1
|
Hnath B, Dokholyan NV. Novel extracellular vesicle release pathway facilitated by toxic superoxide dismutase 1 oligomers. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.07.647611. [PMID: 40291716 PMCID: PMC12026985 DOI: 10.1101/2025.04.07.647611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating neurodegenerative disease resulting in paralysis and death within three to five years. Mutations in over forty different proteins have been linked to ALS, leading to controversy whether ALS is one disease or many diseases with a similar phenotype. Mutations in Cu,Zn superoxide dismutase 1 (SOD1) are only found in 2-3% of ALS cases, yet misfolded SOD1 is found in both sporadic (sALS) and familial (fALS) patients. Yet, mutations in TDP-43 or FUS increase the level of misfolded SOD1 on extracellular vesicles (EVs). Additionally, small EVs isolated from ALS patient samples caused cell death of wild type motor neurons and myotubules. The toxicity and protein alterations of ALS EVs have led to the theory that EVs are responsible for the spread of ALS. We hypothesize that previously-identified toxic trimeric SOD1 is spreading on EVs in ALS and altering the spread of other ALS-related proteins, linking them to a common mechanism. To test our hypothesis, we isolate EVs from motor neuron-like cells expressing trimer stabilizing mutations and perform a sandwich enzyme-linked immunoassay (ELISA) (CD9 capture antibody) to quantify whether misfolded SOD1 and 17 other ALS-related proteins increase or decrease on EVs with trimer stabilization. We identify which EV release pathway is being affected by trimeric SOD1 utilizing endocytosis and exocytosis inhibitors, and determine if any specific EV-related proteins are altered with trimer stabilization. We establish that VAPB, VCP, and Stathmin-2 increase on EVs with trimer stabilization. The common pathway between SOD1 and three other ALS-associated proteins is affected by multiple pathways, including the Caveolae endocytosis pathway, suggesting a novel hybrid pathway of EV release present in ALS.
Collapse
|
2
|
Candor K, Ding L, Balchand S, Hammonds JE, Spearman P. The CLIC/GEEC pathway regulates particle uptake and formation of the virus-containing compartment (VCC) in HIV-1-infected macrophages. PLoS Pathog 2025; 21:e1012564. [PMID: 40067817 PMCID: PMC11925468 DOI: 10.1371/journal.ppat.1012564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 03/20/2025] [Accepted: 02/12/2025] [Indexed: 03/22/2025] Open
Abstract
HIV-1 particles are captured by the immunoglobulin superfamily member Siglec-1 on the surface of macrophages and dendritic cells, leading to particle internalization and facilitating trans-infection of CD4+ T cells. HIV-1-infected macrophages develop a unique intracellular compartment termed the virus-containing compartment (VCC) that exhibits characteristic markers of the late endosome and is enriched in components of the plasma membrane (PM). The VCC has been proposed as the major site of particle assembly in macrophages. Depleting Siglec-1 from macrophages significantly reduces VCC formation, implying a link between the capture and uptake of external HIV-1 particles and the development of VCCs within HIV-infected cells. We found that internalization of particles to the VCC was independent of clathrin, but required dynamin-2. CD98 and CD44, classical markers of the CLIC/GEEC pathway, colocalized with Siglec-1 and HIV-1 particles within the VCC. Virus-like particles (VLPs) were taken up within CD98 and Siglec-1-enriched tubular membranes that migrated centripetally over time to form VCC-like structures. Inhibition of CLIC/GEEC-mediated endocytosis resulted in the arrest of captured HIV-1 particles on the macrophage cell surface, prevented VCC formation, and significantly reduced the efficiency of trans-infection of T cells. These findings indicate that following capture of virus by Siglec-1, particles follow an endocytic route to the VCC that requires both the CLIC/GEEC pathway and dynamin-2. We propose a model in which internalization of HIV-1 particles together with CLIC/GEEC membranes leads to the formation of the VCC in HIV-infected macrophages, creating an intracellular platform that facilitates further particle assembly and budding.
Collapse
Affiliation(s)
- Kathleen Candor
- Immunology Graduate Program, University of Cincinnati, and Infectious Diseases Division, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
- Infectious Diseases Division, Cincinnati Children’s Hospital Medical Center and University of Cincinnati, Cincinnati, Ohio United States of America
| | - Lingmei Ding
- Infectious Diseases Division, Cincinnati Children’s Hospital Medical Center and University of Cincinnati, Cincinnati, Ohio United States of America
| | - Sai Balchand
- Infectious Diseases Division, Cincinnati Children’s Hospital Medical Center and University of Cincinnati, Cincinnati, Ohio United States of America
| | - Jason E. Hammonds
- Infectious Diseases Division, Cincinnati Children’s Hospital Medical Center and University of Cincinnati, Cincinnati, Ohio United States of America
| | - Paul Spearman
- Infectious Diseases Division, Cincinnati Children’s Hospital Medical Center and University of Cincinnati, Cincinnati, Ohio United States of America
| |
Collapse
|
3
|
Sánchez-Arribas N, Velasco Rodríguez B, Aicart E, Guerrero-Martínez A, Junquera E, Taboada P. Lipid nanoparticles as nano-Trojan-horses for siRNA delivery and gene-knockdown. J Colloid Interface Sci 2025; 679:975-987. [PMID: 39488022 DOI: 10.1016/j.jcis.2024.10.115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 10/17/2024] [Accepted: 10/19/2024] [Indexed: 11/04/2024]
Abstract
The therapeutic messenger RNA strategies, such as those using small interfering RNAs, take several advantages (versatility, efficiency and selectivity) over plasmid DNA-based strategies. However, the challenge remains to find nanovectors capable of properly loading the genetic material, transporting it through troublesome environments, like a tumoral site, and delivering it into the cytoplasm of target cells. Here, lipid nanoparticles, consisting of a gemini cationic/neutral helper lipid mixture, are proposed as siRNA nanovector. Cells from cervical and brain cancer overexpressing the green fluorescent protein (GFP) were chosen to analyse the biological response as well as the efficiency and safety of the siRNA-loaded nanovector according to the cell phenotype. Flow cytometry and epifluorescence or confocal microscopy were used to follow the gene knockdown in these overexpressed cells. The effect of the nanovector on cellular proliferation was evaluated with cytotoxicity assays while their potential oxidative stress generation was determined by quantifying the generation of reactive oxygen species. To explore the mechanism of cellular uptake, different inhibitors of endocytic pathways were used during incubation with cells. Finally, nanovectors were incubated in 3D-grown cells (spheroids) to see whether they can penetrate the complex tumoral microenvironments, their efficiency to knockdown GFP expression being monitored by confocal microscopy.
Collapse
Affiliation(s)
- Natalia Sánchez-Arribas
- Dpto. Química Física, Fac. CC. Químicas, Universidad Complutense de Madrid, Av. Complutense s/n, 28040 Madrid, Spain
| | - Brenda Velasco Rodríguez
- Departamento de Física de Partículas-Facultad de Física, Instituto de Materiales (IMATUS) e Instituto de Investigaciones Sanitarias (IDIS), Universidad de Santiago de Compostela, Campus Vida, E-15782 Santiago de Compostela, Spain
| | - Emilio Aicart
- Dpto. Química Física, Fac. CC. Químicas, Universidad Complutense de Madrid, Av. Complutense s/n, 28040 Madrid, Spain
| | - Andrés Guerrero-Martínez
- Dpto. Química Física, Fac. CC. Químicas, Universidad Complutense de Madrid, Av. Complutense s/n, 28040 Madrid, Spain
| | - Elena Junquera
- Dpto. Química Física, Fac. CC. Químicas, Universidad Complutense de Madrid, Av. Complutense s/n, 28040 Madrid, Spain.
| | - Pablo Taboada
- Departamento de Física de Partículas-Facultad de Física, Instituto de Materiales (IMATUS) e Instituto de Investigaciones Sanitarias (IDIS), Universidad de Santiago de Compostela, Campus Vida, E-15782 Santiago de Compostela, Spain.
| |
Collapse
|
4
|
Garcia BM, Douka S, Mertins O, Mastrobattista E, Han SW. Efficacy of Chitosan-N-Arginine Chitosomes in mRNA Delivery and Cell Viability Enhancement. ACS APPLIED BIO MATERIALS 2024; 7:8261-8271. [PMID: 39558637 PMCID: PMC11653394 DOI: 10.1021/acsabm.4c00983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 11/11/2024] [Accepted: 11/13/2024] [Indexed: 11/20/2024]
Abstract
Cationic lipid-based carriers are recognized for their ability to complex with mRNA and effectively deliver the mRNA for vaccination and therapeutic purposes. However, the significant cytotoxicity of these carriers often restricts their practical application. In the present study, polymer-lipid hybrid nanoparticles, termed chitosomes, incorporating chitosan-N-arginine (CSA) with the DOTAP cationic lipid and the DOPE helper lipid, were synthesized and evaluated. The addition of CSA to the lipid formulations improved their physicochemical stability and enhanced mRNA complexation, resulting in high transfection rates in the HeLa and HEK293T cell lines. However, the transfection efficiency was low in the NIH-3T3 cell line, indicating a cell type-specific response to chitosomes. Importantly, CSA significantly reduced the cytotoxicity typically associated with DOTAP. Overall, the present study indicated that optimizing the ratio of CSA to DOTAP is crucial for developing mRNA nanocarriers to achieve high transfection efficiency and reduce cytotoxicity across different cell lines.
Collapse
Affiliation(s)
- Bianca
B. M. Garcia
- Department
of Biophysics, Paulista School of Medicine, Federal University of São Paulo, 04023-062 São Paulo, Brazil
- Pharmaceutics
Division, Utrecht Institute for Pharmaceutical Sciences (UIPS), Faculty
of Science, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
| | - Stefania Douka
- Pharmaceutics
Division, Utrecht Institute for Pharmaceutical Sciences (UIPS), Faculty
of Science, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
| | - Omar Mertins
- Department
of Biophysics, Paulista School of Medicine, Federal University of São Paulo, 04023-062 São Paulo, Brazil
| | - Enrico Mastrobattista
- Pharmaceutics
Division, Utrecht Institute for Pharmaceutical Sciences (UIPS), Faculty
of Science, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
| | - Sang W. Han
- Department
of Biophysics, Paulista School of Medicine, Federal University of São Paulo, 04023-062 São Paulo, Brazil
| |
Collapse
|
5
|
Cavallari N, Johnson A, Nagl C, Seiser S, Rechberger GN, Züllig T, Kufer TA, Elbe-Bürger A, Geiselhart S, Hoffmann-Sommergruber K. Nonspecific lipid-transfer proteins trigger TLR2 and NOD2 signaling and undergo ligand-dependent endocytosis in epithelial cells. J Allergy Clin Immunol 2024; 154:1289-1299. [PMID: 39084297 DOI: 10.1016/j.jaci.2024.07.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 07/10/2024] [Accepted: 07/24/2024] [Indexed: 08/02/2024]
Abstract
BACKGROUND Allergens can cross the epithelial barrier to enter the body but how this cellular passage affects protein structures and the downstream interactions with the immune system are still open questions. OBJECTIVE We sought to show the molecular details and the effects of 3 nonspecific lipid transfer proteins (nsLTPs; Mal d 3 [allergenic nsLTP1 from apple], Cor a 8 [allergenic nsLTP1 from hazelnut], and Pru p 3 [allergenic nsLTP1 from peach]) on epithelial cell uptake and transport. METHODS We used fluorescent imaging, flow cytometry, and proteomic and lipidomic screenings to identify the mechanism involved in nsLTP cellular uptake and signaling on selected epithelial and transgenic cell lines. RESULTS nsLTPs are transported across the epithelium without affecting cell membrane stability or viability, and allergen uptake was largely impaired by inhibition of clathrin-mediated endocytosis. Analysis of the lipidome associated with nsLTPs showed a wide variety of lipid ligands predicted to bind inside the allergen hydrophobic cavity. Importantly, the internalization of nsLTPs was contingent on these ligands in the protein complex. nsLTPs were found to initiate cellular signaling via Toll-like receptor 2 but not the cluster of differentiation 1 protein receptor, despite neither being essential for nsLTP endocytosis. We also provide evidence that the 3 allergens induced intracellular stress signaling through activation of the NOD2 pathway. CONCLUSIONS Our work consolidates the current model on nsLTP-epithelial cell interplay and adds molecular details about cell transport and signaling. In addition, we have developed a versatile toolbox to extend these investigations to other allergens and cell types.
Collapse
Affiliation(s)
- Nicola Cavallari
- Center for Pathophysiology, Infectiology and Immunology, Department of Pathophysiology and Allergy Research, Vienna, Austria
| | - Alexander Johnson
- Center for Anatomy & Cell Biology, Division of Anatomy, Medical University of Vienna, Vienna, Austria; Medical Imaging Cluster, Vienna, Austria
| | - Christoph Nagl
- Center for Pathophysiology, Infectiology and Immunology, Department of Pathophysiology and Allergy Research, Vienna, Austria
| | - Saskia Seiser
- Department of Dermatology, Vienna General Hospital, Medical University of Vienna, Vienna, Austria
| | - Gerald N Rechberger
- Institute of Molecular Biosciences, University of Graz, NAWI Graz, Graz, Austria
| | - Thomas Züllig
- Institute of Molecular Biosciences, University of Graz, NAWI Graz, Graz, Austria
| | - Thomas A Kufer
- Department of Immunology, Institute of Nutritional Medicine, University of Hohenheim, Stuttgart, Germany
| | - Adelheid Elbe-Bürger
- Department of Dermatology, Vienna General Hospital, Medical University of Vienna, Vienna, Austria
| | - Sabine Geiselhart
- Center for Pathophysiology, Infectiology and Immunology, Department of Pathophysiology and Allergy Research, Vienna, Austria
| | - Karin Hoffmann-Sommergruber
- Center for Pathophysiology, Infectiology and Immunology, Department of Pathophysiology and Allergy Research, Vienna, Austria.
| |
Collapse
|
6
|
Sandvig K, Iversen TG, Skotland T. Entry of nanoparticles into cells and tissues: status and challenges. BEILSTEIN JOURNAL OF NANOTECHNOLOGY 2024; 15:1017-1029. [PMID: 39161463 PMCID: PMC11331539 DOI: 10.3762/bjnano.15.83] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 07/23/2024] [Indexed: 08/21/2024]
Abstract
In this article we discuss how nanoparticles (NPs) of different compositions may interact with and be internalized by cells, and the consequences of that for cellular functions. A large number of NPs are made with the intention to improve cancer treatment, the goal being to increase the fraction of injected drug delivered to the tumor and thereby improve the therapeutic effect and decrease side effects. Thus, we discuss how NPs are delivered to tumors and some challenges related to investigations of biodistribution, pharmacokinetics, and excretion. Finally, we discuss requirements for bringing NPs into clinical use and aspects when it comes to usage of complex and slowly degraded or nondegradable NPs.
Collapse
Affiliation(s)
- Kirsten Sandvig
- Department of Molecular Cell Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, 0379 Oslo, Norway
- Centre for Cancer Cell Reprogramming, University of Oslo, 0379 Oslo, Norway
- Department of Biosciences, University of Oslo, 0316 Oslo, Norway
| | - Tore Geir Iversen
- Department of Molecular Cell Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, 0379 Oslo, Norway
- Centre for Cancer Cell Reprogramming, University of Oslo, 0379 Oslo, Norway
| | - Tore Skotland
- Department of Molecular Cell Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, 0379 Oslo, Norway
- Centre for Cancer Cell Reprogramming, University of Oslo, 0379 Oslo, Norway
| |
Collapse
|
7
|
Jiang A, Kudo K, Gormal RS, Ellis S, Guo S, Wallis TP, Longfield SF, Robinson PJ, Johnson ME, Joensuu M, Meunier FA. Dynamin1 long- and short-tail isoforms exploit distinct recruitment and spatial patterns to form endocytic nanoclusters. Nat Commun 2024; 15:4060. [PMID: 38744819 PMCID: PMC11094030 DOI: 10.1038/s41467-024-47677-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Accepted: 04/09/2024] [Indexed: 05/16/2024] Open
Abstract
Endocytosis requires a coordinated framework of molecular interactions that ultimately lead to the fission of nascent endocytic structures. How cytosolic proteins such as dynamin concentrate at discrete sites that are sparsely distributed across the plasma membrane remains poorly understood. Two dynamin-1 major splice variants differ by the length of their C-terminal proline-rich region (short-tail and long-tail). Using sptPALM in PC12 cells, neurons and MEF cells, we demonstrate that short-tail dynamin-1 isoforms ab and bb display an activity-dependent recruitment to the membrane, promptly followed by their concentration into nanoclusters. These nanoclusters are sensitive to both Calcineurin and dynamin GTPase inhibitors, and are larger, denser, and more numerous than that of long-tail isoform aa. Spatiotemporal modelling confirms that dynamin-1 isoforms perform distinct search patterns and undergo dimensional reduction to generate endocytic nanoclusters, with short-tail isoforms more robustly exploiting lateral trapping in the generation of nanoclusters compared to the long-tail isoform.
Collapse
Affiliation(s)
- Anmin Jiang
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Kye Kudo
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Rachel S Gormal
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Sevannah Ellis
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Sikao Guo
- Department of Biophysics, Johns Hopkins University, 3400 N Charles St, Baltimore, MD, 21218, USA
| | - Tristan P Wallis
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Shanley F Longfield
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Phillip J Robinson
- Cell Signalling Unit, Children's Medical Research Institute, The University of Sydney, Sydney, NSW, 2145, Australia
| | - Margaret E Johnson
- Department of Biophysics, Johns Hopkins University, 3400 N Charles St, Baltimore, MD, 21218, USA
| | - Merja Joensuu
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, 4072, Australia.
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, 4072, Australia.
| | - Frédéric A Meunier
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, 4072, Australia.
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, 4072, Australia.
| |
Collapse
|
8
|
Sotodosos-Alonso L, Del Pozo MA. Cancer cell invasion: Caveolae and invadosomes are partners in crime. Curr Biol 2024; 34:R244-R246. [PMID: 38531317 DOI: 10.1016/j.cub.2024.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/28/2024]
Abstract
During cancer progression, tumor cells need to disseminate by remodeling the extracellular tumor matrix. A recent study sheds light on the intricate cooperation between caveolae and invadosomes that facilitates the spread of cancer cells.
Collapse
Affiliation(s)
- Laura Sotodosos-Alonso
- Mechanoadaptation and Caveolae Biology Laboratory, Novel Mechanisms of Atherosclerosis Program, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), 28029 Madrid, Spain
| | - Miguel A Del Pozo
- Mechanoadaptation and Caveolae Biology Laboratory, Novel Mechanisms of Atherosclerosis Program, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), 28029 Madrid, Spain.
| |
Collapse
|
9
|
Kryvenko V, Vadász I. Alveolar-capillary endocytosis and trafficking in acute lung injury and acute respiratory distress syndrome. Front Immunol 2024; 15:1360370. [PMID: 38533500 PMCID: PMC10963603 DOI: 10.3389/fimmu.2024.1360370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 02/29/2024] [Indexed: 03/28/2024] Open
Abstract
Acute respiratory distress syndrome (ARDS) is associated with high morbidity and mortality but lacks specific therapeutic options. Diverse endocytic processes play a key role in all phases of acute lung injury (ALI), including the initial insult, development of respiratory failure due to alveolar flooding, as a consequence of altered alveolar-capillary barrier function, as well as in the resolution or deleterious remodeling after injury. In particular, clathrin-, caveolae-, endophilin- and glycosylphosphatidyl inositol-anchored protein-mediated endocytosis, as well as, macropinocytosis and phagocytosis have been implicated in the setting of acute lung damage. This manuscript reviews our current understanding of these endocytic pathways and subsequent intracellular trafficking in various phases of ALI, and also aims to identify potential therapeutic targets for patients with ARDS.
Collapse
Affiliation(s)
- Vitalii Kryvenko
- Department of Internal Medicine, Justus Liebig University, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
- The Cardio-Pulmonary Institute (CPI), Giessen, Germany
- Institute for Lung Health (ILH), Giessen, Germany
| | - István Vadász
- Department of Internal Medicine, Justus Liebig University, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
- The Cardio-Pulmonary Institute (CPI), Giessen, Germany
- Institute for Lung Health (ILH), Giessen, Germany
| |
Collapse
|
10
|
Štefl M, Takamiya M, Middel V, Tekpınar M, Nienhaus K, Beil T, Rastegar S, Strähle U, Nienhaus GU. Caveolae disassemble upon membrane lesioning and foster cell survival. iScience 2024; 27:108849. [PMID: 38303730 PMCID: PMC10831942 DOI: 10.1016/j.isci.2024.108849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 11/22/2023] [Accepted: 01/03/2024] [Indexed: 02/03/2024] Open
Abstract
Repair of lesions in the plasma membrane is key to sustaining cellular homeostasis. Cells maintain cytoplasmic as well as membrane-bound stores of repair proteins that can rapidly precipitate at the site of membrane lesions. However, little is known about the origins of lipids and proteins for resealing and repair of the plasma membrane. Here we study the dynamics of caveolar proteins after laser-induced lesioning of plasma membranes of mammalian C2C12 tissue culture cells and muscle cells of intact zebrafish embryos. Single-molecule diffusivity measurements indicate that caveolar clusters break up into smaller entities after wounding. Unlike Annexins and Dysferlin, caveolar proteins do not accumulate at the lesion patch. In caveolae-depleted cavin1a knockout zebrafish embryos, lesion patch formation is impaired, and injured cells show reduced survival. Our data suggest that caveolae disassembly releases surplus plasma membrane near the lesion to facilitate membrane repair after initial patch formation for emergency sealing.
Collapse
Affiliation(s)
- Martin Štefl
- Institute of Applied Physics (APH), Karlsruhe Institute of Technology (KIT), Wolfgang Gaede-Strasse 1, 76131 Karlsruhe, Germany
| | - Masanari Takamiya
- Institute of Biological and Chemical Systems (IBCS), Karlsruhe Institute of Technology (KIT), PO Box 3640, 76021 Karlsruhe, Germany
| | - Volker Middel
- Institute of Biological and Chemical Systems (IBCS), Karlsruhe Institute of Technology (KIT), PO Box 3640, 76021 Karlsruhe, Germany
| | - Miyase Tekpınar
- Institute of Applied Physics (APH), Karlsruhe Institute of Technology (KIT), Wolfgang Gaede-Strasse 1, 76131 Karlsruhe, Germany
| | - Karin Nienhaus
- Institute of Applied Physics (APH), Karlsruhe Institute of Technology (KIT), Wolfgang Gaede-Strasse 1, 76131 Karlsruhe, Germany
| | - Tanja Beil
- Institute of Biological and Chemical Systems (IBCS), Karlsruhe Institute of Technology (KIT), PO Box 3640, 76021 Karlsruhe, Germany
| | - Sepand Rastegar
- Institute of Biological and Chemical Systems (IBCS), Karlsruhe Institute of Technology (KIT), PO Box 3640, 76021 Karlsruhe, Germany
| | - Uwe Strähle
- Institute of Biological and Chemical Systems (IBCS), Karlsruhe Institute of Technology (KIT), PO Box 3640, 76021 Karlsruhe, Germany
- Centre for Organismal Studies (COS), Heidelberg University, Im Neuenheimer Feld 230, 69120 Heidelberg, Germany
| | - Gerd Ulrich Nienhaus
- Institute of Applied Physics (APH), Karlsruhe Institute of Technology (KIT), Wolfgang Gaede-Strasse 1, 76131 Karlsruhe, Germany
- Institute of Biological and Chemical Systems (IBCS), Karlsruhe Institute of Technology (KIT), PO Box 3640, 76021 Karlsruhe, Germany
- Institute of Nanotechnology (INT), Karlsruhe Institute of Technology (KIT), PO Box 3640, 76021 Karlsruhe, Germany
- Department of Physics, University of Illinois at Urbana−Champaign, Urbana, IL 61801, USA
| |
Collapse
|
11
|
Gandek TB, van der Koog L, Nagelkerke A. A Comparison of Cellular Uptake Mechanisms, Delivery Efficacy, and Intracellular Fate between Liposomes and Extracellular Vesicles. Adv Healthc Mater 2023; 12:e2300319. [PMID: 37384827 PMCID: PMC11469107 DOI: 10.1002/adhm.202300319] [Citation(s) in RCA: 56] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 06/21/2023] [Accepted: 06/21/2023] [Indexed: 07/01/2023]
Abstract
A key aspect for successful drug delivery via lipid-based nanoparticles is their internalization in target cells. Two prominent examples of such drug delivery systems are artificial phospholipid-based carriers, such as liposomes, and their biological counterparts, the extracellular vesicles (EVs). Despite a wealth of literature, it remains unclear which mechanisms precisely orchestrate nanoparticle-mediated cargo delivery to recipient cells and the subsequent intracellular fate of therapeutic cargo. In this review, internalization mechanisms involved in the uptake of liposomes and EVs by recipient cells are evaluated, also exploring their intracellular fate after intracellular trafficking. Opportunities are highlighted to tweak these internalization mechanisms and intracellular fates to enhance the therapeutic efficacy of these drug delivery systems. Overall, literature to date shows that both liposomes and EVs are predominantly internalized through classical endocytosis mechanisms, sharing a common fate: accumulation inside lysosomes. Studies tackling the differences between liposomes and EVs, with respect to cellular uptake, intracellular delivery and therapy efficacy, remain scarce, despite its importance for the selection of an appropriate drug delivery system. In addition, further exploration of functionalization strategies of both liposomes and EVs represents an important avenue to pursue in order to control internalization and fate, thereby improving therapeutic efficacy.
Collapse
Affiliation(s)
- Timea B. Gandek
- Pharmaceutical AnalysisGroningen Research Institute of PharmacyUniversity of GroningenP.O. Box 196, XB20Groningen9700 ADThe Netherlands
| | - Luke van der Koog
- Molecular PharmacologyGroningen Research Institute of PharmacyUniversity of GroningenP.O. Box 196, XB10Groningen9700 ADThe Netherlands
| | - Anika Nagelkerke
- Pharmaceutical AnalysisGroningen Research Institute of PharmacyUniversity of GroningenP.O. Box 196, XB20Groningen9700 ADThe Netherlands
| |
Collapse
|
12
|
Kenworthy AK, Han B, Ariotti N, Parton RG. The Role of Membrane Lipids in the Formation and Function of Caveolae. Cold Spring Harb Perspect Biol 2023; 15:a041413. [PMID: 37277189 PMCID: PMC10513159 DOI: 10.1101/cshperspect.a041413] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Caveolae are plasma membrane invaginations with a distinct lipid composition. Membrane lipids cooperate with the structural components of caveolae to generate a metastable surface domain. Recent studies have provided insights into the structure of essential caveolar components and how lipids are crucial for the formation, dynamics, and disassembly of caveolae. They also suggest new models for how caveolins, major structural components of caveolae, insert into membranes and interact with lipids.
Collapse
Affiliation(s)
- Anne K Kenworthy
- Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, Virginia 22903, USA
- Department of Molecular Physiology and Biological Physics, University of Virginia School of Medicine, Charlottesville, Virginia 22903, USA
| | - Bing Han
- Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, Virginia 22903, USA
- Department of Molecular Physiology and Biological Physics, University of Virginia School of Medicine, Charlottesville, Virginia 22903, USA
| | - Nicholas Ariotti
- Institute for Molecular Bioscience, The University of Queensland, 4072 Brisbane, Australia
| | - Robert G Parton
- Institute for Molecular Bioscience, The University of Queensland, 4072 Brisbane, Australia
- Centre for Microscopy and Microanalysis, The University of Queensland, 4072 Brisbane, Australia
| |
Collapse
|
13
|
Banushi B, Joseph SR, Lum B, Lee JJ, Simpson F. Endocytosis in cancer and cancer therapy. Nat Rev Cancer 2023:10.1038/s41568-023-00574-6. [PMID: 37217781 DOI: 10.1038/s41568-023-00574-6] [Citation(s) in RCA: 79] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/11/2023] [Indexed: 05/24/2023]
Abstract
Endocytosis is a complex process whereby cell surface proteins, lipids and fluid from the extracellular environment are packaged, sorted and internalized into cells. Endocytosis is also a mechanism of drug internalization into cells. There are multiple routes of endocytosis that determine the fate of molecules, from degradation in the lysosomes to recycling back to the plasma membrane. The overall rates of endocytosis and temporal regulation of molecules transiting through endocytic pathways are also intricately linked with signalling outcomes. This process relies on an array of factors, such as intrinsic amino acid motifs and post-translational modifications. Endocytosis is frequently disrupted in cancer. These disruptions lead to inappropriate retention of receptor tyrosine kinases on the tumour cell membrane, changes in the recycling of oncogenic molecules, defective signalling feedback loops and loss of cell polarity. In the past decade, endocytosis has emerged as a pivotal regulator of nutrient scavenging, response to and regulation of immune surveillance and tumour immune evasion, tumour metastasis and therapeutic drug delivery. This Review summarizes and integrates these advances into the understanding of endocytosis in cancer. The potential to regulate these pathways in the clinic to improve cancer therapy is also discussed.
Collapse
Affiliation(s)
- Blerida Banushi
- Frazer Institute, University of Queensland, Woolloongabba, Queensland, Australia
| | - Shannon R Joseph
- Frazer Institute, University of Queensland, Woolloongabba, Queensland, Australia
| | - Benedict Lum
- Frazer Institute, University of Queensland, Woolloongabba, Queensland, Australia
| | - Jason J Lee
- Frazer Institute, University of Queensland, Woolloongabba, Queensland, Australia
| | - Fiona Simpson
- Frazer Institute, University of Queensland, Woolloongabba, Queensland, Australia.
| |
Collapse
|
14
|
Müller GA, Müller TD. (Patho)Physiology of Glycosylphosphatidylinositol-Anchored Proteins I: Localization at Plasma Membranes and Extracellular Compartments. Biomolecules 2023; 13:biom13050855. [PMID: 37238725 DOI: 10.3390/biom13050855] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 05/11/2023] [Accepted: 05/13/2023] [Indexed: 05/28/2023] Open
Abstract
Glycosylphosphatidylinositol (GPI)-anchored proteins (APs) are anchored at the outer leaflet of plasma membranes (PMs) of all eukaryotic organisms studied so far by covalent linkage to a highly conserved glycolipid rather than a transmembrane domain. Since their first description, experimental data have been accumulating for the capability of GPI-APs to be released from PMs into the surrounding milieu. It became evident that this release results in distinct arrangements of GPI-APs which are compatible with the aqueous milieu upon loss of their GPI anchor by (proteolytic or lipolytic) cleavage or in the course of shielding of the full-length GPI anchor by incorporation into extracellular vesicles, lipoprotein-like particles and (lyso)phospholipid- and cholesterol-harboring micelle-like complexes or by association with GPI-binding proteins or/and other full-length GPI-APs. In mammalian organisms, the (patho)physiological roles of the released GPI-APs in the extracellular environment, such as blood and tissue cells, depend on the molecular mechanisms of their release as well as the cell types and tissues involved, and are controlled by their removal from circulation. This is accomplished by endocytic uptake by liver cells and/or degradation by GPI-specific phospholipase D in order to bypass potential unwanted effects of the released GPI-APs or their transfer from the releasing donor to acceptor cells (which will be reviewed in a forthcoming manuscript).
Collapse
Affiliation(s)
- Günter A Müller
- Institute for Diabetes and Obesity (IDO), Helmholtz Diabetes Center (HDC) at Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstraße 1, 85764 Oberschleissheim, Germany
- German Center for Diabetes Research (DZD), 85764 Oberschleissheim, Germany
| | - Timo D Müller
- Institute for Diabetes and Obesity (IDO), Helmholtz Diabetes Center (HDC) at Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstraße 1, 85764 Oberschleissheim, Germany
- German Center for Diabetes Research (DZD), 85764 Oberschleissheim, Germany
| |
Collapse
|
15
|
Hessien M, Donia T, Tabll AA, Adly E, Abdelhafez TH, Attia A, Alkafaas SS, Kuna L, Glasnovic M, Cosic V, Smolic R, Smolic M. Mechanistic-Based Classification of Endocytosis-Related Inhibitors: Does It Aid in Assigning Drugs against SARS-CoV-2? Viruses 2023; 15:1040. [PMID: 37243127 PMCID: PMC10222743 DOI: 10.3390/v15051040] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/13/2023] [Accepted: 04/19/2023] [Indexed: 05/28/2023] Open
Abstract
Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) canonically utilizes clathrin-mediated endocytosis (CME) and several other endocytic mechanisms to invade airway epithelial cells. Endocytic inhibitors, particularly those targeting CME-related proteins, have been identified as promising antiviral drugs. Currently, these inhibitors are ambiguously classified as chemical, pharmaceutical, or natural inhibitors. However, their varying mechanisms may suggest a more realistic classification system. Herein, we present a new mechanistic-based classification of endocytosis inhibitors, in which they are segregated among four distinct classes including: (i) inhibitors that disrupt endocytosis-related protein-protein interactions, and assembly or dissociation of complexes; (ii) inhibitors of large dynamin GTPase and/or kinase/phosphatase activities associated with endocytosis; (iii) inhibitors that modulate the structure of subcellular components, especially the plasma membrane, and actin; and (iv) inhibitors that cause physiological or metabolic alterations in the endocytosis niche. Excluding antiviral drugs designed to halt SARS-CoV-2 replication, other drugs, either FDA-approved or suggested through basic research, could be systematically assigned to one of these classes. We observed that many anti-SARS-CoV-2 drugs could be included either in class III or IV as they interfere with the structural or physiological integrity of subcellular components, respectively. This perspective may contribute to our understanding of the relative efficacy of endocytosis-related inhibitors and support the optimization of their individual or combined antiviral potential against SARS-CoV-2. However, their selectivity, combined effects, and possible interactions with non-endocytic cellular targets need more clarification.
Collapse
Affiliation(s)
- Mohamed Hessien
- Molecular Cell Biology Unit, Division of Biochemistry, Department of Chemistry, Faculty of Science, Tanta University, Tanta 31527, Egypt
| | - Thoria Donia
- Molecular Cell Biology Unit, Division of Biochemistry, Department of Chemistry, Faculty of Science, Tanta University, Tanta 31527, Egypt
| | - Ashraf A. Tabll
- National Research Centre, Microbial Biotechnology Department, Biotechnology Research Institute, Giza 12622, Egypt
- Egypt Center for Research and Regenerative Medicine (ECRRM), Cairo 11517, Egypt
| | - Eiman Adly
- Molecular Cell Biology Unit, Division of Biochemistry, Department of Chemistry, Faculty of Science, Tanta University, Tanta 31527, Egypt
| | - Tawfeek H. Abdelhafez
- National Research Centre, Microbial Biotechnology Department, Biotechnology Research Institute, Giza 12622, Egypt
| | - Amany Attia
- Molecular Cell Biology Unit, Division of Biochemistry, Department of Chemistry, Faculty of Science, Tanta University, Tanta 31527, Egypt
| | - Samar Sami Alkafaas
- Molecular Cell Biology Unit, Division of Biochemistry, Department of Chemistry, Faculty of Science, Tanta University, Tanta 31527, Egypt
| | - Lucija Kuna
- Department of Pharmacology and Biochemistry, Faculty of Dental Medicine and Health Osijek, University of J. J. Strossmayer Osijek, 31000 Osijek, Croatia
| | - Marija Glasnovic
- Department of Medicine, Family Medicine and History of Medicine, Faculty of Medicine Osijek, University of J. J. Strossmayer Osijek, 31000 Osijek, Croatia
| | - Vesna Cosic
- Department of Paediatrics and Gynaecology with Obstetrics, Faculty of Dental Medicine and Health Osijek, University of J. J. Strossmayer Osijek, 31000 Osijek, Croatia
| | - Robert Smolic
- Department of Pharmacology and Biochemistry, Faculty of Dental Medicine and Health Osijek, University of J. J. Strossmayer Osijek, 31000 Osijek, Croatia
| | - Martina Smolic
- Department of Pharmacology and Biochemistry, Faculty of Dental Medicine and Health Osijek, University of J. J. Strossmayer Osijek, 31000 Osijek, Croatia
- Department of Pharmacology, Faculty of Medicine Osijek, University of Osijek, 31000 Osijek, Croatia
| |
Collapse
|
16
|
Rosenkranz AA, Slastnikova TA. Prospects of Using Protein Engineering for Selective Drug Delivery into a Specific Compartment of Target Cells. Pharmaceutics 2023; 15:pharmaceutics15030987. [PMID: 36986848 PMCID: PMC10055131 DOI: 10.3390/pharmaceutics15030987] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 03/13/2023] [Accepted: 03/17/2023] [Indexed: 03/30/2023] Open
Abstract
A large number of proteins are successfully used to treat various diseases. These include natural polypeptide hormones, their synthetic analogues, antibodies, antibody mimetics, enzymes, and other drugs based on them. Many of them are demanded in clinical settings and commercially successful, mainly for cancer treatment. The targets for most of the aforementioned drugs are located at the cell surface. Meanwhile, the vast majority of therapeutic targets, which are usually regulatory macromolecules, are located inside the cell. Traditional low molecular weight drugs freely penetrate all cells, causing side effects in non-target cells. In addition, it is often difficult to elaborate a small molecule that can specifically affect protein interactions. Modern technologies make it possible to obtain proteins capable of interacting with almost any target. However, proteins, like other macromolecules, cannot, as a rule, freely penetrate into the desired cellular compartment. Recent studies allow us to design multifunctional proteins that solve these problems. This review considers the scope of application of such artificial constructs for the targeted delivery of both protein-based and traditional low molecular weight drugs, the obstacles met on the way of their transport to the specified intracellular compartment of the target cells after their systemic bloodstream administration, and the means to overcome those difficulties.
Collapse
Affiliation(s)
- Andrey A Rosenkranz
- Laboratory of Molecular Genetics of Intracellular Transport, Institute of Gene Biology of Russian Academy of Sciences, 34/5 Vavilov St., 119334 Moscow, Russia
- Department of Biophysics, Faculty of Biology, Lomonosov Moscow State University, 1-12 Leninskie Gory St., 119234 Moscow, Russia
| | - Tatiana A Slastnikova
- Laboratory of Molecular Genetics of Intracellular Transport, Institute of Gene Biology of Russian Academy of Sciences, 34/5 Vavilov St., 119334 Moscow, Russia
| |
Collapse
|
17
|
Placidi G, Mattu C, Ciardelli G, Campa CC. Small molecules targeting endocytic uptake and recycling pathways. Front Cell Dev Biol 2023; 11:1125801. [PMID: 36968200 PMCID: PMC10036367 DOI: 10.3389/fcell.2023.1125801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 02/23/2023] [Indexed: 03/12/2023] Open
Abstract
Over the past years a growing number of studies highlighted the pivotal role of intracellular trafficking in cell physiology. Among the distinct transport itineraries connecting the endocytic system, both internalization (endocytosis) and recycling (endocytic recycling) pathways were found fundamental to ensure cellular sensing, cell-to-cell communication, cellular division, and collective cell migration in tissue specific-contexts. Consistently, the dysregulation of endocytic trafficking pathways is correlated with several human diseases including both cancers and neurodegeneration. Aimed at suppress specific intracellular trafficking routes involved in disease onset and progression, huge efforts have been made to identify small molecule inhibitors with suitable pharmacological properties for in vivo administration. Here, we review most used drugs and recently discovered small molecules able to block endocytosis and endocytic recycling pathways. We characterize such pharmacological inhibitors by emphasizing their target specificity, molecular affinity, biological activity and efficacy in both in vitro and in vivo experimental models.
Collapse
Affiliation(s)
- Giampaolo Placidi
- Italian Institute for Genomic Medicine, Candiolo, Italy
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy
| | - Clara Mattu
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy
| | - Gianluca Ciardelli
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy
- Chemical-Physical Processes, National Research Council (CNR-IPCF), Pisa, Italy
| | - Carlo C. Campa
- Italian Institute for Genomic Medicine, Candiolo, Italy
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| |
Collapse
|
18
|
Shikanai M, Ito S, Nishimura YV, Akagawa R, Fukuda M, Yuzaki M, Nabeshima Y, Kawauchi T. Rab21 regulates caveolin-1-mediated endocytic trafficking to promote immature neurite pruning. EMBO Rep 2023; 24:e54701. [PMID: 36683567 PMCID: PMC9986827 DOI: 10.15252/embr.202254701] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 12/09/2022] [Accepted: 12/21/2022] [Indexed: 01/24/2023] Open
Abstract
Transmembrane proteins are internalized by clathrin- and caveolin-dependent endocytosis. Both pathways converge on early endosomes and are thought to share the small GTPase Rab5 as common regulator. In contrast to this notion, we show here that the clathrin- and caveolin-mediated endocytic pathways are differentially regulated. Rab5 and Rab21 localize to distinct populations of early endosomes in cortical neurons and preferentially regulate clathrin- and caveolin-mediated pathways, respectively, suggesting heterogeneity in the early endosomes, rather than a converging point. Suppression of Rab21, but not Rab5, results in decreased plasma membrane localization and total protein levels of caveolin-1, which perturbs immature neurite pruning of cortical neurons, an in vivo-specific step of neuronal maturation. Taken together, our data indicate that clathrin- and caveolin-mediated endocytic pathways run in parallel in early endosomes, which show different molecular regulation and physiological function.
Collapse
Affiliation(s)
- Mima Shikanai
- Department of PhysiologyKeio University School of MedicineTokyoJapan
| | - Shiho Ito
- Department of Aging Science and Medicine, Graduate School of MedicineKyoto UniversityKyotoJapan
- Laboratory of Molecular Life ScienceInstitute of Biomedical Research and Innovation, FBRI, CLIK‐5FKobeJapan
| | - Yoshiaki V Nishimura
- Division of Neuroscience, Faculty of MedicineTohoku Medical and Pharmaceutical UniversitySendaiJapan
| | - Remi Akagawa
- Laboratory of Molecular Life ScienceInstitute of Biomedical Research and Innovation, FBRI, CLIK‐5FKobeJapan
| | - Mitsunori Fukuda
- Laboratory of Membrane Trafficking Mechanisms, Department of Integrative Life Sciences, Graduate School of Life SciencesTohoku UniversitySendaiJapan
| | - Michisuke Yuzaki
- Department of PhysiologyKeio University School of MedicineTokyoJapan
| | - Yo‐ichi Nabeshima
- Department of Aging Science and Medicine, Graduate School of MedicineKyoto UniversityKyotoJapan
- Laboratory of Molecular Life ScienceInstitute of Biomedical Research and Innovation, FBRI, CLIK‐5FKobeJapan
| | - Takeshi Kawauchi
- Department of PhysiologyKeio University School of MedicineTokyoJapan
- Department of Aging Science and Medicine, Graduate School of MedicineKyoto UniversityKyotoJapan
- Laboratory of Molecular Life ScienceInstitute of Biomedical Research and Innovation, FBRI, CLIK‐5FKobeJapan
| |
Collapse
|
19
|
Transfer of Proteins from Cultured Human Adipose to Blood Cells and Induction of Anabolic Phenotype Are Controlled by Serum, Insulin and Sulfonylurea Drugs. Int J Mol Sci 2023; 24:ijms24054825. [PMID: 36902257 PMCID: PMC10003403 DOI: 10.3390/ijms24054825] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 02/10/2023] [Accepted: 02/27/2023] [Indexed: 03/06/2023] Open
Abstract
Glycosylphosphatidylinositol-anchored proteins (GPI-APs) are anchored at the outer leaflet of eukaryotic plasma membranes (PMs) only by carboxy-terminal covalently coupled GPI. GPI-APs are known to be released from the surface of donor cells in response to insulin and antidiabetic sulfonylureas (SUs) by lipolytic cleavage of the GPI or upon metabolic derangement as full-length GPI-APs with the complete GPI attached. Full-length GPI-APs become removed from extracellular compartments by binding to serum proteins, such as GPI-specific phospholipase D (GPLD1), or insertion into the PMs of acceptor cells. Here, the interplay between the lipolytic release and intercellular transfer of GPI-APs and its potential functional impact was studied using transwell co-culture with human adipocytes as insulin-/SU-responsive donor cells and GPI-deficient erythroleukemia as acceptor cells (ELCs). Measurement of the transfer as the expression of full-length GPI-APs at the ELC PMs by their microfluidic chip-based sensing with GPI-binding α-toxin and GPI-APs antibodies and of the ELC anabolic state as glycogen synthesis upon incubation with insulin, SUs and serum yielded the following results: (i) Loss of GPI-APs from the PM upon termination of their transfer and decline of glycogen synthesis in ELCs, as well as prolongation of the PM expression of transferred GPI-APs upon inhibition of their endocytosis and upregulated glycogen synthesis follow similar time courses. (ii) Insulin and SUs inhibit both GPI-AP transfer and glycogen synthesis upregulation in a concentration-dependent fashion, with the efficacies of the SUs increasing with their blood glucose-lowering activity. (iii) Serum from rats eliminates insulin- and SU-inhibition of both GPI-APs' transfer and glycogen synthesis in a volume-dependent fashion, with the potency increasing with their metabolic derangement. (iv) In rat serum, full-length GPI-APs bind to proteins, among them (inhibited) GPLD1, with the efficacy increasing with the metabolic derangement. (v) GPI-APs are displaced from serum proteins by synthetic phosphoinositolglycans and then transferred to ELCs with accompanying stimulation of glycogen synthesis, each with efficacies increasing with their structural similarity to the GPI glycan core. Thus, both insulin and SUs either block or foster transfer when serum proteins are depleted of or loaded with full-length GPI-APs, respectively, i.e., in the normal or metabolically deranged state. The transfer of the anabolic state from somatic to blood cells over long distance and its "indirect" complex control by insulin, SUs and serum proteins support the (patho)physiological relevance of the intercellular transfer of GPI-APs.
Collapse
|
20
|
Larsson E, Morén B, McMahon KA, Parton RG, Lundmark R. Dynamin2 functions as an accessory protein to reduce the rate of caveola internalization. J Cell Biol 2023; 222:213853. [PMID: 36729022 PMCID: PMC9929934 DOI: 10.1083/jcb.202205122] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 11/14/2022] [Accepted: 01/10/2023] [Indexed: 02/03/2023] Open
Abstract
Caveolae are small membrane invaginations that generally are stably attached to the plasma membrane. Their release is believed to depend on the GTPase dynamin 2 (Dyn2), in analogy with its role in fission of clathrin-coated vesicles. The mechanistic understanding of caveola fission is, however, sparse. Here, we used microscopy-based tracking of individual caveolae in living cells to determine the role of Dyn2 in caveola dynamics. We report that Dyn2 stably associated with the bulb of a subset of caveolae, but was not required for formation or fission of caveolae. Dyn2-positive caveolae displayed longer plasma membrane duration times, whereas depletion of Dyn2 resulted in shorter duration times and increased caveola fission. The stabilizing role of Dyn2 was independent of its GTPase activity and the caveola stabilizing protein EHD2. Thus, we propose that, in contrast to the current view, Dyn2 is not a core component of the caveolae machinery, but rather functions as an accessory protein that restrains caveola internalization.
Collapse
Affiliation(s)
- Elin Larsson
- https://ror.org/05kb8h459Integrative Medical Biology, Umeå University, Umeå, Sweden,Umeå Centre for Microbial Research, Umeå University, Umeå, Sweden
| | - Björn Morén
- https://ror.org/05kb8h459Integrative Medical Biology, Umeå University, Umeå, Sweden,Umeå Centre for Microbial Research, Umeå University, Umeå, Sweden
| | - Kerrie-Ann McMahon
- https://ror.org/00rqy9422Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Robert G. Parton
- https://ror.org/00rqy9422Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia,Centre for Microscopy and Microanalysis, The University of Queensland, Brisbane, Queensland, Australia
| | - Richard Lundmark
- https://ror.org/05kb8h459Integrative Medical Biology, Umeå University, Umeå, Sweden,Umeå Centre for Microbial Research, Umeå University, Umeå, Sweden,Molecular Infection Medicine Sweden, Umeå University, Umeå, Sweden,Correspondence to Richard Lundmark:
| |
Collapse
|
21
|
Alkafaas SS, Abdallah AM, Ghosh S, Loutfy SA, Elkafas SS, Abdel Fattah NF, Hessien M. Insight into the role of clathrin-mediated endocytosis inhibitors in SARS-CoV-2 infection. Rev Med Virol 2023; 33:e2403. [PMID: 36345157 PMCID: PMC9877911 DOI: 10.1002/rmv.2403] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 10/08/2022] [Accepted: 10/10/2022] [Indexed: 11/10/2022]
Abstract
Emergence of SARS-CoV-2 variants warrants sustainable efforts to upgrade both the diagnostic and therapeutic protocols. Understanding the details of cellular and molecular basis of the virus-host cell interaction is essential for developing variant-independent therapeutic options. The internalization of SARS-CoV-2, into lung epithelial cells, is mediated by endocytosis, especially clathrin-mediated endocytosis (CME). Although vaccination is the gold standard strategy against viral infection, selective inhibition of endocytic proteins, complexes, and associated adaptor proteins may present a variant-independent therapeutic strategy. Although clathrin and/or dynamins are the most important proteins involved in CME, other endocytic mechanisms are clathrin and/or dynamin independent and rely on other proteins. Moreover, endocytosis implicates some subcellular structures, like plasma membrane, actin and lysosomes. Also, physiological conditions, such as pH and ion concentrations, represent an additional factor that mediates these events. Accordingly, endocytosis related proteins are potential targets for small molecules that inhibit endocytosis-mediated viral entry. This review summarizes the potential of using small molecules, targeting key proteins, participating in clathrin-dependent and -independent endocytosis, as variant-independent antiviral drugs against SARS-CoV-2 infection. The review takes two approaches. The first outlines the potential role of endocytic inhibitors in preventing endocytosis-mediated viral entry and its mechanism of action, whereas in the second computational analysis was implemented to investigate the selectivity of common inhibitors against endocytic proteins in SARS-CoV-2 endocytosis. The analysis revealed that remdesivir, methyl-β-cyclodextrin, rottlerin, and Bis-T can effectively inhibit clathrin, HMG-CoA reductase, actin, and dynamin I GTPase and are more potent in inhibiting SARS-CoV-2 than chloroquine. CME inhibitors for SARS-CoV-2 infection remain understudied.
Collapse
Affiliation(s)
- Samar Sami Alkafaas
- Molecular Cell Biology UniteDivision of BiochemistryDepartment of ChemistryFaculty of ScienceTanta UniversityTantaEgypt
| | - Abanoub Mosaad Abdallah
- Narcotic Research DepartmentNational Center for Social and Criminological Research (NCSCR)GizaEgypt
| | - Soumya Ghosh
- Department of GeneticsFaculty of Natural and Agricultural SciencesUniversity of the Free StateBloemfonteinSouth Africa
| | - Samah A. Loutfy
- Virology and Immunology UnitCancer Biology DepartmentNational Cancer Institute (NCI)Cairo UniversityCairoEgypt
- Nanotechnology Research CenterBritish UniversityCairoEgypt
| | - Sara Samy Elkafas
- Production Engineering and Mechanical Design DepartmentFaculty of EngineeringMenofia UniversityMenofiaEgypt
| | - Nasra F. Abdel Fattah
- Virology and Immunology UnitCancer Biology DepartmentNational Cancer Institute (NCI)Cairo UniversityCairoEgypt
| | - Mohamed Hessien
- Molecular Cell Biology UniteDivision of BiochemistryDepartment of ChemistryFaculty of ScienceTanta UniversityTantaEgypt
| |
Collapse
|
22
|
Enyong EN, Gurley JM, De Ieso ML, Stamer WD, Elliott MH. Caveolar and non-Caveolar Caveolin-1 in ocular homeostasis and disease. Prog Retin Eye Res 2022; 91:101094. [PMID: 35729002 PMCID: PMC9669151 DOI: 10.1016/j.preteyeres.2022.101094] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 06/03/2022] [Accepted: 06/10/2022] [Indexed: 11/17/2022]
Abstract
Caveolae, specialized plasma membrane invaginations present in most cell types, play important roles in multiple cellular processes including cell signaling, lipid uptake and metabolism, endocytosis and mechanotransduction. They are found in almost all cell types but most abundant in endothelial cells, adipocytes and fibroblasts. Caveolin-1 (Cav1), the signature structural protein of caveolae was the first protein associated with caveolae, and in association with Cavin1/PTRF is required for caveolae formation. Genetic ablation of either Cav1 or Cavin1/PTRF downregulates expression of the other resulting in loss of caveolae. Studies using Cav1-deficient mouse models have implicated caveolae with human diseases such as cardiomyopathies, lipodystrophies, diabetes and muscular dystrophies. While caveolins and caveolae are extensively studied in extra-ocular settings, their contributions to ocular function and disease pathogenesis are just beginning to be appreciated. Several putative caveolin/caveolae functions are relevant to the eye and Cav1 is highly expressed in retinal vascular and choroidal endothelium, Müller glia, the retinal pigment epithelium (RPE), and the Schlemm's canal endothelium and trabecular meshwork cells. Variants at the CAV1/2 gene locus are associated with risk of primary open angle glaucoma and the high risk HTRA1 variant for age-related macular degeneration is thought to exert its effect through regulation of Cav1 expression. Caveolins also play important roles in modulating retinal neuroinflammation and blood retinal barrier permeability. In this article, we describe the current state of caveolin/caveolae research in the context of ocular function and pathophysiology. Finally, we discuss new evidence showing that retinal Cav1 exists and functions outside caveolae.
Collapse
Affiliation(s)
- Eric N Enyong
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Ophthalmology, Dean A. McGee Eye Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Jami M Gurley
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Ophthalmology, Dean A. McGee Eye Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Michael L De Ieso
- Department of Ophthalmology, Duke Eye Center, Duke University, Durham, NC, USA
| | - W Daniel Stamer
- Department of Ophthalmology, Duke Eye Center, Duke University, Durham, NC, USA
| | - Michael H Elliott
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Ophthalmology, Dean A. McGee Eye Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| |
Collapse
|
23
|
Lolo FN, Pavón DM, Grande-García A, Elosegui-Artola A, Segatori VI, Sánchez S, Trepat X, Roca-Cusachs P, del Pozo MA. Caveolae couple mechanical stress to integrin recycling and activation. eLife 2022; 11:e82348. [PMID: 36264062 PMCID: PMC9747151 DOI: 10.7554/elife.82348] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 10/19/2022] [Indexed: 12/15/2022] Open
Abstract
Cells are subjected to multiple mechanical inputs throughout their lives. Their ability to detect these environmental cues is called mechanosensing, a process in which integrins play an important role. During cellular mechanosensing, plasma membrane (PM) tension is adjusted to mechanical stress through the buffering action of caveolae; however, little is known about the role of caveolae in early integrin mechanosensing regulation. Here, we show that Cav1KO fibroblasts increase adhesion to FN-coated beads when pulled with magnetic tweezers, as compared to wild type fibroblasts. This phenotype is Rho-independent and mainly derived from increased active β1-integrin content on the surface of Cav1KO fibroblasts. Florescence recovery after photobleaching analysis and endocytosis/recycling assays revealed that active β1-integrin is mostly endocytosed through the clathrin independent carrier/glycosylphosphatidyl inositol (GPI)-enriched endocytic compartment pathway and is more rapidly recycled to the PM in Cav1KO fibroblasts, in a Rab4 and PM tension-dependent manner. Moreover, the threshold for PM tension-driven β1-integrin activation is lower in Cav1KO mouse embryonic fibroblasts (MEFs) than in wild type MEFs, through a mechanism dependent on talin activity. Our findings suggest that caveolae couple mechanical stress to integrin cycling and activation, thereby regulating the early steps of the cellular mechanosensing response.
Collapse
Affiliation(s)
- Fidel-Nicolás Lolo
- Mechanoadaptation and Caveolae Biology Laboratory, Cell and developmental Biology Area, Centro Nacional de Investigaciones CardiovascularesMadridSpain
| | - Dácil María Pavón
- Mechanoadaptation and Caveolae Biology Laboratory, Cell and developmental Biology Area, Centro Nacional de Investigaciones CardiovascularesMadridSpain
| | - Araceli Grande-García
- Mechanoadaptation and Caveolae Biology Laboratory, Cell and developmental Biology Area, Centro Nacional de Investigaciones CardiovascularesMadridSpain
| | | | - Valeria Inés Segatori
- Mechanoadaptation and Caveolae Biology Laboratory, Cell and developmental Biology Area, Centro Nacional de Investigaciones CardiovascularesMadridSpain
| | - Sara Sánchez
- Mechanoadaptation and Caveolae Biology Laboratory, Cell and developmental Biology Area, Centro Nacional de Investigaciones CardiovascularesMadridSpain
| | - Xavier Trepat
- Institute for Bioengineering of CataloniaBarcelonaSpain
| | | | - Miguel A del Pozo
- Mechanoadaptation and Caveolae Biology Laboratory, Cell and developmental Biology Area, Centro Nacional de Investigaciones CardiovascularesMadridSpain
| |
Collapse
|
24
|
Kim KS, Jeon MT, Kim ES, Lee CH, Kim DG. Activation of NMDA receptors in brain endothelial cells increases transcellular permeability. Fluids Barriers CNS 2022; 19:70. [PMID: 36068542 PMCID: PMC9450318 DOI: 10.1186/s12987-022-00364-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 08/15/2022] [Indexed: 12/04/2022] Open
Abstract
Neurovascular coupling is a precise mechanism that induces increased blood flow to activated brain regions, thereby providing oxygen and glucose. In this study, we hypothesized that N-methyl-D-aspartate (NMDA) receptor signaling, the most well characterized neurotransmitter signaling system which regulates delivery of essential molecules through the blood–brain barrier (BBB). Upon application of NMDA in both in vitro and in vivo models, increased delivery of bioactive molecules that was mediated through modulation of molecules involved in molecular delivery, including clathrin and caveolin were observed. Also, NMDA activation induced structural changes in the BBB and increased transcellular permeability that showed regional heterogeneity in its responses. Moreover, NMDA receptor activation increased endosomal trafficking and facilitated inactivation of lysosomal pathways and consequently increased molecular delivery mediated by activation of calmodulin-dependent protein kinase II (CaMKII) and RhoA/protein kinase C (PKC). Subsequent in vivo experiments using mice specifically lacking NMDA receptor subunit 1 in endothelial cells showed decreased neuronal density in the brain cortex, suggesting that a deficiency in NMDA receptor signaling in brain endothelial cells induces neuronal losses. Together, these results highlight the importance of NMDA-receptor-mediated signaling in the regulation of BBB permeability that surprisingly also affected CD31 staining.
Collapse
Affiliation(s)
- Kyu-Sung Kim
- Neuroimmunology Lab, Dementia Research Group, Korea Brain Research Institute, Daegu, 41062, South Korea.,Department of Brain Science, Daegu Gyeongbuk Institute of Science & Technology (DGIST), Daegu, South Korea
| | - Min Tae Jeon
- Neuroimmunology Lab, Dementia Research Group, Korea Brain Research Institute, Daegu, 41062, South Korea
| | - Eun Seon Kim
- Neuroimmunology Lab, Dementia Research Group, Korea Brain Research Institute, Daegu, 41062, South Korea.,Department of Brain Science, Daegu Gyeongbuk Institute of Science & Technology (DGIST), Daegu, South Korea
| | - Chan Hee Lee
- Neuroimmunology Lab, Dementia Research Group, Korea Brain Research Institute, Daegu, 41062, South Korea
| | - Do-Geun Kim
- Neuroimmunology Lab, Dementia Research Group, Korea Brain Research Institute, Daegu, 41062, South Korea.
| |
Collapse
|
25
|
Griffiths G, Gruenberg J, Marsh M, Wohlmann J, Jones AT, Parton RG. Nanoparticle entry into cells; the cell biology weak link. Adv Drug Deliv Rev 2022; 188:114403. [PMID: 35777667 DOI: 10.1016/j.addr.2022.114403] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 06/22/2022] [Accepted: 06/23/2022] [Indexed: 12/22/2022]
Abstract
Nanoparticles (NP) are attractive options for the therapeutic delivery of active pharmaceutical drugs, proteins and nucleic acids into cells, tissues and organs. Research into the development and application of NP most often starts with a diverse group of scientists, including chemists, bioengineers and material and pharmaceutical scientists, who design, fabricate and characterize NP in vitro (Stage 1). The next step (Stage 2) generally investigates cell toxicity as well as the processes by which NP bind, are internalized and deliver their cargo to appropriate model tissue culture cells. Subsequently, in Stage 3, selected NP are tested in animal systems, mostly mouse. Whereas the chemistry-based development and analysis in Stage 1 is increasingly sophisticated, the investigations in Stage 2 are not what could be regarded as 'state-of-the-art' for the cell biology field and the quality of research into NP interactions with cells is often sub-standard. In this review we describe our current understanding of the mechanisms by which particles gain entry into mammalian cells via endocytosis. We summarize the most important areas for concern, highlight some of the most common mis-conceptions, and identify areas where NP scientists could engage with trained cell biologists. Our survey of the different mechanisms of uptake into cells makes us suspect that claims for roles for caveolae, as well as macropinocytosis, in NP uptake into cells have been exaggerated, whereas phagocytosis has been under-appreciated.
Collapse
Affiliation(s)
- Gareth Griffiths
- Department Biosciences, University of Oslo, Blindernveien 31, PO Box 1041, 0316 Oslo, Norway.
| | - Jean Gruenberg
- Department of Biochemistry, University of Geneva, 30 quai E. Ansermet, 1211-Geneva-4, Switzerland
| | - Mark Marsh
- Laboratory for Molecular Cell Biology, University College London, Gower Street, London, WC1E 6BT, UK
| | - Jens Wohlmann
- Department Biosciences, University of Oslo, Blindernveien 31, PO Box 1041, 0316 Oslo, Norway
| | - Arwyn T Jones
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Redwood Building, Cardiff, Wales CF103NB, UK
| | - Robert G Parton
- Institute for Molecular Bioscience and Centre for Microscopy and Microanalysis, The University of Queensland, Qld 4072, Australia
| |
Collapse
|
26
|
Mishra LC, Pandey U, Gupta A, Gupta J, Sharma M, Mishra G. Alternating exosomes and their mimetics as an emergent strategy for targeted cancer therapy. Front Mol Biosci 2022; 9:939050. [PMID: 36032679 PMCID: PMC9399404 DOI: 10.3389/fmolb.2022.939050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 07/15/2022] [Indexed: 11/18/2022] Open
Abstract
Exosomes, a subtype of the class of extracellular vesicles and nano-sized particles, have a specific membrane structure that makes them an alternative proposition to combat with cancer through slight modification. As constituents of all most all the primary body fluids, exosomes establish the status of intercellular communication. Exosomes have specific proteins/mRNAs and miRNAs which serve as biomarkers, imparting a prognostic tool in clinical and disease pathologies. They have efficient intrinsic targeting potential and efficacy. Engineered exosomes are employed to deliver therapeutic cargos to the targeted tumor cell or the recipient. Exosomes from cancer cells bring about changes in fibroblast via TGFβ/Smad pathway, augmenting the tumor growth. These extracellular vesicles are multidimensional in terms of the functions that they perform. We herein discuss the uptake and biogenesis of exosomes, their role in various facets of cancer studies, cell-to-cell communication and modification for therapeutic and diagnostic use.
Collapse
Affiliation(s)
| | - Utkarsh Pandey
- Department of Zoology, Swami Shraddhanand College, University of Delhi, New Delhi, India
| | - Abhikarsh Gupta
- Department of Microbiology, Swami Shraddhanand College, University of Delhi, New Delhi, India
| | - Jyotsna Gupta
- Department of Microbiology, Swami Shraddhanand College, University of Delhi, New Delhi, India
| | - Monal Sharma
- Betterhumans Inc., Gainesville, FL, United States
| | - Gauri Mishra
- Department of Zoology, Swami Shraddhanand College, University of Delhi, New Delhi, India
- Division Radiopharmaceuticals and Radiation Biology, Institute of Nuclear Medicine and Allied Sciences, New Delhi, India
| |
Collapse
|
27
|
Zhu D, Zhang Z, Zhao J, Liu D, Gan L, Lau WB, Xie D, Meng Z, Yao P, Tsukuda J, Christopher TA, Lopez BL, Gao E, Koch WJ, Wang Y, Ma XL. Targeting Adiponectin Receptor 1 Phosphorylation Against Ischemic Heart Failure. Circ Res 2022; 131:e34-e50. [PMID: 35611695 PMCID: PMC9308652 DOI: 10.1161/circresaha.121.319976] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
BACKGROUND Despite significantly reduced acute myocardial infarction (MI) mortality in recent years, ischemic heart failure continues to escalate. Therapeutic interventions effectively reversing pathological remodeling are an urgent unmet medical need. We recently demonstrated that AdipoR1 (APN [adiponectin] receptor 1) phosphorylation by GRK2 (G-protein-coupled receptor kinase 2) contributes to maladaptive remodeling in the ischemic heart. The current study clarified the underlying mechanisms leading to AdipoR1 phosphorylative desensitization and investigated whether blocking AdipoR1 phosphorylation may restore its protective signaling, reversing post-MI remodeling. METHODS Specific sites and underlying molecular mechanisms responsible for AdipoR1 phosphorylative desensitization were investigated in vitro (neonatal and adult cardiomyocytes). The effects of AdipoR1 phosphorylation inhibition upon APN post-MI remodeling and heart failure progression were investigated in vivo. RESULTS Among 4 previously identified sites sensitive to GRK2 phosphorylation, alanine substitution of Ser205 (AdipoR1S205A), but not other 3 sites, rescued GRK2-suppressed AdipoR1 functions, restoring APN-induced cell salvage kinase activation and reducing oxidative cell death. The molecular investigation followed by functional determination demonstrated that AdipoR1 phosphorylation promoted clathrin-dependent (not caveolae) endocytosis and lysosomal-mediated (not proteasome) degradation, reducing AdipoR1 protein level and suppressing AdipoR1-mediated cytoprotective action. GRK2-induced AdipoR1 endocytosis and degradation were blocked by AdipoR1S205A overexpression. Moreover, AdipoR1S205E (pseudophosphorylation) phenocopied GRK2 effects, promoted AdipoR1 endocytosis and degradation, and inhibited AdipoR1 biological function. Most importantly, AdipoR1 function was preserved during heart failure development in AdipoR1-KO (AdipoR1 knockout) mice reexpressing hAdipoR1S205A. APN administration in the failing heart reversed post-MI remodeling and improved cardiac function. However, reexpressing hAdipoR1WT in AdipoR1-KO mice failed to restore APN cardioprotection. CONCLUSIONS Ser205 is responsible for AdipoR1 phosphorylative desensitization in the failing heart. Blockade of AdipoR1 phosphorylation followed by pharmacological APN administration is a novel therapy effective in reversing post-MI remodeling and mitigating heart failure progression.
Collapse
Affiliation(s)
- Di Zhu
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19107
| | - Zhen Zhang
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19107
| | - Jianli Zhao
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19107
| | - Demin Liu
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19107
| | - Lu Gan
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19107
| | - Wayne Bond Lau
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19107
| | - Dina Xie
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19107
| | - Zhijun Meng
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19107
| | - Peng Yao
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19107
| | - Jumpei Tsukuda
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19107
| | | | - Bernard L. Lopez
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19107
| | - Erhe Gao
- Department of Cardiovascular Sciences, Center for Translational Medicine, Temple University, Philadelphia, PA 19104
| | - Walter J. Koch
- Department of Cardiovascular Sciences, Center for Translational Medicine, Temple University, Philadelphia, PA 19104
| | - Yajing Wang
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19107
- Corresponding Authors: Xinliang (Xin) Ma, M.D., Ph.D, Department of Medicine and, Department of Emergency Medicine, 1025 Walnut Street, College Building 300, Thomas Jefferson University, Philadelphia, PA 19107, Tel: 215-955-4994, Or Yajing Wang, MD,PhD, Department of Emergency Medicine, 1025 Walnut Street, College Building 325, Thomas Jefferson University, Philadelphia, PA 19107, Tel: 215-955-8895,
| | - Xin-Liang Ma
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19107
- Corresponding Authors: Xinliang (Xin) Ma, M.D., Ph.D, Department of Medicine and, Department of Emergency Medicine, 1025 Walnut Street, College Building 300, Thomas Jefferson University, Philadelphia, PA 19107, Tel: 215-955-4994, Or Yajing Wang, MD,PhD, Department of Emergency Medicine, 1025 Walnut Street, College Building 325, Thomas Jefferson University, Philadelphia, PA 19107, Tel: 215-955-8895,
| |
Collapse
|
28
|
Szychowski KA, Skóra B, Pomianek T. Effect of the elastin-derived peptides (VGVAPG and VVGPGA) on breast (MCF-7) and lung (A549) cancer cell lines in vitro. Biomed Pharmacother 2022; 151:113149. [PMID: 35598370 DOI: 10.1016/j.biopha.2022.113149] [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: 03/17/2022] [Revised: 05/11/2022] [Accepted: 05/15/2022] [Indexed: 11/29/2022] Open
Abstract
Tissues are subjected to dynamic communication between cells and the extracellular matrix (ECM), resulting in ECM remodeling. One of the ECM components is elastin, which releases elastin-derived peptides (EDPs) during the aging process. Therefore, the aim of the present study was to evaluate the impact of the VGVAPG hexapeptide and elastin-like peptide VVGPGA (control) on certain metabolism parameters in human breast adenocarcinoma (MCF-7) and human lung carcinoma (A549) cell lines. The results did not show a significant effect of the peptides on metabolic activity and caspase-3 activity. However, more specific analysis revealed that VGVAPG and VVGPGA were able to increase KI67 protein expression in both tested cell lines after 24-h treatment. Moreover, the same correlation was observed at the KI67 gene level. VGVAPG also increased the P53, ATM and SHH gene expression in the A549 cells up to 19.08%, 20.74%, and 28.77%, respectively. Interestingly, the VGVAPG peptide exerted an effect on the expression of antioxidant enzymes SOD2 and CAT in the A549 and MCF-7 cells, especially after the 24-h treatment. Lastly, both peptides influenced the CAV1 and CLTC1 expression. Our results show that the tested EDPs have an effect on both A549 and MCF-7 cells at the cellular level. This may be correlated with the multidrug-resistance (MDR) phenotype in these cancer cells, which is an emerging problem in the current anticancer treatment. However, more research is needed in this field.
Collapse
Affiliation(s)
- Konrad A Szychowski
- Department of Biotechnology and Cell Biology, Medical College, University of Information Technology and Management in Rzeszow, Sucharskiego 2, 35-225 Rzeszow, Poland.
| | - Bartosz Skóra
- Department of Biotechnology and Cell Biology, Medical College, University of Information Technology and Management in Rzeszow, Sucharskiego 2, 35-225 Rzeszow, Poland
| | - Tadeusz Pomianek
- Department of Management, Management College, University of Information Technology and Management in Rzeszow, Sucharskiego 2, Rzeszow 35-225, Poland
| |
Collapse
|
29
|
Roncato R, Angelini J, Pani A, Talotta R. Lipid rafts as viral entry routes and immune platforms: A double-edged sword in SARS-CoV-2 infection? Biochim Biophys Acta Mol Cell Biol Lipids 2022; 1867:159140. [PMID: 35248801 PMCID: PMC8894694 DOI: 10.1016/j.bbalip.2022.159140] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 02/13/2022] [Accepted: 02/25/2022] [Indexed: 12/15/2022]
Abstract
Lipid rafts are nanoscopic compartments of cell membranes that serve a variety of biological functions. They play a crucial role in viral infections, as enveloped viruses such as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) can exploit rafts to enter or quit target cells. On the other hand, lipid rafts contribute to the formation of immune synapses and their proper functioning is a prerequisite for adequate immune response and viral clearance. In this narrative review we dissect the panorama focusing on this singular aspect of cell biology in the context of SARS-CoV-2 infection and therapy. A lipid raft-mediated mechanism can be hypothesized for many drugs recommended or considered for the treatment of SARS-CoV-2 infection, such as glucocorticoids, antimalarials, immunosuppressants and antiviral agents. Furthermore, the additional use of lipid-lowering agents, like statins, may affect the lipid composition of membrane rafts and thus influence the processes occurring in these compartments. The combination of drugs acting on lipid rafts may be successful in the treatment of more severe forms of the disease and should be reserved for further investigation.
Collapse
Affiliation(s)
- Rossana Roncato
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO), Istituto di Ricovero e Cura a carattere Scientifico (IRCCS), via Gallini, 33081 Aviano (PN), Italy
| | - Jacopo Angelini
- Clinical Pharmacology Institute, Azienda Sanitaria Universitaria Friuli Centrale (ASU FC), via Pozzuolo, 33100 Udine, Italy
| | - Arianna Pani
- Toxicology Department of Oncology and Hemato-Oncology, University of Milan, via Vanvitelli, 20133 Milan, Italy
| | - Rossella Talotta
- Department of Clinical and Experimental Medicine, Rheumatology Unit, AOU "Gaetano Martino", University of Messina, 98100 Messina, Italy
| |
Collapse
|
30
|
Szabó I, Yousef M, Soltész D, Bató C, Mező G, Bánóczi Z. Redesigning of Cell-Penetrating Peptides to Improve Their Efficacy as a Drug Delivery System. Pharmaceutics 2022; 14:pharmaceutics14050907. [PMID: 35631493 PMCID: PMC9146218 DOI: 10.3390/pharmaceutics14050907] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 04/11/2022] [Accepted: 04/13/2022] [Indexed: 12/29/2022] Open
Abstract
Cell-penetrating peptides (CPP) are promising tools for the transport of a broad range of compounds into cells. Since the discovery of the first members of this peptide family, many other peptides have been identified; nowadays, dozens of these peptides are known. These peptides sometimes have very different chemical–physical properties, but they have similar drawbacks; e.g., non-specific internalization, fast elimination from the body, intracellular/vesicular entrapment. Although our knowledge regarding the mechanism and structure–activity relationship of internalization is growing, the prediction and design of the cell-penetrating properties are challenging. In this review, we focus on the different modifications of well-known CPPs to avoid their drawbacks, as well as how these modifications may increase their internalization and/or change the mechanism of penetration.
Collapse
Affiliation(s)
- Ildikó Szabó
- MTA-ELTE Research Group of Peptide Chemistry, Eötvös Loránd Research Network (ELKH), Eötvös Loránd University, 1117 Budapest, Hungary;
- Correspondence: (I.S.); (Z.B.)
| | - Mo’ath Yousef
- Department of Organic Chemistry, Institute of Chemistry, Eötvös Loránd University, 1117 Budapest, Hungary; (M.Y.); (D.S.); (C.B.)
| | - Dóra Soltész
- Department of Organic Chemistry, Institute of Chemistry, Eötvös Loránd University, 1117 Budapest, Hungary; (M.Y.); (D.S.); (C.B.)
| | - Csaba Bató
- Department of Organic Chemistry, Institute of Chemistry, Eötvös Loránd University, 1117 Budapest, Hungary; (M.Y.); (D.S.); (C.B.)
| | - Gábor Mező
- MTA-ELTE Research Group of Peptide Chemistry, Eötvös Loránd Research Network (ELKH), Eötvös Loránd University, 1117 Budapest, Hungary;
- Department of Organic Chemistry, Institute of Chemistry, Eötvös Loránd University, 1117 Budapest, Hungary; (M.Y.); (D.S.); (C.B.)
| | - Zoltán Bánóczi
- Department of Organic Chemistry, Institute of Chemistry, Eötvös Loránd University, 1117 Budapest, Hungary; (M.Y.); (D.S.); (C.B.)
- Correspondence: (I.S.); (Z.B.)
| |
Collapse
|
31
|
Cesar-Silva D, Pereira-Dutra FS, Moraes Giannini AL, Jacques G. de Almeida C. The Endolysosomal System: The Acid Test for SARS-CoV-2. Int J Mol Sci 2022; 23:ijms23094576. [PMID: 35562967 PMCID: PMC9105036 DOI: 10.3390/ijms23094576] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 01/13/2022] [Accepted: 01/17/2022] [Indexed: 12/16/2022] Open
Abstract
This review aims to describe and discuss the different functions of the endolysosomal system, from homeostasis to its vital role during viral infections. We will initially describe endolysosomal system's main functions, presenting recent data on how its compartments are essential for host defense to explore later how SARS-CoV-2 (Severe Acute Respiratory Syndrome Coronavirus 2) and other coronaviruses subvert these organelles for their benefit. It is clear that to succeed, pathogens' evolution favored the establishment of ways to avoid, escape, or manipulate lysosomal function. The unavoidable coexistence with such an unfriendly milieu imposed on viruses the establishment of a vast array of strategies to make the most out of the invaded cell's machinery to produce new viruses and maneuvers to escape the host's defense system.
Collapse
Affiliation(s)
- Daniella Cesar-Silva
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Fundação Oswaldo Cruz, Rio de Janeiro 21040-900, Brazil; (D.C.-S.); (F.S.P.-D.)
| | - Filipe S. Pereira-Dutra
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Fundação Oswaldo Cruz, Rio de Janeiro 21040-900, Brazil; (D.C.-S.); (F.S.P.-D.)
| | - Ana Lucia Moraes Giannini
- Laboratory of Functional Genomics and Signal Transduction, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-901, Brazil;
| | - Cecília Jacques G. de Almeida
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Fundação Oswaldo Cruz, Rio de Janeiro 21040-900, Brazil; (D.C.-S.); (F.S.P.-D.)
- Correspondence: or
| |
Collapse
|
32
|
Truskewycz A, Yin H, Halberg N, Lai DTH, Ball AS, Truong VK, Rybicka AM, Cole I. Carbon Dot Therapeutic Platforms: Administration, Distribution, Metabolism, Excretion, Toxicity, and Therapeutic Potential. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2106342. [PMID: 35088534 DOI: 10.1002/smll.202106342] [Citation(s) in RCA: 88] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 12/14/2021] [Indexed: 06/14/2023]
Abstract
Ultrasmall nanoparticles are often grouped under the broad umbrella term of "nanoparticles" when reported in the literature. However, for biomedical applications, their small sizes give them intimate interactions with biological species and endow them with unique functional physiochemical properties. Carbon quantum dots (CQDs) are an emerging class of ultrasmall nanoparticles which have demonstrated considerable biocompatibility and have been employed as potent theragnostic platforms. These particles find application for increasing drug solubility and targeting, along with facilitating the passage of drugs across impermeable membranes (i.e., blood brain barrier). Further functionality can be triggered by various environmental conditions or external stimuli (i.e., pH, temperature, near Infrared (NIR) light, ultrasound), and their intrinsic fluorescence is valuable for diagnostic applications. The focus of this review is to shed light on the therapeutic potential of CQDs and identify how they travel through the body, reach their site of action, administer therapeutic effect, and are excreted. Investigation into their toxicity and compatibility with larger nanoparticle carriers is also examined. The future of CQDs for theragnostic applications is promising due to their multifunctional attributes and documented biocompatibility. As nanomaterial platforms become more commonplace in clinical treatments, the commercialization of CQD therapeutics is anticipated.
Collapse
Affiliation(s)
- Adam Truskewycz
- School of Engineering, Advanced Manufacturing and Fabrication, RMIT University, Melbourne, Victoria, 3000, Australia
- Department of Biomedicine, University of Bergen, Bergen, 5020, Norway
| | - Hong Yin
- School of Engineering, Advanced Manufacturing and Fabrication, RMIT University, Melbourne, Victoria, 3000, Australia
| | - Nils Halberg
- Department of Biomedicine, University of Bergen, Bergen, 5020, Norway
| | - Daniel T H Lai
- Institute of Health and Sport (IHES), Victoria University, Melbourne, Victoria, 3011, Australia
| | - Andrew S Ball
- ARC Training Centre for the Transformation of Australia Biosolids Resource, RMIT University, Melbourne, Victoria, 3000, Australia
| | - Vi Khanh Truong
- School of Science, Engineering and Health, RMIT University, Melbourne, Victoria, 3000, Australia
| | - Agata Marta Rybicka
- Oncovet Clinical Research, Parc Eurasante, 80 Rue du Dr Alexandre Yersin, Loos, F-59120, France
| | - Ivan Cole
- School of Engineering, Advanced Manufacturing and Fabrication, RMIT University, Melbourne, Victoria, 3000, Australia
| |
Collapse
|
33
|
Tian X, Bunda P, Ishibe S. Podocyte Endocytosis in Regulating the Glomerular Filtration Barrier. Front Med (Lausanne) 2022; 9:801837. [PMID: 35223901 PMCID: PMC8866310 DOI: 10.3389/fmed.2022.801837] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 01/06/2022] [Indexed: 12/26/2022] Open
Abstract
Endocytosis is a mechanism that internalizes and recycles plasma membrane components and transmembrane receptors via vesicle formation, which is mediated by clathrin-dependent and clathrin-independent signaling pathways. Podocytes are specialized, terminally differentiated epithelial cells in the kidney, located on the outermost layer of the glomerulus. These cells play an important role in maintaining the integrity of the glomerular filtration barrier in conjunction with the adjacent basement membrane and endothelial cell layers within the glomerulus. An intact podocyte endocytic machinery appears to be necessary for maintaining podocyte function. De novo pathologic human genetic mutations and loss-of-function studies of critical podocyte endocytosis genes in genetically engineered mouse models suggest that this pathway contributes to the pathophysiology of development and progression of proteinuria in chronic kidney disease. Here, we review the mechanism of cellular endocytosis and its regulation in podocyte injury in the context of glomerular diseases. A thorough understanding of podocyte endocytosis may shed novel insights into its biological function in maintaining a functioning filter and offer potential targeted therapeutic strategies for proteinuric glomerular diseases.
Collapse
Affiliation(s)
| | | | - Shuta Ishibe
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, United States
| |
Collapse
|
34
|
Wang X, Zhang Y, Mu X, Tu CR, Chung Y, Tsao SW, Chan GCF, Leung WH, Lau YL, Liu Y, Tu W. Exosomes derived from γδ-T cells synergize with radiotherapy and preserve antitumor activities against nasopharyngeal carcinoma in immunosuppressive microenvironment. J Immunother Cancer 2022; 10:jitc-2021-003832. [PMID: 35105688 PMCID: PMC8808451 DOI: 10.1136/jitc-2021-003832] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/04/2022] [Indexed: 12/30/2022] Open
Abstract
Background Radiotherapy is the first-line treatment for patients nasopharyngeal carcinoma (NPC), but its therapeutic efficacy is poor in some patients due to radioresistance. Adoptive T cell-based immunotherapy has also shown promise to control NPC; however, its antitumor efficacy may be attenuated by an immunosuppressive tumor microenvironment. Exosomes derived from γδ-T cells (γδ-T-Exos) have potent antitumor potentials. However, it remains unknown whether γδ-T-Exos have synergistic effect with radiotherapy and preserve their antitumor activities against NPC in an immunosuppressive tumor microenvironment. Methods γδ-T-Exos were stained with fluorescent membrane dye, and their interactions with NPC were determined both in vitro and in vivo. NPC cell deaths were detected after treatment with γδ-T-Exos and/or irradiation. Moreover, effects of γδ-T-Exos on radioresistant cancer stem-like cells (CSCs) were determined. The therapeutic efficacy of combination therapy using γδ-T-Exos and irradiation on NPC tumor progression was also monitored in vivo. Finally, the tumor-killing and T cell-promoting activities of γδ-T-Exos were determined under the culture in immunosuppressive NPC supernatant. Results γδ-T-Exos effectively interacted with NPC tumor cells in vitro and in vivo. γδ-T-Exos not only killed NPC cells in vitro, which was mainly mediated by Fas/Fas ligand (FasL) and death receptor 5 (DR5)/tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) pathways, but also controlled NPC tumor growth and prolonged tumor-bearing mice survival in vivo. Furthermore, γδ-T-Exos selectively targeted the radioresistant CD44+/high CSCs and induced profound cell apoptosis. The combination of γδ-T-Exos with radiotherapy overcame the radioresistance of CD44+/high NPC cells and significantly improved its therapeutic efficacy against NPC in vitro and in vivo. In addition, γδ-T-Exos promoted T-cell migration into NPC tumors by upregulating CCR5 on T cells that were chemoattracted by CCR5 ligands in the NPC tumor microenvironment. Although NPC tumor cells secreted abundant tumor growth factor beta to suppress T-cell responses, γδ-T-Exos preserved their direct antitumor activities and overcame the immunosuppressive NPC microenvironment to amplify T-cell antitumor immunity. Conclusions γδ-T-Exos synergized with radiotherapy to control NPC by overcoming the radioresistance of NPC CSCs. Moreover, γδ-T-Exos preserved their tumor-killing and T cell-promoting activities in the immunosuppressive NPC microenvironment. This study provides a proof of concept for a novel and potent strategy by combining γδ-T-Exos with radiotherapy in the control of NPC.
Collapse
Affiliation(s)
- Xiwei Wang
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Yanmei Zhang
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Xiaofeng Mu
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Chloe Ran Tu
- Computational and Systems Biology Interdepartmental Program, University of California Los Angeles, Los Angeles, California, USA
| | - Yuet Chung
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Sai Wah Tsao
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China
| | - Godfrey Chi-Fung Chan
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Wing-Hang Leung
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Yu-Lung Lau
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Yinping Liu
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Wenwei Tu
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
35
|
Siddiqui H, Yevstigneyev N, Madani G, McCormick S. Approaches to Visualising Endocytosis of LDL-Related Lipoproteins. Biomolecules 2022; 12:biom12020158. [PMID: 35204658 PMCID: PMC8961563 DOI: 10.3390/biom12020158] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 12/20/2021] [Accepted: 01/12/2022] [Indexed: 02/04/2023] Open
Abstract
Endocytosis is the process by which molecules are actively transported into cells. It can take on a variety of forms depending on the cellular machinery involved ranging from specific receptor-mediated endocytosis to the less selective and actin-driven macropinocytosis. The plasma lipoproteins, which deliver lipids and other cargo to cells, have been intensely studied with respect to their endocytic uptake. One of the first molecules to be visualised undergoing endocytosis via a receptor-mediated, clathrin-dependent pathway was low-density lipoprotein (LDL). The LDL molecule has subsequently been shown to be internalised through multiple endocytic pathways. Dissecting the pathways of lipoprotein endocytosis has been crucial to understanding the regulation of plasma lipid levels and how lipids enter cells in the arterial wall to promote atherosclerosis. It has also aided understanding of the dysregulation that occurs in plasma lipid levels when molecules involved in uptake are defective, as is the case in familial hypercholesterolemia (FH). The aim of this review is to outline the many endocytic pathways utilised for lipoprotein uptake. It explores the various experimental approaches that have been applied to visualise lipoprotein endocytosis with an emphasis on LDL and its more complex counterpart, lipoprotein(a) [Lp(a)]. Finally, we look at new developments in lipoprotein visualisation that hold promise for scrutinising endocytic pathways to finer detail in the future.
Collapse
Affiliation(s)
- Halima Siddiqui
- Department of Biochemistry, School of Biomedical Sciences, University of Otago, Dunedin 9016, New Zealand; (H.S.); (N.Y.); (G.M.)
- HeartOtago, School of Biomedical Sciences, University of Otago, Dunedin 9016, New Zealand
| | - Nikita Yevstigneyev
- Department of Biochemistry, School of Biomedical Sciences, University of Otago, Dunedin 9016, New Zealand; (H.S.); (N.Y.); (G.M.)
- HeartOtago, School of Biomedical Sciences, University of Otago, Dunedin 9016, New Zealand
| | - Golnoush Madani
- Department of Biochemistry, School of Biomedical Sciences, University of Otago, Dunedin 9016, New Zealand; (H.S.); (N.Y.); (G.M.)
- HeartOtago, School of Biomedical Sciences, University of Otago, Dunedin 9016, New Zealand
| | - Sally McCormick
- Department of Biochemistry, School of Biomedical Sciences, University of Otago, Dunedin 9016, New Zealand; (H.S.); (N.Y.); (G.M.)
- HeartOtago, School of Biomedical Sciences, University of Otago, Dunedin 9016, New Zealand
- Correspondence:
| |
Collapse
|
36
|
Makvandi P, Chen M, Sartorius R, Zarrabi A, Ashrafizadeh M, Dabbagh Moghaddam F, Ma J, Mattoli V, Tay FR. Endocytosis of abiotic nanomaterials and nanobiovectors: Inhibition of membrane trafficking. NANO TODAY 2021; 40:101279. [PMID: 34518771 PMCID: PMC8425779 DOI: 10.1016/j.nantod.2021.101279] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 08/05/2021] [Accepted: 08/19/2021] [Indexed: 05/04/2023]
Abstract
Humans are exposed to nanoscopical nanobiovectors (e.g. coronavirus SARS-CoV-2) as well as abiotic metal/carbon-based nanomaterials that enter cells serendipitously or intentionally. Understanding the interactions of cell membranes with these abiotic and biotic nanostructures will facilitate scientists to design better functional nanomaterials for biomedical applications. Such knowledge will also provide important clues for the control of viral infections and the treatment of virus-induced infectious diseases. In the present review, the mechanisms of endocytosis are reviewed in the context of how nanomaterials are uptaken into cells. This is followed by a detailed discussion of the attributes of man-made nanomaterials (e.g. size, shape, surface functional groups and elasticity) that affect endocytosis, as well as the different human cell types that participate in the endocytosis of nanomaterials. Readers are then introduced to the concept of viruses as nature-derived nanoparticles. The mechanisms in which different classes of viruses interact with various cell types to gain entry into the human body are reviewed with examples published over the last five years. These basic tenets will enable the avid reader to design advanced drug delivery and gene transfer nanoplatforms that harness the knowledge acquired from endocytosis to improve their biomedical efficacy. The review winds up with a discussion on the hurdles to be addressed in mimicking the natural mechanisms of endocytosis in nanomaterials design.
Collapse
Affiliation(s)
- Pooyan Makvandi
- Istituto Italiano di Tecnologia, Centre for Materials Interfaces, Viale Rinaldo Piaggio 34, 56025 Pontedera, Pisa, Italy
| | - Meiling Chen
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rossella Sartorius
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council (CNR), Naples 80131, Italy
| | - Ali Zarrabi
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, Istanbul 34956, Turkey
| | - Milad Ashrafizadeh
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, Istanbul 34956, Turkey
- Faculty of Engineering and Natural Sciences, Sabanci University, Orta Mahalle, Üniversite Caddesi No. 27, Orhanlı, Tuzla, 34956 Istanbul, Turkey
| | - Farnaz Dabbagh Moghaddam
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran 1477893855, Iran
| | - Jingzhi Ma
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Virgilio Mattoli
- Istituto Italiano di Tecnologia, Centre for Materials Interfaces, Viale Rinaldo Piaggio 34, 56025 Pontedera, Pisa, Italy
| | - Franklin R Tay
- The Graduate School, Augusta University, Augusta, GA 30912, United States
| |
Collapse
|
37
|
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
|
38
|
Zhou Y, Ariotti N, Rae J, Liang H, Tillu V, Tee S, Bastiani M, Bademosi AT, Collins BM, Meunier FA, Hancock JF, Parton RG. Caveolin-1 and cavin1 act synergistically to generate a unique lipid environment in caveolae. J Cell Biol 2021; 220:211716. [PMID: 33496726 PMCID: PMC7844427 DOI: 10.1083/jcb.202005138] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 11/20/2020] [Accepted: 12/21/2020] [Indexed: 01/09/2023] Open
Abstract
Caveolae are specialized domains of the vertebrate cell surface with a well-defined morphology and crucial roles in cell migration and mechanoprotection. Unique compositions of proteins and lipids determine membrane architectures. The precise caveolar lipid profile and the roles of the major caveolar structural proteins, caveolins and cavins, in selectively sorting lipids have not been defined. Here, we used quantitative nanoscale lipid mapping together with molecular dynamic simulations to define the caveolar lipid profile. We show that caveolin-1 (CAV1) and cavin1 individually sort distinct plasma membrane lipids. Intact caveolar structures composed of both CAV1 and cavin1 further generate a unique lipid nano-environment. The caveolar lipid sorting capability includes selectivities for lipid headgroups and acyl chains. Because lipid headgroup metabolism and acyl chain remodeling are tightly regulated, this selective lipid sorting may allow caveolae to act as transit hubs to direct communications among lipid metabolism, vesicular trafficking, and signaling.
Collapse
Affiliation(s)
- Yong Zhou
- Department of Integrative Biology and Pharmacology, University of Texas Medical School, Houston, TX
| | - Nicholas Ariotti
- University of New South Wales Sydney, Mark Wainwright Analytical Center, Sydney, New South Wales, Australia.,University of New South Wales Sydney, Department of Pathology, School of Medical Sciences, Kensington, Sydney, New South Wales, Australia
| | - James Rae
- The University of Queensland, Institute for Molecular Bioscience, Brisbane, Queensland, Australia
| | - Hong Liang
- Department of Integrative Biology and Pharmacology, University of Texas Medical School, Houston, TX
| | - Vikas Tillu
- The University of Queensland, Institute for Molecular Bioscience, Brisbane, Queensland, Australia
| | - Shern Tee
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland, Australia
| | - Michele Bastiani
- The University of Queensland, Institute for Molecular Bioscience, Brisbane, Queensland, Australia
| | - Adekunle T Bademosi
- Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia.,Clem Jones Centre for Ageing Dementia Research, The University of Queensland, Brisbane, Queensland, Australia
| | - Brett M Collins
- University of New South Wales Sydney, Department of Pathology, School of Medical Sciences, Kensington, Sydney, New South Wales, Australia
| | - Frederic A Meunier
- Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia.,Clem Jones Centre for Ageing Dementia Research, The University of Queensland, Brisbane, Queensland, Australia
| | - John F Hancock
- Department of Integrative Biology and Pharmacology, University of Texas Medical School, Houston, TX.,Program in Cell and Regulatory Biology, University of Texas Graduate School of Biomedical Sciences, Houston, TX
| | - Robert G Parton
- The University of Queensland, Institute for Molecular Bioscience, Brisbane, Queensland, Australia.,The University of Queensland, Centre for Microscopy and Microanalysis, Brisbane, Queensland, Australia
| |
Collapse
|
39
|
The Protein Toxins Ricin and Shiga Toxin as Tools to Explore Cellular Mechanisms of Internalization and Intracellular Transport. Toxins (Basel) 2021; 13:toxins13060377. [PMID: 34070659 PMCID: PMC8227415 DOI: 10.3390/toxins13060377] [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: 04/19/2021] [Revised: 05/12/2021] [Accepted: 05/22/2021] [Indexed: 12/18/2022] Open
Abstract
Protein toxins secreted by bacteria and found in plants can be threats to human health. However, their extreme toxicity can also be exploited in different ways, e.g., to produce hybrid toxins directed against cancer cells and to study transport mechanisms in cells. Investigations during the last decades have shown how powerful these molecules are as tools in cell biological research. Here, we first present a partly historical overview, with emphasis on Shiga toxin and ricin, of how such toxins have been used to characterize processes and proteins of importance for their trafficking. In the second half of the article, we describe how one can now use toxins to investigate the role of lipid classes for intracellular transport. In recent years, it has become possible to quantify hundreds of lipid species using mass spectrometry analysis. Thus, it is also now possible to explore the importance of lipid species in intracellular transport. The detailed analyses of changes in lipids seen under conditions of inhibited toxin transport reveal previously unknown connections between syntheses of lipid classes and demonstrate the ability of cells to compensate under given conditions.
Collapse
|
40
|
Xiu M, Huan X, Ou Y, Ying S, Wang J. The basic route of nuclear-targeted transport of IGF-1/IGF-1R and potential biological functions in intestinal epithelial cells. Cell Prolif 2021; 54:e13030. [PMID: 33932050 PMCID: PMC8168413 DOI: 10.1111/cpr.13030] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 02/08/2021] [Accepted: 02/20/2021] [Indexed: 01/05/2023] Open
Abstract
OBJECTIVES Insulin-like growth factor (IGF-1) plays an important role in many biological processes in the intestinal tract. However, the cellular behaviour and characteristics of IGF-1/IGF-1R in intestinal cells remain unclear. MATERIALS AND METHODS A series of techniques (such as indirect immunofluorescence, co-localization and Western blot) have been used to systematically study the cellular behaviour of IGF-1/IGF-1R on intestinal cells. RESULTS We found that IGF-1 can not only internalize into the cytoplasm, but also transport into the cell nuclei. We systematically studied the detailed molecular pathways of IGF-1/IGF-1R's nuclear translocation. We found that IGF-1R underwent clathrin-mediated endocytosis into cells and then entered into Rab-5-positive endosomes. Dynein/dynactin were used as motors to drive Rab-5-positive endosomes carrying IGF-1R (cargo molecule) to Golgi apparatus (transit station) along the surface of the microtubule. IGF-1 and/or IGF-1R entered the cell nuclei through NPC (nuclear pore complex), a process mediated by NUP358. Further study indicated that nuclear localization of IGF-1 and/or IGF-1R promoted cell proliferation and increased the nuclear residence time of signalling molecules activated by IGF-1. Further experiments showed that IGF-1R may regulate the transcription of genes in the cell nuclei, indicating that nuclear-localized IGF-1R plays an important in cell proliferation. CONCLUSIONS In short, we revealed the molecular mechanism by which IGF-1/IGF-1R transports into the cell nuclei of intestinal cells. More importantly, the current work showed that the nuclear-localized IGF-1R has important biological functions.
Collapse
Affiliation(s)
- Ming Xiu
- Department of Intensive care unit, The first hospital of Jilin University, Changchun, China
| | - Xia Huan
- Department of Respiratory Medicine, The First Hospital of Jilin University, Changchun, China
| | - Yang Ou
- The department of Geriatris, The first hospital of Jilin University, Changchun, China
| | - Sha Ying
- The department of Geriatris, The first hospital of Jilin University, Changchun, China
| | - Jianmeng Wang
- Department of Intensive care unit, The first hospital of Jilin University, Changchun, China.,The department of Geriatris, The first hospital of Jilin University, Changchun, China
| |
Collapse
|
41
|
Albacete-Albacete L, Navarro-Lérida I, López JA, Martín-Padura I, Astudillo AM, Ferrarini A, Van-Der-Heyden M, Balsinde J, Orend G, Vázquez J, Del Pozo MÁ. ECM deposition is driven by caveolin-1-dependent regulation of exosomal biogenesis and cargo sorting. J Cell Biol 2021; 219:211453. [PMID: 33053168 PMCID: PMC7551399 DOI: 10.1083/jcb.202006178] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 07/31/2020] [Accepted: 08/03/2020] [Indexed: 12/16/2022] Open
Abstract
The composition and physical properties of the extracellular matrix (ECM) critically influence tumor progression, but the molecular mechanisms underlying ECM layering are poorly understood. Tumor–stroma interaction critically depends on cell communication mediated by exosomes, small vesicles generated within multivesicular bodies (MVBs). We show that caveolin-1 (Cav1) centrally regulates exosome biogenesis and exosomal protein cargo sorting through the control of cholesterol content at the endosomal compartment/MVBs. Quantitative proteomics profiling revealed that Cav1 is required for exosomal sorting of ECM protein cargo subsets, including Tenascin-C (TnC), and for fibroblast-derived exosomes to efficiently deposit ECM and promote tumor invasion. Cav1-driven exosomal ECM deposition not only promotes local stromal remodeling but also the generation of distant ECM-enriched stromal niches in vivo. Cav1 acts as a cholesterol rheostat in MVBs, determining sorting of ECM components into specific exosome pools and thus ECM deposition. This supports a model by which Cav1 is a central regulatory hub for tumor–stroma interactions through a novel exosome-dependent ECM deposition mechanism.
Collapse
Affiliation(s)
- Lucas Albacete-Albacete
- Mechanoadaptation and Caveolae Biology Laboratory, Cell and Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
| | - Inmaculada Navarro-Lérida
- Mechanoadaptation and Caveolae Biology Laboratory, Cell and Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
| | - Juan Antonio López
- Cardiovascular Proteomics Lab, Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Madrid, Spain
| | - Inés Martín-Padura
- Mechanoadaptation and Caveolae Biology Laboratory, Cell and Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
| | - Alma M Astudillo
- Instituto de Biología y Genética Molecular, Consejo Superior de Investigaciones Científicas, Universidad de Valladolid, Valladolid, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, Madrid, Spain
| | - Alessia Ferrarini
- Cardiovascular Proteomics Lab, Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
| | - Michael Van-Der-Heyden
- Institut National de la Santé et de la Recherche Médicale U1109-MN3T, The Microenvironmental Niche in Tumorigenesis and Targeted Therapy, Université de Strasbourg, LabEx Medalis, Fédération de Médecine Translationnelle de Strasbourg, Strasbourg, France
| | - Jesús Balsinde
- Instituto de Biología y Genética Molecular, Consejo Superior de Investigaciones Científicas, Universidad de Valladolid, Valladolid, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, Madrid, Spain
| | - Gertraud Orend
- Institut National de la Santé et de la Recherche Médicale U1109-MN3T, The Microenvironmental Niche in Tumorigenesis and Targeted Therapy, Université de Strasbourg, LabEx Medalis, Fédération de Médecine Translationnelle de Strasbourg, Strasbourg, France
| | - Jesús Vázquez
- Cardiovascular Proteomics Lab, Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Madrid, Spain
| | - Miguel Ángel Del Pozo
- Mechanoadaptation and Caveolae Biology Laboratory, Cell and Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
| |
Collapse
|
42
|
Rennick JJ, Johnston APR, Parton RG. Key principles and methods for studying the endocytosis of biological and nanoparticle therapeutics. NATURE NANOTECHNOLOGY 2021; 16:266-276. [PMID: 33712737 DOI: 10.1038/s41565-021-00858-8] [Citation(s) in RCA: 704] [Impact Index Per Article: 176.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 01/19/2021] [Indexed: 05/20/2023]
Abstract
Endocytosis is a critical step in the process by which many therapeutic nanomedicines reach their intracellular targets. Our understanding of cellular uptake mechanisms has developed substantially in the past five years. However, these advances in cell biology have not fully translated to the nanoscience and therapeutics literature. Misconceptions surrounding the role of different endocytic pathways and how to study these pathways are hindering progress in developing improved nanoparticle therapies. Here, we summarize the latest insights into cellular uptake mechanisms and pathways. We highlight limitations of current systems to study endocytosis, particularly problems with non-specific inhibitors. We also summarize alternative genetic approaches to robustly probe these pathways and discuss the need to understand how cells endocytose particles in vivo. We hope that this critical assessment of the current methods used in studying nanoparticle uptake will guide future studies at the interface of cell biology and nanomedicine.
Collapse
Affiliation(s)
- Joshua J Rennick
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Brisbane, Queensland, Australia
| | - Angus P R Johnston
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia.
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Brisbane, Queensland, Australia.
| | - Robert G Parton
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Brisbane, Queensland, Australia.
- Institute for Molecular Bioscience and Centre for Microscopy and Microanalysis, The University of Queensland, Brisbane, Queensland, Australia.
| |
Collapse
|
43
|
Abstract
Caveolin-1 (CAV1) has long been implicated in cancer progression, and while widely accepted as an oncogenic protein, CAV1 also has tumor suppressor activity. CAV1 was first identified in an early study as the primary substrate of Src kinase, a potent oncoprotein, where its phosphorylation correlated with cellular transformation. Indeed, CAV1 phosphorylation on tyrosine-14 (Y14; pCAV1) has been associated with several cancer-associated processes such as focal adhesion dynamics, tumor cell migration and invasion, growth suppression, cancer cell metabolism, and mechanical and oxidative stress. Despite this, a clear understanding of the role of Y14-phosphorylated pCAV1 in cancer progression has not been thoroughly established. Here, we provide an overview of the role of Src-dependent phosphorylation of tumor cell CAV1 in cancer progression, focusing on pCAV1 in tumor cell migration, focal adhesion signaling and metabolism, and in the cancer cell response to stress pathways characteristic of the tumor microenvironment. We also discuss a model for Y14 phosphorylation regulation of CAV1 effector protein interactions via the caveolin scaffolding domain.
Collapse
|
44
|
Parton RG, Kozlov MM, Ariotti N. Caveolae and lipid sorting: Shaping the cellular response to stress. J Cell Biol 2020; 219:133844. [PMID: 32328645 PMCID: PMC7147102 DOI: 10.1083/jcb.201905071] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 10/30/2019] [Accepted: 02/05/2020] [Indexed: 02/06/2023] Open
Abstract
Caveolae are an abundant and characteristic surface feature of many vertebrate cells. The uniform shape of caveolae is characterized by a bulb with consistent curvature connected to the plasma membrane (PM) by a neck region with opposing curvature. Caveolae act in mechanoprotection by flattening in response to increased membrane tension, and their disassembly influences the lipid organization of the PM. Here, we review evidence for caveolae as a specialized lipid domain and speculate on mechanisms that link changes in caveolar shape and/or protein composition to alterations in specific lipid species. We propose that high membrane curvature in specific regions of caveolae can enrich specific lipid species, with consequent changes in their localization upon caveolar flattening. In addition, we suggest how changes in the association of lipid-binding caveolar proteins upon flattening of caveolae could allow release of specific lipids into the bulk PM. We speculate that the caveolae-lipid system has evolved to function as a general stress-sensing and stress-protective membrane domain.
Collapse
Affiliation(s)
- Robert G Parton
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia.,Centre for Microscopy and Microanalysis, The University of Queensland, Brisbane, Australia
| | - Michael M Kozlov
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Nicholas Ariotti
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia.,Electron Microscope Unit, Mark Wainwright Analytical Centre, The University of New South Wales, Kensington, Australia.,Department of Pathology, School of Medical Sciences, The University of New South Wales, Kensington, Australia
| |
Collapse
|
45
|
Corrotte M, Cerasoli M, Maeda FY, Andrews NW. Endophilin-A2-dependent tubular endocytosis promotes plasma membrane repair and parasite invasion. J Cell Sci 2020; 134:jcs249524. [PMID: 33093240 PMCID: PMC7725609 DOI: 10.1242/jcs.249524] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 10/12/2020] [Indexed: 12/12/2022] Open
Abstract
Endocytosis of caveolae has previously been implicated in the repair of plasma membrane wounds. Here, we show that caveolin-1-deficient fibroblasts lacking caveolae upregulate a tubular endocytic pathway and have a reduced capacity to reseal after permeabilization with pore-forming toxins compared with wild-type cells. Silencing endophilin-A2 expression inhibited fission of endocytic tubules and further reduced plasma membrane repair in cells lacking caveolin-1, supporting a role for tubular endocytosis as an alternative pathway for the removal of membrane lesions. Endophilin-A2 was visualized in association with cholera toxin B-containing endosomes and was recruited to recently formed intracellular vacuoles containing Trypanosoma cruzi, a parasite that utilizes the plasma membrane wounding repair pathway to invade host cells. Endophilin-A2 deficiency inhibited T. cruzi invasion, and fibroblasts deficient in both caveolin-1 and endophilin-A2 did not survive prolonged exposure to the parasites. These findings reveal a novel crosstalk between caveolin-1 and endophilin-A2 in the regulation of clathrin-independent endocytosis and plasma membrane repair, a process that is subverted by T. cruzi parasites for cell invasion.
Collapse
Affiliation(s)
- Matthias Corrotte
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742, USA
- Department of Veterinary Medicine, VA-MD College of Veterinary Medicine, University of Maryland, College Park, MD 20742, USA
| | - Mark Cerasoli
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742, USA
| | - Fernando Y Maeda
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742, USA
| | - Norma W Andrews
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742, USA
| |
Collapse
|
46
|
Proteins involved in actin filament organization are key host factors for Japanese encephalitis virus life-cycle in human neuronal cells. Microb Pathog 2020; 149:104565. [DOI: 10.1016/j.micpath.2020.104565] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 10/01/2020] [Accepted: 10/01/2020] [Indexed: 12/22/2022]
|
47
|
Toth AE, Holst MR, Nielsen MS. Vesicular Transport Machinery in Brain Endothelial Cells: What We Know and What We Do not. Curr Pharm Des 2020; 26:1405-1416. [PMID: 32048959 DOI: 10.2174/1381612826666200212113421] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 12/03/2019] [Indexed: 12/20/2022]
Abstract
The vesicular transport machinery regulates numerous essential functions in cells such as cell polarity, signaling pathways, and the transport of receptors and their cargoes. From a pharmaceutical perspective, vesicular transport offers avenues to facilitate the uptake of therapeutic agents into cells and across cellular barriers. In order to improve receptor-mediated transcytosis of biologics across the blood-brain barrier and into the diseased brain, a detailed understanding of intracellular transport mechanisms is essential. The vesicular transport machinery is a highly complex network and involves an array of protein complexes, cytosolic adaptor proteins, and the subcellular structures of the endo-lysosomal system. The endo-lysosomal system includes several types of vesicular entities such as early, late, and recycling endosomes, exosomes, ectosomes, retromer-coated vesicles, lysosomes, trans-endothelial channels, and tubules. While extensive research has been done on the trafficking system in many cell types, little is known about vesicular trafficking in brain endothelial cells. Consequently, assumptions on the transport system in endothelial cells are based on findings in polarised epithelial cells, although recent studies have highlighted differences in the endothelial system. This review highlights aspects of the vesicular trafficking machinery in brain endothelial cells, including recent findings, limitations, and opportunities for further studies.
Collapse
Affiliation(s)
- Andrea E Toth
- Department of Biomedicine, Faculty of Health, Aarhus University, Høegh-Guldberg Gade 10, 8000 Aarhus C, Denmark
| | - Mikkel R Holst
- Department of Biomedicine, Faculty of Health, Aarhus University, Høegh-Guldberg Gade 10, 8000 Aarhus C, Denmark
| | - Morten S Nielsen
- Department of Biomedicine, Faculty of Health, Aarhus University, Høegh-Guldberg Gade 10, 8000 Aarhus C, Denmark
| |
Collapse
|
48
|
Buwa N, Mazumdar D, Balasubramanian N. Caveolin1 Tyrosine-14 Phosphorylation: Role in Cellular Responsiveness to Mechanical Cues. J Membr Biol 2020; 253:509-534. [PMID: 33089394 DOI: 10.1007/s00232-020-00143-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 10/05/2020] [Indexed: 02/07/2023]
Abstract
The plasma membrane is a dynamic lipid bilayer that engages with the extracellular microenvironment and intracellular cytoskeleton. Caveolae are distinct plasma membrane invaginations lined by integral membrane proteins Caveolin1, 2, and 3. Caveolae formation and stability is further supported by additional proteins including Cavin1, EHD2, Pacsin2 and ROR1. The lipid composition of caveolar membranes, rich in cholesterol and phosphatidylserine, actively contributes to caveolae formation and function. Post-translational modifications of Cav1, including its phosphorylation of the tyrosine-14 residue (pY14Cav1) are vital to its function in and out of caveolae. Cells that experience significant mechanical stress are seen to have abundant caveolae. They play a vital role in regulating cellular signaling and endocytosis, which could further affect the abundance and distribution of caveolae at the PM, contributing to sensing and/or buffering mechanical stress. Changes in membrane tension in cells responding to multiple mechanical stimuli affects the organization and function of caveolae. These mechanical cues regulate pY14Cav1 levels and function in caveolae and focal adhesions. This review, along with looking at the mechanosensitive nature of caveolae, focuses on the role of pY14Cav1 in regulating cellular mechanotransduction.
Collapse
Affiliation(s)
- Natasha Buwa
- Indian Institute of Science Education and Research, Pune, Dr. Homi Bhabha Road, Pashan, Pune, 411008, India
| | - Debasmita Mazumdar
- Indian Institute of Science Education and Research, Pune, Dr. Homi Bhabha Road, Pashan, Pune, 411008, India
| | - Nagaraj Balasubramanian
- Indian Institute of Science Education and Research, Pune, Dr. Homi Bhabha Road, Pashan, Pune, 411008, India.
| |
Collapse
|
49
|
Mayberry CL, Maginnis MS. Taking the Scenic Route: Polyomaviruses Utilize Multiple Pathways to Reach the Same Destination. Viruses 2020; 12:v12101168. [PMID: 33076363 PMCID: PMC7602598 DOI: 10.3390/v12101168] [Citation(s) in RCA: 5] [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: 09/22/2020] [Revised: 10/08/2020] [Accepted: 10/09/2020] [Indexed: 01/02/2023] Open
Abstract
Members of the Polyomaviridae family differ in their host range, pathogenesis, and disease severity. To date, some of the most studied polyomaviruses include human JC, BK, and Merkel cell polyomavirus and non-human subspecies murine and simian virus 40 (SV40) polyomavirus. Although dichotomies in host range and pathogenesis exist, overlapping features of the infectious cycle illuminate the similarities within this virus family. Of particular interest to human health, JC, BK, and Merkel cell polyomavirus have all been linked to critical, often fatal, illnesses, emphasizing the importance of understanding the underlying viral infections that result in the onset of these diseases. As there are significant overlaps in the capacity of polyomaviruses to cause disease in their respective hosts, recent advancements in characterizing the infectious life cycle of non-human murine and SV40 polyomaviruses are key to understanding diseases caused by their human counterparts. This review focuses on the molecular mechanisms by which different polyomaviruses hijack cellular processes to attach to host cells, internalize, traffic within the cytoplasm, and disassemble within the endoplasmic reticulum (ER), prior to delivery to the nucleus for viral replication. Unraveling the fundamental processes that facilitate polyomavirus infection provides deeper insight into the conserved mechanisms of the infectious process shared within this virus family, while also highlighting critical unique viral features.
Collapse
Affiliation(s)
- Colleen L. Mayberry
- Department of Molecular and Biomedical Sciences, The University of Maine, Orono, ME 04469, USA;
| | - Melissa S. Maginnis
- Department of Molecular and Biomedical Sciences, The University of Maine, Orono, ME 04469, USA;
- Graduate School in Biomedical Sciences and Engineering, The University of Maine, Orono, ME 04469, USA
- Correspondence:
| |
Collapse
|
50
|
A molecular sensor to quantify the localization of proteins, DNA and nanoparticles in cells. Nat Commun 2020; 11:4482. [PMID: 32901011 PMCID: PMC7479595 DOI: 10.1038/s41467-020-18082-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 07/28/2020] [Indexed: 12/22/2022] Open
Abstract
Intracellular trafficking governs receptor signaling, pathogenesis, immune responses and fate of nanomedicines. These processes are typically tracked by observing colocalization of fluorescent markers using confocal microscopy. However, this method is low throughput, limited by the resolution of microscopy, and can miss fleeting interactions. To address this, we developed a localization sensor composed of a quenched SNAP-tag substrate (SNAPSwitch) that can be conjugated to biomolecules using click chemistry. SNAPSwitch enables quantitative detection of trafficking to locations of interest within live cells using flow cytometry. Using SNAPSwitch, we followed the trafficking of DNA complexes from endosomes into the cytosol and nucleus. We show that antibodies against the transferrin or hyaluronan receptor are initially sorted into different compartments following endocytosis. In addition, we can resolve which side of the cellular membrane material was located. These results demonstrate SNAPSwitch is a high-throughput and broadly applicable tool to quantitatively track localization of materials in cells. Determining the trafficking of intracellular material is commonly done by colocalisation analysis using microscopy. Here the authors monitor trafficking of select cargo by measuring the conversion of quenched SNAP-tag substrates by subcellularly-localised SNAP-tag and detection by flow cytometry.
Collapse
|