|
42
|
Li D, Molldrem JJ, Ma Q. LFA-1 regulates CD8+ T cell activation via T cell receptor-mediated and LFA-1-mediated Erk1/2 signal pathways. J Biol Chem 2009; 284:21001-10. [PMID: 19483086 DOI: 10.1074/jbc.m109.002865] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
LFA-1 regulates T cell activation and signal transduction through the immunological synapse. T cell receptor (TCR) stimulation rapidly activates LFA-1, which provides unique LFA-1-dependent signals to promote T cell activation. However, the detailed molecular pathways that regulate these processes and the precise mechanism by which LFA-1 contributes to TCR activation remain unclear. We found LFA-1 directly participates in Erk1/2 signaling upon TCR stimulation in CD8+ T cells. The presence of LFA-1, not ligand binding, is required for the TCR-mediated Erk1/2 signal pathway. LFA-1-deficient T cells have defects in sustained Erk1/2 signaling and TCR/CD3 clustering, which subsequently prevents MTOC reorientation, cell cycle progression, and mitosis. LFA-1 regulates the TCR-mediated Erk1/2 signal pathway in the context of immunological synapse for recruitment and amplification of the Erk1/2 signal. In addition, LFA-1 ligation with ICAM-1 generates an additional Erk1/2 signal, which synergizes with the existing TCR-mediated Erk1/2 signal to enhance T cell activation. Thus, LFA-1 contributes to CD8+ T cell activation through two distinct signal pathways. We demonstrated that the function of LFA-1 is to enhance TCR signaling through the immunological synapse and deliver distinct signals in CD8+ T cell activation.
Collapse
Affiliation(s)
- Dan Li
- Section of Transplantation Immunology, Department of Stem Cell Transplantation and Cellular Therapy, University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030, USA
| | | | | |
Collapse
|
43
|
Abstract
Integrins are cell surface heterodimers that bind adhesion molecules expressed on other cells or in the extracellular matrix. Integrin-mediated interactions are critical for T cell development in the thymus, migration of T cells in the periphery, and induction of T cell effector functions. In resting T cells, integrins are maintained in a low affinity state. Engagement of the T cell receptor or chemokine receptors increases integrin affinity, enabling integrins to bind their ligands and initiate a signaling cascade resulting in altered cell morphology and motility. Our laboratory is interested how adapter proteins, mediators of intracellular signal transduction, regulate both signals from the T cell receptor to integrins (inside-out signaling) and (outside-in) signals from integrins into the cell.
Collapse
|
44
|
Kutsuzawa K, Tada S, Hossain S, Fukuda K, Maruyama K, Akiyama Y, Akaike T, Chowdhury EH. Disrupting actin filaments promotes efficient transfection of a leukemia cell line using cell adhesive protein-embedded carbonate apatite particles. Anal Biochem 2009; 388:164-6. [PMID: 19454213 DOI: 10.1016/j.ab.2009.02.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2008] [Revised: 01/30/2009] [Accepted: 02/03/2009] [Indexed: 11/19/2022]
Abstract
Tumor cells such as leukemia and lymphoma cells are obvious and attractive targets for gene therapy. Gene transfer and expression for cytokine and immunomodulatory molecules in various kinds of tumor cells have been shown to mediate tumor regression and antimetastatic effects. Moreover, genetically modified leukemia cells expressing costimulatory molecules or cytokines are likely to have significant therapeutic roles for patients with leukemia. One of the major hurdles to the successful implementation of these promising approaches is the lack of a suitable nanocarrier for transgene delivery and expression in a safe and effective manner. Recently, we reported on the development of a safe, efficient nanocarrier system of carbonate apatite that can assist both intracellular delivery and release of DNA, leading to very high level of transgene expression in cancer and primary cells. However, its efficiency in human lymphocytes is poor. We show here that nanocrystals of carbonate apatite, when electrostatically associated with fibronectin and/or E-cadherin-Fc, accelerated transgene delivery in a human T leukemia cell line (Jurkat). Moreover, transgene expression efficiency could be enhanced dramatically with the cell adhesive protein-embedded particles finally up to 150 times by selectively disrupting the actin filaments.
Collapse
Affiliation(s)
- K Kutsuzawa
- Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, Midori-ku, Yokohama 226-8501, Japan
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Pospieszalska MK, Ley K. Chapter 8 Modeling Leukocyte Rolling. CURRENT TOPICS IN MEMBRANES 2009. [DOI: 10.1016/s1063-5823(09)64008-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
46
|
Alon R. Chapter 6 Membrane–Cytoskeletal Platforms for Rapid Chemokine Signaling to Integrins. CURRENT TOPICS IN MEMBRANES 2009. [DOI: 10.1016/s1063-5823(09)64006-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
47
|
Craig DH, Gayer CP, Schaubert KL, Wei Y, Li J, Laouar Y, Basson MD. Increased extracellular pressure enhances cancer cell integrin-binding affinity through phosphorylation of beta1-integrin at threonine 788/789. Am J Physiol Cell Physiol 2008; 296:C193-204. [PMID: 19005162 DOI: 10.1152/ajpcell.00355.2008] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Increased extracellular pressure stimulates beta1-integrin-dependent cancer cell adhesion. We asked whether pressure-induced adhesion is mediated by changes in beta1-integrin binding affinity or avidity and whether these changes are phosphorylation dependent. We evaluated integrin affinity and clustering in human SW620 colon cancer cells by measuring differences in binding between soluble Arg-Gly-Asp (RGD)-Fc ligands and RGD-Fc-F(ab')2 multimeric complexes under ambient and 15-mmHg increased pressures. Phosphorylation of beta1-integrin S785 and T788/9 residues in SW620 and primary malignant colonocytes was assessed in parallel. We further used GD25-beta1-integrin-null murine fibroblasts stably transfected with either wild-type beta1A-integrin, S785A, TT788/9AA, or T788D mutants to investigate the role of beta1-integrin site-specific phosphorylation. SW620 binding of RGD-Fc-F(ab')2 multimeric complexes, but not soluble RGD-Fc ligands, was sensitive to integrin clustering. RGD-Fc ligand binding was significantly increased under elevated pressure, suggesting that pressure modulates beta1-integrin affinity. Pressure stimulated both beta1-integrin S785 and T788/9 phosphorylation. GD25-beta1A-integrin wild-type and S785A cells displayed an increase in adhesion to fibronectin under elevated pressure, an effect absent in beta1-integrin-null and TT788/9AA cells. T788D substitution significantly elevated basal cell adhesion but displayed no further increase under pressure. These results suggest pressure-induced cell adhesion is mediated by beta1-integrin T788/9 phosphorylation-dependent changes in integrin binding affinity.
Collapse
Affiliation(s)
- David H Craig
- Department of Surgery, John D. Dingell VA Medical Center, 4646 John R. Street, Detroit, MI 48201-1932, USA
| | | | | | | | | | | | | |
Collapse
|
48
|
Friedrichs J, Manninen A, Muller DJ, Helenius J. Galectin-3 regulates integrin alpha2beta1-mediated adhesion to collagen-I and -IV. J Biol Chem 2008; 283:32264-72. [PMID: 18806266 DOI: 10.1074/jbc.m803634200] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Galectins are a taxonomically widespread family of galactose-binding proteins of which galectin-3 is known to modulate cell adhesion. Using single cell force spectroscopy, the contribution of galectin-3 to the adhesion of Madin-Darby canine kidney (MDCK) cells to different extracellular matrix proteins was investigated. When adhering to collagen-I or -IV, some cells rapidly entered an enhanced adhesion state, marked by a significant increase in the force required for cell detachment. Galectin-3-depleted cells had an increased probability of entering the enhanced adhesion state. Adhesion enhancement was specific to integrin alpha(2)beta(1), as it was not observed when cells adhered to extracellular matrix substrates by other integrins. The adhesion phenotype of galectin-3-depleted cells was mimicked in a galactoside-deficient MDCK cell line and could be complemented by the addition of recombinant galectin-3. We propose that galectin-3 influences integrin alpha(2)beta(1)-mediated adhesion complex formation by altering receptor clustering.
Collapse
Affiliation(s)
- Jens Friedrichs
- Biotechnology Center, University of Technology Dresden, 01307 Dresden Germany
| | | | | | | |
Collapse
|
49
|
Membrane mobility of beta2 integrins and rolling associated adhesion molecules in resting neutrophils. Biophys J 2008; 95:4934-47. [PMID: 18689449 DOI: 10.1529/biophysj.108.132886] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The mobilities of transmembrane adhesion proteins are key underlying physical factors that contribute to neutrophil adhesion and arrest during inflammation. Here we present a novel (to our knowledge) fluorescence recovery after photobleaching system and a complementary analytical model to measure the mobility of the four key receptors involved in the adhesion cascade: L-selectin, PSGL-1, Mac-1, and LFA-1 for resting, spherical, and human neutrophils. In general, we find that beta(2) integrins (Mac-1, LFA-1) have mobilities 3-7 times faster than rolling associated molecules (L-selectin; PSGL-1), but that the mobilities within each of these groups are indistinguishable. Increasing temperature (room temperature versus 37 degrees C) results in increased mobility, in all cases, and the use of a bivalent antibody label (mAb versus Fab) decreases mobility, except in the case of rolling associated molecules at room temperature. Disrupting the actin cytoskeleton increased mobility except that the highest mobilities measured for integrins (D = 1.2 x 10(-9) cm(2)/s; 37 degrees C, Fab) are not affected by actin poisons and approach the expected value for free diffusion. Although evidence of cytoskeletal hindrance of integrin mobility has been found in other systems, our data suggest such hindrance does not limit bulk integrin diffusion in resting neutrophils over distances and times important for adhesive plaque formation.
Collapse
|
50
|
Cairo CW, Golan DE. T cell adhesion mechanisms revealed by receptor lateral mobility. Biopolymers 2008; 89:409-19. [PMID: 18041065 DOI: 10.1002/bip.20898] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cell surface receptors mediate the exchange of information between cells and their environment. In the case of adhesion receptors, the spatial distribution and molecular associations of the receptors are critical to their function. Therefore, understanding the mechanisms regulating the distribution and binding associations of these molecules is necessary to understand their functional regulation. Experiments characterizing the lateral mobility of adhesion receptors have revealed a set of common mechanisms that control receptor function and thus cellular behavior. The T cell provides one of the most dynamic examples of cellular adhesion. An individual T cell makes innumerable intercellular contacts with antigen presenting cells, the vascular endothelium, and many other cell types. We review here the mechanisms that regulate T cell adhesion receptor lateral mobility as a window into the molecular regulation of these systems, and we present a general framework for understanding the principles and mechanisms that are likely to be common among these and other cellular adhesion systems. We suggest that receptor lateral mobility is regulated via four major mechanisms-reorganization, recruitment, dispersion, and anchoring-and we review specific examples of T cell adhesion receptor systems that utilize one or more of these mechanisms.
Collapse
Affiliation(s)
- Christopher W Cairo
- Department of Chemistry, Alberta Ingenuity Centre for Carbohydrate Science, University of Alberta, Edmonton, Alberta, Canada T6G 2G2.
| | | |
Collapse
|