1
|
Vogel GF, Klee KMC, Demir AM, Garczarczyk-Asim D, Hess MW, Huber LA, Müller T, Janecke AR. Congenital enteropathy caused by ezrin deficiency. Hum Genet 2025; 144:505-514. [PMID: 40137958 PMCID: PMC12033192 DOI: 10.1007/s00439-025-02738-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 03/14/2025] [Indexed: 03/29/2025]
Abstract
Ezrin, encoded by EZR, is a central module of epithelial polarity and links membrane proteins to the actin cytoskeleton directly or indirectly through scaffold proteins in the epithelium. Ezrin knockout mice fail to thrive and do not survive past weaning. We identified a homozygous EZR loss-of-function (LoF) variant, c.356dup, by exome sequencing in an infant with intractable diarrhea and failure to thrive, who died from septicemia at 5 months of age. The variant localized within a homozygous region of 13.2 Mb in the proband, is consistent with inheritance identical-by-descent from the consanguineous parents, and segregated with disease in the proband's family. EZR transcript analyses in a heterozygous carrier showed that the variant triggers nonsense-mediated mRNA decay. Homozygous EZR LoF variants have not been reported in public databases. In this study, we generated a Caco-2 EZR knockout cell line to investigate the role of ezrin in human intestinal epithelia. Our analyses used electron and immunofluorescence microscopy to assess structural changes in the knockout cells. We observed significant disorganization of the terminal web region, microvillus rarefaction and abnormal branching. Furthermore, the absence of ezrin resulted in the mislocalization of the ezrin-interacting scaffold protein Na+/H + exchanger regulatory factor-1. In conclusion, this represents the first documentation of complete ezrin deficiency in humans, highlighting the essential and non-redundant functions of the protein in maintaining intestinal physiology.
Collapse
Affiliation(s)
- Georg F Vogel
- Department of Paediatrics I, Medical University of Innsbruck, Anichstrasse 35, Innsbruck, 6020, Austria
- Institute of Cell Biology, Biocenter, Medical University of Innsbruck, Innsbruck, 6020, Austria
| | - Katharina M C Klee
- Institute of Cell Biology, Biocenter, Medical University of Innsbruck, Innsbruck, 6020, Austria
| | - Arzu Meltem Demir
- Ankara University, Faculty of Medicine, Department of Pediatrics, Division of Pediatric Gastroenterology, Hepatology and Nutrition, Ankara, 06110, Türkiye
| | - Dorota Garczarczyk-Asim
- Department of Paediatrics I, Medical University of Innsbruck, Anichstrasse 35, Innsbruck, 6020, Austria
| | - Michael W Hess
- Institute of Histology und Embryology, Medical University of Innsbruck, Innsbruck, Austria
| | - Lukas A Huber
- Institute of Cell Biology, Biocenter, Medical University of Innsbruck, Innsbruck, 6020, Austria
| | - Thomas Müller
- Department of Paediatrics I, Medical University of Innsbruck, Anichstrasse 35, Innsbruck, 6020, Austria
| | - Andreas R Janecke
- Department of Paediatrics I, Medical University of Innsbruck, Anichstrasse 35, Innsbruck, 6020, Austria.
- Division of Human Genetics, Medical University of Innsbruck, Innsbruck, 6020, Austria.
| |
Collapse
|
2
|
Malik S, Ali SA, Mehdi AM, Raza A, Bashir S, Baig DN. A pilot study: Examining cytoskeleton gene expression profiles in Pakistani children with autism spectrum disorder. Int J Dev Neurosci 2024; 84:769-778. [PMID: 39285780 DOI: 10.1002/jdn.10372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 08/14/2024] [Accepted: 08/14/2024] [Indexed: 11/09/2024] Open
Abstract
BACKGROUND Finding effective pharmacological interventions to address the complex array of neurodevelopmental disorders is currently an urgent imperative within the scientific community as these conditions present significant challenges for patients and their families, often impacting cognitive, emotional, and social development. In this study, we aimed to explore non-invasive method to diagnose autism spectrum disorders (ASD) within Pakistan children population and to identify clinical drugs for its treatment. AIMS The current report outlines a comprehensive bidirectional investigation showcasing the successful utilization of saliva samples to quantify the expression patterns of profilins (PFN1, 2, and 3); and ERM (ezrin, radixin, and moesin) proteins; and additionally moesin pseudogene 1 and moesin pseudogene 1 antisense (MSNP1AS). Subsequently, these expression profiles were employed to forecast interactions between drugs and genes in children diagnosed with ASD. METHODS This study sought to delve into the intricate gene expression profiles using qualitative polymerase chain reaction of profilin isoforms (PFN1, 2, and 3) and ERM genes extracted from saliva samples obtained from children diagnosed with ASD. Through this analysis, we aimed to elucidate potential molecular mechanisms underlying ASD pathogenesis, shedding light on novel biomarkers and therapeutic targets for this complex neurological condition. (n = 22). Subsequently, we implemented a diagnostic model utilizing sparse partial least squares discriminant analysis (sPLS-DA) to predict drugs against our genes of interest. Furthermore, connectivity maps were developed to illustrate the predicted associations of 24 drugs with the genes expression. RESULTS Our study results showed varied expression profile of cytoskeleton linked genes. Similarly, sPLS-DA model precisely predicted drug to genes response. Sixteen of the examined drugs had significant positive correlations with the expression of the targeted genes whereas eight of the predicted drugs had shown negative correlations. CONCLUSION Here we report the role of cytoskeleton linked genes (PFN and ERM) in co-relation to ASD. Furthermore, variable yet significant quantitative expression of these genes successfully predicted drug-gene interactions as shown with the help of connectivity maps that can be used to support the clinical use of these drugs to treat individuals with ASD in future studies.
Collapse
Affiliation(s)
- Sana Malik
- Kauser Abdullah Malik School of Life Sciences, Forman Christian College (A Chartered University) Lahore, Lahore, Pakistan
| | - Syed Aoun Ali
- Australian Institute of Bioengineering and Nanotechnology, University of Queensland, Brisbane, Queensland, Australia
| | - Ahmed Murtaza Mehdi
- Diamantina Institute, Faculty of Medicine, Translational Research Institute, University of Queensland, Brisbane, Queensland, Australia
| | - Amir Raza
- Department of Biotechnology, Knowledge Unit of Science, University of Management and Technology (Sialkot Campus), Sialkot, Pakistan
| | - Shahid Bashir
- Neuroscience Center, King Fahad Specialist Hospital, Dammam, Saudi Arabia
| | - Deeba Noreen Baig
- Kauser Abdullah Malik School of Life Sciences, Forman Christian College (A Chartered University) Lahore, Lahore, Pakistan
- University of Western Australia, Perth, Western Australia, Australia
| |
Collapse
|
3
|
Do TL, Tachibana K, Yamamoto N, Ando K, Isoda T, Kihara T. Interaction of SARS-CoV-2 Spike protein with ACE2 induces cortical actin modulation, including dephosphorylation of ERM proteins and reduction of cortical stiffness. Hum Cell 2024; 38:3. [PMID: 39436480 DOI: 10.1007/s13577-024-01142-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 08/30/2024] [Indexed: 10/23/2024]
Abstract
Cell surface cortical actin is a regulatory target for viral infection. We aimed to investigate the effect of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection on host cell cortical stiffness, an indicator of cortical actin structure. The receptor-binding domain (RBD) of SARS-CoV-2 Spike (S) protein induced a reduction in cortical stiffness in ACE2-expressing cells. The interaction of RBD with ACE2 caused the inactivation of Ezrin/Radixin/Moesin (ERM) proteins. We further investigated the effects of the RBD of SARS-CoV-2 Omicron variants, BA.1 and BA.5. These RBDs influenced cortical stiffness depending on their affinity for ACE2. Our study provides the first evidence that the interaction of the SARS-CoV-2 S protein with ACE2 induces mechanobiological signals and attenuates the cortical actin.
Collapse
Affiliation(s)
- Thi Ly Do
- Department of Life and Environment Engineering, Faculty of Environmental Engineering, The University of Kitakyushu, 1-1 Hibikino, Wakamatsu, Kitakyushu, Fukuoka, 808-0135, Japan
| | - Kouichi Tachibana
- Division of Hematology and Oncology, Department of Internal Medicine, Tokai University School of Medicine, 143 Shimokasuya, Isehara, Kanagawa, 259-1193, Japan
| | - Norio Yamamoto
- Department of Microbiology, Tokai University School of Medicine, 143 Shimokasuya, Isehara, Kanagawa, 259-1193, Japan
| | - Kiyoshi Ando
- Division of Hematology and Oncology, Department of Internal Medicine, Tokai University School of Medicine, 143 Shimokasuya, Isehara, Kanagawa, 259-1193, Japan
| | - Takaaki Isoda
- Department of Life and Environment Engineering, Faculty of Environmental Engineering, The University of Kitakyushu, 1-1 Hibikino, Wakamatsu, Kitakyushu, Fukuoka, 808-0135, Japan
| | - Takanori Kihara
- Department of Life and Environment Engineering, Faculty of Environmental Engineering, The University of Kitakyushu, 1-1 Hibikino, Wakamatsu, Kitakyushu, Fukuoka, 808-0135, Japan.
| |
Collapse
|
4
|
De Belly H, Gallen AF, Strickland E, Estrada DC, Zager PJ, Burkhardt JK, Turlier H, Weiner OD. Long range mutual activation establishes Rho and Rac polarity during cell migration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.01.616161. [PMID: 40236007 PMCID: PMC11996577 DOI: 10.1101/2024.10.01.616161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
In migrating cells, the GTPase Rac organizes a protrusive front, whereas Rho organizes a contractile back. How these GTPases are appropriately positioned at the opposite poles of a migrating cell is unknown. Here we leverage optogenetics, manipulation of cell mechanics, and mathematical modeling to reveal a surprising long-range mutual activation of the front and back polarity programs that complements their well-known local mutual inhibition. This long-range activation is rooted in two distinct modes of mechanochemical crosstalk. Local Rac-based protrusion stimulates Rho activation at the opposite side of the cell via membrane tension-based activation of mTORC2. Conversely, local Rho-based contraction induces cortical-flow-based remodeling of membrane-to-cortex interactions leading to PIP2 release, PIP3 generation, and Rac activation at the opposite side of the cell. We develop a minimal unifying mechanochemical model of the cell to explain how this long-range mechanical facilitation complements local biochemical inhibition to enable robust global Rho and Rac partitioning. Finally, we validate the importance of this long-range facilitation in the context of chemoattractant-based cell polarization and migration in primary human lymphocytes. Our findings demonstrate that the actin cortex and plasma membrane function as an integrated mechanochemical system for long-range partitioning of Rac and Rho during cell migration and likely other cellular contexts.
Collapse
|
5
|
Cao X, Lake M, Van der Hoeven G, Claes Z, Del Pino García J, Lemaire S, Greiner EC, Karamanou S, Van Eynde A, Kettenbach AN, Natera de Benito D, Carrera García L, Hernando Davalillo C, Ortez C, Nascimento A, Urreizti R, Bollen M. SDS22 coordinates the assembly of holoenzymes from nascent protein phosphatase-1. Nat Commun 2024; 15:5359. [PMID: 38918402 PMCID: PMC11199634 DOI: 10.1038/s41467-024-49746-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 06/18/2024] [Indexed: 06/27/2024] Open
Abstract
SDS22 forms an inactive complex with nascent protein phosphatase PP1 and Inhibitor-3. SDS22:PP1:Inhibitor-3 is a substrate for the ATPase p97/VCP, which liberates PP1 for binding to canonical regulatory subunits. The exact role of SDS22 in PP1-holoenzyme assembly remains elusive. Here, we show that SDS22 stabilizes nascent PP1. In the absence of SDS22, PP1 is gradually lost, resulting in substrate hyperphosphorylation and a proliferation arrest. Similarly, we identify a female individual with a severe neurodevelopmental disorder bearing an unstable SDS22 mutant, associated with decreased PP1 levels. We furthermore find that SDS22 directly binds to Inhibitor-3 and that this is essential for the stable assembly of SDS22:PP1: Inhibitor-3, the recruitment of p97/VCP, and the extraction of SDS22 during holoenzyme assembly. SDS22 with a disabled Inhibitor-3 binding site co-transfers with PP1 to canonical regulatory subunits, thereby forming non-functional holoenzymes. Our data show that SDS22, through simultaneous interaction with PP1 and Inhibitor-3, integrates the major steps of PP1 holoenzyme assembly.
Collapse
Affiliation(s)
- Xinyu Cao
- Laboratory of Biosignaling & Therapeutics, KU Leuven Department of Cellular and Molecular Medicine, University of Leuven, B-3000, Leuven, Belgium
| | - Madryn Lake
- Laboratory of Biosignaling & Therapeutics, KU Leuven Department of Cellular and Molecular Medicine, University of Leuven, B-3000, Leuven, Belgium
| | - Gerd Van der Hoeven
- Laboratory of Biosignaling & Therapeutics, KU Leuven Department of Cellular and Molecular Medicine, University of Leuven, B-3000, Leuven, Belgium
| | - Zander Claes
- Laboratory of Biosignaling & Therapeutics, KU Leuven Department of Cellular and Molecular Medicine, University of Leuven, B-3000, Leuven, Belgium
| | - Javier Del Pino García
- Laboratory of Biosignaling & Therapeutics, KU Leuven Department of Cellular and Molecular Medicine, University of Leuven, B-3000, Leuven, Belgium
| | - Sarah Lemaire
- Laboratory of Biosignaling & Therapeutics, KU Leuven Department of Cellular and Molecular Medicine, University of Leuven, B-3000, Leuven, Belgium
| | - Elora C Greiner
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
- Dartmouth Cancer Center, Lebanon, NH, USA
| | - Spyridoula Karamanou
- Laboratory of Molecular Bacteriology, KU Leuven Department of Microbiology and Immunology, University of Leuven, Leuven, Belgium
| | - Aleyde Van Eynde
- Laboratory of Biosignaling & Therapeutics, KU Leuven Department of Cellular and Molecular Medicine, University of Leuven, B-3000, Leuven, Belgium
| | - Arminja N Kettenbach
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
- Dartmouth Cancer Center, Lebanon, NH, USA
| | | | - Laura Carrera García
- Neuromuscular Unit, Department of Neurology, Hospital Sant Joan de Deu, Barcelona, Spain
| | | | - Carlos Ortez
- Neuromuscular Unit, Department of Neurology, Hospital Sant Joan de Deu, Barcelona, Spain
| | - Andrés Nascimento
- Neuromuscular Unit, Department of Neurology, Hospital Sant Joan de Deu, Barcelona, Spain
| | - Roser Urreizti
- Clinical Biochemistry Department, Institut de Recerca Sant Joan de Deu, Hospital Sant Joan de Deu, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
| | - Mathieu Bollen
- Laboratory of Biosignaling & Therapeutics, KU Leuven Department of Cellular and Molecular Medicine, University of Leuven, B-3000, Leuven, Belgium.
| |
Collapse
|
6
|
Martinez-Mayer J, Brinkmeier ML, O'Connell SP, Ukagwu A, Marti MA, Miras M, Forclaz MV, Benzrihen MG, Cheung LYM, Camper SA, Ellsworth BS, Raetzman LT, Pérez-Millán MI, Davis SW. Knockout mice with pituitary malformations help identify human cases of hypopituitarism. Genome Med 2024; 16:75. [PMID: 38822427 PMCID: PMC11140907 DOI: 10.1186/s13073-024-01347-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 05/20/2024] [Indexed: 06/03/2024] Open
Abstract
BACKGROUND Congenital hypopituitarism (CH) and its associated syndromes, septo-optic dysplasia (SOD) and holoprosencephaly (HPE), are midline defects that cause significant morbidity for affected people. Variants in 67 genes are associated with CH, but a vast majority of CH cases lack a genetic diagnosis. Whole exome and whole genome sequencing of CH patients identifies sequence variants in genes known to cause CH, and in new candidate genes, but many of these are variants of uncertain significance (VUS). METHODS The International Mouse Phenotyping Consortium (IMPC) is an effort to establish gene function by knocking-out all genes in the mouse genome and generating corresponding phenotype data. We used mouse embryonic imaging data generated by the Deciphering Mechanisms of Developmental Disorders (DMDD) project to screen 209 embryonic lethal and sub-viable knockout mouse lines for pituitary malformations. RESULTS Of the 209 knockout mouse lines, we identified 51 that have embryonic pituitary malformations. These genes not only represent new candidates for CH, but also reveal new molecular pathways not previously associated with pituitary organogenesis. We used this list of candidate genes to mine whole exome sequencing data of a cohort of patients with CH, and we identified variants in two unrelated cases for two genes, MORC2 and SETD5, with CH and other syndromic features. CONCLUSIONS The screening and analysis of IMPC phenotyping data provide proof-of-principle that recessive lethal mouse mutants generated by the knockout mouse project are an excellent source of candidate genes for congenital hypopituitarism in children.
Collapse
Affiliation(s)
- Julian Martinez-Mayer
- Institute of Biosciences, Biotechnology and Translational Biology (iB3), University of Buenos Aires, Intendente Güiraldes 2160, Ciudad Universitaria, C1428EGA, Buenos Aires, Argentina
| | - Michelle L Brinkmeier
- Department of Human Genetics, University of Michigan, 1241 Catherine St., Ann Arbor, MI, 48109-5618, USA
| | - Sean P O'Connell
- Department of Biological Sciences, University of South Carolina, 715 Sumter St., Columbia, SC, 29208, USA
| | - Arnold Ukagwu
- Department of Physiology, Southern Illinois University, 1135 Lincoln Dr, Carbondale, IL, 62901, USA
| | - Marcelo A Marti
- Instituto de Química Biológica de La Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Mirta Miras
- Hospital De Niños de La Santísima Trinidad, Córdoba, Argentina
| | - Maria V Forclaz
- Servicio de Endocrinología, Hospital Posadas, Buenos Aires, Argentina
| | - Maria G Benzrihen
- Servicio de Endocrinología, Hospital Posadas, Buenos Aires, Argentina
| | - Leonard Y M Cheung
- Department of Human Genetics, University of Michigan, 1241 Catherine St., Ann Arbor, MI, 48109-5618, USA
- Department of Physiology and Biophyscis, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Sally A Camper
- Department of Human Genetics, University of Michigan, 1241 Catherine St., Ann Arbor, MI, 48109-5618, USA
| | - Buffy S Ellsworth
- Department of Physiology, Southern Illinois University, 1135 Lincoln Dr, Carbondale, IL, 62901, USA
| | - Lori T Raetzman
- Department of Molecular and Integrative Physiology, University of Illinois, Champaign-Urbana, Urbana, IL, 61801, USA
| | - Maria I Pérez-Millán
- Institute of Biosciences, Biotechnology and Translational Biology (iB3), University of Buenos Aires, Intendente Güiraldes 2160, Ciudad Universitaria, C1428EGA, Buenos Aires, Argentina.
| | - Shannon W Davis
- Department of Biological Sciences, University of South Carolina, 715 Sumter St., Columbia, SC, 29208, USA.
| |
Collapse
|
7
|
Di Meo D, Kundu T, Ravindran P, Shah B, Püschel AW. Pip5k1γ regulates axon formation by limiting Rap1 activity. Life Sci Alliance 2024; 7:e202302383. [PMID: 38438249 PMCID: PMC10912816 DOI: 10.26508/lsa.202302383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 02/26/2024] [Accepted: 02/26/2024] [Indexed: 03/06/2024] Open
Abstract
During their differentiation, neurons establish a highly polarized morphology by forming axons and dendrites. Cortical and hippocampal neurons initially extend several short neurites that all have the potential to become an axon. One of these neurites is then selected as the axon by a combination of positive and negative feedback signals that promote axon formation and prevent the remaining neurites from developing into axons. Here, we show that Pip5k1γ is required for the formation of a single axon as a negative feedback signal that regulates C3G and Rap1 through the generation of phosphatidylinositol-4,5-bisphosphate (PI(4,5)P2). Impairing the function of Pip5k1γ results in a hyper-activation of the Fyn/C3G/Rap1 pathway, which induces the formation of supernumerary axons. Application of a hyper-osmotic shock to modulate membrane tension has a similar effect, increasing Rap1 activity and inducing the formation of supernumerary axons. In both cases, the induction of supernumerary axons can be reverted by expressing constitutively active Pip5k. Our results show that PI(4,5)P2-dependent membrane properties limit the activity of C3G and Rap1 to ensure the extension of a single axon.
Collapse
Affiliation(s)
- Danila Di Meo
- Institut für Integrative Zellbiologie und Physiologie, Universität Münster, Münster, Germany
- Cells-in-Motion Interfaculty Center, University of Münster, Münster, Germany
| | - Trisha Kundu
- Institut für Integrative Zellbiologie und Physiologie, Universität Münster, Münster, Germany
- Cells-in-Motion Interfaculty Center, University of Münster, Münster, Germany
| | - Priyadarshini Ravindran
- Institut für Integrative Zellbiologie und Physiologie, Universität Münster, Münster, Germany
| | - Bhavin Shah
- Institut für Integrative Zellbiologie und Physiologie, Universität Münster, Münster, Germany
| | - Andreas W Püschel
- Institut für Integrative Zellbiologie und Physiologie, Universität Münster, Münster, Germany
- Cells-in-Motion Interfaculty Center, University of Münster, Münster, Germany
| |
Collapse
|
8
|
Arreola J, Pérez-Cornejo P, Segura-Covarrubias G, Corral-Fernández N, León-Aparicio D, Guzmán-Hernández ML. Function and Regulation of the Calcium-Activated Chloride Channel Anoctamin 1 (TMEM16A). Handb Exp Pharmacol 2024; 283:101-151. [PMID: 35768554 DOI: 10.1007/164_2022_592] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Various human tissues express the calcium-activated chloride channel Anoctamin 1 (ANO1), also known as TMEM16A. ANO1 allows the passive chloride flux that controls different physiological functions ranging from muscle contraction, fluid and hormone secretion, gastrointestinal motility, and electrical excitability. Overexpression of ANO1 is associated with pathological conditions such as hypertension and cancer. The molecular cloning of ANO1 has led to a surge in structural, functional, and physiological studies of the channel in several tissues. ANO1 is a homodimer channel harboring two pores - one in each monomer - that work independently. Each pore is activated by voltage-dependent binding of two intracellular calcium ions to a high-affinity-binding site. In addition, the binding of phosphatidylinositol 4,5-bisphosphate to sites scattered throughout the cytosolic side of the protein aids the calcium activation process. Furthermore, many pharmacological studies have established ANO1 as a target of promising compounds that could treat several illnesses. This chapter describes our current understanding of the physiological roles of ANO1 and its regulation under physiological conditions as well as new pharmacological compounds with potential therapeutic applications.
Collapse
Affiliation(s)
- Jorge Arreola
- Physics Institute, Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico.
| | - Patricia Pérez-Cornejo
- Department of Physiology and Biophysics, School of Medicine of Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico
| | - Guadalupe Segura-Covarrubias
- Physics Institute, Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico
- Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, OH, USA
| | - Nancy Corral-Fernández
- Department of Physiology and Biophysics, School of Medicine of Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico
| | - Daniel León-Aparicio
- Physics Institute, Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico
| | | |
Collapse
|
9
|
Quiroga X, Walani N, Disanza A, Chavero A, Mittens A, Tebar F, Trepat X, Parton RG, Geli MI, Scita G, Arroyo M, Le Roux AL, Roca-Cusachs P. A mechanosensing mechanism controls plasma membrane shape homeostasis at the nanoscale. eLife 2023; 12:e72316. [PMID: 37747150 PMCID: PMC10569792 DOI: 10.7554/elife.72316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 09/24/2023] [Indexed: 09/26/2023] Open
Abstract
As cells migrate and experience forces from their surroundings, they constantly undergo mechanical deformations which reshape their plasma membrane (PM). To maintain homeostasis, cells need to detect and restore such changes, not only in terms of overall PM area and tension as previously described, but also in terms of local, nanoscale topography. Here, we describe a novel phenomenon, by which cells sense and restore mechanically induced PM nanoscale deformations. We show that cell stretch and subsequent compression reshape the PM in a way that generates local membrane evaginations in the 100 nm scale. These evaginations are recognized by I-BAR proteins, which triggers a burst of actin polymerization mediated by Rac1 and Arp2/3. The actin polymerization burst subsequently re-flattens the evagination, completing the mechanochemical feedback loop. Our results demonstrate a new mechanosensing mechanism for PM shape homeostasis, with potential applicability in different physiological scenarios.
Collapse
Affiliation(s)
- Xarxa Quiroga
- Institute for Bioengineering of Catalonia, the Barcelona Institute of Technology (BIST)BarcelonaSpain
- Departament de Biomedicina, Unitat de Biofísica i Bioenginyeria, Facultat de Medicina i Ciències de la Salut, Universitat de BarcelonaBarcelonaSpain
| | - Nikhil Walani
- Department of Applied Mechanics, IIT DelhiNew DelhiIndia
| | - Andrea Disanza
- IFOM ETS - The AIRC Institute of Molecular OncologyMilanItaly
| | - Albert Chavero
- Departament de Biomedicina, Unitat de Biologia Cel·lular, Facultat de Medicina i Ciències de la Salut, Centre de Recerca Biomèdica CELLEX, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de BarcelonaBarcelonaSpain
| | - Alexandra Mittens
- Institute for Bioengineering of Catalonia, the Barcelona Institute of Technology (BIST)BarcelonaSpain
| | - Francesc Tebar
- Departament de Biomedicina, Unitat de Biologia Cel·lular, Facultat de Medicina i Ciències de la Salut, Centre de Recerca Biomèdica CELLEX, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de BarcelonaBarcelonaSpain
| | - Xavier Trepat
- Institute for Bioengineering of Catalonia, the Barcelona Institute of Technology (BIST)BarcelonaSpain
| | - Robert G Parton
- Institute for Molecular Bioscience and Centre for Microscopy and Microanalysis, University of QueenslandBrisbaneAustralia
| | | | - Giorgio Scita
- IFOM ETS - The AIRC Institute of Molecular OncologyMilanItaly
- Department of Oncology and Haemato-Oncology, University of MilanMilanItaly
| | - Marino Arroyo
- Institute for Bioengineering of Catalonia, the Barcelona Institute of Technology (BIST)BarcelonaSpain
- Universitat Politècnica de Catalunya (UPC), Campus Nord, Carrer de Jordi GironaBarcelonaSpain
- Centre Internacional de Mètodes Numèrics en Enginyeria (CIMNE)BarcelonaSpain
| | - Anabel-Lise Le Roux
- Institute for Bioengineering of Catalonia, the Barcelona Institute of Technology (BIST)BarcelonaSpain
| | - Pere Roca-Cusachs
- Institute for Bioengineering of Catalonia, the Barcelona Institute of Technology (BIST)BarcelonaSpain
- Departament de Biomedicina, Unitat de Biofísica i Bioenginyeria, Facultat de Medicina i Ciències de la Salut, Universitat de BarcelonaBarcelonaSpain
| |
Collapse
|
10
|
Mohamed OAA, Tesen HS, Hany M, Sherif A, Abdelwahab MM, Elnaggar MH. The role of hypoxia on prostate cancer progression and metastasis. Mol Biol Rep 2023; 50:3873-3884. [PMID: 36787054 PMCID: PMC10042974 DOI: 10.1007/s11033-023-08251-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 01/04/2023] [Indexed: 02/15/2023]
Abstract
Prostate cancer is the second most common cancer diagnosed in men and the fifth-leading cause of cancer death in men worldwide. Like any solid tumor, the hypoxic microenvironment of prostatic cancer drives hypoxia-inducible factors (HIFs) to mediate cell adaptions to hypoxic conditions. HIFs direct different signaling pathways such as PI3K/Akt/mTOR, NOX, and Wnt/β-Catenin to tumor progression depending on the degree of hypoxia. HIFs regulate cytoskeleton protein expression, promoting epithelial-mesenchymal transition (EMT), which occurs when cancer cells lose cell-to-cell adhesions and start invasion and metastasis. Through activating pathways, the hypoxic microenvironment maintains the self-renewal, potency, and anti-apoptotic function of prostate cancer cells and induces tumor metastasis and transformation. These pathways could serve as a potential target for prostate cancer therapy. HIFs increase the expression of androgen receptors on cancer cells maintaining the growth and survival of prostate cancer and the development of its castration resistance. In this review, we elaborate on the role of hypoxia in prostatic cancer pathogenesis and different hypoxia-induced mechanisms.
Collapse
Affiliation(s)
- Osama A A Mohamed
- Biotechnology Department, Faculty of Science, Mansoura University, Dakahlia, Egypt.,Biomedical Research Department, Tetraploid Team, Cairo, Egypt
| | - Heba S Tesen
- Faculty of Medicine, Ain Shams University, Cairo, Egypt.,Biomedical Research Department, Tetraploid Team, Cairo, Egypt
| | - Marwa Hany
- Biotechnology Department, Faculty of Science, Cairo University, Giza, Egypt
| | - Aya Sherif
- Chemistry & Microbiology Department, Faculty of Science, Cairo University, Giza, Egypt.,Biomedical Research Department, Tetraploid Team, Cairo, Egypt
| | - Maya Magdy Abdelwahab
- Faculty of Medicine, Helwan University, Cairo, Egypt. .,Biomedical Research Department, Tetraploid Team, Cairo, Egypt.
| | - Muhammed H Elnaggar
- Biochemistry Department, Faculty of Science, Ain Shams University, Cairo, Egypt.,Biomedical Research Department, Tetraploid Team, Cairo, Egypt
| |
Collapse
|
11
|
Aufderhorst-Roberts A, Staykova M. Scratching beyond the surface - minimal actin assemblies as tools to elucidate mechanical reinforcement and shape change. Emerg Top Life Sci 2022; 6:ETLS20220052. [PMID: 36541184 PMCID: PMC9788373 DOI: 10.1042/etls20220052] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 12/08/2022] [Accepted: 12/09/2022] [Indexed: 12/24/2022]
Abstract
The interaction between the actin cytoskeleton and the plasma membrane in eukaryotic cells is integral to a large number of functions such as shape change, mechanical reinforcement and contraction. These phenomena are driven by the architectural regulation of a thin actin network, directly beneath the membrane through interactions with a variety of binding proteins, membrane anchoring proteins and molecular motors. An increasingly common approach to understanding the mechanisms that drive these processes is to build model systems from reconstituted lipids, actin filaments and associated actin-binding proteins. Here we review recent progress in this field, with a particular emphasis on how the actin cytoskeleton provides mechanical reinforcement, drives shape change and induces contraction. Finally, we discuss potential future developments in the field, which would allow the extension of these techniques to more complex cellular processes.
Collapse
Affiliation(s)
| | - Margarita Staykova
- Centre for Materials Physics, Department of Physics, Durham University, Durham DH1 3LE, U.K
| |
Collapse
|
12
|
Hausrat TJ, Vogl C, Neef J, Schweizer M, Yee BK, Strenzke N, Kneussel M. Monoallelic loss of the F-actin-binding protein radixin facilitates startle reactivity and pre-pulse inhibition in mice. Front Cell Dev Biol 2022; 10:987691. [DOI: 10.3389/fcell.2022.987691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 11/11/2022] [Indexed: 11/29/2022] Open
Abstract
Hearing impairment is one of the most common disorders with a global burden and increasing prevalence in an ever-aging population. Previous research has largely focused on peripheral sensory perception, while the brain circuits of auditory processing and integration remain poorly understood. Mutations in the rdx gene, encoding the F-actin binding protein radixin (Rdx), can induce hearing loss in human patients and homozygous depletion of Rdx causes deafness in mice. However, the precise physiological function of Rdx in hearing and auditory information processing is still ill-defined. Here, we investigated consequences of rdx monoallelic loss in the mouse. Unlike the homozygous (−/−) rdx knockout, which is characterized by the degeneration of actin-based stereocilia and subsequent hearing loss, our analysis of heterozygous (+/−) mutants has revealed a different phenotype. Specifically, monoallelic loss of rdx potentiated the startle reflex in response to acoustic stimulation of increasing intensities, suggesting a gain of function relative to wildtype littermates. The monoallelic loss of the rdx gene also facilitated pre-pulse inhibition of the acoustic startle reflex induced by weak auditory pre-pulse stimuli, indicating a modification to the circuit underlying sensorimotor gating of auditory input. However, the auditory brainstem response (ABR)-based hearing thresholds revealed a mild impairment in peripheral sound perception in rdx (+/-) mice, suggesting minor aberration of stereocilia structural integrity. Taken together, our data suggest a critical role of Rdx in the top-down processing and/or integration of auditory signals, and therefore a novel perspective to uncover further Rdx-mediated mechanisms in central auditory information processing.
Collapse
|
13
|
Winkenbach LP, Parker DM, Williams RTP, Nishimura EO. The ERM-1 membrane-binding domain directs erm-1 mRNA localization to the plasma membrane in the C. elegans embryo. Development 2022; 149:279335. [PMID: 36314842 PMCID: PMC9793419 DOI: 10.1242/dev.200930] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 10/10/2022] [Indexed: 11/06/2022]
Abstract
mRNA localization and transport are integral in regulating gene expression. In Caenorhabditis elegans embryos, the maternally inherited mRNA erm-1 (Ezrin/Radixin/Moesin) becomes concentrated in anterior blastomeres. erm-1 mRNA localizes within those blastomeres to the plasma membrane where the essential ERM-1 protein, a membrane-actin linker, is also found. We demonstrate that the localization of erm-1 mRNA to the plasma membrane is translation dependent and requires its encoded N-terminal, membrane-binding (FERM) domain. By perturbing translation through multiple methods, we found that erm-1 mRNA localization at the plasma membrane persisted only if the nascent peptide remained in complex with the translating mRNA. Indeed, re-coding the erm-1 mRNA coding sequence while preserving the encoded amino acid sequence did not disrupt erm-1 mRNA localization, corroborating that the information directing mRNA localization resides within its membrane-binding protein domain. A single-molecule inexpensive fluorescence in situ hybridization screen of 17 genes encoding similar membrane-binding domains identified three plasma membrane-localized mRNAs in the early embryo. Ten additional transcripts showed potential membrane localization later in development. These findings point to a translation-dependent pathway for localization of mRNAs encoding membrane-associated proteins.
Collapse
Affiliation(s)
- Lindsay P. Winkenbach
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523, USA
| | - Dylan M. Parker
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523, USA,Department of Chemistry and Biochemistry, Howard Hughes Medical Institute, University of Colorado, Boulder, CO 80303, USA
| | - Robert T. P. Williams
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523, USA
| | - Erin Osborne Nishimura
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523, USA,Author for correspondence ()
| |
Collapse
|
14
|
Huang X, Li B, Hu J, Liu Z, Li D, Chen Z, Huang H, Chen Y, Guo X, Cui Y, Huang Q. Advanced glycation endproducts mediate chronic kidney injury with characteristic patterns in different stages. Front Physiol 2022; 13:977247. [PMID: 36160865 PMCID: PMC9500449 DOI: 10.3389/fphys.2022.977247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 08/12/2022] [Indexed: 11/20/2022] Open
Abstract
Advanced glycation endproducts (AGEs) have been confirmed to play a causative role in the development of diabetic nephropathy (DN). In this study, we revealed that AGE-induced kidney injury with characteristic patterns in different stages and moesin phosphorylation plays a role in these processes. In WT mice treated with AGE-modified bovine serum albumin (AGE-BSA), distinct abnormal angiogenesis in Bowman’s capsule of the kidney emerged early after 1 m under AGE-BSA stimulation, while these neovessels became rare after 6 m. AGE-BSA also induced glomerular hypertrophy and mesangial expansion at 1 m but glomerular atrophy and fibrosis at 6 m. Electron microscopy imaging demonstrated the damage of foot process integrity in podocytes and the uneven thickening of the glomerular basement membrane in the AGE-BSA-treated group, which was more significant after 6 m of AGE-BSA treatment than 1 m. The kidney dysfunction appeared along with these AGE-induced morphological changes. However, these AGE-BSA-induced pathological changes were significantly attenuated in RAGE-knockout mice. Moreover, moesin phosphorylation was accompanied by AGE-BSA-induced alterations and moesin deficiency in mice attenuated by AGE-BSA-induced fibrosis. The investigation on glomerular endothelial cells (GECs) also confirmed that the phosphorylation of moesin T558 is critical in AGE-induced tube formation. Overall, this study suggests that AGEs mediate kidney injury with characteristic patterns by binding with RAGE and inducing moesin phosphorylation.
Collapse
Affiliation(s)
- Xiaoxia Huang
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Bingyu Li
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Jiaqing Hu
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Zhuanhua Liu
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Dongping Li
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Zhenfeng Chen
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Hang Huang
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Yanjia Chen
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaohua Guo
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Yun Cui
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- Shunde Hospital, Southern Medical University (The First People’s Hospital of Shunde), Guangzhou, China
- *Correspondence: Yun Cui, ; Qiaobing Huang,
| | - Qiaobing Huang
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- *Correspondence: Yun Cui, ; Qiaobing Huang,
| |
Collapse
|
15
|
De Belly H, Paluch EK, Chalut KJ. Interplay between mechanics and signalling in regulating cell fate. Nat Rev Mol Cell Biol 2022; 23:465-480. [PMID: 35365816 DOI: 10.1038/s41580-022-00472-z] [Citation(s) in RCA: 109] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/04/2022] [Indexed: 12/11/2022]
Abstract
Mechanical signalling affects multiple biological processes during development and in adult organisms, including cell fate transitions, cell migration, morphogenesis and immune responses. Here, we review recent insights into the mechanisms and functions of two main routes of mechanical signalling: outside-in mechanical signalling, such as mechanosensing of substrate properties or shear stresses; and mechanical signalling regulated by the physical properties of the cell surface itself. We discuss examples of how these two classes of mechanical signalling regulate stem cell function, as well as developmental processes in vivo. We also discuss how cell surface mechanics affects intracellular signalling and, in turn, how intracellular signalling controls cell surface mechanics, generating feedback into the regulation of mechanosensing. The cooperation between mechanosensing, intracellular signalling and cell surface mechanics has a profound impact on biological processes. We discuss here our understanding of how these three elements interact to regulate stem cell fate and development.
Collapse
Affiliation(s)
- Henry De Belly
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, USA
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA, USA
| | - Ewa K Paluch
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK.
| | - Kevin J Chalut
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK.
- Wellcome/MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK.
| |
Collapse
|
16
|
Sepers JJ, Ramalho JJ, Kroll JR, Schmidt R, Boxem M. ERM-1 Phosphorylation and NRFL-1 Redundantly Control Lumen Formation in the C. elegans Intestine. Front Cell Dev Biol 2022; 10:769862. [PMID: 35198555 PMCID: PMC8860247 DOI: 10.3389/fcell.2022.769862] [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] [Received: 09/02/2021] [Accepted: 01/04/2022] [Indexed: 11/13/2022] Open
Abstract
Reorganization of the plasma membrane and underlying actin cytoskeleton into specialized domains is essential for the functioning of most polarized cells in animals. Proteins of the ezrin-radixin-moesin (ERM) and Na+/H+ exchanger 3 regulating factor (NHERF) family are conserved regulators of cortical specialization. ERM proteins function as membrane-actin linkers and as molecular scaffolds that organize the distribution of proteins at the membrane. NHERF proteins are PDZ-domain containing adapters that can bind to ERM proteins and extend their scaffolding capability. Here, we investigate how ERM and NHERF proteins function in regulating intestinal lumen formation in the nematode Caenorhabditis elegans. C. elegans has single ERM and NHERF family proteins, termed ERM-1 and NRFL-1, and ERM-1 was previously shown to be critical for intestinal lumen formation. Using CRISPR/Cas9-generated nrfl-1 alleles we demonstrate that NRFL-1 localizes at the intestinal microvilli, and that this localization is depended on an interaction with ERM-1. However, nrfl-1 loss of function mutants are viable and do not show defects in intestinal development. Interestingly, combining nrfl-1 loss with erm-1 mutants that either block or mimic phosphorylation of a regulatory C-terminal threonine causes severe defects in intestinal lumen formation. These defects are not observed in the phosphorylation mutants alone, and resemble the effects of strong erm-1 loss of function. The loss of NRFL-1 did not affect the localization or activity of ERM-1. Together, these data indicate that ERM-1 and NRFL-1 function together in intestinal lumen formation in C. elegans. We postulate that the functioning of ERM-1 in this tissue involves actin-binding activities that are regulated by the C-terminal threonine residue and the organization of apical domain composition through NRFL-1.
Collapse
Affiliation(s)
- Jorian J Sepers
- Division of Developmental Biology, Department of Biology, Faculty of Science, Institute of Biodynamics and Biocomplexity, Utrecht University, Utrecht, Netherlands
| | - João J Ramalho
- Division of Developmental Biology, Department of Biology, Faculty of Science, Institute of Biodynamics and Biocomplexity, Utrecht University, Utrecht, Netherlands.,Laboratory of Biochemistry, Wageningen University and Research, Wageningen, Netherlands
| | - Jason R Kroll
- Division of Developmental Biology, Department of Biology, Faculty of Science, Institute of Biodynamics and Biocomplexity, Utrecht University, Utrecht, Netherlands
| | - Ruben Schmidt
- Division of Developmental Biology, Department of Biology, Faculty of Science, Institute of Biodynamics and Biocomplexity, Utrecht University, Utrecht, Netherlands
| | - Mike Boxem
- Division of Developmental Biology, Department of Biology, Faculty of Science, Institute of Biodynamics and Biocomplexity, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
17
|
A biophysical perspective of the regulatory mechanisms of ezrin/radixin/moesin proteins. Biophys Rev 2022; 14:199-208. [PMID: 35340609 PMCID: PMC8921360 DOI: 10.1007/s12551-021-00928-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 12/14/2021] [Indexed: 02/08/2023] Open
Abstract
Many signal transductions resulting from ligand-receptor interactions occur at the cell surface. These signaling pathways play essential roles in cell polarization, membrane morphogenesis, and the modulation of membrane tension at the cell surface. However, due to the large number of membrane-binding proteins, including actin-membrane linkers, and transmembrane proteins present at the cell surface, the molecular mechanisms underlying the regulation at the cell surface are yet unclear. Here, we describe the molecular functions of one of the key players at the cell surface, ezrin/radixin/moesin (ERM) proteins from a biophysical point of view. We focus our discussion on biophysical properties of ERM proteins revealed by using biophysical tools in live cells and in vitro reconstitution systems. We first describe the structural properties of ERM proteins and then discuss the interactions of ERM proteins with PI(4,5)P2 and the actin cytoskeleton. These properties of ERM proteins revealed by using biophysical approaches have led to a better understanding of their physiological functions in cells and tissues. Supplementary Information The online version contains supplementary material available at 10.1007/s12551-021-00928-0.
Collapse
|
18
|
Li Y, Chen J, Chen Z, Xu X, Weng J, Zhang Y, Mo Y, Liu Y, Wang J, Ke Y. CircGLIS3 Promotes High-Grade Glioma Invasion via Modulating Ezrin Phosphorylation. Front Cell Dev Biol 2021; 9:663207. [PMID: 34540823 PMCID: PMC8446459 DOI: 10.3389/fcell.2021.663207] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 07/30/2021] [Indexed: 12/12/2022] Open
Abstract
High-grade glioma is highly invasive and malignant, resistant to combined therapies, and easy to relapse. A better understanding of circular RNA (circRNA) biological function in high-grade glioma might contribute to the therapeutic efficacy. Here, a circRNA merely upregulated in high-grade glioma, circGLIS3 (hsa_circ_0002874, originating from exon 2 of GLIS3), was validated by microarray and Real-time quantitative reverse transcription PCR (qRT-PCR). The role of circGLIS3 in glioma was assessed by functional experiments both in vitro and in vivo. Fluorescence in situ hybridization (FISH), RNA pull-down, RNA immunoprecipitation (RIP), and immunohistochemical staining were performed for mechanistic study. Cocultured brain endothelial cells with glioma explored the role of exosome-derived circGLIS3 in the glioma microenvironment. We found that upregulation of circGLIS3 promoted glioma cell migration and invasion and showed aggressive characteristics in tumor-bearing mice. Mechanistically, we found that circGLIS3 could promote the Ezrin T567 phosphorylation level. Moreover, circGLIS3 could be excreted by glioma through exosomes and induced endothelial cell angiogenesis. Our findings indicate that circGLIS3 is upregulated in high-grade glioma and contributes to the invasion and angiogenesis of glioma via modulating Ezrin T567 phosphorylation.
Collapse
Affiliation(s)
- Yan Li
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jiansheng Chen
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China.,Department of Neurosurgery, Huizhou Municipal Central Hospital, Huizhou, China
| | - Zetao Chen
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xiangdong Xu
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jun Weng
- Department of Hepticbile Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yuxuan Zhang
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yunzhao Mo
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yang Liu
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jihui Wang
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yiquan Ke
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
19
|
Gibieža P, Petrikaitė V. The regulation of actin dynamics during cell division and malignancy. Am J Cancer Res 2021; 11:4050-4069. [PMID: 34659876 PMCID: PMC8493394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 07/29/2021] [Indexed: 06/13/2023] Open
Abstract
Actin is the most abundant protein in almost all the eukaryotic cells. Actin amino acid sequences are highly conserved and have not changed a lot during the progress of evolution, varying by no more than 20% in the completely different species, such as humans and algae. The network of actin filaments plays a crucial role in regulating cells' cytoskeleton that needs to undergo dynamic tuning and structural changes in order for various functional processes, such as cell motility, migration, adhesion, polarity establishment, cell growth and cell division, to take place in live cells. Owing to its fundamental role in the cell, actin is a prominent regulator of cell division, a process, whose success directly depends on morphological changes of actin cytoskeleton and correct segregation of duplicated chromosomes. Disorganization of actin framework during the last stage of cell division, known as cytokinesis, can lead to multinucleation and formation of polyploidy in post-mitotic cells, eventually developing into cancer. In this review, we will cover the principles of actin regulation during cell division and discuss how the control of actin dynamics is altered during the state of malignancy.
Collapse
Affiliation(s)
- Paulius Gibieža
- Laboratory of Drug Targets Histopathology, Institute of Cardiology, Lithuanian University of Health Sciences Kaunas, LT-50162, Lithuania
| | - Vilma Petrikaitė
- Laboratory of Drug Targets Histopathology, Institute of Cardiology, Lithuanian University of Health Sciences Kaunas, LT-50162, Lithuania
| |
Collapse
|
20
|
Zhang L, Lu SY, Guo R, Ma JX, Tang LY, Wang JJ, Shen CL, Lu LM, Liu J, Wang ZG, Zhang HX. STK10 knockout inhibits cell migration and promotes cell proliferation via modulating the activity of ERM and p38 MAPK in prostate cancer cells. Exp Ther Med 2021; 22:851. [PMID: 34149897 PMCID: PMC8210223 DOI: 10.3892/etm.2021.10283] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 05/25/2021] [Indexed: 12/24/2022] Open
Abstract
Prostate cancer (PCa) is one of the most common types of cancer and is a serious threat to men's health due to the high rate of incidence and metastasis. However, the exact underlying pathology of this malignant disease has yet to be fully elucidated. The ezrin-radixin-moesin (ERM) family of proteins are associated with the development and metastasis of various types of cancer. Serine threonine kinase 10 (STK10) is an ERM kinase that is involved in the activation of ERM proteins and serves essential roles in the aggregation and adhesion of lymphocytes. To evaluate the functional roles of STK10 in the pathogenesis of PCa, a STK10-knockout (KO) DU145 PCa cell line was generated using the CRISPR-Cas9 gene editing system, and the effects of STK10 deletion on tumor biological behaviors were further analyzed. The present data suggested that STK10 KO promoted PCa cell proliferation by inhibiting p38 MAPK activation and suppressed migration primarily via the inhibition of p38 MAPK signaling and ERM protein activation. To the best of our knowledge, this is the first study to provide evidence that STK10 plays important roles in the proliferation and migration of PCa cells, which will be useful for further investigation into the pathogenesis of this disease.
Collapse
Affiliation(s)
- Lu Zhang
- Research Center for Experimental Medicine, State Key Laboratory of Medical Genomics, Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R. China
| | - Shun-Yuan Lu
- Research Center for Experimental Medicine, State Key Laboratory of Medical Genomics, Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R. China
| | - Rui Guo
- Research Center for Experimental Medicine, State Key Laboratory of Medical Genomics, Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R. China
| | - Jin-Xia Ma
- Research Center for Experimental Medicine, State Key Laboratory of Medical Genomics, Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R. China
| | - Ling-Yun Tang
- Research Center for Experimental Medicine, State Key Laboratory of Medical Genomics, Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R. China
| | - Jin-Jin Wang
- Shanghai Model Organisms Center, Shanghai 201321, P.R. China
| | - Chun-Ling Shen
- Research Center for Experimental Medicine, State Key Laboratory of Medical Genomics, Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R. China
| | - Li-Ming Lu
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R. China
| | - Jie Liu
- Shanghai Institute of Orthopaedics and Traumatology, Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R. China
| | - Zhu-Gang Wang
- Research Center for Experimental Medicine, State Key Laboratory of Medical Genomics, Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R. China
| | - Hong-Xin Zhang
- Research Center for Experimental Medicine, State Key Laboratory of Medical Genomics, Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R. China
| |
Collapse
|
21
|
Dupré L, Boztug K, Pfajfer L. Actin Dynamics at the T Cell Synapse as Revealed by Immune-Related Actinopathies. Front Cell Dev Biol 2021; 9:665519. [PMID: 34249918 PMCID: PMC8266300 DOI: 10.3389/fcell.2021.665519] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 04/06/2021] [Indexed: 01/21/2023] Open
Abstract
The actin cytoskeleton is composed of dynamic filament networks that build adaptable local architectures to sustain nearly all cellular activities in response to a myriad of stimuli. Although the function of numerous players that tune actin remodeling is known, the coordinated molecular orchestration of the actin cytoskeleton to guide cellular decisions is still ill defined. T lymphocytes provide a prototypical example of how a complex program of actin cytoskeleton remodeling sustains the spatio-temporal control of key cellular activities, namely antigen scanning and sensing, as well as polarized delivery of effector molecules, via the immunological synapse. We here review the unique knowledge on actin dynamics at the T lymphocyte synapse gained through the study of primary immunodeficiences caused by mutations in genes encoding actin regulatory proteins. Beyond the specific roles of individual actin remodelers, we further develop the view that these operate in a coordinated manner and are an integral part of multiple signaling pathways in T lymphocytes.
Collapse
Affiliation(s)
- Loïc Dupré
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases (LBI-RUD), Vienna, Austria.,Department of Dermatology, Medical University of Vienna, Vienna, Austria.,Toulouse Institute for Infectious and Inflammatory Diseases (INFINITy), INSERM, CNRS, Toulouse III Paul Sabatier University, Toulouse, France
| | - Kaan Boztug
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases (LBI-RUD), Vienna, Austria.,St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria.,CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria.,Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria.,St. Anna Children's Hospital, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Laurène Pfajfer
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases (LBI-RUD), Vienna, Austria.,Department of Dermatology, Medical University of Vienna, Vienna, Austria.,Toulouse Institute for Infectious and Inflammatory Diseases (INFINITy), INSERM, CNRS, Toulouse III Paul Sabatier University, Toulouse, France.,St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria
| |
Collapse
|
22
|
Aalto A, Olguin-Olguin A, Raz E. Zebrafish Primordial Germ Cell Migration. Front Cell Dev Biol 2021; 9:684460. [PMID: 34249937 PMCID: PMC8260996 DOI: 10.3389/fcell.2021.684460] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 05/17/2021] [Indexed: 01/03/2023] Open
Abstract
Similar to many other organisms, zebrafish primordial germ cells (PGCs) are specified at a location distinct from that of gonadal somatic cells. Guided by chemotactic cues, PGCs migrate through embryonic tissues toward the region where the gonad develops. In this process, PGCs employ a bleb-driven amoeboid migration mode, characterized by low adhesion and high actomyosin contractility, a strategy used by other migrating cells, such as leukocytes and certain types of cancer cells. The mechanisms underlying the motility and the directed migration of PGCs should be robust to ensure arrival at the target, thereby contributing to the fertility of the organism. These features make PGCs an excellent model for studying guided single-cell migration in vivo. In this review, we present recent findings regarding the establishment and maintenance of cell polarity that are essential for motility and discuss the mechanisms by which cell polarization and directed migration are controlled by chemical and physical cues.
Collapse
Affiliation(s)
- Anne Aalto
- Institute of Cell Biology, Center for Molecular Biology of Inflammation, University of Münster, Münster, Germany
| | - Adan Olguin-Olguin
- Institute of Cell Biology, Center for Molecular Biology of Inflammation, University of Münster, Münster, Germany
| | - Erez Raz
- Institute of Cell Biology, Center for Molecular Biology of Inflammation, University of Münster, Münster, Germany
| |
Collapse
|
23
|
Makiishi S, Furuichi K, Yamamura Y, Sako K, Shinozaki Y, Toyama T, Kitajima S, Iwata Y, Sakai N, Shimizu M, Hirose-Sugiura T, Kaneko S, Kato Y, Wada T. Carnitine/organic cation transporter 1 precipitates the progression of interstitial fibrosis through oxidative stress in diabetic nephropathy in mice. Sci Rep 2021; 11:9093. [PMID: 33907247 PMCID: PMC8079701 DOI: 10.1038/s41598-021-88724-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Accepted: 04/07/2021] [Indexed: 11/08/2022] Open
Abstract
Carnitine/organic cation transporter 1 (OCTN1) is the only known uptake transporter for ergothioneine which is a food-derived strong antioxidant amino acid that is absorbed by OCTN1. We previously reported the roles of OCTN1/ergothioneine in the progression of kidney fibrosis in ischemic kidney disease. In this study, we evaluated the roles of OCTN1 in the progression of diabetic kidney disease. A diabetic kidney disease model was induced in octn1 knockout and wild-type mice by streptozotocin (STZ). Oxidative stress, represented by urinary 8-hydroxy-2'-deoxyguanosine (8-OHdG), were higher in the octn1 knockout mice. Azan- and Sirius red-positive areas increased significantly in the octn1 knockout mice. Gene expression was evaluated by cluster analysis, and shown to be different in the octn1 knockout mice compared with the wild-type mice. In a pathway analysis, the pathway associated with the cytoskeleton and cell adhesion increased. In accordance with interstitial fibrosis in octn1 knockout mice, gene expression of moesin in the injured kidney, known as an associated protein of cytoskeleton and cell membranes, was doubled 28 weeks after STZ injection. In addition, the moesin protein was expressed in a part of α-SMA-positive renal tubular epithelial cells. These findings were confirmed by cultured murine proximal tubular epithelial cells: The expression of moesin was induced under oxidative stress with hydrogen peroxide. These data indicate that OCTN1 would play some roles in progression of interstitial fibrosis under oxidative stress via moesin expression in diabetic kidney disease.
Collapse
Affiliation(s)
- Shohei Makiishi
- Department of Nephrology and Laboratory Medicine, Faculty of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Kengo Furuichi
- Department of Nephrology, Kanazawa Medical University School of Medicine, Uchinada, Ishikawa, Japan.
| | - Yuta Yamamura
- Department of Nephrology and Laboratory Medicine, Faculty of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Keisuke Sako
- Department of Nephrology and Laboratory Medicine, Faculty of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Yasuyuki Shinozaki
- Department of Nephrology and Laboratory Medicine, Faculty of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Tadashi Toyama
- Department of Nephrology and Laboratory Medicine, Faculty of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Shinji Kitajima
- Department of Nephrology and Laboratory Medicine, Faculty of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Yasunori Iwata
- Department of Nephrology and Laboratory Medicine, Faculty of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Norihiko Sakai
- Department of Nephrology and Laboratory Medicine, Faculty of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Miho Shimizu
- Department of Nephrology and Laboratory Medicine, Faculty of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Tomoko Hirose-Sugiura
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Science, Kanazawa University, Kanazawa, Japan
| | - Shuichi Kaneko
- Department of System Biology, Kanazawa University, Kanazawa, Japan
| | - Yukio Kato
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Science, Kanazawa University, Kanazawa, Japan
| | - Takashi Wada
- Department of Nephrology and Laboratory Medicine, Faculty of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| |
Collapse
|
24
|
Termini CM, Pang A, Batton DM, Chute JP. Proteoglycans regulate protein tyrosine phosphatase receptor σ organization on hematopoietic stem/progenitor cells. Exp Hematol 2021; 96:44-51. [PMID: 33515635 PMCID: PMC10838547 DOI: 10.1016/j.exphem.2021.01.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 01/07/2021] [Accepted: 01/22/2021] [Indexed: 01/08/2023]
Abstract
Protein tyrosine phosphatase receptor σ (PTPσ) is highly expressed by murine and human hematopoietic stem cells (HSCs) and negatively regulates HSC self-renewal and regeneration. Previous studies of the nervous system suggest that heparan sulfate proteoglycans can inactivate PTPσ by clustering PTPσ receptors on neurons, but this finding has yet to be visually verified with adequate resolution. Here, we sought to visualize and quantify how heparan sulfate proteoglycans regulate the organization and activation of PTPσ in hematopoietic stem/progenitor cells (HSPCs). Our study illustrates that syndecan-2 promotes PTPσ clustering, which sustains phospho-tyrosine and phospho-ezrin levels in association with augmentation of hematopoietic colony formation. Strategies that promote clustering of PTPσ on HSPCs may serve to powerfully augment hematopoietic function.
Collapse
Affiliation(s)
- Christina M Termini
- Division of Hematology/Oncology, Department of Medicine, University of California at Los Angeles, Los Angeles, CA; Department of Orthopaedic Surgery, University of California at Los Angeles, Los Angeles, CA; Department of Medicine, Cedars Sinai Medical Center, Los Angeles, CA
| | - Amara Pang
- Division of Hematology/Oncology, Department of Medicine, University of California at Los Angeles, Los Angeles, CA; Department of Medicine, Cedars Sinai Medical Center, Los Angeles, CA
| | - Destiny M Batton
- Division of Hematology/Oncology, Department of Medicine, University of California at Los Angeles, Los Angeles, CA
| | - John P Chute
- Division of Hematology/Oncology, Department of Medicine, University of California at Los Angeles, Los Angeles, CA; Department of Orthopaedic Surgery, University of California at Los Angeles, Los Angeles, CA; Department of Medicine, Cedars Sinai Medical Center, Los Angeles, CA; Broad Stem Cell Research Center, University of California at Los Angeles, Los Angeles, CA; Jonsson Comprehensive Cancer Center, University of California at Los Angeles, Los Angeles, CA.
| |
Collapse
|
25
|
Wang S, Lin CW, Carleton AE, Cortez CL, Johnson C, Taniguchi LE, Sekulovski N, Townshend RF, Basrur V, Nesvizhskii AI, Zou P, Fu J, Gumucio DL, Duncan MC, Taniguchi K. Spatially resolved cell polarity proteomics of a human epiblast model. SCIENCE ADVANCES 2021; 7:7/17/eabd8407. [PMID: 33893097 PMCID: PMC8064645 DOI: 10.1126/sciadv.abd8407] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 03/05/2021] [Indexed: 05/08/2023]
Abstract
Critical early steps in human embryonic development include polarization of the inner cell mass, followed by formation of an expanded lumen that will become the epiblast cavity. Recently described three-dimensional (3D) human pluripotent stem cell-derived cyst (hPSC-cyst) structures can replicate these processes. To gain mechanistic insights into the poorly understood machinery involved in epiblast cavity formation, we interrogated the proteomes of apical and basolateral membrane territories in 3D human hPSC-cysts. APEX2-based proximity bioinylation, followed by quantitative mass spectrometry, revealed a variety of proteins without previous annotation to specific membrane subdomains. Functional experiments validated the requirement for several apically enriched proteins in cyst morphogenesis. In particular, we found a key role for the AP-1 clathrin adaptor complex in expanding the apical membrane domains during lumen establishment. These findings highlight the robust power of this proximity labeling approach for discovering novel regulators of epithelial morphogenesis in 3D stem cell-based models.
Collapse
Affiliation(s)
- Sicong Wang
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Chien-Wei Lin
- Division of Biostatistics, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Amber E Carleton
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Chari L Cortez
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Craig Johnson
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Linnea E Taniguchi
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Nikola Sekulovski
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Ryan F Townshend
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Venkatesha Basrur
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Alexey I Nesvizhskii
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Peng Zou
- College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Jianping Fu
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA.
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Deborah L Gumucio
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Mara C Duncan
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| | - Kenichiro Taniguchi
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| |
Collapse
|
26
|
Ghosh S, Di Bartolo V, Tubul L, Shimoni E, Kartvelishvily E, Dadosh T, Feigelson SW, Alon R, Alcover A, Haran G. ERM-Dependent Assembly of T Cell Receptor Signaling and Co-stimulatory Molecules on Microvilli prior to Activation. Cell Rep 2021; 30:3434-3447.e6. [PMID: 32160548 DOI: 10.1016/j.celrep.2020.02.069] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 12/16/2019] [Accepted: 02/18/2020] [Indexed: 01/25/2023] Open
Abstract
T cell surfaces are covered with microvilli, actin-rich and flexible protrusions. We use super-resolution microscopy to show that ≥90% of T cell receptor (TCR) complex molecules TCRαβ and TCRζ, as well as the co-receptor CD4 (cluster of differentiation 4) and the co-stimulatory molecule CD2, reside on microvilli of resting human T cells. Furthermore, TCR proximal signaling molecules involved in the initial stages of the immune response, including the protein tyrosine kinase Lck (lymphocyte-specific protein tyrosine kinase) and the key adaptor LAT (linker for activation of T cells), are also enriched on microvilli. Notably, phosphorylated proteins of the ERM (ezrin, radixin, and moesin) family colocalize with TCRαβ as well as with actin filaments, implying a role for one or more ERMs in linking the TCR complex to the actin cytoskeleton within microvilli. Our results establish microvilli as key signaling hubs, in which the TCR complex and its proximal signaling molecules and adaptors are preassembled prior to activation in an ERM-dependent manner, facilitating initial antigen sensing.
Collapse
Affiliation(s)
- Shirsendu Ghosh
- Department of Chemical and Biological Physics, Weizmann Institute of Science, Rehovot 7610001, Israel.
| | - Vincenzo Di Bartolo
- Lymphocyte Cell Biology Unit, INSERM U1221, Department of Immunology, Institut Pasteur, Paris 75015, France
| | - Liron Tubul
- Department of Chemical and Biological Physics, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Eyal Shimoni
- Chemical Research Support, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Elena Kartvelishvily
- Chemical Research Support, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Tali Dadosh
- Chemical Research Support, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Sara W Feigelson
- Department of Immunology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Ronen Alon
- Department of Immunology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Andres Alcover
- Lymphocyte Cell Biology Unit, INSERM U1221, Department of Immunology, Institut Pasteur, Paris 75015, France
| | - Gilad Haran
- Department of Chemical and Biological Physics, Weizmann Institute of Science, Rehovot 7610001, Israel.
| |
Collapse
|
27
|
Ferri G, Edreira MM. All Roads Lead to Cytosol: Trypanosoma cruzi Multi-Strategic Approach to Invasion. Front Cell Infect Microbiol 2021; 11:634793. [PMID: 33747982 PMCID: PMC7973469 DOI: 10.3389/fcimb.2021.634793] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Accepted: 01/27/2021] [Indexed: 12/17/2022] Open
Abstract
T. cruzi has a complex life cycle involving four developmental stages namely, epimastigotes, metacyclic trypomastigotes, amastigotes and bloodstream trypomastigotes. Although trypomastigotes are the infective forms, extracellular amastigotes have also shown the ability to invade host cells. Both stages can invade a broad spectrum of host tissues, in fact, almost any nucleated cell can be the target of infection. To add complexity, the parasite presents high genetic variability with differential characteristics such as infectivity. In this review, we address the several strategies T. cruzi has developed to subvert the host cell signaling machinery in order to gain access to the host cell cytoplasm. Special attention is made to the numerous parasite/host protein interactions and to the set of signaling cascades activated during the formation of a parasite-containing vesicle, the parasitophorous vacuole, from which the parasite escapes to the cytosol, where differentiation and replication take place.
Collapse
Affiliation(s)
- Gabriel Ferri
- CONICET-Universidad de Buenos Aires, IQUIBICEN, Ciudad de Buenos Aires, Argentina
| | - Martin M Edreira
- CONICET-Universidad de Buenos Aires, IQUIBICEN, Ciudad de Buenos Aires, Argentina.,Laboratorio de Biología Molecular de Trypanosoma, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos, Ciudad de Buenos Aires, Argentina.,Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
28
|
Wang S, Wang T, Liu T, Xie RG, Zhao XH, Wang L, Yang Q, Jia LT, Han J. Ermin is a p116 RIP -interacting protein promoting oligodendroglial differentiation and myelin maintenance. Glia 2020; 68:2264-2276. [PMID: 32530539 DOI: 10.1002/glia.23838] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 04/17/2020] [Accepted: 04/21/2020] [Indexed: 12/21/2022]
Abstract
Myelin sheaths, which insulate the axons and ensure saltatory conduction of the nerve impulse, are generated and maintained via largely uncharacterized mechanisms. Ermin is an oligodendrocyte-specific protein associated with the cytoskeleton, but how it regulates cytoskeletal remodeling during oligodendrocyte differentiation and its role in myelin maintenance are not clear. To address this, we generated mice constitutively deficient for Ermn, the Ermin-coding gene. We found that aged Ermn-knockout mice exhibit an aberrant myelin architecture, with splitting of myelin layers, peeling of the myelin sheath from axons, and breakdown of myelinated fibers. As a result, these mice had remarkably impaired motor coordination. Ermn knockout also accelerated cuprizone-induced demyelination and exacerbated the associated movement disorders. Ermin was found to contribute to oligodendrocyte morphogenesis by associating with the myosin phosphatase Rho interacting protein (Mprip/p116RIP ) and inactivating RhoA, a GTPase that controls cytoskeletal rearrangement in differentiating cells. These findings provide novel insights into the mechanisms regulating oligodendroglial differentiation, the maintenance of the myelin sheaths, and remyelination.
Collapse
Affiliation(s)
- Shan Wang
- Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, China
| | - Tao Wang
- Department of Neurology, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Tao Liu
- Key Laboratory of Modern Teaching Technology, Shaanxi Normal University, Xi'an, China
| | - Rou-Gang Xie
- Department of Neurobiology and Collaborative Innovation Center for Brain Science, Fourth Military Medical University, Xi'an, China
| | - Xiang-Hui Zhao
- Department of Neurobiology and Collaborative Innovation Center for Brain Science, Fourth Military Medical University, Xi'an, China
| | - Lei Wang
- Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, China
| | - Qian Yang
- Department of Neurology, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Lin-Tao Jia
- Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, China
| | - Jing Han
- Key Laboratory of Modern Teaching Technology, Shaanxi Normal University, Xi'an, China
| |
Collapse
|
29
|
Meng F, Su Y, Xu B. Rho-associated protein kinase-dependent moesin phosphorylation is required for PD-L1 stabilization in breast cancer. Mol Oncol 2020; 14:2701-2712. [PMID: 32941674 PMCID: PMC7607174 DOI: 10.1002/1878-0261.12804] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 07/06/2020] [Accepted: 09/11/2020] [Indexed: 01/12/2023] Open
Abstract
Expression of programmed cell death ligand (PD-L1) is associated with poor prognosis in breast cancer. Understanding the regulation of PD-L1 expression in breast cancer could provide a new strategy for breast cancer treatment. Here, we demonstrate that moesin (MSN) phosphorylation by Rho-associated protein kinase (ROCK) stabilizes PD-L1 protein levels. Our results indicate that phosphorylated MSN may compete with the E3 ubiquitin ligase SPOP for binding PD-L1. ROCK inhibition via the Y-27632 inhibitor or MSN silencing decreased PD-L1 expression, resulting in T-cell activation both in vitro and in vivo. Administration of Y-27632 into immunocompetent Balb/c mice bearing breast tumors suppressed tumor progression and enhanced CD4+ and CD8+ T-cell infiltration. RNA-seq analysis of Y-27632-treated mouse tumors revealed that ROCK inhibition upregulated several immune response genes. However, the combination of Y-27632 and an anti-PD-1 antibody did not show additive or synergistic effects due to reduced PD-L1 in the presence of Y-27632. Our study unravels a previously unappreciated mechanism of PD-L1 regulation through the ROCK-MSN pathway. Moreover, we found that ROCK inhibitors could be combined with breast cancer immunotherapy.
Collapse
Affiliation(s)
- Fanbiao Meng
- Department of Biochemistry and Molecular Biology, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Breast Cancer Prevention and Therapy, Ministry of Education, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Yang Su
- Department of Biochemistry and Molecular Biology, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Breast Cancer Prevention and Therapy, Ministry of Education, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Bo Xu
- Department of Biochemistry and Molecular Biology, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Breast Cancer Prevention and Therapy, Ministry of Education, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Center for Intelligent Oncology, Chongqing University Cancer Hospital, Chongqing University School of Medicine, Chongqing, China
| |
Collapse
|
30
|
Hubrich H, Mey IP, Brückner BR, Mühlenbrock P, Nehls S, Grabenhorst L, Oswald T, Steinem C, Janshoff A. Viscoelasticity of Native and Artificial Actin Cortices Assessed by Nanoindentation Experiments. NANO LETTERS 2020; 20:6329-6335. [PMID: 32786944 DOI: 10.1021/acs.nanolett.0c01769] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Cell cortices are responsible for the resilience and morphological dynamics of cells. Measuring their mechanical properties is impeded by contributions from other filament types, organelles, and the crowded cytoplasm. We established a versatile concept for the precise assessment of cortical viscoelasticity based on force cycle experiments paired with continuum mechanics. Apical cell membranes of confluent MDCK II cells were deposited on porous substrates and locally deformed. Force cycles could be described with a time-dependent area compressibility modulus obeying the same power law as employed for whole cells. The reduced fluidity of apical cell membranes compared to living cells could partially be restored by reactivating myosin motors. A comparison with artificial minimal actin cortices (MACs) reveals lower stiffness and higher fluidity attributed to missing cross-links in MACs.
Collapse
Affiliation(s)
- Hanna Hubrich
- Department of Chemistry, Institute of Physical Chemistry, Göttingen 37077, Germany
| | - Ingo P Mey
- Department of Chemistry, Institute of Organic and Biomolecular Chemistry, Göttingen 37077, Germany
| | - Bastian R Brückner
- Department of Chemistry, Institute of Physical Chemistry, Göttingen 37077, Germany
| | - Peter Mühlenbrock
- Department of Chemistry, Institute of Organic and Biomolecular Chemistry, Göttingen 37077, Germany
| | - Stefan Nehls
- Department of Chemistry, Institute of Physical Chemistry, Göttingen 37077, Germany
| | - Lennart Grabenhorst
- Department of Chemistry, Institute of Physical Chemistry, Göttingen 37077, Germany
| | - Tabea Oswald
- Department of Chemistry, Institute of Organic and Biomolecular Chemistry, Göttingen 37077, Germany
| | - Claudia Steinem
- Department of Chemistry, Institute of Organic and Biomolecular Chemistry, Göttingen 37077, Germany
| | - Andreas Janshoff
- Department of Chemistry, Institute of Physical Chemistry, Göttingen 37077, Germany
| |
Collapse
|
31
|
Ramalho JJ, Sepers JJ, Nicolle O, Schmidt R, Cravo J, Michaux G, Boxem M. C-terminal phosphorylation modulates ERM-1 localization and dynamics to control cortical actin organization and support lumen formation during Caenorhabditiselegans development. Development 2020; 147:dev188011. [PMID: 32586975 PMCID: PMC10755404 DOI: 10.1242/dev.188011] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 06/13/2020] [Indexed: 12/31/2023]
Abstract
ERM proteins are conserved regulators of cortical membrane specialization that function as membrane-actin linkers and molecular hubs. The activity of ERM proteins requires a conformational switch from an inactive cytoplasmic form into an active membrane- and actin-bound form, which is thought to be mediated by sequential PIP2 binding and phosphorylation of a conserved C-terminal threonine residue. Here, we use the single Caenorhabditiselegans ERM ortholog, ERM-1, to study the contribution of these regulatory events to ERM activity and tissue formation in vivo Using CRISPR/Cas9-generated erm-1 mutant alleles, we demonstrate that a PIP2-binding site is crucially required for ERM-1 function. By contrast, dynamic regulation of C-terminal T544 phosphorylation is not essential but modulates ERM-1 apical localization and dynamics in a tissue-specific manner, to control cortical actin organization and support lumen formation in epithelial tubes. Our work highlights the dynamic nature of ERM protein regulation during tissue morphogenesis and the importance of C-terminal phosphorylation in fine-tuning ERM activity in a tissue-specific context.
Collapse
Affiliation(s)
- João J Ramalho
- Division of Developmental Biology, Institute of Biodynamics and Biocomplexity, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH, Utrecht, The Netherlands
| | - Jorian J Sepers
- Division of Developmental Biology, Institute of Biodynamics and Biocomplexity, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH, Utrecht, The Netherlands
| | - Ophélie Nicolle
- Univ Rennes, CNRS, IGDR (Institut de Génétique et de Développement de Rennes), UMR 6290, F-35000 Rennes, France
| | - Ruben Schmidt
- Division of Developmental Biology, Institute of Biodynamics and Biocomplexity, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH, Utrecht, The Netherlands
| | - Janine Cravo
- Division of Developmental Biology, Institute of Biodynamics and Biocomplexity, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH, Utrecht, The Netherlands
| | - Grégoire Michaux
- Univ Rennes, CNRS, IGDR (Institut de Génétique et de Développement de Rennes), UMR 6290, F-35000 Rennes, France
| | - Mike Boxem
- Division of Developmental Biology, Institute of Biodynamics and Biocomplexity, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH, Utrecht, The Netherlands
| |
Collapse
|
32
|
Svitkina TM. Actin Cell Cortex: Structure and Molecular Organization. Trends Cell Biol 2020; 30:556-565. [PMID: 32278656 PMCID: PMC7566779 DOI: 10.1016/j.tcb.2020.03.005] [Citation(s) in RCA: 110] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 03/11/2020] [Accepted: 03/19/2020] [Indexed: 12/20/2022]
Abstract
The actin cytoskeleton consists of structurally and biochemically different actin filament arrays. Among them, the actin cortex is thought to have key roles in cell mechanics, but remains a poorly characterized part of the actin cytoskeleton. The cell cortex is typically defined as a thin layer of actin meshwork that uniformly underlies the plasma membrane of the entire cell. However, this definition applies only to specific cases. In general, the cortex structure and subcellular distribution vary significantly across cell types and physiological states of the cell. In this review, I focus on our current knowledge of the structure and molecular composition of the cell cortex.
Collapse
Affiliation(s)
- Tatyana M Svitkina
- Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
33
|
Medvedev A, Kopylov A, Buneeva O, Kurbatov L, Tikhonova O, Ivanov A, Zgoda V. A Neuroprotective Dose of Isatin Causes Multilevel Changes Involving the Brain Proteome: Prospects for Further Research. Int J Mol Sci 2020; 21:ijms21114187. [PMID: 32545384 PMCID: PMC7313464 DOI: 10.3390/ijms21114187] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 06/08/2020] [Accepted: 06/09/2020] [Indexed: 12/18/2022] Open
Abstract
Isatin (indole-2,3-dione) is an endogenous regulator, exhibiting a wide range of biological and pharmacological activities. At doses of 100 mg/kg and above, isatin is neuroprotective in different experimental models of neurodegeneration. Good evidence exists that its effects are realized via interaction with numerous isatin-binding proteins identified in the brain and peripheral tissues studied. In this study, we investigated the effect of a single dose administration of isatin to mice (100 mg/kg, 24 h) on differentially expressed proteins and a profile of the isatin-binding proteins in brain hemispheres. Isatin administration to mice caused downregulation of 31 proteins. However, these changes cannot be attributed to altered expression of corresponding genes. Although at this time point isatin influenced the expression of more than 850 genes in brain hemispheres (including 433 upregulated and 418 downregulated genes), none of them could account for the changes in the differentially expressed proteins. Comparative proteomic analysis of brain isatin-binding proteins of control and isatin-treated mice revealed representative groups of proteins sensitive to isatin administration. Control-specific proteins (n = 55) represent specific targets that interact directly with isatin. Appearance of brain isatin-binding proteins specific to isatin-treated mice (n = 94) may be attributed to the formation of new clusters of protein–protein interactions and/or novel binding sites induced by a high concentration of this regulator (ligand-induced binding sites). Thus, isatin administration produces multiple effects in the brain, which include changes in gene expression and also profiles of isatin-binding proteins and their interactomes. Further studies are needed for deeper insight into the mechanisms of the multilevel changes in the brain proteome induced by isatin. In the context of the neuroprotective action, these changes may be aimed at interruption of pathological links that begin to form after initiation of pathological processes.
Collapse
|
34
|
Zhang Q, Wang G, Xie Y, Gao Z, Liang Z, Pan Z, Wang G, Feng W. Mechanical Changes and Microfilament Reorganization Involved in Microcystin-LR-Promoted Cell Invasion in DU145 and WPMY Cells. Front Pharmacol 2020; 11:89. [PMID: 32174829 PMCID: PMC7054891 DOI: 10.3389/fphar.2020.00089] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 01/27/2020] [Indexed: 12/16/2022] Open
Abstract
Microcystin-leucine arginine (MC-LR) is a potent tumor initiator that can induce malignant cell transformation. Cellular mechanical characteristics are pivotal parameters that are closely related to cell invasion. The aim of this study is to determine the effect of MC-LR on mechanical parameters, microfilament, and cell invasion in DU145 and WPMY cells. Firstly, 10 μM MC-LR was selected as the appropriate concentration via cell viability assay. Subsequently, after MC-LR treatment, the cellular deformability and viscoelastic parameters were tested using the micropipette aspiration technique. The results showed that MC-LR increased the cellular deformability, reduced the cellular viscoelastic parameter values, and caused the cells to become softer. Furthermore, microfilament and microfilament-associated proteins were examined by immunofluorescence and Western blot, respectively. Our results showed that MC-LR induced microfilament reorganization and increased the expression of p-VASP and p-ezrin. Finally, the impact of MC-LR on cell invasion was evaluated. The results revealed that MC-LR promoted cell invasion. Taken together, our results suggested that mechanical changes and microfilament reorganization were involved in MC-LR-promoted cell invasion in DU145 and WPMY cells. Our data provide novel information to explain the toxicological mechanism of MC-LR.
Collapse
Affiliation(s)
- Qiang Zhang
- College of Bioscience and Technology, Weifang Medical University, Weifang, China
| | - Guihua Wang
- Department of Fundamental Veterinary, College of Veterinary Medicine, Shandong Agricultural University, Tai'an, China
| | - Yongfang Xie
- College of Bioscience and Technology, Weifang Medical University, Weifang, China
| | - Zhiqin Gao
- College of Bioscience and Technology, Weifang Medical University, Weifang, China
| | - Zumu Liang
- College of Bioscience and Technology, Weifang Medical University, Weifang, China
| | - Zhifang Pan
- College of Bioscience and Technology, Weifang Medical University, Weifang, China
| | - Guohui Wang
- College of Bioscience and Technology, Weifang Medical University, Weifang, China
| | - Weiguo Feng
- College of Bioscience and Technology, Weifang Medical University, Weifang, China
| |
Collapse
|
35
|
Suárez H, López-Martín S, Toribio V, Zamai M, Hernández-Riquer MV, Genís L, Arroyo AG, Yáñez-Mó M. Regulation of MT1-MMP Activity through Its Association with ERMs. Cells 2020; 9:cells9020348. [PMID: 32028690 PMCID: PMC7072721 DOI: 10.3390/cells9020348] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 01/31/2020] [Accepted: 02/01/2020] [Indexed: 02/08/2023] Open
Abstract
Membrane-bound proteases play a key role in biology by degrading matrix proteins or shedding adhesion receptors. MT1-MMP metalloproteinase is critical during cancer invasion, angiogenesis, and development. MT1-MMP activity is strictly regulated by internalization, recycling, autoprocessing but also through its incorporation into tetraspanin-enriched microdomains (TEMs), into invadopodia, or by its secretion on extracellular vesicles (EVs). We identified a juxtamembrane positively charged cluster responsible for the interaction of MT1-MMP with ERM (ezrin/radixin/moesin) cytoskeletal connectors in breast carcinoma cells. Linkage to ERMs regulates MT1-MMP subcellular distribution and internalization, but not its incorporation into extracellular vesicles. MT1-MMP association to ERMs and insertion into TEMs are independent phenomena, so that mutation of the ERM-binding motif in the cytoplasmic region of MT1-MMP does not preclude its association with the tetraspanin CD151, but impairs the accumulation and coalescence of CD151/MT1-MMP complexes at actin-rich structures. Conversely, gene deletion of CD151 does not impact on MT1-MMP colocalization with ERM molecules. At the plasma membrane MT1-MMP autoprocessing is severely dependent on ERM association and seems to be the dominant regulator of the enzyme collagenolytic activity. This newly characterized MT1-MMP/ERM association can thus be of relevance for tumor cell invasion.
Collapse
Affiliation(s)
- Henar Suárez
- Molecular Biology Department, Universidad Autónoma de Madrid (UAM), 28049 Madrid, Spain; (H.S.); (V.T.)
- Severo Ochoa Molecular Biology Center (CBM-SO), Instituto de Investigación Sanitaria Princesa (IIS-IP), 28049 Madrid, Spain;
| | - Soraya López-Martín
- Severo Ochoa Molecular Biology Center (CBM-SO), Instituto de Investigación Sanitaria Princesa (IIS-IP), 28049 Madrid, Spain;
| | - Víctor Toribio
- Molecular Biology Department, Universidad Autónoma de Madrid (UAM), 28049 Madrid, Spain; (H.S.); (V.T.)
- Severo Ochoa Molecular Biology Center (CBM-SO), Instituto de Investigación Sanitaria Princesa (IIS-IP), 28049 Madrid, Spain;
| | - Moreno Zamai
- Unit of Microscopy and Dynamic Imaging, Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain;
| | - M. Victoria Hernández-Riquer
- Vascular Pathophysiology Department, Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain; (M.V.H.-R.); (L.G.); (A.G.A.)
| | - Laura Genís
- Vascular Pathophysiology Department, Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain; (M.V.H.-R.); (L.G.); (A.G.A.)
| | - Alicia G. Arroyo
- Vascular Pathophysiology Department, Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain; (M.V.H.-R.); (L.G.); (A.G.A.)
- Molecular Biomedicine Department, Centro de Investigaciones Biológicas Margarita Salas (CIB-CSIC), 28040 Madrid, Spain
| | - María Yáñez-Mó
- Molecular Biology Department, Universidad Autónoma de Madrid (UAM), 28049 Madrid, Spain; (H.S.); (V.T.)
- Severo Ochoa Molecular Biology Center (CBM-SO), Instituto de Investigación Sanitaria Princesa (IIS-IP), 28049 Madrid, Spain;
- Correspondence:
| |
Collapse
|
36
|
Chen L, Cui Y, Li B, Weng J, Wang W, Zhang S, Huang X, Guo X, Huang Q. Advanced glycation end products induce immature angiogenesis in in vivo and ex vivo mouse models. Am J Physiol Heart Circ Physiol 2020; 318:H519-H533. [PMID: 31922896 DOI: 10.1152/ajpheart.00473.2019] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Proliferative diabetic retinopathy (PDR) is a progressive disease predominantly involving pathological angiogenesis and is characterized by the development of immature, fragile, and easily hemorrhagic new vessels. Advanced glycation end products (AGEs) and the receptor for AGEs (RAGE) play important roles in the progression of diabetic retinopathy. Our previous studies demonstrated that AGEs promoted HUVEC angiogenesis by inducing moesin phosphorylation via RhoA/Rho-associated protein kinase (ROCK) pathway. The aim of this study was to further confirm AGE-induced angiogenesis in vivo and the involvement of RAGE, ROCK, and moesin phosphorylation in this process. We performed the study in an AGE-treated mouse model with various angiogenesis assays in multiple in vivo and ex vivo models. The results demonstrated that AGEs promoted significant neovascularization in whole mount retina and mouse aortic ring of adult and postnatal mice and in Matrigel plug as well, which were consistently accompanied by increased moesin phosphorylation. The increase of AGE-evoked neovascularization and moesin phosphorylation were both attenuated by RAGE knockout or ROCK inhibitor Y27632 administration in mice. We also revealed the pathological characteristics of AGE-promoted angiogenesis by demonstrating the decrease of pericyte coverage and the disarranged endothelial alignment in microvessels. In conclusion, this study provides in vivo evidences that AGEs induce immature angiogenesis by binding to RAGE, activating the RhoA/ROCK signal pathway and inducing moesin phosphorylation.NEW & NOTEWORTHY Advanced glycation end product (AGE)-induced formation of neovessels and phosphorylation of moesin in retina and aortic ring required AGE receptors. AGEs increased neovessels and the phosphorylation of moesin in retina and aortic ring via RhoA/ROCK pathway. AGE-induced immature angiogenesis in AGE-treated mouse retina and aortic ring. The AGE-RAGE axis and moesin could be candidate targets for overcoming relative diseases.
Collapse
Affiliation(s)
- Lixian Chen
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Shock and Microcirculation, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Yun Cui
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Shock and Microcirculation, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Bingyu Li
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Shock and Microcirculation, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Jie Weng
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Shock and Microcirculation, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Weiju Wang
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Shock and Microcirculation, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Shuangshuang Zhang
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Shock and Microcirculation, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Xuliang Huang
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Shock and Microcirculation, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Xiaohua Guo
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Shock and Microcirculation, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Qiaobing Huang
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Shock and Microcirculation, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| |
Collapse
|
37
|
Shabardina V, Kashima Y, Suzuki Y, Makalowski W. Emergence and Evolution of ERM Proteins and Merlin in Metazoans. Genome Biol Evol 2020; 12:3710-3724. [PMID: 31851361 PMCID: PMC6978628 DOI: 10.1093/gbe/evz265] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/02/2019] [Indexed: 12/18/2022] Open
Abstract
Ezrin, radixin, moesin, and merlin are cytoskeletal proteins, whose functions are specific to metazoans. They participate in cell cortex rearrangement, including cell-cell contact formation, and play an important role in cancer progression. Here, we have performed a comprehensive phylogenetic analysis of the proteins spanning 87 species. The results describe a possible mechanism for the protein family origin in the root of Metazoa, paralogs diversification in vertebrates, and acquisition of novel functions, including tumor suppression. In addition, a merlin paralog, present in most vertebrates but lost in mammals, has been described here for the first time. We have also highlighted a set of amino acid variations within the conserved motifs as the candidates for determining physiological differences between ERM paralogs.
Collapse
Affiliation(s)
| | - Yukie Kashima
- Department of Computational Biology and Medical Sciences, The University of Tokyo, Kashiwa, Japan
| | - Yutaka Suzuki
- Laboratory of Systems Genomics, Department of Computational Biology and Medical Sciences, The University of Tokyo, Kashiwa, Japan
| | | |
Collapse
|
38
|
Lattner J, Leng W, Knust E, Brankatschk M, Flores-Benitez D. Crumbs organizes the transport machinery by regulating apical levels of PI(4,5)P 2 in Drosophila. eLife 2019; 8:e50900. [PMID: 31697234 PMCID: PMC6881148 DOI: 10.7554/elife.50900] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 10/25/2019] [Indexed: 12/12/2022] Open
Abstract
An efficient vectorial intracellular transport machinery depends on a well-established apico-basal polarity and is a prerequisite for the function of secretory epithelia. Despite extensive knowledge on individual trafficking pathways, little is known about the mechanisms coordinating their temporal and spatial regulation. Here, we report that the polarity protein Crumbs is essential for apical plasma membrane phospholipid-homeostasis and efficient apical secretion. Through recruiting βHeavy-Spectrin and MyosinV to the apical membrane, Crumbs maintains the Rab6-, Rab11- and Rab30-dependent trafficking and regulates the lipid phosphatases Pten and Ocrl. Crumbs knock-down results in increased apical levels of PI(4,5)P2 and formation of a novel, Moesin- and PI(4,5)P2-enriched apical membrane sac containing microvilli-like structures. Our results identify Crumbs as an essential hub required to maintain the organization of the apical membrane and the physiological activity of the larval salivary gland.
Collapse
Affiliation(s)
- Johanna Lattner
- Max-Planck Institute of Molecular Cell Biology and Genetics (MPI-CBG)DresdenGermany
| | - Weihua Leng
- Max-Planck Institute of Molecular Cell Biology and Genetics (MPI-CBG)DresdenGermany
| | - Elisabeth Knust
- Max-Planck Institute of Molecular Cell Biology and Genetics (MPI-CBG)DresdenGermany
| | - Marko Brankatschk
- The Biotechnological Center of the TU Dresden (BIOTEC)DresdenGermany
| | - David Flores-Benitez
- Max-Planck Institute of Molecular Cell Biology and Genetics (MPI-CBG)DresdenGermany
| |
Collapse
|
39
|
Le Roux AL, Quiroga X, Walani N, Arroyo M, Roca-Cusachs P. The plasma membrane as a mechanochemical transducer. Philos Trans R Soc Lond B Biol Sci 2019; 374:20180221. [PMID: 31431176 PMCID: PMC6627014 DOI: 10.1098/rstb.2018.0221] [Citation(s) in RCA: 119] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/22/2019] [Indexed: 12/20/2022] Open
Abstract
Cells are constantly submitted to external mechanical stresses, which they must withstand and respond to. By forming a physical boundary between cells and their environment that is also a biochemical platform, the plasma membrane (PM) is a key interface mediating both cellular response to mechanical stimuli, and subsequent biochemical responses. Here, we review the role of the PM as a mechanosensing structure. We first analyse how the PM responds to mechanical stresses, and then discuss how this mechanical response triggers downstream biochemical responses. The molecular players involved in PM mechanochemical transduction include sensors of membrane unfolding, membrane tension, membrane curvature or membrane domain rearrangement. These sensors trigger signalling cascades fundamental both in healthy scenarios and in diseases such as cancer, which cells harness to maintain integrity, keep or restore homeostasis and adapt to their external environment. This article is part of a discussion meeting issue 'Forces in cancer: interdisciplinary approaches in tumour mechanobiology'.
Collapse
Affiliation(s)
- Anabel-Lise Le Roux
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology (BIST), Barcelona 08028, Spain
| | - Xarxa Quiroga
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology (BIST), Barcelona 08028, Spain
| | - Nikhil Walani
- LaCàN, Universitat Politècnica de Catalunya-BarcelonaTech, Spain
| | - Marino Arroyo
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology (BIST), Barcelona 08028, Spain
- LaCàN, Universitat Politècnica de Catalunya-BarcelonaTech, Spain
| | - Pere Roca-Cusachs
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology (BIST), Barcelona 08028, Spain
- Department of Biomedical Sciences, Universitat de Barcelona, Barcelona 08036, Spain
| |
Collapse
|
40
|
Ogi S, Matsuda A, Otsuka Y, Liu Z, Satoh T, Satoh AK. Syndapin constricts microvillar necks to form a united rhabdomere in Drosophila photoreceptors. Development 2019; 146:dev.169292. [PMID: 31371377 DOI: 10.1242/dev.169292] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 07/22/2019] [Indexed: 01/24/2023]
Abstract
Drosophila photoreceptors develop from polarized epithelial cells that have apical and basolateral membranes. During morphogenesis, the apical membranes subdivide into a united bundle of photosensory microvilli (rhabdomeres) and a surrounding supporting membrane (stalk). By EMS-induced mutagenesis screening, we found that the F-Bin/Amphiphysin/Rvs (F-BAR) protein syndapin is essential for apical membrane segregation. The analysis of the super-resolution microscopy, STORM and the electron microscopy suggest that syndapin localizes to the neck of the microvilli at the base of the rhabdomere. Syndapin and moesin are required to constrict the neck of the microvilli to organize the membrane architecture at the base of the rhabdomere, to exclude the stalk membrane. Simultaneous loss of syndapin along with the microvilli adhesion molecule chaoptin significantly enhanced the disruption of stalk-rhabdomere segregation. However, loss of the factors involving endocytosis do not interfere. These results indicated syndapin is most likely functioning through its membrane curvature properties, and not through endocytic processes for stalk-rhabdomere segregation. Elucidation of the mechanism of this unconventional domain formation will provide novel insights into the field of cell biology.
Collapse
Affiliation(s)
- Sakiko Ogi
- Program of Life and Environmental Science, Graduate School of Integral Science for Life, Hiroshima University, 1-7-1, Kagamiyama, Higashi-hiroshima, Hiroshima 739-8521, Japan
| | - Atsushi Matsuda
- National Institute of Information and Communications Technology, Advanced ICT Research Institute, 588-2, Iwaoka, Nishi-ku, Kobe 651-2492, Japan
| | - Yuna Otsuka
- Program of Life and Environmental Science, Graduate School of Integral Science for Life, Hiroshima University, 1-7-1, Kagamiyama, Higashi-hiroshima, Hiroshima 739-8521, Japan
| | - Ziguang Liu
- Program of Life and Environmental Science, Graduate School of Integral Science for Life, Hiroshima University, 1-7-1, Kagamiyama, Higashi-hiroshima, Hiroshima 739-8521, Japan.,Institute of Animal Husbandry, Heilongjiang Academy of Agricultural Sciences, Xuefu Road No. 368, Nangang District, Harbin, Heilongjiang 150-086, China
| | - Takunori Satoh
- Program of Life and Environmental Science, Graduate School of Integral Science for Life, Hiroshima University, 1-7-1, Kagamiyama, Higashi-hiroshima, Hiroshima 739-8521, Japan
| | - Akiko K Satoh
- Program of Life and Environmental Science, Graduate School of Integral Science for Life, Hiroshima University, 1-7-1, Kagamiyama, Higashi-hiroshima, Hiroshima 739-8521, Japan
| |
Collapse
|
41
|
Wang HY, Yuan H, Liu JH, Wang BL, Xu KL, Huang P, Lin ZH, Xu LH. Comparative analysis of a panel of biomarkers related to protein phosphatase 2A between laryngeal squamous cell carcinoma tissues and adjacent normal tissues. J Zhejiang Univ Sci B 2019; 20:776-780. [PMID: 31379147 DOI: 10.1631/jzus.b1900179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Laryngeal squamous cell carcinoma (LSCC) is the most common type of head and neck squamous cell carcinoma (HNSCC) worldwide. Protein phosphatase 2A (PP2A) dysfunction has been widely reported in a broad range of malignancies due to its distinctive role in miscellaneous cellular processes. However, it is poorly understood whether aberrant alterations of PP2A are involved in the network of oncogenic events in LSCC. Here, we detected a panel of PP2A-associated proteins using western blot in both laryngeal squamous cell carcinoma tissues and paired adjacent normal tissues from patients (Data S1). We found that phospho-PP2A/C (Y307), α4, cancerous inhibitor of protein phosphatase 2A (CIP2A), Akt, ezrin, phospho-ezrin (T567), 14-3-3, and focal adhesion kinase (FAK) showed increased expression levels in carcinoma tissues relative to normal tissues, while phospho-Akt (T308) showed decreased levels. Our study, thus, provides a rationale for targeting PP2A to develop novel therapies and proposes a combination of interrelated biomarkers for the diagnostic evaluation and prognosis prediction in LSCC.
Collapse
Affiliation(s)
- Han-Ying Wang
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China
| | - Hui Yuan
- Department of Otorhinolaryngology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Jing-Hui Liu
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40506, USA
| | - Bei-Lei Wang
- Department of Biochemistry, School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Kai-Lun Xu
- Cancer Institute, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Pu Huang
- Department of Biochemistry, School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Zhi-Hong Lin
- Department of Otorhinolaryngology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Li-Hong Xu
- Department of Biochemistry, School of Medicine, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
42
|
Derouiche A, Geiger KD. Perspectives for Ezrin and Radixin in Astrocytes: Kinases, Functions and Pathology. Int J Mol Sci 2019; 20:ijms20153776. [PMID: 31382374 PMCID: PMC6695708 DOI: 10.3390/ijms20153776] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 07/30/2019] [Accepted: 07/31/2019] [Indexed: 02/06/2023] Open
Abstract
Astrocytes are increasingly perceived as active partners in physiological brain function and behaviour. The structural correlations of the glia–synaptic interaction are the peripheral astrocyte processes (PAPs), where ezrin and radixin, the two astrocytic members of the ezrin-radixin-moesin (ERM) family of proteins are preferentially localised. While the molecular mechanisms of ERM (in)activation appear universal, at least in mammalian cells, and have been studied in great detail, the actual ezrin and radixin kinases, phosphatases and binding partners appear cell type specific and may be multiplexed within a cell. In astrocytes, ezrin is involved in process motility, which can be stimulated by the neurotransmitter glutamate, through activation of the glial metabotropic glutamate receptors (mGluRs) 3 or 5. However, it has remained open how this mGluR stimulus is transduced to ezrin activation. Knowing upstream signals of ezrin activation, ezrin kinase(s), and membrane-bound binding partners of ezrin in astrocytes might open new approaches to the glial role in brain function. Ezrin has also been implicated in invasive behaviour of astrocytomas, and glial activation. Here, we review data pertaining to potential molecular interaction partners of ezrin in astrocytes, with a focus on PKC and GRK2, and in gliomas and other diseases, to stimulate further research on their potential roles in glia-synaptic physiology and pathology.
Collapse
Affiliation(s)
- Amin Derouiche
- Institute of Anatomy II, Goethe-University Frankfurt, D-60590 Frankfurt am Main, Germany.
| | - Kathrin D Geiger
- Neuropathology, Institute for Pathology, Carl Gustav Carus University Hospital, TU Dresden, D-01307 Dresden, Germany
| |
Collapse
|
43
|
Polarity signaling ensures epidermal homeostasis by coupling cellular mechanics and genomic integrity. Nat Commun 2019; 10:3362. [PMID: 31358743 PMCID: PMC6662827 DOI: 10.1038/s41467-019-11325-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 07/05/2019] [Indexed: 02/07/2023] Open
Abstract
Epithelial homeostasis requires balanced progenitor cell proliferation and differentiation, whereas disrupting this equilibrium fosters degeneration or cancer. Here we studied how cell polarity signaling orchestrates epidermal self-renewal and differentiation. Using genetic ablation, quantitative imaging, mechanochemical reconstitution and atomic force microscopy, we find that mammalian Par3 couples genome integrity and epidermal fate through shaping keratinocyte mechanics, rather than mitotic spindle orientation. Par3 inactivation impairs RhoA activity, actomyosin contractility and viscoelasticity, eliciting mitotic failures that trigger aneuploidy, mitosis-dependent DNA damage responses, p53 stabilization and premature differentiation. Importantly, reconstituting myosin activity is sufficient to restore mitotic fidelity, genome integrity, and balanced differentiation and stratification. Collectively, this study deciphers a mechanical signaling network in which Par3 acts upstream of Rho/actomyosin contractility to promote intrinsic force generation, thereby maintaining mitotic accuracy and cellular fitness at the genomic level. Disturbing this network may compromise not only epidermal homeostasis but potentially also that of other self-renewing epithelia. Many developing tissues require Par-driven polarization, but its role in mammalian tissue maintenance is unclear. Here, the authors show that in mouse epidermis, Par3 governs tissue homeostasis not via orientation of cell division but by coupling cell mechanics with mitotic accuracy and genome integrity.
Collapse
|
44
|
Barbera S, Nardi F, Elia I, Realini G, Lugano R, Santucci A, Tosi GM, Dimberg A, Galvagni F, Orlandini M. The small GTPase Rab5c is a key regulator of trafficking of the CD93/Multimerin-2/β1 integrin complex in endothelial cell adhesion and migration. Cell Commun Signal 2019; 17:55. [PMID: 31138217 PMCID: PMC6537425 DOI: 10.1186/s12964-019-0375-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 05/20/2019] [Indexed: 02/06/2023] Open
Abstract
Background In the endothelium, the single-pass membrane protein CD93, through its interaction with the extracellular matrix protein Multimerin-2, activates signaling pathways that are critical for vascular development and angiogenesis. Trafficking of adhesion molecules through endosomal compartments modulates their signaling output. However, the mechanistic basis coordinating CD93 recycling and its implications for endothelial cell (EC) function remain elusive. Methods Human umbilical vein ECs (HUVECs) and human dermal blood ECs (HDBEC) were used in this study. Fluorescence confocal microscopy was employed to follow CD93 retrieval, recycling, and protein colocalization in spreading cells. To better define CD93 trafficking, drug treatments and transfected chimeric wild type and mutant CD93 proteins were used. The scratch assay was used to evaluate cell migration. Gene silencing strategies, flow citometry, and quantification of migratory capability were used to determine the role of Rab5c during CD93 recycling to the cell surface. Results Here, we identify the recycling pathway of CD93 following EC adhesion and migration. We show that the cytoplasmic domain of CD93, by its interaction with Moesin and F-actin, is instrumental for CD93 retrieval in adhering and migrating cells and that aberrant endosomal trafficking of CD93 prevents its localization at the leading edge of migration. Moreover, the small GTPase Rab5c turns out to be a key component of the molecular machinery that is able to drive CD93 recycling to the EC surface. Finally, in the Rab5c endosomal compartment CD93 forms a complex with Multimerin-2 and active β1 integrin, which is recycled back to the basolaterally-polarized cell surface by clathrin-independent endocytosis. Conclusions Our findings, focusing on the pro-angiogenic receptor CD93, unveil the mechanisms of its polarized trafficking during EC adhesion and migration, opening novel therapeutic opportunities for angiogenic diseases. Electronic supplementary material The online version of this article (10.1186/s12964-019-0375-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Stefano Barbera
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via A. Moro, 2, 53100, Siena, Italy
| | - Federica Nardi
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via A. Moro, 2, 53100, Siena, Italy
| | - Ines Elia
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via A. Moro, 2, 53100, Siena, Italy
| | - Giulia Realini
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via A. Moro, 2, 53100, Siena, Italy
| | - Roberta Lugano
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Rudbeck Laboratory, Uppsala University, SE-751 85, Uppsala, Sweden
| | - Annalisa Santucci
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via A. Moro, 2, 53100, Siena, Italy
| | - Gian Marco Tosi
- Department of Medicine, Surgery and Neuroscience, Ophthalmology Unit, University of Siena, Policlinico "Le Scotte", Viale Bracci, 53100, Siena, Italy
| | - Anna Dimberg
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Rudbeck Laboratory, Uppsala University, SE-751 85, Uppsala, Sweden
| | - Federico Galvagni
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via A. Moro, 2, 53100, Siena, Italy.
| | - Maurizio Orlandini
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via A. Moro, 2, 53100, Siena, Italy.
| |
Collapse
|
45
|
Michie KA, Bermeister A, Robertson NO, Goodchild SC, Curmi PMG. Two Sides of the Coin: Ezrin/Radixin/Moesin and Merlin Control Membrane Structure and Contact Inhibition. Int J Mol Sci 2019; 20:ijms20081996. [PMID: 31018575 PMCID: PMC6515277 DOI: 10.3390/ijms20081996] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 04/16/2019] [Accepted: 04/19/2019] [Indexed: 12/21/2022] Open
Abstract
The merlin-ERM (ezrin, radixin, moesin) family of proteins plays a central role in linking the cellular membranes to the cortical actin cytoskeleton. Merlin regulates contact inhibition and is an integral part of cell–cell junctions, while ERM proteins, ezrin, radixin and moesin, assist in the formation and maintenance of specialized plasma membrane structures and membrane vesicle structures. These two protein families share a common evolutionary history, having arisen and separated via gene duplication near the origin of metazoa. During approximately 0.5 billion years of evolution, the merlin and ERM family proteins have maintained both sequence and structural conservation to an extraordinary level. Comparing crystal structures of merlin-ERM proteins and their complexes, a picture emerges of the merlin-ERM proteins acting as switchable interaction hubs, assembling protein complexes on cellular membranes and linking them to the actin cytoskeleton. Given the high level of structural conservation between the merlin and ERM family proteins we speculate that they may function together.
Collapse
Affiliation(s)
- Katharine A Michie
- School of Physics, University of New South Wales, Sydney 2052, Australia.
| | - Adam Bermeister
- School of Physics, University of New South Wales, Sydney 2052, Australia.
| | - Neil O Robertson
- School of Physics, University of New South Wales, Sydney 2052, Australia.
| | - Sophia C Goodchild
- Department of Molecular Sciences, Macquarie University, Sydney 2109, Australia.
| | - Paul M G Curmi
- School of Physics, University of New South Wales, Sydney 2052, Australia.
| |
Collapse
|
46
|
Zimmermann D, Kovar DR. Feeling the force: formin's role in mechanotransduction. Curr Opin Cell Biol 2019; 56:130-140. [PMID: 30639952 DOI: 10.1016/j.ceb.2018.12.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 12/13/2018] [Accepted: 12/16/2018] [Indexed: 11/15/2022]
Abstract
Fundamental cellular processes such as division, polarization, and motility require the tightly regulated spatial and temporal assembly and disassembly of the underlying actin cytoskeleton. The actin cytoskeleton has been long viewed as a central player facilitating diverse mechanotransduction pathways due to the notion that it is capable of receiving, processing, transmitting, and generating mechanical stresses. Recent work has begun to uncover the roles of mechanical stresses in modulating the activity of key regulatory actin-binding proteins and their interactions with actin filaments, thereby controlling the assembly (formin and Arp2/3 complex) and disassembly (ADF/Cofilin) of actin filament networks. In this review, we will focus on discussing the current molecular understanding of how members of the formin protein family sense and respond to forces and the potential implications for formin-mediated mechanotransduction in cells.
Collapse
Affiliation(s)
- Dennis Zimmermann
- Massachusetts Institute of Technology, David H. Koch Institute for Integrative Cancer Research, 77 Massachusetts Ave, 76-361F, Cambridge, MA 02139-4307, United States.
| | - David R Kovar
- The University of Chicago, Department of Molecular Genetics and Cell Biology, 90 E. 58th Street, CSLC 212, Chicago, IL 60637, United States.
| |
Collapse
|
47
|
Kopp S, Krüger M, Bauer J, Wehland M, Corydon TJ, Sahana J, Nassef MZ, Melnik D, Bauer TJ, Schulz H, Schütte A, Schmitz B, Oltmann H, Feldmann S, Infanger M, Grimm D. Microgravity Affects Thyroid Cancer Cells during the TEXUS-53 Mission Stronger than Hypergravity. Int J Mol Sci 2018; 19:E4001. [PMID: 30545079 PMCID: PMC6321223 DOI: 10.3390/ijms19124001] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 12/06/2018] [Accepted: 12/09/2018] [Indexed: 12/24/2022] Open
Abstract
Thyroid cancer is the most abundant tumor of the endocrine organs. Poorly differentiated thyroid cancer is still difficult to treat. Human cells exposed to long-term real (r-) and simulated (s-) microgravity (µg) revealed morphological alterations and changes in the expression profile of genes involved in several biological processes. The objective of this study was to examine the effects of short-term µg on poorly differentiated follicular thyroid cancer cells (FTC-133 cell line) resulting from 6 min of exposure to µg on a sounding rocket flight. As sounding rocket flights consist of several flight phases with different acceleration forces, rigorous control experiments are mandatory. Hypergravity (hyper-g) experiments were performed at 18g on a centrifuge in simulation of the rocket launch and s-µg was simulated by a random positioning machine (RPM). qPCR analyses of selected genes revealed no remarkable expression changes in controls as well as in hyper-g samples taken at the end of the first minute of launch. Using a centrifuge initiating 18g for 1 min, however, presented moderate gene expression changes, which were significant for COL1A1, VCL, CFL1, PTK2, IL6, CXCL8 and MMP14. We also identified a network of mutual interactions of the investigated genes and proteins by employing in-silico analyses. Lastly, µg-samples indicated that microgravity is a stronger regulator of gene expression than hyper-g.
Collapse
Affiliation(s)
- Sascha Kopp
- Clinic for Plastic, Aesthetic and Hand Surgery, Otto von Guericke University Magdeburg, Leipziger Str. 44, D-39120 Magdeburg, Germany.
| | - Marcus Krüger
- Clinic for Plastic, Aesthetic and Hand Surgery, Otto von Guericke University Magdeburg, Leipziger Str. 44, D-39120 Magdeburg, Germany.
| | - Johann Bauer
- Max Planck Institute of Biochemistry, D-82152 Martinsried, Germany.
| | - Markus Wehland
- Clinic for Plastic, Aesthetic and Hand Surgery, Otto von Guericke University Magdeburg, Leipziger Str. 44, D-39120 Magdeburg, Germany.
| | - Thomas J Corydon
- Department of Biomedicine, Aarhus University, Wilhelm Meyers Allé 4, DK-8000 Aarhus C, Denmark.
- Department of Ophthalmology, Aarhus University Hospital, Aarhus, 8000 Aarhus C, Denmark.
| | - Jayashree Sahana
- Department of Biomedicine, Aarhus University, Wilhelm Meyers Allé 4, DK-8000 Aarhus C, Denmark.
| | - Mohamed Zakaria Nassef
- Clinic for Plastic, Aesthetic and Hand Surgery, Otto von Guericke University Magdeburg, Leipziger Str. 44, D-39120 Magdeburg, Germany.
| | - Daniela Melnik
- Clinic for Plastic, Aesthetic and Hand Surgery, Otto von Guericke University Magdeburg, Leipziger Str. 44, D-39120 Magdeburg, Germany.
| | - Thomas J Bauer
- Clinic for Plastic, Aesthetic and Hand Surgery, Otto von Guericke University Magdeburg, Leipziger Str. 44, D-39120 Magdeburg, Germany.
| | - Herbert Schulz
- Cologne Center for Genomics, University of Cologne, D-50931 Cologne, Germany.
| | - Andreas Schütte
- Airbus Defence and Space GmbH, Airbus-Allee 1, D-28199 Bremen, Germany.
| | - Burkhard Schmitz
- Airbus Defence and Space GmbH, Airbus-Allee 1, D-28199 Bremen, Germany.
| | - Hergen Oltmann
- Airbus Defence and Space GmbH, Airbus-Allee 1, D-28199 Bremen, Germany.
| | - Stefan Feldmann
- Airbus Defence and Space GmbH, Airbus-Allee 1, D-28199 Bremen, Germany.
| | - Manfred Infanger
- Clinic for Plastic, Aesthetic and Hand Surgery, Otto von Guericke University Magdeburg, Leipziger Str. 44, D-39120 Magdeburg, Germany.
| | - Daniela Grimm
- Clinic for Plastic, Aesthetic and Hand Surgery, Otto von Guericke University Magdeburg, Leipziger Str. 44, D-39120 Magdeburg, Germany.
- Department of Biomedicine, Aarhus University, Wilhelm Meyers Allé 4, DK-8000 Aarhus C, Denmark.
- Gravitational Biology and Translational Regenerative Medicine, Faculty of Medicine and Mechanical Engineering, Otto-von-Guericke-University Magdeburg, D-39120 Magdeburg, Germany.
| |
Collapse
|
48
|
Simonova VV, Vetchinova AS, Novosadova EV, Khaspekov LG, Illarioshkin SN. Genome Editing and the Problem of Tetraploidy in Cell Modeling of the Genetic Form of Parkinsonism. BIOCHEMISTRY (MOSCOW) 2018; 83:1040-1045. [PMID: 30472942 DOI: 10.1134/s0006297918090055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The prevalent form of familial parkinsonism is caused by mutations in the LRRK2 gene encoding for the mitochondrial protein kinase. In the review, we discuss possible causes of appearance of tetraploid cells in neuronal precursors obtained from induced pluripotent stem cells from patients with the LRRK2-associated form of parkinsonism after genome editing procedure. As LRRK2 protein participates in cell proliferation and maintenance of the nuclear envelope, spindle fibers, and cytoskeleton, mutations in the LRRK2 gene can affect protein functions and lead, via various mechanisms, to the mitotic machinery disintegration and chromosomal aberration. These abnormalities can appear at different stages of fibroblast reprogramming; therefore, editing of the LRRK2 nucleotide sequence should be done during or before the reprogramming stage.
Collapse
Affiliation(s)
- V V Simonova
- Research Center of Neurology, Moscow, 125367, Russia
| | | | - E V Novosadova
- Institute of Molecular Genetics, Russian Academy of Sciences, Moscow, 123182, Russia.
| | - L G Khaspekov
- Research Center of Neurology, Moscow, 125367, Russia.
| | | |
Collapse
|
49
|
Halova I, Bambouskova M, Draberova L, Bugajev V, Draber P. The transmembrane adaptor protein NTAL limits mast cell chemotaxis toward prostaglandin E2. Sci Signal 2018; 11:11/556/eaao4354. [DOI: 10.1126/scisignal.aao4354] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Chemotaxis of mast cells is one of the crucial steps in their development and function. Non–T cell activation linker (NTAL) is a transmembrane adaptor protein that inhibits the activation of mast cells and B cells in a phosphorylation-dependent manner. Here, we studied the role of NTAL in the migration of mouse mast cells stimulated by prostaglandin E2 (PGE2). Although PGE2 does not induce the tyrosine phosphorylation of NTAL, unlike IgE immune complex antigens, we found that loss of NTAL increased the chemotaxis of mast cells toward PGE2. Stimulation of mast cells that lacked NTAL with PGE2 enhanced the phosphorylation of AKT and the production of phosphatidylinositol 3,4,5-trisphosphate. In resting NTAL-deficient mast cells, phosphorylation of an inhibitory threonine in ERM family proteins accompanied increased activation of β1-containing integrins, which are features often associated with increased invasiveness in tumors. Rescue experiments indicated that only full-length, wild-type NTAL restored the chemotaxis of NTAL-deficient cells toward PGE2. Together, these data suggest that NTAL is a key inhibitor of mast cell chemotaxis toward PGE2, which may act through the RHOA/ERM/β1-integrin and PI3K/AKT axes.
Collapse
|
50
|
Chiasson-MacKenzie C, Morris ZS, Liu CH, Bradford WB, Koorman T, McClatchey AI. Merlin/ERM proteins regulate growth factor-induced macropinocytosis and receptor recycling by organizing the plasma membrane:cytoskeleton interface. Genes Dev 2018; 32:1201-1214. [PMID: 30143526 PMCID: PMC6120716 DOI: 10.1101/gad.317354.118] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 07/20/2018] [Indexed: 12/19/2022]
Abstract
The architectural and biochemical features of the plasma membrane are governed by its intimate association with the underlying cortical cytoskeleton. The neurofibromatosis type 2 (NF2) tumor suppressor merlin and closely related membrane:cytoskeleton-linking protein ezrin organize the membrane:cytoskeleton interface, a critical cellular compartment that both regulates and is regulated by growth factor receptors. An example of this poorly understood interrelationship is macropinocytosis, an ancient process of nutrient uptake and membrane remodeling that can both be triggered by growth factors and manage receptor availability. We show that merlin deficiency primes the membrane:cytoskeleton interface for epidermal growth factor (EGF)-induced macropinocytosis via a mechanism involving increased cortical ezrin, altered actomyosin, and stabilized cholesterol-rich membranes. These changes profoundly alter EGF receptor (EGFR) trafficking in merlin-deficient cells, favoring increased membrane levels of its heterodimerization partner, ErbB2; clathrin-independent internalization; and recycling. Our work suggests that, unlike Ras transformed cells, merlin-deficient cells do not depend on macropinocytic protein scavenging and instead exploit macropinocytosis for receptor recycling. Finally, we provide evidence that the macropinocytic proficiency of NF2-deficient cells can be used for therapeutic uptake. This work provides new insight into fundamental mechanisms of macropinocytic uptake and processing and suggests new ways to interfere with or exploit macropinocytosis in NF2 mutant and other tumors.
Collapse
Affiliation(s)
- Christine Chiasson-MacKenzie
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, Massachusetts 02129, USA
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Zachary S Morris
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, Massachusetts 02129, USA
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Ching-Hui Liu
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, Massachusetts 02129, USA
| | - William B Bradford
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, Massachusetts 02129, USA
| | - Thijs Koorman
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, Massachusetts 02129, USA
| | - Andrea I McClatchey
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, Massachusetts 02129, USA
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| |
Collapse
|