1
|
Sato Y, Yoshimura K, Matsuda K, Haraguchi T, Marumo A, Yamagishi M, Sato S, Ito K, Yajima J. Membrane-bound myosin IC drives the chiral rotation of the gliding actin filament around its longitudinal axis. Sci Rep 2023; 13:19908. [PMID: 37963943 PMCID: PMC10646037 DOI: 10.1038/s41598-023-47125-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 11/09/2023] [Indexed: 11/16/2023] Open
Abstract
Myosin IC, a single-headed member of the myosin I family, specifically interacts with anionic phosphatidylinositol 4,5-bisphosphate (PI[4,5]P2) in the cell membrane via the pleckstrin homology domain located in the myosin IC tail. Myosin IC is widely expressed and physically links the cell membrane to the actin cytoskeleton; it plays various roles in membrane-associated physiological processes, including establishing cellular chirality, lipid transportation, and mechanosensing. In this study, we evaluated the motility of full-length myosin IC of Drosophila melanogaster via the three-dimensional tracking of quantum dots bound to actin filaments that glided over a membrane-bound myosin IC-coated surface. The results revealed that myosin IC drove a left-handed rotational motion in the gliding actin filament around its longitudinal axis, indicating that myosin IC generated a torque perpendicular to the gliding direction of the actin filament. The quantification of the rotational motion of actin filaments on fluid membranes containing different PI(4,5)P2 concentrations revealed that the rotational pitch was longer at lower PI(4,5)P2 concentrations. These results suggest that the torque generated by membrane-bound myosin IC molecules can be modulated based on the phospholipid composition of the cell membrane.
Collapse
Affiliation(s)
- Yusei Sato
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo, 153-8902, Japan
| | - Kohei Yoshimura
- Department of Biology, Graduate School of Science, Chiba University, 1-33, Inage, Chiba, Japan
| | - Kyohei Matsuda
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo, 153-8902, Japan
| | - Takeshi Haraguchi
- Department of Biology, Graduate School of Science, Chiba University, 1-33, Inage, Chiba, Japan
| | - Akisato Marumo
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo, 153-8902, Japan
| | - Masahiko Yamagishi
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo, 153-8902, Japan
| | - Suguru Sato
- Department of Biology, Graduate School of Science, Chiba University, 1-33, Inage, Chiba, Japan
| | - Kohji Ito
- Department of Biology, Graduate School of Science, Chiba University, 1-33, Inage, Chiba, Japan.
| | - Junichiro Yajima
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo, 153-8902, Japan.
- Komaba Institute for Science, The University of Tokyo, 3-8-1, Komaba, Meguro-ku, Tokyo, 153-8902, Japan.
- Research Center for Complex Systems Biology, The University of Tokyo, 3-8-1, Komaba, Meguro-ku, Tokyo, 153-8902, Japan.
| |
Collapse
|
2
|
Diaz-Valencia JD, Estrada-Abreo LA, Rodríguez-Cruz L, Salgado-Aguayo AR, Patiño-López G. Class I Myosins, molecular motors involved in cell migration and cancer. Cell Adh Migr 2022; 16:1-12. [PMID: 34974807 PMCID: PMC8741282 DOI: 10.1080/19336918.2021.2020705] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 12/02/2021] [Accepted: 12/16/2021] [Indexed: 01/13/2023] Open
Abstract
Class I Myosins are a subfamily of motor proteins with ATPase activity and a characteristic structure conserved in all myosins: A N-Terminal Motor Domain, a central Neck and a C terminal Tail domain. Humans have eight genes for these myosins. Class I Myosins have different functions: regulate membrane tension, participate in endocytosis, exocytosis, intracellular trafficking and cell migration. Cell migration is influenced by many cellular components including motor proteins, like myosins. Recently has been reported that changes in myosin expression have an impact on the migration of cancer cells, the formation of infiltrates and metastasis. We propose that class I myosins might be potential markers for future diagnostic, prognostic or even as therapeutic targets in leukemia and other cancers.Abbreviations: Myo1g: Myosin 1g; ALL: Acute Lymphoblastic Leukemia, TH1: Tail Homology 1; TH2: Tail Homology 2; TH3: Tail Homology 3.
Collapse
Affiliation(s)
- Juan D. Diaz-Valencia
- Immunology and Proteomics Laboratory, Children’s Hospital of Mexico, Mexico City, Mexico
| | - Laura A. Estrada-Abreo
- Immunology and Proteomics Laboratory, Children’s Hospital of Mexico, Mexico City, Mexico
- Cell Biology and Flow Cytometry Laboratory, Metropolitan Autonomous University, México City, Mexico
| | - Leonor Rodríguez-Cruz
- Cell Biology and Flow Cytometry Laboratory, Metropolitan Autonomous University, México City, Mexico
| | - Alfonso R. Salgado-Aguayo
- Rheumatic Diseases Laboratory, National Institute of Respiratory Diseases “Ismael Cosío Villegas”, Mexico City, Mexico
| | - Genaro Patiño-López
- Immunology and Proteomics Laboratory, Children’s Hospital of Mexico, Mexico City, Mexico
| |
Collapse
|
3
|
Matozo T, Kogachi L, de Alencar BC. Myosin motors on the pathway of viral infections. Cytoskeleton (Hoboken) 2022; 79:41-63. [PMID: 35842902 DOI: 10.1002/cm.21718] [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: 04/27/2022] [Revised: 06/25/2022] [Accepted: 07/07/2022] [Indexed: 01/30/2023]
Abstract
Molecular motors are microscopic machines that use energy from adenosine triphosphate (ATP) hydrolysis to generate movement. While kinesins and dynein are molecular motors associated with microtubule tracks, myosins bind to and move on actin filaments. Mammalian cells express several myosin motors. They power cellular processes such as endo- and exocytosis, intracellular trafficking, transcription, migration, and cytokinesis. As viruses navigate through cells, they may take advantage or be hindered by host components and machinery, including the cytoskeleton. This review delves into myosins' cell roles and compares them to their reported functions in viral infections. In most cases, the previously described myosin functions align with their reported role in viral infections, although not in all cases. This opens the possibility that knowledge obtained from studying myosins in viral infections might shed light on new physiological roles for myosins in cells. However, given the high number of myosins expressed and the variety of viruses investigated in the different studies, it is challenging to infer whether the interactions found are specific to a single virus or can be applied to other viruses with the same characteristics. We conclude that the participation of myosins in viral cycles is still a largely unexplored area, especially concerning unconventional myosins.
Collapse
Affiliation(s)
- Tais Matozo
- Departamento de Imunologia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Leticia Kogachi
- Departamento de Imunologia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Bruna Cunha de Alencar
- Departamento de Imunologia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
4
|
Myosin 1D and the branched actin network control the condensation of p62 bodies. Cell Res 2022; 32:659-669. [PMID: 35477997 DOI: 10.1038/s41422-022-00662-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 04/01/2022] [Indexed: 11/08/2022] Open
Abstract
Biomolecular condensation driven by liquid-liquid phase separation (LLPS) is key to assembly of membraneless organelles in numerous crucial pathways. It is largely unknown how cellular structures or components spatiotemporally regulate LLPS and condensate formation. Here we reveal that cytoskeletal dynamics can control the condensation of p62 bodies comprising the autophagic adaptor p62/SQSTM1 and poly-ubiquitinated cargos. Branched actin networks are associated with p62 bodies and are required for their condensation. Myosin 1D, a branched actin-associated motor protein, drives coalescence of small nanoscale p62 bodies into large micron-scale condensates along the branched actin network. Impairment of actin cytoskeletal networks compromises the condensation of p62 bodies and retards substrate degradation by autophagy in both cellular models and Myosin 1D knockout mice. Coupling of LLPS scaffold to cytoskeleton systems may represent a general mechanism by which cells exert spatiotemporal control over phase condensation processes.
Collapse
|
5
|
Kulkarni R, Wiemer EAC, Chang W. Role of Lipid Rafts in Pathogen-Host Interaction - A Mini Review. Front Immunol 2022; 12:815020. [PMID: 35126371 PMCID: PMC8810822 DOI: 10.3389/fimmu.2021.815020] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Accepted: 12/31/2021] [Indexed: 12/25/2022] Open
Abstract
Lipid rafts, also known as microdomains, are important components of cell membranes and are enriched in cholesterol, glycophospholipids and receptors. They are involved in various essential cellular processes, including endocytosis, exocytosis and cellular signaling. Receptors are concentrated at lipid rafts, through which cellular signaling can be transmitted. Pathogens exploit these signaling mechanisms to enter cells, proliferate and egress. However, lipid rafts also play an important role in initiating antimicrobial responses by sensing pathogens via clustered pathogen-sensing receptors and triggering downstream signaling events such as programmed cell death or cytokine production for pathogen clearance. In this review, we discuss how both host and pathogens use lipid rafts and associated proteins in an arms race to survive. Special attention is given to the involvement of the major vault protein, the main constituent of a ribonucleoprotein complex, which is enriched in lipid rafts upon infection with vaccinia virus.
Collapse
Affiliation(s)
- Rakesh Kulkarni
- Molecular and Cell Biology, Taiwan International Graduate Program, National Defense Medical Center, Academia Sinica and Graduate Institute of Life Science, Taipei, Taiwan
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
- *Correspondence: Rakesh Kulkarni, ; Wen Chang,
| | - Erik A. C. Wiemer
- Medical Oncology, Erasmus MC Cancer Institute, University Medical Center, Rotterdam, Netherlands
| | - Wen Chang
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
- *Correspondence: Rakesh Kulkarni, ; Wen Chang,
| |
Collapse
|
6
|
Liu X, Bennison SA, Robinson L, Toyo-oka K. Responsible Genes for Neuronal Migration in the Chromosome 17p13.3: Beyond Pafah1b1(Lis1), Crk and Ywhae(14-3-3ε). Brain Sci 2021; 12:brainsci12010056. [PMID: 35053800 PMCID: PMC8774252 DOI: 10.3390/brainsci12010056] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 12/13/2021] [Accepted: 12/23/2021] [Indexed: 01/07/2023] Open
Abstract
The 17p13.3 chromosome region is often deleted or duplicated in humans, resulting in severe neurodevelopmental disorders such as Miller–Dieker syndrome (MDS) and 17p13.3 duplication syndrome. Lissencephaly can also be caused by gene mutations or deletions of a small piece of the 17p13.3 region, including a single gene or a few genes. PAFAH1B1 gene, coding for LIS1 protein, is a responsible gene for lissencephaly and MDS and regulates neuronal migration by controlling microtubules (MTs) and cargo transport along MTs via dynein. CRK is a downstream regulator of the reelin signaling pathways and regulates neuronal migration. YWHAE, coding for 14-3-3ε, is also responsible for MDS and regulates neuronal migration by binding to LIS1-interacting protein, NDEL1. Although these three proteins are known to be responsible for neuronal migration defects in MDS, there are 23 other genes in the MDS critical region on chromosome 17p13.3, and little is known about their functions in neurodevelopment, especially in neuronal migration. This review will summarize the recent progress on the functions of LIS1, CRK, and 14-3-3ε and describe the recent findings of other molecules in the MDS critical regions in neuronal migration.
Collapse
Affiliation(s)
- Xiaonan Liu
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19129, USA;
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, USA; (S.A.B.); (L.R.)
| | - Sarah A. Bennison
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, USA; (S.A.B.); (L.R.)
| | - Lozen Robinson
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, USA; (S.A.B.); (L.R.)
| | - Kazuhito Toyo-oka
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, USA; (S.A.B.); (L.R.)
- Correspondence: ; Tel.: +1-(215)-991-8288
| |
Collapse
|
7
|
mDia1 Assembles a Linear F-Actin Coat at Membrane Invaginations To Drive Listeria monocytogenes Cell-to-Cell Spreading. mBio 2021; 12:e0293921. [PMID: 34781738 PMCID: PMC8593688 DOI: 10.1128/mbio.02939-21] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Direct cell-to-cell spreading of Listeria monocytogenes requires the bacteria to induce actin-based finger-like membrane protrusions in donor host cells that are endocytosed through caveolin-rich membrane invaginations by adjacent receiving cells. An actin shell surrounds these endocytic sites; however, its structure, composition, and functional significance remain elusive. Here, we show that the formin mDia1, but surprisingly not the Arp2/3 complex, is enriched at the membrane invaginations generated by L. monocytogenes during HeLa and Jeg-3 cell infections. Electron microscopy reveals a band of linear actin filaments that run along the longitudinal axis of the invagination membrane. Mechanistically, mDia1 expression is vital for the assembly of this F-actin shell. mDia1 is also required for the recruitment of Filamin A, a caveola-associated F-actin cross-linking protein, and caveolin-1 to the invaginations. Importantly, mixed-cell infection assays show that optimal caveolin-based L. monocytogenes cell-to-cell spreading correlates with the formation of the linear actin filament-containing shell by mDia1. IMPORTANCE Listeria monocytogenes spreads from one cell to another to colonize tissues. This cell-to-cell movement requires the propulsive force of an actin-rich comet tail behind the advancing bacterium, which ultimately distends the host plasma membrane into a slender bacterium-containing membrane protrusion. These membrane protrusions induce a corresponding invagination in the membrane of the adjacent host cell. The host cell that receives the protrusion utilizes caveolin-based endocytosis to internalize the structures, and filamentous actin lines these membrane invaginations. Here, we set out to determine the structure and function of this filamentous actin "shell." We demonstrate that the formin mDia1, but not the Arp2/3 complex, localizes to the invaginations. Morphologically, we show that this actin is organized into linear arrays and not branched dendritic networks. Mechanistically, we show that the actin shell is assembled by mDia1 and that mDia1 is required for efficient cell-to-cell transfer of L. monocytogenes.
Collapse
|
8
|
Cook AW, Toseland CP. The roles of nuclear myosin in the DNA damage response. J Biochem 2021; 169:265-271. [PMID: 33035317 DOI: 10.1093/jb/mvaa113] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 09/22/2020] [Indexed: 02/06/2023] Open
Abstract
Myosin within the nucleus has often been overlooked due to their importance in cytoplasmic processes and a lack of investigation. However, more recently, it has been shown that their nuclear roles are just as fundamental to cell function and survival with roles in transcription, DNA damage and viral replication. Myosins can act as molecular transporters and anchors that rely on their actin binding and ATPase capabilities. Their roles within the DNA damage response can varies from a transcriptional response, moving chromatin and stabilizing chromosome contacts. This review aims to highlight their key roles in the DNA damage response and how they impact nuclear organization and transcription.
Collapse
Affiliation(s)
- Alexander W Cook
- Department of Oncology and Metabolism, University of Sheffield, Sheffield S10 2RX, UK
| | | |
Collapse
|
9
|
Folylpoly-ɣ-glutamate synthetase association to the cytoskeleton: Implications to folate metabolon compartmentalization. J Proteomics 2021; 239:104169. [PMID: 33676037 DOI: 10.1016/j.jprot.2021.104169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 02/03/2021] [Accepted: 02/19/2021] [Indexed: 11/23/2022]
Abstract
Folates are essential for nucleotide biosynthesis, amino acid metabolism and cellular proliferation. Following carrier-mediated uptake, folates are polyglutamylated by folylpoly-ɣ-glutamate synthetase (FPGS), resulting in their intracellular retention. FPGS appears as a long isoform, directed to mitochondria via a leader sequence, and a short isoform reported as a soluble cytosolic protein (cFPGS). However, since folates are labile and folate metabolism is compartmentalized, we herein hypothesized that cFPGS is associated with the cytoskeleton, to couple folate uptake and polyglutamylation and channel folate polyglutamates to metabolon compartments. We show that cFPGS is a cytoskeleton-microtubule associated protein: Western blot analysis revealed that endogenous cFPGS is associated with the insoluble cellular fraction, i.e., cytoskeleton and membranes, but not with the cytosol. Mass spectrometry analysis identified the putative cFPGS interactome primarily consisting of microtubule subunits and cytoskeletal motor proteins. Consistently, immunofluorescence microscopy with cytosol-depleted cells demonstrated the association of cFPGS with the cytoskeleton and unconventional myosin-1c. Furthermore, since anti-microtubule, anti-actin cytoskeleton, and coatomer dissociation-inducing agents yielded perinuclear pausing of cFPGS, we propose an actin- and microtubule-dependent transport of cFPGS between the ER-Golgi and the plasma membrane. These novel findings support the coupling of folate transport with polyglutamylation and folate channeling to intracellular metabolon compartments. SIGNIFICANCE: FPGS, an essential enzyme catalyzing intracellular folate polyglutamylation and efficient retention, was described as a soluble cytosolic enzyme in the past 40 years. However, based on the lability of folates and the compartmentalization of folate metabolism and nucleotide biosynthesis, we herein hypothesized that cytoplasmic FPGS is associated with the cytoskeleton, to couple folate transport and polyglutamylation as well as channel folate polyglutamates to biosynthetic metabolon compartments. Indeed, using complementary techniques including Mass-spectrometry proteomics and fluorescence microscopy, we show that cytoplasmic FPGS is associated with the cytoskeleton and unconventional myosin-1c. This novel cytoskeletal localization of cytoplasmic FPGS supports the dynamic channeling of polyglutamylated folates to metabolon compartments to avoid oxidation and intracellular dilution of folates, while enhancing folate-dependent de novo biosynthesis of nucleotides and DNA/protein methylation.
Collapse
|
10
|
Pillon M, Doublet P. Myosins, an Underestimated Player in the Infectious Cycle of Pathogenic Bacteria. Int J Mol Sci 2021; 22:ijms22020615. [PMID: 33435466 PMCID: PMC7826972 DOI: 10.3390/ijms22020615] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 01/04/2021] [Accepted: 01/06/2021] [Indexed: 12/15/2022] Open
Abstract
Myosins play a key role in many cellular processes such as cell migration, adhesion, intracellular trafficking and internalization processes, making them ideal targets for bacteria. Through selected examples, such as enteropathogenic E. coli (EPEC), Neisseria, Salmonella, Shigella, Listeria or Chlamydia, this review aims to illustrate how bacteria target and hijack host cell myosins in order to adhere to the cell, to enter the cell by triggering their internalization, to evade from the cytosolic autonomous cell defense, to promote the biogenesis of intracellular replicative niche, to disseminate in tissues by cell-to-cell spreading, to exit out the host cell, and also to evade from macrophage phagocytosis. It highlights the diversity and sophistication of the strategy evolved by bacteria to manipulate one of their privileged targets, the actin cytoskeleton.
Collapse
Affiliation(s)
- Margaux Pillon
- CIRI, Centre International de Recherche en Infectiologie, Legionella Pathogenesis Group, Université de Lyon, 69007 Lyon, France;
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1111, 69007 Lyon, France
- Ecole Normale Supérieure de Lyon, 69007 Lyon, France
- Centre International de Recherche en Infectiologie, Université Claude Bernard Lyon 1, 69007 Lyon, France
- Centre National de la Recherche Scientifique, UMR5308, 69007 Lyon, France
| | - Patricia Doublet
- CIRI, Centre International de Recherche en Infectiologie, Legionella Pathogenesis Group, Université de Lyon, 69007 Lyon, France;
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1111, 69007 Lyon, France
- Ecole Normale Supérieure de Lyon, 69007 Lyon, France
- Centre International de Recherche en Infectiologie, Université Claude Bernard Lyon 1, 69007 Lyon, France
- Centre National de la Recherche Scientifique, UMR5308, 69007 Lyon, France
- Correspondence:
| |
Collapse
|
11
|
Abstract
Myosins constitute a superfamily of actin-based molecular motor proteins that mediates a variety of cellular activities including muscle contraction, cell migration, intracellular transport, the formation of membrane projections, cell adhesion, and cell signaling. The 12 myosin classes that are expressed in humans share sequence similarities especially in the N-terminal motor domain; however, their enzymatic activities, regulation, ability to dimerize, binding partners, and cellular functions differ. It is becoming increasingly apparent that defects in myosins are associated with diseases including cardiomyopathies, colitis, glomerulosclerosis, neurological defects, cancer, blindness, and deafness. Here, we review the current state of knowledge regarding myosins and disease.
Collapse
|
12
|
Ju Y, Guo H, Edman M, Hamm-Alvarez SF. Application of advances in endocytosis and membrane trafficking to drug delivery. Adv Drug Deliv Rev 2020; 157:118-141. [PMID: 32758615 PMCID: PMC7853512 DOI: 10.1016/j.addr.2020.07.026] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 07/28/2020] [Accepted: 07/29/2020] [Indexed: 12/12/2022]
Abstract
Multidisciplinary research efforts in the field of drug delivery have led to the development of a variety of drug delivery systems (DDS) designed for site-specific delivery of diagnostic and therapeutic agents. Since efficient uptake of drug carriers into target cells is central to effective drug delivery, a comprehensive understanding of the biological pathways for cellular internalization of DDS can facilitate the development of DDS capable of precise tissue targeting and enhanced therapeutic outcomes. Diverse methods have been applied to study the internalization mechanisms responsible for endocytotic uptake of extracellular materials, which are also the principal pathways exploited by many DDS. Chemical inhibitors remain the most commonly used method to explore endocytotic internalization mechanisms, although genetic methods are increasingly accessible and may constitute more specific approaches. This review highlights the molecular basis of internalization pathways most relevant to internalization of DDS, and the principal methods used to study each route. This review also showcases examples of DDS that are internalized by each route, and reviews the general effects of biophysical properties of DDS on the internalization efficiency. Finally, options for intracellular trafficking and targeting of internalized DDS are briefly reviewed, representing an additional opportunity for multi-level targeting to achieve further specificity and therapeutic efficacy.
Collapse
Affiliation(s)
- Yaping Ju
- Department of Pharmacology and Pharmaceutical Sciences, USC School of Pharmacy, USA
| | - Hao Guo
- Department of Pharmacology and Pharmaceutical Sciences, USC School of Pharmacy, USA
| | - Maria Edman
- Department of Ophthalmology, Roski Eye Institute, Keck School of Medicine, University of Southern California, USA
| | - Sarah F Hamm-Alvarez
- Department of Pharmacology and Pharmaceutical Sciences, USC School of Pharmacy, USA; Department of Ophthalmology, Roski Eye Institute, Keck School of Medicine, University of Southern California, USA.
| |
Collapse
|
13
|
Tian Y, Wang Z, Wang Y, Yin B, Yuan J, Qiang B, Han W, Peng X. Glioma-derived endothelial cells promote glioma cells migration via extracellular vesicles-mediated transfer of MYO1C. Biochem Biophys Res Commun 2020; 525:S0006-291X(20)30283-7. [PMID: 32081419 DOI: 10.1016/j.bbrc.2020.02.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 02/04/2020] [Indexed: 12/11/2022]
Abstract
Extracellular vesicles (EV), as the intercellular information transfer molecules which can regulate the tumor microenvironment, promote migration and tumor progression. Previous studies reported that EV from endothelial cells was used to guide the fate and survival of gliomas, but many researches focus on normal human endothelial cells (NhEC) rather than tumor-derived endothelial cells. Our laboratory isolated human endothelial cells from glioma issue (GhEC). We have previously demonstrated that EV from GhEC and NhEC, which both can promote glioma stem cells (GSC) proliferation and tumorsphere formation in vitro and tumourigenicity in vivo by the transfer of CD9. However, NhEC-EV or GhEC-EV could suppress glioma cells (GC) proliferation in vitro. It demonstrates the undifferentiated impact of EV. Here, we first compared GhEC-EV proteins with NhEC-EV (Screening criteria: GhEC-EV/NhEC-EV, FC > 1.5), and obtained 70 differential expression proteins, most of which were associated with invasion and migration. We found that GhEC or GhEC-EV preferred promoting GC migration than treating with NhEC or NhEC-EV. In terms of mechanism, we further revealed that EV-mediated transfer of MYO1C induced glioma cell LN229 migration. Knockdown of MYO1C in GhEC or GhEC-EV suppressed this effect. Overexpression of MYO1C promoted migration on the contrary. MYO1C was also detected in glioma cerebrospinal fluid (CSF), which is more suitable as a liquid biopsy biomarker and contributes to early diagnosis and monitoring in glioma. Our findings provide a new protein-MYO1C in EV to target tumor blood vessels, and bring a new point-cut to the treatment of gliomablastoma (GBM).
Collapse
Affiliation(s)
- Yuan Tian
- State Key Laboratory of Medical Molecular Biology, Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Medical Primate Research Center, Neuroscience Center, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China
| | - Zhixing Wang
- State Key Laboratory of Medical Molecular Biology, Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Medical Primate Research Center, Neuroscience Center, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China
| | - Yuxin Wang
- State Key Laboratory of Medical Molecular Biology, Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Medical Primate Research Center, Neuroscience Center, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China
| | - Bin Yin
- State Key Laboratory of Medical Molecular Biology, Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Medical Primate Research Center, Neuroscience Center, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China
| | - Jiangang Yuan
- State Key Laboratory of Medical Molecular Biology, Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Medical Primate Research Center, Neuroscience Center, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China
| | - Boqin Qiang
- State Key Laboratory of Medical Molecular Biology, Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Medical Primate Research Center, Neuroscience Center, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China
| | - Wei Han
- State Key Laboratory of Medical Molecular Biology, Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Medical Primate Research Center, Neuroscience Center, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China.
| | - Xiaozhong Peng
- State Key Laboratory of Medical Molecular Biology, Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Medical Primate Research Center, Neuroscience Center, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China; Institute of Medical Biology, Chinese Academy of Medical Sciences, Peking Union Medical College, Kunming, China.
| |
Collapse
|
14
|
Ceramide Domains in Health and Disease: A Biophysical Perspective. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1159:79-108. [DOI: 10.1007/978-3-030-21162-2_6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
15
|
Roles of Myosin-Mediated Membrane Trafficking in TGF-β Signaling. Int J Mol Sci 2019; 20:ijms20163913. [PMID: 31408934 PMCID: PMC6719161 DOI: 10.3390/ijms20163913] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Revised: 08/08/2019] [Accepted: 08/09/2019] [Indexed: 12/17/2022] Open
Abstract
Recent findings have revealed the role of membrane traffic in the signaling of transforming growth factor-β (TGF-β). These findings originate from the pivotal function of TGF-β in development, cell proliferation, tumor metastasis, and many other processes essential in malignancy. Actin and unconventional myosin have crucial roles in subcellular trafficking of receptors; research has also revealed a growing number of unconventional myosins that have crucial roles in TGF-β signaling. Unconventional myosins modulate the spatial organization of endocytic trafficking and tether membranes or transport them along the actin cytoskeletons. Current models do not fully explain how membrane traffic forms a bridge between TGF-β and the downstream effectors that produce its functional responsiveness, such as cell migration. In this review, we present a brief overview of the current knowledge of the TGF-β signaling pathway and the molecular components that comprise the core pathway as follows: ligands, receptors, and Smad mediators. Second, we highlight key role(s) of myosin motor-mediated protein trafficking and membrane domain segregation in the modulation of the TGF-β signaling pathway. Finally, we review future challenges and provide future prospects in this field.
Collapse
|
16
|
Cota Teixeira S, Silva Lopes D, Santos da Silva M, Cordero da Luz FA, Cirilo Gimenes SN, Borges BC, Alves da Silva A, Alves Martins F, Alves Dos Santos M, Teixeira TL, Oliveira RA, de Melo Rodrigues Ávila V, Barbosa Silva MJ, Elias MC, Martin R, Vieira da Silva C, Knölker HJ. Pentachloropseudilin Impairs Angiogenesis by Disrupting the Actin Cytoskeleton, Integrin Trafficking and the Cell Cycle. Chembiochem 2019; 20:2390-2401. [PMID: 31026110 DOI: 10.1002/cbic.201900203] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Indexed: 12/21/2022]
Abstract
Class 1 myosins (Myo1s) were the first unconventional myosins identified and humans have eight known Myo1 isoforms. The Myo1 family is involved in the regulation of gene expression, cytoskeletal rearrangements, delivery of proteins to the cell surface, cell migration and spreading. Thus, the important role of Myo1s in different biological processes is evident. In this study, we have investigated the effects of pentachloropseudilin (PClP), a reversible and allosteric potent inhibitor of Myo1s, on angiogenesis. We demonstrated that treatment of cells with PClP promoted a decrease in the number of vessels. The observed inhibition of angiogenesis is likely to be related to the inhibition of cell proliferation, migration and adhesion, as well as to alteration of the actin cytoskeleton pattern, as shown on a PClP-treated HUVEC cell line. Moreover, we also demonstrated that PClP treatment partially prevented the delivery of integrins to the plasma membrane. Finally, we showed that PClP caused DNA strand breaks, which are probably repaired during the cell cycle arrest in the G1 phase. Taken together, our results suggest that Myo1s participate directly in the angiogenesis process.
Collapse
Affiliation(s)
- Samuel Cota Teixeira
- Department of Immunology, Biomedical Sciences Institute, Federal University of Uberlândia, Rua Piauí, Bloco 2B, sala 200, Campus Umuarama, Uberlândia, 38400-902, MG, Brazil
| | - Daiana Silva Lopes
- Multidisciplinary Institute of Health, Anísio Teixeira Campus, Federal University of Bahia, Rua Hormindo Barros, 58, Candeias, Vitória da Conquista, 45029-094, BA, Brazil
| | - Marcelo Santos da Silva
- Special Laboratory of Cell Cycle (LECC), Center of Toxins, Immune Response and Cell Signaling (CeTICS), Butantan Institute, Av. Vital Brasil, 1500 - Butantã, São Paulo, 05503-900, SP, Brazil.,The Wellcome Centre for Molecular Parasitology, Institute of Infection, Immunity and Inflammation, Sir Graeme Davies Building, University of Glasgow, 120 University Place, Glasgow, G12 8TA, UK
| | - Felipe Andrés Cordero da Luz
- Department of Immunology, Biomedical Sciences Institute, Federal University of Uberlândia, Rua Piauí, Bloco 2B, sala 200, Campus Umuarama, Uberlândia, 38400-902, MG, Brazil
| | - Sarah Natalie Cirilo Gimenes
- Imunopathology Laboratory, Butantan Institute, Av. Vital Brasil, 1500 - Butantã, São Paulo, 05503-900, SP, Brazil
| | - Bruna Cristina Borges
- Department of Immunology, Biomedical Sciences Institute, Federal University of Uberlândia, Rua Piauí, Bloco 2B, sala 200, Campus Umuarama, Uberlândia, 38400-902, MG, Brazil
| | - Aline Alves da Silva
- Department of Immunology, Biomedical Sciences Institute, Federal University of Uberlândia, Rua Piauí, Bloco 2B, sala 200, Campus Umuarama, Uberlândia, 38400-902, MG, Brazil
| | - Flávia Alves Martins
- Department of Immunology, Biomedical Sciences Institute, Federal University of Uberlândia, Rua Piauí, Bloco 2B, sala 200, Campus Umuarama, Uberlândia, 38400-902, MG, Brazil
| | - Marlus Alves Dos Santos
- Department of Immunology, Biomedical Sciences Institute, Federal University of Uberlândia, Rua Piauí, Bloco 2B, sala 200, Campus Umuarama, Uberlândia, 38400-902, MG, Brazil
| | - Thaise Lara Teixeira
- Department of Immunology, Biomedical Sciences Institute, Federal University of Uberlândia, Rua Piauí, Bloco 2B, sala 200, Campus Umuarama, Uberlândia, 38400-902, MG, Brazil
| | - Ricardo A Oliveira
- Medical School, Federal University of Uberlândia, Av. Pará, Bloco 2u, 1720 - Umuarama, Uberlândia, 38400-902, MG, Brazil
| | - Veridiana de Melo Rodrigues Ávila
- Institute of Biotechnology, Federal University of Uberlândia, Av. Pará, 1720 - Bloco 2E - Sala(s) 246 - Campus Umuarama, Uberlândia, 38405-320, MG, Brazil
| | - Marcelo José Barbosa Silva
- Department of Immunology, Biomedical Sciences Institute, Federal University of Uberlândia, Rua Piauí, Bloco 2B, sala 200, Campus Umuarama, Uberlândia, 38400-902, MG, Brazil
| | - Maria Carolina Elias
- Special Laboratory of Cell Cycle (LECC), Center of Toxins, Immune Response and Cell Signaling (CeTICS), Butantan Institute, Av. Vital Brasil, 1500 - Butantã, São Paulo, 05503-900, SP, Brazil
| | - René Martin
- Fakultät Chemie, Technische Universität Dresden, Bergstraße 66, 01069, Dresden, Germany
| | - Claudio Vieira da Silva
- Department of Immunology, Biomedical Sciences Institute, Federal University of Uberlândia, Rua Piauí, Bloco 2B, sala 200, Campus Umuarama, Uberlândia, 38400-902, MG, Brazil
| | - Hans-Joachim Knölker
- Fakultät Chemie, Technische Universität Dresden, Bergstraße 66, 01069, Dresden, Germany
| |
Collapse
|
17
|
Kumar M, Sharma S, Sil P, Kushwaha M, Mayor S, Vishwakarma RA, Singh PP. C-H Arylation of N
-Heteroarenes under Metal-Free Conditions and its Application towards the Synthesis of Pentabromo- and Pentachloropseudilins. European J Org Chem 2019. [DOI: 10.1002/ejoc.201900353] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Mukesh Kumar
- Medicinal Chemistry Division; Academy of Scientific and Innovative Research; CSIR-Indian Institute of Integrative Medicine; Canal Road -180001 Jammu India
| | - Shweta Sharma
- Medicinal Chemistry Division; Academy of Scientific and Innovative Research; CSIR-Indian Institute of Integrative Medicine; Canal Road -180001 Jammu India
| | - Parijat Sil
- National Centre for Biological Sciences; Tata Institute of Fundamental Research; GKVK; Bellary Road 560065 Bangalore- India
| | - Manoj Kushwaha
- Medicinal Chemistry Division; Academy of Scientific and Innovative Research; CSIR-Indian Institute of Integrative Medicine; Canal Road -180001 Jammu India
| | - Satyajit Mayor
- National Centre for Biological Sciences; Tata Institute of Fundamental Research; GKVK; Bellary Road 560065 Bangalore- India
| | - Ram A. Vishwakarma
- Medicinal Chemistry Division; Academy of Scientific and Innovative Research; CSIR-Indian Institute of Integrative Medicine; Canal Road -180001 Jammu India
| | - Parvinder Pal Singh
- Medicinal Chemistry Division; Academy of Scientific and Innovative Research; CSIR-Indian Institute of Integrative Medicine; Canal Road -180001 Jammu India
| |
Collapse
|
18
|
Arif E, Solanki AK, Srivastava P, Rahman B, Tash BR, Holzman LB, Janech MG, Martin R, Knölker HJ, Fitzgibbon WR, Deng P, Budisavljevic MN, Syn WK, Wang C, Lipschutz JH, Kwon SH, Nihalani D. The motor protein Myo1c regulates transforming growth factor-β-signaling and fibrosis in podocytes. Kidney Int 2019; 96:139-158. [PMID: 31097328 DOI: 10.1016/j.kint.2019.02.014] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 02/07/2019] [Accepted: 02/14/2019] [Indexed: 01/19/2023]
Abstract
Transforming growth factor-β (TGF-β) is known to play a critical role in the pathogenesis of many progressive podocyte diseases. However, the molecular mechanisms regulating TGF-β signaling in podocytes remain unclear. Using a podocyte-specific myosin (Myo)1c knockout, we demonstrate whether Myo1c is critical for TGF-β-signaling in podocyte disease pathogenesis. Specifically, podocyte-specific Myo1c knockout mice were resistant to fibrotic injury induced by Adriamycin or nephrotoxic serum. Further, loss of Myo1c also protected from injury in the TGF-β-dependent unilateral ureteral obstruction mouse model of renal interstitial fibrosis. Mechanistic analyses showed that loss of Myo1c significantly blunted TGF-β signaling through downregulation of canonical and non-canonical TGF-β pathways. Interestingly, nuclear rather than the cytoplasmic Myo1c was found to play a central role in controlling TGF-β signaling through transcriptional regulation. Differential expression analysis of nuclear Myo1c-associated gene promoters showed that nuclear Myo1c targeted the TGF-β responsive gene growth differentiation factor (GDF)-15 and directly bound to the GDF-15 promoter. Importantly, GDF15 was found to be involved in podocyte pathogenesis, where GDF15 was upregulated in glomeruli of patients with focal segmental glomerulosclerosis. Thus, Myo1c-mediated regulation of TGF-β-responsive genes is central to the pathogenesis of podocyte injury. Hence, inhibiting this process may have clinical application in treating podocytopathies.
Collapse
Affiliation(s)
- Ehtesham Arif
- Department of Medicine, Nephrology Division, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Ashish K Solanki
- Department of Medicine, Nephrology Division, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Pankaj Srivastava
- Department of Medicine, Nephrology Division, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Bushra Rahman
- Department of Medicine, Nephrology Division, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Brian R Tash
- Department of Medicine, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Lawrence B Holzman
- Department of Medicine, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Michael G Janech
- Department of Medicine, Nephrology Division, Medical University of South Carolina, Charleston, South Carolina, USA; College of Charleston, Charleston, South Carolina, USA
| | - René Martin
- Department of Chemistry, TU Dresden, Dresden, Germany
| | | | - Wayne R Fitzgibbon
- Department of Medicine, Nephrology Division, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Peifeng Deng
- Department of Medicine, Nephrology Division, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Milos N Budisavljevic
- Department of Medicine, Nephrology Division, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Wing-Kin Syn
- Department of Gastroenterology & Hepatology, Medical University of South Carolina, Charleston, South Carolina, USA; Section of Gastroenterology, Ralph H Johnson VA Medical Center, Charleston, South Carolina, USA; Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country, (UPV/EHU), Vizcaya, Spain
| | - Cindy Wang
- Department of Gastroenterology & Hepatology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Joshua H Lipschutz
- Department of Medicine, Nephrology Division, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Sang-Ho Kwon
- Department of Cellular Biology and Anatomy, Augusta University, Augusta, Georgia, USA
| | - Deepak Nihalani
- Department of Medicine, Nephrology Division, Medical University of South Carolina, Charleston, South Carolina, USA.
| |
Collapse
|
19
|
Girón-Pérez DA, Piedra-Quintero ZL, Santos-Argumedo L. Class I myosins: Highly versatile proteins with specific functions in the immune system. J Leukoc Biol 2019; 105:973-981. [PMID: 30821871 DOI: 10.1002/jlb.1mr0918-350rrr] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 02/04/2019] [Accepted: 02/06/2019] [Indexed: 12/20/2022] Open
Abstract
Connections established between cytoskeleton and plasma membrane are essential in cellular processes such as cell migration, vesicular trafficking, and cytokinesis. Class I myosins are motor proteins linking the actin-cytoskeleton with membrane phospholipids. Previous studies have implicated these molecules in cell functions including endocytosis, exocytosis, release of extracellular vesicles and the regulation of cell shape and membrane elasticity. In immune cells, those proteins also are involved in the formation and maintenance of immunological synapse-related signaling. Thus, these proteins are master regulators of actin cytoskeleton dynamics in different scenarios. Although the localization of class I myosins has been described in vertebrates, their functions, regulation, and mechanical properties are not very well understood. In this review, we focused on and summarized the current understanding of class I myosins in vertebrates with particular emphasis in leukocytes.
Collapse
Affiliation(s)
- Daniel Alberto Girón-Pérez
- Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México, México
| | - Zayda Lizbeth Piedra-Quintero
- Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México, México
| | - Leopoldo Santos-Argumedo
- Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México, México
| |
Collapse
|
20
|
Capmany A, Yoshimura A, Kerdous R, Caorsi V, Lescure A, Nery ED, Coudrier E, Goud B, Schauer K. MYO1C stabilizes actin and facilitates arrival of transport carriers at the Golgi apparatus. J Cell Sci 2019; 132:jcs.225029. [DOI: 10.1242/jcs.225029] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 03/01/2019] [Indexed: 12/22/2022] Open
Abstract
We aim to identify the myosin motor proteins that control trafficking at the Golgi apparatus. In addition to the known Golgi-associated myosins MYO6, MYO18A and MYH9 (myosin IIA), we identify MYO1C as a novel player at the Golgi. We demonstrate that depletion of MYO1C induces Golgi apparatus fragmentation and decompaction. MYO1C accumulates at dynamic structures around the Golgi apparatus that colocalize with Golgi-associated actin dots. MYO1C depletion leads to loss of cellular F-actin, and Golgi apparatus decompaction is also observed after the inhibition or loss of the Arp2/3 complex. We show that the functional consequences of MYO1C depletion is a delay in the arrival of incoming transport carriers, both from the anterograde and retrograde routes. We propose that MYO1C stabilizes actin at the Golgi apparatus facilitating the arrival of incoming transport carriers at the Golgi.
Collapse
Affiliation(s)
- Anahi Capmany
- Institut Curie, PSL Research University, Molecular Mechanisms of Intracellular Transport group, 75248 Paris Cedex 05, France
- Centre National de la Recherche Scientifique, Unité Mixte de Recherche144, 75005 Paris, France
| | - Azumi Yoshimura
- Institut Curie, PSL Research University, Molecular Mechanisms of Intracellular Transport group, 75248 Paris Cedex 05, France
- Centre National de la Recherche Scientifique, Unité Mixte de Recherche144, 75005 Paris, France
| | - Rachid Kerdous
- Institut Curie, PSL Research University, Molecular Mechanisms of Intracellular Transport group, 75248 Paris Cedex 05, France
- Centre National de la Recherche Scientifique, Unité Mixte de Recherche144, 75005 Paris, France
| | | | - Aurianne Lescure
- Institut Curie, PSL Research University, Molecular Mechanisms of Intracellular Transport group, 75248 Paris Cedex 05, France
- Department of Translational Research, BioPhenics High-Content Screening Laboratory, Cell and Tissue Imaging Facility (PICT-IBiSA), Paris, France
| | - Elaine Del Nery
- Institut Curie, PSL Research University, Molecular Mechanisms of Intracellular Transport group, 75248 Paris Cedex 05, France
- Department of Translational Research, BioPhenics High-Content Screening Laboratory, Cell and Tissue Imaging Facility (PICT-IBiSA), Paris, France
| | - Evelyne Coudrier
- Institut Curie, PSL Research University, Molecular Mechanisms of Intracellular Transport group, 75248 Paris Cedex 05, France
- Centre National de la Recherche Scientifique, Unité Mixte de Recherche144, 75005 Paris, France
| | - Bruno Goud
- Institut Curie, PSL Research University, Molecular Mechanisms of Intracellular Transport group, 75248 Paris Cedex 05, France
- Centre National de la Recherche Scientifique, Unité Mixte de Recherche144, 75005 Paris, France
| | - Kristine Schauer
- Institut Curie, PSL Research University, Molecular Mechanisms of Intracellular Transport group, 75248 Paris Cedex 05, France
- Centre National de la Recherche Scientifique, Unité Mixte de Recherche144, 75005 Paris, France
| |
Collapse
|
21
|
Truong D, Boddy KC, Canadien V, Brabant D, Fairn GD, D'Costa VM, Coyaud E, Raught B, Pérez-Sala D, Park WS, Heo WD, Grinstein S, Brumell JH. Salmonella
exploits host Rho GTPase signalling pathways through the phosphatase activity of SopB. Cell Microbiol 2018; 20:e12938. [DOI: 10.1111/cmi.12938] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 06/11/2018] [Accepted: 07/06/2018] [Indexed: 12/20/2022]
Affiliation(s)
- Dorothy Truong
- Cell Biology Program; Hospital for Sick Children; Toronto ON Canada
- Department of Molecular Genetics; University of Toronto; Toronto ON Canada
| | - Kirsten C. Boddy
- Cell Biology Program; Hospital for Sick Children; Toronto ON Canada
- Institute of Medical Science; University of Toronto; Toronto ON Canada
| | | | - Danielle Brabant
- Cell Biology Program; Hospital for Sick Children; Toronto ON Canada
| | - Gregory D. Fairn
- Institute of Medical Science; University of Toronto; Toronto ON Canada
- Keenan Research Centre for Biomedical Science; St. Michael's Hospital; Toronto ON Canada
| | | | - Etienne Coyaud
- Princess Margaret Cancer Centre; University Health Network; Toronto Ontario Canada
| | - Brian Raught
- Princess Margaret Cancer Centre; University Health Network; Toronto Ontario Canada
- Department of Medical Biophysics; University of Toronto; Toronto Ontario Canada
| | - Dolores Pérez-Sala
- Department of Structural and Chemical Biology; Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas; Madrid Spain
| | - Wei Sun Park
- Department of Biological Sciences; Korea Advanced Institute of Science and Technology (KAIST); Daejeon Republic of Korea
| | - Won Do Heo
- Department of Biological Sciences; Korea Advanced Institute of Science and Technology (KAIST); Daejeon Republic of Korea
- Center for Cognition and Sociality; Institute of Basic Science (IBS); Daejeon Republic of Korea
| | - Sergio Grinstein
- Cell Biology Program; Hospital for Sick Children; Toronto ON Canada
- Institute of Medical Science; University of Toronto; Toronto ON Canada
- Keenan Research Centre for Biomedical Science; St. Michael's Hospital; Toronto ON Canada
- Department of Biochemistry; University of Toronto; Toronto ON Canada
| | - John H. Brumell
- Cell Biology Program; Hospital for Sick Children; Toronto ON Canada
- Department of Molecular Genetics; University of Toronto; Toronto ON Canada
- Institute of Medical Science; University of Toronto; Toronto ON Canada
- Sickkids IBD Centre; Hospital for Sick Children; Toronto ON Canada
| |
Collapse
|
22
|
Weimer BC, Chen P, Desai PT, Chen D, Shah J. Whole Cell Cross-Linking to Discover Host-Microbe Protein Cognate Receptor/Ligand Pairs. Front Microbiol 2018; 9:1585. [PMID: 30072965 PMCID: PMC6060266 DOI: 10.3389/fmicb.2018.01585] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 06/26/2018] [Indexed: 12/31/2022] Open
Abstract
Bacterial surface ligands mediate interactions with the host cell during association that determines the specific outcome for the host–microbe association. The association begins with receptors on the host cell binding ligands on the microbial cell to form a partnership that initiates responses in both cells. Methods to determine the specific cognate partnerships are lacking. Determining these molecular interactions between the host and microbial surfaces are difficult, yet crucial in defining biologically important events that are triggered during association of the microbiome, and critical in defining the initiating signal from the host membrane that results in pathology or commensal association. In this study, we designed an approach to discover cognate host–microbe receptor/ligand pairs using a covalent cross-linking strategy with whole cells. Protein/protein cross-linking occurred when the interacting molecules were within 9–12 Å, allowing for identification of specific pairs of proteins from the host and microbe that define the molecular interaction during association. To validate the method three different bacteria with three previously known protein/protein partnerships were examined. The exact interactions were confirmed and led to discovery of additional partnerships that were not recognized as cognate partners, but were previously reported to be involved in bacterial interactions. Additionally, three unknown receptor/ligand partners were discovered and validated with in vitro infection assays by blocking the putative host receptor and deleting the bacterial ligand. Subsequently, Salmonella enterica sv. Typhimurium was cross-linked to differentiated colonic epithelial cells (caco-2) to discover four previously unknown host receptors bound to three previously undefined host ligands for Salmonella. This approach resulted in a priori discovery of previously unknown and biologically important molecules for host/microbe association that were casually reported to mediate bacterial invasion. The whole cell cross-linking approach promises to enable discovery of possible targets to modulate interaction of the microbiome with the host that are important in infection and commensalism, both of with initiate a host response.
Collapse
Affiliation(s)
- Bart C Weimer
- Department of Population Health and Reproduction, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| | - Poyin Chen
- Department of Population Health and Reproduction, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| | - Prerak T Desai
- Department of Population Health and Reproduction, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States.,Department of Dietetics, Nutrition and Food Sciences, Utah State University, Logan, UT, United States
| | - Dong Chen
- Department of Biology, Utah State University, Logan, UT, United States
| | - Jigna Shah
- Department of Population Health and Reproduction, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States.,Department of Dietetics, Nutrition and Food Sciences, Utah State University, Logan, UT, United States
| |
Collapse
|
23
|
Li L, Dong X, Peng F, Shen L. Integrin β1 regulates the invasion and radioresistance of laryngeal cancer cells by targeting CD147. Cancer Cell Int 2018; 18:80. [PMID: 29930482 PMCID: PMC5992723 DOI: 10.1186/s12935-018-0578-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 06/02/2018] [Indexed: 11/29/2022] Open
Abstract
Background Increased expression of integrin β1 has been reported to correlate with progression and therapy resistance in many types of cancers. The aim of this study was to investigate the effects of integrin β1 on the invasion and radioresistance of laryngeal cancer cells. Methods The expression of integrin β1 in the tumor specimens of laryngeal cancer patients was assessed by immunohistochemical assays. The invasion ability of laryngeal cancer cells was detected by transwell and wound healing assays. The radiosensitivity of laryngeal cancer cells was evaluated by flow cytometry and colony formation assays. Results High expression of integrin β1 was significantly associated with lymph node metastasis, TNM stage and poor clinical outcomes (all p < 0.05). Knockdown of integrin β1 in laryngeal cancer cells inhibited invasion and increased radiosensitivity. Mechanistically, these effects were caused by suppression of the downstream focal adhesion kinase (FAK)/cortactin pathway. In addition, integrin β1 could interact with CD147 and the antibody blockade of CD147 led to the deactivation of FAK/cortactin signaling. Further studies revealed that the interaction between integrin β1 and CD147 relied on intact lipid rafts. Disruption of lipid rafts by methyl beta cyclodextrin in laryngeal cancer cells was able to reverse integrin β1-mediated malignant phenotypes. Conclusions Integrin β1 has potential as a therapeutic target in prevention and treatment of laryngeal cancer.
Collapse
Affiliation(s)
- Li Li
- 1The Functional Science Laboratory, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000 Hubei People's Republic of China
| | - Xiaoxia Dong
- 2Department of Pharmacology, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000 Hubei People's Republic of China
| | - Feng Peng
- 3Department of Clinical Oncology, Taihe Hospital, Hubei University of Medicine, 30 South Renmin Road, Shiyan, 442000 Hubei People's Republic of China
| | - Li Shen
- 3Department of Clinical Oncology, Taihe Hospital, Hubei University of Medicine, 30 South Renmin Road, Shiyan, 442000 Hubei People's Republic of China
| |
Collapse
|
24
|
Abstract
Actin cytoskeleton dynamics play vital roles in most forms of intracellular trafficking by promoting the biogenesis and transport of vesicular cargoes. Mounting evidence indicates that actin dynamics and membrane-cytoskeleton scaffolds also have essential roles in macroautophagy, the process by which cellular waste is isolated inside specialized vesicles called autophagosomes for recycling and degradation. Branched actin polymerization is necessary for the biogenesis of autophagosomes from the endoplasmic reticulum (ER) membrane. Actomyosin-based transport is then used to feed the growing phagophore with pre-selected cargoes and debris derived from different membranous organelles inside the cell. Finally, mature autophagosomes detach from the ER membrane by an as yet unknown mechanism, undergo intracellular transport and then fuse with lysosomes, endosomes and multivesicular bodies through mechanisms that involve actin- and microtubule-mediated motility, cytoskeleton-membrane scaffolds and signaling proteins. In this review, we highlight the considerable progress made recently towards understanding the diverse roles of the cytoskeleton in autophagy.
Collapse
|
25
|
Chung CL, Wang SW, Martin R, Knölker HJ, Kao YC, Lin MH, Chen JJ, Huang YB, Wu DC, Chen CL. Pentachloropseudilin Inhibits Transforming Growth Factor-β (TGF-β) Activity by Accelerating Cell-Surface Type II TGF-β Receptor Turnover in Target Cells. Chembiochem 2018; 19:851-864. [DOI: 10.1002/cbic.201700693] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Indexed: 12/22/2022]
Affiliation(s)
- Chih-Ling Chung
- Department of Biological Sciences; National Sun Yat-sen University; Kaohsiung 80424 ROC Taiwan
| | - Shih-Wei Wang
- Department of Biological Sciences; National Sun Yat-sen University; Kaohsiung 80424 ROC Taiwan
| | - René Martin
- Department of Chemistry; Technische Universität Dresden; Bergstrasse 66 01069 Dresden Germany
| | - Hans-Joachim Knölker
- Department of Chemistry; Technische Universität Dresden; Bergstrasse 66 01069 Dresden Germany
| | - Yu-Chen Kao
- Department of Biological Sciences; National Sun Yat-sen University; Kaohsiung 80424 ROC Taiwan
| | - Ming-Hong Lin
- Department of Microbiology and Immunology; Faculty of Medicine; Kaohsiung Medical University Hospital; Kaohsiung 80708 ROC Taiwan
| | - Jih-Jung Chen
- Faculty of Pharmacy; School of Pharmaceutical Sciences; National Yang-Ming University; Taipei 11221 ROC Taiwan
| | - Yaw-Bin Huang
- Department of Biological Sciences; National Sun Yat-sen University; Kaohsiung 80424 ROC Taiwan
- Department of Pharmacy; School of Pharmacy; Kaohsiung Medical University; Kaohsiung 80708 ROC Taiwan
- Center for Stem Cell Research; Kaohsiung Medical University; Kaohsiung 80708 ROC Taiwan
| | - Deng-Chyang Wu
- Division of Gastroenterology; Department of Internal Medicine; Kaohsiung Medical University Hospital; Kaohsiung 80708 ROC Taiwan
- Center for Stem Cell Research; Kaohsiung Medical University; Kaohsiung 80708 ROC Taiwan
| | - Chun-Lin Chen
- Department of Biological Sciences; National Sun Yat-sen University; Kaohsiung 80424 ROC Taiwan
- Doctoral Degree Program in Marine Biotechnology; National Sun Yat-sen University and Academia Sinica; Kaohsiung 80424 ROC Taiwan
| |
Collapse
|
26
|
Tang G, Chen Y, Xu JR, Kistler HC, Ma Z. The fungal myosin I is essential for Fusarium toxisome formation. PLoS Pathog 2018; 14:e1006827. [PMID: 29357387 PMCID: PMC5794197 DOI: 10.1371/journal.ppat.1006827] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 02/01/2018] [Accepted: 12/18/2017] [Indexed: 01/12/2023] Open
Abstract
Myosin-I molecular motors are proposed to function as linkers between membranes and the actin cytoskeleton in several cellular processes, but their role in the biosynthesis of fungal secondary metabolites remain elusive. Here, we found that the myosin I of Fusarium graminearum (FgMyo1), the causal agent of Fusarium head blight, plays critical roles in mycotoxin biosynthesis. Inhibition of myosin I by the small molecule phenamacril leads to marked reduction in deoxynivalenol (DON) biosynthesis. FgMyo1 also governs translation of the DON biosynthetic enzyme Tri1 by interacting with the ribosome-associated protein FgAsc1. Disruption of the ATPase activity of FgMyo1 either by the mutation E420K, down-regulation of FgMyo1 expression or deletion of FgAsc1 results in reduced Tri1 translation. The DON biosynthetic enzymes Tri1 and Tri4 are mainly localized to subcellular structures known as toxisomes in response to mycotoxin induction and the FgMyo1-interacting protein, actin, participates in toxisome formation. The actin polymerization disruptor latrunculin A inhibits toxisome assembly. Consistent with this observation, deletion of the actin-associated proteins FgPrk1 and FgEnd3 also results in reduced toxisome formation. Unexpectedly, the FgMyo1-actin cytoskeleton is not involved in biosynthesis of another secondary metabolite tested. Taken together, this study uncovers a novel function of myosin I in regulating mycotoxin biosynthesis in filamentous fungi. The mycotoxin deoxynivalenol (DON) is the most frequently detected secondary metabolite produced by Fusarium graminearum and other Fusarium spp. To date, relatively few studies have addressed how mycotoxin biosynthesis occurs in fungal cells. Here we found that myosin I governs translation of DON biosynthetic enzyme Tri1 via interacting with the ribosome-associated protein FgAsc1. Moreover, the key DON biosynthetic enzymes Tri1 and Tri4 are mainly localized to the toxisomes derived from endoplasmic reticulum under toxin inducing conditions. We further found that the FgMyo1-actin cytoskeleton was involved in toxisome formation but not for the biosynthesis of another secondary metabolite tested. Taken together, these results indicate for the first time that myosin I plays critical roles in mycotoxin biosynthesis.
Collapse
Affiliation(s)
- Guangfei Tang
- Institute of Biotechnology, Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Zhejiang University, Hangzhou, China
| | - Yun Chen
- Institute of Biotechnology, Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Zhejiang University, Hangzhou, China
| | - Jin-Rong Xu
- Department of Botany and Plant Pathology, Purdue University, West Lafayette, Indiana, United States of America
| | - H. Corby Kistler
- Department of Plant Pathology, University of Minnesota, St. Paul, Minnesota, United States of America
| | - Zhonghua Ma
- Institute of Biotechnology, Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Zhejiang University, Hangzhou, China
- State Key Laboratory of Rice Biology, Zhejiang University, Hangzhou, China
- * E-mail:
| |
Collapse
|
27
|
McIntosh BB, Pyrpassopoulos S, Holzbaur ELF, Ostap EM. Opposing Kinesin and Myosin-I Motors Drive Membrane Deformation and Tubulation along Engineered Cytoskeletal Networks. Curr Biol 2018; 28:236-248.e5. [PMID: 29337076 DOI: 10.1016/j.cub.2017.12.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 09/29/2017] [Accepted: 12/07/2017] [Indexed: 01/22/2023]
Abstract
Microtubule and actin filament molecular motors such as kinesin-1 and myosin-Ic (Myo1c) transport and remodel membrane-bound vesicles; however, it is unclear how they coordinate to accomplish these tasks. We introduced kinesin-1- and Myo1c-bound giant unilamellar vesicles (GUVs) into a micropatterned in vitro cytoskeletal matrix modeled after the subcellular architecture where vesicular sorting and membrane remodeling are observed. This array was composed of sparse microtubules intersecting regions dense with actin filaments, and revealed that Myo1c-dependent tethering of GUVs enabled kinesin-1-driven membrane deformation and tubulation. Membrane remodeling at actin/microtubule intersections was modulated by lipid composition and the addition of the Bin-Amphiphysin-Rvs-domain (BAR-domain) proteins endophilin or FCH-domain-only (FCHo). Myo1c not only tethered microtubule-transported cargo, but also transported, deformed, and tubulated GUVs along actin filaments in a lipid-composition- and BAR-protein-responsive manner. These results suggest a mechanism for actin-based involvement in vesicular transport and remodeling of intracellular membranes, and implicate lipid composition as a key factor in determining whether vesicles will undergo transport, deformation, or tubulation driven by opposing actin and microtubule motors and BAR-domain proteins.
Collapse
Affiliation(s)
- Betsy B McIntosh
- The Pennsylvania Muscle Institute and Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-6085, USA; Center for Engineering MechanoBiology, University of Pennsylvania, Philadelphia, PA 19104-6085, USA
| | - Serapion Pyrpassopoulos
- The Pennsylvania Muscle Institute and Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-6085, USA; Center for Engineering MechanoBiology, University of Pennsylvania, Philadelphia, PA 19104-6085, USA
| | - Erika L F Holzbaur
- The Pennsylvania Muscle Institute and Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-6085, USA; Center for Engineering MechanoBiology, University of Pennsylvania, Philadelphia, PA 19104-6085, USA.
| | - E Michael Ostap
- The Pennsylvania Muscle Institute and Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-6085, USA; Center for Engineering MechanoBiology, University of Pennsylvania, Philadelphia, PA 19104-6085, USA.
| |
Collapse
|
28
|
López-Ortega O, Santos-Argumedo L. Myosin 1g Contributes to CD44 Adhesion Protein and Lipid Rafts Recycling and Controls CD44 Capping and Cell Migration in B Lymphocytes. Front Immunol 2017; 8:1731. [PMID: 29321775 PMCID: PMC5732150 DOI: 10.3389/fimmu.2017.01731] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 11/23/2017] [Indexed: 12/30/2022] Open
Abstract
Cell migration and adhesion are critical for immune system function and involve many proteins, which must be continuously transported and recycled in the cell. Recycling of adhesion molecules requires the participation of several proteins, including actin, tubulin, and GTPases, and of membrane components such as sphingolipids and cholesterol. However, roles of actin motor proteins in adhesion molecule recycling are poorly understood. In this study, we identified myosin 1g as one of the important motor proteins that drives recycling of the adhesion protein CD44 in B lymphocytes. We demonstrate that the lack of Myo1g decreases the cell-surface levels of CD44 and of the lipid raft surrogate GM1. In cells depleted of Myo1g, the recycling of CD44 was delayed, the delay seems to be caused at the level of formation of recycling complex and entry into recycling endosomes. Moreover, a defective lipid raft recycling in Myo1g-deficient cells had an impact both on the capping of CD44 and on cell migration. Both processes required the transportation of lipid rafts to the cell surface to deliver signaling components. Furthermore, the extramembrane was essential for cell expansion and remodeling of the plasma membrane topology. Therefore, Myo1g is important during the recycling of lipid rafts to the membrane and to the accompanied proteins that regulate plasma membrane plasticity. Thus, Myosin 1g contributes to cell adhesion and cell migration through CD44 recycling in B lymphocytes.
Collapse
Affiliation(s)
- Orestes López-Ortega
- Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México, México
| | - Leopoldo Santos-Argumedo
- Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México, México
| |
Collapse
|
29
|
Abstract
Macroautophagy is an intracellular pathway used for targeting of cellular components to the lysosome for their degradation and involves sequestration of cytoplasmic material into autophagosomes formed from a double membrane structure called the phagophore. The nucleation and elongation of the phagophore is tightly regulated by several autophagy-related (ATG) proteins, but also involves vesicular trafficking from different subcellular compartments to the forming autophagosome. Such trafficking must be tightly regulated by various intra- and extracellular signals to respond to different cellular stressors and metabolic states, as well as the nature of the cargo to become degraded. We are only starting to understand the interconnections between different membrane trafficking pathways and macroautophagy. This review will focus on the membrane trafficking machinery found to be involved in delivery of membrane, lipids, and proteins to the forming autophagosome and in the subsequent autophagosome fusion with endolysosomal membranes. The role of RAB proteins and their regulators, as well as coat proteins, vesicle tethers, and SNARE proteins in autophagosome biogenesis and maturation will be discussed.
Collapse
|
30
|
Zattelman L, Regev R, Ušaj M, Reinke PYA, Giese S, Samson AO, Taft MH, Manstein DJ, Henn A. N-terminal splicing extensions of the human MYO1C gene fine-tune the kinetics of the three full-length myosin IC isoforms. J Biol Chem 2017; 292:17804-17818. [PMID: 28893906 DOI: 10.1074/jbc.m117.794008] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 09/06/2017] [Indexed: 12/28/2022] Open
Abstract
The MYO1C gene produces three alternatively spliced isoforms, differing only in their N-terminal regions (NTRs). These isoforms, which exhibit both specific and overlapping nuclear and cytoplasmic functions, have different expression levels and nuclear-cytoplasmic partitioning. To investigate the effect of NTR extensions on the enzymatic behavior of individual isoforms, we overexpressed and purified the three full-length human isoforms from suspension-adapted HEK cells. MYO1CC favored the actomyosin closed state (AMC), MYO1C16 populated the actomyosin open state (AMO) and AMC equally, and MYO1C35 favored the AMO state. Moreover, the full-length constructs isomerized before ADP release, which has not been observed previously in truncated MYO1CC constructs. Furthermore, global numerical simulation analysis predicted that MYO1C35 populated the actomyosin·ADP closed state (AMDC) 5-fold more than the actomyosin·ADP open state (AMDO) and to a greater degree than MYO1CC and MYO1C16 (4- and 2-fold, respectively). On the basis of a homology model of the 35-amino acid NTR of MYO1C35 (NTR35) docked to the X-ray structure of MYO1CC, we predicted that MYO1C35 NTR residue Arg-21 would engage in a specific interaction with post-relay helix residue Glu-469, which affects the mechanics of the myosin power stroke. In addition, we found that adding the NTR35 peptide to MYO1CC yielded a protein that transiently mimics MYO1C35 kinetic behavior. By contrast, NTR35, which harbors the R21G mutation, was unable to confer MYO1C35-like kinetic behavior. Thus, the NTRs affect the specific nucleotide-binding properties of MYO1C isoforms, adding to their kinetic diversity. We propose that this level of fine-tuning within MYO1C broadens its adaptability within cells.
Collapse
Affiliation(s)
- Lilach Zattelman
- From the Faculty of Biology, Technion-Israel Institute of Technology, Haifa 3200003, Israel
| | - Ronit Regev
- From the Faculty of Biology, Technion-Israel Institute of Technology, Haifa 3200003, Israel
| | - Marko Ušaj
- From the Faculty of Biology, Technion-Israel Institute of Technology, Haifa 3200003, Israel
| | - Patrick Y A Reinke
- the Institute for Biophysical Chemistry, Hannover Medical School, OE 4350, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| | - Sven Giese
- the Institute for Biophysical Chemistry, Hannover Medical School, OE 4350, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| | - Abraham O Samson
- the Faculty of Medicine in the Galilee, Bar-Ilan University, Safed 1311520, Israel, and
| | - Manuel H Taft
- the Institute for Biophysical Chemistry, Hannover Medical School, OE 4350, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| | - Dietmar J Manstein
- the Institute for Biophysical Chemistry, Hannover Medical School, OE 4350, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| | - Arnon Henn
- From the Faculty of Biology, Technion-Israel Institute of Technology, Haifa 3200003, Israel,
| |
Collapse
|
31
|
Myosin isoform expressed in metastatic prostate cancer stimulates cell invasion. Sci Rep 2017; 7:8476. [PMID: 28814772 PMCID: PMC5559518 DOI: 10.1038/s41598-017-09158-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 07/21/2017] [Indexed: 12/15/2022] Open
Abstract
During metastasis, tumor cells migrate out of their original tissue to invade other organs. Secretion of exosomes and metalloproteases is essential for extracellular matrix remodeling, enabling migration through tissue barriers. Metastatic prostate cancer is differentiated by expression of the rare isoform A of the molecular motor myosin IC, however the function of this isoform remained unknown. Here we show that it contributes causatively to the invasive motility of prostate cancer cells. We found that the isoform associates with metalloprotease-containing exosomes and stimulates their secretion. While the data show that myosin IC is involved in prostate cancer cell migration, migration outside extracellular matrix in vitro proves little affected specifically by isoform A. Nevertheless, this isoform stimulates invasion through extracellular matrix, pointing to a critical role in secretion. Both the secretion and invasion depend on the integrity of the motor and lipid-binding domains of the protein. Our results demonstrate how myosin IC isoform A is likely to function in metastasis, driving secretion of exosomes that enable invasion of prostate cancer cells across extracellular matrix barriers. The new data identify a molecule suitable for a mechanistically grounded development into a marker and target for prognosis, detection, and treatment of invasive prostate cancer.
Collapse
|
32
|
Hume PJ, Singh V, Davidson AC, Koronakis V. Swiss Army Pathogen: The Salmonella Entry Toolkit. Front Cell Infect Microbiol 2017; 7:348. [PMID: 28848711 PMCID: PMC5552672 DOI: 10.3389/fcimb.2017.00348] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 07/21/2017] [Indexed: 02/04/2023] Open
Abstract
Salmonella causes disease in humans and animals ranging from mild self-limiting gastroenteritis to potentially life-threatening typhoid fever. Salmonellosis remains a considerable cause of morbidity and mortality globally, and hence imposes a huge socio-economic burden worldwide. A key property of all pathogenic Salmonella strains is the ability to invade non-phagocytic host cells. The major determinant of this invasiveness is a Type 3 Secretion System (T3SS), a molecular syringe that injects virulence effector proteins directly into target host cells. These effectors cooperatively manipulate multiple host cell signaling pathways to drive pathogen internalization. Salmonella does not only rely on these injected effectors, but also uses several other T3SS-independent mechanisms to gain entry into host cells. This review summarizes our current understanding of the methods used by Salmonella for cell invasion, with a focus on the host signaling networks that must be coordinately exploited for the pathogen to achieve its goal.
Collapse
Affiliation(s)
- Peter J Hume
- Department of Pathology, University of CambridgeCambridge, United Kingdom
| | - Vikash Singh
- Department of Pathology, University of CambridgeCambridge, United Kingdom
| | - Anthony C Davidson
- Department of Pathology, University of CambridgeCambridge, United Kingdom
| | - Vassilis Koronakis
- Department of Pathology, University of CambridgeCambridge, United Kingdom
| |
Collapse
|
33
|
Huang FC. The Role of Sphingolipids on Innate Immunity to Intestinal Salmonella Infection. Int J Mol Sci 2017; 18:ijms18081720. [PMID: 28783107 PMCID: PMC5578110 DOI: 10.3390/ijms18081720] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 07/25/2017] [Accepted: 07/27/2017] [Indexed: 02/06/2023] Open
Abstract
Salmonella spp. remains a major public health problem for the whole world. To reduce the use of antimicrobial agents and drug-resistant Salmonella, a better strategy is to explore alternative therapy rather than to discover another antibiotic. Sphingolipid- and cholesterol-enriched lipid microdomains attract signaling proteins and orchestrate them toward cell signaling and membrane trafficking pathways. Recent studies have highlighted the crucial role of sphingolipids in the innate immunity against infecting pathogens. It is therefore mandatory to exploit the role of the membrane sphingolipids in the innate immunity of intestinal epithelia infected by this pathogen. In the present review, we focus on the role of sphingolipids in the innate immunity of intestinal epithelia against Salmonella infection, including adhesion, autophagy, bactericidal effect, barrier function, membrane trafficking, cytokine and antimicrobial peptide expression. The intervention of sphingolipid-enhanced foods to make our life healthy or pharmacological agents regulating sphingolipids is provided at the end.
Collapse
Affiliation(s)
- Fu-Chen Huang
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan.
| |
Collapse
|
34
|
Gupta P, Martin R, Knölker HJ, Nihalani D, Kumar Sinha D. Myosin-1 inhibition by PClP affects membrane shape, cortical actin distribution and lipid droplet dynamics in early Zebrafish embryos. PLoS One 2017; 12:e0180301. [PMID: 28678859 PMCID: PMC5498032 DOI: 10.1371/journal.pone.0180301] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 06/13/2017] [Indexed: 12/22/2022] Open
Abstract
Myosin-1 (Myo1) represents a mechanical link between the membrane and actin-cytoskeleton in animal cells. We have studied the effect of Myo1 inhibitor PClP in 1-8 cell Zebrafish embryos. Our results indicate a unique involvement of Myo1 in early development of Zebrafish embryos. Inhibition of Myo1 (by PClP) and Myo2 (by Blebbistatin) lead to arrest in cell division. While Myo1 isoforms appears to be important for both the formation and the maintenance of cleavage furrows, Myo2 is required only for the formation of furrows. We found that the blastodisc of the embryo, which contains a thick actin cortex (~13 μm), is loaded with cortical Myo1. Myo1 appears to be crucial for maintaining the blastodisc morphology and the actin cortex thickness. In addition to cell division and furrow formation, inhibition of Myo1 has a drastic effect on the dynamics and distribution of lipid droplets (LDs) in the blastodisc near the cleavage furrow. All these results above are effects of Myo1 inhibition exclusively; Myo2 inhibition by blebbistatin does not show such phenotypes. Therefore, our results demonstrate a potential role for Myo1 in the maintenance and formation of furrow, blastodisc morphology, cell-division and LD organization within the blastodisc during early embryogenesis.
Collapse
MESH Headings
- Actin Cytoskeleton/drug effects
- Actin Cytoskeleton/metabolism
- Actins/genetics
- Actins/metabolism
- Animals
- Blastomeres/cytology
- Blastomeres/metabolism
- Blastomeres/ultrastructure
- Blotting, Western
- Cell Division/drug effects
- Cell Division/genetics
- Cell Membrane/metabolism
- Embryo, Nonmammalian/embryology
- Embryo, Nonmammalian/metabolism
- Embryo, Nonmammalian/ultrastructure
- Gene Expression Regulation, Developmental
- Heterocyclic Compounds, 4 or More Rings/pharmacology
- Hydrocarbons, Chlorinated/pharmacology
- Lipid Droplets/metabolism
- Microscopy, Electron, Scanning
- Microscopy, Fluorescence
- Myosin Heavy Chains/antagonists & inhibitors
- Myosin Heavy Chains/genetics
- Myosin Heavy Chains/metabolism
- Pyrroles/pharmacology
- Reverse Transcriptase Polymerase Chain Reaction
- Zebrafish/embryology
- Zebrafish/genetics
- Zebrafish/metabolism
- Zebrafish Proteins/antagonists & inhibitors
- Zebrafish Proteins/genetics
- Zebrafish Proteins/metabolism
Collapse
Affiliation(s)
| | - René Martin
- Department Chemie, TU Dresden, Dresden, Germany
| | | | - Deepak Nihalani
- Dept. Medicine, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | | |
Collapse
|
35
|
Sala S, Van Troys M, Medves S, Catillon M, Timmerman E, Staes A, Schaffner-Reckinger E, Gevaert K, Ampe C. Expanding the Interactome of TES by Exploiting TES Modules with Different Subcellular Localizations. J Proteome Res 2017; 16:2054-2071. [DOI: 10.1021/acs.jproteome.7b00034] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Stefano Sala
- Department of Biochemistry, Ghent University, 9000 Gent, Belgium
| | | | - Sandrine Medves
- Cytoskeleton
and Cell Plasticity Lab, Life Sciences Research Unit − FSTC, University of Luxembourg, 4365 Esch-sur-Alzette, Luxembourg
- Laboratory of Experimental Cancer Research, LIH, 1445 Strassen, Luxembourg
| | - Marie Catillon
- Cytoskeleton
and Cell Plasticity Lab, Life Sciences Research Unit − FSTC, University of Luxembourg, 4365 Esch-sur-Alzette, Luxembourg
| | - Evy Timmerman
- Department of Biochemistry, Ghent University, 9000 Gent, Belgium
- VIB Medical Biotechnology Center, 9000 Gent, Belgium
| | - An Staes
- Department of Biochemistry, Ghent University, 9000 Gent, Belgium
- VIB Medical Biotechnology Center, 9000 Gent, Belgium
| | - Elisabeth Schaffner-Reckinger
- Cytoskeleton
and Cell Plasticity Lab, Life Sciences Research Unit − FSTC, University of Luxembourg, 4365 Esch-sur-Alzette, Luxembourg
| | - Kris Gevaert
- Department of Biochemistry, Ghent University, 9000 Gent, Belgium
- VIB Medical Biotechnology Center, 9000 Gent, Belgium
| | - Christophe Ampe
- Department of Biochemistry, Ghent University, 9000 Gent, Belgium
| |
Collapse
|
36
|
MYO6 is targeted by Salmonella virulence effectors to trigger PI3-kinase signaling and pathogen invasion into host cells. Proc Natl Acad Sci U S A 2017; 114:3915-3920. [PMID: 28348208 DOI: 10.1073/pnas.1616418114] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
To establish infections, Salmonella injects virulence effectors that hijack the host actin cytoskeleton and phosphoinositide signaling to drive pathogen invasion. How effectors reprogram the cytoskeleton network remains unclear. By reconstituting the activities of the Salmonella effector SopE, we recapitulated Rho GTPase-driven actin polymerization at model phospholipid membrane bilayers in cell-free extracts and identified the network of Rho-recruited cytoskeleton proteins. Knockdown of network components revealed a key role for myosin VI (MYO6) in Salmonella invasion. SopE triggered MYO6 localization to invasion foci, and SopE-mediated activation of PAK recruited MYO6 to actin-rich membranes. We show that the virulence effector SopB requires MYO6 to regulate the localization of PIP3 and PI(3)P phosphoinositides and Akt activation. SopE and SopB target MYO6 to coordinate phosphoinositide production at invasion foci, facilitating the recruitment of cytoskeleton adaptor proteins to mediate pathogen uptake.
Collapse
|
37
|
Masters TA, Kendrick-Jones J, Buss F. Myosins: Domain Organisation, Motor Properties, Physiological Roles and Cellular Functions. Handb Exp Pharmacol 2017; 235:77-122. [PMID: 27757761 DOI: 10.1007/164_2016_29] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Myosins are cytoskeletal motor proteins that use energy derived from ATP hydrolysis to generate force and movement along actin filaments. Humans express 38 myosin genes belonging to 12 classes that participate in a diverse range of crucial activities, including muscle contraction, intracellular trafficking, cell division, motility, actin cytoskeletal organisation and cell signalling. Myosin malfunction has been implicated a variety of disorders including deafness, hypertrophic cardiomyopathy, Usher syndrome, Griscelli syndrome and cancer. In this chapter, we will first discuss the key structural and kinetic features that are conserved across the myosin family. Thereafter, we summarise for each member in turn its unique functional and structural adaptations, cellular roles and associated pathologies. Finally, we address the broad therapeutic potential for pharmacological interventions that target myosin family members.
Collapse
Affiliation(s)
- Thomas A Masters
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, CB2 0XY, UK.
| | | | - Folma Buss
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, CB2 0XY, UK
| |
Collapse
|
38
|
Visuttijai K, Pettersson J, Mehrbani Azar Y, van den Bout I, Örndal C, Marcickiewicz J, Nilsson S, Hörnquist M, Olsson B, Ejeskär K, Behboudi A. Lowered Expression of Tumor Suppressor Candidate MYO1C Stimulates Cell Proliferation, Suppresses Cell Adhesion and Activates AKT. PLoS One 2016; 11:e0164063. [PMID: 27716847 PMCID: PMC5055341 DOI: 10.1371/journal.pone.0164063] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2016] [Accepted: 09/18/2016] [Indexed: 12/12/2022] Open
Abstract
Myosin-1C (MYO1C) is a tumor suppressor candidate located in a region of recurrent losses distal to TP53. Myo1c can tightly and specifically bind to PIP2, the substrate of Phosphoinositide 3-kinase (PI3K), and to Rictor, suggesting a role for MYO1C in the PI3K pathway. This study was designed to examine MYO1C expression status in a panel of well-stratified endometrial carcinomas as well as to assess the biological significance of MYO1C as a tumor suppressor in vitro. We found a significant correlation between the tumor stage and lowered expression of MYO1C in endometrial carcinoma samples. In cell transfection experiments, we found a negative correlation between MYO1C expression and cell proliferation, and MYO1C silencing resulted in diminished cell migration and adhesion. Cells expressing excess of MYO1C had low basal level of phosphorylated protein kinase B (PKB, a.k.a. AKT) and cells with knocked down MYO1C expression showed a quicker phosphorylated AKT (pAKT) response in reaction to serum stimulation. Taken together the present study gives further evidence for tumor suppressor activity of MYO1C and suggests MYO1C mediates its tumor suppressor function through inhibition of PI3K pathway and its involvement in loss of contact inhibition.
Collapse
Affiliation(s)
- Kittichate Visuttijai
- School of Bioscience, Tumor Biology research group, University of Skövde, SE-541 28, Skövde, Sweden
- Department of Medical and Clinical Genetics, Sahlgrenska Academy, University of Gothenburg, SE-405 30, Gothenburg, Sweden
| | - Jennifer Pettersson
- Department of Medical and Clinical Genetics, Sahlgrenska Academy, University of Gothenburg, SE-405 30, Gothenburg, Sweden
| | - Yashar Mehrbani Azar
- School of Bioscience, Tumor Biology research group, University of Skövde, SE-541 28, Skövde, Sweden
| | - Iman van den Bout
- Department of physiology, Faculty of Health Sciences, University of Pretoria, Pretoria, 0007, South Africa
| | - Charlotte Örndal
- Department of Pathology, Sahlgrenska University Hospital, SE-413 45, Gothenburg, Sweden
| | - Janusz Marcickiewicz
- Department of Obstetrics and Gynecology, Halland Hospital Varberg, SE- 432 37, Varberg, Sweden
| | - Staffan Nilsson
- Institute of Mathematical Statistics, Chalmers University of Technology, SE-412 96, Gothenburg, Sweden
| | - Michael Hörnquist
- Department of Science and Technology, University of Linköping, ITN, SE-601 74, Norrköping, Sweden
| | - Björn Olsson
- School of Bioscience, Tumor Biology research group, University of Skövde, SE-541 28, Skövde, Sweden
| | - Katarina Ejeskär
- School of Bioscience, Tumor Biology research group, University of Skövde, SE-541 28, Skövde, Sweden
| | - Afrouz Behboudi
- School of Bioscience, Tumor Biology research group, University of Skövde, SE-541 28, Skövde, Sweden
- * E-mail:
| |
Collapse
|
39
|
Coutts AS, La Thangue NB. Regulation of actin nucleation and autophagosome formation. Cell Mol Life Sci 2016; 73:3249-63. [PMID: 27147468 PMCID: PMC4967107 DOI: 10.1007/s00018-016-2224-z] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 03/21/2016] [Accepted: 04/08/2016] [Indexed: 01/08/2023]
Abstract
Autophagy is a process of self-eating, whereby cytosolic constituents are enclosed by a double-membrane vesicle before delivery to the lysosome for degradation. This is an important process which allows for recycling of nutrients and cellular components and thus plays a critical role in normal cellular homeostasis as well as cell survival during stresses such as starvation or hypoxia. A large number of proteins regulate various stages of autophagy in a complex and still incompletely understood series of events. In this review, we will discuss recent studies which provide a growing body of evidence that actin dynamics and proteins that influence actin nucleation play an important role in the regulation of autophagosome formation and maturation.
Collapse
Affiliation(s)
- Amanda S Coutts
- Laboratory of Cancer Biology, Medical Sciences Division, Department of Oncology, University of Oxford, Old Road Campus Research Building, Old Road Campus, Off Roosevelt Drive, Oxford, OX3 7DQ, UK
| | - Nicholas B La Thangue
- Laboratory of Cancer Biology, Medical Sciences Division, Department of Oncology, University of Oxford, Old Road Campus Research Building, Old Road Campus, Off Roosevelt Drive, Oxford, OX3 7DQ, UK.
| |
Collapse
|
40
|
Venit T, Kalendová A, Petr M, Dzijak R, Pastorek L, Rohožková J, Malohlava J, Hozák P. Nuclear myosin I regulates cell membrane tension. Sci Rep 2016; 6:30864. [PMID: 27480647 PMCID: PMC4969604 DOI: 10.1038/srep30864] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 07/12/2016] [Indexed: 11/09/2022] Open
Abstract
Plasma membrane tension is an important feature that determines the cell shape and influences processes such as cell motility, spreading, endocytosis and exocytosis. Unconventional class 1 myosins are potent regulators of plasma membrane tension because they physically link the plasma membrane with adjacent cytoskeleton. We identified nuclear myosin 1 (NM1) - a putative nuclear isoform of myosin 1c (Myo1c) - as a new player in the field. Although having specific nuclear functions, NM1 localizes predominantly to the plasma membrane. Deletion of NM1 causes more than a 50% increase in the elasticity of the plasma membrane around the actin cytoskeleton as measured by atomic force microscopy. This higher elasticity of NM1 knock-out cells leads to 25% higher resistance to short-term hypotonic environment and rapid cell swelling. In contrast, overexpression of NM1 in wild type cells leads to an additional 30% reduction of their survival. We have shown that NM1 has a direct functional role in the cytoplasm as a dynamic linker between the cell membrane and the underlying cytoskeleton, regulating the degree of effective plasma membrane tension.
Collapse
Affiliation(s)
- Tomáš Venit
- Department of Biology of the Cell Nucleus, Institute of Molecular Genetics, AS CR, v.v.i., Videnska 1083, 142 20 Prague, Czech Republic.,Faculty of Science, Charles University in Prague, Albertov 6, 128 43 Prague, Czech Republic
| | - Alžběta Kalendová
- Department of Biology of the Cell Nucleus, Institute of Molecular Genetics, AS CR, v.v.i., Videnska 1083, 142 20 Prague, Czech Republic
| | - Martin Petr
- Department of Biology of the Cell Nucleus, Institute of Molecular Genetics, AS CR, v.v.i., Videnska 1083, 142 20 Prague, Czech Republic
| | - Rastislav Dzijak
- Department of Biology of the Cell Nucleus, Institute of Molecular Genetics, AS CR, v.v.i., Videnska 1083, 142 20 Prague, Czech Republic
| | - Lukáš Pastorek
- Department of Biology of the Cell Nucleus, Institute of Molecular Genetics, AS CR, v.v.i., Videnska 1083, 142 20 Prague, Czech Republic
| | - Jana Rohožková
- Department of Biology of the Cell Nucleus, Institute of Molecular Genetics, AS CR, v.v.i., Videnska 1083, 142 20 Prague, Czech Republic
| | - Jakub Malohlava
- Department of Medical Biophysics, Faculty of Medicine and Dentistry, Palacky University in Olomouc, Hnevotinska 3, 775 15 Olomouc, Czech Republic
| | - Pavel Hozák
- Department of Biology of the Cell Nucleus, Institute of Molecular Genetics, AS CR, v.v.i., Videnska 1083, 142 20 Prague, Czech Republic
| |
Collapse
|
41
|
Tunneling nanotubes mediate the transfer of stem cell marker CD133 between hematopoietic progenitor cells. Exp Hematol 2016; 44:1092-1112.e2. [PMID: 27473566 DOI: 10.1016/j.exphem.2016.07.006] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2016] [Revised: 07/15/2016] [Accepted: 07/16/2016] [Indexed: 12/24/2022]
Abstract
Deciphering all mechanisms of intercellular communication used by hematopoietic progenitors is important, not only for basic stem cell research, but also in view of their therapeutic relevance. Here, we investigated whether these cells can produce the thin F-actin-based plasma membrane protrusions referred to as tunneling nanotubes (TNTs), which are known to bridge cells over long distances without contact with the substratum and transfer cargo molecules along them in various biological processes. We found that human primary CD34+ hematopoietic progenitors and leukemic KG1a cells develop such structures upon culture on primary mesenchymal stromal cells or specific extracellular-matrix-based substrata. Time-lapse video microscopy revealed that cell dislodgement is the primary mechanism responsible for TNT biogenesis. Surprisingly, we found that, among various cluster of differentiation (CD) markers, only the stem cell antigen CD133 is transferred between cells. It is selectively and directionally transported along the surface of TNTs in small clusters, such as cytoplasmic phospho-myosin light chain 2, suggesting that the latter actin motor protein might be implicated in this process. Our data provide new insights into the biology of hematopoietic progenitors that can contribute to our understanding of all facets of intercellular communication in the bone marrow microenvironment under healthy or cancerous conditions.
Collapse
|
42
|
Abstract
Myosin-I molecular motors are proposed to play various cellular roles related to membrane dynamics and trafficking. In this Cell Science at a Glance article and the accompanying poster, we review and illustrate the proposed cellular functions of metazoan myosin-I molecular motors by examining the structural, biochemical, mechanical and cell biological evidence for their proposed molecular roles. We highlight evidence for the roles of myosin-I isoforms in regulating membrane tension and actin architecture, powering plasma membrane and organelle deformation, participating in membrane trafficking, and functioning as a tension-sensitive dock or tether. Collectively, myosin-I motors have been implicated in increasingly complex cellular phenomena, yet how a single isoform accomplishes multiple types of molecular functions is still an active area of investigation. To fully understand the underlying physiology, it is now essential to piece together different approaches of biological investigation. This article will appeal to investigators who study immunology, metabolic diseases, endosomal trafficking, cell motility, cancer and kidney disease, and to those who are interested in how cellular membranes are coupled to the underlying actin cytoskeleton in a variety of different applications.
Collapse
Affiliation(s)
- Betsy B McIntosh
- Pennsylvania Muscle Institute and Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-6085, USA
| | - E Michael Ostap
- Pennsylvania Muscle Institute and Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-6085, USA
| |
Collapse
|
43
|
Kruppa AJ, Kendrick-Jones J, Buss F. Myosins, Actin and Autophagy. Traffic 2016; 17:878-90. [PMID: 27146966 PMCID: PMC4957615 DOI: 10.1111/tra.12410] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 05/02/2016] [Accepted: 05/02/2016] [Indexed: 12/20/2022]
Abstract
Myosin motor proteins working together with the actin cytoskeleton drive a wide range of cellular processes. In this review, we focus on their roles in autophagy – the pathway the cell uses to ensure homeostasis by targeting pathogens, misfolded proteins and damaged organelles for degradation. The actin cytoskeleton regulated by a host of nucleating, anchoring and stabilizing proteins provides the filament network for the delivery of essential membrane vesicles from different cellular compartments to the autophagosome. Actin networks have also been implicated in structurally supporting the expanding phagophore, moving autophagosomes and enabling efficient fusion with the lysosome. Only a few myosins have so far been shown to play a role in autophagy. Non‐muscle myosin IIA functions in the early stages delivering membrane for the initial formation of the autophagosome, whereas myosin IC and myosin VI are involved in the final stages providing specific membranes for autophagosome maturation and its fusion with the lysosome.
Collapse
Affiliation(s)
- Antonina J Kruppa
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge Biomedical Campus, Wellcome Trust/MRC Building, Hills Road, Cambridge, CB2 0XY, UK
| | - John Kendrick-Jones
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge Biomedical Campus, Cambridge, CB2 0QH, UK
| | - Folma Buss
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge Biomedical Campus, Wellcome Trust/MRC Building, Hills Road, Cambridge, CB2 0XY, UK
| |
Collapse
|
44
|
Structural Analysis of the Myo1c and Neph1 Complex Provides Insight into the Intracellular Movement of Neph1. Mol Cell Biol 2016; 36:1639-54. [PMID: 27044863 DOI: 10.1128/mcb.00020-16] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Accepted: 03/21/2016] [Indexed: 11/20/2022] Open
Abstract
The Myo1c motor functions as a cargo transporter supporting various cellular events, including vesicular trafficking, cell migration, and stereociliary movements of hair cells. Although its partial crystal structures were recently described, the structural details of its interaction with cargo proteins remain unknown. This study presents the first structural demonstration of a cargo protein, Neph1, attached to Myo1c, providing novel insights into the role of Myo1c in intracellular movements of this critical slit diaphragm protein. Using small angle X-ray scattering studies, models of predominant solution conformation of unliganded full-length Myo1c and Myo1c bound to Neph1 were constructed. The resulting structures show an extended S-shaped Myo1c with Neph1 attached to its C-terminal tail. Importantly, binding of Neph1 did not induce a significant shape change in Myo1c, indicating this as a spontaneous process or event. Analysis of interaction surfaces led to the identification of a critical residue in Neph1 involved in binding to Myo1c. Indeed, a point mutant from this site abolished interaction between Neph1 and Myo1c when tested in the in vitro and in live-cell binding assays. Live-cell imaging, including fluorescence recovery after photobleaching, provided further support for the role of Myo1c in intracellular vesicular movement of Neph1 and its turnover at the membrane.
Collapse
|
45
|
López-Ortega O, Ovalle-García E, Ortega-Blake I, Antillón A, Chávez-Munguía B, Patiño-López G, Fragoso-Soriano R, Santos-Argumedo L. Myo1g is an active player in maintaining cell stiffness in B-lymphocytes. Cytoskeleton (Hoboken) 2016; 73:258-68. [DOI: 10.1002/cm.21299] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Revised: 04/11/2016] [Accepted: 04/13/2016] [Indexed: 12/11/2022]
Affiliation(s)
- O. López-Ortega
- Departamento De Biomedicina Molecular; Centro De Investigación Y De Estudios Avanzados Del Instituto Politécnico Nacional; Ciudad De México C. P. 07360 México
- Facultad De Medicina, Universidad Nacional Autónoma De México; Ciudad De México C. P. 04510 México
| | - E. Ovalle-García
- Universidad Autónoma De Nuevo León, UANL. Facultad De Ingeniería Mecánica Y Eléctrica, Av. Universidad S/N, Ciudad Universitaria, San Nicolás De Los Garza; Nuevo León C. P. 66451 México
| | - I. Ortega-Blake
- Instituto De Ciencias Físicas, UNAM; Cuernavaca Morelos C. P. 62210 México
| | - A. Antillón
- Instituto De Ciencias Físicas, UNAM; Cuernavaca Morelos C. P. 62210 México
| | - B. Chávez-Munguía
- Departamento De Infectómica Y Patogénesis Molecular; Centro De Investigación Y De Estudios Avanzados Del Instituto Politécnico Nacional; Ciudad De México C. P. 07360 México
| | - G. Patiño-López
- Laboratorio De Investigación En Inmunología Y Proteómica, Hospital Infantil De México, “Federico Gómez”; Ciudad De México C. P. 06720 México
| | - R. Fragoso-Soriano
- Departamento De Física; Centro De Investigación Y De Estudios Avanzados Del Instituto Politécnico Nacional; Ciudad De México C. P. 07360 México
| | - L. Santos-Argumedo
- Departamento De Biomedicina Molecular; Centro De Investigación Y De Estudios Avanzados Del Instituto Politécnico Nacional; Ciudad De México C. P. 07360 México
| |
Collapse
|
46
|
Abstract
Vertebrate myosin-IC (Myo1c) is a type-1 myosin that links cell membranes to the cytoskeleton via its actin-binding motor domain and its phosphatidylinositol 4,5-bisphosphate (PtdIns(4,5)P2)-binding tail domain. While it is known that Myo1c bound to PtdIns(4,5)P2 in fluid-lipid bilayers can propel actin filaments in an unloaded motility assay, its ability to develop forces against external load on actin while bound to fluid bilayers has not been explored. Using optical tweezers, we measured the diffusion coefficient of single membrane-bound Myo1c molecules by force-relaxation experiments, and the ability of ensembles of membrane-bound Myo1c molecules to develop and sustain forces. To interpret our results, we developed a computational model that recapitulates the basic features of our experimental ensemble data and suggests that Myo1c ensembles can generate forces parallel to lipid bilayers, with larger forces achieved when the myosin works away from the plane of the membrane or when anchored to slowly diffusing regions.
Collapse
|
47
|
Kittelberger N, Breunig M, Martin R, Knölker HJ, Miklavc P. The role of myosin 1c and myosin 1b in surfactant exocytosis. J Cell Sci 2016; 129:1685-96. [PMID: 26940917 PMCID: PMC4852769 DOI: 10.1242/jcs.181313] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 02/25/2016] [Indexed: 12/19/2022] Open
Abstract
Actin and actin-associated proteins have a pivotal effect on regulated exocytosis in secretory cells and influence pre-fusion as well as post-fusion stages of exocytosis. Actin polymerization on secretory granules during the post-fusion phase (formation of an actin coat) is especially important in cells with large secretory vesicles or poorly soluble secretions. Alveolar type II (ATII) cells secrete hydrophobic lipo-protein surfactant, which does not easily diffuse from fused vesicles. Previous work showed that compression of actin coat is necessary for surfactant extrusion. Here, we investigate the role of class 1 myosins as possible linkers between actin and membranes during exocytosis. Live-cell microscopy showed translocation of fluorescently labeled myosin 1b and myosin 1c to the secretory vesicle membrane after fusion. Myosin 1c translocation was dependent on its pleckstrin homology domain. Expression of myosin 1b and myosin 1c constructs influenced vesicle compression rate, whereas only the inhibition of myosin 1c reduced exocytosis. These findings suggest that class 1 myosins participate in several stages of ATII cell exocytosis and link actin coats to the secretory vesicle membrane to influence vesicle compression.
Collapse
Affiliation(s)
- Nadine Kittelberger
- Institute of General Physiology, Ulm University, Albert-Einstein Allee 11, Ulm 89081, Germany
| | - Markus Breunig
- Institute of General Physiology, Ulm University, Albert-Einstein Allee 11, Ulm 89081, Germany
| | - René Martin
- Department of Chemistry, Technische Universität Dresden, Bergstr. 66, Dresden 01069, Germany
| | - Hans-Joachim Knölker
- Department of Chemistry, Technische Universität Dresden, Bergstr. 66, Dresden 01069, Germany
| | - Pika Miklavc
- Institute of General Physiology, Ulm University, Albert-Einstein Allee 11, Ulm 89081, Germany
| |
Collapse
|
48
|
Brandstaetter H, Kishi-Itakura C, Tumbarello DA, Manstein DJ, Buss F. Loss of functional MYO1C/myosin 1c, a motor protein involved in lipid raft trafficking, disrupts autophagosome-lysosome fusion. Autophagy 2015; 10:2310-23. [PMID: 25551774 PMCID: PMC4502697 DOI: 10.4161/15548627.2014.984272] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
MYO1C, a single-headed class I myosin, associates with cholesterol-enriched lipid rafts and facilitates their recycling from intracellular compartments to the cell surface. Absence of functional MYO1C disturbs the cellular distribution of lipid rafts, causes the accumulation of cholesterol-enriched membranes in the perinuclear recycling compartment, and leads to enlargement of endolysosomal membranes. Several feeder pathways, including classical endocytosis but also the autophagy pathway, maintain the health of the cell by selective degradation of cargo through fusion with the lysosome. Here we show that loss of functional MYO1C leads to an increase in total cellular cholesterol and its disrupted subcellular distribution. We observe an accumulation of autophagic structures caused by a block in fusion with the lysosome and a defect in autophagic cargo degradation. Interestingly, the loss of MYO1C has no effect on degradation of endocytic cargo such as EGFR, illustrating that although the endolysosomal compartment is enlarged in size, it is functional, contains active hydrolases, and the correct pH. Our results highlight the importance of correct lipid composition in autophagosomes and lysosomes to enable them to fuse. Ablating MYO1C function causes abnormal cholesterol distribution, which has a major selective impact on the autophagy pathway.
Collapse
Key Words
- BafA1, bafilomycin A1
- EGF, epidermal growth factor
- EGFR, epidermal growth factor receptor
- EM, electron microscopy
- GFP, green fluorescent protein
- KD, knockdown
- LAMP1, lysosomal-associated membrane protein 1
- LC3, microtubule-associated protein 1 light chain 3
- MVB, multivesicular body
- MYO1C, myosin IC
- PB, phosphate buffer
- PCIP, pentachloropseudilin
- PtdIns(4, 5)P2, phosphatidylinositol 4, 5-bisphosphate
- RFP, red fluorescent protein
- RPE, retinal pigment epithelium
- autophagy
- cholesterol
- electron microscopy
- lipid raft
- lysosome, MYO1C
Collapse
Affiliation(s)
- Hemma Brandstaetter
- a Cambridge Institute for Medical Research ; University of Cambridge ; Cambridge , UK
| | | | | | | | | |
Collapse
|
49
|
Hedberg Oldfors C, Dios DG, Linder A, Visuttijai K, Samuelson E, Karlsson S, Nilsson S, Behboudi A. Analysis of an independent tumor suppressor locus telomeric to Tp53 suggested Inpp5k and Myo1c as novel tumor suppressor gene candidates in this region. BMC Genet 2015; 16:80. [PMID: 26170120 PMCID: PMC4501283 DOI: 10.1186/s12863-015-0238-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Accepted: 06/23/2015] [Indexed: 01/26/2023] Open
Abstract
Background Several reports indicate a commonly deleted chromosomal region independent from, and distal to the TP53 locus in a variety of human tumors. In a previous study, we reported a similar finding in a rat tumor model for endometrial carcinoma (EC) and through developing a deletion map, narrowed the candidate region to 700 kb, harboring 19 genes. In the present work real-time qPCR analysis, Western blot, semi-quantitative qPCR, sequencing, promoter methylation analysis, and epigenetic gene expression restoration analyses (5-aza-2´-deoxycytidine and/or trichostatin A treatments) were used to analyze the 19 genes located within the candidate region in a panel of experimental tumors compared to control samples. Results Real-time qPCR analysis suggested Hic1 (hypermethylated in cancer 1), Inpp5k (inositol polyphosphate-5-phosphatase K; a.k.a. Skip, skeletal muscle and kidney enriched inositol phosphatase) and Myo1c (myosin 1c) as the best targets for the observed deletions. No mutation in coding sequences of these genes was detected, hence the observed low expression levels suggest a haploinsufficient mode of function for these potential tumor suppressor genes. Both Inpp5k and Myo1c were down regulated at mRNA and/or protein levels, which could be rescued in gene expression restoration assays. This could not be shown for Hic1. Conclusion Innp5k and Myo1c were identified as the best targets for the deletions in the region. INPP5K and MYO1C are located adjacent to each other within the reported independent region of tumor suppressor activity located at chromosome arm 17p distal to TP53 in human tumors. There is no earlier report on the potential tumor suppressor activity of INPP5K and MYO1C, however, overlapping roles in phosphoinositide (PI) 3-kinase/Akt signaling, known to be vital for the cell growth and survival, are reported for both. Moreover, there are reports on tumor suppressor activity of other members of the gene families that INPP5K and MYO1C belong to. Functional significance of these two candidate tumor suppressor genes in cancerogenesis pathways remains to be investigated. Electronic supplementary material The online version of this article (doi:10.1186/s12863-015-0238-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Carola Hedberg Oldfors
- Department of Medical and Clinical Genetics, Sahlgrenska Academy, University of Gothenburg, SE-40530, Gothenburg, Sweden.
| | - Diego Garcia Dios
- Department of Medical and Clinical Genetics, Sahlgrenska Academy, University of Gothenburg, SE-40530, Gothenburg, Sweden.
| | - Anna Linder
- Department of Medical and Clinical Genetics, Sahlgrenska Academy, University of Gothenburg, SE-40530, Gothenburg, Sweden.
| | - Kittichate Visuttijai
- Department of Medical and Clinical Genetics, Sahlgrenska Academy, University of Gothenburg, SE-40530, Gothenburg, Sweden. .,Tumor Biology Research Group, School of Bioscience, University of Skövde, SE-54128, Skövde, Sweden.
| | - Emma Samuelson
- Department of Medical and Clinical Genetics, Sahlgrenska Academy, University of Gothenburg, SE-40530, Gothenburg, Sweden.
| | - Sandra Karlsson
- Tumor Biology Research Group, School of Bioscience, University of Skövde, SE-54128, Skövde, Sweden.
| | - Staffan Nilsson
- Institute of Mathematical Statistics, Chalmers University of Technology, SE-41296, Gothenburg, Sweden.
| | - Afrouz Behboudi
- Tumor Biology Research Group, School of Bioscience, University of Skövde, SE-54128, Skövde, Sweden.
| |
Collapse
|
50
|
Echarri A, Del Pozo MA. Caveolae - mechanosensitive membrane invaginations linked to actin filaments. J Cell Sci 2015; 128:2747-58. [PMID: 26159735 DOI: 10.1242/jcs.153940] [Citation(s) in RCA: 126] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
An essential property of the plasma membrane of mammalian cells is its plasticity, which is required for sensing and transmitting of signals, and for accommodating the tensional changes imposed by its environment or its own biomechanics. Caveolae are unique invaginated membrane nanodomains that play a major role in organizing signaling, lipid homeostasis and adaptation to membrane tension. Caveolae are frequently associated with stress fibers, a major regulator of membrane tension and cell shape. In this Commentary, we discuss recent studies that have provided new insights into the function of caveolae and have shown that trafficking and organization of caveolae are tightly regulated by stress-fiber regulators, providing a functional link between caveolae and stress fibers. Furthermore, the tension in the plasma membrane determines the curvature of caveolae because they flatten at high tension and invaginate at low tension, thus providing a tension-buffering system. Caveolae also regulate multiple cellular pathways, including RhoA-driven actomyosin contractility and other mechanosensitive pathways, suggesting that caveolae could couple mechanotransduction pathways to actin-controlled changes in tension through their association with stress fibers. Therefore, we argue here that the association of caveolae with stress fibers could provide an important strategy for cells to deal with mechanical stress.
Collapse
Affiliation(s)
- Asier Echarri
- Integrin Signaling Laboratory, Cell Biology & Physiology Program, Cell & Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernández Almagro, 3, Madrid 28029, Spain
| | - Miguel A Del Pozo
- Integrin Signaling Laboratory, Cell Biology & Physiology Program, Cell & Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernández Almagro, 3, Madrid 28029, Spain
| |
Collapse
|