51
|
Groen J, Palanca A, Aires A, Conesa JJ, Maestro D, Rehbein S, Harkiolaki M, Villar AV, Cortajarena AL, Pereiro E. Correlative 3D cryo X-ray imaging reveals intracellular location and effect of designed antifibrotic protein-nanomaterial hybrids. Chem Sci 2021; 12:15090-15103. [PMID: 34909150 PMCID: PMC8612387 DOI: 10.1039/d1sc04183e] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 10/17/2021] [Indexed: 12/20/2022] Open
Abstract
Revealing the intracellular location of novel therapeutic agents is paramount for the understanding of their effect at the cell ultrastructure level. Here, we apply a novel correlative cryo 3D imaging approach to determine the intracellular fate of a designed protein–nanomaterial hybrid with antifibrotic properties that shows great promise in mitigating myocardial fibrosis. Cryo 3D structured illumination microscopy (cryo-3D-SIM) pinpoints the location and cryo soft X-ray tomography (cryo-SXT) reveals the ultrastructural environment and subcellular localization of this nanomaterial with spatial correlation accuracy down to 70 nm in whole cells. This novel high resolution 3D cryo correlative approach unambiguously locates the nanomaterial after overnight treatment within multivesicular bodies which have been associated with endosomal trafficking events by confocal microscopy. Moreover, this approach allows assessing the cellular response towards the treatment by evaluating the morphological changes induced. This is especially relevant for the future usage of nanoformulations in clinical practices. This correlative super-resolution and X-ray imaging strategy joins high specificity, by the use of fluorescence, with high spatial resolution at 30 nm (half pitch) provided by cryo-SXT in whole cells, without the need of staining or fixation, and can be of particular benefit to locate specific molecules in the native cellular environment in bio-nanomedicine. A novel 3D cryo correlative approach locates designed therapeutic protein–nanomaterial hybrids in whole cells with high specificity and resolution. Detection of treatment-induced morphological changes, crucial for pre-clinical studies, are revealed.![]()
Collapse
Affiliation(s)
- J Groen
- MISTRAL Beamline, Experiments Division, ALBA Synchrotron Light Source Cerdanyola del Valles 08290 Barcelona Spain
| | - A Palanca
- Instituto de Biomedicina y Biotecnologia de Cantabria (IBBTEC), University of Cantabria, CSIC 39011 Santander Spain.,Department of Anatomy and Cell Biology, University of Cantabria 39011 Santander Spain
| | - A Aires
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA) Paseo de Miramón 194 20014 Donostia San Sebastian Spain
| | - J J Conesa
- MISTRAL Beamline, Experiments Division, ALBA Synchrotron Light Source Cerdanyola del Valles 08290 Barcelona Spain .,National Center for Biotechnology CSIC (CNB-CSIC), Department of Macromolecular Structures Cantoblanco 28049 Madrid Spain
| | - D Maestro
- Instituto de Biomedicina y Biotecnologia de Cantabria (IBBTEC), University of Cantabria, CSIC 39011 Santander Spain
| | - S Rehbein
- Helmholtz-Zentrum Berlin für Materialien und Energie, Bessy II D-12489 Berlin Germany
| | - M Harkiolaki
- Beamline B24, Diamond Light Source, Harwell Science and Innovation Campus Didcot Oxfordshire OX11 0DE UK
| | - A V Villar
- Instituto de Biomedicina y Biotecnologia de Cantabria (IBBTEC), University of Cantabria, CSIC 39011 Santander Spain.,Department of Physiology and Pharmacology, University of Cantabria Avd. Herrera Oria s/n Santander Spain
| | - A L Cortajarena
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA) Paseo de Miramón 194 20014 Donostia San Sebastian Spain .,Ikerbasque, Basque Foundation for Science 48009 Bilbao Spain
| | - E Pereiro
- MISTRAL Beamline, Experiments Division, ALBA Synchrotron Light Source Cerdanyola del Valles 08290 Barcelona Spain
| |
Collapse
|
52
|
Sousa D, Lima RT, Lopes-Rodrigues V, Gonzalez E, Royo F, Xavier CPR, Falcón-Pérez JM, Vasconcelos MH. Different Ability of Multidrug-Resistant and -Sensitive Counterpart Cells to Release and Capture Extracellular Vesicles. Cells 2021; 10:cells10112886. [PMID: 34831110 PMCID: PMC8616370 DOI: 10.3390/cells10112886] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 10/20/2021] [Accepted: 10/21/2021] [Indexed: 12/24/2022] Open
Abstract
Cancer multidrug resistance (MDR) is one of the main challenges for cancer treatment efficacy. MDR is a phenomenon by which tumor cells become resistant to several unrelated drugs. Some studies have previously described the important role of extracellular vesicles (EVs) in the dissemination of a MDR phenotype. EVs’ cargo may include different players of MDR, such as microRNAS and drug-efflux pumps, which may be transferred from donor MDR cells to recipient drug-sensitive counterparts. The present work aimed to: (i) compare the ability of drug-sensitive and their MDR counterpart cells to release and capture EVs and (ii) study and relate those differences with possible distinct fate of the endocytic pathway in these counterpart cells. Our results showed that MDR cells released more EVs than their drug-sensitive counterparts and also that the drug-sensitive cells captured more EVs than their MDR counterparts. This difference in the release and capture of EVs may be associated with differences in the endocytic pathway between drug-sensitive and MDR cells. Importantly, manipulation of the recycling pathway influenced the response of drug-sensitive cells to doxorubicin treatment.
Collapse
Affiliation(s)
- Diana Sousa
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (D.S.); (R.T.L.); (V.L.-R.); (C.P.R.X.)
- Cancer Drug Resistance Group, IPATIMUP—Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal
- Department of Biological Sciences, FFUP—Faculty of Pharmacy of the University of Porto, 4050-313 Porto, Portugal
| | - Raquel T. Lima
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (D.S.); (R.T.L.); (V.L.-R.); (C.P.R.X.)
- Cancer Drug Resistance Group, IPATIMUP—Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal
- Department of Pathology, FMUP—Faculty of Medicine of the University of Porto, 4200-319 Porto, Portugal
- Cancer Signaling & Metabolism Group, IPATIMUP—Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal
| | - Vanessa Lopes-Rodrigues
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (D.S.); (R.T.L.); (V.L.-R.); (C.P.R.X.)
- Cancer Drug Resistance Group, IPATIMUP—Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal
- ICBAS-UP—Institute of Biomedical Sciences Abel Salazar of the University of Porto, 4099-003 Porto, Portugal
| | - Esperanza Gonzalez
- Exosomes Lab. & Metabolomics Platform, CIC bioGUNE, CIBERehd, 28160 Derio, Spain; (E.G.); (F.R.); (J.M.F.-P.)
| | - Félix Royo
- Exosomes Lab. & Metabolomics Platform, CIC bioGUNE, CIBERehd, 28160 Derio, Spain; (E.G.); (F.R.); (J.M.F.-P.)
| | - Cristina P. R. Xavier
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (D.S.); (R.T.L.); (V.L.-R.); (C.P.R.X.)
- Cancer Drug Resistance Group, IPATIMUP—Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal
| | - Juan M. Falcón-Pérez
- Exosomes Lab. & Metabolomics Platform, CIC bioGUNE, CIBERehd, 28160 Derio, Spain; (E.G.); (F.R.); (J.M.F.-P.)
- IKERBASQUE Basque Foundation for Science, 48013 Bilbao, Spain
| | - M. Helena Vasconcelos
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (D.S.); (R.T.L.); (V.L.-R.); (C.P.R.X.)
- Cancer Drug Resistance Group, IPATIMUP—Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal
- Department of Biological Sciences, FFUP—Faculty of Pharmacy of the University of Porto, 4050-313 Porto, Portugal
- Correspondence: ; Tel.: +351-225-570-772
| |
Collapse
|
53
|
Vitry J, Paré G, Murru A, Charest-Morin X, Maaroufi H, McLeish KR, Naccache PH, Fernandes MJ. Regulation of the Expression, Oligomerisation and Signaling of the Inhibitory Receptor CLEC12A by Cysteine Residues in the Stalk Region. Int J Mol Sci 2021; 22:ijms221910207. [PMID: 34638548 PMCID: PMC8508511 DOI: 10.3390/ijms221910207] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 09/11/2021] [Accepted: 09/15/2021] [Indexed: 01/26/2023] Open
Abstract
CLEC12A is a myeloid inhibitory receptor that negatively regulates inflammation in mouse models of autoimmune and autoinflammatory arthritis. Reduced CLEC12A expression enhances myeloid cell activation and inflammation in CLEC12A knock-out mice with collagen antibody-induced or gout-like arthritis. Similarly to other C-type lectin receptors, CLEC12A harbours a stalk domain between its ligand binding and transmembrane domains. While it is presumed that the cysteines in the stalk domain have multimerisation properties, their role in CLEC12A expression and/or signaling remain unknown. We thus used site-directed mutagenesis to determine whether the stalk domain cysteines play a role in CLEC12A expression, internalisation, oligomerisation, and/or signaling. Mutation of C118 blocks CLEC12A transport through the secretory pathway diminishing its cell-surface expression. In contrast, mutating C130 does not affect CLEC12A cell-surface expression but increases its oligomerisation, inducing ligand-independent phosphorylation of the receptor. Moreover, we provide evidence that CLEC12A dimerisation is regulated in a redox-dependent manner. We also show that antibody-induced CLEC12A cross-linking induces flotillin oligomerisation in insoluble membrane domains in which CLEC12A signals. Taken together, these data indicate that the stalk cysteines in CLEC12A differentially modulate this inhibitory receptor’s expression, oligomerisation and signaling, suggestive of the regulation of CLEC12A in a redox-dependent manner during inflammation.
Collapse
Affiliation(s)
- Julien Vitry
- CHU de Québec Research Center, Division of Infectious Diseases and Immunology, Laval University, Québec, QC G1V 4G2, Canada; (J.V.); (G.P.); (A.M.); (P.H.N.)
- Department of Microbiology-Infectious Diseases and Immunology, Faculty of Medicine, Laval University, Québec, QC G1V 4G2, Canada;
| | - Guillaume Paré
- CHU de Québec Research Center, Division of Infectious Diseases and Immunology, Laval University, Québec, QC G1V 4G2, Canada; (J.V.); (G.P.); (A.M.); (P.H.N.)
- Department of Microbiology-Infectious Diseases and Immunology, Faculty of Medicine, Laval University, Québec, QC G1V 4G2, Canada;
| | - Andréa Murru
- CHU de Québec Research Center, Division of Infectious Diseases and Immunology, Laval University, Québec, QC G1V 4G2, Canada; (J.V.); (G.P.); (A.M.); (P.H.N.)
- Department of Microbiology-Infectious Diseases and Immunology, Faculty of Medicine, Laval University, Québec, QC G1V 4G2, Canada;
| | - Xavier Charest-Morin
- Department of Microbiology-Infectious Diseases and Immunology, Faculty of Medicine, Laval University, Québec, QC G1V 4G2, Canada;
| | - Halim Maaroufi
- Institute of Integrative Biology and Systems, Laval University, Québec, QC G1V 0A6, Canada;
| | - Kenneth R. McLeish
- Department of Medicine, University of Louisville School of Medicine, Louisville, KY 40206, USA;
| | - Paul H. Naccache
- CHU de Québec Research Center, Division of Infectious Diseases and Immunology, Laval University, Québec, QC G1V 4G2, Canada; (J.V.); (G.P.); (A.M.); (P.H.N.)
- Department of Microbiology-Infectious Diseases and Immunology, Faculty of Medicine, Laval University, Québec, QC G1V 4G2, Canada;
| | - Maria J. Fernandes
- CHU de Québec Research Center, Division of Infectious Diseases and Immunology, Laval University, Québec, QC G1V 4G2, Canada; (J.V.); (G.P.); (A.M.); (P.H.N.)
- Department of Microbiology-Infectious Diseases and Immunology, Faculty of Medicine, Laval University, Québec, QC G1V 4G2, Canada;
- Correspondence: ; Tel.: +1-418-656-4141 (ext. 46106)
| |
Collapse
|
54
|
Charpentier JC, King PD. Mechanisms and functions of endocytosis in T cells. Cell Commun Signal 2021; 19:92. [PMID: 34503523 PMCID: PMC8427877 DOI: 10.1186/s12964-021-00766-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 07/17/2021] [Indexed: 11/11/2022] Open
Abstract
Once thought of primarily as a means to neutralize pathogens or to facilitate feeding, endocytosis is now known to regulate a wide range of eukaryotic cell processes. Among these are regulation of signal transduction, mitosis, lipid homeostasis, and directed migration, among others. Less well-appreciated are the roles various forms of endocytosis plays in regulating αβ and, especially, γδ T cell functions, such as T cell receptor signaling, antigen discovery by trogocytosis, and activated cell growth. Herein we examine the contribution of both clathrin-mediated and clathrin-independent mechanisms of endocytosis to T cell biology. Video Abstract
Collapse
Affiliation(s)
- John C Charpentier
- Department of Microbiology and Immunology, University of Michigan Medical School, 6606 Med Sci II, 1150 West Medical Center Drive, Ann Arbor, MI, 48109-5620, USA
| | - Philip D King
- Department of Microbiology and Immunology, University of Michigan Medical School, 6606 Med Sci II, 1150 West Medical Center Drive, Ann Arbor, MI, 48109-5620, USA.
| |
Collapse
|
55
|
Molecular characterization of direct interactions between MPP1 and flotillins. Sci Rep 2021; 11:14751. [PMID: 34285255 PMCID: PMC8292550 DOI: 10.1038/s41598-021-93982-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Accepted: 06/28/2021] [Indexed: 12/14/2022] Open
Abstract
Flotillins are the major structural proteins in erythroid raft domains. We have shown previously that the dynamic nanoscale organization of raft domains in erythroid cells may depend on flotillin-MPP1 interactions. Here, by using molecular dynamic simulations and a surface plasmon resonance-based approach we determined that high-affinity complexes of MPP1 and flotillins are formed via a so far unidentified region within the D5 domain of MPP1. Significantly, this particular “flotillin binding motif” is of key physiological importance, as overexpression of peptides containing this motif inhibited endogenous MPP1-flotillin interaction in erythroid precursor cells, thereby causing lateral disorganization of raft domains. This was reflected by both reduction in the plasma membrane order and markedly decreased activation of signal transduction via the raft-dependent insulin receptor pathway. Our data highlight new molecular details concerning the mechanism whereby MPP1 functionally links flotillins to exert their physiological role in raft domain formation.
Collapse
|
56
|
Varma S, Dey S, S P D. Cellular Uptake Pathways of Nanoparticles: Process of Endocytosis and Factors Affecting Their Fate. Curr Pharm Biotechnol 2021; 23:679-706. [PMID: 34264182 DOI: 10.2174/1389201022666210714145356] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 05/06/2021] [Accepted: 05/07/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Efficient and controlled internalization of NPs into the cells depends on their physicochemical properties and dynamics of the plasma membrane. NPs-cell interaction is a complex process that decides the fate of NPs internalization through different endocytosis pathways. OBJECTIVE The aim of this review is to highlight the physicochemical properties of synthesized nanoparticles (NPs) and their interaction with the cellular-dynamics and pathways like phagocytosis, pinocytosis, macropinocytosis, clathrin, and caveolae-mediated endocytosis and the involvement of effector proteins domain such as clathrin, AP2, caveolin, Arf6, Cdc42, dynamin and cell surface receptors during the endocytosis process of NPs. METHOD An electronic search was performed to explore the focused reviews and research articles on types of endocytosis and physicochemical properties of nanoparticles and their impact on cellular internalizations. The search was limited to peer-reviewed journals in the PubMed database. RESULTS This article discusses in detail how different types of NPs and their physicochemical properties such as size, shape, aspect ratio, surface charge, hydrophobicity, elasticity, stiffness, corona formation, surface functionalization changes the pattern of endocytosis in the presence of different pharmacological blockers. Some external forces like a magnetic field, electric field, and ultrasound exploit the cell membrane dynamics to permeabilize them for efficient internalization with respect to fundamental principles of membrane bending and pore formation. CONCLUSION This review will be useful to attract and guide the audience to understand the endocytosis mechanism and their pattern with respect to physicochemical properties of NPs to improve their efficacy and targeting to achieve the impactful outcome in drug-delivery and theranostics applications.
Collapse
Affiliation(s)
- Sameer Varma
- Department of Pharmaceutical Biotechnology, JSS Academy of Higher Education & Research- JSS College of Pharmacy, Ooty-643001, Tamil Nadu, India
| | - Smita Dey
- Department of Pharmaceutical Biotechnology, JSS Academy of Higher Education & Research- JSS College of Pharmacy, Ooty-643001, Tamil Nadu, India
| | - Dhanabal S P
- Department of Pharmacognosy & Phytopharmacy, JSS Academy of Higher Education & Research- JSS College of Pharmacy, Ooty-643001, Tamil Nadu, India
| |
Collapse
|
57
|
Wan B, Belghazi M, Lemauf S, Poirié M, Gatti JL. Proteomics of purified lamellocytes from Drosophila melanogaster HopT um-l identifies new membrane proteins and networks involved in their functions. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2021; 134:103584. [PMID: 34033897 DOI: 10.1016/j.ibmb.2021.103584] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/20/2021] [Accepted: 04/22/2021] [Indexed: 06/12/2023]
Abstract
In healthy Drosophila melanogaster larvae, plasmatocytes and crystal cells account for 95% and 5% of the hemocytes, respectively. A third type of hemocytes, lamellocytes, are rare, but their number increases after oviposition by parasitoid wasps. The lamellocytes form successive layers around the parasitoid egg, leading to its encapsulation and melanization, and finally the death of this intruder. However, the total number of lamellocytes per larva remains quite low even after parasitoid infestation, making direct biochemical studies difficult. Here, we used the HopTum-l mutant strain that constitutively produces large numbers of lamellocytes to set up a purification method and analyzed their major proteins by 2D gel electrophoresis and their plasma membrane surface proteins by 1D SDS-PAGE after affinity purification. Mass spectrometry identified 430 proteins from 2D spots and 344 affinity-purified proteins from 1D bands, for a total of 639 unique proteins. Known lamellocyte markers such as PPO3 and the myospheroid integrin were among the components identified with specific chaperone proteins. Affinity purification detected other integrins, as well as a wide range of integrin-associated proteins involved in the formation and function of cell-cell junctions. Overall, the newly identified proteins indicate that these cells are highly adapted to the encapsulation process (recognition, motility, adhesion, signaling), but may also have several other physiological functions (such as secretion and internalization of vesicles) under different signaling pathways. These results provide the basis for further in vivo and in vitro studies of lamellocytes, including the development of new markers to identify coexisting populations and their respective origins and functions in Drosophila immunity.
Collapse
Affiliation(s)
- Bin Wan
- Université Côte d'Azur, INRAE, CNRS, Institute Sophia-Agrobiotech, Sophia Antipolis, France
| | - Maya Belghazi
- Institute of NeuroPhysiopathology (INP), UMR7051, CNRS, Aix-Marseille Université, Marseille, 13015, France
| | - Séverine Lemauf
- Université Côte d'Azur, INRAE, CNRS, Institute Sophia-Agrobiotech, Sophia Antipolis, France
| | - Marylène Poirié
- Université Côte d'Azur, INRAE, CNRS, Institute Sophia-Agrobiotech, Sophia Antipolis, France
| | - Jean-Luc Gatti
- Université Côte d'Azur, INRAE, CNRS, Institute Sophia-Agrobiotech, Sophia Antipolis, France.
| |
Collapse
|
58
|
Paré G, Vitry J, Merchant ML, Vaillancourt M, Murru A, Shen Y, Elowe S, Lahoud MH, Naccache PH, McLeish KR, Fernandes MJ. The Inhibitory Receptor CLEC12A Regulates PI3K-Akt Signaling to Inhibit Neutrophil Activation and Cytokine Release. Front Immunol 2021; 12:650808. [PMID: 34234773 PMCID: PMC8256872 DOI: 10.3389/fimmu.2021.650808] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 05/21/2021] [Indexed: 12/25/2022] Open
Abstract
The myeloid inhibitory C-type lectin receptor CLEC12A limits neutrophil activation, pro-inflammatory pathways and disease in mouse models of inflammatory arthritis by a molecular mechanism that remains poorly understood. We addressed how CLEC12A-mediated inhibitory signaling counteracts activating signaling by cross-linking CLEC12A in human neutrophils. CLEC12A cross-linking induced its translocation to flotillin-rich membrane domains where its ITIM was phosphorylated in a Src-dependent manner. Phosphoproteomic analysis identified candidate signaling molecules regulated by CLEC12A that include MAPKs, phosphoinositol kinases and members of the JAK-STAT pathway. Stimulating neutrophils with uric acid crystals, the etiological agent of gout, drove the hyperphosphorylation of p38 and Akt. Ultimately, one of the pathways through which CLEC12A regulates uric acid crystal-stimulated release of IL-8 by neutrophils is through a p38/PI3K-Akt signaling pathway. In summary this work defines early molecular events that underpin CLEC12A signaling in human neutrophils to modulate cytokine synthesis. Targeting this pathway could be useful therapeutically to dampen inflammation.
Collapse
Affiliation(s)
- Guillaume Paré
- Division of Infectious Diseases and Immunology, Laval University, Centres Hospitaliers Universitaires (CHU) de Québec Research Center, Québec, QC, Canada
| | - Julien Vitry
- Division of Infectious Diseases and Immunology, Laval University, Centres Hospitaliers Universitaires (CHU) de Québec Research Center, Québec, QC, Canada.,Department of Microbiology and Immunology, Faculty of Medicine, Laval University, CHU de Québec Research Center, Québec, QC, Canada
| | - Michael L Merchant
- Department of Medicine, University of Louisville School of Medicine, Louisville, KY, United States
| | - Myriam Vaillancourt
- Division of Infectious Diseases and Immunology, Laval University, Centres Hospitaliers Universitaires (CHU) de Québec Research Center, Québec, QC, Canada
| | - Andréa Murru
- Division of Infectious Diseases and Immunology, Laval University, Centres Hospitaliers Universitaires (CHU) de Québec Research Center, Québec, QC, Canada.,Department of Microbiology and Immunology, Faculty of Medicine, Laval University, CHU de Québec Research Center, Québec, QC, Canada
| | - Yunyun Shen
- Division of Infectious Diseases and Immunology, Laval University, Centres Hospitaliers Universitaires (CHU) de Québec Research Center, Québec, QC, Canada.,Department of Microbiology and Immunology, Faculty of Medicine, Laval University, CHU de Québec Research Center, Québec, QC, Canada
| | - Sabine Elowe
- Department of Pediatrics, Faculty of Medicine, Laval University, CHU de Québec Research Center, Québec, QC, Canada.,Reproduction, Mother and Youth Health Division, Laval University, CHU de Québec Research Center, Québec, QC, Canada
| | - Mireille H Lahoud
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Paul H Naccache
- Division of Infectious Diseases and Immunology, Laval University, Centres Hospitaliers Universitaires (CHU) de Québec Research Center, Québec, QC, Canada.,Department of Microbiology and Immunology, Faculty of Medicine, Laval University, CHU de Québec Research Center, Québec, QC, Canada
| | - Kenneth R McLeish
- Department of Medicine, University of Louisville School of Medicine, Louisville, KY, United States
| | - Maria J Fernandes
- Division of Infectious Diseases and Immunology, Laval University, Centres Hospitaliers Universitaires (CHU) de Québec Research Center, Québec, QC, Canada.,Department of Microbiology and Immunology, Faculty of Medicine, Laval University, CHU de Québec Research Center, Québec, QC, Canada
| |
Collapse
|
59
|
Song Q, Wang XQ, Holmes TR, Bonkowski M, Roth EW, Ponedal A, Mirkin C, Paller AS. Epidermal SR-A Complexes Are Lipid Raft Based and Promote Nucleic Acid Nanoparticle Uptake. J Invest Dermatol 2021; 141:1428-1437.e8. [PMID: 33385397 PMCID: PMC8154648 DOI: 10.1016/j.jid.2020.10.027] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 09/13/2020] [Accepted: 10/05/2020] [Indexed: 01/22/2023]
Abstract
Scavenger receptors clear pathogens, transport lipid, and mediate polyanionic ligand uptake in macrophages, but their expression and role in the skin are poorly understood. Although the epidermal barrier typically excludes nucleic acid entry, topically applied, spherically arranged oligonucleotide nanoconjugates (spherical nucleic acids [SNAs]) penetrate mouse skin, three-dimensional (3D) skin equivalents, and human skin. We explored the mechanism of SNA uptake in normal human epidermal keratinocytes and 3D skin equivalents. Normal human epidermal keratinocytes and 3D raft treatment with SR-A inhibitors reduced SNA uptake by >80%. The human epidermis expresses SR-As SCARA3 and, to a lesser extent, MARCO. Simultaneous lentiviral knockdown of SCARA3 and MARCO reduced SNA uptake in normal human epidermal keratinocytes and 3D rafts after topical application, affirming a role for SR-As in SNA uptake and 3D raft penetration. Incubation of normal human epidermal keratinocytes at 4oC or with sodium azide prevented SNA uptake, suggesting active endocytosis. Endocytosis inhibitors, immunofluorescence, immunoprecipitation, and knockdown studies localized functional SR-As to FLOT-1-containing lipid rafts throughout the epidermis and CAV-1-containing rafts only in the upper epidermis. These studies suggest a central role for SR-A complexes in epidermal lipid rafts in mediating the uptake of nucleic acid‒laden nanoparticles.
Collapse
Affiliation(s)
- Qian Song
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Xiao-Qi Wang
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Thomas R Holmes
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Michael Bonkowski
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Eric W Roth
- Northwestern University Atomic and Nanoscale Characterization Experimental Center (NUANCE), Evanston, Illinois, USA
| | - Adam Ponedal
- Department of Chemical & Biological Engineering, Northwestern University, Evanston, Illinois, USA; International Institute for Nanotechnology, Northwestern University, Evanston, Illinois, USA
| | - Chad Mirkin
- International Institute for Nanotechnology, Northwestern University, Evanston, Illinois, USA; Department of Chemistry, Northwestern University, Evanston, Illinois, USA
| | - Amy S Paller
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA; International Institute for Nanotechnology, Northwestern University, Evanston, Illinois, USA.
| |
Collapse
|
60
|
Flotillin-Dependent Membrane Microdomains Are Required for Functional Phagolysosomes against Fungal Infections. Cell Rep 2021; 32:108017. [PMID: 32814035 PMCID: PMC10054021 DOI: 10.1016/j.celrep.2020.108017] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 04/03/2020] [Accepted: 07/17/2020] [Indexed: 11/23/2022] Open
Abstract
Lipid rafts form signaling platforms on biological membranes with incompletely characterized role in immune response to infection. Here we report that lipid-raft microdomains are essential components of phagolysosomal membranes of macrophages and depend on flotillins. Genetic deletion of flotillins demonstrates that the assembly of both major defense complexes vATPase and NADPH oxidase requires membrane microdomains. Furthermore, we describe a virulence mechanism leading to dysregulation of membrane microdomains by melanized wild-type conidia of the important human-pathogenic fungus Aspergillus fumigatus resulting in reduced phagolysosomal acidification. We show that phagolysosomes with ingested melanized conidia contain a reduced amount of free Ca2+ ions and that inhibition of Ca2+-dependent calmodulin activity led to reduced lipid-raft formation. We identify a single-nucleotide polymorphism in the human FLOT1 gene resulting in heightened susceptibility for invasive aspergillosis in hematopoietic stem cell transplant recipients. Collectively, flotillin-dependent microdomains on the phagolysosomal membrane play an essential role in protective antifungal immunity.
Collapse
|
61
|
Sousa de Almeida M, Susnik E, Drasler B, Taladriz-Blanco P, Petri-Fink A, Rothen-Rutishauser B. Understanding nanoparticle endocytosis to improve targeting strategies in nanomedicine. Chem Soc Rev 2021; 50:5397-5434. [PMID: 33666625 PMCID: PMC8111542 DOI: 10.1039/d0cs01127d] [Citation(s) in RCA: 489] [Impact Index Per Article: 122.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Indexed: 12/19/2022]
Abstract
Nanoparticles (NPs) have attracted considerable attention in various fields, such as cosmetics, the food industry, material design, and nanomedicine. In particular, the fast-moving field of nanomedicine takes advantage of features of NPs for the detection and treatment of different types of cancer, fibrosis, inflammation, arthritis as well as neurodegenerative and gastrointestinal diseases. To this end, a detailed understanding of the NP uptake mechanisms by cells and intracellular localization is essential for safe and efficient therapeutic applications. In the first part of this review, we describe the several endocytic pathways involved in the internalization of NPs and we discuss the impact of the physicochemical properties of NPs on this process. In addition, the potential challenges of using various inhibitors, endocytic markers and genetic approaches to study endocytosis are addressed along with the principal (semi) quantification methods of NP uptake. The second part focuses on synthetic and bio-inspired substances, which can stimulate or decrease the cellular uptake of NPs. This approach could be interesting in nanomedicine where a high accumulation of drugs in the target cells is desirable and clearance by immune cells is to be avoided. This review contributes to an improved understanding of NP endocytic pathways and reveals potential substances, which can be used in nanomedicine to improve NP delivery.
Collapse
Affiliation(s)
- Mauro Sousa de Almeida
- Adolphe Merkle Institute, University of FribourgChemin des Verdiers 41700 FribourgSwitzerland
| | - Eva Susnik
- Adolphe Merkle Institute, University of FribourgChemin des Verdiers 41700 FribourgSwitzerland
| | - Barbara Drasler
- Adolphe Merkle Institute, University of FribourgChemin des Verdiers 41700 FribourgSwitzerland
| | | | - Alke Petri-Fink
- Adolphe Merkle Institute, University of FribourgChemin des Verdiers 41700 FribourgSwitzerland
- Department of Chemistry, University of FribourgChemin du Musée 91700 FribourgSwitzerland
| | | |
Collapse
|
62
|
Tau internalization: A complex step in tau propagation. Ageing Res Rev 2021; 67:101272. [PMID: 33571704 DOI: 10.1016/j.arr.2021.101272] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 12/04/2020] [Accepted: 02/04/2021] [Indexed: 12/16/2022]
Abstract
Aggregation of microtubule-associated protein Tau (MAPT) may underlie abnormalities of the intracellular matrix and neuronal death in tauopathies. Tau proteins can be secreted to the extracellular space and internalized into adjacent cells. The internalization of Tau is a complex but critical step in Tau propagation. This review summarizes the internalization pathways of Tau, including macropinocytosis, Clathrin-mediated endocytosis (CME), lipid raft dependent endocytosis, Tunneling nanotubes dependent endocytosis (TNTs) and phagocytosis. The conformation of Tau fibrils and the types of recipient cell determine the internalization pathway. However, the HSPGs-dependent endocytosis seems to be the predominant pathway of Tau internalization. After internalization, Tau fibrils undergo clearance and seeding. Imbalance among Tau secretion, internalization and clearance may result in the propagation of misfolded Tau in the brain, thereby inducing Tauopathies. A better understanding of the internalization of Tau proteins may facilitate the discovery of novel therapeutic strategies to block the propagation of Tau pathology.
Collapse
|
63
|
Li X, Chen R, Kemper S, Brigstock DR. Structural and Functional Characterization of Fibronectin in Extracellular Vesicles From Hepatocytes. Front Cell Dev Biol 2021; 9:640667. [PMID: 33816490 PMCID: PMC8012540 DOI: 10.3389/fcell.2021.640667] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 02/22/2021] [Indexed: 12/21/2022] Open
Abstract
Extracellular vesicles (EVs) are membrane-limited nanoparticles that are liberated by cells and contain a complex molecular payload comprising proteins, microRNA, RNAs, and lipids. EVs may be taken up by other cells resulting in their phenotypic or functional reprogramming. In the liver, EVs produced by non-injured hepatocytes are involved in the maintenance of hepatic homeostasis or therapeutic outcomes following injury while EVs produced by damaged hepatocytes may drive or exacerbate liver injury. In this study, we examined the contribution of EV fibronectin (FN1) to the biogenesis, release, uptake, and action of hepatocyte-derived EVs. While FN1 is classically viewed as a component of the extracellular matrix that regulates processes such as cell adhesion, differentiation, and wound healing and can exist in cell-associated or soluble plasma forms, we report that FN1 is also a constituent of hepatocyte EVs that functions in EV uptake by target cells such as hepatocytes and hepatic stellate cells (HSC). FN1 co-purified with EVs when EVs were enriched from conditioned medium of human or mouse hepatocytes and a direct association between FN1 and hepatocyte EVs was established by immunoprecipitation and proteinase protection. FN1 ablation in mouse hepatocytes using CRISPR-Cas9 did not alter EV biogenesis but EV uptake by HSC was significantly reduced for FN1 knockout EVs (EVΔFN1) as compared to EVs from wild type hepatocytes (EVWT). The uptake by hepatocytes or HSC of either EVWT or EVΔFN1 required clathrin- and caveolin-mediated endocytosis, cholesterol, lysosomal acidic lipase activity, and low pH, while macropinocytosis was also involved in EVΔFN1 uptake in HSC. Despite their differences in rate and mechanisms of uptake, EVΔFN1 functioned comparably to EVWT in ameliorating CCl4-induced hepatic fibrosis in mice. In conclusion, FN1 is a constituent of hepatocyte EVs that facilitates EV uptake by target cells but is dispensable for EV-mediated anti-fibrotic activity in vivo.
Collapse
Affiliation(s)
- Xinlei Li
- Center for Clinical and Translational Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, United States
| | - Ruju Chen
- Center for Clinical and Translational Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, United States
| | - Sherri Kemper
- Center for Clinical and Translational Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, United States
| | - David R Brigstock
- Center for Clinical and Translational Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, United States.,Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| |
Collapse
|
64
|
Live-cell monitoring of protein localization to membrane rafts using protein-fragment complementation. Biosci Rep 2021; 40:221616. [PMID: 31850494 PMCID: PMC6944658 DOI: 10.1042/bsr20191290] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 12/13/2019] [Accepted: 12/17/2019] [Indexed: 12/12/2022] Open
Abstract
The plasma membrane consists of a variety of discrete domains differing from the surrounding membrane in composition and properties. Selective partitioning of protein to these microdomains is essential for membrane functioning and integrity. Studying the nanoscale size and dynamic nature of the membrane microdomains requires advanced imaging approaches with a high spatiotemporal resolution and, consequently, expensive and specialized equipment, unavailable for most researchers and unsuited for large-scale studies. Thus, understanding of protein partitioning to the membrane microdomains in health and disease is still hampered by the lack of inexpensive live-cell approaches with an appropriate spatial resolution. Here, we have developed a novel approach based on Gaussia princeps luciferase protein-fragment complementation assay to quantitively investigate protein partitioning to cholesterol and sphingomyelin-rich domains, sometimes called ‘lipid rafts’, in intact living cells with a high-spatial resolution. In the assay, the reporter construct, carrying one half of the luciferase protein, is targeted to lipid microdomains through the fused acetylation motif from Src-family kinase Fyn. A protein of interest carries the second half of the luciferase protein. Together, this serves as a reversible real-time sensor of raft recruitment for the studied protein. We demonstrated that the assay can efficiently detect the dynamic alterations in raft localization of two disease-associated proteins: Akt and APP. Importantly, this method can be used in high-throughput screenings and other large-scale studies in living cells. This inexpensive, and easy to implement raft localization assay will benefit all researchers interested in protein partitioning in rafts.
Collapse
|
65
|
Brandel A, Aigal S, Lagies S, Schlimpert M, Meléndez AV, Xu M, Lehmann A, Hummel D, Fisch D, Madl J, Eierhoff T, Kammerer B, Römer W. The Gb3-enriched CD59/flotillin plasma membrane domain regulates host cell invasion by Pseudomonas aeruginosa. Cell Mol Life Sci 2021; 78:3637-3656. [PMID: 33555391 PMCID: PMC8038999 DOI: 10.1007/s00018-021-03766-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 12/22/2020] [Accepted: 01/15/2021] [Indexed: 12/11/2022]
Abstract
The opportunistic pathogen Pseudomonas aeruginosa has gained precedence over the years due to its ability to develop resistance to existing antibiotics, thereby necessitating alternative strategies to understand and combat the bacterium. Our previous work identified the interaction between the bacterial lectin LecA and its host cell glycosphingolipid receptor globotriaosylceramide (Gb3) as a crucial step for the engulfment of P. aeruginosa via the lipid zipper mechanism. In this study, we define the LecA-associated host cell membrane domain by pull-down and mass spectrometry analysis. We unraveled a predilection of LecA for binding to saturated, long fatty acyl chain-containing Gb3 species in the extracellular membrane leaflet and an induction of dynamic phosphatidylinositol (3,4,5)-trisphosphate (PIP3) clusters at the intracellular leaflet co-localizing with sites of LecA binding. We found flotillins and the GPI-anchored protein CD59 not only to be an integral part of the LecA-interacting membrane domain, but also majorly influencing bacterial invasion as depletion of either of these host cell proteins resulted in about 50% reduced invasiveness of the P. aeruginosa strain PAO1. In summary, we report that the LecA-Gb3 interaction at the extracellular leaflet induces the formation of a plasma membrane domain enriched in saturated Gb3 species, CD59, PIP3 and flotillin thereby facilitating efficient uptake of PAO1.
Collapse
Affiliation(s)
- Annette Brandel
- Faculty of Biology, University of Freiburg, Schänzlestraße 1, 79104, Freiburg, Germany
- BIOSS, Centre for Biological Signalling Studies, University of Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany
- CIBSS, Centre for Integrative Biological Signalling Studies, University of Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany
| | - Sahaja Aigal
- Faculty of Biology, University of Freiburg, Schänzlestraße 1, 79104, Freiburg, Germany
- BIOSS, Centre for Biological Signalling Studies, University of Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany
- Institute of Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Simon Lagies
- Faculty of Biology, University of Freiburg, Schänzlestraße 1, 79104, Freiburg, Germany
- Center for Biological Systems Analysis, University of Freiburg, Habsburgerstraße 49, 79104, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine, University of Freiburg, Albertstraße 19a, 79104, Freiburg, Germany
| | - Manuel Schlimpert
- Faculty of Biology, University of Freiburg, Schänzlestraße 1, 79104, Freiburg, Germany
- Center for Biological Systems Analysis, University of Freiburg, Habsburgerstraße 49, 79104, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine, University of Freiburg, Albertstraße 19a, 79104, Freiburg, Germany
| | - Ana Valeria Meléndez
- Faculty of Biology, University of Freiburg, Schänzlestraße 1, 79104, Freiburg, Germany
- BIOSS, Centre for Biological Signalling Studies, University of Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany
- CIBSS, Centre for Integrative Biological Signalling Studies, University of Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine, University of Freiburg, Albertstraße 19a, 79104, Freiburg, Germany
| | - Maokai Xu
- Faculty of Biology, University of Freiburg, Schänzlestraße 1, 79104, Freiburg, Germany
- BIOSS, Centre for Biological Signalling Studies, University of Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany
- CIBSS, Centre for Integrative Biological Signalling Studies, University of Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany
| | - Anika Lehmann
- Faculty of Biology, University of Freiburg, Schänzlestraße 1, 79104, Freiburg, Germany
- BIOSS, Centre for Biological Signalling Studies, University of Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany
- CIBSS, Centre for Integrative Biological Signalling Studies, University of Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany
| | - Daniel Hummel
- Faculty of Biology, University of Freiburg, Schänzlestraße 1, 79104, Freiburg, Germany
- BIOSS, Centre for Biological Signalling Studies, University of Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany
- Department of Biochemistry, University of Geneva, 30 Quai Ernest-Ansermet, 1211, Geneva, Switzerland
| | - Daniel Fisch
- Faculty of Biology, University of Freiburg, Schänzlestraße 1, 79104, Freiburg, Germany
- BIOSS, Centre for Biological Signalling Studies, University of Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany
- Host-Toxoplasma Interaction Laboratory, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
- Department of Infectious Disease, MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London, SW7 2AZ, UK
| | - Josef Madl
- Faculty of Biology, University of Freiburg, Schänzlestraße 1, 79104, Freiburg, Germany
- BIOSS, Centre for Biological Signalling Studies, University of Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg - Bad Krozingen, and Faculty of Medicine, University of Freiburg, Elsässer Straße 2q, 79110, Freiburg, Germany
| | - Thorsten Eierhoff
- Faculty of Biology, University of Freiburg, Schänzlestraße 1, 79104, Freiburg, Germany
- BIOSS, Centre for Biological Signalling Studies, University of Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany
- Clinic for Vascular and Endovascular Surgery, University Hospital Münster, Albert Schweitzer Campus 1, 48149, Münster, Germany
| | - Bernd Kammerer
- BIOSS, Centre for Biological Signalling Studies, University of Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany
- Center for Biological Systems Analysis, University of Freiburg, Habsburgerstraße 49, 79104, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine, University of Freiburg, Albertstraße 19a, 79104, Freiburg, Germany
| | - Winfried Römer
- Faculty of Biology, University of Freiburg, Schänzlestraße 1, 79104, Freiburg, Germany.
- BIOSS, Centre for Biological Signalling Studies, University of Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany.
- CIBSS, Centre for Integrative Biological Signalling Studies, University of Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany.
- Spemann Graduate School of Biology and Medicine, University of Freiburg, Albertstraße 19a, 79104, Freiburg, Germany.
| |
Collapse
|
66
|
Kwok ZH, Wang C, Jin Y. Extracellular Vesicle Transportation and Uptake by Recipient Cells: A Critical Process to Regulate Human Diseases. Processes (Basel) 2021; 9. [PMID: 34336602 PMCID: PMC8323758 DOI: 10.3390/pr9020273] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Emerging evidence highlights the relevance of extracellular vesicles
(EVs) in modulating human diseases including but not limited to cancer,
inflammation, and neurological disorders. EVs can be found in almost all types
of human body fluids, suggesting that their trafficking may allow for their
targeting to remote recipient cells. While molecular processes underlying EV
biogenesis and secretion are increasingly elucidated, mechanisms governing EV
transportation, target finding and binding, as well as uptake into recipient
cells remain to be characterized. Understanding the specificity of EV transport
and uptake is critical to facilitating the development of EVs as valuable
diagnostics and therapeutics. In this mini review, we focus on EV uptake
mechanisms and specificities, as well as their implications in human
diseases.
Collapse
|
67
|
Greenlee JD, Subramanian T, Liu K, King MR. Rafting Down the Metastatic Cascade: The Role of Lipid Rafts in Cancer Metastasis, Cell Death, and Clinical Outcomes. Cancer Res 2021; 81:5-17. [PMID: 32999001 PMCID: PMC7952000 DOI: 10.1158/0008-5472.can-20-2199] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 09/01/2020] [Accepted: 09/21/2020] [Indexed: 11/16/2022]
Abstract
Lipid rafts are tightly packed, cholesterol- and sphingolipid-enriched microdomains within the plasma membrane that play important roles in many pathophysiologic processes. Rafts have been strongly implicated as master regulators of signal transduction in cancer, where raft compartmentalization can promote transmembrane receptor oligomerization, shield proteins from enzymatic degradation, and act as scaffolds to enhance intracellular signaling cascades. Cancer cells have been found to exploit these mechanisms to initiate oncogenic signaling and promote tumor progression. This review highlights the roles of lipid rafts within the metastatic cascade, specifically within tumor angiogenesis, cell adhesion, migration, epithelial-to-mesenchymal transition, and transendothelial migration. In addition, the interplay between lipid rafts and different modes of cancer cell death, including necrosis, apoptosis, and anoikis, will be described. The clinical role of lipid raft-specific proteins, caveolin and flotillin, in assessing patient prognosis and evaluating metastatic potential of various cancers will be presented. Collectively, elucidation of the complex roles of lipid rafts and raft components within the metastatic cascade may be instrumental for therapeutic discovery to curb prometastatic processes.
Collapse
Affiliation(s)
- Joshua D Greenlee
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
| | - Tejas Subramanian
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
| | - Kevin Liu
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
| | - Michael R King
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee.
| |
Collapse
|
68
|
Caveolin-1, tetraspanin CD81 and flotillins in lymphocyte cell membrane organization, signaling and immunopathology. Biochem Soc Trans 2020; 48:2387-2397. [PMID: 33242069 DOI: 10.1042/bst20190387] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 10/08/2020] [Accepted: 10/12/2020] [Indexed: 12/24/2022]
Abstract
The adaptive immune system relies on B and T lymphocytes to ensure a specific and long-lasting protection of an individual from a wide range of potential pathogenic hits. Lymphocytes are highly potent and efficient in eliminating pathogens. However, lymphocyte activation must be tightly regulated to prevent incorrect activity that could result in immunopathologies, such as autoimmune disorders or cancers. Comprehensive insight into the molecular events underlying lymphocyte activation is of enormous importance to better understand the function of the immune system. It provides the basis to design therapeutics to regulate lymphocyte activation in pathological scenarios. Most reported defects in immunopathologies affect the regulation of intracellular signaling pathways. This highlights the importance of these molecules, which control lymphocyte activation and homeostasis impacting lymphocyte tolerance to self, cytokine production and responses to infections. Most evidence for these defects comes from studies of disease models in genetically engineered mice. There is an increasing number of studies focusing on lymphocytes derived from patients which supports these findings. Many indirectly involved proteins are emerging as unexpected regulators of the immune system. In this mini-review, we focus in proteins that regulate plasma membrane (PM) compartmentalization and thereby impact the steady state and the activation of immunoreceptors, namely the T cell antigen receptor (TCR) and the B cell antigen receptor (BCR). Some of these membrane proteins are shown to be involved in immune abnormalities; others, however, are not thoroughly investigated in the context of immune pathogenesis. We aim to highlight them and stimulate future research avenues.
Collapse
|
69
|
Taga H, Dallaire MP, Gervais R, Richard FJ, Ma L, Corl BA, Chouinard PY. Characterization of raft microdomains in bovine mammary tissue during lactation: How they are modulated by fatty acid treatments. J Dairy Sci 2020; 104:2384-2395. [PMID: 33246605 DOI: 10.3168/jds.2020-19267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 09/03/2020] [Indexed: 11/19/2022]
Abstract
The objective of the current study was first to characterize lipid raft microdomains isolated as detergent-resistant membranes (DRM) from mammary gland tissue, and second to determine how dietary fatty acids (FA) such as conjugated linoleic acid (CLA), 19:1 cyclo, and long-chain n-3 polyunsaturated FA affect lipid raft markers of mammary cells, and to finally establish relationships between these markers and lactation performance in dairy cows. Eight Holstein cows were used in a replicated 4 × 4 Latin square design with periods of 28 d. For the first 14 d, cows received daily an abomasal infusion of (1) 406 g of a saturated FA supplement (112 g of 16:0 + 230 g of 18:0) used as a control; (2) 36 g of a CLA supplement (13.9 g of trans-10,cis-12 18:2) + 370 g of saturated FA; (3) 7 g of Sterculia fetida oil (3.1 g of 19:1 cyclo, STO) + 399 g of saturated FA; or (4) 406 g of fish oil (55.2 g of cis-5,cis-8,cis-11,cis-14,cis-17 20:5 + 59.3 g of cis-4,cis-7,cis-10,cis-13,cis-16,cis-19 22:6, FO). Mammary biopsies were harvested on d 14 of each infusion period and were followed by a 14-d washout interval. Cholera toxin subunit B, which specifically binds to ganglioside M-1 (GM-1), a lipid raft marker, was used to assess its distribution in DRM. Infusions of CLA, STO, and FO were individually compared with the control, and significance was declared at P ≤ 0.05. Milk fat yield was decreased with CLA and FO, but was not affected by STO. Milk lactose yield was decreased with CLA and STO, but was not affected by FO. Mammary tissue shows a strong GM-1-signal enrichment in isolated DRM from mammary gland tissue. Caveolin (CAV) and flotillin (FLOT) are 2 proteins considered as lipid raft markers and they are present in DRM from mammary gland tissue. Distributions of GM-1, CAV-1, and FLOT-1 showed an effect of treatments determined by their subcellular distributions in sucrose gradient fractions. Regardless of treatments, data showed positive relationships between the yield of milk fat, protein, and lactose, and the abundance GM-1 in DRM fraction. Milk protein yield was positively correlated with relative proportion of FLOT-1 in the soluble fraction, whereas lactose yield was positively correlated with relative proportion of CAV-1 in the DRM fractions. Infusion of CLA decreased mRNA abundance of CAV-1, FLOT-1, and FLOT-2. Regardless of treatments, a positive relationship was observed between fat yield and mRNA abundance of FLOT-2. In conclusion, although limited to a few markers, results of the current experiment raised potential links between variation in specific biologically active component of raft microdomains in bovine mammary gland and lactation performances in dairy cows.
Collapse
Affiliation(s)
- H Taga
- Département des Sciences Animales, Université Laval, Québec, QC, G1V 0A6 Canada
| | - M P Dallaire
- Département des Sciences Animales, Université Laval, Québec, QC, G1V 0A6 Canada
| | - R Gervais
- Département des Sciences Animales, Université Laval, Québec, QC, G1V 0A6 Canada
| | - F J Richard
- Département des Sciences Animales, Université Laval, Québec, QC, G1V 0A6 Canada
| | - L Ma
- Department of Dairy Science, Virginia Tech, Blacksburg 24061
| | - B A Corl
- Department of Dairy Science, Virginia Tech, Blacksburg 24061
| | - P Y Chouinard
- Département des Sciences Animales, Université Laval, Québec, QC, G1V 0A6 Canada.
| |
Collapse
|
70
|
Chhuon C, Zhang SY, Jung V, Lewandowski D, Lipecka J, Pawlak A, Sahali D, Ollero M, Guerrera IC. A sensitive S-Trap-based approach to the analysis of T cell lipid raft proteome. J Lipid Res 2020; 61:1512-1523. [PMID: 32769147 PMCID: PMC7604723 DOI: 10.1194/jlr.d120000672] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The analysis of T cell lipid raft proteome is challenging due to the highly dynamic nature of rafts and the hydrophobic character of raft-resident proteins. We explored an innovative strategy for bottom-up lipid raftomics based on suspension-trapping (S-Trap) sample preparation. Mouse T cells were prepared from splenocytes by negative immunoselection, and rafts were isolated by a detergent-free method and OptiPrep gradient ultracentrifugation. Microdomains enriched in flotillin-1, LAT, and cholesterol were subjected to proteomic analysis through an optimized protocol based on S-Trap and high pH fractionation, followed by nano-LC-MS/MS. Using this method, we identified 2,680 proteins in the raft-rich fraction and established a database of 894 T cell raft proteins. We then performed a differential analysis on the raft-rich fraction from nonstimulated versus anti-CD3/CD28 T cell receptor (TCR)-stimulated T cells. Our results revealed 42 proteins present in one condition and absent in the other. For the first time, we performed a proteomic analysis on rafts from ex vivo T cells obtained from individual mice, before and after TCR activation. This work demonstrates that the proposed method utilizing an S-Trap-based approach for sample preparation increases the specificity and sensitivity of lipid raftomics.
Collapse
Affiliation(s)
- Cerina Chhuon
- Proteomic Platform Necker, Structure Fédérative de Recherche SFR Necker US24, Paris, France
- Institut Mondor de Recherche Biomédicale, INSERM, U955, Créteil, France
| | - Shao-Yu Zhang
- Institut Mondor de Recherche Biomédicale, INSERM, U955, Créteil, France
| | - Vincent Jung
- Proteomic Platform Necker, Structure Fédérative de Recherche SFR Necker US24, Paris, France
| | - Daniel Lewandowski
- CEA/DRF/IBFJ/iRCM/LRTS, Fontenay-aux-Roses Cedex, France
- CEA/DRF/IBFJ/iRCM/LRTS, Fontenay-aux-Roses Cedex, France
- CEA/DRF/IBFJ/iRCM/LRTS, Fontenay-aux-Roses Cedex, France
- Université Paris-Sud, Paris, France
| | - Joanna Lipecka
- Proteomic Platform Necker, Structure Fédérative de Recherche SFR Necker US24, Paris, France
| | - André Pawlak
- Institut Mondor de Recherche Biomédicale, INSERM, U955, Créteil, France
| | - Dil Sahali
- Institut Mondor de Recherche Biomédicale, INSERM, U955, Créteil, France
- AP-HP (Assistance Publique des Hôpitaux de Paris), Department of Nephrology and Renal Transplantation, Groupe Hospitalier Henri-Mondor, Créteil, France
- Université Paris Est Créteil, Créteil, France
| | - Mario Ollero
- Institut Mondor de Recherche Biomédicale, INSERM, U955, Créteil, France
- Université Paris Est Créteil, Créteil, France
| | - Ida Chiara Guerrera
- Proteomic Platform Necker, Structure Fédérative de Recherche SFR Necker US24, Paris, France
| |
Collapse
|
71
|
Tsai YT, Moore W, Kim H, Budin I. Bringing rafts to life: Lessons learned from lipid organization across diverse biological membranes. Chem Phys Lipids 2020; 233:104984. [PMID: 33203526 DOI: 10.1016/j.chemphyslip.2020.104984] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 09/13/2020] [Accepted: 09/28/2020] [Indexed: 10/23/2022]
Abstract
The ability of lipids to drive lateral organization is a remarkable feature of membranes and has been hypothesized to underlie the architecture of cells. Models for lipid rafts and related domains were originally based on the mammalian plasma membrane, but the nature of heterogeneity in this system is still not fully resolved. However, the concept of lipid-driven organization has been highly influential across biology, and has led to discoveries in organisms that feature a diversity of lipid chemistries and physiological needs. Here we review several emerging and instructive cases of membrane organization in non-mammalian systems. In bacteria, several types of membrane domains that act in metabolism and signaling have been elucidated. These widen our view of what constitutes a raft, but also introduce new questions about the relationship between organization and function. In yeast, observable membrane organization is found in both the plasma membrane and the vacuole. The latter serves as the best example of classic membrane phase partitioning in a living system to date, suggesting that internal organelles are important membranes to investigate across eukaryotes. Finally, we highlight plants as powerful model systems for complex membrane interactions in multicellular organisms. Plant membranes are organized by unique glycosphingolipids, supporting the importance of carbohydrate interactions in organizing lateral domains. These examples demonstrate that membrane organization is a potentially universal phenonenon in biology and argue for the continued broadening of lipid physical chemistry research into a wide range of systems.
Collapse
Affiliation(s)
- Yi-Ting Tsai
- Department of Chemistry & Biochemistry, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, United States
| | - William Moore
- Department of Chemistry & Biochemistry, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, United States
| | - Hyesoo Kim
- Department of Chemistry & Biochemistry, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, United States
| | - Itay Budin
- Department of Chemistry & Biochemistry, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, United States.
| |
Collapse
|
72
|
Tetraspanins, More than Markers of Extracellular Vesicles in Reproduction. Int J Mol Sci 2020; 21:ijms21207568. [PMID: 33066349 PMCID: PMC7589920 DOI: 10.3390/ijms21207568] [Citation(s) in RCA: 134] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 10/08/2020] [Accepted: 10/08/2020] [Indexed: 02/07/2023] Open
Abstract
The participation of extracellular vesicles in many cellular processes, including reproduction, is unquestionable. Although currently, the tetraspanin proteins found in extracellular vesicles are mostly applied as markers, increasing evidence points to their role in extracellular vesicle biogenesis, cargo selection, cell targeting, and cell uptake under both physiological and pathological conditions. In this review, we bring other insight into the involvement of tetraspanin proteins in extracellular vesicle physiology in mammalian reproduction. We provide knowledge regarding the involvement of extracellular vesicle tetraspanins in these processes in somatic cells. Furthermore, we discuss the future direction towards an understanding of their functions in the tissues and fluids of the mammalian reproductive system in gamete maturation, fertilization, and embryo development; their involvement in mutual cell contact and communication in their complexity.
Collapse
|
73
|
Yao N, Wang CR, Liu MQ, Li YJ, Chen WM, Li ZQ, Qi Q, Lu JJ, Fan CL, Chen MF, Qi M, Li XB, Hong J, Zhang DM, Ye WC. Discovery of a novel EGFR ligand DPBA that degrades EGFR and suppresses EGFR-positive NSCLC growth. Signal Transduct Target Ther 2020; 5:214. [PMID: 33033232 PMCID: PMC7544691 DOI: 10.1038/s41392-020-00251-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 05/25/2020] [Accepted: 06/15/2020] [Indexed: 12/22/2022] Open
Abstract
Epidermal growth factor receptor (EGFR) activation plays a pivotal role in EGFR-driven non-small cell lung cancer (NSCLC) and is considered as a key target of molecular targeted therapy. EGFR tyrosine kinase inhibitors (TKIs) have been canonically used in NSCLC treatment. However, prevalent innate and acquired resistances and EGFR kinase-independent pro-survival properties limit the clinical efficacy of EGFR TKIs. Therefore, the discovery of novel EGFR degraders is a promising approach towards improving therapeutic efficacy and overcoming drug resistance. Here, we identified a 23-hydroxybetulinic acid derivative, namely DPBA, as a novel EGFR small-molecule ligand. It exerted potent in vitro and in vivo anticancer activity in both EGFR wild type and mutant NSCLC by degrading EGFR. Mechanistic studies disclosed that DPBA binds to the EGFR extracellular domain at sites differing from those of EGF and EGFR. DPBA did not induce EGFR dimerization, phosphorylation, and ubiquitination, but it significantly promoted EGFR degradation and repressed downstream survival pathways. Further analyses showed that DPBA induced clathrin-independent EGFR endocytosis mediated by flotillin-dependent lipid rafts and unaffected by EGFR TKIs. Activation of the early and late endosome markers rab5 and rab7 but not the recycling endosome marker rab11 was involved in DPBA-induced EGFR lysosomal degradation. The present study offers a new EGFR ligand for EGFR pharmacological degradation and proposes it as a potential treatment for EGFR-positive NSCLC, particularly NSCLC with innate or acquired EGFR TKI resistance. DPBA can also serve as a chemical probe in the studies on EGFR trafficking and degradation.
Collapse
Affiliation(s)
- Nan Yao
- College of Pharmacy, Jinan University, Guangzhou, China.,Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, China
| | - Chen-Ran Wang
- College of Pharmacy, Jinan University, Guangzhou, China.,Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, China
| | - Ming-Qun Liu
- College of Pharmacy, Jinan University, Guangzhou, China
| | - Ying-Jie Li
- College of Pharmacy, Jinan University, Guangzhou, China
| | - Wei-Min Chen
- College of Pharmacy, Jinan University, Guangzhou, China
| | - Zheng-Qiu Li
- College of Pharmacy, Jinan University, Guangzhou, China
| | - Qi Qi
- School of Medicine, Jinan University, Guangzhou, China
| | - Jin-Jian Lu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Chun-Lin Fan
- College of Pharmacy, Jinan University, Guangzhou, China.,Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, China
| | - Min-Feng Chen
- College of Pharmacy, Jinan University, Guangzhou, China.,Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, China
| | - Ming Qi
- College of Pharmacy, Jinan University, Guangzhou, China.,Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, China
| | - Xiao-Bo Li
- College of Pharmacy, Jinan University, Guangzhou, China.,Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, China
| | - Jian Hong
- School of Medicine, Jinan University, Guangzhou, China
| | - Dong-Mei Zhang
- College of Pharmacy, Jinan University, Guangzhou, China. .,Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, China.
| | - Wen-Cai Ye
- College of Pharmacy, Jinan University, Guangzhou, China. .,Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, China.
| |
Collapse
|
74
|
Skryabin GO, Komelkov AV, Galetsky SA, Bagrov DV, Evtushenko EG, Nikishin II, Zhordaniia KI, Savelyeva EE, Akselrod ME, Paianidi IG, Tchevkina EM. Stomatin is highly expressed in exosomes of different origin and is a promising candidate as an exosomal marker. J Cell Biochem 2020; 122:100-115. [PMID: 32951259 DOI: 10.1002/jcb.29834] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 04/13/2020] [Accepted: 06/30/2020] [Indexed: 01/05/2023]
Abstract
Proteins involved in the organizing of lipid rafts can be found in exosomes, as shown for caveolin-1, and they could contribute to exosomal cargo sorting, as shown for flotillins. Stomatin belongs to the same stomatin/prohibitin/flotillin/HflK/C family of lipid rafts proteins, but it has never been studied in exosomes except for extracellular vesicles (EVs) originating from blood cells. Here we first show the presence of stomatin in exosomes produced by epithelial cancer cells (non-small cell lung cancer, breast, and ovarian cancer cells) as well as in EVs from biological fluids, including blood plasma, ascitic fluids, and uterine flushings. A high abundance of stomatin in EVs of various origins and its enrichment in exosomes make stomatin a promising exosomal marker. Comparison with other lipid raft proteins and exosomal markers showed that the level of stomatin protein in exosomes from different sources corresponds well to that of CD9, while it differs essentially from flotillin-1 and flotillin-2 homologs, which in turn are present in exosomes in nearly equal proportions. In contrast, the level of vesicular caveolin-1 as well as its EV-to-cellular ratio vary drastically depending on cell type.
Collapse
Affiliation(s)
- Gleb O Skryabin
- Oncogene Regulation Department of Institute of Carcinogenesis, N. N. Blokhin National Medical Research Center of Oncology, Moscow, Russia
| | - Andrei V Komelkov
- Oncogene Regulation Department of Institute of Carcinogenesis, N. N. Blokhin National Medical Research Center of Oncology, Moscow, Russia
| | - Sergey A Galetsky
- Oncogene Regulation Department of Institute of Carcinogenesis, N. N. Blokhin National Medical Research Center of Oncology, Moscow, Russia
| | - Dmitry V Bagrov
- Department of Bioengineering, Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Evgeniy G Evtushenko
- Department of Chemical Enzymology, Faculty of Chemistry, Lomonosov Moscow State University, Moscow, Russia
| | - Igor I Nikishin
- Department of Bioengineering, Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Kirill I Zhordaniia
- Oncogene Regulation Department of Institute of Carcinogenesis, N. N. Blokhin National Medical Research Center of Oncology, Moscow, Russia
| | - Elizaveta E Savelyeva
- Oncogene Regulation Department of Institute of Carcinogenesis, N. N. Blokhin National Medical Research Center of Oncology, Moscow, Russia
| | - Maria E Akselrod
- Oncogene Regulation Department of Institute of Carcinogenesis, N. N. Blokhin National Medical Research Center of Oncology, Moscow, Russia
| | - Iulia G Paianidi
- Oncogene Regulation Department of Institute of Carcinogenesis, N. N. Blokhin National Medical Research Center of Oncology, Moscow, Russia
| | - Elena M Tchevkina
- Oncogene Regulation Department of Institute of Carcinogenesis, N. N. Blokhin National Medical Research Center of Oncology, Moscow, Russia
| |
Collapse
|
75
|
Regulation of invadosomes by microtubules: Not only a matter of railways. Eur J Cell Biol 2020; 99:151109. [DOI: 10.1016/j.ejcb.2020.151109] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 07/02/2020] [Accepted: 07/13/2020] [Indexed: 12/19/2022] Open
|
76
|
Greig J, Bulgakova NA. Arf6 determines tissue architecture by stabilizing intercellular adhesion. Philos Trans R Soc Lond B Biol Sci 2020; 375:20190682. [PMID: 32829688 DOI: 10.1098/rstb.2019.0682] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Correct cell shape is indispensable for tissue architecture, with cell shape being determined by cortical actin and surface adhesion. The role of adhesion in remodelling tissue is to counteract the deformation of cells by force, resulting from actomyosin contractility, and to maintain tissue integrity. The dynamics of this adhesion are critical to the processes of cell shape formation and maintenance. Here, we show that the trafficking molecule Arf6 has a direct impact on cell elongation, by acting to stabilize E-cadherin-based adhesion complexes at the cell surface, in addition to its canonical role in endocytosis. We demonstrate that these functions of Arf6 are dependent on the molecule Flotillin1, which recruits Arf6 to the plasma membrane. Our data suggest that Arf6 and Flotillin1 operate in a pathway distinct from clathrin-mediated endocytosis. Altogether, we demonstrate that Arf6- and Flotillin1-dependent regulation of the dynamics of cell adhesion contribute to moulding tissue in vivo. This article is part of the discussion meeting issue 'Contemporary morphogenesis'.
Collapse
Affiliation(s)
- Joshua Greig
- Department of Biomedical Science and Bateson Centre, University of Sheffield, Sheffield S10 2TN, UK
| | - Natalia A Bulgakova
- Department of Biomedical Science and Bateson Centre, University of Sheffield, Sheffield S10 2TN, UK
| |
Collapse
|
77
|
Corbeil D, Santos MF, Karbanová J, Kurth T, Rappa G, Lorico A. Uptake and Fate of Extracellular Membrane Vesicles: Nucleoplasmic Reticulum-Associated Late Endosomes as a New Gate to Intercellular Communication. Cells 2020; 9:cells9091931. [PMID: 32825578 PMCID: PMC7563309 DOI: 10.3390/cells9091931] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/18/2020] [Accepted: 08/20/2020] [Indexed: 02/06/2023] Open
Abstract
Extracellular membrane vesicles (EVs) are emerging as new vehicles in intercellular communication, but how the biological information contained in EVs is shared between cells remains elusive. Several mechanisms have been described to explain their release from donor cells and the initial step of their uptake by recipient cells, which triggers a cellular response. Yet, the intracellular routes and subcellular fate of EV content upon internalization remain poorly characterized. This is particularly true for EV-associated proteins and nucleic acids that shuttle to the nucleus of host cells. In this review, we will describe and discuss the release of EVs from donor cells, their uptake by recipient cells, and the fate of their cargoes, focusing on a novel intracellular route wherein small GTPase Rab7+ late endosomes containing endocytosed EVs enter into nuclear envelope invaginations and deliver their cargo components to the nucleoplasm of recipient cells. A tripartite protein complex composed of (VAMP)-associated protein A (VAP-A), oxysterol-binding protein (OSBP)-related protein-3 (ORP3), and Rab7 is essential for the transfer of EV-derived components to the nuclear compartment by orchestrating the particular localization of late endosomes in the nucleoplasmic reticulum.
Collapse
Affiliation(s)
- Denis Corbeil
- Biotechnology Center (BIOTEC) and Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, Tatzberg 47-49, 01307 Dresden, Germany; (J.K.)
- Correspondence: (D.C.); (A.L.); Tel.: +49-(0)351-463-40118 (D.C.); +1-(702)-777-3942 (A.L.); Fax: +49-(0)351-463-40244 (D.C.); +1-(702)-777-1758 (A.L.)
| | - Mark F. Santos
- College of Osteopathic Medicine, Touro University Nevada, 874 American Pacific Drive, Henderson, NV 89014, USA; (M.F.S.); (G.R.)
| | - Jana Karbanová
- Biotechnology Center (BIOTEC) and Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, Tatzberg 47-49, 01307 Dresden, Germany; (J.K.)
| | - Thomas Kurth
- Center for Regenerative Therapies Dresden and CMCB, Technische Universität Dresden, Fetscherstraße 105, 01307 Dresden, Germany; (T.K.)
| | - Germana Rappa
- College of Osteopathic Medicine, Touro University Nevada, 874 American Pacific Drive, Henderson, NV 89014, USA; (M.F.S.); (G.R.)
| | - Aurelio Lorico
- College of Osteopathic Medicine, Touro University Nevada, 874 American Pacific Drive, Henderson, NV 89014, USA; (M.F.S.); (G.R.)
- Mediterranean Institute of Oncology, Via Penninazzo, 11, 95029 Viagrande, Italy
- Correspondence: (D.C.); (A.L.); Tel.: +49-(0)351-463-40118 (D.C.); +1-(702)-777-3942 (A.L.); Fax: +49-(0)351-463-40244 (D.C.); +1-(702)-777-1758 (A.L.)
| |
Collapse
|
78
|
Flemming JP, Hill BL, Haque MW, Raad J, Bonder CS, Harshyne LA, Rodeck U, Luginbuhl A, Wahl JK, Tsai KY, Wermuth PJ, Overmiller AM, Mahoney MG. miRNA- and cytokine-associated extracellular vesicles mediate squamous cell carcinomas. J Extracell Vesicles 2020; 9:1790159. [PMID: 32944178 PMCID: PMC7480578 DOI: 10.1080/20013078.2020.1790159] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Exosomes, or small extracellular vesicles (sEVs), serve as intercellular messengers with key roles in normal and pathological processes. Our previous work had demonstrated that Dsg2 expression in squamous cell carcinoma (SCC) cells enhanced both sEV secretion and loading of pro-mitogenic cargo. In this study, using wild-type Dsg2 and a mutant form that is unable to be palmitoylated (Dsg2cacs), we investigated the mechanism by which Dsg2 modulates SCC tumour development and progression through sEVs. We demonstrate that palmitoylation was required for Dsg2 to regulate sub-cellular localisation of lipid raft and endosomal proteins necessary for sEV biogenesis. Pharmacological inhibition of the endosomal pathway abrogated Dsg2-mediated sEV release. In murine xenograft models, Dsg2-expressing cells generated larger xenograft tumours as compared to cells expressing GFP or Dsg2cacs. Co-treatment with sEVs derived from Dsg2-over-expressing cells increased xenograft size. Cytokine profiling revealed, Dsg2 enhanced both soluble and sEV-associated IL-8 and miRNA profiling revealed, Dsg2 down-regulated both cellular and sEV-loaded miR-146a. miR-146a targets IRAK1, a serine-threonine kinase involved in IL-8 signalling. Treatment with a miR-146a inhibitor up-regulated both IRAK1 and IL-8 expression. RNAseq analysis of HNSCC tumours revealed a correlation between Dsg2 and IL-8. Finally, elevated IL-8 plasma levels were detected in a subset of HNSCC patients who did not respond to immune checkpoint therapy, suggesting that these patients may benefit from prior anti-IL-8 treatment. In summary, these results suggest that intercellular communication through cell-cell adhesion, cytokine release and secretion of EVs are coordinated, and critical for tumour growth and development, and may serve as potential prognostic markers to inform treatment options. Abbreviations Basal cell carcinomas, BCC; Betacellulin, BTC; 2-bromopalmitate, 2-Bromo; Cluster of differentiation, CD; Cytochrome c oxidase IV, COX IV; Desmoglein 2, Dsg2; Early endosome antigen 1, EEA1; Epidermal growth factor receptor substrate 15, EPS15; Extracellular vesicle, EV; Flotillin 1, Flot1; Glyceraldehyde-3-phosphate dehydrogenase, GAPH; Green fluorescent protein, GFP; Head and neck squamous cell carcinoma, HNSCC; Interleukin-1 receptor-associated kinase 1, IRAK1; Interleukin 8, IL-8; Large EV, lEV; MicroRNA, miR; Palmitoylacyltransferase, PAT; Ras-related protein 7 Rab7; Small EV, sEV; Squamous cell carcinoma, SCC; Tissue inhibitor of metalloproteinases, TIMP; Tumour microenvironment, TME
Collapse
Affiliation(s)
- Joseph P Flemming
- Department of Dermatology and Cutaneous Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Brianna L Hill
- Department of Dermatology and Cutaneous Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Mohammed W Haque
- Department of Dermatology and Cutaneous Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Jessica Raad
- Department of Dermatology and Cutaneous Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Claudine S Bonder
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA, Australia
| | - Larry A Harshyne
- Department of Medical Oncology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Ulrich Rodeck
- Department of Dermatology and Cutaneous Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Adam Luginbuhl
- Department of Otolaryngology-Head and Neck Surgery, Thomas Jefferson University, Philadelphia, PA, USA
| | - James K Wahl
- Department of Oral Biology, University of Nebraska Medical Center, Lincoln, NE, USA
| | - Kenneth Y Tsai
- Department of Tumor Biology, Moffitt Cancer Center, Tampa, FL, USA
| | - Peter J Wermuth
- Department of Dermatology and Cutaneous Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Andrew M Overmiller
- Department of Dermatology and Cutaneous Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Mỹ G Mahoney
- Department of Dermatology and Cutaneous Biology, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
79
|
Bosmani C, Leuba F, Hanna N, Bach F, Burdet F, Pagni M, Hagedorn M, Soldati T. Vacuolins and myosin VII are required for phagocytic uptake and phagosomal membrane recycling in Dictyostelium discoideum. J Cell Sci 2020; 133:jcs242974. [PMID: 32482795 DOI: 10.1242/jcs.242974] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 05/18/2020] [Indexed: 12/17/2022] Open
Abstract
Flotillins are lipid raft residents involved in membrane trafficking and recycling of plasma membrane proteins. Dictyostelium discoideum uses phagocytosis to kill, digest and feed on bacteria. It possesses three flotillin-like vacuolins that are strongly associated with membranes and that gradually accumulate on maturing phagosomes. Absence of vacuolins reduced adhesion and particle recognition resulting in a drastic reduction in the uptake of various types of particles. This was caused by a block in the recycling of plasma membrane components and the absence of their specific cortex-associated proteins. In addition, absence of vacuolins also impaired phagolysosome biogenesis, without significantly impacting killing and digestion of a range of bacteria. Strikingly, both absence and overexpression of vacuolins induced a strong downregulation of myosin VII (also known as MyoI) expression, as well as its binding partner talin A. Episomal expression of myosin VII fully rescued defects in uptake and adhesion but not in phagosome maturation. These results suggest a dual role for vacuolins: a novel mechanism involving membrane microdomains and myosin VII-talin A in clustering phagosomal receptors and adhesion molecules at the plasma membrane, and a role in phagolysosomal biogenesis.
Collapse
Affiliation(s)
- Cristina Bosmani
- Départment de Biochimie, Faculté des Sciences, Université de Genève, CH-1205 Geneva, Switzerland
| | - Florence Leuba
- Départment de Biochimie, Faculté des Sciences, Université de Genève, CH-1205 Geneva, Switzerland
| | - Nabil Hanna
- Départment de Biochimie, Faculté des Sciences, Université de Genève, CH-1205 Geneva, Switzerland
| | - Frauke Bach
- Section Parasitology, Bernhard Nocht Institute for Tropical Medicine, D-20359 Hamburg, Germany
| | - Frédéric Burdet
- Vital-IT, Swiss Institute of Bioinformatics, University of Lausanne, CH-1015 Lausanne, Switzerland
| | - Marco Pagni
- Vital-IT, Swiss Institute of Bioinformatics, University of Lausanne, CH-1015 Lausanne, Switzerland
| | - Monica Hagedorn
- Section Parasitology, Bernhard Nocht Institute for Tropical Medicine, D-20359 Hamburg, Germany
| | - Thierry Soldati
- Départment de Biochimie, Faculté des Sciences, Université de Genève, CH-1205 Geneva, Switzerland
| |
Collapse
|
80
|
Samson GPB, Legler DF. Membrane Compartmentalization and Scaffold Proteins in Leukocyte Migration. Front Cell Dev Biol 2020; 8:285. [PMID: 32411706 PMCID: PMC7198906 DOI: 10.3389/fcell.2020.00285] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 04/02/2020] [Indexed: 01/14/2023] Open
Abstract
Leukocyte migration across vessels into and within peripheral and lymphoid tissues is essential for host defense against invading pathogens. Leukocytes are specialized in sensing a variety of guidance cues and to integrate environmental stimuli to navigate in a timely and spatially controlled manner. These extracellular signals must be transmitted across the leukocyte’s plasma membrane in a way that intracellular signaling cascades enable directional cell movement. Therefore, the composition of the membrane in concert with proteins that influence the compartmentalization of the plasma membrane or contribute to delineate intracellular signaling molecules are key in controlling leukocyte navigation. This becomes evident by the fact that mislocalization of membrane proteins is known to deleteriously affect cellular functions that may cause diseases. In this review we summarize recent advances made in the understanding of how membrane cholesterol levels modulate chemokine receptor signaling and hence leukocyte trafficking. Moreover, we provide an overview on the role of membrane scaffold proteins, particularly tetraspanins, flotillins/reggies, and caveolins in controlling leukocyte migration both in vitro and in vivo.
Collapse
Affiliation(s)
- Guerric P B Samson
- Biotechnology Institute Thurgau at the University of Konstanz, Kreuzlingen, Switzerland
| | - Daniel F Legler
- Biotechnology Institute Thurgau at the University of Konstanz, Kreuzlingen, Switzerland.,Faculty of Biology, University of Konstanz, Konstanz, Germany.,Theodor Kocher Institute, University of Bern, Bern, Switzerland
| |
Collapse
|
81
|
Membrane dynamics in cell migration. Essays Biochem 2020; 63:469-482. [PMID: 31350382 DOI: 10.1042/ebc20190014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 06/27/2019] [Accepted: 07/11/2019] [Indexed: 12/20/2022]
Abstract
Migration of cells is required in multiple tissue-level processes, such as in inflammation or cancer metastasis. Endocytosis is an extremely regulated cellular process by which cells uptake extracellular molecules or internalise cell surface receptors. While the role of endocytosis of focal adhesions (FA) and plasma membrane (PM) turnover at the leading edge of migratory cells is wide known, the contribution of endocytic proteins per se in migration has been frequently disregarded. In this review, we describe the novel functions of the most well-known endocytic proteins in cancer cell migration, focusing on clathrin, caveolin, flotillins and GRAF1. In addition, we highlight the relevance of the macropinocytic pathway in amoeboid-like cell migration.
Collapse
|
82
|
Skryabin GO, Komelkov AV, Savelyeva EE, Tchevkina EM. Lipid Rafts in Exosome Biogenesis. BIOCHEMISTRY (MOSCOW) 2020; 85:177-191. [PMID: 32093594 DOI: 10.1134/s0006297920020054] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Exosomes (secreted extracellular vesicles formed in the intracellular vesicular transport system) play a crucial role in distant cell-cell communication. Exosomes transfer active forms of various biomolecules; the molecular composition of the exosomal cargo is a result of targeted selection and depends on the type of producer cells. The mechanisms underlying exosome formation and cargo selection are poorly understood. It is believed that there are several pathways for exosome biogenesis, although the questions about their independence and simultaneous coexistence in the cell still remain open. The least studied topic is the recently discovered mechanism of exosome formation associated with lipid rafts, or membrane lipid microdomains. Here, we present modern concepts and basic hypotheses on the mechanisms of exosome biogenesis and secretion and summarize current data on the involvement of lipid rafts and their constituent molecules in these processes. Special attention is paid to the analysis of possible role in the exosome formation of raft-forming proteins of the SPFH family, components of planar rafts, and caveolin, the main component of caveolae.
Collapse
Affiliation(s)
- G O Skryabin
- Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, Moscow, 115478, Russia
| | - A V Komelkov
- Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, Moscow, 115478, Russia.
| | - E E Savelyeva
- Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, Moscow, 115478, Russia
| | - E M Tchevkina
- Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, Moscow, 115478, Russia
| |
Collapse
|
83
|
Flotillins: At the Intersection of Protein S-Palmitoylation and Lipid-Mediated Signaling. Int J Mol Sci 2020; 21:ijms21072283. [PMID: 32225034 PMCID: PMC7177705 DOI: 10.3390/ijms21072283] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 03/19/2020] [Accepted: 03/20/2020] [Indexed: 02/07/2023] Open
Abstract
Flotillin-1 and flotillin-2 are ubiquitously expressed, membrane-associated proteins involved in multifarious cellular events from cell signaling, endocytosis, and protein trafficking to gene expression. They also contribute to oncogenic signaling. Flotillins bind the cytosolic leaflet of the plasma membrane and endomembranes and, upon hetero-oligomerization, serve as scaffolds facilitating the assembly of multiprotein complexes at the membrane-cytosol interface. Additional functions unique to flotillin-1 have been discovered recently. The membrane-binding of flotillins is regulated by S-palmitoylation and N-myristoylation, hydrophobic interactions involving specific regions of the polypeptide chain and, to some extent, also by their oligomerization. All these factors endow flotillins with an ability to associate with the sphingolipid/cholesterol-rich plasma membrane domains called rafts. In this review, we focus on the critical input of lipids to the regulation of the flotillin association with rafts and thereby to their functioning. In particular, we discuss how the recent developments in the field of protein S-palmitoylation have contributed to the understanding of flotillin1/2-mediated processes, including endocytosis, and of those dependent exclusively on flotillin-1. We also emphasize that flotillins affect directly or indirectly the cellular levels of lipids involved in diverse signaling cascades, including sphingosine-1-phosphate and PI(4,5)P2. The mutual relations between flotillins and distinct lipids are key to the regulation of their involvement in numerous cellular processes.
Collapse
|
84
|
Dam DHM, Jelsma SA, Yu JM, Liu H, Kong B, Paller AS. Flotillin and AP2A1/2 Promote IGF-1 Receptor Association with Clathrin and Internalization in Primary Human Keratinocytes. J Invest Dermatol 2020; 140:1743-1752.e4. [PMID: 32027876 DOI: 10.1016/j.jid.2020.01.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Revised: 01/13/2020] [Accepted: 01/20/2020] [Indexed: 12/21/2022]
Abstract
IGF-1 receptor (IGF1R) signaling promotes keratinocyte proliferation, migration, and survival. However, the mechanism of IGF1R endocytosis in normal keratinocytes remains unclear. Confocal, super resolution structured illumination microscopy, total internal reflection fluorescence microscopy, and coimmunoprecipitation studies reveal that IGF1R associates with flotillin-1 (Flot-1), which currently has no known role in normal receptor tyrosine kinase endocytosis, under basal conditions in monolayer keratinocyte cultures. Ligand stimulation of IGF1R promotes its clathrin-dependent endocytosis, mediated by two distinct adaptors, Flot-1 in noncaveolar lipid rafts and the AP2A1/2 complex in clathrin vesicles. Concurrent, but not individual, short hairpin RNA knockdown of FLOT1/2 and AP2A1/2 reduced IGF1R association with clathrin, internalization, and pathway activation by more than 50% (of phosphorylated IGF1R, phosphorylated protein kinase B, and phosphorylated MAPK kinase), suggesting the complementarity of these two adaptor-specific pathways. The Flot-1 pathway is more responsive to low IGF-1 concentrations, whereas the AP2A1/2 pathway predominates at higher IGF-1 concentrations. Selective association of IGF1R-Flot-1-clathrin with Rab4, but IGF1R-AP2A1/2-clathrin with Rab11, implicates Flot-1 as the adaptor for faster recycling and AP2A1/2 as the adaptor for slower IGF1R recycling. These dual pathways, particularly flotillin-dependent, clathrin-mediated endocytosis, provide a new avenue for drug targeting in disorders with aberrant regulation of IGF1R signaling.
Collapse
Affiliation(s)
- Duncan Hieu M Dam
- Department of Dermatology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Sophia A Jelsma
- Department of Dermatology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Jeong Min Yu
- Department of Dermatology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Haoming Liu
- Department of Dermatology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Betty Kong
- Department of Dermatology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Amy S Paller
- Department of Dermatology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA.
| |
Collapse
|
85
|
Cacciottolo M, Morgan TE, Saffari AA, Shirmohammadi F, Forman HJ, Sioutas C, Finch CE. Traffic-related air pollutants (TRAP-PM) promote neuronal amyloidogenesis through oxidative damage to lipid rafts. Free Radic Biol Med 2020; 147:242-251. [PMID: 31883973 PMCID: PMC7075030 DOI: 10.1016/j.freeradbiomed.2019.12.023] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 12/10/2019] [Accepted: 12/20/2019] [Indexed: 12/21/2022]
Abstract
Traffic-related air pollution particulate matter (TRAP-PM) is associated with increased risk of Alzheimer Disease (AD). Rodent models respond to nano-sized TRAP-PM (nPM) with increased production of amyloid Aβ peptides, concurrently with oxidative damage. Because pro-Aβ processing of the amyloid precursor protein (APP) occurs on subcellular lipid rafts, we hypothesized that oxidative stress from nPM exposure would alter lipid rafts to favor Aβ production. This hypothesis was tested with J20 mice and N2a cells transgenic for hAPPswe (familial AD). Exposure of J20-APPswe mice to nPM for 150 h caused increased lipid oxidation (4-HNE) and increased the pro-amyloidogenic processing of APP in lipid raft fractions in cerebral cortex; the absence of these changes in cerebellum parallels the AD brain region selectivity for Aβ deposits. In vitro, nPM induced similar oxidative responses in N2a-APPswe cells, with dose-dependent production of NO, oxidative damage (4-HNE, 3NT), and lipid raft alterations of APP with increased Aβ peptides. The antioxidant N-acetyl-cysteine (NAC) attenuated nPM-induced oxidative damage and lipid raft alterations of APP processing. These findings identify neuronal lipid rafts as novel targets of oxidative damage in the pro-amyloidogenic effects of air pollution.
Collapse
Affiliation(s)
- Mafalda Cacciottolo
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Todd E Morgan
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Arian A Saffari
- Viterbi School of Engineering, University of Southern California, Los Angeles, CA, USA
| | - Farimah Shirmohammadi
- Viterbi School of Engineering, University of Southern California, Los Angeles, CA, USA
| | - Henry Jay Forman
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Costantinos Sioutas
- Viterbi School of Engineering, University of Southern California, Los Angeles, CA, USA
| | - Caleb E Finch
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA; Dornsife College, Dept. Biological Sciences, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
86
|
Vocelle D, Chan C, Walton SP. Endocytosis Controls siRNA Efficiency: Implications for siRNA Delivery Vehicle Design and Cell-Specific Targeting. Nucleic Acid Ther 2020; 30:22-32. [PMID: 31718426 PMCID: PMC6987736 DOI: 10.1089/nat.2019.0804] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 10/10/2019] [Indexed: 12/19/2022] Open
Abstract
While small interfering RNAs (siRNAs) are commonly used for laboratory studies, development of siRNA therapeutics has been slower than expected, due, in part, to a still limited understanding of the endocytosis and intracellular trafficking of siRNA-containing complexes. With the recent characterization of multiple clathrin-/caveolin-independent endocytic pathways, that is, those mediated by Graf1, Arf6, and flotillin, it has become clear that the endocytic mechanism influences subsequent intracellular processing of the internalized cargo. To explore siRNA delivery in light of these findings, we developed a novel assay that differentiates uptake by each of the endocytic pathways and can be used to determine whether endocytosis by a pathway leads to the initiation of RNA interference (RNAi). Using Lipofectamine 2000 (LF2K), we determined the endocytosis pathway leading to active silencing (whether by clathrin, caveolin, Arf6, Graf1, flotillin, or macropinocytosis) across multiple cell types (HeLa, H1299, HEK293, and HepG2). We showed that LF2K is internalized by Graf1-, Arf6-, or flotillin-mediated endocytosis for the initiation of RNAi, depending on cell type. In addition, we found that a portion of siRNA-containing complexes is internalized by pathways that do not lead to initiation of silencing. Inhibition of these pathways enhanced intracellular levels of siRNAs with concomitant enhancement of silencing.
Collapse
Affiliation(s)
- Daniel Vocelle
- Department of Chemical Engineering and Materials Science, Michigan State University, East Lansing, Michigan
| | - Christina Chan
- Department of Chemical Engineering and Materials Science, Michigan State University, East Lansing, Michigan
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan
| | - S. Patrick Walton
- Department of Chemical Engineering and Materials Science, Michigan State University, East Lansing, Michigan
| |
Collapse
|
87
|
Yokoyama H, Matsui I. The lipid raft markers stomatin, prohibitin, flotillin, and HflK/C (SPFH)-domain proteins form an operon with NfeD proteins and function with apolar polyisoprenoid lipids. Crit Rev Microbiol 2020; 46:38-48. [PMID: 31983249 DOI: 10.1080/1040841x.2020.1716682] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
SPFH-domain proteins are found in almost all organisms across three domains: archaea, bacteria, and eukaryotes. In eukaryotic organelles, their subfamilies exhibit overlapping distribution and functions; thus, the rationality of annotation to discriminate these subfamilies remains unclear. In this review, the binding ability of prokaryotic SPFH-domain proteins towards nonpolar polyisoprenoides such as squalene and lycopene, rather than cholesterol, is discussed. The hydrophobic region at the C-terminus of SPFH-domain proteins constitutes the main region that binds apolar polyisoprenoid lipids as well as cholesterol and substantively contributes towards lipid raft formation as these regions are self-assembled together with specific lipids. Because the scaffolding proteins caveolins show common topological properties with SPFH-domain proteins such as stomatin and flotillin, the α-helical segments of stomatin proteins can flexibly move along with the membrane surface, with such movement potentially leading to membrane bending via lipid raft clustering through the formation of high order homo-oligomeric complexes of SPFH-domain proteins. We also discuss the functional significance and ancient origin of SPFH-domain proteins and the NfeD protein (STOPP) operon, which can be traced back to the ancient living cells that diverged and evolved to archaea and bacteria. Based on the molecular mechanism whereby the STOPP-protease degrades the C-terminal hydrophobic clusters of SPFH-domain proteins, it is conceivable that STOPP-protease might control the physicochemical properties of lipid rafts.
Collapse
Affiliation(s)
- Hideshi Yokoyama
- Department of Medical and Life Sciences, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba, Japan
| | - Ikuo Matsui
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Japan
| |
Collapse
|
88
|
Qiao Z, Zogli P, Libault M. Plant Hormones Differentially Control the Sub-Cellular Localization of Plasma Membrane Microdomains during the Early Stage of Soybean Nodulation. Genes (Basel) 2019; 10:E1012. [PMID: 31817452 PMCID: PMC6947267 DOI: 10.3390/genes10121012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 11/30/2019] [Accepted: 12/02/2019] [Indexed: 01/31/2023] Open
Abstract
Phytohormones regulate the mutualistic symbiotic interaction between legumes and rhizobia, nitrogen-fixing soil bacteria, notably by controlling the formation of the infection thread in the root hair (RH). At the cellular level, the formation of the infection thread is promoted by the translocation of plasma membrane microdomains at the tip of the RH. We hypothesize that phytohormones regulate the translocation of plasma membrane microdomains to regulate infection thread formation. Accordingly, we treated with hormone and hormone inhibitors transgenic soybean roots expressing fusions between the Green Fluorescent Protein (GFP) and GmFWL1 or GmFLOT2/4, two microdomain-associated proteins translocated at the tip of the soybean RH in response to rhizobia. Auxin and cytokinin treatments are sufficient to trigger or inhibit the translocation of GmFWL1 and GmFLOT2/4 to the RH tip independently of the presence of rhizobia, respectively. Unexpectedly, the application of salicylic acid, a phytohormone regulating the plant defense system, also promotes the translocation of GmFWL1 and GmFLOT2/4 to the RH tip regardless of the presence of rhizobia. These results suggest that phytohormones are playing a central role in controlling the early stages of rhizobia infection by regulating the translocation of plasma membrane microdomains. They also support the concept of crosstalk of phytohormones to control nodulation.
Collapse
Affiliation(s)
- Zhenzhen Qiao
- Department of Microbiology and Plant Biology, University of Oklahoma, Norman, OK 73019, USA;
- Biosciences Division, Oak Ridge National Laboratory, Oak Ridge, TN 37830, USA
| | - Prince Zogli
- Department of Agronomy and Horticulture, University of Nebraska-Lincoln, Beadle Center, Lincoln, NE 68503, USA;
| | - Marc Libault
- Department of Agronomy and Horticulture, University of Nebraska-Lincoln, Beadle Center, Lincoln, NE 68503, USA;
| |
Collapse
|
89
|
Fekri F, Abousawan J, Bautista S, Orofiamma L, Dayam RM, Antonescu CN, Karshafian R. Targeted enhancement of flotillin-dependent endocytosis augments cellular uptake and impact of cytotoxic drugs. Sci Rep 2019; 9:17768. [PMID: 31780775 PMCID: PMC6882852 DOI: 10.1038/s41598-019-54062-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 11/07/2019] [Indexed: 12/23/2022] Open
Abstract
Cellular uptake is limiting for the efficacy of many cytotoxic drugs used to treat cancer. Identifying endocytic mechanisms that can be modulated with targeted, clinically-relevant interventions is important to enhance the efficacy of various cancer drugs. We identify that flotillin-dependent endocytosis can be targeted and upregulated by ultrasound and microbubble (USMB) treatments to enhance uptake and efficacy of cancer drugs such as cisplatin. USMB involves targeted ultrasound following administration of encapsulated microbubbles, used clinically for enhanced ultrasound image contrast. USMB treatments robustly enhanced internalization of the molecular scaffold protein flotillin, as well as flotillin-dependent fluid-phase internalization, a phenomenon dependent on the protein palmitoyltransferase DHHC5 and the Src-family kinase Fyn. USMB treatment enhanced DNA damage and cell killing elicited by the cytotoxic agent cisplatin in a flotillin-dependent manner. Thus, flotillin-dependent endocytosis can be modulated by clinically-relevant USMB treatments to enhance drug uptake and efficacy, revealing an important new strategy for targeted drug delivery for cancer treatment.
Collapse
Affiliation(s)
- Farnaz Fekri
- Department of Chemistry and Biology, Ryerson University, Toronto, Canada
- Graduate Program in Molecular Science, Ryerson University, Toronto, Ontario, M5B 2K3, Canada
| | - John Abousawan
- Department of Chemistry and Biology, Ryerson University, Toronto, Canada
- Graduate Program in Molecular Science, Ryerson University, Toronto, Ontario, M5B 2K3, Canada
| | - Stephen Bautista
- Department of Chemistry and Biology, Ryerson University, Toronto, Canada
- Graduate Program in Molecular Science, Ryerson University, Toronto, Ontario, M5B 2K3, Canada
| | - Laura Orofiamma
- Department of Chemistry and Biology, Ryerson University, Toronto, Canada
- Graduate Program in Molecular Science, Ryerson University, Toronto, Ontario, M5B 2K3, Canada
| | - Roya M Dayam
- Department of Chemistry and Biology, Ryerson University, Toronto, Canada
- Graduate Program in Molecular Science, Ryerson University, Toronto, Ontario, M5B 2K3, Canada
| | - Costin N Antonescu
- Department of Chemistry and Biology, Ryerson University, Toronto, Canada.
- Graduate Program in Molecular Science, Ryerson University, Toronto, Ontario, M5B 2K3, Canada.
- Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, Ontario, Canada.
| | - Raffi Karshafian
- Graduate Program in Molecular Science, Ryerson University, Toronto, Ontario, M5B 2K3, Canada.
- Department of Physics, Ryerson University, Toronto, Ontario, M5B 2K3, Canada.
- Institute for Biomedical Engineering, Science and Technology (iBEST), a partnership between Ryerson University and St. Michael's Hospital, Toronto, Ontario, Canada.
- Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, Ontario, Canada.
| |
Collapse
|
90
|
Thalwieser Z, Király N, Fonódi M, Csortos C, Boratkó A. Protein phosphatase 2A-mediated flotillin-1 dephosphorylation up-regulates endothelial cell migration and angiogenesis regulation. J Biol Chem 2019; 294:20196-20206. [PMID: 31753918 DOI: 10.1074/jbc.ra119.007980] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 11/04/2019] [Indexed: 12/31/2022] Open
Abstract
Endothelial cells have key functions in endothelial barrier integrity and in responses to angiogenic signals that promote cell proliferation, cell migration, cytoskeletal reorganization, and formation of new blood vessels. These functions highly depend on protein-protein interactions in cell-cell junction and cell attachment complexes and on interactions with cytoskeletal proteins. Protein phosphatase 2A (PP2A) dephosphorylates several target proteins involved in cytoskeletal dynamics and cell adhesion. Our goal was to find new interacting and substrate proteins of the PP2A-B55α holoenzyme in bovine pulmonary endothelial cells. Using LC-MS/MS analysis, we identified flotillin-1 as a protein that binds recombinant GSH S-transferase-tagged PP2A-B55α. Immunoprecipitation experiments, proximity ligation assays, and immunofluorescent staining confirmed the interaction between these two endogenous proteins in endothelial cells. Originally, flotillins were described as regulatory proteins for axon regeneration, but they appear to function in many cellular processes, such as membrane receptor signaling, endocytosis, and cell adhesion. Ser315 is a known PKC-targeted site in flotillin-1. Utilizing phosphomutants of flotillin-1 and the NanoBiT luciferase assay, we show here that phosphorylation/dephosphorylation of Ser315 in flotillin-1 significantly affects its interaction with PP2A-B55α and that PP2A-B55α dephosphorylates phospho-Ser315 Spreading, attachment, migration, and in vitro tube formation rates of S315A variant-overexpressing cells were faster than those of nontransfected or S315D-transfected cells. These results indicate that the PP2A-flotillin-1 interaction identified here affects major physiological activities of pulmonary endothelial cells.
Collapse
Affiliation(s)
- Zsófia Thalwieser
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Egyetem tér 1, 4032 Debrecen, Hungary
| | - Nikolett Király
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Egyetem tér 1, 4032 Debrecen, Hungary
| | - Márton Fonódi
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Egyetem tér 1, 4032 Debrecen, Hungary
| | - Csilla Csortos
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Egyetem tér 1, 4032 Debrecen, Hungary
| | - Anita Boratkó
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Egyetem tér 1, 4032 Debrecen, Hungary
| |
Collapse
|
91
|
Hudák A, Kusz E, Domonkos I, Jósvay K, Kodamullil AT, Szilák L, Hofmann-Apitius M, Letoha T. Contribution of syndecans to cellular uptake and fibrillation of α-synuclein and tau. Sci Rep 2019; 9:16543. [PMID: 31719623 PMCID: PMC6851098 DOI: 10.1038/s41598-019-53038-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 10/28/2019] [Indexed: 11/09/2022] Open
Abstract
Scientific evidence suggests that α-synuclein and tau have prion-like properties and that prion-like spreading and seeding of misfolded protein aggregates constitutes a central mechanism for neurodegeneration. Heparan sulfate proteoglycans (HSPGs) in the plasma membrane support this process by attaching misfolded protein fibrils. Despite of intense studies, contribution of specific HSPGs to seeding and spreading of α-synuclein and tau has not been explored yet. Here we report that members of the syndecan family of HSPGs mediate cellular uptake of α-synuclein and tau fibrils via a lipid-raft dependent and clathrin-independent endocytic route. Among syndecans, the neuron predominant syndecan-3 exhibits the highest affinity for both α-synuclein and tau. Syndecan-mediated internalization of α-synuclein and tau depends heavily on conformation as uptake via syndecans start to dominate once fibrils are formed. Overexpression of syndecans, on the other hand, reduces cellular uptake of monomeric α-synuclein and tau, yet exerts a fibril forming effect on both proteins. Data obtained from syndecan overexpressing cellular models presents syndecans, especially the neuron predominant syndecan-3, as important mediators of seeding and spreading of α-synuclein and tau and reveal how syndecans contribute to fundamental molecular events of α-synuclein and tau pathology.
Collapse
Affiliation(s)
| | | | - Ildikó Domonkos
- Biological Research Centre of the Hungarian Academy of Sciences, Szeged, H-6726, Hungary
| | - Katalin Jósvay
- Biological Research Centre of the Hungarian Academy of Sciences, Szeged, H-6726, Hungary
| | - Alpha Tom Kodamullil
- Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Sankt Augustin, 53754, Germany
| | - László Szilák
- Szilak Laboratories, Bioinformatics and Molecule-Design, Szeged, H-6723, Hungary
| | - Martin Hofmann-Apitius
- Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Sankt Augustin, 53754, Germany
| | | |
Collapse
|
92
|
Stremmel W, Staffer S, Fricker G, Weiskirchen R. The Bile Acid-Phospholipid Conjugate Ursodeoxycholyl-Lysophosphatidylethanolamide (UDCA-LPE) Disintegrates the Lipid Backbone of Raft Plasma Membrane Domains by the Removal of the Membrane Phospholipase A2. Int J Mol Sci 2019; 20:ijms20225631. [PMID: 31717968 PMCID: PMC6888454 DOI: 10.3390/ijms20225631] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 10/28/2019] [Accepted: 11/06/2019] [Indexed: 01/07/2023] Open
Abstract
The bile acid-phospholipid conjugate ursodeoxycholyl-lysophosphatidylethanolamide (UDCA-LPE) was shown to have anti-inflammatory, antisteatotic, and antifibrotic properties, rendering it as a drug targeting non-alcoholic steatohepatitis (NASH). On a molecular level, it disrupted the heterotetrameric fatty acid uptake complex localized in detergent-resistant membrane domains of the plasma membrane (DRM-PM). However, its mode of action was unclear. Methodologically, UDCA-LPE was incubated with the liver tumor cell line HepG2 as well as their isolated DRM-PM and all other cellular membranes (non-DRM). The membrane cholesterol and phospholipids were quantified as well as the DRM-PM protein composition by Western blotting. The results show a loss of DRM-PM by UDCA-LPE (50 µM) with a 63.13 ± 7.14% reduction of phospholipids and an 81.94 ± 8.30% reduction of cholesterol in relation to mg total protein. The ratio of phospholipids to cholesterol changed from 2:1 to 4:1, resembling those of non-DRM fractions. Among the members of the fatty acid uptake complex, the calcium-independent membrane phospholipase A2 (iPLA2β) abandoned DRM-PM most rapidly. As a consequence, the other members of this transport system disappeared as well as the DRM-PM anchored fibrosis regulating proteins integrin β-1 and lysophospholipid receptor 1 (LPAR-1). It is concluded that UDCA-LPE executes its action by iPLA2β removal from DRM-PM and consequent dissolution of the raft lipid platform.
Collapse
Affiliation(s)
- Wolfgang Stremmel
- Institute of Pharmacy and Molecular Biotechnology, University of Heidelberg, 69120 Heidelberg, Germany;
- Correspondence: ; Tel.: +49-152-34349907
| | - Simone Staffer
- Department of Internal Medicine IV, University Hospital of Heidelberg, 69120 Heidelberg, Germany;
| | - Gert Fricker
- Institute of Pharmacy and Molecular Biotechnology, University of Heidelberg, 69120 Heidelberg, Germany;
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry, RWTH University Hospital Aachen, 52074 Aachen, Germany;
| |
Collapse
|
93
|
Thottacherry JJ, Sathe M, Prabhakara C, Mayor S. Spoiled for Choice: Diverse Endocytic Pathways Function at the Cell Surface. Annu Rev Cell Dev Biol 2019; 35:55-84. [PMID: 31283376 PMCID: PMC6917507 DOI: 10.1146/annurev-cellbio-100617-062710] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Endocytosis has long been identified as a key cellular process involved in bringing in nutrients, in clearing cellular debris in tissue, in the regulation of signaling, and in maintaining cell membrane compositional homeostasis. While clathrin-mediated endocytosis has been most extensively studied, a number of clathrin-independent endocytic pathways are continuing to be delineated. Here we provide a current survey of the different types of endocytic pathways available at the cell surface and discuss a new classification and plausible molecular mechanisms for some of the less characterized pathways. Along with an evolutionary perspective of the origins of some of these pathways, we provide an appreciation of the distinct roles that these pathways play in various aspects of cellular physiology, including the control of signaling and membrane tension.
Collapse
Affiliation(s)
- Joseph Jose Thottacherry
- National Centre for Biological Science, Tata Institute for Fundamental Research, Bangalore 560065, India;
| | - Mugdha Sathe
- National Centre for Biological Science, Tata Institute for Fundamental Research, Bangalore 560065, India;
| | - Chaitra Prabhakara
- National Centre for Biological Science, Tata Institute for Fundamental Research, Bangalore 560065, India;
| | - Satyajit Mayor
- National Centre for Biological Science, Tata Institute for Fundamental Research, Bangalore 560065, India;
- Institute for Stem Cell Science and Regenerative Medicine, Bangalore, 560065, India
| |
Collapse
|
94
|
Flotillins promote T cell receptor sorting through a fast Rab5-Rab11 endocytic recycling axis. Nat Commun 2019; 10:4392. [PMID: 31558725 PMCID: PMC6763463 DOI: 10.1038/s41467-019-12352-w] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 08/28/2019] [Indexed: 12/25/2022] Open
Abstract
The targeted endocytic recycling of the T cell receptor (TCR) to the immunological synapse is essential for T cell activation. Despite this, the mechanisms that underlie the sorting of internalised receptors into recycling endosomes remain poorly understood. To build a comprehensive picture of TCR recycling during T cell activation, we developed a suite of new imaging and quantification tools centred on photoactivation of fluorescent proteins. We show that the membrane-organising proteins, flotillin-1 and -2, are required for TCR to reach Rab5-positive endosomes immediately after endocytosis and for transfer from Rab5- to Rab11a-positive compartments. We further observe that after sorting into in Rab11a-positive vesicles, TCR recycles to the plasma membrane independent of flotillin expression. Our data suggest a mechanism whereby flotillins delineate a fast Rab5-Rab11a endocytic recycling axis and functionally contribute to regulate the spatial organisation of these endosomes. Internalized receptors are recycled back to the cell surface, but their precise mechanisms are unclear. Here, the authors show that the flotillin membrane proteins may regulate the transfer of internalized T cell receptors into Rab5 and Rab11-positive endosomes to support its rapid recycling.
Collapse
|
95
|
Ficht X, Ruef N, Stolp B, Samson GPB, Moalli F, Page N, Merkler D, Nichols BJ, Diz-Muñoz A, Legler DF, Niggli V, Stein JV. In Vivo Function of the Lipid Raft Protein Flotillin-1 during CD8 + T Cell-Mediated Host Surveillance. THE JOURNAL OF IMMUNOLOGY 2019; 203:2377-2387. [PMID: 31548330 DOI: 10.4049/jimmunol.1900075] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 08/24/2019] [Indexed: 01/12/2023]
Abstract
Flotillin-1 (Flot1) is an evolutionary conserved, ubiquitously expressed lipid raft-associated scaffolding protein. Migration of Flot1-deficient neutrophils is impaired because of a decrease in myosin II-mediated contractility. Flot1 also accumulates in the uropod of polarized T cells, suggesting an analogous role in T cell migration. In this study, we analyzed morphology and migration parameters of murine wild-type and Flot1-/- CD8+ T cells using in vitro assays and intravital two-photon microscopy of lymphoid and nonlymphoid tissues. Flot1-/- CD8+ T cells displayed significant alterations in cell shape and motility parameters in vivo but showed comparable homing to lymphoid organs and intact in vitro migration to chemokines. Furthermore, their clonal expansion and infiltration into nonlymphoid tissues during primary and secondary antiviral immune responses was comparable to wild-type CD8+ T cells. Taken together, Flot1 plays a detectable but unexpectedly minor role for CD8+ T cell behavior under physiological conditions.
Collapse
Affiliation(s)
- Xenia Ficht
- Theodor Kocher Institute, University of Bern, 3012 Bern, Switzerland
| | - Nora Ruef
- Department of Oncology, Microbiology and Immunology, University of Fribourg, 1700 Fribourg, Switzerland
| | - Bettina Stolp
- Theodor Kocher Institute, University of Bern, 3012 Bern, Switzerland.,Department for Infectious Diseases, Integrative Virology, Center for Integrative Infectious Disease Research, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Guerric P B Samson
- Biotechnology Institute Thurgau at the University of Konstanz, 8280 Kreuzlingen, Switzerland
| | - Federica Moalli
- Theodor Kocher Institute, University of Bern, 3012 Bern, Switzerland.,Scientific Institute for Research and Healthcare, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Nicolas Page
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva, Switzerland
| | - Doron Merkler
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva, Switzerland
| | - Ben J Nichols
- Medical Research Council Laboratory of Molecular Biology, Cambridge CB2 0QH, United Kingdom
| | - Alba Diz-Muñoz
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany; and
| | - Daniel F Legler
- Biotechnology Institute Thurgau at the University of Konstanz, 8280 Kreuzlingen, Switzerland
| | - Verena Niggli
- Institute of Pathology, University of Bern, 3008 Bern, Switzerland
| | - Jens V Stein
- Department of Oncology, Microbiology and Immunology, University of Fribourg, 1700 Fribourg, Switzerland;
| |
Collapse
|
96
|
Corso G, Heusermann W, Trojer D, Görgens A, Steib E, Voshol J, Graff A, Genoud C, Lee Y, Hean J, Nordin JZ, Wiklander OPB, El Andaloussi S, Meisner-Kober N. Systematic characterization of extracellular vesicle sorting domains and quantification at the single molecule - single vesicle level by fluorescence correlation spectroscopy and single particle imaging. J Extracell Vesicles 2019; 8:1663043. [PMID: 31579435 PMCID: PMC6758720 DOI: 10.1080/20013078.2019.1663043] [Citation(s) in RCA: 112] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 08/20/2019] [Accepted: 08/21/2019] [Indexed: 02/07/2023] Open
Abstract
Extracellular vesicles (EV) convey biological information by transmitting macromolecules between cells and tissues and are of great promise as pharmaceutical nanocarriers, and as therapeutic per se. Strategies for customizing the EV surface and cargo are being developed to enable their tracking, visualization, loading with pharmaceutical agents and decoration of the surface with tissue targeting ligands. While much progress has been made in the engineering of EVs, an exhaustive comparative analysis of the most commonly exploited EV-associated proteins, as well as a quantification at the molecular level are lacking. Here, we selected 12 EV-related proteins based on MS-proteomics data for comparative quantification of their EV engineering potential. All proteins were expressed with fluorescent protein (FP) tags in EV-producing cells; both parent cells as well as the recovered vesicles were characterized biochemically and biophysically. Using Fluorescence Correlation Spectroscopy (FCS) we quantified the number of FP-tagged molecules per vesicle. We observed different loading efficiencies and specificities for the different proteins into EVs. For the candidates showing the highest loading efficiency in terms of engineering, the molecular levels in the vesicles did not exceed ca 40-60 fluorescent proteins per vesicle upon transient overexpression in the cells. Some of the GFP-tagged EV reporters showed quenched fluorescence and were either non-vesicular, despite co-purification with EVs, or comprised a significant fraction of truncated GFP. The co-expression of each target protein with CD63 was further quantified by widefield and confocal imaging of single vesicles after double transfection of parent cells. In summary, we provide a quantitative comparison for the most commonly used sorting proteins for bioengineering of EVs and introduce a set of biophysical techniques for straightforward quantitative and qualitative characterization of fluorescent EVs to link single vesicle analysis with single molecule quantification.
Collapse
Affiliation(s)
- Giulia Corso
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Wolf Heusermann
- Novartis Institutes for Biomedical Research, Basel, Switzerland.,Imaging Core Facility, Biozentrum, University of Basel, Basel, Switzerland
| | - Dominic Trojer
- Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - André Görgens
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden.,Institute for Transfusion Medicine, University Hospital Essen, Essen, Germany
| | - Emmanuelle Steib
- Novartis Institutes for Biomedical Research, Basel, Switzerland.,Department of Cell Biology, Sciences III, University of Geneva, Geneva Switzerland
| | - Johannes Voshol
- Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Alexandra Graff
- Facility for advanced imaging and microscopy, Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Christel Genoud
- Facility for advanced imaging and microscopy, Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Yi Lee
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden.,Department of Cancer and Stratified Oncology 5, Astar Genome Institute of Singapore, Singapore
| | - Justin Hean
- Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Joel Z Nordin
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | | | | | - Nicole Meisner-Kober
- Novartis Institutes for Biomedical Research, Basel, Switzerland.,Department of Biosciences, University of Salzburg, Salzburg, Austria
| |
Collapse
|
97
|
Li HT, Ye C, Zhou M, Yang Y, Jin Q, Pan CF. Moxifloxacin suppresses airway inflammation and modulates expression of caveolin-1 and flotillin-1 in airway smooth muscle cells of asthmatic rats. ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:469. [PMID: 31700905 DOI: 10.21037/atm.2019.08.43] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Background Moxifloxacin (MXF) possesses anti-inflammatory properties on asthmatic airway smooth muscle cells (ASMCs) beyond their antimicrobial effects, but the mechanisms are still unknown. This study was to investigate effects of MXF on expression of caveolin-1 (Cav-1) and flotillin-1 (FLOT1) in ASMCs in asthmatic rats. Methods ASMCs were collected from the airway and cultured in vitro. Cells from normal rats were treated with normal saline (Group N); cells from asthmatic rats were incubated with normal saline (Group A) or MXF (20 mg/L) (Group M); Cav-1 expression was up-regulated by transferring Cav-1 expressing lentivirus (Group L) and FLOT1 expression down-regulated by using siRNA in cells from asthmatic rats (Group S). The expressions of Cav-1, FLOT1 and p65 NF-κB were measured by Western blotting and quantificational real-time polymerase chain reaction (qRT-PCR), and interleukin-8 (IL-8) and eotaxin contents were measured by enzyme-linked immunosorbent assay (ELISA). Results Compared with normal control, Cav-1 expression significantly decreased in asthmatic groups (P<0.01); MXF up-regulated Cav-1 expression in asthmatic groups (P<0.01). However, compared with normal control, the expression of FLOT1 and p65 NF-κB dramatically increased in asthmatic groups (P<0.01); MXF down-regulated the expression of FLOT1 and p65 NF-κB in asthmatic groups (P<0.01); meanwhile, the expressions of FLOT1 and p65 NF-κB decreased after up-regulation of Cav-1 expression in asthmatic groups (P=0.01). Compared with asthmatic groups, the IL-8 and eotaxin contents significantly decreased in MXF Groups, Cav-1 up-regulation asthmatic groups and FLOT1 down-regulation asthmatic groups (P<0.01). Conclusions MXF can modulate the airway inflammation, upregulate Cav-1 expression, downregulate the expression of FLOT1 and p65 NF-κB in asthmatic rat ASMCs, which may be related to the anti-inflammatory effects of MXF in asthmatic ASMCs.
Collapse
Affiliation(s)
- Hui-Ting Li
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, Tongji University School, Shanghai 200433, China
| | - Cong Ye
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, Tongji University School, Shanghai 200433, China
| | - Min Zhou
- Department of Respiratory Medicine, Shanghai Jinshan Central Hospital, Shanghai 201500, China
| | - Yan Yang
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, Tongji University School, Shanghai 200433, China
| | - Quan Jin
- Department of Respiratory Medicine, Jinshan Hospital, Fudan University, Shanghai 201508, China
| | - Chun-Feng Pan
- Department of Respiratory Medicine, Jinshan Hospital, Fudan University, Shanghai 201508, China
| |
Collapse
|
98
|
Kim DY, Park JA, Kim Y, Noh M, Park S, Lie E, Kim E, Kim YM, Kwon YG. SALM4 regulates angiogenic functions in endothelial cells through VEGFR2 phosphorylation at Tyr1175. FASEB J 2019; 33:9842-9857. [PMID: 31170000 PMCID: PMC6704462 DOI: 10.1096/fj.201802516rr] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Angiogenesis depends on VEGF-mediated signaling. However, the regulatory mechanisms and functions of individual VEGF receptor 2 (VEGFR2) phosphorylation sites remain unclear. Here, we report that synaptic adhesion-like molecule 4 (SALM4) regulates a specific VEGFR2 phosphorylation site. SALM4 silencing in HUVECs and Salm4 knockout (KO) in lung endothelial cells (ECs) of Salm4−/− mice suppressed phosphorylation of VEGFR2 tyrosine (Y) 1175 (Y1173 in mice) and downstream signaling upon VEGF-A stimulation. However, VEGFR2 phosphorylation at Y951 (Y949 in mice) and Y1214 (Y1212 in mice) remained unchanged. Knockdown and KO of SALM4 inhibited VEGF-A–induced angiogenic functions of ECs. SALM4 depletion reduced endothelial leakage, sprouting, and migratory activities. Furthermore, in an ischemia and reperfusion (I/R) model, brain injury was attenuated in Salm4−/− mice compared with wild-type (WT) mice. In brain lysates after I/R, VEGFR2 phosphorylation at Y949, Y1173, and Y1212 were induced in WT brains, but only Y1173 phosphorylation of VEGFR2 was reduced in Salm4−/− brains. Taken together, our results demonstrate that SALM4 specifically regulates VEGFR2 phosphorylation at Y1175 (Y1173 in mice), thereby fine-tuning VEGF signaling in ECs.—Kim, D. Y., Park, J. A., Kim, Y., Noh, M., Park, S., Lie, E., Kim, E., Kim, Y.-M., Kwon, Y.-G. SALM4 regulates angiogenic functions in endothelial cells through VEGFR2 phosphorylation at Tyr1175.
Collapse
Affiliation(s)
- Dong Young Kim
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, South Korea
| | - Jeong Ae Park
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, South Korea
| | - Yeomyung Kim
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, South Korea
| | - Minyoung Noh
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, South Korea
| | - Songyi Park
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, South Korea
| | - Eunkyung Lie
- Department of Biological Sciences, Korea Advanced Institute for Science and Technology, Daejeon, South Korea
| | - Eunjoon Kim
- Department of Biological Sciences, Korea Advanced Institute for Science and Technology, Daejeon, South Korea
| | - Young-Myeong Kim
- Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon-si, South Korea
| | - Young-Guen Kwon
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, South Korea
| |
Collapse
|
99
|
Fan W, Guo J, Gao B, Zhang W, Ling L, Xu T, Pan C, Li L, Chen S, Wang H, Zhang J, Wang X. Flotillin-mediated endocytosis and ALIX-syntenin-1-mediated exocytosis protect the cell membrane from damage caused by necroptosis. Sci Signal 2019; 12:12/583/eaaw3423. [PMID: 31138766 DOI: 10.1126/scisignal.aaw3423] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Necroptosis is a form of regulated necrosis that is implicated in various human diseases including Alzheimer's disease. Necroptosis requires the translocation of the pseudokinase MLKL from the cytosol to the plasma membrane after its phosphorylation by the kinase RIPK3. Using protein cross-linking followed by affinity purification, we detected the lipid raft-associated proteins flotillin-1 and flotillin-2 and the ESCRT-associated proteins ALIX and syntenin-1 in membrane-localized MLKL immunoprecipitates. Phosphorylated MLKL was removed from membranes through either flotillin-mediated endocytosis followed by lysosomal degradation or ALIX-syntenin-1-mediated exocytosis. Thus, cells undergoing necroptosis need to overcome these independent suppressive mechanisms before plasma membrane disruption can occur.
Collapse
Affiliation(s)
- Weiliang Fan
- National Institute of Biological Sciences, Beijing, No. 7 Science Park Road, Zhongguancun Life Science Park, Beijing 102206, China.,Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing 102206, China
| | - Jia Guo
- National Institute of Biological Sciences, Beijing, No. 7 Science Park Road, Zhongguancun Life Science Park, Beijing 102206, China.,Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing 102206, China
| | - Beichen Gao
- National Institute of Biological Sciences, Beijing, No. 7 Science Park Road, Zhongguancun Life Science Park, Beijing 102206, China.,Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing 102206, China
| | - Wenbin Zhang
- National Institute of Biological Sciences, Beijing, No. 7 Science Park Road, Zhongguancun Life Science Park, Beijing 102206, China.,Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing 102206, China
| | - Liucong Ling
- National Institute of Biological Sciences, Beijing, No. 7 Science Park Road, Zhongguancun Life Science Park, Beijing 102206, China.,Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing 102206, China
| | - Tao Xu
- National Institute of Biological Sciences, Beijing, No. 7 Science Park Road, Zhongguancun Life Science Park, Beijing 102206, China.,Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing 102206, China
| | - Chenjie Pan
- National Institute of Biological Sciences, Beijing, No. 7 Science Park Road, Zhongguancun Life Science Park, Beijing 102206, China.,Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing 102206, China
| | - Lin Li
- National Institute of Biological Sciences, Beijing, No. 7 Science Park Road, Zhongguancun Life Science Park, Beijing 102206, China.,Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing 102206, China
| | - She Chen
- National Institute of Biological Sciences, Beijing, No. 7 Science Park Road, Zhongguancun Life Science Park, Beijing 102206, China.,Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing 102206, China
| | - Hua Wang
- Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China.,Department of Pathology, Peking University Third Hospital, Beijing 100191, China
| | - Jing Zhang
- Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China.,Department of Pathology, University of Washington School of Medicine, Seattle, WA, USA
| | - Xiaodong Wang
- National Institute of Biological Sciences, Beijing, No. 7 Science Park Road, Zhongguancun Life Science Park, Beijing 102206, China. .,Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing 102206, China
| |
Collapse
|
100
|
Zhang L, Tian XY, Chan CKW, Bai Q, Cheng CK, Chen FM, Cheung MSH, Yin B, Yang H, Yung WY, Chen Z, Ding F, Leung KCF, Zhang C, Huang Y, Lau JYW, Choi CHJ. Promoting the Delivery of Nanoparticles to Atherosclerotic Plaques by DNA Coating. ACS APPLIED MATERIALS & INTERFACES 2019; 11:13888-13904. [PMID: 30516979 DOI: 10.1021/acsami.8b17928] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Many nanoparticle-based carriers to atherosclerotic plaques contain peptides, lipoproteins, and sugars, yet the application of DNA-based nanostructures for targeting plaques remains infrequent. In this work, we demonstrate that DNA-coated superparamagnetic iron oxide nanoparticles (DNA-SPIONs), prepared by attaching DNA oligonucleotides to poly(ethylene glycol)-coated SPIONs (PEG-SPIONs), effectively accumulate in the macrophages of atherosclerotic plaques following an intravenous injection into apolipoprotein E knockout (ApoE-/-) mice. DNA-SPIONs enter RAW 264.7 macrophages faster and more abundantly than PEG-SPIONs. DNA-SPIONs mostly enter RAW 264.7 cells by engaging Class A scavenger receptors (SR-A) and lipid rafts and traffic inside the cell along the endolysosomal pathway. ABS-SPIONs, nanoparticles with a similarly polyanionic surface charge as DNA-SPIONs but bearing abasic oligonucleotides also effectively bind to SR-A and enter RAW 264.7 cells. Near-infrared fluorescence imaging reveals evident localization of DNA-SPIONs in the heart and aorta 30 min post-injection. Aortic iron content for DNA-SPIONs climbs to the peak (∼60% ID/g) 2 h post-injection (accompanied by profuse accumulation in the aortic root), but it takes 8 h for PEG-SPIONs to reach the peak aortic amount (∼44% ID/g). ABS-SPIONs do not appreciably accumulate in the aorta or aortic root, suggesting that the DNA coating (not the surface charge) dictates in vivo plaque accumulation. Flow cytometry analysis reveals more pronounced uptake of DNA-SPIONs by hepatic endothelial cells, splenic macrophages and dendritic cells, and aortic M2 macrophages (the cell type with the highest uptake in the aorta) than PEG-SPIONs. In summary, coating nanoparticles with DNA is an effective strategy of promoting their systemic delivery to atherosclerotic plaques.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Wing-Yin Yung
- Department of Chemistry , Hong Kong Baptist University , Kowloon, Hong Kong China
| | | | - Fei Ding
- School of Chemistry and Chemical Engineering, State Key Laboratory of Metal Matrix Composites , Shanghai Jiao Tong University , Shanghai , China
| | - Ken Cham-Fai Leung
- Department of Chemistry , Hong Kong Baptist University , Kowloon, Hong Kong China
| | - Chuan Zhang
- School of Chemistry and Chemical Engineering, State Key Laboratory of Metal Matrix Composites , Shanghai Jiao Tong University , Shanghai , China
| | | | | | | |
Collapse
|