1
|
Kageler L, Perr J, Flynn RA. Tools to investigate the cell surface: Proximity as a central concept in glycoRNA biology. Cell Chem Biol 2024; 31:1132-1144. [PMID: 38772372 PMCID: PMC11193615 DOI: 10.1016/j.chembiol.2024.04.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 04/02/2024] [Accepted: 04/25/2024] [Indexed: 05/23/2024]
Abstract
Proximity is a fundamental concept in chemistry and biology, referring to the convergence of molecules to facilitate new molecular interactions or reactions. Hybrid biopolymers like glycosylphosphatidylinositol (GPI)-anchored proteins, ubiquitinated proteins, glycosylated RNAs (glycoRNAs), and RNAylated proteins exemplify this by covalent bonding of moieties that are often orthogonally active. Hybrid molecules like glycoRNAs are localized to new physical spaces, generating new interfaces for biological functions. To fully investigate the compositional and spatial features of molecules like glycoRNAs, flexible genetic and chemical tools that encompass different encoding and targeting biopolymers are required. Here we discuss concepts of molecular proximity and explore newer proximity labeling technologies that facilitate applications in RNA biology, cell surface biology, and the interface therein with a particular focus on glycoRNA biology. We review the advantages and disadvantages of methods pertaining to cell surface RNA identification and provide insights into the vast opportunities for method development in this area.
Collapse
Affiliation(s)
- Lauren Kageler
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA
| | - Jonathan Perr
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA
| | - Ryan A Flynn
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
2
|
Blake LA, De La Cruz A, Wu B. Imaging spatiotemporal translation regulation in vivo. Semin Cell Dev Biol 2024; 154:155-164. [PMID: 36963991 PMCID: PMC10514244 DOI: 10.1016/j.semcdb.2023.03.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 03/08/2023] [Accepted: 03/15/2023] [Indexed: 03/26/2023]
Abstract
Translation is regulated spatiotemporally to direct protein synthesis when and where it is needed. RNA localization and local translation have been observed in various subcellular compartments, allowing cells to rapidly and finely adjust their proteome post-transcriptionally. Local translation on membrane-bound organelles is important to efficiently synthesize proteins targeted to the organelles. Protein-RNA phase condensates restrict RNA spatially in membraneless organelles and play essential roles in translation regulation and RNA metabolism. In addition, the temporal translation kinetics not only determine the amount of protein produced, but also serve as an important checkpoint for the quality of ribosomes, mRNAs, and nascent proteins. Translation imaging provides a unique capability to study these fundamental processes in the native environment. Recent breakthroughs in imaging enabled real-time visualization of translation of single mRNAs, making it possible to determine the spatial distribution and key biochemical parameters of in vivo translation dynamics. Here we reviewed the recent advances in translation imaging methods and their applications to study spatiotemporal translation regulation in vivo.
Collapse
Affiliation(s)
- Lauren A Blake
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; The Center for Cell Dynamics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ana De La Cruz
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; The Center for Cell Dynamics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Bin Wu
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; The Solomon H Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; The Center for Cell Dynamics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
3
|
Phillips AT, Boumil EF, Venkatesan A, Tilstra-Smith C, Castro N, Knox BE, Henty-Ridilla JL, Bernstein AM. The formin DAAM1 regulates the deubiquitinase activity of USP10 and integrin homeostasis. Eur J Cell Biol 2023; 102:151347. [PMID: 37562219 PMCID: PMC10839120 DOI: 10.1016/j.ejcb.2023.151347] [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: 07/12/2022] [Revised: 08/02/2023] [Accepted: 08/04/2023] [Indexed: 08/12/2023] Open
Abstract
The differentiation of fibroblasts into pathological myofibroblasts during wound healing is characterized by increased cell surface expression of αv-integrins. Our previous studies found that the deubiquitinase (DUB) USP10 removes ubiquitin from αv-integrins, leading to cell surface integrin accumulation, subsequent TGFβ1 activation, and pathological myofibroblast differentiation. In this study, a yeast two-hybrid screen revealed a novel binding partner for USP10, the formin, DAAM1. We found that DAAM1 binds to and inhibits USP10's DUB activity through the FH2 domain of DAAM1 independent of its actin functions. The USP10/DAAM1 interaction was also supported by proximity ligation assay (PLA) in primary human corneal fibroblasts. Treatment with TGFβ1 significantly increased USP10 and DAAM1 protein expression, PLA signal, and co-localization to actin stress fibers. DAAM1 siRNA knockdown significantly reduced co-precipitation of USP10 and DAAM1 on purified actin stress fibers, and β1- and β5-integrin ubiquitination. This resulted in increased αv-, β1-, and β5-integrin total protein levels, αv-integrin recycling, and extracellular fibronectin (FN) deposition. Together, our data demonstrate that DAAM1 inhibits USP10's DUB activity on integrins subsequently regulating cell surface αv-integrin localization and FN accumulation.
Collapse
Affiliation(s)
- Andrew T Phillips
- SUNY Upstate Medical University, Department of Ophthalmology and Visual Sciences, 750 East Adams Street, Syracuse, NY 13210, USA
| | - Edward F Boumil
- SUNY Upstate Medical University, Department of Ophthalmology and Visual Sciences, 750 East Adams Street, Syracuse, NY 13210, USA
| | - Arunkumar Venkatesan
- SUNY Upstate Medical University, Department of Ophthalmology and Visual Sciences, 750 East Adams Street, Syracuse, NY 13210, USA
| | - Christine Tilstra-Smith
- SUNY Upstate Medical University, Department of Ophthalmology and Visual Sciences, 750 East Adams Street, Syracuse, NY 13210, USA
| | - Nileyma Castro
- SUNY Upstate Medical University, Department of Ophthalmology and Visual Sciences, 750 East Adams Street, Syracuse, NY 13210, USA; New York VA Health Care, Syracuse VA Medical Center, 800 Irving Ave, Syracuse 13210, USA
| | - Barry E Knox
- SUNY Upstate Medical University, Department of Ophthalmology and Visual Sciences, 750 East Adams Street, Syracuse, NY 13210, USA; SUNY Upstate Medical University, Biochemistry and Molecular Biology, 750 East Adams Street, Syracuse, NY 13210, USA
| | - Jessica L Henty-Ridilla
- SUNY Upstate Medical University, Biochemistry and Molecular Biology, 750 East Adams Street, Syracuse, NY 13210, USA
| | - Audrey M Bernstein
- SUNY Upstate Medical University, Department of Ophthalmology and Visual Sciences, 750 East Adams Street, Syracuse, NY 13210, USA; SUNY Upstate Medical University, Biochemistry and Molecular Biology, 750 East Adams Street, Syracuse, NY 13210, USA; New York VA Health Care, Syracuse VA Medical Center, 800 Irving Ave, Syracuse 13210, USA.
| |
Collapse
|
4
|
Phan QT, Solis NV, Cravener MV, Swidergall M, Lin J, Huang MY, Liu H, Singh S, Ibrahim AS, Mazzone M, Mitchell AP, Filler SG. Candida albicans stimulates formation of a multi-receptor complex that mediates epithelial cell invasion during oropharyngeal infection. PLoS Pathog 2023; 19:e1011579. [PMID: 37611070 PMCID: PMC10479894 DOI: 10.1371/journal.ppat.1011579] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 09/05/2023] [Accepted: 07/25/2023] [Indexed: 08/25/2023] Open
Abstract
Fungal invasion of the oral epithelium is central to the pathogenesis of oropharyngeal candidiasis (OPC). Candida albicans invades the oral epithelium by receptor-induced endocytosis but this process is incompletely understood. We found that C. albicans infection of oral epithelial cells induces c-Met to form a multi-protein complex with E-cadherin and the epidermal growth factor receptor (EGFR). E-cadherin is necessary for C. albicans to activate both c-Met and EGFR and to induce the endocytosis of C. albicans. Proteomics analysis revealed that c-Met interacts with C. albicans Hyr1, Als3 and Ssa1. Both Hyr1 and Als3 are required for C. albicans to stimulate c-Met and EGFR in oral epithelial cells in vitro and for full virulence during OPC in mice. Treating mice with small molecule inhibitors of c-Met and EGFR ameliorates OPC, demonstrating the potential therapeutic efficacy of blocking these host receptors for C. albicans.
Collapse
Affiliation(s)
- Quynh T. Phan
- Institute for Infection and Immunity, Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California, United States of America
| | - Norma V. Solis
- Institute for Infection and Immunity, Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California, United States of America
| | - Max V. Cravener
- Department of Microbiology, University of Georgia, Athens, Georgia United States of America
| | - Marc Swidergall
- Institute for Infection and Immunity, Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California, United States of America
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - Jianfeng Lin
- Institute for Infection and Immunity, Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California, United States of America
| | - Manning Y. Huang
- Department of Biochemistry and Biophysics, University of California, San Francisco, California, United States of America
| | - Hong Liu
- Institute for Infection and Immunity, Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California, United States of America
| | - Shakti Singh
- Institute for Infection and Immunity, Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California, United States of America
| | - Ashraf S. Ibrahim
- Institute for Infection and Immunity, Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California, United States of America
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - Massimiliano Mazzone
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Aaron P. Mitchell
- Department of Microbiology, University of Georgia, Athens, Georgia United States of America
| | - Scott G. Filler
- Institute for Infection and Immunity, Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California, United States of America
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| |
Collapse
|
5
|
Sachsenweger J, Jansche R, Merk T, Heitmeir B, Deniz M, Faust U, Roggia C, Tzschach A, Schroeder C, Riess A, Pospiech H, Peltoketo H, Pylkäs K, Winqvist R, Wiesmüller L. ABRAXAS1 orchestrates BRCA1 activities to counter genome destabilizing repair pathways-lessons from breast cancer patients. Cell Death Dis 2023; 14:328. [PMID: 37198153 DOI: 10.1038/s41419-023-05845-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 04/27/2023] [Accepted: 05/02/2023] [Indexed: 05/19/2023]
Abstract
It has been well-established that mutations in BRCA1 and BRCA2, compromising functions in DNA double-strand break repair (DSBR), confer hereditary breast and ovarian cancer risk. Importantly, mutations in these genes explain only a minor fraction of the hereditary risk and of the subset of DSBR deficient tumors. Our screening efforts identified two truncating germline mutations in the gene encoding the BRCA1 complex partner ABRAXAS1 in German early-onset breast cancer patients. To unravel the molecular mechanisms triggering carcinogenesis in these carriers of heterozygous mutations, we examined DSBR functions in patient-derived lymphoblastoid cells (LCLs) and in genetically manipulated mammary epithelial cells. By use of these strategies we were able to demonstrate that these truncating ABRAXAS1 mutations exerted dominant effects on BRCA1 functions. Interestingly, we did not observe haploinsufficiency regarding homologous recombination (HR) proficiency (reporter assay, RAD51-foci, PARP-inhibitor sensitivity) in mutation carriers. However, the balance was shifted to use of mutagenic DSBR-pathways. The dominant effect of truncated ABRAXAS1 devoid of the C-terminal BRCA1 binding site can be explained by retention of the N-terminal interaction sites for other BRCA1-A complex partners like RAP80. In this case BRCA1 was channeled from the BRCA1-A to the BRCA1-C complex, which induced single-strand annealing (SSA). Further truncation, additionally deleting the coiled-coil region of ABRAXAS1, unleashed excessive DNA damage responses (DDRs) de-repressing multiple DSBR-pathways including SSA and non-homologous end-joining (NHEJ). Our data reveal de-repression of low-fidelity repair activities as a common feature of cells from patients with heterozygous mutations in genes encoding BRCA1 and its complex partners.
Collapse
Affiliation(s)
- Juliane Sachsenweger
- Department of Obstetrics and Gynecology, Ulm University, Ulm, Germany
- Laboratory of Cancer Genetics and Tumor Biology, Translational Medicine Research Unit, Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Rebecca Jansche
- Department of Obstetrics and Gynecology, Ulm University, Ulm, Germany
| | - Tatjana Merk
- Department of Obstetrics and Gynecology, Ulm University, Ulm, Germany
| | - Benedikt Heitmeir
- Department of Obstetrics and Gynecology, Ulm University, Ulm, Germany
| | - Miriam Deniz
- Department of Obstetrics and Gynecology, Ulm University, Ulm, Germany
| | - Ulrike Faust
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - Cristiana Roggia
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - Andreas Tzschach
- Institute of Human Genetics, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Christopher Schroeder
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - Angelika Riess
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - Helmut Pospiech
- Leibniz Institute on Aging - Fritz Lipmann Institute, Jena, Germany
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Hellevi Peltoketo
- Laboratory of Cancer Genetics and Tumor Biology, Translational Medicine Research Unit, Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Katri Pylkäs
- Laboratory of Cancer Genetics and Tumor Biology, Translational Medicine Research Unit, Biocenter Oulu, University of Oulu, Oulu, Finland
- Laboratory of Cancer Genetics and Tumor Biology, Northern Finland Laboratory Centre, Oulu, Finland
| | - Robert Winqvist
- Laboratory of Cancer Genetics and Tumor Biology, Translational Medicine Research Unit, Biocenter Oulu, University of Oulu, Oulu, Finland
- Laboratory of Cancer Genetics and Tumor Biology, Northern Finland Laboratory Centre, Oulu, Finland
| | - Lisa Wiesmüller
- Department of Obstetrics and Gynecology, Ulm University, Ulm, Germany.
| |
Collapse
|
6
|
Phan QT, Solis NV, Cravener MV, Swidergall M, Lin J, Huang MY, Liu H, Singh S, Ibrahim AS, Mazzone M, Mitchell AP, Filler SG. Candida albicans stimulates the formation of a multi-receptor complex that mediates epithelial cell invasion during oropharyngeal infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.23.529756. [PMID: 36865306 PMCID: PMC9980113 DOI: 10.1101/2023.02.23.529756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
Abstract
Fungal invasion of the oral epithelium is central to the pathogenesis of oropharyngeal candidiasis (OPC). Candida albicans invades the oral epithelium by receptor-induced endocytosis but this process is incompletely understood. We found that C. albicans infection of oral epithelial cells induces c-Met to form a multi-protein complex with E-cadherin and the epidermal growth factor receptor (EGFR). E-cadherin is necessary for C. albicans to activate both c-Met and EGFR and to induce the endocytosis of C. albicans . Proteomics analysis revealed that c-Met interacts with C. albicans Hyr1, Als3 and Ssa1. Both Hyr1 and Als3 were required for C. albicans stimulation of c-Met and EGFR in oral epithelial cells in vitro and for full virulence during OPC in mice. Treating mice with small molecule inhibitors of c-Met and EGFR ameliorated OPC, demonstrating the potential therapeutic efficacy of blocking these host receptors for C. albicans . Graphical abstract Highlights c-Met is an oral epithelial cell receptor for Candida albicans C. albicans infection causes c-Met and the epidermal growth factor receptor (EGFR) to form a complex with E-cadherin, which is required for c-Met and EGFR function C. albicans Hyr1 and Als3 interact with c-Met and EGFR, inducing oral epithelial cell endocytosis and virulence during oropharyngeal candidiasis Dual blockade of c-Met and EGFR ameliorates oropharyngeal candidiasis.
Collapse
Affiliation(s)
- Quynh T. Phan
- Institute for Infection and Immunity, Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Norma V. Solis
- Institute for Infection and Immunity, Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Max V. Cravener
- Department of Microbiology, University of Georgia, Athens, Georgia 30602, USA
| | - Marc Swidergall
- Institute for Infection and Immunity, Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Jianfeng Lin
- Institute for Infection and Immunity, Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Manning Y. Huang
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA, USA
| | - Hong Liu
- Institute for Infection and Immunity, Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Shakti Singh
- Institute for Infection and Immunity, Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Ashraf S. Ibrahim
- Institute for Infection and Immunity, Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Massimiliano Mazzone
- 1Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven B3000, Belgium
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven B3000, Belgium
| | - Aaron P. Mitchell
- Department of Microbiology, University of Georgia, Athens, Georgia 30602, USA
| | - Scott G. Filler
- Institute for Infection and Immunity, Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| |
Collapse
|
7
|
Hayat U, Siddiqui AA, Farhan ML, Haris A, Hameed N. Genome Editing and Fatty Liver. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1396:191-206. [DOI: 10.1007/978-981-19-5642-3_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
|
8
|
Phan QT, Lin J, Solis NV, Eng M, Swidergall M, Wang F, Li S, Gaffen SL, Chou TF, Filler SG. The Globular C1q Receptor Is Required for Epidermal Growth Factor Receptor Signaling during Candida albicans Infection. mBio 2021; 12:e0271621. [PMID: 34724825 PMCID: PMC8561387 DOI: 10.1128/mbio.02716-21] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Accepted: 09/21/2021] [Indexed: 02/07/2023] Open
Abstract
During oropharyngeal candidiasis, Candida albicans activates the epidermal growth factor receptor (EGFR), which induces oral epithelial cells to endocytose the fungus and synthesize proinflammatory mediators. To elucidate EGFR signaling pathways that are stimulated by C. albicans, we used proteomics to identify 1,214 proteins that were associated with EGFR in C. albicans-infected cells. Seven of these proteins were selected for additional study. Among these proteins, WW domain-binding protein 2, Toll-interacting protein, interferon-induced transmembrane protein 3 (IFITM3), and the globular C1q receptor (gC1qR) were found to associate with EGFR in viable oral epithelial cells. Each of these proteins was required for maximal endocytosis of C. albicans, and all regulated fungus-induced production of interleukin-1β (IL-1β) and/or IL-8, either positively or negatively. gC1qR was found to function as a key coreceptor with EGFR. Interacting with the C. albicans Als3 invasin, gC1qR was required for the fungus to induce autophosphorylation of both EGFR and the ephrin type A receptor 2. The combination of gC1qR and EGFR was necessary for maximal endocytosis of C. albicans and secretion of IL-1β, IL-8, and granulocyte-macrophage colony-stimulating factor (GM-CSF) by human oral epithelial cells. In mouse oral epithelial cells, inhibition of gC1qR failed to block C. albicans-induced phosphorylation, and knockdown of IFITM3 did not inhibit C. albicans endocytosis, indicating that gC1qR and IFITM3 function differently in mouse versus human oral epithelial cells. Thus, this work provides an atlas of proteins that associate with EGFR and identifies several that play a central role in the response of human oral epithelial cells to C. albicans infection. IMPORTANCE Oral epithelial cells play a key role in the pathogenesis of oropharyngeal candidiasis. In addition to being target host cells for C. albicans adherence and invasion, they secrete proinflammatory cytokines and chemokines that recruit T cells and activated phagocytes to foci of infection. It is known that C. albicans activates EGFR on oral epithelial cells, which induces these cells to endocytose the organism and stimulates them to secrete proinflammatory mediators. To elucidate the EGFR signaling pathways that govern these responses, we analyzed the epithelial cell proteins that associate with EGFR in C. albicans-infected epithelial cells. We identified four proteins that physically associate with EGFR and that regulate different aspects of the epithelial response to C. albicans. One of these is gC1qR, which is required for C. albicans to activate EGFR, induce endocytosis, and stimulate the secretion of proinflammatory mediators, indicating that gC1qR functions as a key coreceptor with EGFR.
Collapse
Affiliation(s)
- Quynh T. Phan
- Institute for Infection and Immunity, Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California, USA
| | - Jianfeng Lin
- Institute for Infection and Immunity, Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California, USA
| | - Norma V. Solis
- Institute for Infection and Immunity, Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California, USA
| | - Michael Eng
- Institute for Infection and Immunity, Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California, USA
| | - Marc Swidergall
- Institute for Infection and Immunity, Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California, USA
- David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Feng Wang
- Department of Pediatrics, Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California, USA
| | - Shan Li
- Department of Pediatrics, Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California, USA
| | - Sarah L. Gaffen
- Division of Rheumatology and Clinical Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Tsui-Fen Chou
- Department of Pediatrics, Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California, USA
| | - Scott G. Filler
- Institute for Infection and Immunity, Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California, USA
- David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| |
Collapse
|
9
|
Sundaria N, Upadhyay A, Prasad A, Prajapati VK, Poluri KM, Mishra A. Neurodegeneration & imperfect ageing: Technological limitations and challenges? Mech Ageing Dev 2021; 200:111574. [PMID: 34562507 DOI: 10.1016/j.mad.2021.111574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 08/29/2021] [Accepted: 09/21/2021] [Indexed: 11/18/2022]
Abstract
Cellular homeostasis is regulated by the protein quality control (PQC) machinery, comprising multiple chaperones and enzymes. Studies suggest that the loss of the PQC mechanisms in neurons may lead to the formation of abnormal inclusions that may lead to neurological disorders and defective aging. The questions could be raised how protein aggregate formation precisely engenders multifactorial molecular pathomechanism in neuronal cells and affects different brain regions? Such questions await thorough investigation that may help us understand how aberrant proteinaceous bodies lead to neurodegeneration and imperfect aging. However, these studies face multiple technological challenges in utilizing available tools for detailed characterizations of the protein aggregates or amyloids and developing new techniques to understand the biology and pathology of proteopathies. The lack of detection and analysis methods has decelerated the pace of the research in amyloid biology. Here, we address the significance of aggregation and inclusion formation, followed by exploring the evolutionary contribution of these structures. We also provide a detailed overview of current state-of-the-art techniques and advances in studying amyloids in the diseased brain. A comprehensive understanding of the structural, pathological, and clinical characteristics of different types of aggregates (inclusions, fibrils, plaques, etc.) will aid in developing future therapies.
Collapse
Affiliation(s)
- Naveen Sundaria
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology, Jodhpur, Rajasthan, 342037, India
| | - Arun Upadhyay
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology, Jodhpur, Rajasthan, 342037, India
| | - Amit Prasad
- School of Basic Sciences, Indian Institute of Technology Mandi, Mandi, India
| | - Vijay Kumar Prajapati
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, NH‑8 Bandarsindri, Ajmer, Rajasthan, 305817, India
| | - Krishna Mohan Poluri
- Department of Biotechnology, Indian Institute of Technology Roorkee, Roorkee, 247667, Uttarakhand, India
| | - Amit Mishra
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology, Jodhpur, Rajasthan, 342037, India.
| |
Collapse
|
10
|
Åberg M, Edén D, Siegbahn A. Activation of β1 integrins and caveolin-1 by TF/FVIIa promotes IGF-1R signaling and cell survival. Apoptosis 2021; 25:519-534. [PMID: 32458278 PMCID: PMC7347522 DOI: 10.1007/s10495-020-01611-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The tissue factor/coagulation factor VIIa (TF/FVIIa) complex induces transactivation of the IGF-1 receptor (IGF-1R) in a number of different cell types. The mechanism is largely unknown. The transactivation leads to protection from apoptosis and nuclear translocation of the IGF-1R. The aim of this study was to clarify the signaling pathway between TF and IGF-1R after FVIIa treatment with PC3 and DU145 prostate or MDA-MB-231 breast cancer cells as model systems. Protein interactions, levels, and phosphorylations were assessed by proximity ligation assay or flow cytometry in intact cells and by western blot on cell lysates. The transactivation of the IGF-1R was found dependent on TF/FVIIa-induced activation of β1-integrins. A series of experiments led to the conclusion that the caveolae protein caveolin-1 prevented IGF-1R activation in resting cells via its scaffolding domain. TF/FVIIa/β1-integrins terminated this inhibition by activation of Src family kinases and subsequent phosphorylation of caveolin-1 on tyrosine 14. This phosphorylation was not seen after treatment with PAR1 or PAR2 agonists. Consequently, the protective effect of FVIIa against apoptosis induced by the death receptor agonist TRAIL and the de novo synthesis of cyclin D1 induced by nuclear IGF-1R accumulation were both significantly reduced by down-regulation of β1-integrins or overexpression of the caveolin-1 scaffolding domain. In conclusion, we present a plausible mechanism for the interplay between TF and IGF-1R involving FVIIa, β1-integrins, Src family proteins, and caveolin-1. Our results increase the knowledge of diseases associated with TF and IGF-1R overexpression in general but specifically of TF-mediated signaling with focus on cell survival.
Collapse
Affiliation(s)
- Mikael Åberg
- Department of Medical Sciences, Clinical Chemistry and Science for Life Laboratory, Uppsala University Hospital, Entr. 61 3rd floor, 751 85, Uppsala, Sweden.
| | - Desirée Edén
- Department of Medical Sciences, Clinical Chemistry and Science for Life Laboratory, Uppsala University Hospital, Entr. 61 3rd floor, 751 85, Uppsala, Sweden
| | - Agneta Siegbahn
- Department of Medical Sciences, Clinical Chemistry and Science for Life Laboratory, Uppsala University Hospital, Entr. 61 3rd floor, 751 85, Uppsala, Sweden
| |
Collapse
|
11
|
Jain U, Saxena K, Chauhan N. Helicobacter pylori induced reactive oxygen Species: A new and developing platform for detection. Helicobacter 2021; 26:e12796. [PMID: 33666321 DOI: 10.1111/hel.12796] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 01/11/2021] [Accepted: 02/10/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND Gastric cancer is the third leading cause of cancer-related deaths worldwide. Approximately 70% of cases are caused by a microaerophilic gram-negative bacteria, Helicobacter pylori (H. pylori), which potentially infect almost 50% of world's population. H. pylori is mainly responsible for persistent oxidative stress in stomach and induction of chronic immune responses which ultimately result into DNA damage that eventually can lead to gastric cancer. Oxidative stress is the result of excessive release of ROS/RNS by activated neutrophils whereas bacteria itself also produce ROS in host cells. Therefore, ROS detection is an important factor for development of new strategies related to identification of H. pylori infection. METHODS The review summarizes the various available techniques for ROS detection with their advantages, disadvantages, and limitations. All of the information included in this review have been retrieved from published studies on ROS generation and its detection methods. RESULTS Precisely, 71 articles have been incorporated and evaluated for this review. The studied articles were divided into two major categories including articles on H. pylori-related pathogenesis and various ROS detection methods for example probe-based methods, immunoassays, gene expression profiling, and other techniques. The major part of probe activity is based on fluorescence, chemiluminescence, or bioluminescence and detected by complementary techniques such as LC-MS, HPLC, EPR, and redox blotting. CONCLUSION The review describes the methods for ROS detection but due to some limitations in conventional methods, there is a need of cost-effective, early and fast detection methods like biosensors to diagnose the infection at its initial stage.
Collapse
Affiliation(s)
- Utkarsh Jain
- Amity Institute of Nanotechnology (AINT), Amity University Uttar Pradesh, Noida, India
| | - Kirti Saxena
- Amity Institute of Nanotechnology (AINT), Amity University Uttar Pradesh, Noida, India
| | - Nidhi Chauhan
- Amity Institute of Nanotechnology (AINT), Amity University Uttar Pradesh, Noida, India
| |
Collapse
|
12
|
Vo DK, Engler A, Stoimenovski D, Hartig R, Kaehne T, Kalinski T, Naumann M, Haybaeck J, Nass N. Interactome Mapping of eIF3A in a Colon Cancer and an Immortalized Embryonic Cell Line Using Proximity-Dependent Biotin Identification. Cancers (Basel) 2021; 13:cancers13061293. [PMID: 33799492 PMCID: PMC7999522 DOI: 10.3390/cancers13061293] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 03/08/2021] [Accepted: 03/11/2021] [Indexed: 12/12/2022] Open
Abstract
Translation initiation comprises complex interactions of eukaryotic initiation factor (eIF) subunits and the structural elements of the mRNAs. Translation initiation is a key process for building the cell's proteome. It not only determines the total amount of protein synthesized but also controls the translation efficiency for individual transcripts, which is important for cancer or ageing. Thus, understanding protein interactions during translation initiation is one key that contributes to understanding how the eIF subunit composition influences translation or other pathways not yet attributed to eIFs. We applied the BioID technique to two rapidly dividing cell lines (the immortalized embryonic cell line HEK-293T and the colon carcinoma cell line HCT-166) in order to identify interacting proteins of eIF3A, a core subunit of the eukaryotic initiation factor 3 complex. We identified a total of 84 interacting proteins, with very few proteins being specific to one cell line. When protein biosynthesis was blocked by thapsigargin-induced endoplasmic reticulum (ER) stress, the interacting proteins were considerably smaller in number. In terms of gene ontology, although eIF3A interactors are mainly part of the translation machinery, protein folding and RNA binding were also found. Cells suffering from ER-stress show a few remaining interactors which are mainly ribosomal proteins or involved in RNA-binding.
Collapse
Affiliation(s)
- Diep-Khanh Vo
- Department of Pathology, Medical Faculty, Otto-von-Guericke University Magdeburg, D-39120 Magdeburg, Germany; (D.-K.V.); (D.S.); (T.K.); (J.H.)
| | - Alexander Engler
- Institute of Experimental Internal Medicine, Medical Faculty, Otto von Guericke University, D-39120 Magdeburg, Germany; (A.E.); (T.K.); (M.N.)
| | - Darko Stoimenovski
- Department of Pathology, Medical Faculty, Otto-von-Guericke University Magdeburg, D-39120 Magdeburg, Germany; (D.-K.V.); (D.S.); (T.K.); (J.H.)
| | - Roland Hartig
- Institute of Molecular and Clinical Immunology, Otto von Guericke University Magdeburg, Leipziger Str. 44, D-39120 Magdeburg, Germany;
| | - Thilo Kaehne
- Institute of Experimental Internal Medicine, Medical Faculty, Otto von Guericke University, D-39120 Magdeburg, Germany; (A.E.); (T.K.); (M.N.)
| | - Thomas Kalinski
- Department of Pathology, Medical Faculty, Otto-von-Guericke University Magdeburg, D-39120 Magdeburg, Germany; (D.-K.V.); (D.S.); (T.K.); (J.H.)
| | - Michael Naumann
- Institute of Experimental Internal Medicine, Medical Faculty, Otto von Guericke University, D-39120 Magdeburg, Germany; (A.E.); (T.K.); (M.N.)
| | - Johannes Haybaeck
- Department of Pathology, Medical Faculty, Otto-von-Guericke University Magdeburg, D-39120 Magdeburg, Germany; (D.-K.V.); (D.S.); (T.K.); (J.H.)
- Department of Pathology, Neuropathology, and Molecular Pathology, Medical University of Innsbruck, A-6020 Innsbruck, Austria
- Department of Pathology, Diagnostic & Research Center for Molecular BioMedicine, Institute of Pathology, Medical University of Graz, A-8010 Graz, Austria
- Center for Biomarker Research in Medicine, A-8010 Graz, Austria
| | - Norbert Nass
- Department of Pathology, Medical Faculty, Otto-von-Guericke University Magdeburg, D-39120 Magdeburg, Germany; (D.-K.V.); (D.S.); (T.K.); (J.H.)
- Correspondence:
| |
Collapse
|
13
|
Swidergall M, Solis NV, Millet N, Huang MY, Lin J, Phan QT, Lazarus MD, Wang Z, Yeaman MR, Mitchell AP, Filler SG. Activation of EphA2-EGFR signaling in oral epithelial cells by Candida albicans virulence factors. PLoS Pathog 2021; 17:e1009221. [PMID: 33471869 PMCID: PMC7850503 DOI: 10.1371/journal.ppat.1009221] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 02/01/2021] [Accepted: 12/05/2020] [Indexed: 12/12/2022] Open
Abstract
During oropharyngeal candidiasis (OPC), Candida albicans invades and damages oral epithelial cells, which respond by producing proinflammatory mediators that recruit phagocytes to foci of infection. The ephrin type-A receptor 2 (EphA2) detects β-glucan and plays a central role in stimulating epithelial cells to release proinflammatory mediators during OPC. The epidermal growth factor receptor (EGFR) also interacts with C. albicans and is known to be activated by the Als3 adhesin/invasin and the candidalysin pore-forming toxin. Here, we investigated the interactions among EphA2, EGFR, Als3 and candidalysin during OPC. We found that EGFR and EphA2 constitutively associate with each other as part of a heteromeric physical complex and are mutually dependent for C. albicans-induced activation. Als3-mediated endocytosis of a C. albicans hypha leads to the formation of an endocytic vacuole where candidalysin accumulates at high concentration. Thus, Als3 potentiates targeting of candidalysin, and both Als3 and candidalysin are required for C. albicans to cause maximal damage to oral epithelial cells, sustain activation of EphA2 and EGFR, and stimulate pro-inflammatory cytokine and chemokine secretion. In the mouse model of OPC, C. albicans-induced production of CXCL1/KC and CCL20 is dependent on the presence of candidalysin and EGFR, but independent of Als3. The production of IL-1α and IL-17A also requires candidalysin but is independent of Als3 and EGFR. The production of TNFα requires Als1, Als3, and candidalysin. Collectively, these results delineate the complex interplay among host cell receptors EphA2 and EGFR and C. albicans virulence factors Als1, Als3 and candidalysin during the induction of OPC and the resulting oral inflammatory response. Oropharyngeal candidiasis occurs when the fungus Candida albicans proliferates in the mouth to a point at which tissue damage occurs. The disease is characterized by fungal invasion of the superficial epithelium and a localized inflammatory response. Two C. albicans virulence factors contribute to the pathogenesis of OPC, Als3 which enables the organism to adhere to and invade host cells, and candidalysin which is a pore-forming toxin that damages host cells. Two epithelial cell receptors, ephrin type-A receptor 2 (EphA2) and the epidermal growth factor receptor (EGFR) are activated by C. albicans. Here, we show that EphA2 and EGFR form part of complex wherein these co-receptors are required to activate each other. Als3 enhances the host cell targeting of candidalysin by stimulating epithelial cell endocytosis of C. albicans, leading to the formation of an endocytic vacuole in which candidalysin accumulates. Thus, Als3 and candidalysin synergize to damage epithelial cells, activate EphA2 and EGFR, and stimulate the production of inflammatory mediators. In the mouse model of OPC, candidalysin elicits of a subset of the oral inflammatory response molecular repertoire. Of the cytokines and chemokines induced by this toxin, some require the activation of EGFR while others are induced independently of EGFR. Collectively, this work provides a deeper understanding of the interactions among C. albicans virulence factors, host cell receptors and immune responses during OPC.
Collapse
Affiliation(s)
- Marc Swidergall
- Division of Infectious Diseases, Harbor-UCLA Medical Center, Torrance, California, United States of America
- Institute for Infection and Immunity, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California, United States of America
- David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
- * E-mail: (MS); (SGF)
| | - Norma V. Solis
- Division of Infectious Diseases, Harbor-UCLA Medical Center, Torrance, California, United States of America
- Institute for Infection and Immunity, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California, United States of America
| | - Nicolas Millet
- Division of Infectious Diseases, Harbor-UCLA Medical Center, Torrance, California, United States of America
- Institute for Infection and Immunity, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California, United States of America
| | - Manning Y. Huang
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States of America
| | - Jianfeng Lin
- Division of Infectious Diseases, Harbor-UCLA Medical Center, Torrance, California, United States of America
- Institute for Infection and Immunity, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California, United States of America
| | - Quynh T. Phan
- Division of Infectious Diseases, Harbor-UCLA Medical Center, Torrance, California, United States of America
- Institute for Infection and Immunity, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California, United States of America
| | - Michael D. Lazarus
- Division of Infectious Diseases, Harbor-UCLA Medical Center, Torrance, California, United States of America
| | - Zeping Wang
- Division of Infectious Diseases, Harbor-UCLA Medical Center, Torrance, California, United States of America
| | - Michael R. Yeaman
- Division of Infectious Diseases, Harbor-UCLA Medical Center, Torrance, California, United States of America
- Institute for Infection and Immunity, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California, United States of America
- David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
- Division of Molecular Medicine, Harbor-UCLA Medical Center, Torrance, California, United States of America
| | - Aaron P. Mitchell
- Department of Microbiology, University of Georgia, Athens, Georgia, United States of America
| | - Scott G. Filler
- Division of Infectious Diseases, Harbor-UCLA Medical Center, Torrance, California, United States of America
- Institute for Infection and Immunity, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California, United States of America
- David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
- * E-mail: (MS); (SGF)
| |
Collapse
|
14
|
Evans HT, Blackmore D, Götz J, Bodea LG. De novo proteomic methods for examining the molecular mechanisms underpinning long-term memory. Brain Res Bull 2021; 169:94-103. [PMID: 33465403 DOI: 10.1016/j.brainresbull.2020.12.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 12/07/2020] [Accepted: 12/21/2020] [Indexed: 01/19/2023]
Abstract
Memory formation is a fundamental function of the nervous system that enables the experience-based adaptation of behaviour. The formation, recall and updating of long-term memory (LTM) requires new protein synthesis through its direct involvement in neuronal processes, such as long-term potentiation (LTP), long-term depression (LTD) and synaptic scaling. We discuss the advantages and limitations of several emerging techniques which enable the tagging of newly synthesised proteins, including stable isotope labelling with amino acids in cell culture (SILAC), puromycin labelling, and non-canonical amino acid (NCAA) labelling. We further present how these methods allow for the identification and visualisation of proteins which are newly synthesised during different stages of memory formation. These emerging techniques will continue to expand our understanding of how memories are formed, consolidated and retrieved.
Collapse
Affiliation(s)
- Harrison Tudor Evans
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane QLD 4072, Australia
| | - Daniel Blackmore
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane QLD 4072, Australia
| | - Jürgen Götz
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane QLD 4072, Australia.
| | - Liviu-Gabriel Bodea
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane QLD 4072, Australia.
| |
Collapse
|
15
|
A Novel Mechanism Underlying Antiviral Activity of an Influenza Virus M2-Specific Antibody. J Virol 2020; 95:JVI.01277-20. [PMID: 33055251 DOI: 10.1128/jvi.01277-20] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 10/08/2020] [Indexed: 11/20/2022] Open
Abstract
Protective immunity against influenza A viruses (IAVs) generally depends on antibodies to the major envelope glycoprotein, hemagglutinin (HA), whose antigenicity is distinctive among IAV subtypes. On the other hand, the matrix 2 (M2) protein is antigenically highly conserved and has been studied as an attractive vaccine antigen to confer cross-protective immunity against multiple subtypes of IAVs. However, antiviral mechanisms of M2-specific antibodies are not fully understood. Here, we report the molecular basis of antiviral activity of an M2-specific monoclonal antibody (MAb), rM2ss23. We first found that rM2ss23 inhibited A/Aichi/2/1968 (H3N2) (Aichi) but not A/PR/8/1934 (H1N1) (PR8) replication. rM2ss23 altered the cell surface distribution of M2, likely by cross-linking the molecules, and interfered with the colocalization of HA and M2, resulting in reduced budding of progeny viruses. However, these effects were not observed for another strain, PR8, despite the binding capacity of rM2ss23 to PR8 M2. Interestingly, HA was also involved in the resistance of PR8 to rM2ss23. We also found that two amino acid residues at positions 54 and 57 in the M2 cytoplasmic tail were critical for the insensitivity of PR8 to rM2ss2. These findings suggest that the disruption of the M2-HA colocalization on infected cells and subsequent reduction of virus budding is one of the principal mechanisms of antiviral activity of M2-specific antibodies and that anti-M2 antibody-sensitive and -resistant IAVs have different properties in the interaction between M2 and HA.IMPORTANCE Although the IAV HA is the major target of neutralizing antibodies, most of the antibodies are HA subtype specific, restricting the potential of HA-based vaccines. On the contrary, the IAV M2 protein has been studied as a vaccine antigen to confer cross-protective immunity against IAVs with multiple HA subtypes, since M2 is antigenically conserved. Although a number of studies highlight the protective role of anti-HA neutralizing and nonneutralizing antibodies, precise information on the molecular mechanism of action of M2-specific antibodies is still obscure. In this study, we found that an anti-M2 antibody interfered with the HA-M2 association, which is important for efficient budding of progeny virus particles from infected cells. The antiviral activity was IAV strain dependent despite the similar binding capacity of the antibody to M2, and, interestingly, HA was involved in susceptibility to the antibody. Our data provide a novel mechanism underlying antiviral activity of M2-specific antibodies.
Collapse
|
16
|
Kim U, Kim N, Shin HY. Modeling Non-Alcoholic Fatty Liver Disease (NAFLD) Using "Good-Fit" Genome-Editing Tools. Cells 2020; 9:cells9122572. [PMID: 33271878 PMCID: PMC7760008 DOI: 10.3390/cells9122572] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 11/27/2020] [Accepted: 11/30/2020] [Indexed: 12/16/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD), which affects both adults and children, is the most common liver disorder worldwide. NAFLD is characterized by excess fat accumulation in the liver in the absence of significant alcohol use. NAFLD is strongly associated with obesity, insulin resistance, metabolic syndrome, as well as specific genetic polymorphisms. Severe NAFLD cases can further progress to cirrhosis, hepatocellular carcinoma (HCC), or cardiovascular complications. Here, we describe the pathophysiological features and critical genetic variants associated with NAFLD. Recent advances in genome-engineering technology have provided a new opportunity to generate in vitro and in vivo models that reflect the genetic abnormalities of NAFLD. We review the currently developed NAFLD models generated using clustered regularly interspaced short palindromic repeats/CRISPR-associated protein 9 (CRISPR/Cas9) genome editing. We further discuss unique features of CRISPR/Cas9 and Cas9 variants, including base editors and prime editor, that are useful for replicating genetic features specific to NAFLD. We also compare advantages and limitations of currently available methods for delivering genome-editing tools necessary for optimal genome editing. This review should provide helpful guidance for selecting “good fit” genome-editing tools and appropriate gene-delivery methods for the successful development of NAFLD models and clinical therapeutics.
Collapse
|
17
|
Ikari S, Lu SL, Hao F, Imai K, Araki Y, Yamamoto YH, Tsai CY, Nishiyama Y, Shitan N, Yoshimori T, Otomo T, Noda T. Starvation-induced autophagy via calcium-dependent TFEB dephosphorylation is suppressed by Shigyakusan. PLoS One 2020; 15:e0230156. [PMID: 32134989 PMCID: PMC7058311 DOI: 10.1371/journal.pone.0230156] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 02/22/2020] [Indexed: 12/11/2022] Open
Abstract
Kampo, a system of traditional Japanese therapy utilizing mixtures of herbal medicine, is widely accepted in the Japanese medical system. Kampo originated from traditional Chinese medicine, and was gradually adopted into a Japanese style. Although its effects on a variety of diseases are appreciated, the underlying mechanisms remain mostly unclear. Using a quantitative tf-LC3 system, we conducted a high-throughput screen of 128 kinds of Kampo to evaluate the effects on autophagy. The results revealed a suppressive effect of Shigyakusan/TJ-35 on autophagic activity. TJ-35 specifically suppressed dephosphorylation of ULK1 and TFEB, among several TORC1 substrates, in response to nutrient deprivation. TFEB was dephosphorylated by calcineurin in a Ca2+ dependent manner. Cytosolic Ca2+ concentration was increased in response to nutrient starvation, and TJ-35 suppressed this increase. Thus, TJ-35 prevents the starvation-induced Ca2+ increase, thereby suppressing induction of autophagy.
Collapse
Affiliation(s)
- Sumiko Ikari
- Center for Frontier Oral Science, Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka, Japan
| | - Shiou-Ling Lu
- Center for Frontier Oral Science, Graduate School of Dentistry, Osaka University, Suita, Osaka, Japan
| | - Feike Hao
- Center for Frontier Oral Science, Graduate School of Dentistry, Osaka University, Suita, Osaka, Japan
- China National Research Institute of Food and Fermentation Industries, Beijing, China
| | - Kenta Imai
- Department of Intracellular Membrane Dynamics, Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka, Japan
| | - Yasuhiro Araki
- Center for Frontier Oral Science, Graduate School of Dentistry, Osaka University, Suita, Osaka, Japan
| | - Yo-hei Yamamoto
- Center for Frontier Oral Science, Graduate School of Dentistry, Osaka University, Suita, Osaka, Japan
| | - Chao-Yuan Tsai
- Laboratory of Immune Regulation, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
| | - Yumi Nishiyama
- Medicinal Botanical Garden, Kobe Pharmaceutical University, Kobe, Hyogo, Japan
| | - Nobukazu Shitan
- Laboratory of Medicinal Cell Biology, Kobe Pharmaceutical University, Kobe, Hyogo, Japan
| | - Tamotsu Yoshimori
- Department of Intracellular Membrane Dynamics, Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka, Japan
| | - Takanobu Otomo
- Department of Molecular and Genetic Medicine, Kawasaki Medical School, Kurashiki, Okayama, Japan
| | - Takeshi Noda
- Center for Frontier Oral Science, Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka, Japan
- Center for Frontier Oral Science, Graduate School of Dentistry, Osaka University, Suita, Osaka, Japan
- * E-mail:
| |
Collapse
|
18
|
Yu X, Martella A, Kolovos P, Stevens M, Stadhouders R, Grosveld FG, Andrieu-Soler C. The dynamic emergence of GATA1 complexes identified in in vitro embryonic stem cell differentiation and in vivo mouse fetal liver. Haematologica 2019; 105:1802-1812. [PMID: 31582556 PMCID: PMC7327653 DOI: 10.3324/haematol.2019.216010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 10/03/2019] [Indexed: 01/20/2023] Open
Abstract
GATA1 is an essential transcriptional regulator of myeloid hematopoietic differentiation towards red blood cells. During erythroid differentiation, GATA1 forms different complexes with other transcription factors such as LDB1, TAL1, E2A and LMO2 ("the LDB1 complex") or with FOG1. The functions of GATA1 complexes have been studied extensively in definitive erythroid differentiation; however, the temporal and spatial formation of these complexes during erythroid development is unknown. We applied proximity ligation assay (PLA) to detect, localize and quantify individual interactions during embryonic stem cell differentiation and in mouse fetal liver (FL) tissue. We show that GATA1/LDB1 interactions appear before the proerythroblast stage and increase in a subset of the CD71+/TER119- cells to activate the terminal erythroid differentiation program in 12.5 day FL. Using Ldb1 and Gata1 knockdown FL cells, we studied the functional contribution of the GATA1/LDB1 complex during differentiation. This shows that the active LDB1 complex appears quite late at the proerythroblast stage of differentiation and confirms the power of PLA in studying the dynamic interaction of proteins in cell differentiation at the single cell level. We provide dynamic insight into the temporal and spatial formation of the GATA1 and LDB1 transcription factor complexes during hematopoietic development and differentiation.
Collapse
Affiliation(s)
- Xiao Yu
- Department of Cell Biology, ErasmusMC, Rotterdam, the Netherlands.,Current address: Department of Medical Microbiology, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Andrea Martella
- Department of Cell Biology, ErasmusMC, Rotterdam, the Netherlands.,AstraZeneca, R&D Innovative Medicines, Cambridge Science Park, Milton Road, Cambridge, UK
| | - Petros Kolovos
- Department of Cell Biology, ErasmusMC, Rotterdam, the Netherlands.,Biotech Research & Innovation Centre, University of Copenhagen, Copenhagen, Denmark
| | - Mary Stevens
- Department of Cell Biology, ErasmusMC, Rotterdam, the Netherlands
| | - Ralph Stadhouders
- Department of Cell Biology, ErasmusMC, Rotterdam, the Netherlands.,Department of Pulmonary Medicine, Erasmus MC, Rotterdam, the Netherlands
| | - Frank G Grosveld
- Department of Cell Biology, ErasmusMC, Rotterdam, the Netherlands
| | - Charlotte Andrieu-Soler
- Department of Cell Biology, ErasmusMC, Rotterdam, the Netherlands .,Institut de Génétique Moléculaire Montpellier, Université de Montpellier, CNRS, Montpellier, France.,Université de Paris, Laboratoire d'excellence (LabEx) du globule rouge GR-Ex, Paris, France
| |
Collapse
|
19
|
Zhang BK, Moran AM, Bailey CG, Rasko JEJ, Holst J, Wang Q. EGF-activated PI3K/Akt signalling coordinates leucine uptake by regulating LAT3 expression in prostate cancer. Cell Commun Signal 2019; 17:83. [PMID: 31345230 PMCID: PMC6659227 DOI: 10.1186/s12964-019-0400-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 07/16/2019] [Indexed: 02/06/2023] Open
Abstract
Background Growth factors, such as EGF, activate the PI3K/Akt/mTORC1 signalling pathway, which regulates a distinct program of protein synthesis leading to cell growth. This pathway relies on mTORC1 sensing sufficient levels of intracellular amino acids, such as leucine, which are required for mTORC1 activation. However, it is currently unknown whether there is a direct link between these external growth signals and intracellular amino acid levels. In primary prostate cancer cells, intracellular leucine levels are regulated by L-type amino acid transporter 3 (LAT3/SLC43A1), and we therefore investigated whether LAT3 is regulated by growth factor signalling. Methods To investigate how PI3K/Akt signalling regulates leucine transport, prostate cancer cells were treated with different PI3K/Akt inhibitors, or stable knock down of LAT3 by shRNA, followed by analysis of leucine uptake, western blotting, immunofluorescent staining and proximity ligation assay. Results Inhibition of PI3K/Akt signalling significantly reduced leucine transport in LNCaP and PC-3 human prostate cancer cell lines, while growth factor addition significantly increased leucine uptake. These effects appeared to be mediated by LAT3 transport, as LAT3 knockdown blocked leucine uptake, and was not rescued by growth factor activation or further inhibited by signalling pathway inhibition. We further demonstrated that EGF significantly increased LAT3 protein levels when Akt was phosphorylated, and that Akt and LAT3 co-localised on the plasma membrane in EGF-activated LNCaP cells. These effects were likely due to stabilisation of LAT3 protein levels on the plasma membrane, with EGF treatment preventing ubiquitin-mediated LAT3 degradation. Conclusion Growth factor-activated PI3K/Akt signalling pathway regulates leucine transport through LAT3 in prostate cancer cell lines. These data support a direct link between growth factor and amino acid uptake, providing a mechanism by which the cells rapidly coordinate amino acid uptake for cell growth. Electronic supplementary material The online version of this article (10.1186/s12964-019-0400-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Blake K Zhang
- Centenary Institute, University of Sydney, Camperdown, Australia.,Sydney Medical School, University of Sydney, Camperdown, Australia
| | - Anne M Moran
- Centenary Institute, University of Sydney, Camperdown, Australia.,Sydney Medical School, University of Sydney, Camperdown, Australia
| | - Charles G Bailey
- Sydney Medical School, University of Sydney, Camperdown, Australia.,Gene & Stem Cell Therapy Program Centenary Institute, University of Sydney, Camperdown, Australia
| | - John E J Rasko
- Sydney Medical School, University of Sydney, Camperdown, Australia.,Gene & Stem Cell Therapy Program Centenary Institute, University of Sydney, Camperdown, Australia.,Cell and Molecular Therapies, Royal Prince Alfred Hospital, Camperdown, Australia
| | - Jeff Holst
- Translational Cancer Metabolism Laboratory, Lowy Cancer Research Centre, School of Medical Sciences and Prince of Wales Clinical School, University of New South Wales, Sydney, Australia. .,Origins of Cancer Program Centenary Institute, University of Sydney, Camperdown, Australia.
| | - Qian Wang
- Sydney Medical School, University of Sydney, Camperdown, Australia. .,Translational Cancer Metabolism Laboratory, Lowy Cancer Research Centre, School of Medical Sciences and Prince of Wales Clinical School, University of New South Wales, Sydney, Australia.
| |
Collapse
|
20
|
Evans HT, Benetatos J, van Roijen M, Bodea L, Götz J. Decreased synthesis of ribosomal proteins in tauopathy revealed by non-canonical amino acid labelling. EMBO J 2019; 38:e101174. [PMID: 31268600 PMCID: PMC6600635 DOI: 10.15252/embj.2018101174] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [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: 04/12/2019] [Accepted: 04/17/2019] [Indexed: 01/06/2023] Open
Abstract
Tau is a scaffolding protein that serves multiple cellular functions that are perturbed in neurodegenerative diseases, including Alzheimer's disease (AD) and frontotemporal dementia (FTD). We have recently shown that amyloid-β, the second hallmark of AD, induces de novo protein synthesis of tau. Importantly, this activation was found to be tau-dependent, raising the question of whether FTD-tau by itself affects protein synthesis. We therefore applied non-canonical amino acid labelling to visualise and identify newly synthesised proteins in the K369I tau transgenic K3 mouse model of FTD. This revealed massively decreased protein synthesis in neurons containing pathologically phosphorylated tau, a finding confirmed in P301L mutant tau transgenic rTg4510 mice. Using quantitative SWATH-MS proteomics, we identified changes in 247 proteins of the de novo proteome of K3 mice. These included decreased synthesis of the ribosomal proteins RPL23, RPLP0, RPL19 and RPS16, a finding that was validated in both K3 and rTg4510 mice. Together, our findings present a potential pathomechanism by which pathological tau interferes with cellular functions through the dysregulation of ribosomal protein synthesis.
Collapse
Affiliation(s)
- Harrison Tudor Evans
- Clem Jones Centre for Ageing Dementia ResearchQueensland Brain InstituteThe University of QueenslandBrisbaneQldAustralia
| | - Joseph Benetatos
- Clem Jones Centre for Ageing Dementia ResearchQueensland Brain InstituteThe University of QueenslandBrisbaneQldAustralia
| | | | - Liviu‐Gabriel Bodea
- Clem Jones Centre for Ageing Dementia ResearchQueensland Brain InstituteThe University of QueenslandBrisbaneQldAustralia
| | - Jürgen Götz
- Clem Jones Centre for Ageing Dementia ResearchQueensland Brain InstituteThe University of QueenslandBrisbaneQldAustralia
| |
Collapse
|
21
|
Squarzanti DF, Sorrentino R, Landini MM, Chiesa A, Pinato S, Rocchio F, Mattii M, Penengo L, Azzimonti B. Human papillomavirus type 16 E6 and E7 oncoproteins interact with the nuclear p53-binding protein 1 in an in vitro reconstructed 3D epithelium: new insights for the virus-induced DNA damage response. Virol J 2018; 15:176. [PMID: 30445982 PMCID: PMC6240266 DOI: 10.1186/s12985-018-1086-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 10/22/2018] [Indexed: 12/28/2022] Open
Abstract
Background Despite vaccination and screening measures, anogenital cancer, mainly promoted by HPV16 oncoproteins, still represents the fourth tumor and the second cause of death among women. Cell replication fidelity is the result of the host DNA damage response (DDR). Unlike many DNA viruses that promote their life cycle through the DDR inactivation, HR-HPVs encourage cells proliferation despite the DDR turned on. Why and how it occurs has been only partially elucidated. During HPV16 infection, E6 links and degrades p53 via the binding to the E6AP LXXLL sequence; unfortunately, E6 direct role in the DDR response has not clearly identified yet. Similarly, E7 increases DDR by competing with E2F1-pRb interaction, thus leading to the inactivation of pRb, and promotion, E2F1 mediated, of DDR genes translation, by binding to the pRb-like proteins CBP/p300 and p107, that also harbour LXXLL sequence, and via the interaction and activation of several DDR proteins. Methods To gain information regarding E6 and E7 contribution in DDR activation, we produced an in vitro 3D HPV16-E6E7 infected epithelium, already consolidated study model for HPVs, and validated it by assessing H&E staining and BrdU, HPV16 DNA, E6E7 proteins and γH2A.X/53BP1 double-strand break (DSBs) sensors expression; then we made an immuno-colocalization of E6 and E7 with cyclin E2 and B1. Since 53BP1, like E6 and E7, also binds p53 and pRb, we supposed their possible direct binding. To explore this hypothesis, we performed a double immunofluorescence of E6 and E7 with 53BP1, a sequence analysis of 53BP1 within its BRCT2 domain and then an in situ PLA within CaSki, E6E7HPV16 NHEKs and the 3D model. Results The in vitro epithelium resembled the histology and the events typical of in vivo infected tissues. E6E7HPV16 were both expressed in basal and differentiated strata and induced H2A.X phosphorylation and 53BP1 increment into nuclear foci. After highlighting E6 and E7 co-expression with 53BP1 and a LKVLL sequence within the 53BP1 BRCT2 domain, we demonstrated the bindings via the PLA technique. Conclusions Our results reinforce E6 and E7 role in cellular function control providing potentially new insights into the activity of this tumor virus.
Collapse
Affiliation(s)
- Diletta Francesca Squarzanti
- Laboratory of applied Microbiology, Department of Health Sciences, University of Piemonte Orientale (UPO), Via Solaroli 17, 28100, Novara, Italy
| | - Rita Sorrentino
- Laboratory of Biomedical Materials, Department of Health Sciences, University of Piemonte Orientale (UPO), Novara, Italy
| | - Manuela Miriam Landini
- Laboratory of applied Microbiology, Department of Health Sciences, University of Piemonte Orientale (UPO), Via Solaroli 17, 28100, Novara, Italy
| | - Andrea Chiesa
- Laboratory of applied Microbiology, Department of Health Sciences, University of Piemonte Orientale (UPO), Via Solaroli 17, 28100, Novara, Italy
| | - Sabrina Pinato
- Laboratory of Molecular Biology, Department of Pharmaceutical Sciences, University of Piemonte Orientale (UPO), Novara, Italy
| | - Francesca Rocchio
- Laboratory of Molecular Biology, Department of Pharmaceutical Sciences, University of Piemonte Orientale (UPO), Novara, Italy
| | - Martina Mattii
- Laboratory of applied Microbiology, Department of Health Sciences, University of Piemonte Orientale (UPO), Via Solaroli 17, 28100, Novara, Italy
| | - Lorenza Penengo
- Institute of Molecular Cancer Research, University of Zurich, Zurich, Switzerland
| | - Barbara Azzimonti
- Laboratory of applied Microbiology, Department of Health Sciences, University of Piemonte Orientale (UPO), Via Solaroli 17, 28100, Novara, Italy. .,Consorzio Interuniversitario Nazionale per la Scienza e Tecnologia dei Materiali (INSTM, Firenze, Italy- Local Unit of Piemonte Orientale, UPO, Novara, Italy.
| |
Collapse
|
22
|
Luff SA, Kao CY, Papoutsakis ET. Role of p53 and transcription-independent p53-induced apoptosis in shear-stimulated megakaryocytic maturation, particle generation, and platelet biogenesis. PLoS One 2018; 13:e0203991. [PMID: 30231080 PMCID: PMC6145578 DOI: 10.1371/journal.pone.0203991] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 09/02/2018] [Indexed: 12/18/2022] Open
Abstract
Megakaryocytes (Mks) derive from hematopoietic stem and progenitor cells (HSPCs) in the bone marrow and develop into large, polyploid cells that eventually give rise to platelets. As Mks mature, they migrate from the bone marrow niche into the vasculature, where they are exposed to shear forces from blood flow, releasing Mk particles (platelet-like particles (PLPs), pro/preplatelets (PPTs), and Mk microparticles (MkMPs)) into circulation. We have previously shown that transcription factor p53 is important in Mk maturation, and that physiological levels of shear promote Mk particle generation and platelet biogenesis. Here we examine the role of p53 in the Mk shear-stress response. We show that p53 is acetylated in response to shear in both immature and mature Mks, and that decreased expression of deacetylase HDAC1, and increased expression of the acetyltransferases p300 and PCAF might be responsible for these changes. We also examined the hypothesis that p53 might be involved in the shear-induced Caspase 3 activation, phosphatidylserine (PS) externalization, and increased biogenesis of PLPs, PPTs, and MkMPs. We show that p53 is involved in all these shear-induced processes. We show that in response to shear, acetyl-p53 binds Bax, cytochrome c is released from mitochondria, and Caspase 9 is activated. We also show that shear-stimulated Caspase 9 activation and Mk particle biogenesis depend on transcription-independent p53-induced apoptosis (TIPA), but PS externalization is not. This is the first report to show that shear flow stimulates TIPA and that Caspase 9 activation and Mk-particle biogenesis are directly modulated by TIPA.
Collapse
Affiliation(s)
- Stephanie A. Luff
- Department of Biological Sciences, University of Delaware, Newark, Delaware, United States of America
- Delaware Biotechnology Institute, University of Delaware, Newark, Delaware, United States of America
| | - Chen-Yuan Kao
- Delaware Biotechnology Institute, University of Delaware, Newark, Delaware, United States of America
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, Delaware, United States of America
| | - Eleftherios T. Papoutsakis
- Department of Biological Sciences, University of Delaware, Newark, Delaware, United States of America
- Delaware Biotechnology Institute, University of Delaware, Newark, Delaware, United States of America
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, Delaware, United States of America
| |
Collapse
|
23
|
Xue L, Lu X, He J, Zhang T, Wu X, Zhang Y, Wang N, An Z, Xu J, Geng Y. Serum CK 18-M30 reflect liver pathological severity during NAFLD progression in a rat model. Pathol Res Pract 2018; 214:1778-1786. [PMID: 30149902 DOI: 10.1016/j.prp.2018.08.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 08/04/2018] [Accepted: 08/18/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND CK 18-M30 was increased in patients with NAFLD. However, little is known about the relationship between CK 18-M30 and NAFLD progression. We aimed to analyze the variety of CK 18-M30 and other metabolism indices during NAFLD progression. Meanwhile, we aimed to investigate the correlation between CK 18-M30 and liver pathology during NAFLD progression. MATERIALS AND METHODS Rats were fed with high sucrose and high fat diet for building NAFLD models. We detected liver pathology by hematoxylin-eosin (HE) staining. We also detected serum CK 18-M30 and metabolism indices including liver enzymes, serum lipids and glycometabolism indices. RESULTS The aggravating degree of liver pathology appeared with prolonged feeding period. The relevance of CK 18-M30 to the severity of liver pathology were higher relative to other indices. CONCLUSION Our results suggested the significance of CK 18-M30 in the progression of NAFLD and provided new evidence for the early diagnosis and prognostic estimation of NAFLD.
Collapse
Affiliation(s)
- Li Xue
- Department of Laboratory, The Second Affiliated Hospital of Medical College of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, PR China
| | - Xiaolan Lu
- Department of Gastroenterology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, PR China
| | - Juntao He
- Department of Laboratory, The Second Affiliated Hospital of Medical College of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, PR China
| | - Ting Zhang
- Department of Laboratory, Wuxi Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu 214002, PR China.
| | - Xiaokang Wu
- Department of Laboratory, The Second Affiliated Hospital of Medical College of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, PR China
| | - Yanping Zhang
- Department of Laboratory, The Second Affiliated Hospital of Medical College of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, PR China
| | - Ningning Wang
- Department of Infection Control, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, PR China.
| | - Zhe An
- Department of Laboratory, The Second Affiliated Hospital of Medical College of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, PR China
| | - Jiru Xu
- Department of Immunology and Pathogenic Biology, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China.
| | - Yan Geng
- Department of Laboratory, The Second Affiliated Hospital of Medical College of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, PR China.
| |
Collapse
|
24
|
Yan J, Zhang H, Xiang J, Zhao Y, Yuan X, Sun B, Lin A. The BH3-only protein BAD mediates TNFα cytotoxicity despite concurrent activation of IKK and NF-κB in septic shock. Cell Res 2018; 28:701-718. [PMID: 29795446 PMCID: PMC6028455 DOI: 10.1038/s41422-018-0041-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2018] [Revised: 03/27/2018] [Accepted: 04/18/2018] [Indexed: 12/13/2022] Open
Abstract
The inflammatory cytokine TNFα plays a crucial role in the pathology of many inflammatory and infectious diseases. However, the mechanism underlying TNFα cytotoxicity in these diseases is incompletely understood. Here we report that the pro-apoptotic BCL-2 family member BAD mediates TNFα cytotoxicity despite concurrent activation of IKK and NF-κB in vitro by inducing apoptosis in cultured cells and in vivo by eliciting tissue damage of multiple organs and contributing to mortality in septic shock. At high doses, TNFα significantly inactivates RhoA through activation of the Src-p190GAP pathway, resulting in massive actin stress fiber destabilization, followed by substantial BAD release from the cytoskeleton to the cytosol. Under this condition, activated IKK fails to phosphorylate all cytosolic BAD, allowing translocation of non-phosphorylated BAD to mitochondria to trigger apoptosis. Polymicrobial infection utilizes the same mechanism as high-dose TNFα to elicit apoptosis-associated tissue damage of multiple organs. Consequently, loss of Bad or elimination of BAD pro-apoptotic activity protects mice from tissue damage of multiple organs and reduces mortality rates. Our results support a model in which BAD mediates TNFα cytotoxicity despite concurrent activation of the IKK-NF-κB pathway in cultured mammalian cells and in septic shock.
Collapse
Affiliation(s)
- Jie Yan
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL, 60637, USA.,The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allery & Clinical Immunology, Guangzhou Medical University, Guangzhou, Guangdong, 510260, China
| | - Hao Zhang
- The State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Jialing Xiang
- Department of Biology, Illinois Institute of Technology, Chicago, IL, 60616, USA
| | - Yu Zhao
- Department of Biology, Illinois Institute of Technology, Chicago, IL, 60616, USA
| | - Xiang Yuan
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL, 60637, USA.,The State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Beicheng Sun
- Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Anning Lin
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL, 60637, USA. .,The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allery & Clinical Immunology, Guangzhou Medical University, Guangzhou, Guangdong, 510260, China.
| |
Collapse
|
25
|
Hao F, Kondo K, Itoh T, Ikari S, Nada S, Okada M, Noda T. Rheb localized on the Golgi membrane activates lysosome-localized mTORC1 at the Golgi-lysosome contact site. J Cell Sci 2018; 131:jcs.208017. [PMID: 29222112 DOI: 10.1242/jcs.208017] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 11/26/2017] [Indexed: 12/31/2022] Open
Abstract
In response to amino acid supply, mTORC1, a master regulator of cell growth, is recruited to the lysosome and activated by the small GTPase Rheb. However, the intracellular localization of Rheb is controversial. In this study, we showed that a significant portion of Rheb is localized on the Golgi but not on the lysosome. GFP-Rheb could activate mTORC1, even when forced to exclusively localize to the Golgi. Likewise, artificial recruitment of mTORC1 to the Golgi allowed its activation. Accordingly, the Golgi was in contact with the lysosome at an newly discovered area of the cell that we term the Golgi-lysosome contact site (GLCS). The number of GLCSs increased in response to amino acid supply, whereas GLCS perturbation suppressed mTORC1 activation. These results suggest that inter-organelle communication between the Golgi and lysosome is important for mTORC1 regulation and the Golgi-localized Rheb may activate mTORC1 at GLCSs.
Collapse
Affiliation(s)
- Feike Hao
- Center for Frontier Oral Science, Graduate School of Dentistry, Osaka University, Osaka 565-0871, Japan
| | - Kazuhiko Kondo
- Graduate School of Frontier Bioscience, Osaka University, Osaka 565-0871, Japan
| | - Takashi Itoh
- Center for Frontier Oral Science, Graduate School of Dentistry, Osaka University, Osaka 565-0871, Japan
| | - Sumiko Ikari
- Graduate School of Frontier Bioscience, Osaka University, Osaka 565-0871, Japan
| | - Shigeyuki Nada
- Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
| | - Masato Okada
- Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
| | - Takeshi Noda
- Center for Frontier Oral Science, Graduate School of Dentistry, Osaka University, Osaka 565-0871, Japan .,Graduate School of Frontier Bioscience, Osaka University, Osaka 565-0871, Japan
| |
Collapse
|
26
|
Jorge-Finnigan A, Kleppe R, Jung-Kc K, Ying M, Marie M, Rios-Mondragon I, Salvatore MF, Saraste J, Martinez A. Phosphorylation at serine 31 targets tyrosine hydroxylase to vesicles for transport along microtubules. J Biol Chem 2017. [PMID: 28637871 DOI: 10.1074/jbc.m116.762344] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Tyrosine hydroxylase (TH) catalyzes the conversion of l-tyrosine into l-DOPA, which is the rate-limiting step in the synthesis of catecholamines, such as dopamine, in dopaminergergic neurons. Low dopamine levels and death of the dopaminergic neurons are hallmarks of Parkinson's disease (PD), where α-synuclein is also a key player. TH is highly regulated, notably by phosphorylation of several Ser/Thr residues in the N-terminal tail. However, the functional role of TH phosphorylation at the Ser-31 site (THSer(P)-31) remains unclear. Here, we report that THSer(P)-31 co-distributes with the Golgi complex and synaptic-like vesicles in rat and human dopaminergic cells. We also found that the TH microsomal fraction content decreases after inhibition of cyclin-dependent kinase 5 (Cdk5) and ERK1/2. The cellular distribution of an overexpressed phospho-null mutant, TH1-S31A, was restricted to the soma of neuroblastoma cells, with decreased association with the microsomal fraction, whereas a phospho-mimic mutant, TH1-S31E, was distributed throughout the soma and neurites. TH1-S31E associated with vesicular monoamine transporter 2 (VMAT2) and α-synuclein in neuroblastoma cells, and endogenous THSer(P)-31 was detected in VMAT2- and α-synuclein-immunoprecipitated mouse brain samples. Microtubule disruption or co-transfection with α-synuclein A53T, a PD-associated mutation, caused TH1-S31E accumulation in the cell soma. Our results indicate that Ser-31 phosphorylation may regulate TH subcellular localization by enabling its transport along microtubules, notably toward the projection terminals. These findings disclose a new mechanism of TH regulation by phosphorylation and reveal its interaction with key players in PD, opening up new research avenues for better understanding dopamine synthesis in physiological and pathological states.
Collapse
Affiliation(s)
- Ana Jorge-Finnigan
- From the Department of Biomedicine, University of Bergen, Jonas Lies vei 91, 5009 Bergen, Norway; K. G. Jebsen Centre for Neuropsychiatric Disorders, Jonas Lies vei 91, 5009 Bergen, Norway.
| | - Rune Kleppe
- From the Department of Biomedicine, University of Bergen, Jonas Lies vei 91, 5009 Bergen, Norway; K. G. Jebsen Centre for Neuropsychiatric Disorders, Jonas Lies vei 91, 5009 Bergen, Norway
| | - Kunwar Jung-Kc
- From the Department of Biomedicine, University of Bergen, Jonas Lies vei 91, 5009 Bergen, Norway; K. G. Jebsen Centre for Neuropsychiatric Disorders, Jonas Lies vei 91, 5009 Bergen, Norway
| | - Ming Ying
- From the Department of Biomedicine, University of Bergen, Jonas Lies vei 91, 5009 Bergen, Norway
| | - Michael Marie
- Department of Molecular Biology, University of Bergen, Thormøhlensgaten 55, 5020 Bergen Norway
| | - Ivan Rios-Mondragon
- From the Department of Biomedicine, University of Bergen, Jonas Lies vei 91, 5009 Bergen, Norway
| | - Michael F Salvatore
- Institute for Healthy Aging, University of North Texas Health Science Center, Fort Worth, Texas 76107
| | - Jaakko Saraste
- From the Department of Biomedicine, University of Bergen, Jonas Lies vei 91, 5009 Bergen, Norway
| | - Aurora Martinez
- From the Department of Biomedicine, University of Bergen, Jonas Lies vei 91, 5009 Bergen, Norway; K. G. Jebsen Centre for Neuropsychiatric Disorders, Jonas Lies vei 91, 5009 Bergen, Norway
| |
Collapse
|
27
|
Lahiri P, Schmidt V, Smole C, Kufferath I, Denk H, Strnad P, Rülicke T, Fröhlich LF, Zatloukal K. p62/Sequestosome-1 Is Indispensable for Maturation and Stabilization of Mallory-Denk Bodies. PLoS One 2016; 11:e0161083. [PMID: 27526095 PMCID: PMC4985067 DOI: 10.1371/journal.pone.0161083] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Accepted: 07/31/2016] [Indexed: 11/19/2022] Open
Abstract
Mallory-Denk bodies (MDBs) are hepatocytic protein aggregates found in steatohepatitis and several other chronic liver diseases as well as hepatocellular carcinoma. MDBs are mainly composed of phosphorylated keratins and stress protein p62/Sequestosome-1 (p62), which is a common component of cytoplasmic aggregates in a variety of protein aggregation diseases. In contrast to the well-established role of keratins, the role of p62 in MDB pathogenesis is still elusive. We have generated total and hepatocyte-specific p62 knockout mice, fed them with 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC) to induce MDBs and allowed the mice to recover from DDC intoxication on a standard diet to investigate the role of p62 in MDB formation and elimination. In the absence of p62, smaller, granular and less distinct MDBs appeared, which failed to mature to larger and compact inclusions. Moreover, p62 deficiency impaired the binding of other proteins such as NBR1 and Hsp25 to MDBs and altered the cellular defense mechanism by downregulation of Nrf2 target genes. Upon recovery from DDC intoxication on a standard diet, there was an enhanced reduction of p62-deficient MDBs, which was accompanied by a pronounced decrease in ubiquitinated proteins. Our data provide strong evidence that keratin aggregation is the initial step in MDB formation in steatohepatitis-related mouse models. Interaction of p62 with keratin aggregates then leads to maturation i.e., enlargement and stabilization of the MDBs as well as recruitment of other MDB-associated proteins.
Collapse
Affiliation(s)
- Pooja Lahiri
- Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Volker Schmidt
- Institute of Laboratory Animal Science, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Claudia Smole
- Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Iris Kufferath
- Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Helmut Denk
- Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Pavel Strnad
- IZKF and Department of Internal Medicine III, Aachen, Germany
| | - Thomas Rülicke
- Institute of Laboratory Animal Science, University of Veterinary Medicine Vienna, Vienna, Austria
| | | | - Kurt Zatloukal
- Institute of Pathology, Medical University of Graz, Graz, Austria
- * E-mail:
| |
Collapse
|
28
|
Dharan A, Talley S, Tripathi A, Mamede JI, Majetschak M, Hope TJ, Campbell EM. KIF5B and Nup358 Cooperatively Mediate the Nuclear Import of HIV-1 during Infection. PLoS Pathog 2016; 12:e1005700. [PMID: 27327622 PMCID: PMC4915687 DOI: 10.1371/journal.ppat.1005700] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 05/21/2016] [Indexed: 12/23/2022] Open
Abstract
Following envelope mediated fusion, the HIV-1 core is released into the cytoplasm of the target cell and undergoes a series of trafficking and replicative steps that result in the nuclear import of the viral genome, which ultimately leads to the integration of the proviral DNA into the host cell genome. Previous studies have found that disruption of microtubules, or depletion of dynein or kinesin motors, perturb the normal uncoating and trafficking of the viral genome. Here, we show that the Kinesin-1 motor, KIF5B, induces a relocalization of the nuclear pore component Nup358 into the cytoplasm during HIV-1 infection. This relocalization of NUP358 is dependent on HIV-1 capsid, and NUP358 directly associates with viral cores following cytoplasmic translocation. This interaction between NUP358 and the HIV-1 core is dependent on multiple capsid binding surfaces, as this association is not observed following infection with capsid mutants in which a conserved hydrophobic binding pocket (N74D) or the cyclophilin A binding loop (P90A) is disrupted. KIF5B knockdown also prevents the nuclear entry and infection by HIV-1, but does not exert a similar effect on the N74D or P90A capsid mutants which do not rely on Nup358 for nuclear import. Finally, we observe that the relocalization of Nup358 in response to CA is dependent on cleavage protein and polyadenylation factor 6 (CPSF6), but independent of cyclophilin A. Collectively, these observations identify a previously unappreciated role for KIF5B in mediating the Nup358 dependent nuclear import of the viral genome during infection. Fusion of viral and target cell membranes releases the HIV-1 viral capsid, which houses the viral RNA and proteins necessary for viral reverse transcription and integration, into the cytoplasm of target cells. To complete infection, the viral capsid must ultimately traffic to the nucleus and undergo a process known as uncoating to allow the nuclear import of the viral genome into the nucleus, where it subsequently integrates into the genome of the target cell. Here, we show that the concerted actions of microtubule motor KIF5B and the nuclear pore component Nup358 cooperatively facilitate the uncoating and nuclear import of the viral genome. Moreover, we also identify the determinants in the viral capsid protein, which forms the viral capsid core, that are required for KIF5B dependent nuclear entry. These studies reveal a novel role for the microtubule motor KIF5B in the nuclear import of the viral genome and reveal potential intervention targets for therapeutic intervention.
Collapse
Affiliation(s)
- Adarsh Dharan
- Department of Microbiology and Immunology, Stritch School of Medicine, Loyola University, Chicago, Illinois, United States of America
| | - Sarah Talley
- Integrative Cell Biology Program, Stritch School of Medicine, Loyola University, Chicago, Illinois, United States of America
| | - Abhishek Tripathi
- Burn and Shock Trauma Research Institute, Department of Surgery, Stritch School of Medicine, Loyola University, Chicago, Illinois, United States of America
| | - João I. Mamede
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Matthias Majetschak
- Burn and Shock Trauma Research Institute, Department of Surgery, Stritch School of Medicine, Loyola University, Chicago, Illinois, United States of America
| | - Thomas J. Hope
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Edward M. Campbell
- Department of Microbiology and Immunology, Stritch School of Medicine, Loyola University, Chicago, Illinois, United States of America
- Integrative Cell Biology Program, Stritch School of Medicine, Loyola University, Chicago, Illinois, United States of America
- * E-mail:
| |
Collapse
|
29
|
Mu Y, Xie H, Wan Y. Sensitive and Specific Neutrophil Gelatinase-associated Lipocalin Detection by Solid-phase Proximity Ligation Assay. ANAL SCI 2016; 31:475-9. [PMID: 26063008 DOI: 10.2116/analsci.31.475] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Neutrophil gelatinase-associated lipocalin (NGAL) is a candidate diagnostic biomarker for acute kidney injury (AKI). Since there is no specific treatment to reverse AKI, a good biomarker such as NGAL can increase the performance of clinical care. Therefore, a timely, specific and sensitive assay for detecting NGAL is critical for clinical determination. In this study, we established a solid-phase proximity ligation assay for the detection of NGAL using polyclonal antibodies conjugated with a pair of oligonucleotides. The data are read out as the Ct value via quantitative real-time polymerase chain reaction (qPCR). Our results demonstrate that this new assay performs with good specificity and sensitivity for detection of NGAL spiked in buffer or serum, which indicates that the solid-phase proximity ligation technique is a promising tool for diagnostics in clinical decisions.
Collapse
Affiliation(s)
- Yawen Mu
- The Key Laboratory of Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University
| | | | | |
Collapse
|
30
|
|
31
|
|
32
|
Examining Roles of Glycans in Chemokine-Mediated Dendritic-Endothelial Cell Interactions. Methods Enzymol 2015. [PMID: 26921954 DOI: 10.1016/bs.mie.2015.09.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Interactions between glycosaminoglycans (GAGs) and chemokines play a critical role in multiple physiological and pathological processes, including tumor metastasis and immune-cell trafficking. During our studies examining the genetic importance of the GAG subtype known as heparan sulfate (HS) on lymphatic endothelial cells (LECs), we established a repertoire of methods to assess how HS affects chemokine-mediated cell-cell interactions. In this chapter, we describe methods for monitoring migration and adhesion interactions of dendritic cells (DCs), the most potent antigen-presenting cells, with LECs. We will also report a methodology to assess chemokine-receptor interactions while incorporating approaches to target HS in the system. This includes in situ methods to visualize and quantify direct interactions between chemokines and chemokine receptors on DC surfaces, and how targeting HS produced by LECs or even DCs affects these interactions. These methods enable the mechanistic and functional characterization of GAG-chemokine interactions in cell-based studies that model physiologic interactions ex vivo. They may also be used to obtain novel insights into GAG-mediated biological processes.
Collapse
|