1
|
Heydecker M, Shitara A, Chen D, Tran DT, Masedunskas A, Tora MS, Ebrahim S, Appaduray MA, Galeano Niño JL, Bhardwaj A, Narayan K, Hardeman EC, Gunning PW, Weigert R. Coordination of force-generating actin-based modules stabilizes and remodels membranes in vivo. J Cell Biol 2024; 223:e202401091. [PMID: 39172125 PMCID: PMC11344176 DOI: 10.1083/jcb.202401091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 06/18/2024] [Accepted: 08/07/2024] [Indexed: 08/23/2024] Open
Abstract
Membrane remodeling drives a broad spectrum of cellular functions, and it is regulated through mechanical forces exerted on the membrane by cytoplasmic complexes. Here, we investigate how actin filaments dynamically tune their structure to control the active transfer of membranes between cellular compartments with distinct compositions and biophysical properties. Using intravital subcellular microscopy in live rodents we show that a lattice composed of linear filaments stabilizes the granule membrane after fusion with the plasma membrane and a network of branched filaments linked to the membranes by Ezrin, a regulator of membrane tension, initiates and drives to completion the integration step. Our results highlight how the actin cytoskeleton tunes its structure to adapt to dynamic changes in the biophysical properties of membranes.
Collapse
Affiliation(s)
- Marco Heydecker
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- School of Biomedical Sciences, University of New South Wales Sydney, Sydney, Australia
| | - Akiko Shitara
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Department of Pharmacology, Asahi University School of Dentistry, Gifu, Japan
| | - Desu Chen
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Duy T Tran
- NIDCR Imaging Core, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - Andrius Masedunskas
- School of Biomedical Sciences, University of New South Wales Sydney, Sydney, Australia
| | - Muhibullah S Tora
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Seham Ebrahim
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Department of Molecular Physiology and Biological Physics, Center for Membrane and Cell Physiology, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Mark A Appaduray
- School of Biomedical Sciences, University of New South Wales Sydney, Sydney, Australia
| | - Jorge Luis Galeano Niño
- EMBL Australia, Single Molecule Science node, University of New South Wales Sydney, Sydney, Australia
| | - Abhishek Bhardwaj
- Center for Molecular Microscopy, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Kedar Narayan
- Center for Molecular Microscopy, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Edna C Hardeman
- School of Biomedical Sciences, University of New South Wales Sydney, Sydney, Australia
| | - Peter W Gunning
- School of Biomedical Sciences, University of New South Wales Sydney, Sydney, Australia
| | - Roberto Weigert
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
2
|
Thamizhchelvan AM, Ma H, Wu T, Nguyen D, Padelford J, Whitworth TJ, Li Y, Yang L, Mao H. Shape-dependent cellular uptake of iron oxide nanorods: mechanisms of endocytosis and implications on cell labeling and cellular delivery. NANOSCALE 2024. [PMID: 39329423 PMCID: PMC11429166 DOI: 10.1039/d4nr02408g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 09/11/2024] [Indexed: 09/28/2024]
Abstract
The effects of nanoparticle morphology, especially size and shape, on their interactions with cells are of great interest in understanding the fate of nanoparticles in biological systems and designing them for biomedical applications. While size and shape-dependent cell behavior, endocytosis mechanism, and subcellular distribution of nanoparticles have been investigated extensively with gold and other nanoparticles, studies on iron oxide nanoparticles (IONP), one of the most promising and well-thought-of nanomaterials in biomedical applications, were limited. In this study, we synthesized oligosaccharide-coated water-soluble iron oxide nanorods (IONR) with different core sizes (nm) and different aspect ratios (i.e., length/width), such as IONR(L) at 140/6 nm and IONR(S) at 50/7 nm as well as spherical IONP (20 nm). We investigated how their sizes and shapes affect uptake mechanisms, localization, and cell viability in different cell lines. The results of transmission electron microscopy (TEM) and confocal fluorescence microscopic imaging confirmed the internalization of these nanoparticles in different types of cells and subsequent accumulation in the subcellular compartments, such as the endosomes, and into the cytosol. Specifically, IONR(L) exhibited the highest cellular uptake compared to IONR(S) and spherical IONP, 1.36-fold and 1.17-fold higher than that of spherical IONP in macrophages and pediatric brain tumor medulloblastoma cells, respectively. To examine the cellular uptake mechanisms preferred by the different IONR and IONP, we used different endocytosis inhibitors to block specific cellular internalization pathways when cells were treated with different nanoparticles. The results from these blocking experiments showed that IONR(L) enter macrophages and normal kidney cells through clathrin-mediated, dynamin-dependent, and macropinocytosis/phagocytosis pathways, while they are internalized in cancer cells primarily via clathrin/caveolae-mediated and phagocytosis mechanisms. Overall, our findings provide new insights into further development of magnetic IONR-based imaging probes and drug delivery systems for biomedical applications.
Collapse
Affiliation(s)
- Anbu Mozhi Thamizhchelvan
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, Georgia 30322, USA.
| | - Hedi Ma
- 5M Biomed, LLC, Atlanta, Georgia 30303, USA
| | - Tianhe Wu
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, Georgia 30322, USA.
| | - Darlene Nguyen
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, Georgia 30322, USA.
| | | | - Ted J Whitworth
- Robert P. Apkarian Integrated Electron Microscopy Core, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | - Yuancheng Li
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, Georgia 30322, USA.
- 5M Biomed, LLC, Atlanta, Georgia 30303, USA
| | - Lily Yang
- Department of Surgery Emory University School of Medicine, Atlanta, Georgia 30322, USA
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | - Hui Mao
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, Georgia 30322, USA.
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| |
Collapse
|
3
|
Wang X, Berro J, Ma R. Vesiculation pathways in clathrin-mediated endocytosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.13.607731. [PMID: 39185216 PMCID: PMC11343097 DOI: 10.1101/2024.08.13.607731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
During clathrin-mediated endocytosis, a patch of flat plasma membrane is internalized to form a vesicle. In mammalian cells, how the clathrin coat deforms the membrane into a vesicle remains unclear and two main hypotheses have been debated. The "constant area" hypothesis assumes that clathrin molecules initially form a flat lattice on the membrane and deform the membrane by changing its intrinsic curvature while keeping the coating area constant. The alternative "constant curvature" hypothesis assumes that the intrinsic curvature of the clathrin lattice remains constant during the formation of a vesicle while the surface area it covers increases. Previous experimental studies were unable to unambiguously determine which hypothesis is correct. In this paper, we show that these two hypotheses are only two extreme cases of a continuum of vesiculation pathways if we account for the free energies associated with clathrin assembly and curvature generation. By tracing the negative gradient of the free energy, we define vesiculation pathways in the phase space of the coating area and the intrinsic curvature of clathrin coat. Our results show that, overall, the differences in measurable membrane morphology between the different models are not as big as expected, and the main differences are most salient at the early stage of endocytosis. Furthermore, the best fitting pathway to experimental data is not compatible with the constant-curvature model and resembles a constant-area-like pathway where the coating area initially expands with minor changes in the intrinsic curvature, later followed by a dramatic increase in the intrinsic curvature and minor change in the coating area. Our results also suggest that experimental measurement of the tip radius and the projected area of the clathrin coat will be the key to distinguish between models.
Collapse
Affiliation(s)
- Xinran Wang
- Department of Physics, Xiamen University, Xiamen 361005, China
- Fujian Provincial Key Lab for Soft Functional Materials Research, Xiamen University, 361005, China
| | - Julien Berro
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06520, USA
- Nanobiology Institute, Yale University, West Haven, CT 06516, USA
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Rui Ma
- Department of Physics, Xiamen University, Xiamen 361005, China
- Fujian Provincial Key Lab for Soft Functional Materials Research, Xiamen University, 361005, China
| |
Collapse
|
4
|
Wan C, Puscher H, Ouyang Y, Wu J, Tian Y, Li S, Yin Q, Shen J. An AAGAB-to-CCDC32 handover mechanism controls the assembly of the AP2 adaptor complex. Proc Natl Acad Sci U S A 2024; 121:e2409341121. [PMID: 39145939 PMCID: PMC11348294 DOI: 10.1073/pnas.2409341121] [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: 05/09/2024] [Accepted: 07/13/2024] [Indexed: 08/16/2024] Open
Abstract
Vesicular transport relies on multimeric trafficking complexes to capture cargo and drive vesicle budding and fusion. Faithful assembly of the trafficking complexes is essential to their functions but remains largely unexplored. Assembly of AP2 adaptor, a heterotetrameric protein complex regulating clathrin-mediated endocytosis, is assisted by the chaperone AAGAB. Here, we found that AAGAB initiates AP2 assembly by stabilizing its α and σ2 subunits, but the AAGAB:α:σ2 complex cannot recruit additional AP2 subunits. We identified CCDC32 as another chaperone regulating AP2 assembly. CCDC32 recognizes the AAGAB:α:σ2 complex, and its binding leads to the formation of an α:σ2:CCDC32 ternary complex. The α:σ2:CCDC32 complex serves as a template that sequentially recruits the µ2 and β2 subunits of AP2 to complete AP2 assembly, accompanied by CCDC32 release. The AP2-regulating function of CCDC32 is disrupted by a disease-causing mutation. These findings demonstrate that AP2 is assembled by a handover mechanism switching from AAGAB-based initiation complexes to CCDC32-based template complexes. A similar mechanism may govern the assembly of other trafficking complexes exhibiting the same configuration as AP2.
Collapse
Affiliation(s)
- Chun Wan
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO80309
| | - Harrison Puscher
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO80309
| | - Yan Ouyang
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO80309
| | - Jingyi Wu
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO80309
| | - Yuan Tian
- Department of Biological Sciences and Institute of Molecular Biophysics, Florida State University, Tallahassee, FL32306
| | - Suzhao Li
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO80045
| | - Qian Yin
- Department of Biological Sciences and Institute of Molecular Biophysics, Florida State University, Tallahassee, FL32306
| | - Jingshi Shen
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO80309
| |
Collapse
|
5
|
Wang C, Zhou G, Guo X, Zhang W, Wu C. Electrical Stimulation Promotes Endocytosis of Magnetic Nanoparticles by Cancer Cells. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024:e2403381. [PMID: 39126240 DOI: 10.1002/smll.202403381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Indexed: 08/12/2024]
Abstract
Nanomaterials are increasingly used in biomedical imaging and cancer therapy, and how to improve the endocytosis of nanomaterials by cells is a key issue. The application of alternating current (AC) electrical stimulation to osteosarcoma cells (MG-63) here can increase the cellular endocytosis of Fe3O4 nanoparticles (diameter: 50 nm) by 52.46% via macropinocytosis. This can be ascribed to the decrease in F-actin content and the increase in intracellular Ca2+ concentration. Transmission electron microscope, immunofluorescence staining, western blot, flow cytometry, and inductively coupled plasma emission spectrometer analyses support this interpretation. The application of electrical stimulation decreases the cell viability in magnetic hyperthermia by 47.6% and increases the signal intensity of magnetic resonance imaging by 29%. Similar enhanced endocytosis is observed for breast cancer cells (MCF-7), glioblastoma cells (U-87 MG), melanoma cells (A-375), and bladder cancer cells (TCCSUP), and also for Fe3O4 nanoparticles with the diameters of 20 and 100 nm, and Zn0.54Co0.46Cr0.65Fe1.35O4 nanoparticles with the diameter of 70 nm. It seems the electrical stimulation has the potential to improve the diagnostic and therapeutic effects of magnetic nanoparticles by promoting endocytosis.
Collapse
Affiliation(s)
- Chao Wang
- State Key Laboratory of Structural Analysis, Optimization and CAE Software for Industrial Equipment, School of Mechanics and Aerospace Engineering, Dalian University of Technology, Dalian, 116024, China
| | - Guanlin Zhou
- State Key Laboratory of Structural Analysis, Optimization and CAE Software for Industrial Equipment, School of Mechanics and Aerospace Engineering, Dalian University of Technology, Dalian, 116024, China
| | - Xu Guo
- State Key Laboratory of Structural Analysis, Optimization and CAE Software for Industrial Equipment, School of Mechanics and Aerospace Engineering, Dalian University of Technology, Dalian, 116024, China
| | - Wei Zhang
- State Key Laboratory of Structural Analysis, Optimization and CAE Software for Industrial Equipment, School of Mechanics and Aerospace Engineering, Dalian University of Technology, Dalian, 116024, China
| | - Chengwei Wu
- State Key Laboratory of Structural Analysis, Optimization and CAE Software for Industrial Equipment, School of Mechanics and Aerospace Engineering, Dalian University of Technology, Dalian, 116024, China
| |
Collapse
|
6
|
Centner CS, Belott CJ, Patel RK, Menze MA, Yaddanapudi K, Kopechek JA. Biomodulatory Effects of Molecular Delivery in Human T Cells Using 3D-Printed Acoustofluidic Devices. ULTRASOUND IN MEDICINE & BIOLOGY 2024:S0301-5629(24)00256-4. [PMID: 39107206 DOI: 10.1016/j.ultrasmedbio.2024.06.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 05/28/2024] [Accepted: 06/21/2024] [Indexed: 08/09/2024]
Abstract
OBJECTIVE Cell-based therapies have shown significant promise for treating many diseases, including cancer. Current cell therapy manufacturing processes primarily utilize viral transduction to insert genomic material into cells, which has limitations, including variable transduction efficiency and extended processing times. Non-viral transfection techniques are also limited by high variability or reduced molecular delivery efficiency. Novel 3D-printed acoustofluidic devices are in development to address these challenges by delivering biomolecules into cells within seconds via sonoporation. METHODS In this study, we assessed biological parameters that influence the ultrasound-mediated delivery of fluorescent molecules (i.e., calcein and 150 kDa FITC-Dextran) to human T cells using flow cytometry and confocal imaging. RESULTS Low cell plating densities (100,000 cells/mL) enhanced molecular delivery compared to higher cell plating densities (p < 0.001), even though cells were resuspended at equal concentrations for acoustofluidic processing. Additionally, cells in the S phase of the cell cycle had enhanced intracellular delivery compared to cells in the G2/M phase (p < 0.001) and G0/G1 phase (p < 0.01), while also maintaining higher viability compared to G0/G1 phase (p < 0.001). Furthermore, the calcium chelator (EGTA) decreased overall molecular delivery levels. Confocal imaging indicated that the actin cytoskeleton had important implications on plasma membrane recovery dynamics after sonoporation. In addition, confocal imaging indicates that acoustofluidic treatment can permeabilize the nuclear membrane, which could enable rapid intranuclear delivery of nucleic acids. CONCLUSIONS The results of this study demonstrate that a 3D-printed acoustofluidic device can enhance molecular delivery to human T cells, which may enable improved techniques for non-viral processing of cell therapies.
Collapse
Affiliation(s)
- Connor S Centner
- Department of Bioengineering, University of Louisville, Louisville, KY, USA
| | - Clinton J Belott
- Department of Biology, University of Louisville, Louisville, KY, USA
| | - Riyakumari K Patel
- Department of Bioengineering, University of Louisville, Louisville, KY, USA
| | - Michael A Menze
- Department of Biology, University of Louisville, Louisville, KY, USA
| | | | - Jonathan A Kopechek
- Department of Bioengineering, University of Louisville, Louisville, KY, USA.
| |
Collapse
|
7
|
Liu J. Roles of membrane mechanics-mediated feedback in membrane traffic. Curr Opin Cell Biol 2024; 89:102401. [PMID: 39018789 PMCID: PMC11297666 DOI: 10.1016/j.ceb.2024.102401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 06/23/2024] [Accepted: 06/26/2024] [Indexed: 07/19/2024]
Abstract
Synthesizing the recent progresses, we present our perspectives on how local modulations of membrane curvature, tension, and bending energy define the feedback controls over membrane traffic processes. We speculate the potential mechanisms of, and the control logic behind, the different membrane mechanics-mediated feedback in endocytosis and exo-endocytosis coupling. We elaborate the path forward with the open questions for theoretical considerations and the grand challenges for experimental validations.
Collapse
Affiliation(s)
- Jian Liu
- Department of Cell Biology, Center for Cell Dynamics, School of Medicine, Johns Hopkins University, Baltimore, MD, United States.
| |
Collapse
|
8
|
Maninger JK, Nowak K, Goberdhan S, O'Donoghue R, Connor-Robson N. Cell type-specific functions of Alzheimer's disease endocytic risk genes. Philos Trans R Soc Lond B Biol Sci 2024; 379:20220378. [PMID: 38368934 PMCID: PMC10874703 DOI: 10.1098/rstb.2022.0378] [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: 03/17/2023] [Accepted: 09/12/2023] [Indexed: 02/20/2024] Open
Abstract
Endocytosis is a key cellular pathway required for the internalization of cellular nutrients, lipids and receptor-bound cargoes. It is also critical for the recycling of cellular components, cellular trafficking and membrane dynamics. The endocytic pathway has been consistently implicated in Alzheimer's disease (AD) through repeated genome-wide association studies and the existence of rare coding mutations in endocytic genes. BIN1 and PICALM are two of the most significant late-onset AD risk genes after APOE and are both key to clathrin-mediated endocytic biology. Pathological studies also demonstrate that endocytic dysfunction is an early characteristic of late-onset AD, being seen in the prodromal phase of the disease. Different cell types of the brain have specific requirements of the endocytic pathway. Neurons require efficient recycling of synaptic vesicles and microglia use the specialized form of endocytosis-phagocytosis-for their normal function. Therefore, disease-associated changes in endocytic genes will have varied impacts across different cell types, which remains to be fully explored. Given the genetic and pathological evidence for endocytic dysfunction in AD, understanding how such changes and the related cell type-specific vulnerabilities impact normal cellular function and contribute to disease is vital and could present novel therapeutic opportunities. This article is part of a discussion meeting issue 'Understanding the endo-lysosomal network in neurodegeneration'.
Collapse
Affiliation(s)
| | - Karolina Nowak
- Cardiff University, Dementia Research Institute, Cardiff University¸ Cardiff, CF24 4HQ, UK
| | - Srilakshmi Goberdhan
- Cardiff University, Dementia Research Institute, Cardiff University¸ Cardiff, CF24 4HQ, UK
| | - Rachel O'Donoghue
- Cardiff University, Dementia Research Institute, Cardiff University¸ Cardiff, CF24 4HQ, UK
| | - Natalie Connor-Robson
- Cardiff University, Dementia Research Institute, Cardiff University¸ Cardiff, CF24 4HQ, UK
| |
Collapse
|
9
|
Nolte DD. Coherent light scattering from cellular dynamics in living tissues. REPORTS ON PROGRESS IN PHYSICS. PHYSICAL SOCIETY (GREAT BRITAIN) 2024; 87:036601. [PMID: 38433567 DOI: 10.1088/1361-6633/ad2229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 01/24/2024] [Indexed: 03/05/2024]
Abstract
This review examines the biological physics of intracellular transport probed by the coherent optics of dynamic light scattering from optically thick living tissues. Cells and their constituents are in constant motion, composed of a broad range of speeds spanning many orders of magnitude that reflect the wide array of functions and mechanisms that maintain cellular health. From the organelle scale of tens of nanometers and upward in size, the motion inside living tissue is actively driven rather than thermal, propelled by the hydrolysis of bioenergetic molecules and the forces of molecular motors. Active transport can mimic the random walks of thermal Brownian motion, but mean-squared displacements are far from thermal equilibrium and can display anomalous diffusion through Lévy or fractional Brownian walks. Despite the average isotropic three-dimensional environment of cells and tissues, active cellular or intracellular transport of single light-scattering objects is often pseudo-one-dimensional, for instance as organelle displacement persists along cytoskeletal tracks or as membranes displace along the normal to cell surfaces, albeit isotropically oriented in three dimensions. Coherent light scattering is a natural tool to characterize such tissue dynamics because persistent directed transport induces Doppler shifts in the scattered light. The many frequency-shifted partial waves from the complex and dynamic media interfere to produce dynamic speckle that reveals tissue-scale processes through speckle contrast imaging and fluctuation spectroscopy. Low-coherence interferometry, dynamic optical coherence tomography, diffusing-wave spectroscopy, diffuse-correlation spectroscopy, differential dynamic microscopy and digital holography offer coherent detection methods that shed light on intracellular processes. In health-care applications, altered states of cellular health and disease display altered cellular motions that imprint on the statistical fluctuations of the scattered light. For instance, the efficacy of medical therapeutics can be monitored by measuring the changes they induce in the Doppler spectra of livingex vivocancer biopsies.
Collapse
Affiliation(s)
- David D Nolte
- Department of Physics and Astronomy, Purdue University, West Lafayette, IN 47907, United States of America
| |
Collapse
|
10
|
Vizcaíno-Castillo A, Kotila T, Kogan K, Yanase R, Como J, Antenucci L, Michelot A, Sunter JD, Lappalainen P. Leishmania profilin interacts with actin through an unusual structural mechanism to control cytoskeletal dynamics in parasites. J Biol Chem 2024; 300:105740. [PMID: 38340794 PMCID: PMC10907219 DOI: 10.1016/j.jbc.2024.105740] [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: 10/20/2023] [Revised: 02/02/2024] [Accepted: 02/05/2024] [Indexed: 02/12/2024] Open
Abstract
Diseases caused by Leishmania and Trypanosoma parasites are a major health problem in tropical countries. Because of their complex life cycle involving both vertebrate and insect hosts, and >1 billion years of evolutionarily distance, the cell biology of trypanosomatid parasites exhibits pronounced differences to animal cells. For example, the actin cytoskeleton of trypanosomatids is divergent when compared with other eukaryotes. To understand how actin dynamics are regulated in trypanosomatid parasites, we focused on a central actin-binding protein profilin. Co-crystal structure of Leishmania major actin in complex with L. major profilin revealed that, although the overall folds of actin and profilin are conserved in eukaryotes, Leishmania profilin contains a unique α-helical insertion, which interacts with the target binding cleft of actin monomer. This insertion is conserved across the Trypanosomatidae family and is similar to the structure of WASP homology-2 (WH2) domain, a small actin-binding motif found in many other cytoskeletal regulators. The WH2-like motif contributes to actin monomer binding and enhances the actin nucleotide exchange activity of Leishmania profilin. Moreover, Leishmania profilin inhibited formin-catalyzed actin filament assembly in a mechanism that is dependent on the presence of the WH2-like motif. By generating profilin knockout and knockin Leishmania mexicana strains, we show that profilin is important for efficient endocytic sorting in parasites, and that the ability to bind actin monomers and proline-rich proteins, and the presence of a functional WH2-like motif, are important for the in vivo function of Leishmania profilin. Collectively, this study uncovers molecular principles by which profilin regulates actin dynamics in trypanosomatids.
Collapse
Affiliation(s)
| | - Tommi Kotila
- HiLIFE Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Konstantin Kogan
- HiLIFE Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Ryuji Yanase
- Oxford Brookes University, Department of Biological and Medical Sciences, Oxford, UK
| | - Juna Como
- Aix Marseille University, CNRS, IBDM, Turing Centre for Living Systems, Marseille, France
| | - Lina Antenucci
- HiLIFE Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Alphee Michelot
- Aix Marseille University, CNRS, IBDM, Turing Centre for Living Systems, Marseille, France
| | - Jack D Sunter
- Oxford Brookes University, Department of Biological and Medical Sciences, Oxford, UK.
| | - Pekka Lappalainen
- HiLIFE Institute of Biotechnology, University of Helsinki, Helsinki, Finland; Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
11
|
Meyer T, Knittelfelder O, Smolnig M, Rockenfeller P. Quantifying yeast lipidomics by high-performance thin-layer chromatography (HPTLC) and comparison to mass spectrometry-based shotgun lipidomics. MICROBIAL CELL (GRAZ, AUSTRIA) 2024; 11:57-68. [PMID: 38384676 PMCID: PMC10879857 DOI: 10.15698/mic2024.02.815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 01/23/2024] [Accepted: 01/29/2024] [Indexed: 02/23/2024]
Abstract
Lipidomic analysis in diverse biological settings has become a frequent tool to increase our understanding of the processes of life. Cellular lipids play important roles not only as being the main components of cellular membranes, but also in the regulation of cell homeostasis as lipid signaling molecules. Yeast has been harnessed for biomedical research based on its good conservation of genetics and fundamental cell organisation principles and molecular pathways. Further application in so-called humanised yeast models have been developed which take advantage of yeast as providing the basics of a living cell with full control over heterologous expression. Here we present evidence that high-performance thin-layer chromatography (HPTLC) represents an effective alternative to replace cost intensive mass spectrometry-based lipidomic analyses. We provide statistical comparison of identical samples by both methods, which support the use of HPTLC for quantitative analysis of the main yeast lipid classes.
Collapse
Affiliation(s)
- Thorsten Meyer
- Chair of Biochemistry and Molecular Medicine, Center for Biomedical Education and Research (ZBAF), University of Witten/Herdecke (UW/H), Stockumer Str. 10, 58453 Witten, Germany
| | - Oskar Knittelfelder
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| | - Martin Smolnig
- Chair of Biochemistry and Molecular Medicine, Center for Biomedical Education and Research (ZBAF), University of Witten/Herdecke (UW/H), Stockumer Str. 10, 58453 Witten, Germany
| | - Patrick Rockenfeller
- Chair of Biochemistry and Molecular Medicine, Center for Biomedical Education and Research (ZBAF), University of Witten/Herdecke (UW/H), Stockumer Str. 10, 58453 Witten, Germany
| |
Collapse
|
12
|
Yu Y, Chen D, Farmer SM, Xu S, Rios B, Solbach A, Ye X, Ye L, Zhang S. Endolysosomal trafficking controls yolk granule biogenesis in vitellogenic Drosophila oocytes. PLoS Genet 2024; 20:e1011152. [PMID: 38315726 PMCID: PMC10898735 DOI: 10.1371/journal.pgen.1011152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 02/27/2024] [Accepted: 01/22/2024] [Indexed: 02/07/2024] Open
Abstract
Endocytosis and endolysosomal trafficking are essential for almost all aspects of physiological functions of eukaryotic cells. As our understanding on these membrane trafficking events are mostly from studies in yeast and cultured mammalian cells, one challenge is to systematically evaluate the findings from these cell-based studies in multicellular organisms under physiological settings. One potentially valuable in vivo system to address this challenge is the vitellogenic oocyte in Drosophila, which undergoes extensive endocytosis by Yolkless (Yl), a low-density lipoprotein receptor (LDLR), to uptake extracellular lipoproteins into oocytes and package them into a specialized lysosome, the yolk granule, for storage and usage during later development. However, by now there is still a lack of sufficient understanding on the molecular and cellular processes that control yolk granule biogenesis. Here, by creating genome-tagging lines for Yl receptor and analyzing its distribution in vitellogenic oocytes, we observed a close association of different endosomal structures with distinct phosphoinositides and actin cytoskeleton dynamics. We further showed that Rab5 and Rab11, but surprisingly not Rab4 and Rab7, are essential for yolk granules biogenesis. Instead, we uncovered evidence for a potential role of Rab7 in actin regulation and observed a notable overlap of Rab4 and Rab7, two Rab GTPases that have long been proposed to have distinct spatial distribution and functional roles during endolysosomal trafficking. Through a small-scale RNA interference (RNAi) screen on a set of reported Rab5 effectors, we showed that yolk granule biogenesis largely follows the canonical endolysosomal trafficking and maturation processes. Further, the data suggest that the RAVE/V-ATPase complexes function upstream of or in parallel with Rab7, and are involved in earlier stages of endosomal trafficking events. Together, our study provides s novel insights into endolysosomal pathways and establishes vitellogenic oocyte in Drosophila as an excellent in vivo model for dissecting the highly complex membrane trafficking events in metazoan.
Collapse
Affiliation(s)
- Yue Yu
- The Brown Foundation Institute of Molecular Medicine, McGovern Medical School at the University of Texas Health Science Center at Houston (UTHealth), Houston, Texas, United States of America
- Program in Neuroscience, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences (MD Anderson UTHealth GSBS), Houston, Texas, United States of America
| | - Dongsheng Chen
- The Brown Foundation Institute of Molecular Medicine, McGovern Medical School at the University of Texas Health Science Center at Houston (UTHealth), Houston, Texas, United States of America
- The College of Life Sciences, Anhui Normal University, #1 Beijing East Road, Wuhu, Anhui, People’s Republic of China
| | - Stephen M. Farmer
- The Brown Foundation Institute of Molecular Medicine, McGovern Medical School at the University of Texas Health Science Center at Houston (UTHealth), Houston, Texas, United States of America
- Program in Neuroscience, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences (MD Anderson UTHealth GSBS), Houston, Texas, United States of America
- Program in Biochemistry and Cell Biology, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences (MD Anderson UTHealth GSBS), Houston, Texas, United States of America
| | - Shiyu Xu
- The Brown Foundation Institute of Molecular Medicine, McGovern Medical School at the University of Texas Health Science Center at Houston (UTHealth), Houston, Texas, United States of America
| | - Beatriz Rios
- The Brown Foundation Institute of Molecular Medicine, McGovern Medical School at the University of Texas Health Science Center at Houston (UTHealth), Houston, Texas, United States of America
- Program in Neuroscience, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences (MD Anderson UTHealth GSBS), Houston, Texas, United States of America
| | - Amanda Solbach
- The Brown Foundation Institute of Molecular Medicine, McGovern Medical School at the University of Texas Health Science Center at Houston (UTHealth), Houston, Texas, United States of America
- Program in Neuroscience, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences (MD Anderson UTHealth GSBS), Houston, Texas, United States of America
- Programs in Genetics and Epigenetics, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences (MD Anderson UTHealth GSBS), Houston, Texas, United States of America
| | - Xin Ye
- The Brown Foundation Institute of Molecular Medicine, McGovern Medical School at the University of Texas Health Science Center at Houston (UTHealth), Houston, Texas, United States of America
| | - Lili Ye
- The Brown Foundation Institute of Molecular Medicine, McGovern Medical School at the University of Texas Health Science Center at Houston (UTHealth), Houston, Texas, United States of America
| | - Sheng Zhang
- The Brown Foundation Institute of Molecular Medicine, McGovern Medical School at the University of Texas Health Science Center at Houston (UTHealth), Houston, Texas, United States of America
- Program in Neuroscience, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences (MD Anderson UTHealth GSBS), Houston, Texas, United States of America
- Programs in Genetics and Epigenetics, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences (MD Anderson UTHealth GSBS), Houston, Texas, United States of America
- Department of Neurobiology and Anatomy, McGovern Medical School at the University of Texas Health Science Center at Houston (UTHealth), Houston, Texas, United States of America
| |
Collapse
|
13
|
Lin Z, Mao Z, Ma R. Inferring biophysical properties of membranes during endocytosis using machine learning. SOFT MATTER 2024; 20:651-660. [PMID: 38164011 DOI: 10.1039/d3sm01221b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2024]
Abstract
Endocytosis is a fundamental cellular process in eukaryotic cells that facilitates the transport of molecules into the cell. With the help of fluorescence microscopy and electron tomography, researchers have accumulated extensive geometric data of membrane shapes during endocytosis. These data contain rich information about the mechanical properties of membranes, which are hard to access via experiments due to the small dimensions of the endocytic patch. In this study, we propose an approach that combines machine learning with the Helfrich theory of membranes to infer the mechanical properties of membranes during endocytosis from a dataset of membrane shapes extracted from electron tomography. Our results demonstrate that machine learning can output solutions that both match the experimental profile and satisfy the membrane shape equations derived from Helfrich theory. The learning results show that during the early stage of endocytosis, the inferred membrane tension is negative, indicating the presence of strong compressive forces at the boundary of the endocytic invagination. Our method presents a generic framework for extracting membrane information from super-resolution imaging.
Collapse
Affiliation(s)
- Zhiwei Lin
- Department of Physics, College of Physical Science and Technology, Xiamen University, Xiamen 361005, China.
| | - Zhiping Mao
- School of Mathematical Sciences, Fujian Provincial Key Laboratory of Mathematical Modeling and High-Performance Scientific Computing, Xiamen University, Xiamen 361005, China.
| | - Rui Ma
- Department of Physics, College of Physical Science and Technology, Xiamen University, Xiamen 361005, China.
- Fujian Provincial Key Laboratory for Soft Functional Materials Research, Research Institute for Biomimetics and Soft Matter, Xiamen University, Xiamen 361005, China
| |
Collapse
|
14
|
Heydecker M, Shitara A, Chen D, Tran D, Masedunskas A, Tora M, Ebrahim S, Appaduray MA, Galeano Niño JL, Bhardwaj A, Narayan K, Hardeman EC, Gunning PW, Weigert R. Spatial and Temporal Coordination of Force-generating Actin-based Modules Drives Membrane Remodeling In Vivo. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.04.569944. [PMID: 38168275 PMCID: PMC10760165 DOI: 10.1101/2023.12.04.569944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Membrane remodeling drives a broad spectrum of cellular functions, and it is regulated through mechanical forces exerted on the membrane by cytoplasmic complexes. Here, we investigate how actin filaments dynamically tune their structure to control the active transfer of membranes between cellular compartments with distinct compositions and biophysical properties. Using intravital subcellular microscopy in live rodents we show that: a lattice composed of linear filaments stabilizes the granule membrane after fusion with the plasma membrane; and a network of branched filaments linked to the membranes by Ezrin, a regulator of membrane tension, initiates and drives to completion the integration step. Our results highlight how the actin cytoskeleton tunes its structure to adapt to dynamic changes in the biophysical properties of membranes.
Collapse
|
15
|
Liu Q, Liu M, Yang T, Wang X, Cheng P, Zhou H. What can we do to optimize mitochondrial transplantation therapy for myocardial ischemia-reperfusion injury? Mitochondrion 2023; 72:72-83. [PMID: 37549815 DOI: 10.1016/j.mito.2023.08.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 05/20/2023] [Accepted: 08/04/2023] [Indexed: 08/09/2023]
Abstract
Mitochondrial transplantation is a promising solution for the heart following ischemia-reperfusion injury due to its capacity to replace damaged mitochondria and restore cardiac function. However, many barriers (such as inadequate mitochondrial internalization, poor survival of transplanted mitochondria, few mitochondria colocalized with cardiac cells) compromise the replacement of injured mitochondria with transplanted mitochondria. Therefore, it is necessary to optimize mitochondrial transplantation therapy to improve clinical effectiveness. By analogy, myocardial ischemia-reperfusion injury is like a withered flower, it needs to absorb enough nutrients to recover and bloom. In this review, we present a comprehensive overview of "nutrients" (source of exogenous mitochondria and different techniques for mitochondrial isolation), "absorption" (mitochondrial transplantation approaches, mitochondrial transplantation dose and internalization mechanism), and "flowering" (the mechanism of mitochondrial transplantation in cardioprotection) for myocardial ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Qian Liu
- Institute of Cardiovascular Disease of Integrated Traditional Chinese Medicine and Western Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Meng Liu
- Comprehensive treatment area of Traditional Chinese Medicine, Guanghua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Tianshu Yang
- Institute of Cardiovascular Disease of Integrated Traditional Chinese Medicine and Western Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xinting Wang
- Institute of Cardiovascular Disease of Integrated Traditional Chinese Medicine and Western Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Peipei Cheng
- Institute of Cardiovascular Disease of Integrated Traditional Chinese Medicine and Western Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hua Zhou
- Institute of Cardiovascular Disease of Integrated Traditional Chinese Medicine and Western Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
16
|
Madarász T, Brunner B, Halász H, Telek E, Matkó J, Nyitrai M, Szabó-Meleg E. Molecular Relay Stations in Membrane Nanotubes: IRSp53 Involved in Actin-Based Force Generation. Int J Mol Sci 2023; 24:13112. [PMID: 37685917 PMCID: PMC10487789 DOI: 10.3390/ijms241713112] [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: 07/04/2023] [Revised: 07/28/2023] [Accepted: 08/12/2023] [Indexed: 09/10/2023] Open
Abstract
Membrane nanotubes are cell protrusions that grow to tens of micrometres and functionally connect cells. Actin filaments are semi-flexible polymers, and their polymerisation provides force for the formation and growth of membrane nanotubes. The molecular bases for the provision of appropriate force through such long distances are not yet clear. Actin filament bundles are likely involved in these processes; however, even actin bundles weaken when growing over long distances, and there must be a mechanism for their regeneration along the nanotubes. We investigated the possibility of the formation of periodic molecular relay stations along membrane nanotubes by describing the interactions of actin with full-length IRSp53 protein and its N-terminal I-BAR domain. We concluded that I-BAR is involved in the early phase of the formation of cell projections, while IRSp53 is also important for the elongation of protrusions. Considering that IRSp53 binds to the membrane along the nanotubes and nucleates actin polymerisation, we propose that, in membrane nanotubes, IRSp53 establishes molecular relay stations for actin polymerisation and, as a result, supports the generation of force required for the growth of nanotubes.
Collapse
Affiliation(s)
- Tamás Madarász
- Department of Biophysics, Medical School, University of Pécs, H-7624 Pécs, Hungary
| | - Brigitta Brunner
- Institute of Biology, Faculty of Sciences, University of Pécs, H-7624 Pécs, Hungary
| | - Henriett Halász
- Department of Biophysics, Medical School, University of Pécs, H-7624 Pécs, Hungary
| | - Elek Telek
- Department of Biophysics, Medical School, University of Pécs, H-7624 Pécs, Hungary
| | - János Matkó
- Department of Immunology, Faculty of Science, Eötvös Loránd University, H-1117 Budapest, Hungary
| | - Miklós Nyitrai
- Department of Biophysics, Medical School, University of Pécs, H-7624 Pécs, Hungary
| | - Edina Szabó-Meleg
- Department of Biophysics, Medical School, University of Pécs, H-7624 Pécs, Hungary
| |
Collapse
|
17
|
Baschieri F, Illand A, Barbazan J, Zajac O, Henon C, Loew D, Dingli F, Vignjevic DM, Lévêque-Fort S, Montagnac G. Fibroblasts generate topographical cues that steer cancer cell migration. SCIENCE ADVANCES 2023; 9:eade2120. [PMID: 37585527 PMCID: PMC10431708 DOI: 10.1126/sciadv.ade2120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 07/14/2023] [Indexed: 08/18/2023]
Abstract
Fibroblasts play a fundamental role in tumor development. Among other functions, they regulate cancer cells' migration through rearranging the extracellular matrix, secreting soluble factors, and establishing direct physical contacts with cancer cells. Here, we report that migrating fibroblasts deposit on the substrate a network of tubular structures that serves as a guidance cue for cancer cell migration. Such membranous tubular network, hereafter called tracks, is stably anchored to the substrate in a β5-integrin-dependent manner. We found that cancer cells specifically adhere to tracks by using clathrin-coated structures that pinch and engulf tracks. Tracks thus represent a spatial memory of fibroblast migration paths that is read and erased by cancer cells directionally migrating along them. We propose that fibroblast tracks represent a topography-based intercellular communication system capable of steering cancer cell migration.
Collapse
Affiliation(s)
- Francesco Baschieri
- Inserm U1279, Gustave Roussy Institute, Université Paris-Saclay, Villejuif, France
| | - Abigail Illand
- Université Paris Saclay, CNRS, Institut des sciences moléculaires d’Orsay, UMR8214, Orsay, France
| | - Jorge Barbazan
- Translational Medical Oncology Group (ONCOMET), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Olivier Zajac
- Institut Curie, UMR144, PSL Research University, Centre Universitaire, Paris, France
| | - Clémence Henon
- Inserm U981, Gustave Roussy Institute, Université Paris-Saclay, Villejuif, France
| | - Damarys Loew
- Institut Curie, PSL Research University, Centre de Recherche, Laboratoire de Spectrométrie de Masse Protéomique, Paris, France
| | - Florent Dingli
- Institut Curie, PSL Research University, Centre de Recherche, Laboratoire de Spectrométrie de Masse Protéomique, Paris, France
| | | | - Sandrine Lévêque-Fort
- Université Paris Saclay, CNRS, Institut des sciences moléculaires d’Orsay, UMR8214, Orsay, France
| | - Guillaume Montagnac
- Inserm U1279, Gustave Roussy Institute, Université Paris-Saclay, Villejuif, France
| |
Collapse
|
18
|
Liashkovich I, Stefanello ST, Vidyadharan R, Haufe G, Erofeev A, Gorelkin PV, Kolmogorov V, Mizdal CR, Dulebo A, Bulk E, Kouzel IU, Shahin V. Pitstop-2 and its novel derivative RVD-127 disrupt global cell dynamics and nuclear pores integrity by direct interaction with small GTPases. Bioeng Transl Med 2023; 8:e10425. [PMID: 37476059 PMCID: PMC10354767 DOI: 10.1002/btm2.10425] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 09/26/2022] [Accepted: 10/03/2022] [Indexed: 07/22/2023] Open
Abstract
Clathrin-mediated endocytosis (CME) is an essential cell physiological process of broad biomedical relevance. Since the recent introduction of Pitstop-2 as a potent CME inhibitor, we and others have reported on substantial clathrin-independent inhibitory effects. Herein, we developed and experimentally validated a novel fluorescent derivative of Pitstop-2, termed RVD-127, to clarify Pitstop-2 diverse effects. Using RVD-127, we were able to trace additional protein targets of Pitstop-2. Besides inhibiting CME, Pitstop-2 and RVD-127 proved to directly and reversibly bind to at least two members of the small GTPase superfamily Ran and Rac1 with particularly high efficacy. Binding locks the GTPases in a guanosine diphosphate (GDP)-like conformation disabling their interaction with their downstream effectors. Consequently, overall cell motility, mechanics and nucleocytoplasmic transport integrity are rapidly disrupted at inhibitor concentrations well below those required to significantly reduce CME. We conclude that Pitstop-2 is a highly potent, reversible inhibitor of small GTPases. The inhibition of these molecular switches of diverse crucial signaling pathways, including nucleocytoplasmic transport and overall cell dynamics and motility, clarifies the diversity of Pitstop-2 activities. Moreover, considering the fundamental importance and broad implications of small GTPases in physiology, pathophysiology and drug development, Pitstop-2 and RVD-127 open up novel avenues.
Collapse
Affiliation(s)
| | | | | | - Günter Haufe
- Organic Chemistry Institute, University of MünsterMünsterGermany
| | - Alexander Erofeev
- National University of Science and Technology «MISiS»MoscowRussia
- Department of ChemistryLomonosov Moscow State UniversityMoscowRussia
| | | | | | | | | | - Etmar Bulk
- Institute of Physiology II, University of MünsterMünsterGermany
| | | | - Victor Shahin
- Institute of Physiology II, University of MünsterMünsterGermany
| |
Collapse
|
19
|
Benz PM, Frömel T, Laban H, Zink J, Ulrich L, Groneberg D, Boon RA, Poley P, Renne T, de Wit C, Fleming I. Cardiovascular Functions of Ena/VASP Proteins: Past, Present and Beyond. Cells 2023; 12:1740. [PMID: 37443774 PMCID: PMC10340426 DOI: 10.3390/cells12131740] [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: 04/26/2023] [Revised: 06/18/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
Actin binding proteins are of crucial importance for the spatiotemporal regulation of actin cytoskeletal dynamics, thereby mediating a tremendous range of cellular processes. Since their initial discovery more than 30 years ago, the enabled/vasodilator-stimulated phosphoprotein (Ena/VASP) family has evolved as one of the most fascinating and versatile family of actin regulating proteins. The proteins directly enhance actin filament assembly, but they also organize higher order actin networks and link kinase signaling pathways to actin filament assembly. Thereby, Ena/VASP proteins regulate dynamic cellular processes ranging from membrane protrusions and trafficking, and cell-cell and cell-matrix adhesions, to the generation of mechanical tension and contractile force. Important insights have been gained into the physiological functions of Ena/VASP proteins in platelets, leukocytes, endothelial cells, smooth muscle cells and cardiomyocytes. In this review, we summarize the unique and redundant functions of Ena/VASP proteins in cardiovascular cells and discuss the underlying molecular mechanisms.
Collapse
Affiliation(s)
- Peter M. Benz
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, 60596 Frankfurt am Main, Germany
- German Centre of Cardiovascular Research (DZHK), Partner Site Rhein-Main, 60596 Frankfurt am Main, Germany
| | - Timo Frömel
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, 60596 Frankfurt am Main, Germany
| | - Hebatullah Laban
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, 60596 Frankfurt am Main, Germany
| | - Joana Zink
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, 60596 Frankfurt am Main, Germany
| | - Lea Ulrich
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, 60596 Frankfurt am Main, Germany
| | - Dieter Groneberg
- Institute of Physiology I, University of Würzburg, 97070 Würzburg, Germany
| | - Reinier A. Boon
- German Centre of Cardiovascular Research (DZHK), Partner Site Rhein-Main, 60596 Frankfurt am Main, Germany
- Cardiopulmonary Institute, 60596 Frankfurt am Main, Germany
- Centre of Molecular Medicine, Institute of Cardiovascular Regeneration, Goethe-University, 60596 Frankfurt am Main, Germany
- Department of Physiology, Amsterdam Cardiovascular Sciences, VU University Medical Centre, 1081 HZ Amsterdam, The Netherlands
| | - Philip Poley
- Institut für Physiologie, Universität zu Lübeck, 23562 Lübeck, Germany
- German Centre of Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 23562 Lübeck, Germany
| | - Thomas Renne
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- Center for Thrombosis and Hemostasis (CTH), Johannes Gutenberg University Medical Center, 55131 Mainz, Germany
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, D02 VN51 Dublin, Ireland
| | - Cor de Wit
- Institut für Physiologie, Universität zu Lübeck, 23562 Lübeck, Germany
- German Centre of Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 23562 Lübeck, Germany
| | - Ingrid Fleming
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, 60596 Frankfurt am Main, Germany
- German Centre of Cardiovascular Research (DZHK), Partner Site Rhein-Main, 60596 Frankfurt am Main, Germany
- Cardiopulmonary Institute, 60596 Frankfurt am Main, Germany
| |
Collapse
|
20
|
Stoops EH, Ferrin MA, Jorgens DM, Drubin DG. Self-organizing actin networks drive sequential endocytic protein recruitment and vesicle release on synthetic lipid bilayers. Proc Natl Acad Sci U S A 2023; 120:e2302622120. [PMID: 37216532 PMCID: PMC10235984 DOI: 10.1073/pnas.2302622120] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 04/21/2023] [Indexed: 05/24/2023] Open
Abstract
Forces generated by actin assembly assist membrane invagination during clathrin-mediated endocytosis (CME). The sequential recruitment of core endocytic proteins and regulatory proteins, and assembly of the actin network, are well documented in live cells and are highly conserved from yeasts to humans. However, understanding of CME protein self-organization, as well as the biochemical and mechanical principles that underlie actin's role in CME, is lacking. Here, we show that supported lipid bilayers coated with purified yeast Wiskott Aldrich Syndrome Protein (WASP), an endocytic actin assembly regulator, and incubated in cytoplasmic yeast extracts, recruit downstream endocytic proteins and assemble actin networks. Time-lapse imaging of WASP-coated bilayers revealed sequential recruitment of proteins from different endocytic modules, faithfully replicating in vivo behavior. Reconstituted actin networks assemble in a WASP-dependent manner and deform lipid bilayers, as seen by electron microscopy. Time-lapse imaging revealed that vesicles are released from the lipid bilayers with a burst of actin assembly. Actin networks pushing on membranes have previously been reconstituted; here, we have reconstituted a biologically important variation of these actin networks that self-organize on bilayers and produce pulling forces sufficient to bud off membrane vesicles. We propose that actin-driven vesicle generation may represent an ancient evolutionary precursor to diverse vesicle forming processes adapted for a wide array of cellular environments and applications.
Collapse
Affiliation(s)
- Emily H. Stoops
- Department of Molecular and Cell Biology, University of California, Berkeley, CA94720
| | - Michael A. Ferrin
- Department of Molecular and Cell Biology, University of California, Berkeley, CA94720
| | | | - David G. Drubin
- Department of Molecular and Cell Biology, University of California, Berkeley, CA94720
| |
Collapse
|
21
|
Porello I, Cellesi F. Intracellular delivery of therapeutic proteins. New advancements and future directions. Front Bioeng Biotechnol 2023; 11:1211798. [PMID: 37304137 PMCID: PMC10247999 DOI: 10.3389/fbioe.2023.1211798] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 05/16/2023] [Indexed: 06/13/2023] Open
Abstract
Achieving the full potential of therapeutic proteins to access and target intracellular receptors will have enormous benefits in advancing human health and fighting disease. Existing strategies for intracellular protein delivery, such as chemical modification and nanocarrier-based protein delivery approaches, have shown promise but with limited efficiency and safety concerns. The development of more effective and versatile delivery tools is crucial for the safe and effective use of protein drugs. Nanosystems that can trigger endocytosis and endosomal disruption, or directly deliver proteins into the cytosol, are essential for successful therapeutic effects. This article aims to provide a brief overview of the current methods for intracellular protein delivery to mammalian cells, highlighting current challenges, new developments, and future research opportunities.
Collapse
|
22
|
Shang P, Simpson JD, Taylor GM, Sutherland DM, Welsh OL, Aravamudhan P, Natividade RDS, Schwab K, Michel JJ, Poholek AC, Wu Y, Rajasundaram D, Koehler M, Alsteens D, Dermody TS. Paired immunoglobulin-like receptor B is an entry receptor for mammalian orthoreovirus. Nat Commun 2023; 14:2615. [PMID: 37147336 PMCID: PMC10163058 DOI: 10.1038/s41467-023-38327-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 04/25/2023] [Indexed: 05/07/2023] Open
Abstract
Mammalian orthoreovirus (reovirus) infects most mammals and is associated with celiac disease in humans. In mice, reovirus infects the intestine and disseminates systemically to cause serotype-specific patterns of disease in the brain. To identify receptors conferring reovirus serotype-dependent neuropathogenesis, we conducted a genome-wide CRISPRa screen and identified paired immunoglobulin-like receptor B (PirB) as a receptor candidate. Ectopic expression of PirB allowed reovirus binding and infection. PirB extracelluar D3D4 region is required for reovirus attachment and infectivity. Reovirus binds to PirB with nM affinity as determined by single molecule force spectroscopy. Efficient reovirus endocytosis requires PirB signaling motifs. In inoculated mice, PirB is required for maximal replication in the brain and full neuropathogenicity of neurotropic serotype 3 (T3) reovirus. In primary cortical neurons, PirB expression contributes to T3 reovirus infectivity. Thus, PirB is an entry receptor for reovirus and contributes to T3 reovirus replication and pathogenesis in the murine brain.
Collapse
Affiliation(s)
- Pengcheng Shang
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Institute of Infection, Inflammation, and Immunity, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Joshua D Simpson
- Louvain Institute of Biomolecular Science and Technology, Université catholique de Louvain, Louvain-la-Neuve, Belgium
| | - Gwen M Taylor
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Institute of Infection, Inflammation, and Immunity, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Danica M Sutherland
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Institute of Infection, Inflammation, and Immunity, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Olivia L Welsh
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Institute of Infection, Inflammation, and Immunity, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Pavithra Aravamudhan
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Institute of Infection, Inflammation, and Immunity, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Rita Dos Santos Natividade
- Louvain Institute of Biomolecular Science and Technology, Université catholique de Louvain, Louvain-la-Neuve, Belgium
| | - Kristina Schwab
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Joshua J Michel
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Amanda C Poholek
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Yijen Wu
- Department of Developmental Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Dhivyaa Rajasundaram
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Melanie Koehler
- Leibniz Institute for Food Systems Biology at the Technical University Munich, Freising, Germany
| | - David Alsteens
- Louvain Institute of Biomolecular Science and Technology, Université catholique de Louvain, Louvain-la-Neuve, Belgium
- WELBIO Department, WEL Research Institute, Wavre, Belgium
| | - Terence S Dermody
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
- Institute of Infection, Inflammation, and Immunity, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA.
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
23
|
Su H, Xu J, Li J, Yi Z. Four ciliate-specific expansion events occurred during actin gene family evolution of eukaryotes. Mol Phylogenet Evol 2023; 184:107789. [PMID: 37105243 DOI: 10.1016/j.ympev.2023.107789] [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: 01/20/2023] [Revised: 03/21/2023] [Accepted: 04/14/2023] [Indexed: 04/29/2023]
Abstract
Actin gene family is a divergent and ancient eukaryotic cellular cytoskeletal gene family, and participates in many essential cellular processes. Ciliated protists offer us an excellent opportunity to investigate gene family evolution, since their gene families evolved faster in ciliates than in other eukaryotes. Nonetheless, actin gene family is well studied in few model ciliate species but little is known about its evolutionary patterns in ciliates. Here, we analyzed the evolutionary pattern of eukaryotic actin gene family based on genomes/transcriptomes of 36 species covering ten ciliate classes, as well as those of nine non-ciliate eukaryotic species. Results showed: (1) Except for conventional actins and actin-related proteins (Arps) shared by various eukaryotes, at least four ciliate-specific subfamilies occurred during evolution of actin gene family. Expansions of Act2 and ArpC were supposed to have happen in the ciliate common ancestor, while expansions of ActI and ActII may have occurred in the ancestor of Armophorea, Muranotrichea, and Spirotrichea. (2) The number of actin isoforms varied greatly among ciliate species. Environmental adaptability, whole genome duplication (WGD) or segmental duplication events, distinct spatial and temporal patterns of expression might play driving forces for the increasement of isoform numbers. (3) The 'birth and death' model of evolution could explain the evolution of actin gene family in ciliates. And actin genes have been generally under strong negative selection to maintain protein structures and physiological functions. Collectively, we provided meaningful information for understanding the evolution of eukaryotic actin gene family.
Collapse
Affiliation(s)
- Hua Su
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, School of Life Science, South China Normal University, Guangzhou 510631, China
| | - Jiahui Xu
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, School of Life Science, South China Normal University, Guangzhou 510631, China
| | - Jia Li
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, School of Life Science, South China Normal University, Guangzhou 510631, China
| | - Zhenzhen Yi
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, School of Life Science, South China Normal University, Guangzhou 510631, China.
| |
Collapse
|
24
|
Rayens NT, Cook KJ, McKinley SA, Payne CK. Palmitate-mediated disruption of the endoplasmic reticulum decreases intracellular vesicle motility. Biophys J 2023; 122:1355-1363. [PMID: 36869590 PMCID: PMC10111363 DOI: 10.1016/j.bpj.2023.03.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 02/02/2023] [Accepted: 02/28/2023] [Indexed: 03/05/2023] Open
Abstract
Essential cellular processes such as metabolism, protein synthesis, and autophagy require the intracellular transport of membrane-bound vesicles. The importance of the cytoskeleton and associated molecular motors for transport is well documented. Recent research has suggested that the endoplasmic reticulum (ER) may also play a role in vesicle transport through a tethering of vesicles to the ER. We use single-particle tracking fluorescence microscopy and a Bayesian change-point algorithm to characterize vesicle motility in response to the disruption of the ER, actin, and microtubules. This high-throughput change-point algorithm allows us to efficiently analyze thousands of trajectory segments. We find that palmitate-mediated disruption of the ER leads to a significant decrease in vesicle motility. A comparison with the disruption of actin and microtubules shows that disruption of the ER has a significant impact on vesicle motility, greater than the disruption of actin. Vesicle motility was dependent on cellular region, with greater motility in the cell periphery than the perinuclear region, possibly due to regional differences in actin and the ER. Overall, these results suggest that the ER is an important factor in vesicle transport.
Collapse
Affiliation(s)
- Nathan T Rayens
- Thomas Lord Department of Mechanical Engineering and Materials Science, Duke University, Durham, North Carolina
| | - Keisha J Cook
- School of Mathematical and Statistical Sciences, Clemson University, Clemson, South Carolina
| | - Scott A McKinley
- Department of Mathematics, Tulane University, New Orleans, Louisiana
| | - Christine K Payne
- Thomas Lord Department of Mechanical Engineering and Materials Science, Duke University, Durham, North Carolina.
| |
Collapse
|
25
|
Joseph BB, Naslavsky N, Binti S, Conquest S, Robison L, Bai G, Homer RO, Grant BD, Caplan S, Fay DS. Conserved NIMA kinases regulate multiple steps of endocytic trafficking. PLoS Genet 2023; 19:e1010741. [PMID: 37099601 PMCID: PMC10166553 DOI: 10.1371/journal.pgen.1010741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 05/08/2023] [Accepted: 04/11/2023] [Indexed: 04/27/2023] Open
Abstract
Human NIMA-related kinases have primarily been studied for their roles in cell cycle progression (NEK1/2/6/7/9), checkpoint-DNA-damage control (NEK1/2/4/5/10/11), and ciliogenesis (NEK1/4/8). We previously showed that Caenorhabditis elegans NEKL-2 (NEK8/9 homolog) and NEKL-3 (NEK6/7 homolog) regulate apical clathrin-mediated endocytosis (CME) in the worm epidermis and are essential for molting. Here we show that NEKL-2 and NEKL-3 also have distinct roles in controlling endosome function and morphology. Specifically, loss of NEKL-2 led to enlarged early endosomes with long tubular extensions but showed minimal effects on other compartments. In contrast, NEKL-3 depletion caused pronounced defects in early, late, and recycling endosomes. Consistently, NEKL-2 was strongly localized to early endosomes, whereas NEKL-3 was localized to multiple endosomal compartments. Loss of NEKLs also led to variable defects in the recycling of two resident cargoes of the trans-Golgi network (TGN), MIG-14/Wntless and TGN-38/TGN38, which were missorted to lysosomes after NEKL depletion. In addition, defects were observed in the uptake of clathrin-dependent (SMA-6/Type I BMP receptor) and independent cargoes (DAF-4/Type II BMP receptor) from the basolateral surface of epidermal cells after NEKL-2 or NEKL-3 depletion. Complementary studies in human cell lines further showed that siRNA knockdown of the NEKL-3 orthologs NEK6 and NEK7 led to missorting of the mannose 6-phosphate receptor from endosomes. Moreover, in multiple human cell types, depletion of NEK6 or NEK7 disrupted both early and recycling endosomal compartments, including the presence of excess tubulation within recycling endosomes, a defect also observed after NEKL-3 depletion in worms. Thus, NIMA family kinases carry out multiple functions during endocytosis in both worms and humans, consistent with our previous observation that human NEKL-3 orthologs can rescue molting and trafficking defects in C. elegans nekl-3 mutants. Our findings suggest that trafficking defects could underlie some of the proposed roles for NEK kinases in human disease.
Collapse
Affiliation(s)
- Braveen B. Joseph
- Department of Molecular Biology, College of Agriculture Life Sciences, and Natural Resources, University of Wyoming, Laramie, Wyoming, United States of America
| | - Naava Naslavsky
- Department of Biochemistry & Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- The Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Shaonil Binti
- Department of Molecular Biology, College of Agriculture Life Sciences, and Natural Resources, University of Wyoming, Laramie, Wyoming, United States of America
| | - Sylvia Conquest
- Department of Molecular Biology, College of Agriculture Life Sciences, and Natural Resources, University of Wyoming, Laramie, Wyoming, United States of America
| | - Lexi Robison
- Department of Molecular Biology, College of Agriculture Life Sciences, and Natural Resources, University of Wyoming, Laramie, Wyoming, United States of America
| | - Ge Bai
- Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, New Jersey, United States of America
| | - Rafael O. Homer
- Department of Molecular Biology, College of Agriculture Life Sciences, and Natural Resources, University of Wyoming, Laramie, Wyoming, United States of America
| | - Barth D. Grant
- Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, New Jersey, United States of America
| | - Steve Caplan
- Department of Biochemistry & Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- The Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - David S. Fay
- Department of Molecular Biology, College of Agriculture Life Sciences, and Natural Resources, University of Wyoming, Laramie, Wyoming, United States of America
| |
Collapse
|
26
|
Kudryashova I. Presynaptic Plasticity Is Associated with Actin Polymerization. BIOCHEMISTRY (MOSCOW) 2023; 88:392-403. [PMID: 37076285 DOI: 10.1134/s0006297923030082] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2023]
Abstract
Modulation of presynaptic short-term plasticity induced by actin polymerization was studied in rat hippocampal slices using the paired-pulse paradigm. Schaffer collaterals were stimulated with paired pulses with a 70-ms interstimulus interval every 30 s before and during perfusion with jasplakinolide, an activator of actin polymerization. Jasplakinolide application resulted in the increase in the amplitudes of CA3-CA1 responses (potentiation) accompanied by a decrease in the paired-pulse facilitation, suggesting induction of presynaptic modifications. Jasplakinolide-induced potentiation depended on the initial paired-pulse rate. These data indicate that the jasplakinolide-mediated changes in actin polymerization increased the probability of neurotransmitter release. Less typical for CA3-CA1 synapses responses, such as a very low paired-pulse ratio (close to 1 or even lower) or even paired-pulse depression, were affected differently. Thus, jasplakinolide caused potentiation of the second, but not the first response to the paired stimulus, which increased the paired-pulse ratio from 0.8 to 1.0 on average, suggesting a negative impact of jasplakinolide on the mechanisms promoting paired-pulse depression. In general, actin polymerization facilitated potentiation, although the patterns of potentiation differed depending on the initial synapse characteristics. We conclude that in addition to the increase in the neurotransmitter release probability, jasplakinolide induced other actin polymerization-dependent mechanisms, including those involved in the paired-pulse depression.
Collapse
Affiliation(s)
- Irina Kudryashova
- Laboratory of Functional Biochemistry of the Nervous System, Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, Moscow, 119991, Russia.
| |
Collapse
|
27
|
Wang S, Chen X, Crisman L, Dou X, Winborn CS, Wan C, Puscher H, Yin Q, Kennedy MJ, Shen J. Regulation of cargo exocytosis by a Reps1-Ralbp1-RalA module. SCIENCE ADVANCES 2023; 9:eade2540. [PMID: 36812304 PMCID: PMC9946360 DOI: 10.1126/sciadv.ade2540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 01/20/2023] [Indexed: 06/18/2023]
Abstract
Surface levels of membrane proteins are determined by a dynamic balance between exocytosis-mediated surface delivery and endocytosis-dependent retrieval from the cell surface. Imbalances in surface protein levels perturb surface protein homeostasis and cause major forms of human disease such as type 2 diabetes and neurological disorders. Here, we found a Reps1-Ralbp1-RalA module in the exocytic pathway broadly regulating surface protein levels. Reps1 and Ralbp1 form a binary complex that recognizes RalA, a vesicle-bound small guanosine triphosphatases (GTPase) promoting exocytosis through interacting with the exocyst complex. RalA binding results in Reps1 release and formation of a Ralbp1-RalA binary complex. Ralbp1 selectively recognizes GTP-bound RalA but is not a RalA effector. Instead, Ralbp1 binding maintains RalA in an active GTP-bound state. These studies uncovered a segment in the exocytic pathway and, more broadly, revealed a previously unrecognized regulatory mechanism for small GTPases, GTP state stabilization.
Collapse
Affiliation(s)
- Shifeng Wang
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO 80309, USA
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Xu Chen
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO 80309, USA
| | - Lauren Crisman
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO 80309, USA
| | - Ximing Dou
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO 80309, USA
| | - Christina S. Winborn
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Chun Wan
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO 80309, USA
| | - Harrison Puscher
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO 80309, USA
| | - Qian Yin
- Department of Biological Sciences and Institute of Molecular Biophysics, Florida State University, Tallahassee, FL 32306, USA
| | - Matthew J. Kennedy
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Jingshi Shen
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO 80309, USA
| |
Collapse
|
28
|
Diot C, Cosentino G, Rameix-Welti MA. Ribonucleoprotein transport in Negative Strand RNA viruses. Biol Cell 2023; 115:e2200059. [PMID: 36192136 DOI: 10.1111/boc.202200059] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 08/01/2022] [Accepted: 09/09/2022] [Indexed: 01/07/2023]
Abstract
Negative-sense, single-stranded RNA (-ssRNA) viruses comprise some of the deadliest human pathogens (Ebola, rabies, influenza A viruses etc.). Developing therapeutic tools relies on a better understanding of their multiplication cycle. For these viruses, the genome replication and transcription activities most-often segregate in membrane-less environments called inclusion bodies (IBs) or viral factories. These "organelles" usually locate far from the cell surface from where new virions are released, and -ssRNA viruses do not encode for transport factors. The efficient trafficking of the genome progeny toward the cell surface is most often ensured by mechanisms co-opting the cellular machineries. In this review, for each -ssRNA viral family, we cover the methods employed to characterize these host-virus interactions, the strategies used by the viruses to promote the virus genome transport, and the current gaps in the literature. Finally, we highlight how Rab11 has emerged as a target of choice for the intracellular transport of -ssRNA virus genomes.
Collapse
Affiliation(s)
- Cédric Diot
- Université Paris-Saclay - Université de Versailles St. Quentin, UMR 1173 (2I), INSERM, Montigny-le-Bretonneux, France
| | - Gina Cosentino
- Université Paris-Saclay - Université de Versailles St. Quentin, UMR 1173 (2I), INSERM, Montigny-le-Bretonneux, France
| | - Marie-Anne Rameix-Welti
- Université Paris-Saclay - Université de Versailles St. Quentin, UMR 1173 (2I), INSERM, Montigny-le-Bretonneux, France.,Assistance Publique des Hôpitaux de Paris, Hôpital Ambroise Paré, Laboratoire de Microbiologie, DMU15, Versailles, France
| |
Collapse
|
29
|
Abu Rass R, Kembou-Ringert JE, Zamostiano R, Eldar A, Ehrlich M, Bacharach E. Mapping of Tilapia Lake Virus entry pathways with inhibitors reveals dependence on dynamin activity and cholesterol but not endosomal acidification. Front Cell Dev Biol 2022; 10:1075364. [PMID: 36605723 PMCID: PMC9809973 DOI: 10.3389/fcell.2022.1075364] [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: 10/20/2022] [Accepted: 12/05/2022] [Indexed: 12/23/2022] Open
Abstract
Tilapia Lake Virus (TiLV) is an emerging virus lethal to tilapia, which threatens the global tilapia aquaculture with severe implications for food security. TiLV possesses similar features to orthomyxoviruses but is classified in the sole and the monotypic genus Tilapinevirus of the family Amnoonviridae. TiLV enveloped virions encapsidate a genome comprising ten segments of single-stranded, negative RNA. Remarkably, nine of TiLV's ten major proteins lack sequence homology to any known viral or cellular proteins. The mode of TiLV entry into tilapia cells is not known. Following the measurement of the entry window of TiLV (∼3 h), we applied a panel of inhibitors of known regulators of endocytic functions to map the molecular requirements for TiLV entry. We identified productive entry by quantification of TiLV nucleoprotein expression and the generation of infectious particles. Inhibition of dynamin activity with dynasore or dynole, or depletion of cholesterol with methyl-β-cyclodextrin, strongly inhibited TiLV protein synthesis and infectious virion production. Moreover, inhibition of actin cytoskeleton polymerization with latrunculin A or microtubule polymerization with nocodazole within the entry window resulted in partial inhibition of TiLV infection. In contrast, inhibitors of endosomal acidification (NH4Cl, bafilomycin A1, or chloroquine), an inhibitor of clathrin-coated pit assembly (pitstop 2), and erlotinib-an inhibitor of the endocytic Cyclin G-associated kinase (GAK), did not affect TiLV entry. Altogether, these results suggest that TiLV enters via dynamin-mediated endocytosis in a cholesterol-, cytoskeleton-dependent manner, and clathrin-, pH-independent manner. Thus, despite being an orthomyxo-like virus, when compared to the prototypical orthomyxovirus (influenza A virus), TiLV shows a distinct set of requirements for entry into cells.
Collapse
Affiliation(s)
- Reem Abu Rass
- The Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv-Yafo, Israel
| | - Japhette Esther Kembou-Ringert
- The Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv-Yafo, Israel
| | - Rachel Zamostiano
- The Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv-Yafo, Israel
| | - Avi Eldar
- Department of Virology, The Kimron Veterinary Institute, Beit Dagan, Israel
| | - Marcelo Ehrlich
- The Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv-Yafo, Israel,*Correspondence: Marcelo Ehrlich, ; Eran Bacharach,
| | - Eran Bacharach
- The Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv-Yafo, Israel,*Correspondence: Marcelo Ehrlich, ; Eran Bacharach,
| |
Collapse
|
30
|
Etheridge RD. Protozoan phagotrophy from predators to parasites: An overview of the enigmatic cytostome-cytopharynx complex of Trypanosoma cruzi. J Eukaryot Microbiol 2022; 69:e12896. [PMID: 35175673 PMCID: PMC11110969 DOI: 10.1111/jeu.12896] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 02/14/2022] [Accepted: 02/14/2022] [Indexed: 11/28/2022]
Abstract
Eating is fundamental and from this basic principle, living organisms have evolved innumerable strategies to capture energy and nutrients from their environment. As part of the world's aquatic ecosystems, the expansive family of heterotrophic protozoans uses self-generated currents to funnel prokaryotic prey into an ancient, yet highly enigmatic, oral apparatus known as the cytostome-cytopharynx complex prior to digestion. Despite its near ubiquitous presence in protozoans, little is known mechanistically about how this feeding organelle functions. Intriguingly, one class of these flagellated phagotrophic predators known as the kinetoplastids gave rise to a lineage of obligate parasitic protozoa, the trypanosomatids, that can infect a wide variety of organisms ranging from plants to humans. One parasitic species of humans, Trypanosoma cruzi, has retained this ancestral organelle much like its free-living relatives and continues to use it as its primary mode of endocytosis. In this review, we will highlight foundational observations made regarding the cytostome-cytopharynx complex and examine some of the most pressing questions regarding the mechanistic basis for its function. We propose that T. cruzi has the potential to serve as an excellent model system to dissect the enigmatic process of protozoal phagotrophy and thus enhance our overall understanding of fundamental eukaryotic biology.
Collapse
Affiliation(s)
- Ronald Drew Etheridge
- Department of Cellular Biology, Center for Tropical and Emerging Global Diseases (CTEGD), University of Georgia, Athens, Georgia, USA
| |
Collapse
|
31
|
Yang C, Colosi P, Hugelier S, Zabezhinsky D, Lakadamyali M, Svitkina T. Actin polymerization promotes invagination of flat clathrin-coated lattices in mammalian cells by pushing at lattice edges. Nat Commun 2022; 13:6127. [PMID: 36253374 PMCID: PMC9576739 DOI: 10.1038/s41467-022-33852-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 10/05/2022] [Indexed: 12/24/2022] Open
Abstract
Clathrin-mediated endocytosis (CME) requires energy input from actin polymerization in mechanically challenging conditions. The roles of actin in CME are poorly understood due to inadequate knowledge of actin organization at clathrin-coated structures (CCSs). Using platinum replica electron microscopy of mammalian cells, we show that Arp2/3 complex-dependent branched actin networks, which often emerge from microtubule tips, assemble along the CCS perimeter, lack interaction with the apical clathrin lattice, and have barbed ends oriented toward the CCS. This structure is hardly compatible with the widely held "apical pulling" model describing actin functions in CME. Arp2/3 complex inhibition or epsin knockout produce large flat non-dynamic CCSs, which split into invaginating subdomains upon recovery from Arp2/3 inhibition. Moreover, epsin localization to CCSs depends on Arp2/3 activity. We propose an "edge pushing" model for CME, wherein branched actin polymerization promotes severing and invagination of flat CCSs in an epsin-dependent manner by pushing at the CCS boundary, thus releasing forces opposing the intrinsic curvature of clathrin lattices.
Collapse
Affiliation(s)
- Changsong Yang
- grid.25879.310000 0004 1936 8972Department of Biology, University of Pennsylvania, Philadelphia, PA USA
| | - Patricia Colosi
- grid.25879.310000 0004 1936 8972Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA
| | - Siewert Hugelier
- grid.25879.310000 0004 1936 8972Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA
| | - Daniel Zabezhinsky
- grid.25879.310000 0004 1936 8972Department of Biology, University of Pennsylvania, Philadelphia, PA USA
| | - Melike Lakadamyali
- grid.25879.310000 0004 1936 8972Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA
| | - Tatyana Svitkina
- grid.25879.310000 0004 1936 8972Department of Biology, University of Pennsylvania, Philadelphia, PA USA
| |
Collapse
|
32
|
Onofre TS, Loch L, Ferreira Rodrigues JP, Macedo S, Yoshida N. Gp35/50 mucin molecules of Trypanosoma cruzi metacyclic forms that mediate host cell invasion interact with annexin A2. PLoS Negl Trop Dis 2022; 16:e0010788. [PMID: 36190932 PMCID: PMC9529151 DOI: 10.1371/journal.pntd.0010788] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 09/05/2022] [Indexed: 11/07/2022] Open
Abstract
Host cell invasion is a critical step for infection by Trypanosoma cruzi, the agent of Chagas disease. In natural infection, T. cruzi metacyclic trypomastigote (MT) forms establish the first interaction with host cells. The gp35/50 mucin molecules expressed in MT have been implicated in cell invasion process, but the mechanisms involved are not well understood. We performed a series of experiments to elucidate the mode of gp35/50-mediated MT internalization. Comparing two parasite strains from genetically divergent groups, G strain (TcI) and CL strain (TcVI), expressing variant forms of mucins, we demonstrated that G strain mucins participate in MT invasion. Only G strain-derived mucins bound to HeLa cells in a receptor-dependent manner and significantly inhibited G strain MT invasion. CL strain MT internalization was not affected by mucins from either strain. HeLa cell invasion by G strain MT was associated with actin recruitment and did not rely on lysosome mobilization. To examine the involvement of annexin A2, which plays a role in actin dynamic, annexin A2-depleted HeLa cells were generated. Annexin A2-deficient cell lines were significantly more resistant than wild type controls to G strain MT invasion. In a co-immunoprecipitation assay, to check whether annexin A2 might be the receptor for mucins, protein A/G magnetic beads crosslinked with monoclonal antibody to G strain mucins were incubated with detergent extracts of MT and HeLa cells. Binding of gp35/50 mucins to annexin A2 was detected. Both G strain MT and purified mucins induced focal adhesion kinase activation in HeLa cells. By confocal immunofluorescence microscopy, colocalization of invading G strain MT with clathrin was visualized. Inhibition of clathrin-coated vesicle formation reduced parasite internalization. Taken together, our data indicate that gp35/50-mediated MT invasion is accomplished through interaction with host cell annexin A2 and clathrin-dependent endocytosis. Host cell invasion by Trypanosoma cruzi, the agent of Chagas disease, is critical for the establishment of infection. Metacyclic trypomastigote (MT) forms are responsible for the initial T. cruzi-host cell interaction. Mucin molecules expressed on MT surface have been implicated in target cell invasion process, but the underlying mechanism are not fully understood. In this study, we aimed at elucidating the mode of mucin-mediated MT internalization. We found that requirement of mucins for MT invasion is T. cruzi strain-dependent. Experiments with G strain MTs, which rely on mucins and on target cell actin for internalization, revealed that mucin molecules bind to annexin A2, a protein that plays a role in actin dynamic. Annexin A2-deficient cell lines were generated and found to be significantly more resistant than wild type controls to MT invasion. Both MT and purified mucins induced focal adhesion kinase activation in host cells. By confocal immunofluorescence microscopy, invading MT was found to colocalize with clathrin, a protein that plays a role in endocytosis. Inhibition of clathrin-coated vesicle formation reduced parasite internalization. From these data we infer that mucin-mediated MT invasion is accomplished through interaction with host cell annexin A2 and clathrin-dependent endocytosis.
Collapse
Affiliation(s)
- Thiago Souza Onofre
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, São Paulo, Brazil
| | - Leonardo Loch
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, São Paulo, Brazil
| | - João Paulo Ferreira Rodrigues
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, São Paulo, Brazil
| | - Silene Macedo
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, São Paulo, Brazil
| | - Nobuko Yoshida
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, São Paulo, Brazil,* E-mail:
| |
Collapse
|
33
|
Hatano T, Lim TC, Billault-Chaumartin I, Dhar A, Gu Y, Massam-Wu T, Scott W, Adishesha S, Chapa-y-Lazo B, Springall L, Sivashanmugam L, Mishima M, Martin SG, Oliferenko S, Palani S, Balasubramanian MK. mNG-tagged fusion proteins and nanobodies to visualize tropomyosins in yeast and mammalian cells. J Cell Sci 2022; 135:jcs260288. [PMID: 36148799 PMCID: PMC9592052 DOI: 10.1242/jcs.260288] [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: 05/27/2022] [Accepted: 08/15/2022] [Indexed: 11/20/2022] Open
Abstract
Tropomyosins are structurally conserved α-helical coiled-coil proteins that bind along the length of filamentous actin (F-actin) in fungi and animals. Tropomyosins play essential roles in the stability of actin filaments and in regulating myosin II contractility. Despite the crucial role of tropomyosin in actin cytoskeletal regulation, in vivo investigations of tropomyosin are limited, mainly due to the suboptimal live-cell imaging tools currently available. Here, we report on an mNeonGreen (mNG)-tagged tropomyosin, with native promoter and linker length configuration, that clearly reports tropomyosin dynamics in Schizosaccharomyces pombe (Cdc8), Schizosaccharomyces japonicus (Cdc8) and Saccharomyces cerevisiae (Tpm1 and Tpm2). We also describe a fluorescent probe to visualize mammalian tropomyosin (TPM2 isoform). Finally, we generated a camelid nanobody against S. pombe Cdc8, which mimics the localization of mNG-Cdc8 in vivo. Using these tools, we report the presence of tropomyosin in previously unappreciated patch-like structures in fission and budding yeasts, show flow of tropomyosin (F-actin) cables to the cytokinetic actomyosin ring and identify rearrangements of the actin cytoskeleton during mating. These powerful tools and strategies will aid better analyses of tropomyosin and F-actin cables in vivo.
Collapse
Affiliation(s)
- Tomoyuki Hatano
- Centre for Mechanochemical Cell Biology and Division of Biomedical Sciences, Warwick Medical School, Warwick CV4 7AL, UK
| | - Tzer Chyn Lim
- Centre for Mechanochemical Cell Biology and Division of Biomedical Sciences, Warwick Medical School, Warwick CV4 7AL, UK
| | - Ingrid Billault-Chaumartin
- Department of Fundamental Microbiology, Faculty of Biology and Medicine, University of Lausanne, Biophore Building, CH-1015 Lausanne, Switzerland
| | - Anubhav Dhar
- Department of Biochemistry, Indian Institute of Science, Bangalore 560012, India
| | - Ying Gu
- The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
- Randall Centre for Cell and Molecular Biophysics, School of Basic and Medical Biosciences, King's College London, London, SE1 1UL, UK
| | - Teresa Massam-Wu
- Centre for Mechanochemical Cell Biology and Division of Biomedical Sciences, Warwick Medical School, Warwick CV4 7AL, UK
| | - William Scott
- Centre for Mechanochemical Cell Biology and Division of Biomedical Sciences, Warwick Medical School, Warwick CV4 7AL, UK
| | - Sushmitha Adishesha
- Department of Biochemistry, Indian Institute of Science, Bangalore 560012, India
| | - Bernardo Chapa-y-Lazo
- Centre for Mechanochemical Cell Biology and Division of Biomedical Sciences, Warwick Medical School, Warwick CV4 7AL, UK
| | - Luke Springall
- Centre for Mechanochemical Cell Biology and Division of Biomedical Sciences, Warwick Medical School, Warwick CV4 7AL, UK
| | - Lavanya Sivashanmugam
- Centre for Mechanochemical Cell Biology and Division of Biomedical Sciences, Warwick Medical School, Warwick CV4 7AL, UK
| | - Masanori Mishima
- Centre for Mechanochemical Cell Biology and Division of Biomedical Sciences, Warwick Medical School, Warwick CV4 7AL, UK
| | - Sophie G. Martin
- Department of Fundamental Microbiology, Faculty of Biology and Medicine, University of Lausanne, Biophore Building, CH-1015 Lausanne, Switzerland
| | - Snezhana Oliferenko
- The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
- Randall Centre for Cell and Molecular Biophysics, School of Basic and Medical Biosciences, King's College London, London, SE1 1UL, UK
| | - Saravanan Palani
- Department of Biochemistry, Indian Institute of Science, Bangalore 560012, India
| | - Mohan K. Balasubramanian
- Centre for Mechanochemical Cell Biology and Division of Biomedical Sciences, Warwick Medical School, Warwick CV4 7AL, UK
| |
Collapse
|
34
|
Skruzny M. The endocytic protein machinery as an actin-driven membrane-remodeling machine. Eur J Cell Biol 2022; 101:151267. [PMID: 35970066 DOI: 10.1016/j.ejcb.2022.151267] [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: 06/02/2022] [Revised: 08/02/2022] [Accepted: 08/02/2022] [Indexed: 12/14/2022] Open
Abstract
In clathrin-mediated endocytosis, a principal membrane trafficking route of all eukaryotic cells, forces are applied to invaginate the plasma membrane and form endocytic vesicles. These forces are provided by specific endocytic proteins and the polymerizing actin cytoskeleton. One of the best-studied endocytic systems is endocytosis in yeast, known for its simplicity, experimental amenability, and overall similarity to human endocytosis. Importantly, the yeast endocytic protein machinery generates and transmits tremendous force to bend the plasma membrane, making this system beneficial for mechanistic studies of cellular force-driven membrane reshaping. This review summarizes important protein players, molecular functions, applied forces, and open questions and perspectives of this robust, actin-powered membrane-remodeling protein machine.
Collapse
Affiliation(s)
- Michal Skruzny
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory (EMBL), Meyerhofstrasse 1, 69117 Heidelberg, Germany.
| |
Collapse
|
35
|
Dabie bandavirus Nonstructural Protein Interacts with Actin to Induce F-Actin Rearrangement and Inhibit Viral Adsorption and Entry. J Virol 2022; 96:e0078822. [PMID: 35862701 PMCID: PMC9327694 DOI: 10.1128/jvi.00788-22] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Dabie bandavirus (DBV) is an emerging Bandavirus that causes multiorgan failure with a high fatality rate in humans. While many viruses can manipulate the actin cytoskeleton to facilitate viral growth, the regulation pattern of the actin cytoskeleton and the molecular mechanisms involved in DBV entry into the host cells remain unclear. In this study, we demonstrate that expression of nonstructural protein (NSs) or infection with DBV induces actin rearrangement, which presents a point-like distribution, and this destruction is dependent on inclusion bodies (IBs). Further experiments showed that NSs inhibits viral adsorption by destroying the filopodium structure. In addition, NSs also compromised the viral entry by inhibiting clathrin aggregation on the cell surface and capturing clathrin into IBs. Furthermore, NSs induced clathrin light chain B (CLTB) degradation through the K48-linked ubiquitin proteasome pathway, which could negatively regulate clathrin-mediated endocytosis, inhibiting the viral entry. Finally, we confirmed that this NSs-induced antiviral mechanism is broadly applicable to other viruses, such as enterovirus 71 (EV71) and influenza virus, A/PR8/34 (PR8), which use the same clathrin-mediated endocytosis to enter host cells. In conclusion, our study provides new insights into the role of NSs in inhibiting endocytosis and a novel strategy for treating DBV infections. IMPORTANCEDabie bandavirus (DBV), a member of the Phenuiviridae family, is a newly emerging tick-borne pathogen that causes multifunctional organ failure and even death in humans. The actin cytoskeleton is involved in various crucial cellular processes and plays an important role in viral life activities. However, the relationship between DBV infection and the actin cytoskeleton has not been described in detail. Here, we show for the first time the interaction between NSs and actin to induce actin rearrangement, which inhibits the viral adsorption and entry. We also identify a key mechanism underlying NSs-induced entry inhibition in which NSs prevents clathrin aggregation on the cell surface by hijacking clathrin into the inclusion body and induces CLTB degradation through the K48-linked ubiquitination modification. This paper is the first to reveal the antiviral mechanism of NSs and provides a theoretical basis for the search for new antiviral targets.
Collapse
|
36
|
Serwas D, Akamatsu M, Moayed A, Vegesna K, Vasan R, Hill JM, Schöneberg J, Davies KM, Rangamani P, Drubin DG. Mechanistic insights into actin force generation during vesicle formation from cryo-electron tomography. Dev Cell 2022; 57:1132-1145.e5. [PMID: 35504288 PMCID: PMC9165722 DOI: 10.1016/j.devcel.2022.04.012] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 01/18/2022] [Accepted: 04/07/2022] [Indexed: 01/26/2023]
Abstract
Actin assembly provides force for a multitude of cellular processes. Compared to actin-assembly-based force production during cell migration, relatively little is understood about how actin assembly generates pulling forces for vesicle formation. Here, cryo-electron tomography identified actin filament number, organization, and orientation during clathrin-mediated endocytosis in human SK-MEL-2 cells, showing that force generation is robust despite variance in network organization. Actin dynamics simulations incorporating a measured branch angle indicate that sufficient force to drive membrane internalization is generated through polymerization and that assembly is triggered from ∼4 founding "mother" filaments, consistent with tomography data. Hip1R actin filament anchoring points are present along the entire endocytic invagination, where simulations show that it is key to pulling force generation, and along the neck, where it targets filament growth and makes internalization more robust. Actin organization described here allowed direct translation of structure to mechanism with broad implications for other actin-driven processes.
Collapse
Affiliation(s)
- Daniel Serwas
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA.
| | - Matthew Akamatsu
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Amir Moayed
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Karthik Vegesna
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Ritvik Vasan
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, CA, USA
| | - Jennifer M Hill
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Johannes Schöneberg
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Karen M Davies
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA; Molecular Biophysics and Integrative Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Padmini Rangamani
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, CA, USA
| | - David G Drubin
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA.
| |
Collapse
|
37
|
Chen N, He Y, Zang M, Zhang Y, Lu H, Zhao Q, Wang S, Gao Y. Approaches and materials for endocytosis-independent intracellular delivery of proteins. Biomaterials 2022; 286:121567. [DOI: 10.1016/j.biomaterials.2022.121567] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 04/26/2022] [Accepted: 05/03/2022] [Indexed: 12/12/2022]
|
38
|
Mayya C, Naveena AH, Sinha P, Wunder C, Johannes L, Bhatia D. The roles of dynein and myosin VI motor proteins in endocytosis. J Cell Sci 2022; 135:274777. [DOI: 10.1242/jcs.259387] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
ABSTRACT
Endocytosis is indispensable for multiple cellular processes, including signalling, cell adhesion, migration, as well as the turnover of plasma membrane lipids and proteins. The dynamic interplay and regulation of different endocytic entry routes requires multiple cytoskeletal elements, especially motor proteins that bind to membranes and transport vesicles along the actin and microtubule cytoskeletons. Dynein and kinesin motor proteins transport vesicles along microtubules, whereas myosins drive vesicles along actin filaments. Here, we present a brief overview of multiple endocytic pathways and our current understanding of the involvement of these motor proteins in the regulation of the different cellular entry routes. We particularly focus on structural and mechanistic details of the retrograde motor proteins dynein and myosin VI (also known as MYO6), along with their adaptors, which have important roles in the early events of endocytosis. We conclude by highlighting the key challenges in elucidating the involvement of motor proteins in endocytosis and intracellular membrane trafficking.
Collapse
Affiliation(s)
- Chaithra Mayya
- Biological Engineering, Indian Institute of Technology Gandhinagar, Palaj, 382355 Gandhinagar, India
| | - A. Hema Naveena
- Biological Engineering, Indian Institute of Technology Gandhinagar, Palaj, 382355 Gandhinagar, India
| | - Pankhuri Sinha
- Biological Engineering, Indian Institute of Technology Gandhinagar, Palaj, 382355 Gandhinagar, India
| | - Christian Wunder
- Institut Curie, PSL Research University, U1143 INSERM, UMR3666 CNRS, Cellular and Chemical Biology Unit, 26 rue d'Ulm, 75248 Paris CEDEX 05, France
| | - Ludger Johannes
- Institut Curie, PSL Research University, U1143 INSERM, UMR3666 CNRS, Cellular and Chemical Biology Unit, 26 rue d'Ulm, 75248 Paris CEDEX 05, France
| | - Dhiraj Bhatia
- Biological Engineering, Indian Institute of Technology Gandhinagar, Palaj, 382355 Gandhinagar, India
- Center for Biomedical Engineering, Indian Institute of Technology Gandhinagar, Palaj, 382355 Gandhinagar, India
| |
Collapse
|
39
|
Lu Y, Huang D, Wang B, Zheng B, Liu J, Song J, Zheng S. FAM21C Promotes Hepatocellular Carcinoma Invasion and Metastasis by Driving Actin Cytoskeleton Remodeling via Inhibiting Capping Ability of CAPZA1. Front Oncol 2022; 11:809195. [PMID: 35096613 PMCID: PMC8793146 DOI: 10.3389/fonc.2021.809195] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 12/21/2021] [Indexed: 12/24/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is characterized by a high incidence of metastasis. The dynamic remodeling of the actin cytoskeleton plays an important role in the invasion and migration of HCC cells. In previous studies, we found that CAPZA1, a capping protein, can promote EMT of HCC cells by regulating the remodeling of the actin filament (F-actin) cytoskeleton, thus promoting the invasion and migration of HCC cells. In this study, we found that FAM21C may have a regulatory effect on CAPZA1, and we conducted an in-depth study on its potential regulatory mechanism. First, we found that FAM21C is highly expressed in HCC tissues and its high expression could promote the malignant progression of HCC. Meanwhile, the high expression of FAM21C promoted the invasion and migration of HCC cells in vitro and in vivo. Further, FAM21C interacted with CAPZA1, and their binding inhibited the capping capacity of CAPZA1, thus promoting the invasion and migration of HCC cells. This effect of FAM21C was abolished by mutating the CP-interacting (CPI) domain, the CAPZA1 binding site on FAM21C. In conclusion, high expression of FAM21C in HCC tissues can promote malignant progression of HCC and its potential mechanism involves FAM21C inhibition of CAPZA1 capping capacity by binding to CAPZA1, which drives F-actin cytoskeleton remodeling, and thus promotes invasion and migration of HCC cells.
Collapse
Affiliation(s)
- Yao Lu
- Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Deng Huang
- Department of Hepatobiliary, General Hospital of Tibet Military Command Area, Tibet, China
| | - Baolin Wang
- Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Bowen Zheng
- Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Jialong Liu
- Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Juxian Song
- Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Shuguo Zheng
- Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| |
Collapse
|
40
|
Roy A, Zhang W, Jahed Z, Tsai CT, Cui B, Moerner WE. Exploring Cell Surface-Nanopillar Interactions with 3D Super-Resolution Microscopy. ACS NANO 2022; 16:192-210. [PMID: 34582687 PMCID: PMC8830212 DOI: 10.1021/acsnano.1c05313] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Plasma membrane topography has been shown to strongly influence the behavior of many cellular processes such as clathrin-mediated endocytosis, actin rearrangements, and others. Recent studies have used three-dimensional (3D) nanostructures such as nanopillars to imprint well-defined membrane curvatures (the "nano-bio interface"). In these studies, proteins and their interactions were probed by two-dimensional fluorescence microscopy. However, the low resolution and limited axial detail of such methods are not optimal to determine the relative spatial position and distribution of proteins along a 100 nm-diameter object, which is below the optical diffraction limit. Here, we introduce a general method to explore the nanoscale distribution of proteins at the nano-bio interface with 10-20 nm precision using 3D single-molecule super-resolution (SR) localization microscopy. This is achieved by combining a silicone-oil immersion objective and 3D double-helix point spread function microscopy. We carefully adjust the objective to minimize spherical aberrations between quartz nanopillars and the cell. To validate the 3D SR method, we imaged the 3D shape of surface-labeled nanopillars and compared the results with electron microscopy measurements. Turning to transmembrane-anchored labels in cells, the high quality 3D SR reconstructions reveal the membrane tightly wrapping around the nanopillars. Interestingly, the cytoplasmic protein AP-2 involved in clathrin-mediated endocytosis accumulates along the nanopillar above a specific threshold of 1/R (the reciprocal of the radius) membrane curvature. Finally, we observe that AP-2 and actin preferentially accumulate at positive Gaussian curvature near the pillar caps. Our results establish a general method to investigate the nanoscale distribution of proteins at the nano-bio interface using 3D SR microscopy.
Collapse
|
41
|
Ulfo L, Costantini PE, Di Giosia M, Danielli A, Calvaresi M. EGFR-Targeted Photodynamic Therapy. Pharmaceutics 2022; 14:241. [PMID: 35213974 PMCID: PMC8879084 DOI: 10.3390/pharmaceutics14020241] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/13/2022] [Accepted: 01/14/2022] [Indexed: 12/04/2022] Open
Abstract
The epidermal growth factor receptor (EGFR) plays a pivotal role in the proliferation and metastatization of cancer cells. Aberrancies in the expression and activation of EGFR are hallmarks of many human malignancies. As such, EGFR-targeted therapies hold significant potential for the cure of cancers. In recent years, photodynamic therapy (PDT) has gained increased interest as a non-invasive cancer treatment. In PDT, a photosensitizer is excited by light to produce reactive oxygen species, resulting in local cytotoxicity. One of the critical aspects of PDT is to selectively transport enough photosensitizers to the tumors environment. Accordingly, an increasing number of strategies have been devised to foster EGFR-targeted PDT. Herein, we review the recent nanobiotechnological advancements that combine the promise of PDT with EGFR-targeted molecular cancer therapy. We recapitulate the chemistry of the sensitizers and their modes of action in PDT, and summarize the advantages and pitfalls of different targeting moieties, highlighting future perspectives for EGFR-targeted photodynamic treatment of cancer.
Collapse
Affiliation(s)
- Luca Ulfo
- Dipartimento di Farmacia e Biotecnologie, Alma Mater Studiorum—Università di Bologna, Via Francesco Selmi 3, 40126 Bologna, Italy; (L.U.); (P.E.C.)
| | - Paolo Emidio Costantini
- Dipartimento di Farmacia e Biotecnologie, Alma Mater Studiorum—Università di Bologna, Via Francesco Selmi 3, 40126 Bologna, Italy; (L.U.); (P.E.C.)
| | - Matteo Di Giosia
- Dipartimento di Chimica “Giacomo Ciamician”, Alma Mater Studiorum—Università di Bologna, Via Francesco Selmi 2, 40126 Bologna, Italy;
| | - Alberto Danielli
- Dipartimento di Farmacia e Biotecnologie, Alma Mater Studiorum—Università di Bologna, Via Francesco Selmi 3, 40126 Bologna, Italy; (L.U.); (P.E.C.)
| | - Matteo Calvaresi
- Dipartimento di Chimica “Giacomo Ciamician”, Alma Mater Studiorum—Università di Bologna, Via Francesco Selmi 2, 40126 Bologna, Italy;
| |
Collapse
|
42
|
Zhang Y, Li L, Wang J. Role of Ligand Distribution in the Cytoskeleton-Associated Endocytosis of Ellipsoidal Nanoparticles. MEMBRANES 2021; 11:membranes11120993. [PMID: 34940494 PMCID: PMC8705050 DOI: 10.3390/membranes11120993] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 12/16/2021] [Accepted: 12/17/2021] [Indexed: 11/27/2022]
Abstract
Nanoparticle (NP)–cell interaction mediated by receptor–ligand bonds is a crucial phenomenon in pathology, cellular immunity, and drug delivery systems, and relies strongly on the shape of NPs and the stiffness of the cell. Given this significance, a fundamental question is raised on how the ligand distribution may affect the membrane wrapping of non-spherical NPs under the influence of cytoskeleton deformation. To address this issue, in this work we use a coupled elasticity–diffusion model to systematically investigate the role of ligand distribution in the cytoskeleton-associated endocytosis of ellipsoidal NPs for different NP shapes, sizes, cytoskeleton stiffness, and the initial receptor densities. In this model, we have taken into account the effects of receptor diffusion, receptor–ligand binding, cytoskeleton and membrane deformations, and changes in the configuration entropy of receptors. By solving this model, we find that the uptake process can be significantly influenced by the ligand distribution. Additionally, there exists an optimal state of such a distribution, which corresponds to the fastest uptake efficiency and depends on the NP aspect ratio and cytoskeleton stiffness. We also find that the optimal distribution usually needs local ligand density to be sufficiently high at the large curvature region. Furthermore, the optimal state of NP entry into cells can tolerate slight changes to the corresponding optimal distribution of the ligands. The tolerance to such a change is enhanced as the average receptor density and NP size increase. These results may provide guidelines to control NP–cell interactions and improve the efficiency of target drug delivery systems.
Collapse
Affiliation(s)
| | - Long Li
- Correspondence: (L.L.); (J.W.)
| | | |
Collapse
|
43
|
Mehidi A, Kage F, Karatas Z, Cercy M, Schaks M, Polesskaya A, Sainlos M, Gautreau AM, Rossier O, Rottner K, Giannone G. Forces generated by lamellipodial actin filament elongation regulate the WAVE complex during cell migration. Nat Cell Biol 2021; 23:1148-1162. [PMID: 34737443 DOI: 10.1038/s41556-021-00786-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 09/27/2021] [Indexed: 12/17/2022]
Abstract
Actin filaments generate mechanical forces that drive membrane movements during trafficking, endocytosis and cell migration. Reciprocally, adaptations of actin networks to forces regulate their assembly and architecture. Yet, a demonstration of forces acting on actin regulators at actin assembly sites in cells is missing. Here we show that local forces arising from actin filament elongation mechanically control WAVE regulatory complex (WRC) dynamics and function, that is, Arp2/3 complex activation in the lamellipodium. Single-protein tracking revealed WRC lateral movements along the lamellipodium tip, driven by elongation of actin filaments and correlating with WRC turnover. The use of optical tweezers to mechanically manipulate functional WRC showed that piconewton forces, as generated by single-filament elongation, dissociated WRC from the lamellipodium tip. WRC activation correlated with its trapping, dwell time and the binding strength at the lamellipodium tip. WRC crosslinking, hindering its mechanical dissociation, increased WRC dwell time and Arp2/3-dependent membrane protrusion. Thus, forces generated by individual actin filaments on their regulators can mechanically tune their turnover and hence activity during cell migration.
Collapse
Affiliation(s)
- Amine Mehidi
- University Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, Bordeaux, France
| | - Frieda Kage
- Division of Molecular Cell Biology, Zoological Institute, Technische Universität Braunschweig, Braunschweig, Germany.,Department of Cell Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Zeynep Karatas
- University Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, Bordeaux, France
| | - Maureen Cercy
- University Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, Bordeaux, France
| | - Matthias Schaks
- Division of Molecular Cell Biology, Zoological Institute, Technische Universität Braunschweig, Braunschweig, Germany.,Department of Cell Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Anna Polesskaya
- CNRS, Ecole Polytechnique, Institut Polytechnique de Paris, Palaiseau, France
| | - Matthieu Sainlos
- University Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, Bordeaux, France
| | - Alexis M Gautreau
- CNRS, Ecole Polytechnique, Institut Polytechnique de Paris, Palaiseau, France
| | - Olivier Rossier
- University Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, Bordeaux, France
| | - Klemens Rottner
- Division of Molecular Cell Biology, Zoological Institute, Technische Universität Braunschweig, Braunschweig, Germany.,Department of Cell Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Grégory Giannone
- University Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, Bordeaux, France.
| |
Collapse
|
44
|
Liu W, Tang D, Xu XX, Liu YJ, Jiu Y. How Physical Factors Coordinate Virus Infection: A Perspective From Mechanobiology. Front Bioeng Biotechnol 2021; 9:764516. [PMID: 34778236 PMCID: PMC8585752 DOI: 10.3389/fbioe.2021.764516] [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: 08/25/2021] [Accepted: 09/28/2021] [Indexed: 11/13/2022] Open
Abstract
Pandemics caused by viruses have threatened lives of thousands of people. Understanding the complicated process of viral infection provides significantly directive implication to epidemic prevention and control. Viral infection is a complex and diverse process, and substantial studies have been complemented in exploring the biochemical and molecular interactions between viruses and hosts. However, the physical microenvironment where infections implement is often less considered, and the role of mechanobiology in viral infection remains elusive. Mechanobiology focuses on sensation, transduction, and response to intracellular and extracellular physical factors by tissues, cells, and extracellular matrix. The intracellular cytoskeleton and mechanosensors have been proven to be extensively involved in the virus life cycle. Furthermore, innovative methods based on micro- and nanofabrication techniques are being utilized to control and modulate the physical and chemical cell microenvironment, and to explore how extracellular factors including stiffness, forces, and topography regulate viral infection. Our current review covers how physical factors in the microenvironment coordinate viral infection. Moreover, we will discuss how this knowledge can be harnessed in future research on cross-fields of mechanobiology and virology.
Collapse
Affiliation(s)
- Wei Liu
- Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Department of Systems Biology for Medicine, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Daijiao Tang
- The Center for Microbes, Development and Health, Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xin-Xin Xu
- Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Department of Systems Biology for Medicine, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Yan-Jun Liu
- Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Department of Systems Biology for Medicine, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Yaming Jiu
- The Center for Microbes, Development and Health, Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
45
|
Li Y, Tang Y, Liu J, Meng X, Wang Y, Min Q, Hong R, Tsubata T, Hase K, Wang JY. Glia maturation factor-γ is involved in S1P-induced marginal zone B cell chemotaxis and optimal T-independent type II antigen-induced IgM production. Int Immunol 2021; 34:35-43. [PMID: 34673932 DOI: 10.1093/intimm/dxab097] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 10/20/2021] [Indexed: 11/14/2022] Open
Abstract
Marginal zone B cells (MZB) represent a unique B cell subpopulation that rapidly differentiate into IgM-secreting plasma cells in response to T-independent (T-I) antigen. Sphingosine 1-phosphate (S1P) promotes MZB localization to the marginal zone. However, intracellular molecules involved in MZB localization and migration remain largely unknown. Here we show that MZB lacking the Glia maturation factor-γ (GMFG) are impaired in chemotaxis toward S1P under both in vitro and in vivo conditions, suggesting that GMFG is an effector downstream of S1P receptors. GMFG undergoes serine phosphorylation upon S1P stimulation and is required for S1P-induced desensitization of S1P receptor 1 (S1PR1). Compared with wild type mice, Gmfg -/- mice produce elevated levels of 4-hydroxy-3-nitrophenyl-acetyl (NP)-specific IgM against a T-I type II antigen, NP-Ficoll, accompanied by dysregulated MZB localization. These results identify GMFG as a regulator of S1P-induced MZB chemotaxis and reveal a role for MZB localization in the marginal zone for optimal IgM production against a T-I antigen.
Collapse
Affiliation(s)
- Yingqian Li
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Yue Tang
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Jun Liu
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Xin Meng
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Ying Wang
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Qing Min
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Rongjian Hong
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai200030, China
| | - Takeshi Tsubata
- Department of Immunology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo 113-8510, Japan
| | - Koji Hase
- Division of Biochemistry, Faculty of Pharmacy, Keio University, Tokyo, Japan
| | - Ji-Yang Wang
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China.,Department of Immunology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo 113-8510, Japan.,Department of Clinical Immunology, Children's Hospital of Fudan University, Shanghai, 200032, China.,Department of Microbiology and Immunology, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
| |
Collapse
|
46
|
Wan C, Crisman L, Wang B, Tian Y, Wang S, Yang R, Datta I, Nomura T, Li S, Yu H, Yin Q, Shen J. AAGAB is an assembly chaperone regulating AP1 and AP2 clathrin adaptors. J Cell Sci 2021; 134:272394. [PMID: 34494650 DOI: 10.1242/jcs.258587] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 08/31/2021] [Indexed: 11/20/2022] Open
Abstract
Multimeric cargo adaptors such as AP2 play central roles in intracellular membrane trafficking. We recently discovered that the assembly of the AP2 adaptor complex, a key player in clathrin-mediated endocytosis, is a highly organized process controlled by alpha- and gamma-adaptin-binding protein (AAGAB, also known as p34). In this study, we demonstrate that besides AP2, AAGAB also regulates the assembly of AP1, a cargo adaptor involved in clathrin-mediated transport between the trans-Golgi network and the endosome. However, AAGAB is not involved in the formation of other adaptor complexes, including AP3. AAGAB promotes AP1 assembly by binding and stabilizing the γ and σ subunits of AP1, and its mutation abolishes AP1 assembly and disrupts AP1-mediated cargo trafficking. Comparative proteomic analyses indicate that AAGAB mutation massively alters surface protein homeostasis, and its loss-of-function phenotypes reflect the synergistic effects of AP1 and AP2 deficiency. Taken together, these findings establish AAGAB as an assembly chaperone for both AP1 and AP2 adaptors and pave the way for understanding the pathogenesis of AAGAB-linked diseases.
Collapse
Affiliation(s)
- Chun Wan
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO 80309, USA
| | - Lauren Crisman
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO 80309, USA
| | - Bing Wang
- Department of Biological Sciences and Institute of Molecular Biophysics, Florida State University, Tallahassee, FL 32306, USA
| | - Yuan Tian
- Department of Biological Sciences and Institute of Molecular Biophysics, Florida State University, Tallahassee, FL 32306, USA
| | - Shifeng Wang
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO 80309, USA
| | - Rui Yang
- Department of Biological Sciences and Institute of Molecular Biophysics, Florida State University, Tallahassee, FL 32306, USA
| | - Ishara Datta
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO 80309, USA
| | - Toshifumi Nomura
- Department of Dermatology, University of Tsukuba, Tsukuba, 305-8575, Japan
| | - Suzhao Li
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Haijia Yu
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO 80309, USA
| | - Qian Yin
- Department of Biological Sciences and Institute of Molecular Biophysics, Florida State University, Tallahassee, FL 32306, USA
| | - Jingshi Shen
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO 80309, USA
| |
Collapse
|
47
|
Yoshida N, Ogura I, Nagano M, Ando T, Toshima JY, Toshima J. Cooperative regulation of endocytic vesicle transport by yeast Eps15-like protein Pan1p and epsins. J Biol Chem 2021; 297:101254. [PMID: 34592316 PMCID: PMC8628263 DOI: 10.1016/j.jbc.2021.101254] [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: 07/04/2021] [Revised: 09/23/2021] [Accepted: 09/24/2021] [Indexed: 11/28/2022] Open
Abstract
Dynamic actin filaments are required for the formation and internalization of endocytic vesicles. Yeast actin cables serve as a track for the translocation of endocytic vesicles to early endosomes, but the molecular mechanisms regulating the interaction between vesicles and the actin cables remain ambiguous. Previous studies have demonstrated that the yeast Eps15-like protein Pan1p plays an important role in this interaction, and that interaction is not completely lost even after deletion of the Pan1p actin-binding domain, suggesting that additional proteins mediate association of the vesicle with the actin cable. Other candidates for mediating the interaction are endocytic coat proteins Sla2p (yeast Hip1R) and Ent1p/2p (yeast epsins), as these proteins can bind to both the plasma membrane and the actin filament. Here, we investigated the degree of redundancy in the actin-binding activities of Pan1p, Sla2p, and Ent1p/2p involved in the internalization and transport of endocytic vesicles. Expression of the nonphosphorylatable form of Pan1p, Pan1-18TA, caused abnormal accumulation of both actin cables and endocytic vesicles, and this accumulation was additively suppressed by deletion of the actin-binding domains of both Pan1p and Ent1p. Interestingly, deletion of the actin-binding domains of Pan1p and Ent1p in cells lacking the ENT2 gene resulted in severely defective internalization of endocytic vesicles and recruitment of actin cables to the site of endocytosis. These results suggest that Pan1p and Ent1p/2p cooperatively regulate the interaction between the endocytic vesicle and the actin cable.
Collapse
Affiliation(s)
- Nao Yoshida
- Department of Biological Science and Technology, Tokyo University of Science, Katsushika-ku, Tokyo, Japan
| | - Ippo Ogura
- Department of Biological Science and Technology, Tokyo University of Science, Katsushika-ku, Tokyo, Japan
| | - Makoto Nagano
- Department of Biological Science and Technology, Tokyo University of Science, Katsushika-ku, Tokyo, Japan
| | - Tadashi Ando
- Department of Applied Electronics, Tokyo University of Science, Katsushika-ku, Tokyo, Japan
| | - Junko Y Toshima
- Department of Biological Science and Technology, Tokyo University of Science, Katsushika-ku, Tokyo, Japan; School of Health Science, Tokyo University of Technology, Ota-ku, Tokyo, Japan.
| | - Jiro Toshima
- Department of Biological Science and Technology, Tokyo University of Science, Katsushika-ku, Tokyo, Japan.
| |
Collapse
|
48
|
Ganguly A, Sharma R, Boyer NP, Wernert F, Phan S, Boassa D, Parra L, Das U, Caillol G, Han X, Yates JR, Ellisman MH, Leterrier C, Roy S. Clathrin packets move in slow axonal transport and deliver functional payloads to synapses. Neuron 2021; 109:2884-2901.e7. [PMID: 34534453 PMCID: PMC8457040 DOI: 10.1016/j.neuron.2021.08.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 06/10/2021] [Accepted: 08/13/2021] [Indexed: 12/25/2022]
Abstract
In non-neuronal cells, clathrin has established roles in endocytosis, with clathrin cages enclosing plasma membrane infoldings, followed by rapid disassembly and reuse of monomers. However, in neurons, clathrin is conveyed in slow axonal transport over days to weeks, and the underlying transport/targeting mechanisms, mobile cargo structures, and even its precise presynaptic localization and physiologic role are unclear. Combining live imaging, photobleaching/conversion, mass spectrometry, electron microscopy, and super-resolution imaging, we found that unlike in dendrites, where clathrin cages rapidly assemble and disassemble, in axons, clathrin and related proteins organize into stable "transport packets" that are unrelated to endocytosis and move intermittently on microtubules, generating an overall slow anterograde flow. At synapses, multiple clathrin packets abut synaptic vesicle (SV) clusters, and clathrin packets also exchange between synaptic boutons in a microtubule-dependent "superpool." Within synaptic boundaries, clathrin is surprisingly dynamic, continuously exchanging between local clathrin assemblies, and its depletion impairs SV recycling. Our data provide a conceptual framework for understanding clathrin trafficking and presynaptic targeting that has functional implications.
Collapse
Affiliation(s)
- Archan Ganguly
- Department of Pathology, University of California, San Diego, La Jolla, CA, USA
| | - Rohan Sharma
- Department of Pathology, University of California, San Diego, La Jolla, CA, USA
| | - Nicholas P Boyer
- Department of Pathology, University of California, San Diego, La Jolla, CA, USA
| | - Florian Wernert
- Aix Marseille Université, CNRS, INP UMR7051, NeuroCyto, Marseille, France
| | - Sébastien Phan
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA; National Center for Microscopy and Imaging Research, University of California, San Diego, La Jolla, CA, USA
| | - Daniela Boassa
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA; National Center for Microscopy and Imaging Research, University of California, San Diego, La Jolla, CA, USA
| | - Leonardo Parra
- Department of Pathology, University of California, San Diego, La Jolla, CA, USA
| | - Utpal Das
- Department of Pathology, University of California, San Diego, La Jolla, CA, USA; Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Ghislaine Caillol
- Aix Marseille Université, CNRS, INP UMR7051, NeuroCyto, Marseille, France
| | - Xuemei Han
- Department of Cell Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - John R Yates
- Department of Cell Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Mark H Ellisman
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA; National Center for Microscopy and Imaging Research, University of California, San Diego, La Jolla, CA, USA
| | | | - Subhojit Roy
- Department of Pathology, University of California, San Diego, La Jolla, CA, USA; Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
49
|
Cross Talk between Viruses and Insect Cells Cytoskeleton. Viruses 2021; 13:v13081658. [PMID: 34452522 PMCID: PMC8402729 DOI: 10.3390/v13081658] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/18/2021] [Accepted: 08/19/2021] [Indexed: 12/18/2022] Open
Abstract
Viruses are excellent manipulators of host cellular machinery, behavior, and life cycle, with the host cell cytoskeleton being a primordial viral target. Viruses infecting insects generally enter host cells through clathrin-mediated endocytosis or membrane fusion mechanisms followed by transport of the viral particles to the corresponding replication sites. After viral replication, the viral progeny egresses toward adjacent cells and reaches the different target tissues. Throughout all these steps, actin and tubulin re-arrangements are driven by viruses. The mechanisms used by viruses to manipulate the insect host cytoskeleton are well documented in the case of alphabaculoviruses infecting Lepidoptera hosts and plant viruses infecting Hemiptera vectors, but they are not well studied in case of other insect-virus systems such as arboviruses-mosquito vectors. Here, we summarize the available knowledge on how viruses manipulate the insect host cell cytoskeleton, and we emphasize the primordial role of cytoskeleton components in insect virus motility and the need to expand the study of this interaction.
Collapse
|
50
|
Benwell CJ, Taylor JAGE, Robinson SD. Endothelial neuropilin-2 influences angiogenesis by regulating actin pattern development and α5-integrin-p-FAK complex recruitment to assembling adhesion sites. FASEB J 2021; 35:e21679. [PMID: 34314542 DOI: 10.1096/fj.202100286r] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 05/03/2021] [Accepted: 05/04/2021] [Indexed: 01/02/2023]
Abstract
The ability to form a variety of cell-matrix connections is crucial for angiogenesis to take place. Without stable anchorage to the extracellular matrix (ECM), endothelial cells (ECs) are unable to sense, integrate and disseminate growth factor stimulated responses that drive growth of a vascular bed. Neuropilin-2 (NRP2) is a widely expressed membrane-bound multifunctional non-tyrosine kinase receptor, which has previously been implicated in influencing cell adhesion and migration by interacting with α5-integrin and regulating adhesion turnover. α5-integrin, and its ECM ligand fibronectin (FN) are both known to be upregulated during the formation of neo-vasculature. Despite being descriptively annotated as a candidate biomarker for aggressive cancer phenotypes, the EC-specific roles for NRP2 during developmental and pathological angiogenesis remain unexplored. The data reported here support a model whereby NRP2 actively promotes EC adhesion and migration by regulating dynamic cytoskeletal remodeling and by stimulating Rab11-dependent recycling of α5-integrin-p-FAK complexes to newly assembling adhesion sites. Furthermore, temporal depletion of EC-NRP2 in vivo impairs primary tumor growth by disrupting vessel formation. We also demonstrate that EC-NRP2 is required for normal postnatal retinal vascular development, specifically by regulating cell-matrix adhesion. Upon loss of endothelial NRP2, vascular outgrowth from the optic nerve during superficial plexus formation is disrupted, likely due to reduced FAK phosphorylation within sprouting tip cells.
Collapse
Affiliation(s)
- Christopher J Benwell
- Gut Microbes and Health, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
| | - James A G E Taylor
- Gut Microbes and Health, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
| | - Stephen D Robinson
- Gut Microbes and Health, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK.,School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, UK
| |
Collapse
|