1
|
Hemel IMGM, Steen C, Denil SLIJ, Ertaylan G, Kutmon M, Adriaens M, Gerards M. The unusual suspect: A novel role for intermediate filament proteins in mitochondrial morphology. Mitochondrion 2025; 81:102008. [PMID: 39909388 DOI: 10.1016/j.mito.2025.102008] [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: 12/20/2024] [Revised: 01/30/2025] [Accepted: 02/01/2025] [Indexed: 02/07/2025]
Abstract
Mitochondrial dynamics is crucial for cellular homeostasis. However, not all proteins involved are known. Using a protein-protein interaction (PPI) approach, we identified ITPRIPL2 for involvement in mitochondrial dynamics. ITPRIPL2 co-localizes with intermediate filament protein vimentin, supported by protein simulations. ITPRIPL2 knockdown reveals mitochondrial elongation, disrupts vimentin processing, intermediate filament formation, and alters vimentin-related pathways. Interestingly, vimentin knockdown also leads to mitochondrial elongation. These findings highlight ITPRIPL2 as vimentin-associated protein essential for intermediate filament structure and suggest a role for intermediate filaments in mitochondrial morphology. Our study demonstrates that PPI analysis is a powerful approach for identifying novel mitochondrial dynamics proteins.
Collapse
Affiliation(s)
- Irene M G M Hemel
- Maastricht Centre for Systems Biology (MaCSBio), Maastricht University, Maastricht 6229 EN the Netherlands
| | - Carlijn Steen
- Maastricht Centre for Systems Biology (MaCSBio), Maastricht University, Maastricht 6229 EN the Netherlands
| | - Simon L I J Denil
- Flemish Institute for Technological Research (VITO) 2400 Mol, Belgium
| | - Gökhan Ertaylan
- Flemish Institute for Technological Research (VITO) 2400 Mol, Belgium
| | - Martina Kutmon
- Maastricht Centre for Systems Biology (MaCSBio), Maastricht University, Maastricht 6229 EN the Netherlands
| | - Michiel Adriaens
- Maastricht Centre for Systems Biology (MaCSBio), Maastricht University, Maastricht 6229 EN the Netherlands
| | - Mike Gerards
- Maastricht Centre for Systems Biology (MaCSBio), Maastricht University, Maastricht 6229 EN the Netherlands.
| |
Collapse
|
2
|
Yuan Z, Smith P, McCulloch CA. Extracellular vimentin amplifies inflammation: Perspectives for immune injury and therapeutics for periodontitis. FASEB J 2025; 39:e70286. [PMID: 39758044 DOI: 10.1096/fj.202402322r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 11/23/2024] [Accepted: 12/20/2024] [Indexed: 01/07/2025]
Abstract
Periodontitis is an inflammatory disease triggered by microbial biofilms that promote immune dysfunction and tissue destruction of tooth-supporting tissues. The search for soluble mediators that amplify inflammatory responses and matrix degradation in periodontal tissues has implicated extracellular vimentin (ECV) as a signaling ligand and damage-associated molecular pattern in the pathogenesis of periodontitis. Intracellular vimentin filaments are essential for the structural integrity of cells and the preservation of matrix homeostasis. These are important determinants of health in the periodontium and many other organs. But in inflamed tissues, intracellular vimentin filaments are disassembled. Vimentin is subsequently released from cells into the extracellular space in a soluble form where it drives immune signaling and tissue destruction. We discuss the role of ECV as a signaling molecule in several tissues. We apply these data to understand how in inflammatory diseases like periodontitis, ECV amplifies immune responses that contribute to disease progression. Arising from these data, we consider novel therapeutic opportunities for limiting tissue destruction by targeting ECV for treatment of inflammatory disorders like periodontitis.
Collapse
Affiliation(s)
- Zhiyao Yuan
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Research Institute of Stomatology, Nanjing University, Nanjing, China
| | - Patricio Smith
- Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | | |
Collapse
|
3
|
Kuburich NA, Kiselka JM, den Hollander P, Karam AA, Mani SA. The Cancer Chimera: Impact of Vimentin and Cytokeratin Co-Expression in Hybrid Epithelial/Mesenchymal Cancer Cells on Tumor Plasticity and Metastasis. Cancers (Basel) 2024; 16:4158. [PMID: 39766058 PMCID: PMC11674825 DOI: 10.3390/cancers16244158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 12/02/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
The epithelial-mesenchymal transition (EMT) program is critical to metastatic cancer progression. EMT results in the expression of mesenchymal proteins and enhances migratory and invasive capabilities. In a small percentage of cells, EMT results in the expression of stemness-associated genes that provide a metastatic advantage. Although EMT had been viewed as a binary event, it has recently become clear that the program leads to a spectrum of phenotypes, including hybrid epithelial/mesenchymal (E/M) cells that have significantly greater metastatic capability than cells on the epithelial or mesenchymal ends of the spectrum. As hybrid E/M cells are rarely observed in physiological, non-diseased states in the adult human body, these cells are potential biomarkers and drug targets. Hybrid E/M cells are distinguished by the co-expression of epithelial and mesenchymal proteins, such as the intermediate filament proteins cytokeratin (CK; epithelial) and vimentin (VIM; mesenchymal). Although these intermediate filaments have been extensively used for pathological characterization and detection of aggressive carcinomas, little is known regarding the interactions between CK and VIM when co-expressed in hybrid E/M cells. This review describes the characteristics of hybrid E/M cells with a focus on the unique co-expression of VIM and CK. We will discuss the structures and functions of these two intermediate filament proteins and how they may interact when co-expressed in hybrid E/M cells. Additionally, we review what is known about cell-surface expression of these intermediate filament proteins and discuss their potential as predictive biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Nick A. Kuburich
- Legorreta Cancer Center, The Warren Alpert Medical School, Brown University, Providence, RI 02912, USA; (N.A.K.); (J.M.K.); (P.d.H.); (A.A.K.)
- Department of Pathology and Lab Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
| | - Julia M. Kiselka
- Legorreta Cancer Center, The Warren Alpert Medical School, Brown University, Providence, RI 02912, USA; (N.A.K.); (J.M.K.); (P.d.H.); (A.A.K.)
- Department of Pathology and Lab Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
| | - Petra den Hollander
- Legorreta Cancer Center, The Warren Alpert Medical School, Brown University, Providence, RI 02912, USA; (N.A.K.); (J.M.K.); (P.d.H.); (A.A.K.)
- Department of Pathology and Lab Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
| | - Andrew A. Karam
- Legorreta Cancer Center, The Warren Alpert Medical School, Brown University, Providence, RI 02912, USA; (N.A.K.); (J.M.K.); (P.d.H.); (A.A.K.)
- Department of Pathology and Lab Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
| | - Sendurai A. Mani
- Legorreta Cancer Center, The Warren Alpert Medical School, Brown University, Providence, RI 02912, USA; (N.A.K.); (J.M.K.); (P.d.H.); (A.A.K.)
- Department of Pathology and Lab Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
| |
Collapse
|
4
|
Wat LW, Svensson KJ. Novel secreted regulators of glucose and lipid metabolism in the development of metabolic diseases. Diabetologia 2024; 67:2626-2636. [PMID: 39180580 DOI: 10.1007/s00125-024-06253-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 07/01/2024] [Indexed: 08/26/2024]
Abstract
The tight regulation of glucose and lipid metabolism is crucial for maintaining metabolic health. Dysregulation of these processes can lead to the development of metabolic diseases. Secreted factors, or hormones, play an essential role in the regulation of glucose and lipid metabolism, thus also playing an important role in the development of metabolic diseases such as type 2 diabetes and obesity. Given the important roles of secreted factors, there has been significant interest in identifying new secreted factors and new functions for existing secreted factors that control glucose and lipid metabolism. In this review, we evaluate novel secreted factors or novel functions of existing factors that regulate glucose and lipid metabolism discovered in the last decade, including secreted isoform of endoplasmic reticulum membrane complex subunit 10, vimentin, cartilage intermediate layer protein 2, isthmin-1, lipocalin-2, neuregulin-1 and neuregulin-4. We discuss their discovery, tissues of origin, mechanisms of action and sex differences, emphasising their potential to regulate metabolic processes central to diabetes. Additionally, we discuss the translational barriers, particularly the absence of identified receptors, that hamper their functional characterisation and further therapeutic development. Ultimately, the identification of new secreted factors may give insights into previously unidentified pathways of disease progression and mechanisms of glucose and lipid homeostasis.
Collapse
Affiliation(s)
- Lianna W Wat
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Diabetes Research Center, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Katrin J Svensson
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA.
- Stanford Diabetes Research Center, Stanford University School of Medicine, Stanford, CA, USA.
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
5
|
Akaberi-Nasrabadi S, Sabbaghi A, M. Toosi B, Ghorbanifaraz P, Mahmoudiasl GR, Aliaghaei A, Faghihi Hosseinabadi H, Paktinat S, Abdollahifar MA. Vimentin as a contributing factor in SARS-CoV-2-induced orchitis on postmortem testicular autopsy of COVID-19 cases: A case-control study. Int J Reprod Biomed 2024; 22:895-906. [PMID: 39866583 PMCID: PMC11757669 DOI: 10.18502/ijrm.v22i11.17822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 09/22/2024] [Accepted: 10/24/2024] [Indexed: 01/28/2025] Open
Abstract
Background Coronavirus disease 2019 (COVID-19) was identified in China in late December 2019 and led to a pandemic that resulted in millions of confirmed cases and deaths. The causative agent, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), uses distinct receptors and co-receptors to enter host cells. Vimentin has emerged as a potential co-receptor for SARS-CoV-2 due to the high level of vimentin expression in testis tissue. Objective The present study investigated the link between vimentin expression level and SARS-CoV-2-induced orchitis. Materials and Methods In this case-control study, testis autopsy samples were collected immediately after the death of both COVID-19 cases and a control group that included individuals who died due to accidental causes. Gene expression and immunohistochemical assays were conducted to evaluate the level of vimentin expression, cell proliferation, and leukocyte infiltration. Results A significant expression of vimentin and infiltration of immune cells (CD68+, CD38+, and CD138+) in the testicular tissue of COVID-19 cases, along with extensive immunoglobulin G precipitation and reduced inhibin expression (p = 0.001) were observed. Additionally, gene expression analysis revealed increased expression of vimentin and decreased expression of the proliferation markers Ki67 and proliferating cell nuclear antigen, suggesting that SARS-CoV-2 may disrupt spermatogenesis through immune responses and the arrest of cell proliferation. Conclusion There may be a strong link between vimentin expression and COVID-19-induced orchitis. Further studies are needed to confirm these findings. Considering some limitations, vimentin can be used as a biomarker option for testicular damage following COVID-19-induced orchitis.
Collapse
Affiliation(s)
- Soheila Akaberi-Nasrabadi
- Urology and Nephrology Research Center, Research Institute for Urology and Nephrology, Shahid Beheshti University of Medical Sciences (SBMU), Tehran, Iran
| | - Azam Sabbaghi
- Urology and Nephrology Research Center, Research Institute for Urology and Nephrology, Shahid Beheshti University of Medical Sciences (SBMU), Tehran, Iran
| | - Behzad M. Toosi
- Department of Small Animal Clinical Sciences, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Parsa Ghorbanifaraz
- Urology and Nephrology Research Center, Research Institute for Urology and Nephrology, Shahid Beheshti University of Medical Sciences (SBMU), Tehran, Iran
| | | | - Abbas Aliaghaei
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hajarsadat Faghihi Hosseinabadi
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shahrokh Paktinat
- Urology and Nephrology Research Center, Research Institute for Urology and Nephrology, Shahid Beheshti University of Medical Sciences (SBMU), Tehran, Iran
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad-Amin Abdollahifar
- Urology and Nephrology Research Center, Research Institute for Urology and Nephrology, Shahid Beheshti University of Medical Sciences (SBMU), Tehran, Iran
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
6
|
Coelho-Rato LS, Parvanian S, Andrs Salajkova S, Medalia O, Eriksson JE. Intermediate filaments at a glance. J Cell Sci 2024; 137:jcs261386. [PMID: 39206824 DOI: 10.1242/jcs.261386] [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] [Indexed: 09/04/2024] Open
Abstract
Intermediate filaments (IFs) comprise a large family of versatile cytoskeletal proteins, divided into six subtypes with tissue-specific expression patterns. IFs have a wide repertoire of cellular functions, including providing structural support to cells, as well as active roles in mechanical support and signaling pathways. Consequently, defects in IFs are associated with more than 100 diseases. In this Cell Science at a Glance article, we discuss the established classes of IFs and their general features, their functions beyond structural support, and recent advances in the field. We also highlight their involvement in disease and potential use as clinical markers of pathological conditions. Finally, we provide our view on current knowledge gaps and the future directions of the IF field.
Collapse
Affiliation(s)
- Leila S Coelho-Rato
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520 Turku, Finland
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, 20520 Turku, Finland
| | - Sepideh Parvanian
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520 Turku, Finland
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, 20520 Turku, Finland
- Center for Systems Biology, Massachusetts General Hospital Research Institute and Harvard Medical School, Boston, MA 02114, USA
| | - Sarka Andrs Salajkova
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Ohad Medalia
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - John E Eriksson
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520 Turku, Finland
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, 20520 Turku, Finland
- Euro-Bioimaging ERIC, 20520 Turku, Finland
| |
Collapse
|
7
|
Coelho-Rato LS, Parvanian S, Modi MK, Eriksson JE. Vimentin at the core of wound healing. Trends Cell Biol 2024; 34:239-254. [PMID: 37748934 DOI: 10.1016/j.tcb.2023.08.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 08/25/2023] [Accepted: 08/25/2023] [Indexed: 09/27/2023]
Abstract
As a member of the large family of intermediate filaments (IFs), vimentin has emerged as a highly dynamic and versatile cytoskeletal protein involved in many key processes of wound healing. It is well established that vimentin is involved in epithelial-mesenchymal transition (EMT) during wound healing and metastasis, during which epithelial cells acquire more dynamic and motile characteristics. Moreover, vimentin participates in multiple cellular activities supporting growth, proliferation, migration, cell survival, and stress resilience. Here, we explore the role of vimentin at each phase of wound healing, with focus on how it integrates different signaling pathways and protects cells in the fluctuating and challenging environments that characterize a healing tissue.
Collapse
Affiliation(s)
- Leila S Coelho-Rato
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520 Turku, Finland; Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, 20520 Turku, Finland
| | - Sepideh Parvanian
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520 Turku, Finland; Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, 20520 Turku, Finland; Center for Systems Biology, Massachusetts General Hospital Research Institute and Harvard Medical School, Boston, MA 02114, USA
| | - Mayank Kumar Modi
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520 Turku, Finland; Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, 20520 Turku, Finland
| | - John E Eriksson
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520 Turku, Finland; Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, 20520 Turku, Finland; Euro-Bioimaging ERIC, 20520 Turku, Finland.
| |
Collapse
|
8
|
Suprewicz Ł, Zakrzewska M, Okła S, Głuszek K, Sadzyńska A, Deptuła P, Fiedoruk K, Bucki R. Extracellular vimentin as a modulator of the immune response and an important player during infectious diseases. Immunol Cell Biol 2024; 102:167-178. [PMID: 38211939 DOI: 10.1111/imcb.12721] [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: 09/23/2023] [Revised: 11/27/2023] [Accepted: 12/21/2023] [Indexed: 01/13/2024]
Abstract
Vimentin, an intermediate filament protein primarily recognized for its intracellular role in maintaining cellular structure, has recently garnered increased attention and emerged as a pivotal extracellular player in immune regulation and host-pathogen interactions. While the functions of extracellular vimentin were initially overshadowed by its cytoskeletal role, accumulating evidence now highlights its significance in diverse physiological and pathological events. This review explores the multifaceted role of extracellular vimentin in modulating immune responses and orchestrating interactions between host cells and pathogens. It delves into the mechanisms underlying vimentin's release into the extracellular milieu, elucidating its unconventional secretion pathways and identifying critical molecular triggers. In addition, the future perspectives of using extracellular vimentin in diagnostics and as a target protein in the treatment of diseases are discussed.
Collapse
Affiliation(s)
- Łukasz Suprewicz
- Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Białystok, Białystok, Poland
| | - Magdalena Zakrzewska
- Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Białystok, Białystok, Poland
| | - Sławomir Okła
- Institute of Medical Sciences, Collegium Medicum, Jan Kochanowski University of Kielce, Kielce, Poland
| | - Katarzyna Głuszek
- Institute of Medical Sciences, Collegium Medicum, Jan Kochanowski University of Kielce, Kielce, Poland
| | - Alicja Sadzyńska
- State Higher Vocational School of Prof. Edward F. Szczepanik in Suwałki, Suwałki, Poland
| | - Piotr Deptuła
- Independent Laboratory of Nanomedicine, Medical University of Białystok, Białystok, Poland
| | - Krzysztof Fiedoruk
- Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Białystok, Białystok, Poland
| | - Robert Bucki
- Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Białystok, Białystok, Poland
| |
Collapse
|
9
|
Carpo N, Tran V, Biancotti JC, Cepeda C, Espinosa-Jeffrey A. Space Flight Enhances Stress Pathways in Human Neural Stem Cells. Biomolecules 2024; 14:65. [PMID: 38254665 PMCID: PMC10813251 DOI: 10.3390/biom14010065] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 12/23/2023] [Accepted: 12/27/2023] [Indexed: 01/24/2024] Open
Abstract
Mammalian cells have evolved to function under Earth's gravity, but how they respond to microgravity remains largely unknown. Neural stem cells (NSCs) are essential for the maintenance of central nervous system (CNS) functions during development and the regeneration of all CNS cell populations. Here, we examined the behavior of space (SPC)-flown NSCs as they readapted to Earth's gravity. We found that most of these cells survived the space flight and self-renewed. Yet, some showed enhanced stress responses as well as autophagy-like behavior. To ascertain if the secretome from SPC-flown NSCs contained molecules inducing these responses, we incubated naïve, non-starved NSCs in a medium containing SPC-NSC secretome. We found a four-fold increase in stress responses. Proteomic analysis of the secretome revealed that the protein of the highest content produced by SPC-NSCs was secreted protein acidic and rich in cysteine (SPARC), which induces endoplasmic reticulum (ER) stress, resulting in the cell's demise. These results offer novel knowledge on the response of neural cells, particularly NSCs, subjected to space microgravity. Moreover, some secreted proteins have been identified as microgravity sensing, paving a new venue for future research aiming at targeting the SPARC metabolism. Although we did not establish a direct relationship between microgravity-induced stress and SPARC as a potential marker, these results represent the first step in the identification of gravity sensing molecules as targets to be modulated and to design effective countermeasures to mitigate intracranial hypertension in astronauts using structure-based protein design.
Collapse
Affiliation(s)
- Nicholas Carpo
- Department of Psychiatry, UCLA, Los Angeles, CA 90095, USA (V.T.); (C.C.)
| | - Victoria Tran
- Department of Psychiatry, UCLA, Los Angeles, CA 90095, USA (V.T.); (C.C.)
| | - Juan Carlos Biancotti
- Department of Surgery, Division of Pediatric Surgery, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA;
| | - Carlos Cepeda
- Department of Psychiatry, UCLA, Los Angeles, CA 90095, USA (V.T.); (C.C.)
| | | |
Collapse
|
10
|
Thalla DG, Lautenschläger F. Extracellular vimentin: Battle between the devil and the angel. Curr Opin Cell Biol 2023; 85:102265. [PMID: 37866018 DOI: 10.1016/j.ceb.2023.102265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 08/31/2023] [Accepted: 09/24/2023] [Indexed: 10/24/2023]
Abstract
Vimentin, an intracellular cytoskeletal protein, can be secreted by various cells in response to conditions such as injury, stress, senescence, and cancer. Once vimentin is secreted outside of the cell, it is called extracellular vimentin. This extracellular vimentin is significantly involved in pathological conditions, particularly in the areas of viral infection, cancer, immune response, and wound healing. The effects of extracellular vimentin can be either positive or negative, for example it can enhance axonal repair but also mediates SARS-CoV-2 infection. In this review, we categorize the functional implications of extracellular vimentin based on its localization outside the cell. Specifically, we classify extracellular vimentin into two distinct forms: surface vimentin, which remains bound to the cell surface, and secreted vimentin, which refers to the free form that is completely released outside the cell. Overall, extracellular vimentin has a dual nature that encompasses both beneficial and detrimental effects on the functionality of cells, organs and whole organisms. Here, we summarize its effects in viral infection, cancer, immune response and wound healing. We find that surface vimentin is often associated with negative consequences, whereas secreted vimentin manifests predominantly with positive influences. We found that the observed effects of extracellular vimentin strongly depend on the specific circumstances under which its expression occurs in cells.
Collapse
Affiliation(s)
| | - Franziska Lautenschläger
- Experimental Physics, Saarland University, Saarbrücken, Germany; Centre for Biophysics, Saarland University, Saarbrücken, Germany.
| |
Collapse
|
11
|
Parvanian S, Coelho-Rato LS, Patteson AE, Eriksson JE. Vimentin takes a hike - Emerging roles of extracellular vimentin in cancer and wound healing. Curr Opin Cell Biol 2023; 85:102246. [PMID: 37783033 PMCID: PMC11214764 DOI: 10.1016/j.ceb.2023.102246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 08/28/2023] [Accepted: 09/04/2023] [Indexed: 10/04/2023]
Abstract
Vimentin is a cytoskeletal protein important for many cellular processes, including proliferation, migration, invasion, stress resistance, signaling, and many more. The vimentin-deficient mouse has revealed many of these functions as it has numerous severe phenotypes, many of which are found only following a suitable challenge or stress. While these functions are usually related to vimentin as a major intracellular protein, vimentin is also emerging as an extracellular protein, exposed at the cell surface in an oligomeric form or secreted to the extracellular environment in soluble and vesicle-bound forms. Thus, this review explores the roles of the extracellular pool of vimentin (eVIM), identified in both normal and pathological states. It focuses specifically on the recent advances regarding the role of eVIM in wound healing and cancer. Finally, it discusses new technologies and future perspectives for the clinical application of eVIM.
Collapse
Affiliation(s)
- Sepideh Parvanian
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520, Turku, Finland; Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, 20520 Turku, Finland; Center for Systems Biology, Massachusetts General Hospital Research Institute and Harvard Medical School, Boston, MA 02114, USA
| | - Leila S Coelho-Rato
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520, Turku, Finland; Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, 20520 Turku, Finland
| | - Alison E Patteson
- Physics Department and BioInspired Institute, Syracuse University, Syracuse, NY, 13244, USA
| | - John E Eriksson
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520, Turku, Finland; Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, 20520 Turku, Finland; Euro-Bioimaging ERIC, 20520 Turku, Finland.
| |
Collapse
|
12
|
Parvanian S, Coelho-Rato LS, Eriksson JE, Patteson AE. The molecular biophysics of extracellular vimentin and its role in pathogen-host interactions. Curr Opin Cell Biol 2023; 85:102233. [PMID: 37677998 PMCID: PMC10841047 DOI: 10.1016/j.ceb.2023.102233] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 08/08/2023] [Accepted: 08/10/2023] [Indexed: 09/09/2023]
Abstract
Vimentin, an intermediate filament protein typically located in the cytoplasm of mesenchymal cells, can also be secreted as an extracellular protein. The organization of extracellular vimentin strongly determines its functions in physiological and pathological conditions, making it a promising target for future therapeutic interventions. The extracellular form of vimentin has been found to play a role in the interaction between host cells and pathogens. In this review, we first discuss the molecular biophysics of extracellular vimentin, including its structure, secretion, and adhesion properties. We then provide a general overview of the role of extracellular vimentin in mediating pathogen-host interactions, with a focus on its interactions with viruses and bacteria. We also discuss the implications of these findings for the development of new therapeutic strategies for combating infectious diseases.
Collapse
Affiliation(s)
- Sepideh Parvanian
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520, Turku, Finland; Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, 20520, Turku, Finland; Center for Systems Biology, Massachusetts General Hospital Research Institute and Harvard Medical School, Boston, MA 02114, USA
| | - Leila S Coelho-Rato
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520, Turku, Finland; Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, 20520, Turku, Finland
| | - John E Eriksson
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520, Turku, Finland; Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, 20520, Turku, Finland; Euro-Bioimaging ERIC, 20520, Turku, Finland
| | - Alison E Patteson
- Physics Department and BioInspired Institute, Syracuse University, Syracuse, NY, 13244, USA.
| |
Collapse
|
13
|
van Loon K, van Breest Smallenburg ME, Huijbers EJM, Griffioen AW, van Beijnum JR. Extracellular vimentin as a versatile immune suppressive protein in cancer. Biochim Biophys Acta Rev Cancer 2023; 1878:188985. [PMID: 37717859 DOI: 10.1016/j.bbcan.2023.188985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 08/24/2023] [Accepted: 08/29/2023] [Indexed: 09/19/2023]
Abstract
The interest in finding new targets in the tumor microenvironment for anti-cancer therapy has increased rapidly over the years. More specifically, the tumor-associated blood vessels are a promising target. We recently found that the intermediate filament protein vimentin is externalized by endothelial cells of the tumor vasculature. Extracellular vimentin was shown to sustain angiogenesis by mimicking VEGF and supporting cell migration, as well as endothelial cell anergy, the unresponsiveness of the endothelium to proinflammatory cytokines. The latter hampers immune cell infiltration and subsequently provides escape from tumor immunity. Other studies showed that extracellular vimentin plays a role in sustained systemic and local inflammation. Here we will review the reported roles of extracellular vimentin with a particular emphasis on its involvement in the interactions between immune cells and the endothelium in the tumor microenvironment. To this end, we discuss the different ways by which extracellular vimentin modulates the immune system. Moreover, we review how this protein can alter immune cell-vessel wall adhesion by altering the expression of adhesion proteins, attenuating immune cell infiltration into the tumor parenchyma. Finally, we discuss how vimentin-targeting therapy can reverse endothelial cell anergy and promote immune infiltration, supporting anti-tumor immunity.
Collapse
Affiliation(s)
- Karlijn van Loon
- Angiogenesis Laboratory, Department of Medical Oncology, Amsterdam University Medical Center, Cancer Center Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Mathilda E van Breest Smallenburg
- Angiogenesis Laboratory, Department of Medical Oncology, Amsterdam University Medical Center, Cancer Center Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Elisabeth J M Huijbers
- Angiogenesis Laboratory, Department of Medical Oncology, Amsterdam University Medical Center, Cancer Center Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands; CimCure BV, Amsterdam, the Netherlands
| | - Arjan W Griffioen
- Angiogenesis Laboratory, Department of Medical Oncology, Amsterdam University Medical Center, Cancer Center Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands; CimCure BV, Amsterdam, the Netherlands
| | - Judy R van Beijnum
- Angiogenesis Laboratory, Department of Medical Oncology, Amsterdam University Medical Center, Cancer Center Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands; CimCure BV, Amsterdam, the Netherlands.
| |
Collapse
|
14
|
Bucki R, Iwamoto DV, Shi X, Kerr KE, Byfield FJ, Suprewicz Ł, Skłodowski K, Sutaria J, Misiak P, Wilczewska AZ, Ramachandran S, Wolfe A, Thanh MTH, Whalen E, Patteson AE, Janmey PA. Extracellular vimentin is sufficient to promote cell attachment, spreading, and motility by a mechanism involving N-acetyl glucosamine-containing structures. J Biol Chem 2023; 299:104963. [PMID: 37356720 PMCID: PMC10392088 DOI: 10.1016/j.jbc.2023.104963] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 05/29/2023] [Accepted: 06/06/2023] [Indexed: 06/27/2023] Open
Abstract
Vimentin intermediate filaments form part of the cytoskeleton of mesenchymal cells, but under pathological conditions often associated with inflammation, vimentin filaments depolymerize as the result of phosphorylation or citrullination, and vimentin oligomers are secreted or released into the extracellular environment. In the extracellular space, vimentin can bind surfaces of cells and the extracellular matrix, and the interaction between extracellular vimentin and cells can trigger changes in cellular functions, such as activation of fibroblasts to a fibrotic phenotype. The mechanism by which extracellular vimentin binds external cell membranes and whether vimentin alone can act as an adhesive anchor for cells is largely uncharacterized. Here, we show that various cell types (normal and vimentin null fibroblasts, mesenchymal stem cells, and A549 lung carcinoma cells) attach to and spread on polyacrylamide hydrogel substrates covalently linked to vimentin. Using traction force microscopy and spheroid expansion assays, we characterize how different cell types respond to extracellular vimentin. Cell attachment to and spreading on vimentin-coated surfaces is inhibited by hyaluronic acid degrading enzymes, hyaluronic acid synthase inhibitors, soluble heparin or N-acetyl glucosamine, all of which are treatments that have little or no effect on the same cell types binding to collagen-coated hydrogels. These studies highlight the effectiveness of substrate-bound vimentin as a ligand for cells and suggest that carbohydrate structures, including the glycocalyx and glycosylated cell surface proteins that contain N-acetyl glucosamine, form a novel class of adhesion receptors for extracellular vimentin that can either directly support cell adhesion to a substrate or fine-tune the glycocalyx adhesive properties.
Collapse
Affiliation(s)
- Robert Bucki
- Department of Physiology, Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA; Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Białystok, Białystok, Poland.
| | - Daniel V Iwamoto
- Department of Physiology, Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Xuechen Shi
- Department of Physiology, Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Katherine E Kerr
- Department of Physiology, Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Fitzroy J Byfield
- Department of Physiology, Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Łukasz Suprewicz
- Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Białystok, Białystok, Poland
| | - Karol Skłodowski
- Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Białystok, Białystok, Poland
| | - Julian Sutaria
- Department of Physiology, Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Paweł Misiak
- Faculty of Chemistry, University of Białystok, Białystok, Poland
| | | | | | - Aaron Wolfe
- Ichor Life Sciences, Inc, LaFayette, New York, USA; Lewis School of Health Sciences, Clarkson University, Potsdam, New York, USA
| | - Minh-Tri Ho Thanh
- Physics Department, BioInspired Institute, Syracuse University, Syracuse, New York, USA
| | - Eli Whalen
- Physics Department, BioInspired Institute, Syracuse University, Syracuse, New York, USA
| | - Alison E Patteson
- Physics Department, BioInspired Institute, Syracuse University, Syracuse, New York, USA.
| | - Paul A Janmey
- Department of Physiology, Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
15
|
Peng K, Liao Y, Li X, Zeng D, Ye Y, Chen L, Zeng Z, Zeng Y. Vimentin Is an Attachment Receptor for Mycoplasma pneumoniae P1 Protein. Microbiol Spectr 2023; 11:e0448922. [PMID: 36912679 PMCID: PMC10100666 DOI: 10.1128/spectrum.04489-22] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 02/06/2023] [Indexed: 03/14/2023] Open
Abstract
Mycoplasma pneumoniae is the most common pathogen causing respiratory tract infection, and the P1 protein on its adhesion organelle plays a crucial role during the pathogenic process. Currently, there are many studies on P1 and receptors on host cells, but the adhesion mechanism of P1 protein is still unclear. In this study, a modified virus overlay protein binding assay (VOPBA) and liquid chromatography-mass spectrometry (LC-MS) were performed to screen for proteins that specifically bind to the region near the carboxyl terminus of the recombinant P1 protein (rP1-C). The interaction between rP1-C and vimentin or β-4-tubulin were confirmed by far-Western blotting and coimmunoprecipitation. Results verified that vimentin and β-4-tubulin were mainly distributed on the cell membrane and cytoplasm of human bronchial epithelial (BEAS-2B) cells, but only vimentin could interact with rP1-C. The results of the adhesion and adhesion inhibition assays indicated that the adhesion of M. pneumoniae and rP1-C to cells could be partly inhibited by vimentin and its antibody. When vimentin was downregulated with the corresponding small interfering RNA (siRNA) or overexpressed in BEAS-2B cells, the adhesion of M. pneumoniae and rP1-C to cells was decreased or increased, respectively, which indicated that vimentin was closely associated with the adhesion of M. pneumoniae and rP1-C to BEAS-2B cells. Our results demonstrate that vimentin could be a receptor on human bronchial epithelial cells for the P1 protein and plays an essential role in the adhesion of M. pneumoniae to cells, which may clarify the pathogenesis of M. pneumoniae. IMPORTANCE Mycoplasma pneumoniae is the most common pathogen causing respiratory tract infection, and the P1 protein on its adhesion organelle plays a crucial role during the pathogenic process. A variety of experiments, including enzyme-linked immunosorbent assay (ELISA), coimmunoprecipitation, adhesion, and adhesion inhibition assay, have demonstrated that the M. pneumoniae P1 protein can interact with vimentin, that the adhesion of M. pneumoniae and recombinant P1 protein to BEAS-2B cells was affected by the expression level of vimentin. This provides a new idea for the prevention and treatment of Mycoplasma pneumoniae infection.
Collapse
Affiliation(s)
- Kailan Peng
- Institute of Pathogenic Biology, Basic Medical School, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, Hunan Province, People’s Republic of China
| | - Yating Liao
- Institute of Pathogenic Biology, Basic Medical School, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, Hunan Province, People’s Republic of China
| | - Xia Li
- Institute of Pathogenic Biology, Basic Medical School, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, Hunan Province, People’s Republic of China
| | - Dongdong Zeng
- Department of Cardiocascular Medicine, the Third Affiliated Hospital, University of South China, Hengyang, Hunan Province, People’s Republic of China
| | - Youyuan Ye
- Institute of Pathogenic Biology, Basic Medical School, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, Hunan Province, People’s Republic of China
| | - Li Chen
- Institute of Pathogenic Biology, Basic Medical School, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, Hunan Province, People’s Republic of China
| | - Zhuo Zeng
- Institute of Pathogenic Biology, Basic Medical School, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, Hunan Province, People’s Republic of China
| | - Yanhua Zeng
- Institute of Pathogenic Biology, Basic Medical School, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, Hunan Province, People’s Republic of China
| |
Collapse
|
16
|
Malfeld K, Armbrecht N, Pich A, Volk HA, Lenarz T, Scheper V. Prevention of Noise-Induced Hearing Loss In Vivo: Continuous Application of Insulin-like Growth Factor 1 and Its Effect on Inner Ear Synapses, Auditory Function and Perilymph Proteins. Int J Mol Sci 2022; 24:ijms24010291. [PMID: 36613734 PMCID: PMC9820558 DOI: 10.3390/ijms24010291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 12/19/2022] [Accepted: 12/19/2022] [Indexed: 12/28/2022] Open
Abstract
As noise-induced hearing loss (NIHL) is a leading cause of occupational diseases, there is an urgent need for the development of preventive and therapeutic interventions. To avoid user-compliance-based problems occurring with conventional protection devices, the pharmacological prevention is currently in the focus of hearing research. Noise exposure leads to an increase in reactive oxygen species (ROS) in the cochlea. This way antioxidant agents are a promising option for pharmacological interventions. Previous animal studies reported preventive as well as therapeutic effects of Insulin-like growth factor 1 (IGF-1) in the context of NIHL. Unfortunately, in patients the time point of the noise trauma cannot always be predicted, and additive effects may occur. Therefore, continuous prevention seems to be beneficial. The present study aimed to investigate the preventive potential of continuous administration of low concentrations of IGF-1 to the inner ear in an animal model of NIHL. Guinea pigs were unilaterally implanted with an osmotic minipump. One week after surgery they received noise trauma, inducing a temporary threshold shift. Continuous IGF-1 delivery lasted for seven more days. It did not lead to significantly improved hearing thresholds compared to control animals. Quite the contrary, there is a hint for a higher noise susceptibility. Nevertheless, changes in the perilymph proteome indicate a reduced damage and better repair mechanisms through the IGF-1 treatment. Thus, future studies should investigate delivery methods enabling continuous prevention but reducing the risk of an overdosage.
Collapse
Affiliation(s)
- Kathrin Malfeld
- Department of Otolaryngology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
- Department of Small Animal Medicine and Surgery, University of Veterinary Medicine Hannover, Foundation, 30559 Hannover, Germany
| | - Nina Armbrecht
- Department of Otolaryngology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Andreas Pich
- Core Facility Proteomics, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Holger A. Volk
- Department of Small Animal Medicine and Surgery, University of Veterinary Medicine Hannover, Foundation, 30559 Hannover, Germany
| | - Thomas Lenarz
- Department of Otolaryngology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
- Cluster of Excellence “Hearing4all”, German Research Foundation (DFG; “Deutsche Forschungsgemeinschaft”), Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Verena Scheper
- Department of Otolaryngology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
- Cluster of Excellence “Hearing4all”, German Research Foundation (DFG; “Deutsche Forschungsgemeinschaft”), Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
- Correspondence:
| |
Collapse
|
17
|
Chen KZ, Liu SX, Li YW, He T, Zhao J, Wang T, Qiu XX, Wu HF. Vimentin as a potential target for diverse nervous system diseases. Neural Regen Res 2022; 18:969-975. [PMID: 36254976 PMCID: PMC9827761 DOI: 10.4103/1673-5374.355744] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Vimentin is a major type III intermediate filament protein that plays important roles in several basic cellular functions including cell migration, proliferation, and division. Although vimentin is a cytoplasmic protein, it also exists in the extracellular matrix and at the cell surface. Previous studies have shown that vimentin may exert multiple physiological effects in different nervous system injuries and diseases. For example, the studies of vimentin in spinal cord injury and stroke mainly focus on the formation of reactive astrocytes. Reduced glial scar, increased axonal regeneration, and improved motor function have been noted after spinal cord injury in vimentin and glial fibrillary acidic protein knockout (GFAP-/-VIM-/-) mice. However, attenuated glial scar formation in post-stroke in GFAP-/- VIM-/- mice resulted in abnormal neuronal network restoration and worse neurological recovery. These opposite results have been attributed to the multiple roles of glial scar in different temporal and spatial conditions. In addition, extracellular vimentin may be a neurotrophic factor that promotes axonal extension by interaction with the insulin-like growth factor 1 receptor. In the pathogenesis of bacterial meningitis, cell surface vimentin is a meningitis facilitator, acting as a receptor of multiple pathogenic bacteria, including E. coli K1, Listeria monocytogenes, and group B streptococcus. Compared with wild type mice, VIM-/- mice are less susceptible to bacterial infection and exhibit a reduced inflammatory response, suggesting that vimentin is necessary to induce the pathogenesis of meningitis. Recently published literature showed that vimentin serves as a double-edged sword in the nervous system, regulating axonal regrowth, myelination, apoptosis, and neuroinflammation. This review aims to provide an overview of vimentin in spinal cord injury, stroke, bacterial meningitis, gliomas, and peripheral nerve injury and to discuss the potential therapeutic methods involving vimentin manipulation in improving axonal regeneration, alleviating infection, inhibiting brain tumor progression, and enhancing nerve myelination.
Collapse
Affiliation(s)
- Kang-Zhen Chen
- Department of Anesthesiology, Guangzhou Huadu Hospital Affiliated to Guangdong Medical University (Guangzhou Huadu District Maternal and Child Health Care Hospital), Guangzhou, Guangdong Province, China,Dongguan City Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Guangdong Medical University, Dongguan, Guangdong Province, China
| | - Shu-Xian Liu
- Department of Anesthesiology, Guangzhou Huadu Hospital Affiliated to Guangdong Medical University (Guangzhou Huadu District Maternal and Child Health Care Hospital), Guangzhou, Guangdong Province, China
| | - Yan-Wei Li
- Department of Anesthesiology, Guangzhou Huadu Hospital Affiliated to Guangdong Medical University (Guangzhou Huadu District Maternal and Child Health Care Hospital), Guangzhou, Guangdong Province, China
| | - Tao He
- Dongguan City Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Guangdong Medical University, Dongguan, Guangdong Province, China
| | - Jie Zhao
- Dongguan City Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Guangdong Medical University, Dongguan, Guangdong Province, China
| | - Tao Wang
- Department of Surgery, the Third Hospital of Guangdong Medical University (Longjiang Hospital of Shunde District), Foshan, Guangdong Province, China,Correspondence to: Hong-Fu Wu, ; Xian-Xiu Qiu, ; Tao Wang, .
| | - Xian-Xiu Qiu
- Dongguan City Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Guangdong Medical University, Dongguan, Guangdong Province, China,Correspondence to: Hong-Fu Wu, ; Xian-Xiu Qiu, ; Tao Wang, .
| | - Hong-Fu Wu
- Department of Anesthesiology, Guangzhou Huadu Hospital Affiliated to Guangdong Medical University (Guangzhou Huadu District Maternal and Child Health Care Hospital), Guangzhou, Guangdong Province, China,Dongguan City Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Guangdong Medical University, Dongguan, Guangdong Province, China,Correspondence to: Hong-Fu Wu, ; Xian-Xiu Qiu, ; Tao Wang, .
| |
Collapse
|
18
|
Extracellular vimentin mimics VEGF and is a target for anti-angiogenic immunotherapy. Nat Commun 2022; 13:2842. [PMID: 35606362 PMCID: PMC9126915 DOI: 10.1038/s41467-022-30063-7] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Accepted: 04/14/2022] [Indexed: 12/12/2022] Open
Abstract
Anti-angiogenic cancer therapies possess immune-stimulatory properties by counteracting pro-angiogenic molecular mechanisms. We report that tumor endothelial cells ubiquitously overexpress and secrete the intermediate filament protein vimentin through type III unconventional secretion mechanisms. Extracellular vimentin is pro-angiogenic and functionally mimics VEGF action, while concomitantly acting as inhibitor of leukocyte-endothelial interactions. Antibody targeting of extracellular vimentin shows inhibition of angiogenesis in vitro and in vivo. Effective and safe inhibition of angiogenesis and tumor growth in several preclinical and clinical studies is demonstrated using a vaccination strategy against extracellular vimentin. Targeting vimentin induces a pro-inflammatory condition in the tumor, exemplified by induction of the endothelial adhesion molecule ICAM1, suppression of PD-L1, and altered immune cell profiles. Our findings show that extracellular vimentin contributes to immune suppression and functions as a vascular immune checkpoint molecule. Targeting of extracellular vimentin presents therefore an anti-angiogenic immunotherapy strategy against cancer. The pro-tumorigenic effects of vimentin have been attributed to intracellular functions in tumour cells so far. Here, the authors show that tumour endothelial cells can secrete vimentin as a pro-angiogenic factor and that targeting of vimentin can be used as an immunotherapeutic strategy.
Collapse
|
19
|
Lalioti V, González-Sanz S, Lois-Bermejo I, González-Jiménez P, Viedma-Poyatos Á, Merino A, Pajares MA, Pérez-Sala D. Cell surface detection of vimentin, ACE2 and SARS-CoV-2 Spike proteins reveals selective colocalization at primary cilia. Sci Rep 2022; 12:7063. [PMID: 35487944 PMCID: PMC9052736 DOI: 10.1038/s41598-022-11248-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 04/04/2022] [Indexed: 12/24/2022] Open
Abstract
The SARS-CoV-2 Spike protein mediates docking of the virus onto cells prior to viral invasion. Several cellular receptors facilitate SARS-CoV-2 Spike docking at the cell surface, of which ACE2 plays a key role in many cell types. The intermediate filament protein vimentin has been reported to be present at the surface of certain cells and act as a co-receptor for several viruses; furthermore, its potential involvement in interactions with Spike proteins has been proposed. Nevertheless, the potential colocalization of vimentin with Spike and its receptors on the cell surface has not been explored. Here we have assessed the binding of Spike protein constructs to several cell types. Incubation of cells with tagged Spike S or Spike S1 subunit led to discrete dotted patterns at the cell surface, which consistently colocalized with endogenous ACE2, but sparsely with a lipid raft marker. Vimentin immunoreactivity mostly appeared as spots or patches unevenly distributed at the surface of diverse cell types. Of note, vimentin could also be detected in extracellular particles and in the cytoplasm underlying areas of compromised plasma membrane. Interestingly, although overall colocalization of vimentin-positive spots with ACE2 or Spike was moderate, a selective enrichment of the three proteins was detected at elongated structures, positive for acetylated tubulin and ARL13B. These structures, consistent with primary cilia, concentrated Spike binding at the top of the cells. Our results suggest that a vimentin-Spike interaction could occur at selective locations of the cell surface, including ciliated structures, which can act as platforms for SARS-CoV-2 docking.
Collapse
Affiliation(s)
- Vasiliki Lalioti
- Department of Structural and Chemical Biology, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Ramiro de Maeztu, 9, 28040, Madrid, Spain
| | - Silvia González-Sanz
- Department of Structural and Chemical Biology, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Ramiro de Maeztu, 9, 28040, Madrid, Spain
| | - Irene Lois-Bermejo
- Department of Structural and Chemical Biology, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Ramiro de Maeztu, 9, 28040, Madrid, Spain
| | - Patricia González-Jiménez
- Department of Structural and Chemical Biology, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Ramiro de Maeztu, 9, 28040, Madrid, Spain
| | - Álvaro Viedma-Poyatos
- Department of Structural and Chemical Biology, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Ramiro de Maeztu, 9, 28040, Madrid, Spain
| | - Andrea Merino
- Department of Structural and Chemical Biology, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Ramiro de Maeztu, 9, 28040, Madrid, Spain
| | - María A Pajares
- Department of Structural and Chemical Biology, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Ramiro de Maeztu, 9, 28040, Madrid, Spain
| | - Dolores Pérez-Sala
- Department of Structural and Chemical Biology, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Ramiro de Maeztu, 9, 28040, Madrid, Spain.
| |
Collapse
|
20
|
Menko AS, Romisher A, Walker JL. The Pro-fibrotic Response of Mesenchymal Leader Cells to Lens Wounding Involves Hyaluronic Acid, Its Receptor RHAMM, and Vimentin. Front Cell Dev Biol 2022; 10:862423. [PMID: 35386200 PMCID: PMC8977891 DOI: 10.3389/fcell.2022.862423] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 03/08/2022] [Indexed: 12/31/2022] Open
Abstract
Hyaluronic Acid/Hyaluronan (HA) is a major component of the provisional matrix deposited by cells post-wounding with roles both in regulating cell migration to repair a wound and in promoting a fibrotic outcome to wounding. Both are mediated through its receptors CD44 and RHAMM. We now showed that HA is present in the provisional matrix assembled on the substrate surface in a lens post-cataract surgery explant wound model in which mesenchymal leader cells populate the wound edges to direct migration of the lens epithelium across the adjacent culture substrate onto which this matrix is assembled. Inhibiting HA expression with 4-MU blocked assembly of FN-EDA and collagen I by the wound-responsive mesenchymal leader cells and their migration. These cells express both the HA receptors CD44 and RHAMM. CD44 co-localized with HA at their cell-cell interfaces. RHAMM was predominant in the lamellipodial protrusions extended by the mesenchymal cells at the leading edge, and along HA fibrils organized on the substrate surface. Within a few days post-lens wounding the leader cells are induced to transition to αSMA+ myofibroblasts. Since HA/RHAMM is implicated in both cell migration and inducing fibrosis we examined the impact of blocking HA synthesis on myofibroblast emergence and discovered that it was dependent on HA. While RHAMM has not been previously linked to the intermediate filament protein vimentin, our studies with these explant cultures have shown that vimentin in the cells’ lamellipodial protrusions regulate their transition to myofibroblast. PLA studies now revealed that RHAMM was complexed with both HA and vimentin in the lamellipodial protrusions of leader cells, implicating this HA/RHAMM/vimentin complex in the regulation of leader cell function post-wounding, both in promoting cell migration and in the transition of these cells to myofibroblasts. These results increase our understanding of how the post-wounding matrix environment interacts with receptor/cytoskeletal complexes to determine whether injury outcomes are regenerative or fibrotic.
Collapse
Affiliation(s)
- A Sue Menko
- Department of Pathology, Anatomy and Cell Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, United States.,Department of Ophthalmology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, United States
| | - Alison Romisher
- Department of Pathology, Anatomy and Cell Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, United States
| | - Janice L Walker
- Department of Pathology, Anatomy and Cell Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, United States.,Department of Ophthalmology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, United States
| |
Collapse
|
21
|
Vimentin: Regulation and pathogenesis. Biochimie 2022; 197:96-112. [DOI: 10.1016/j.biochi.2022.02.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 01/11/2022] [Accepted: 02/09/2022] [Indexed: 12/18/2022]
|
22
|
Suprewicz Ł, Swoger M, Gupta S, Piktel E, Byfield FJ, Iwamoto DV, Germann D, Reszeć J, Marcińczyk N, Carroll RJ, Janmey PA, Schwarz JM, Bucki R, Patteson AE. Extracellular Vimentin as a Target Against SARS-CoV-2 Host Cell Invasion. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2105640. [PMID: 34866333 PMCID: PMC9252327 DOI: 10.1002/smll.202105640] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 10/29/2021] [Indexed: 05/07/2023]
Abstract
Infection of human cells by pathogens, including SARS-CoV-2, typically proceeds by cell surface binding to a crucial receptor. The primary receptor for SARS-CoV-2 is the angiotensin-converting enzyme 2 (ACE2), yet new studies reveal the importance of additional extracellular co-receptors that mediate binding and host cell invasion by SARS-CoV-2. Vimentin is an intermediate filament protein that is increasingly recognized as being present on the extracellular surface of a subset of cell types, where it can bind to and facilitate pathogens' cellular uptake. Biophysical and cell infection studies are done to determine whether vimentin might bind SARS-CoV-2 and facilitate its uptake. Dynamic light scattering shows that vimentin binds to pseudovirus coated with the SARS-CoV-2 spike protein, and antibodies against vimentin block in vitro SARS-CoV-2 pseudovirus infection of ACE2-expressing cells. The results are consistent with a model in which extracellular vimentin acts as a co-receptor for SARS-CoV-2 spike protein with a binding affinity less than that of the spike protein with ACE2. Extracellular vimentin may thus serve as a critical component of the SARS-CoV-2 spike protein-ACE2 complex in mediating SARS-CoV-2 cell entry, and vimentin-targeting agents may yield new therapeutic strategies for preventing and slowing SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Łukasz Suprewicz
- Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Białystok, Poland
| | - Maxx Swoger
- Physics Department and BioInspired Institute, Syracuse University
| | - Sarthak Gupta
- Physics Department and BioInspired Institute, Syracuse University
| | - Ewelina Piktel
- Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Białystok, Poland
| | - Fitzroy J. Byfield
- Institute for Medicine and Engineering and Department of Physiology, University of Pennsylvania
| | - Daniel V. Iwamoto
- Institute for Medicine and Engineering and Department of Physiology, University of Pennsylvania
| | - Danielle Germann
- Physics Department and BioInspired Institute, Syracuse University
| | - Joanna Reszeć
- Department of Medical Pathomorphology, Medical University of Białystok, PL-15269 Białystok, Poland
| | - Natalia Marcińczyk
- Department of Biopharmacy, Medical University of Białystok, Białystok, Poland
| | | | - Paul A. Janmey
- Institute for Medicine and Engineering and Department of Physiology, University of Pennsylvania
| | - J. M. Schwarz
- Physics Department and BioInspired Institute, Syracuse University
- Indian Creek Farm, Ithaca, NY
| | - Robert Bucki
- Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Białystok, Poland
- Institute for Medicine and Engineering and Department of Physiology, University of Pennsylvania
| | | |
Collapse
|
23
|
Ren YS, Li HL, Piao XH, Yang ZY, Wang SM, Ge YW. Drug affinity responsive target stability (DARTS) accelerated small molecules target discovery: Principles and application. Biochem Pharmacol 2021; 194:114798. [PMID: 34678227 DOI: 10.1016/j.bcp.2021.114798] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 09/28/2021] [Accepted: 09/29/2021] [Indexed: 12/19/2022]
Abstract
Drug affinity responsive target stability (DARTS) is a novel target discovery approach and is particularly adept at screening small molecule (SM) targets without requiring any structural modifications. The DARTS method is capable of revealing drug-target interactions from cells or tissues by tracking changes in the stability of proteins acting as receptors of bioactive SMs. Due to its simple operation and high efficiency, the DARTS method has been applied to uncover the drug-action mechanism. This review summarized analytical principles, protocols, validation approaches, applications, and challenges involved in the DARTS method. Due to the innate advantages of the DARTS method, it is expected to be a powerful tool to accelerate SM target discovery, especially for bioactive natural products with unknown mechanisms.
Collapse
Affiliation(s)
- Ying-Shan Ren
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China; Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM, Guangdong Pharmaceutical University, Guangzhou 510006, China; Engineering & Technology Research Center for Chinese Materia Medica Quality of the Universities of Guangdong Province, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Hui-Lin Li
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China; Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM, Guangdong Pharmaceutical University, Guangzhou 510006, China; Engineering & Technology Research Center for Chinese Materia Medica Quality of the Universities of Guangdong Province, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Xiu-Hong Piao
- Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM, Guangdong Pharmaceutical University, Guangzhou 510006, China; School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| | - Zhi-You Yang
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China
| | - Shu-Mei Wang
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China; Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM, Guangdong Pharmaceutical University, Guangzhou 510006, China; Engineering & Technology Research Center for Chinese Materia Medica Quality of the Universities of Guangdong Province, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| | - Yue-Wei Ge
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China; Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM, Guangdong Pharmaceutical University, Guangzhou 510006, China; Engineering & Technology Research Center for Chinese Materia Medica Quality of the Universities of Guangdong Province, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| |
Collapse
|
24
|
Role of Extracellular Vimentin in Cancer-Cell Functionality and Its Influence on Cell Monolayer Permeability Changes Induced by SARS-CoV-2 Receptor Binding Domain. Int J Mol Sci 2021; 22:ijms22147469. [PMID: 34299089 PMCID: PMC8303762 DOI: 10.3390/ijms22147469] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 07/04/2021] [Accepted: 07/08/2021] [Indexed: 12/23/2022] Open
Abstract
The cytoskeletal protein vimentin is secreted under various physiological conditions. Extracellular vimentin exists primarily in two forms: attached to the outer cell surface and secreted into the extracellular space. While surface vimentin is involved in processes such as viral infections and cancer progression, secreted vimentin modulates inflammation through reduction of neutrophil infiltration, promotes bacterial elimination in activated macrophages, and supports axonal growth in astrocytes through activation of the IGF-1 receptor. This receptor is overexpressed in cancer cells, and its activation pathway has significant roles in general cellular functions. In this study, we investigated the functional role of extracellular vimentin in non-tumorigenic (MCF-10a) and cancer (MCF-7) cells through the evaluation of its effects on cell migration, proliferation, adhesion, and monolayer permeability. Upon treatment with extracellular recombinant vimentin, MCF-7 cells showed increased migration, proliferation, and adhesion, compared to MCF-10a cells. Further, MCF-7 monolayers showed reduced permeability, compared to MCF-10a monolayers. It has been shown that the receptor binding domain of SARS-CoV-2 spike protein can alter blood-brain barrier integrity. Surface vimentin also acts as a co-receptor between the SARS-CoV-2 spike protein and the cell-surface angiotensin-converting enzyme 2 receptor. Therefore, we also investigated the permeability of MCF-10a and MCF-7 monolayers upon treatment with extracellular recombinant vimentin, and its modulation of the SARS-CoV-2 receptor binding domain. These findings show that binding of extracellular recombinant vimentin to the cell surface enhances the permeability of both MCF-10a and MCF-7 monolayers. However, with SARS-CoV-2 receptor binding domain addition, this effect is lost with MCF-7 monolayers, as the extracellular vimentin binds directly to the viral domain. This defines an influence of extracellular vimentin in SARS-CoV-2 infections.
Collapse
|
25
|
Suprewicz Ł, Swoger M, Gupta S, Piktel E, Byfield FJ, Iwamoto DV, Germann D, Reszeć J, Marcińczyk N, Carroll RJ, Lenart M, Pyre K, Janmey P, Schwarz JM, Bucki R, Patteson A. Extracellular vimentin as a target against SARS-CoV-2 host cell invasion. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021:2021.01.08.425793. [PMID: 33442680 PMCID: PMC7805437 DOI: 10.1101/2021.01.08.425793] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Infection of human cells by pathogens, including SARS-CoV-2, typically proceeds by cell surface binding to a crucial receptor. In the case of SARS-CoV-2, angiotensin-converting enzyme 2 (ACE2) has been identified as a necessary receptor, but not all ACE2-expressing cells are equally infected, suggesting that other extracellular factors are involved in host cell invasion by SARS-CoV-2. Vimentin is an intermediate filament protein that is increasingly recognized as being present on the extracellular surface of a subset of cell types, where it can bind to and facilitate pathogens' cellular uptake. Here, we present evidence that extracellular vimentin might act as a critical component of the SARS-CoV-2 spike protein-ACE2 complex in mediating SARS-CoV-2 cell entry. We demonstrate direct binding between vimentin and SARS-CoV-2 pseudovirus coated with the SARS-CoV-2 spike protein and show that antibodies against vimentin block in vitro SARS-CoV-2 pseudovirus infection of ACE2-expressing cells. Our results suggest new therapeutic strategies for preventing and slowing SARS-CoV-2 infection, focusing on targeting cell host surface vimentin.
Collapse
Affiliation(s)
- Łukasz Suprewicz
- Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Białystok, Poland
| | - Maxx Swoger
- Physics Department and BioInspired Institute, Syracuse University
| | - Sarthak Gupta
- Physics Department and BioInspired Institute, Syracuse University
| | - Ewelina Piktel
- Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Białystok, Poland
| | - Fitzroy J Byfield
- Institute for Medicine and Engineering and Department of Physiology, University of Pennsylvania
| | - Daniel V Iwamoto
- Institute for Medicine and Engineering and Department of Physiology, University of Pennsylvania
| | - Danielle Germann
- Physics Department and BioInspired Institute, Syracuse University
| | - Joanna Reszeć
- Department of Medical Pathomorphology, Medical University of Białystok, PL-15269 Białystok, Poland
| | - Natalia Marcińczyk
- Department of Biopharmacy, Medical University of Białystok, Białystok, Poland
| | - Robert J Carroll
- Physics Department and BioInspired Institute, Syracuse University
| | - Marzena Lenart
- Małopolska Centre of Biotechnology; Jagiellonian University; Kraków, Poland
| | - Krzysztof Pyre
- Małopolska Centre of Biotechnology; Jagiellonian University; Kraków, Poland
| | - Paul Janmey
- Institute for Medicine and Engineering and Department of Physiology, University of Pennsylvania
| | - J M Schwarz
- Physics Department and BioInspired Institute, Syracuse University
- Indian Creek Farm, Ithaca, NY
| | - Robert Bucki
- Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Białystok, Poland
- Institute for Medicine and Engineering and Department of Physiology, University of Pennsylvania
| | - Alison Patteson
- Physics Department and BioInspired Institute, Syracuse University
| |
Collapse
|
26
|
Markasz L, Olsson KW, Holmström G, Sindelar R. Cluster Analysis of Early Postnatal Biochemical Markers May Predict Development of Retinopathy of Prematurity. Transl Vis Sci Technol 2020; 9:14. [PMID: 33344058 PMCID: PMC7726592 DOI: 10.1167/tvst.9.13.14] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 10/19/2020] [Indexed: 12/15/2022] Open
Abstract
Purpose Growth factors and inflammatory and angiogenetic proteins are involved in the development of retinopathy of prematurity (ROP). However, no early biochemical markers are in clinical use to predict ROP. By performing cluster analysis of multiple biomarkers, we aimed to determine patient groups with high and low risk for developing ROP. Methods In total, 202 protein markers in plasma were quantified by proximity extension assay from 35 extremely preterm infants on day 2 of life. Infants were sorted in groups by automated two-dimensional hierarchical clustering of all biomarkers. ROP was classified as stages I to III with or without surgical treatment. Predictive biomarkers were evaluated by analysis of variance and detected differences by two-sided paired t-test with Bonferroni corrections for multiple comparisons. Results Differences in 39 biochemical markers divided infants without ROP into two control groups (control 1, n = 7; control 2, n = 5; P < 0.05). Sixty-six biochemical markers defined differences between the control groups (n = 13) and all ROP infants (n = 23; P < 0.05). PARK7, VIM, MPO, CD69, and NEMO were markedly increased in control 1 compared to all ROP infants (P < 0.001). Lower TNFRSF4 and higher HER2 and GAL appeared in infants with ROP as compared to control 1 and/or 2 (P < 0.05, respectively). Conclusions Our data suggest that early elevated levels of PARK7, VIM, MPO, CD69, and NEMO may be associated with lower risk of developing ROP. Lower levels of TNFRSF4 with higher levels of HER2 and GAL may predict ROP development. Translational Relevance Cluster analysis of early postnatal biomarkers may help to identify infants with low or high risk of developing ROP.
Collapse
Affiliation(s)
- Laszlo Markasz
- Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden
| | - Karl-Wilhelm Olsson
- Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden.,Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Gerd Holmström
- Department of Neuroscience/Ophthalmology, Uppsala University, Uppsala, Sweden
| | - Richard Sindelar
- Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden
| |
Collapse
|
27
|
Adolf A, Turko P, Rohrbeck A, Just I, Vida I, Ahnert-Hilger G, Höltje M. The Higher Sensitivity of GABAergic Compared to Glutamatergic Neurons to Growth-Promoting C3bot Treatment Is Mediated by Vimentin. Front Cell Neurosci 2020; 14:596072. [PMID: 33240046 PMCID: PMC7669547 DOI: 10.3389/fncel.2020.596072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 10/06/2020] [Indexed: 11/16/2022] Open
Abstract
The current study investigates the neurotrophic effects of Clostridium botulinum C3 transferase (C3bot) on highly purified, glia-free, GABAergic, and glutamatergic neurons. Incubation with nanomolar concentrations of C3bot promotes dendrite formation as well as dendritic and axonal outgrowth in rat GABAergic neurons. A comparison of C3bot effects on sorted mouse GABAergic and glutamatergic neurons obtained from newly established NexCre;Ai9xVGAT Venus mice revealed a higher sensitivity of GABAergic cells to axonotrophic and dendritic effects of C3bot in terms of process length and branch formation. Protein biochemical analysis of known C3bot binding partners revealed comparable amounts of β1 integrin in both cell types but a higher expression of vimentin in GABAergic neurons. Accordingly, binding of C3bot to GABAergic neurons was stronger than binding to glutamatergic neurons. A combinatory treatment of glutamatergic neurons with C3bot and vimentin raised the amount of bound C3bot to levels comparable to the ones in GABAergic neurons, thereby confirming the specificity of effects. Overall, different surface vimentin levels between GABAergic and glutamatergic neurons exist that mediate neurotrophic C3bot effects.
Collapse
Affiliation(s)
- Andrej Adolf
- Institute of Integrative Neuroanatomy, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Paul Turko
- Institute of Integrative Neuroanatomy, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Astrid Rohrbeck
- Institute of Toxicology, Hannover Medical School (MHH), Hannover, Germany
| | - Ingo Just
- Institute of Toxicology, Hannover Medical School (MHH), Hannover, Germany
| | - Imre Vida
- Institute of Integrative Neuroanatomy, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Gudrun Ahnert-Hilger
- Institute of Integrative Neuroanatomy, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Markus Höltje
- Institute of Integrative Neuroanatomy, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
28
|
Patteson AE, Vahabikashi A, Goldman RD, Janmey PA. Mechanical and Non-Mechanical Functions of Filamentous and Non-Filamentous Vimentin. Bioessays 2020; 42:e2000078. [PMID: 32893352 PMCID: PMC8349470 DOI: 10.1002/bies.202000078] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 08/04/2020] [Indexed: 12/20/2022]
Abstract
Intermediate filaments (IFs) formed by vimentin are less understood than their cytoskeletal partners, microtubules and F-actin, but the unique physical properties of IFs, especially their resistance to large deformations, initially suggest a mechanical function. Indeed, vimentin IFs help regulate cell mechanics and contractility, and in crowded 3D environments they protect the nucleus during cell migration. Recently, a multitude of studies, often using genetic or proteomic screenings show that vimentin has many non-mechanical functions within and outside of cells. These include signaling roles in wound healing, lipogenesis, sterol processing, and various functions related to extracellular and cell surface vimentin. Extracellular vimentin is implicated in marking circulating tumor cells, promoting neural repair, and mediating the invasion of host cells by viruses, including SARS-CoV, or bacteria such as Listeria and Streptococcus. These findings underscore the fundamental role of vimentin in not only cell mechanics but also a range of physiological functions. Also see the video abstract here https://youtu.be/YPfoddqvz-g.
Collapse
Affiliation(s)
- Alison E Patteson
- Physics Department, Syracuse University, Syracuse, NY 13244
- BioInspired Institute, Syracuse University, Syracuse, NY 13244
| | - Amir Vahabikashi
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago IL 60611
| | - Robert D Goldman
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago IL 60611
| | - Paul A. Janmey
- Institute for Medicine and Engineering, Department of Physiology, University of Pennsylvania, Philadelphia, PA 19104
| |
Collapse
|
29
|
Ma Z, Cai Y, Zhang L, Tian C, Lyu L. LINC00319 Promotes Cervical Cancer Progression Via Targeting miR-147a/IGF1R Pathway. Cancer Biother Radiopharm 2020:cbr.2020.3722. [PMID: 32644822 DOI: 10.1089/cbr.2020.3722] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Background: Cervical cancer is identified as the fourth most common female malignancy worldwide. Recently, Linc00319 was reported to play an important role in the development and progression of cervical cancer. However, little is known about the molecular mechanism and clinical significance of Linc00319 in the carcinogenesis of cervical cancer. This study aims to reveal the biological function and molecular mechanisms of Linc00319 in cell proliferation, invasion, and migration of cervical cancer. Materials and Methods: In the current study, gene expression levels of Linc00319, miR-147a, and IFG1R were detected by quantitative real-time PCR in clinical tissue samples and cervical cancer cell lines. Protein levels were also determined by western blot assay in cervical cancer cells. CCK-8, transwell, and wound healing assays were used to test the proliferation, invasion, and migration of cervical cancer cell lines in vitro. Target genes were predicted through bioinformatics methods and then verified by gene engineering technology. Results: The authors' results showed that Linc00319 was upregulated in cervical cancer tissues and cell lines, while Linc00319silencing could inhibit cervical cancer cell proliferation, invasion, and migration. Further investigations showed that Linc00319 interacted with miR-147a and inhibited its expression, unregulated IGF1R to induce progression of cervical cancer. Conclusions: Their research indicated that Linc00319 might play an oncogenic role in cervical cancer and regulate the progression of cervical tumor growth by inhibiting the expression of miR-147a and activating IGF1R-related pathway. The findings suggest a novel molecular biomarker and therapeutic target for cervical tumor and may provide a novel therapeutic strategy for preventing the metastasis of cervical cancer.
Collapse
Affiliation(s)
- Zhe Ma
- Department of Obstetrics and Gynecology, Affiliated Hospital of Beihua University, Jilin City, China
| | - Yufei Cai
- Department of Obstetrics and Gynecology, Affiliated Hospital of Beihua University, Jilin City, China
| | - Limei Zhang
- Department of Obstetrics and Gynecology, Affiliated Hospital of Beihua University, Jilin City, China
| | - Chenchen Tian
- Department of Obstetrics and Gynecology, Affiliated Hospital of Beihua University, Jilin City, China
| | - Lin Lyu
- Department of Gynecology, The First Affiliated Hospital of Chengdu Medical College, Chengdu City, China
| |
Collapse
|
30
|
Ramos I, Stamatakis K, Oeste CL, Pérez-Sala D. Vimentin as a Multifaceted Player and Potential Therapeutic Target in Viral Infections. Int J Mol Sci 2020; 21:E4675. [PMID: 32630064 PMCID: PMC7370124 DOI: 10.3390/ijms21134675] [Citation(s) in RCA: 118] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Revised: 06/28/2020] [Accepted: 06/29/2020] [Indexed: 12/17/2022] Open
Abstract
Vimentin is an intermediate filament protein that plays key roles in integration of cytoskeletal functions, and therefore in basic cellular processes such as cell division and migration. Consequently, vimentin has complex implications in pathophysiology. Vimentin is required for a proper immune response, but it can also act as an autoantigen in autoimmune diseases or as a damage signal. Although vimentin is a predominantly cytoplasmic protein, it can also appear at extracellular locations, either in a secreted form or at the surface of numerous cell types, often in relation to cell activation, inflammation, injury or senescence. Cell surface targeting of vimentin appears to associate with the occurrence of certain posttranslational modifications, such as phosphorylation and/or oxidative damage. At the cell surface, vimentin can act as a receptor for bacterial and viral pathogens. Indeed, vimentin has been shown to play important roles in virus attachment and entry of severe acute respiratory syndrome-related coronavirus (SARS-CoV), dengue and encephalitis viruses, among others. Moreover, the presence of vimentin in specific virus-targeted cells and its induction by proinflammatory cytokines and tissue damage contribute to its implication in viral infection. Here, we recapitulate some of the pathophysiological implications of vimentin, including the involvement of cell surface vimentin in interaction with pathogens, with a special focus on its role as a cellular receptor or co-receptor for viruses. In addition, we provide a perspective on approaches to target vimentin, including antibodies or chemical agents that could modulate these interactions to potentially interfere with viral pathogenesis, which could be useful when multi-target antiviral strategies are needed.
Collapse
Affiliation(s)
- Irene Ramos
- Department of Neurology and Center for Advanced Research on Diagnostic Assays, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
| | - Konstantinos Stamatakis
- Centro de Biología Molecular Severo Ochoa, UAM-CSIC. Nicolás Cabrera, 1, Campus de la Universidad Autónoma de Madrid, 28049 Madrid, Spain; (K.S.); (C.L.O.)
| | - Clara L. Oeste
- Centro de Biología Molecular Severo Ochoa, UAM-CSIC. Nicolás Cabrera, 1, Campus de la Universidad Autónoma de Madrid, 28049 Madrid, Spain; (K.S.); (C.L.O.)
| | - Dolores Pérez-Sala
- Department of Structural and Chemical Biology, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Ramiro de Maeztu, 9, 28040 Madrid, Spain
| |
Collapse
|
31
|
Mak HK, Yung JSY, Weinreb RN, Ng SH, Cao X, Ho TYC, Ng TK, Chu WK, Yung WH, Choy KW, Wang CC, Lee TL, Leung CKS. MicroRNA-19a-PTEN Axis Is Involved in the Developmental Decline of Axon Regenerative Capacity in Retinal Ganglion Cells. MOLECULAR THERAPY. NUCLEIC ACIDS 2020; 21:251-263. [PMID: 32599451 PMCID: PMC7327411 DOI: 10.1016/j.omtn.2020.05.031] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 04/15/2020] [Accepted: 05/28/2020] [Indexed: 11/23/2022]
Abstract
Irreversible blindness from glaucoma and optic neuropathies is attributed to retinal ganglion cells (RGCs) losing the ability to regenerate axons. While several transcription factors and proteins have demonstrated enhancement of axon regeneration after optic nerve injury, mechanisms contributing to the age-related decline in axon regenerative capacity remain elusive. In this study, we show that microRNAs are differentially expressed during RGC development and identify microRNA-19a (miR-19a) as a heterochronic marker; developmental decline of miR-19a relieves suppression of phosphatase and tensin homolog (PTEN), a key regulator of axon regeneration, and serves as a temporal indicator of decreasing axon regenerative capacity. Intravitreal injection of miR-19a promotes axon regeneration after optic nerve crush in adult mice, and it increases axon extension in RGCs isolated from aged human donors. This study uncovers a previously unrecognized involvement of the miR-19a-PTEN axis in RGC axon regeneration, and it demonstrates therapeutic potential of microRNA-mediated restoration of axon regenerative capacity in optic neuropathies.
Collapse
Affiliation(s)
- Heather K Mak
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, PRC
| | - Jasmine S Y Yung
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, PRC
| | - Robert N Weinreb
- Hamilton Glaucoma Center, Shiley Eye Institute, University of California, San Diego, La Jolla, CA, USA; Department of Ophthalmology, University of California, San Diego, La Jolla, CA, USA
| | - Shuk Han Ng
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, PRC
| | - Xu Cao
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, PRC
| | - Tracy Y C Ho
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, PRC
| | - Tsz Kin Ng
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, PRC
| | - Wai Kit Chu
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, PRC
| | - Wing Ho Yung
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, PRC; Gerald Choa Neuroscience Centre, The Chinese University of Hong Kong, Hong Kong, PRC
| | - Kwong Wai Choy
- Department of Obstetrics and Gynecology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, PRC
| | - Chi Chiu Wang
- Department of Obstetrics and Gynecology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, PRC
| | - Tin Lap Lee
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, PRC
| | | |
Collapse
|
32
|
Strouhalova K, Přechová M, Gandalovičová A, Brábek J, Gregor M, Rosel D. Vimentin Intermediate Filaments as Potential Target for Cancer Treatment. Cancers (Basel) 2020; 12:E184. [PMID: 31940801 PMCID: PMC7017239 DOI: 10.3390/cancers12010184] [Citation(s) in RCA: 164] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 01/08/2020] [Accepted: 01/09/2020] [Indexed: 02/06/2023] Open
Abstract
Intermediate filaments constitute the third component of the cellular skeleton. Unlike actin and microtubule cytoskeletons, the intermediate filaments are composed of a wide variety of structurally related proteins showing distinct expression patterns in tissues and cell types. Changes in the expression patterns of intermediate filaments are often associated with cancer progression; in particular with phenotypes leading to increased cellular migration and invasion. In this review we will describe the role of vimentin intermediate filaments in cancer cell migration, cell adhesion structures, and metastasis formation. The potential for targeting vimentin in cancer treatment and the development of drugs targeting vimentin will be reviewed.
Collapse
Affiliation(s)
- Katerina Strouhalova
- Department of Cell Biology, Charles University, Viničná 7, 12843 Prague, Czech Republic; (K.S.); (A.G.); (J.B.)
- Biotechnology and Biomedicine Centre of the Academy of Sciences and Charles University (BIOCEV), Průmyslová 595, 25242 Vestec u Prahy, Czech Republic
| | - Magdalena Přechová
- Laboratory of Integrative Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, 14220 Prague, Czech Republic;
| | - Aneta Gandalovičová
- Department of Cell Biology, Charles University, Viničná 7, 12843 Prague, Czech Republic; (K.S.); (A.G.); (J.B.)
- Biotechnology and Biomedicine Centre of the Academy of Sciences and Charles University (BIOCEV), Průmyslová 595, 25242 Vestec u Prahy, Czech Republic
| | - Jan Brábek
- Department of Cell Biology, Charles University, Viničná 7, 12843 Prague, Czech Republic; (K.S.); (A.G.); (J.B.)
- Biotechnology and Biomedicine Centre of the Academy of Sciences and Charles University (BIOCEV), Průmyslová 595, 25242 Vestec u Prahy, Czech Republic
| | - Martin Gregor
- Laboratory of Integrative Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, 14220 Prague, Czech Republic;
| | - Daniel Rosel
- Department of Cell Biology, Charles University, Viničná 7, 12843 Prague, Czech Republic; (K.S.); (A.G.); (J.B.)
- Biotechnology and Biomedicine Centre of the Academy of Sciences and Charles University (BIOCEV), Průmyslová 595, 25242 Vestec u Prahy, Czech Republic
| |
Collapse
|
33
|
Nissen NI, Karsdal M, Willumsen N. Post-translational modifications of vimentin reflect different pathological processes associated with non-small cell lung cancer and chronic obstructive pulmonary disease. Oncotarget 2019; 10:6829-6841. [PMID: 31827725 PMCID: PMC6887574 DOI: 10.18632/oncotarget.27332] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 11/06/2019] [Indexed: 12/13/2022] Open
Abstract
INTRODUCTION Vimentin has shown to be highly implicated in cancer initiation and progression. Vimentin is often a target of post-translational modifications (PTMs) which can be disease specific, thus targeting these specific modifications can be of high biomarker potential. In this study we set out to evaluate the biological relevance and serum biomarker potential of citrullinated vimentin (VICM) and non-citrullinated vimentin (VIM) in non-small cell lung cancer (NSCLC) and chronic obstructive pulmonary disease (COPD). METHODS A competitive ELISA targeting VIM was developed and quantified in serum from patients with NSCLC and COPD. VIM was compared with levels of VICM in the same indications. RESULTS VIM was significantly increased in NSCLC (n = 100) compared to healthy controls (n = 67) in two independent cohorts (p = 0.0003 and p < 0.0001). Furthermore, VIM was highly increased in late stages of NSCLC (p = 0.001), however VIM was not increased in COPD patients (n = 10). Contrarily, serum levels of VICM was not increased in late stages of NSCLC, but highly elevated in patients with COPD (p < 0.0001). CONCLUSIONS These findings suggest a biomarker potential of VIM in NSCLC. Our findings also indicate that PTMs of vimentin are highly relevant and that targeting these modifications can have differential biomarker potential.
Collapse
Affiliation(s)
- Neel Ingemann Nissen
- Biotech Research & Innovation Centre (BRIC), University of Copenhagen, DK-2200 Copenhagen, Denmark.,Nordic Bioscience, Biomarkers and Research, DK-2730 Herlev, Denmark
| | - Morten Karsdal
- Nordic Bioscience, Biomarkers and Research, DK-2730 Herlev, Denmark
| | | |
Collapse
|
34
|
Zhao XM, He XY, Liu J, Xu Y, Xu FF, Tan YX, Zhang ZB, Wang TH. Neural Stem Cell Transplantation Improves Locomotor Function in Spinal Cord Transection Rats Associated with Nerve Regeneration and IGF-1 R Expression. Cell Transplant 2019; 28:1197-1211. [PMID: 31271053 PMCID: PMC6767897 DOI: 10.1177/0963689719860128] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Transplantation of neural stem cells (NSCs) is a potential strategy for the treatment of
spinal cord transection (SCT). Here we investigated whether transplanted NSCs would
improve motor function of rats with SCT and explored the underlying mechanism. First, the
rats were divided into sham, SCT, and NSC groups. Rats in the SCT and NSC groups were all
subjected to SCT in T10, and were administered with media and NSC transplantation into the
lesion site, respectively. Immunohistochemistry was used to label Nestin-, TUNEL-, and
NeuN-positive cells and reveal the expression and location of type I insulin-like growth
factor receptor (IGF-1 R). Locomotor function of hind limbs was assessed by Basso,
Beattie, Bresnahan (BBB) score and inclined plane test. The conduction velocity and
amplitude of spinal nerve fibers were measured by electrophysiology and the anatomical
changes were measured using magnetic resonance imaging. Moreover, expression of IGF-1 R
was determined by real-time polymerase chain reaction and Western blotting. The results
showed that NSCs could survive and differentiate into neurons in vitro and in vivo.
SCT-induced deficits were reduced by NSC transplantation, including increase in
NeuN-positive cells and decrease in apoptotic cells. Moreover, neurophysiological profiles
indicated that the latent period was decreased and the peak-to-peak amplitude of spinal
nerve fibers conduction was increased in transplanted rats, while morphological measures
indicated that fractional anisotropy and the number of nerve fibers in the site of spinal
cord injury were increased after NSC transplantation. In addition, mRNA and protein level
of IGF-1 R were increased in the rostral segment in the NSC group, especially in neurons.
Therefore, we concluded that NSC transplantation promotes motor function improvement of
SCT, which might be associated with activated IGF-1 R, especially in the rostral site. All
of the above suggests that this approach has potential for clinical treatment of spinal
cord injury.
Collapse
Affiliation(s)
- Xiao-Ming Zhao
- Department of Histology, Embryology and Neurobiology, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu, China.,Both the author contributed equally to this article
| | - Xiu-Ying He
- Institute of Neurological Disease, Department of Anesthesiology, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, China.,Both the author contributed equally to this article
| | - Jia Liu
- Laboratory Zoology Department, Institute of Neuroscience, Kunming Medical University, Kunming, China
| | - Yang Xu
- Institute of Neurological Disease, Department of Anesthesiology, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, China
| | - Fei-Fei Xu
- Institute of Neurological Disease, Department of Anesthesiology, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, China
| | - Ya-Xin Tan
- Laboratory Zoology Department, Institute of Neuroscience, Kunming Medical University, Kunming, China
| | - Zi-Bin Zhang
- Department of Anesthesiology, Qilu Hospital of Shandong University, Jinan, China
| | - Ting-Hua Wang
- Department of Histology, Embryology and Neurobiology, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu, China.,Institute of Neurological Disease, Department of Anesthesiology, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, China.,Laboratory Zoology Department, Institute of Neuroscience, Kunming Medical University, Kunming, China
| |
Collapse
|
35
|
Krishnamurthy A, Ytterberg AJ, Sun M, Sakuraba K, Steen J, Joshua V, Tarasova NK, Malmström V, Wähämaa H, Réthi B, Catrina AI. Citrullination Controls Dendritic Cell Transdifferentiation into Osteoclasts. THE JOURNAL OF IMMUNOLOGY 2019; 202:3143-3150. [PMID: 31019059 DOI: 10.4049/jimmunol.1800534] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 03/21/2019] [Indexed: 11/19/2022]
Abstract
An increased repertoire of potential osteoclast (OC) precursors could accelerate the development of bone-erosive OCs and the consequent bone damage in rheumatoid arthritis (RA). Immature dendritic cells (DCs) can develop into OCs, however, the mechanisms underlying this differentiation switch are poorly understood. We investigated whether protein citrullination and RA-specific anti-citrullinated protein Abs (ACPAs) could regulate human blood-derived DC-OC transdifferentiation. We show that plasticity toward the OC lineage correlated with peptidyl arginine deiminase (PAD) activity and protein citrullination in DCs. Citrullinated actin and vimentin were present in DCs and DC-derived OCs, and both proteins were deposited on the cell surface, colocalizing with ACPAs binding to the cells. ACPAs enhanced OC differentiation from monocyte-derived or circulating CD1c+ DCs by increasing the release of IL-8. Blocking IL-8 binding or the PAD enzymes completely abolished the stimulatory effect of ACPAs, whereas PAD inhibition reduced steady-state OC development, as well, suggesting an essential role for protein citrullination in DC-OC transdifferentiation. Protein citrullination and ACPA binding to immature DCs might thus promote differentiation plasticity toward the OC lineage, which can facilitate bone erosion in ACPA-positive RA.
Collapse
Affiliation(s)
- Akilan Krishnamurthy
- Rheumatology Unit, Karolinska University Hospital, Karolinska Institutet, S-171 76 Stockholm, Sweden
| | - A Jimmy Ytterberg
- Rheumatology Unit, Karolinska University Hospital, Karolinska Institutet, S-171 76 Stockholm, Sweden.,Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE-171 77 Stockholm, Sweden; and
| | - Meng Sun
- Rheumatology Unit, Karolinska University Hospital, Karolinska Institutet, S-171 76 Stockholm, Sweden
| | - Koji Sakuraba
- Rheumatology Unit, Karolinska University Hospital, Karolinska Institutet, S-171 76 Stockholm, Sweden.,Clinical Research Institute, National Hospital Organisation, Kyushu Medical Center, Fukuoka 810-8563, Japan
| | - Johanna Steen
- Rheumatology Unit, Karolinska University Hospital, Karolinska Institutet, S-171 76 Stockholm, Sweden
| | - Vijay Joshua
- Rheumatology Unit, Karolinska University Hospital, Karolinska Institutet, S-171 76 Stockholm, Sweden
| | - Nataliya K Tarasova
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE-171 77 Stockholm, Sweden; and
| | - Vivianne Malmström
- Rheumatology Unit, Karolinska University Hospital, Karolinska Institutet, S-171 76 Stockholm, Sweden
| | - Heidi Wähämaa
- Rheumatology Unit, Karolinska University Hospital, Karolinska Institutet, S-171 76 Stockholm, Sweden
| | - Bence Réthi
- Rheumatology Unit, Karolinska University Hospital, Karolinska Institutet, S-171 76 Stockholm, Sweden;
| | - Anca I Catrina
- Rheumatology Unit, Karolinska University Hospital, Karolinska Institutet, S-171 76 Stockholm, Sweden
| |
Collapse
|
36
|
Dutta S, Sinha A, Dasgupta S, Mukherjee AK. Binding of a Naja naja venom acidic phospholipase A 2 cognate complex to membrane-bound vimentin of rat L6 cells: Implications in cobra venom-induced cytotoxicity. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2019; 1861:958-977. [PMID: 30776333 DOI: 10.1016/j.bbamem.2019.02.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Revised: 01/25/2019] [Accepted: 02/05/2019] [Indexed: 01/28/2023]
Abstract
An acidic phospholipase A2 enzyme (NnPLA2-I) interacts with three finger toxins (cytotoxin and neurotoxin) from Naja naja venom to form cognate complexes to enhance its cytotoxicity towards rat L6 myogenic cells. The cytotoxicity was further enhanced in presence of trace quantity of venom nerve growth factor. The purified rat myoblast cell membrane protein showing interaction with NnPLA2-I was identified as vimentin by LC-MS/MS analysis. The ELISA, immunoblot and spectrofluorometric analyses showed greater binding of NnPLA2-I cognate complex to vimentin as compared to the binding of individual NnPLA2-I. The immunofluorescence and confocal microscopy studies evidenced the internalization of NnPLA2-I to partially differentiated myoblasts post binding with vimentin in a time-dependent manner. Pre-incubation of polyvalent antivenom with NnPLA2-I cognate complex demonstrated better neutralization of cytotoxicity towards L6 cells as compared to exogenous addition of polyvalent antivenom 60-240 min post treatment of L6 cells with cognate complex suggesting clinical advantage of early antivenom treatment to prevent cobra venom-induced cytotoxicity. The in silico analysis showed that 19-22 residues, inclusive of Asp48 residue, of NnPLA2-I preferentially binds with the rod domain (99-189 and 261-335 regions) of vimentin with a predicted free binding energy (ΔG) and dissociation constant (KD) values of -12.86 kcal/mol and 3.67 × 10-10 M, respectively; however, NnPLA2-I cognate complex showed greater binding with the same regions of vimentin indicating the pathophysiological significance of cognate complex in cobra venom-induced cytotoxicity.
Collapse
Affiliation(s)
- Sumita Dutta
- Microbial Biotechnology and Protein Research Laboratory, Department of Molecular Biology and Biotechnology, Tezpur University, Tezpur 784028, Assam, India
| | - Archana Sinha
- Molecular Endocrinology and Metabolism Laboratory, Department of Molecular Biology and Biotechnology, Tezpur University, Tezpur 784028, Assam, India
| | - Suman Dasgupta
- Molecular Endocrinology and Metabolism Laboratory, Department of Molecular Biology and Biotechnology, Tezpur University, Tezpur 784028, Assam, India
| | - Ashis K Mukherjee
- Microbial Biotechnology and Protein Research Laboratory, Department of Molecular Biology and Biotechnology, Tezpur University, Tezpur 784028, Assam, India.
| |
Collapse
|
37
|
Baud A, Little D, Wen TQ, Heywood WE, Gissen P, Mills K. An Optimized Method for the Proteomic Analysis of Low Volumes of Cell Culture Media and the Secretome: The Application and the Demonstration of Altered Protein Expression in iPSC-Derived Neuronal Cell Lines from Parkinson's Disease Patients. J Proteome Res 2019; 18:1198-1207. [PMID: 30562036 DOI: 10.1021/acs.jproteome.8b00831] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Traditionally, cell culture medium in iPSC-derived cell work is not the main focus of the research and often is considered as just "food for cells". We demonstrate that by manipulation of the media and optimized methodology, it is possible to use this solution to study the proteins that the cell secretes (the "secretome"). This is particularly useful in the study of iPSC-derived neurons, which require long culture time. We demonstrate that media can be used to model diseases with optimized incubation and sampling times. The ability not to sacrifice cells allows significant cost and research benefits. In this manuscript we describe an optimized method for the analysis of the cell media from iPSC-derived neuronal lines from control and Parkinson's disease patients. We have evaluated the use of standard and supplement B27-free cell media as well as five different sample preparation techniques for proteomic analysis of the cell secretome. Mass spectral analysis of culture media allowed for the identification of >500 proteins, in 500 μL of media, which is less volume than reported previously (20-40 mL). Using shorter incubation times and our optimized methodology, we describe the use of this technique to study and describe potential disease mechanisms in Parkinson's disease.
Collapse
Affiliation(s)
- Anna Baud
- Centre for Translational Omics , UCL Great Ormond Street Institute of Child Health , London , WC1N 1EH , U.K
| | - Daniel Little
- MRC Laboratory for Molecular Cell Biology , University College London , London , WC1E 6BT , U.K
| | - Teo Qi Wen
- Centre for Translational Omics , UCL Great Ormond Street Institute of Child Health , London , WC1N 1EH , U.K
| | - Wendy E Heywood
- Centre for Translational Omics , UCL Great Ormond Street Institute of Child Health , London , WC1N 1EH , U.K
| | - Paul Gissen
- MRC Laboratory for Molecular Cell Biology , University College London , London , WC1E 6BT , U.K
| | - Kevin Mills
- Centre for Translational Omics , UCL Great Ormond Street Institute of Child Health , London , WC1N 1EH , U.K
| |
Collapse
|
38
|
Adolf A, Rohrbeck A, Münster-Wandowski A, Johansson M, Kuhn HG, Kopp MA, Brommer B, Schwab JM, Just I, Ahnert-Hilger G, Höltje M. Release of astroglial vimentin by extracellular vesicles: Modulation of binding and internalization of C3 transferase in astrocytes and neurons. Glia 2018; 67:703-717. [PMID: 30485542 DOI: 10.1002/glia.23566] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 10/15/2018] [Accepted: 10/19/2018] [Indexed: 12/24/2022]
Abstract
Clostridium botulinum C3 transferase (C3bot) ADP-ribosylates rho proteins to change cellular functions in a variety of cell types including astrocytes and neurons. The intermediate filament protein vimentin as well as transmembrane integrins are involved in internalization of C3bot into cells. The exact contribution, however, of these proteins to binding of C3bot to the cell surface and subsequent cellular uptake remains to be unraveled. By comparing primary astrocyte cultures derived from wild-type with Vim-/- mice, we demonstrate that astrocytes lacking vimentin exhibited a delayed ADP-ribosylation of rhoA concurrent with a blunted morphological response. This functional impairment was rescued by the extracellular excess of recombinant vimentin. Binding assays using C3bot harboring a mutated integrin-binding RGD motif (C3bot-G89I) revealed the involvement of integrins in astrocyte binding of C3bot. Axonotrophic effects of C3bot are vimentin dependent and postulate an underlying mechanism entertaining a molecular cross-talk between astrocytes and neurons. We present functional evidence for astrocytic release of vimentin by exosomes using an in vitro scratch wound model. Exosomal vimentin+ particles released from wild-type astrocytes promote the interaction of C3bot with neuronal membranes. This effect vanished when culturing Vim-/- astrocytes. Specificity of these findings was confirmed by recombinant vimentin propagating enhanced binding of C3bot to synaptosomes from rat spinal cord and mouse brain. We hypothesize that vimentin+ exosomes released by reactive astrocytes provide a novel molecular mechanism constituting axonotrophic (neuroprotective) and plasticity augmenting effects of C3bot after spinal cord injury.
Collapse
Affiliation(s)
- Andrej Adolf
- Institute of Integrative Neuroanatomy, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Astrid Rohrbeck
- Institute of Toxicology, Hannover Medical School (MHH), Hanover, Germany
| | - Agnieszka Münster-Wandowski
- Institute of Integrative Neuroanatomy, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Malin Johansson
- Department of Clinical Neuroscience at Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| | - Hans-Georg Kuhn
- Department of Clinical Neuroscience at Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden.,Department of Neurology and Experimental Neurology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Marcel Alexander Kopp
- Department of Neurology and Experimental Neurology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Berlin Institute of Health, QUEST - Center for Transforming Biomedical Research, Berlin, Germany
| | - Benedikt Brommer
- Department of Neurology and Experimental Neurology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Jan Markus Schwab
- Department of Neurology and Experimental Neurology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Paraplegiology (Spinal Cord Injury Division), Belford Spinal Cord Injury Center, Departments of Neurology, Neuroscience and Physical Medicine and Rehabilitation, The Neurological Institute, The Ohio State University, Wexner Medical Center, Columbus, Ohio
| | - Ingo Just
- Institute of Toxicology, Hannover Medical School (MHH), Hanover, Germany
| | - Gudrun Ahnert-Hilger
- Institute of Integrative Neuroanatomy, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Markus Höltje
- Institute of Integrative Neuroanatomy, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
39
|
Yu MB, Guerra J, Firek A, Langridge WHR. Extracellular vimentin modulates human dendritic cell activation. Mol Immunol 2018; 104:37-46. [PMID: 30399492 DOI: 10.1016/j.molimm.2018.09.017] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Revised: 08/04/2018] [Accepted: 09/25/2018] [Indexed: 12/13/2022]
Abstract
Vimentin is an intermediate filament protein traditionally considered to be an intracellular protein with a structural role. However, recent evidence suggests that vimentin can also be found outside the cell in disease conditions such as cancer, traumatic tissue injury, and inflammation. Extracellular vimentin was previously found to stimulate innate immunity by increasing monocyte and macrophage ability to kill bacteria. However, vimentin has also been previously found to decrease neutrophil infiltration into inflamed tissue. How extracellular vimentin affects the initiation of adaptive immune responses is unknown. Initiation of adaptive immunity involves priming of naïve T cells by antigen-presenting cells, the most effective of which are dendritic cells (DCs). In this study, we demonstrate how extracellular vimentin modulates lipopolysaccharide (LPS) - induced activation of human DCs. Using cytometric bead arrays, we show that extracellular vimentin decreases LPS-activated DC secretion of pro-inflammatory cytokines IL-6 and IL-12 while increasing secretion of the anti-inflammatory cytokine IL-10. Using flow cytometry, we show that extracellular vimentin does not significantly affect LPS-induced DC surface expression of MHC I (HLA-ABC) or MHC II (HLA-DR) presentation molecules, costimulatory factors (CD80, CD86), or the DC maturation marker (CD83). Further, LPS-stimulated DCs co-cultured with allogeneic naïve CD4 + T cells (Th0) induced less secretion of the pro-inflammatory Th1 effector cytokine IFN-γ in the presence of vimentin than in the presence of LPS alone. This result suggests that vimentin reduces Th1 differentiation. Taken together, our data suggest that extracellular vimentin may inhibit pro-inflammatory adaptive immune responses, by blocking DC secretion of pro-inflammatory cytokines. Thus, extracellular vimentin may play an important role in cancer or trauma-complications by inducing suppression of the adaptive immune response. In a positive sense, the presence of extracellular vimentin may prevent tissue-damage from contributing to the development of autoimmunity. Consequently, extracellular vimentin may become a novel drug target for treatment of a variety of pro- and anti-inflammatory disease conditions.
Collapse
Affiliation(s)
- Mary Beth Yu
- Center for Health Disparities and Molecular Medicine, Department of Basic Sciences, Loma Linda, University School of Medicine, Loma Linda, CA, 92354, USA; Division of Biochemistry, Department of Basic Sciences, Loma Linda, University School of Medicine, Loma Linda, CA, 92354, USA
| | - Joshua Guerra
- Center for Health Disparities and Molecular Medicine, Department of Basic Sciences, Loma Linda, University School of Medicine, Loma Linda, CA, 92354, USA; University of Texas at San Antonio, San Antonio, TX, USA
| | - Anthony Firek
- Section of Endocrinology, Riverside University Health System Medical Center, Moreno Valley, CA, USA
| | - William H R Langridge
- Center for Health Disparities and Molecular Medicine, Department of Basic Sciences, Loma Linda, University School of Medicine, Loma Linda, CA, 92354, USA; Division of Biochemistry, Department of Basic Sciences, Loma Linda, University School of Medicine, Loma Linda, CA, 92354, USA.
| |
Collapse
|
40
|
Tanie Y, Tanabe N, Kuboyama T, Tohda C. Extracellular Neuroleukin Enhances Neuroleukin Secretion From Astrocytes and Promotes Axonal Growth in vitro and in vivo. Front Pharmacol 2018; 9:1228. [PMID: 30459611 PMCID: PMC6232869 DOI: 10.3389/fphar.2018.01228] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 10/08/2018] [Indexed: 12/23/2022] Open
Abstract
Under pathological conditions in the central nervous system (CNS), including spinal cord injury, astrocytes show detrimental effects against neurons. It is also known that astrocytes sometimes exert beneficial effects, such as neuroprotection and secretion of axonal growth factors. If beneficial effects of astrocytes after injury could be induced, dysfunction of the injured CNS may improve. However, a way of promoting beneficial functions in astrocytes has not been elucidated. In the current study, we focused on neuroleukin (NLK), which is known to have axonal growth activities in neurons. Although NLK is secreted from astrocytes, the function of NLK in astrocytes is poorly understood. We aimed to clarify the mechanism of NLK secretion in astrocytes and the functional significance of secreted NLK from astrocytes. Stimulation of cultured astrocytes with recombinant NLK significantly elevated the secretion of NLK from astrocytes. Furthermore, astrocyte conditioned medium treated with NLK increased axonal density in cultured cortical neurons. Recombinant NLK itself directly increased axonal density in cultured neurons. These results indicated that NLK secreted from astrocytes acted as an axonal growth factor and that secretion was stimulated by extracellular NLK. To elucidate a direct binding molecule of NLK on astrocytes, drug affinity responsive target stability (DARTS) analysis was performed. A 78 kDa glucose regulated protein (GRP78) was identified as a receptor for NLK, which was related to the secretion of NLK from astrocytes. When NLK was injected into the lesion site of spinal cord injured mice, axonal density in the injured region was significantly increased and hindlimb motor function improved. These results suggested that NLK-GRP78 signalling was important for the beneficial effects of astrocytes. This study strengthens the potential of astrocytes for use as therapeutic targets in CNS traumatic injury.
Collapse
Affiliation(s)
- Yoshitaka Tanie
- Division of Neuromedical Science, Department of Bioscience, Institute of Natural Medicine, University of Toyama, Toyama, Japan
| | - Norio Tanabe
- Division of Neuromedical Science, Department of Bioscience, Institute of Natural Medicine, University of Toyama, Toyama, Japan
| | - Tomoharu Kuboyama
- Division of Neuromedical Science, Department of Bioscience, Institute of Natural Medicine, University of Toyama, Toyama, Japan
| | - Chihiro Tohda
- Division of Neuromedical Science, Department of Bioscience, Institute of Natural Medicine, University of Toyama, Toyama, Japan
| |
Collapse
|
41
|
Danielsson F, Peterson MK, Caldeira Araújo H, Lautenschläger F, Gad AKB. Vimentin Diversity in Health and Disease. Cells 2018; 7:E147. [PMID: 30248895 PMCID: PMC6210396 DOI: 10.3390/cells7100147] [Citation(s) in RCA: 180] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 09/16/2018] [Accepted: 09/17/2018] [Indexed: 12/11/2022] Open
Abstract
Vimentin is a protein that has been linked to a large variety of pathophysiological conditions, including cataracts, Crohn's disease, rheumatoid arthritis, HIV and cancer. Vimentin has also been shown to regulate a wide spectrum of basic cellular functions. In cells, vimentin assembles into a network of filaments that spans the cytoplasm. It can also be found in smaller, non-filamentous forms that can localise both within cells and within the extracellular microenvironment. The vimentin structure can be altered by subunit exchange, cleavage into different sizes, re-annealing, post-translational modifications and interacting proteins. Together with the observation that different domains of vimentin might have evolved under different selection pressures that defined distinct biological functions for different parts of the protein, the many diverse variants of vimentin might be the cause of its functional diversity. A number of review articles have focussed on the biology and medical aspects of intermediate filament proteins without particular commitment to vimentin, and other reviews have focussed on intermediate filaments in an in vitro context. In contrast, the present review focusses almost exclusively on vimentin, and covers both ex vivo and in vivo data from tissue culture and from living organisms, including a summary of the many phenotypes of vimentin knockout animals. Our aim is to provide a comprehensive overview of the current understanding of the many diverse aspects of vimentin, from biochemical, mechanical, cellular, systems biology and medical perspectives.
Collapse
Affiliation(s)
- Frida Danielsson
- Science for Life Laboratory, Royal Institute of Technology, 17165 Stockholm, Sweden.
| | | | | | - Franziska Lautenschläger
- Campus D2 2, Leibniz-Institut für Neue Materialien gGmbH (INM) and Experimental Physics, NT Faculty, E 2 6, Saarland University, 66123 Saarbrücken, Germany.
| | - Annica Karin Britt Gad
- Centro de Química da Madeira, Universidade da Madeira, 9020105 Funchal, Portugal.
- Department of Medical Biochemistry and Microbiology, Uppsala University, 75237 Uppsala, Sweden.
| |
Collapse
|
42
|
Lin YP, Wu JI, Tseng CW, Chen HJ, Wang LH. Gjb4 serves as a novel biomarker for lung cancer and promotes metastasis and chemoresistance via Src activation. Oncogene 2018; 38:822-837. [PMID: 30177841 DOI: 10.1038/s41388-018-0471-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 06/11/2018] [Accepted: 07/03/2018] [Indexed: 12/15/2022]
Abstract
Most lung cancer patients are diagnosed late with metastasis, which is the major cause of cancer-related death and recurrent tumors that often exhibit chemoresistance. In the present study, we initially identified gap junction beta-4 protein (Gjb4) to be overexpressed in highly metastatic cancer cells selected by their enhanced binding to serum components. Overexpression or knockdown of Gjb4 increased or decreased lung metastasis of syngeneic mice, respectively. We found that Gjb4 expression was higher in lung tumors than normal tissues (p = 0.0026), and Gjb4 levels in blood buffy coat samples showed significant performance in diagnosing stage I-III (p = 0.002814) and stage IV (p < 0.0001) lung cancer. Moreover, high Gjb4 expression levels were correlated with poor prognosis (p = 1.4e-4) and recurrence (p = 1.9e-12). Using syngeneic mouse model, we observed that Gjb4 was able to promote tumor growth. High molecular weight serum fraction containing the major growth factor component IGF1 was able to induce Gjb4 via PKC pathway. Gjb4 activated Src signaling via MET, and overexpression of Gjb4 enhanced sphere-forming ability and anchorage-independent growth, which were reversed by inhibition of Src. In addition, we demonstrated that Gjb4-mediated Src activation enhanced chemoresistance of cancer cells toward gemcitabine and etoposide. The combination of Gjb4 knockdown, gemcitabine, and dasatinib further enhanced the inhibition of cancer cell viability. Together, our study has identified Gjb4 as a potential novel diagnostic and prognostic biomarker for lung cancer. Targeting Gjb4 may be exploited as a modality for improving lung cancer therapy.
Collapse
Affiliation(s)
- Yi-Pei Lin
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli, Taiwan.,Institute of Molecular and Cellular Biology, National Taiwan University, Taipei, Taiwan
| | - Jun-I Wu
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli, Taiwan.,Department of Life Sciences, National Central University, Taoyuan, Taiwan
| | - Chien-Wei Tseng
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli, Taiwan.,Chinese Medicine Research Center, China Medical University, Taichung, Taiwan
| | - Huei-Jane Chen
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli, Taiwan
| | - Lu-Hai Wang
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli, Taiwan. .,Institute of Molecular and Cellular Biology, National Taiwan University, Taipei, Taiwan. .,Department of Life Sciences, National Central University, Taoyuan, Taiwan. .,Chinese Medicine Research Center, China Medical University, Taichung, Taiwan.
| |
Collapse
|
43
|
Gaowa S, Bao N, Da M, Qiburi Q, Ganbold T, Chen L, Altangerel A, Temuqile T, Baigude H. Traditional Mongolian medicine Eerdun Wurile improves stroke recovery through regulation of gene expression in rat brain. JOURNAL OF ETHNOPHARMACOLOGY 2018; 222:249-260. [PMID: 29758340 DOI: 10.1016/j.jep.2018.05.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 04/28/2018] [Accepted: 05/10/2018] [Indexed: 06/08/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Eerdun Wurile (EW) is one of the key Mongolian medicines for treatment of neurological and cardiological disorders. EW is ranked most regularly used Mongolian medicine in clinic. Components of EW which mainly originate from natural products are well defined and are unique to Mongolian medicine. AIM OF THE STUDY Although the recipe of EW contains known neuroactive chemicals originated from plants, its mechanism of action has never been elucidated at molecular level. The objective of the present study is to explore the mechanism of neuroregenerative activity of EW by focusing on the regulation of gene expression in the brain of rat model of stroke. MATERIALS AND METHODS Rat middle cerebral artery occlusion (MCAO) models were treated with EW for 15 days. Then, total RNAs from the cerebral cortex of rat MCAO models treated with either EW or control (saline) were extracted and analyzed by transcriptome sequencing. Differentially expressed genes were analyzed for their functions during the recovery of ischemic stroke. The expression level of significantly differentially expressed genes such as growth factors, microglia markers and secretive enzymes in the lesion was further validated by RT-qPCR and immunohistochemistry. RESULTS Previously identified neuroactive compounds, such as geniposide (Yu et al., 2009), myristicin (Shin et al., 1988), costunolide (Okugawa et al., 1996), toosendanin (Shi and Chen, 1999) were detected in EW formulation. Bederson scale indicated that the treatment of rat MCAO models with EW showed significantly lowered neurological deficits (p < 0.01). The regional cerebral blood circulation was also remarkably higher in rat MCAO models treated with EW compared to the control group. A total of 186 genes were upregulated in the lesion of rat MCAO models treated with EW compared to control group. Among them, growth factors such as Igf1 (p < 0.05), Igf2 (p < 0.01), Grn (p < 0.01) were significantly upregulated in brain after treatment of rat MCAO models with EW. Meanwhile, greatly enhanced expression of microglia markers, as well as complementary components and secretive proteases were also detected. CONCLUSION Our data collectively indicated that EW enhances expression of growth factors including Igf1 and Igf2 in neurons and microglia, and may stimulate microglia polarization in the brain. The consequences of such activity include stimulation of neuron growth, hydrolysis and clearance of cell debris at the lesion, as well as the angiogenesis.
Collapse
Affiliation(s)
- Saren Gaowa
- School of Basic Medical Science, Beijing University of Chinese Medicine, Chaoyang District, Beijing 100029, PR China; Inner Mongolia Medical University, Hohhot, Inner Mongolia 010020, PR China; International Hospital of Mongolian Medicine, Hohhot, Inner Mongolia 010021, PR China
| | - Narisi Bao
- School of Basic Medical Science, Beijing University of Chinese Medicine, Chaoyang District, Beijing 100029, PR China; Inner Mongolia Medical University, Hohhot, Inner Mongolia 010020, PR China
| | - Man Da
- International Hospital of Mongolian Medicine, Hohhot, Inner Mongolia 010021, PR China
| | - Qiburi Qiburi
- Institute of Mongolian Medicinal Chemistry, School of Chemistry & Chemical Engineering, Inner Mongolia University, Hohhot, Inner Mongolia 010020, PR China
| | - Tsogzolmaa Ganbold
- Institute of Mongolian Medicinal Chemistry, School of Chemistry & Chemical Engineering, Inner Mongolia University, Hohhot, Inner Mongolia 010020, PR China
| | - Lu Chen
- Institute of Mongolian Medicinal Chemistry, School of Chemistry & Chemical Engineering, Inner Mongolia University, Hohhot, Inner Mongolia 010020, PR China
| | - Altanzul Altangerel
- Institute of Mongolian Medicinal Chemistry, School of Chemistry & Chemical Engineering, Inner Mongolia University, Hohhot, Inner Mongolia 010020, PR China
| | - Temuqile Temuqile
- Inner Mongolia Medical University, Hohhot, Inner Mongolia 010020, PR China; International Hospital of Mongolian Medicine, Hohhot, Inner Mongolia 010021, PR China
| | - Huricha Baigude
- Institute of Mongolian Medicinal Chemistry, School of Chemistry & Chemical Engineering, Inner Mongolia University, Hohhot, Inner Mongolia 010020, PR China.
| |
Collapse
|
44
|
Tanabe N, Kuboyama T, Tohda C. Matrine Directly Activates Extracellular Heat Shock Protein 90, Resulting in Axonal Growth and Functional Recovery in Spinal Cord Injured-Mice. Front Pharmacol 2018; 9:446. [PMID: 29867458 PMCID: PMC5949560 DOI: 10.3389/fphar.2018.00446] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 04/17/2018] [Indexed: 01/28/2023] Open
Abstract
After spinal cord injury (SCI), reconstruction of neuronal tracts is very difficult because an inhibitory scar is formed at the lesion site, in which several axonal growth inhibitors, such as chondroitin sulfate proteoglycans (CSPG), accumulate. We previously found that matrine, a major alkaloid in Sophora flavescens, enhanced axonal growth in neurons seeded on CSPG coating. The aims of this study were to investigate therapeutic effects of matrine in SCI mice and to clarify the underlying mechanism. Matrine was orally administered to contusion SCI mice. In the matrine-treated mice, motor dysfunction of the hindlimbs was improved, and the density of 5-HT-positive tracts was increased in the injured spinal cord. We explored putative direct binding proteins of matrine in cultured neurons using drug affinity responsive target stability (DARTS). As a result, heat shock protein 90 (HSP90) was identified, and matrine enhanced HSP90 chaperon activity. We then presumed that extracellular HSP90 is a matrine-targeting signaling molecule, and found that specific blocking of extracellular HSP90 by a neutralizing antibody completely diminished matrine-induced axonal growth and SCI amelioration. Our results suggest that matrine enhances axonal growth and functional recovery in SCI mice by direct activation of extracellular HSP90. Matrine could be a significant candidate for therapeutic drugs for SCI with a novel mechanism.
Collapse
Affiliation(s)
- Norio Tanabe
- Division of Neuromedical Science, Department of Bioscience, Institute of Natural Medicine, University of Toyama, Toyama, Japan
| | - Tomoharu Kuboyama
- Division of Neuromedical Science, Department of Bioscience, Institute of Natural Medicine, University of Toyama, Toyama, Japan
| | - Chihiro Tohda
- Division of Neuromedical Science, Department of Bioscience, Institute of Natural Medicine, University of Toyama, Toyama, Japan
| |
Collapse
|
45
|
Walker JL, Bleaken BM, Romisher AR, Alnwibit AA, Menko AS. In wound repair vimentin mediates the transition of mesenchymal leader cells to a myofibroblast phenotype. Mol Biol Cell 2018; 29:1555-1570. [PMID: 29718762 PMCID: PMC6080657 DOI: 10.1091/mbc.e17-06-0364] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Following injury, mesenchymal repair cells are activated to function as leader cells that modulate wound healing. These cells have the potential to differentiate to myofibroblasts, resulting in fibrosis and scarring. The signals underlying these differing pathways are complex and incompletely understood. The ex vivo mock cataract surgery cultures are an attractive model with which to address this question. With this model we study, concurrently, the mechanisms that control mesenchymal leader cell function in injury repair within their native microenvironment and the signals that induce this same cell population to acquire a myofibroblast phenotype when these cells encounter the environment of the adjacent tissue culture platform. Here we show that on injury, the cytoskeletal protein vimentin is released into the extracellular space, binds to the cell surface of the mesenchymal leader cells located at the wound edge in the native matrix environment, and supports wound closure. In profibrotic environments, the extracellular vimentin pool also links specifically to the mesenchymal leader cells and has an essential role in signaling their fate change to a myofibroblast. These findings suggest a novel role for extracellular, cell-surface–associated vimentin in mediating repair-cell function in wound repair and in transitioning these cells to a myofibroblast phenotype.
Collapse
Affiliation(s)
- J L Walker
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107
| | - B M Bleaken
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107
| | - A R Romisher
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107
| | - A A Alnwibit
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107
| | - A S Menko
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107
| |
Collapse
|
46
|
Bastounis EE, Yeh YT, Theriot JA. Matrix stiffness modulates infection of endothelial cells by Listeria monocytogenes via expression of cell surface vimentin. Mol Biol Cell 2018; 29:1571-1589. [PMID: 29718765 PMCID: PMC6080647 DOI: 10.1091/mbc.e18-04-0228] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Extracellular matrix stiffness (ECM) is one of the many mechanical forces acting on mammalian adherent cells and an important determinant of cellular function. While the effect of ECM stiffness on many aspects of cellular behavior has been studied previously, how ECM stiffness might mediate susceptibility of host cells to infection by bacterial pathogens is hitherto unexplored. To address this open question, we manufactured hydrogels of varying physiologically relevant stiffness and seeded human microvascular endothelial cells (HMEC-1) on them. We then infected HMEC-1 with the bacterial pathogen Listeria monocytogenes (Lm) and found that adhesion of Lm to host cells increases monotonically with increasing matrix stiffness, an effect that requires the activity of focal adhesion kinase (FAK). We identified cell surface vimentin as a candidate surface receptor mediating stiffness-dependent adhesion of Lm to HMEC-1 and found that bacterial infection of these host cells is decreased when the amount of surface vimentin is reduced. Our results provide the first evidence that ECM stiffness can mediate the susceptibility of mammalian host cells to infection by a bacterial pathogen.
Collapse
Affiliation(s)
- Effie E Bastounis
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA 94305
| | - Yi-Ting Yeh
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093
| | - Julie A Theriot
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA 94305.,Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305.,Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305
| |
Collapse
|
47
|
Secretory factors from OP9 stromal cells delay differentiation and increase the expansion potential of adult erythroid cells in vitro. Sci Rep 2018; 8:1983. [PMID: 29386568 PMCID: PMC5792592 DOI: 10.1038/s41598-018-20491-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 01/19/2018] [Indexed: 12/12/2022] Open
Abstract
Development of in vitro culture systems for the generation of red blood cells is a goal of scientists globally with the aim of producing clinical grade products for transfusion. Although mature reticulocytes can be efficiently generated by such systems, the numbers produced fall short of that required for therapeutics, due to limited proliferative capacity of the erythroblasts. To overcome this hurdle, approaches are required to increase the expansion potential of such culture systems. The OP9 mouse stromal cell line is known to promote haematopoietic differentiation of pluripotent stem cells, however an effect of OP9 cells on erythropoiesis has not been explored. In this study, we show not only OP9 co-culture, but factors secreted by OP9 cells in isolation increase the proliferative potential of adult erythroid cells by delaying differentiation and hence maintaining self-renewing cells for an extended duration. The number of reticulocytes obtained was increased by approximately 3.5-fold, bringing it closer to that required for a therapeutic product. To identify the factors responsible, we analysed the OP9 cell secretome using comparative proteomics, identifying 18 candidate proteins. These data reveal the potential to increase erythroid cell numbers from in vitro culture systems without the need for genetic manipulation or co-culture.
Collapse
|
48
|
Rohrbeck A, Höltje M, Adolf A, Oms E, Hagemann S, Ahnert-Hilger G, Just I. The Rho ADP-ribosylating C3 exoenzyme binds cells via an Arg-Gly-Asp motif. J Biol Chem 2017; 292:17668-17680. [PMID: 28882889 PMCID: PMC5663871 DOI: 10.1074/jbc.m117.798231] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 08/28/2017] [Indexed: 12/11/2022] Open
Abstract
The Rho ADP-ribosylating C3 exoenzyme (C3bot) is a bacterial protein toxin devoid of a cell-binding or -translocation domain. Nevertheless, C3 can efficiently enter intact cells, including neurons, but the mechanism of C3 binding and uptake is not yet understood. Previously, we identified the intermediate filament vimentin as an extracellular membranous interaction partner of C3. However, uptake of C3 into cells still occurs (although reduced) in the absence of vimentin, indicating involvement of an additional host cell receptor. C3 harbors an Arg–Gly–Asp (RGD) motif, which is the major integrin-binding site, present in a variety of integrin ligands. To check whether the RGD motif of C3 is involved in binding to cells, we performed a competition assay with C3 and RGD peptide or with a monoclonal antibody binding to β1-integrin subunit and binding assays in different cell lines, primary neurons, and synaptosomes with C3-RGD mutants. Here, we report that preincubation of cells with the GRGDNP peptide strongly reduced C3 binding to cells. Moreover, mutation of the RGD motif reduced C3 binding to intact cells and also to recombinant vimentin. Anti-integrin antibodies also lowered the C3 binding to cells. Our results indicate that the RGD motif of C3 is at least one essential C3 motif for binding to host cells and that integrin is an additional receptor for C3 besides vimentin.
Collapse
Affiliation(s)
- Astrid Rohrbeck
- From the Institute of Toxicology, Hannover Medical School, Carl-Neuberg-Strasse 1, D-30625 Hannover and
| | - Markus Höltje
- the Institute of Integrative Neuroanatomy, Charité-Universitätsmedizin, D-10115 Berlin, Germany
| | - Andrej Adolf
- the Institute of Integrative Neuroanatomy, Charité-Universitätsmedizin, D-10115 Berlin, Germany
| | - Elisabeth Oms
- From the Institute of Toxicology, Hannover Medical School, Carl-Neuberg-Strasse 1, D-30625 Hannover and
| | - Sandra Hagemann
- From the Institute of Toxicology, Hannover Medical School, Carl-Neuberg-Strasse 1, D-30625 Hannover and
| | - Gudrun Ahnert-Hilger
- the Institute of Integrative Neuroanatomy, Charité-Universitätsmedizin, D-10115 Berlin, Germany
| | - Ingo Just
- From the Institute of Toxicology, Hannover Medical School, Carl-Neuberg-Strasse 1, D-30625 Hannover and
| |
Collapse
|
49
|
Yang Z, Kuboyama T, Tohda C. A Systematic Strategy for Discovering a Therapeutic Drug for Alzheimer's Disease and Its Target Molecule. Front Pharmacol 2017; 8:340. [PMID: 28674493 PMCID: PMC5474478 DOI: 10.3389/fphar.2017.00340] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Accepted: 05/19/2017] [Indexed: 11/13/2022] Open
Abstract
Natural medicines are attractive sources of leading compounds that can be used as interventions for neurodegenerative disorders. The complexity of their chemical components and undetermined bio-metabolism have greatly hindered both the use of natural medicines and the identification of their active constituents. Here, we report a systematic strategy for evaluating the bioactive candidates in natural medicines used for Alzheimer's disease (AD). We found that Drynaria Rhizome could enhance memory function and ameliorate AD pathologies in 5XFAD mice. Biochemical analysis led to the identification of the bio-effective metabolites that are transferred to the brain, namely, naringenin and its glucuronides. To explore the mechanism of action, we combined the drug affinity responsive target stability with immunoprecipitation-liquid chromatography/mass spectrometry analysis, identifying the collapsin response mediator protein 2 protein as a target of naringenin. Our study indicates that biochemical analysis coupled with pharmacological methods can be used in the search for new targets for AD intervention.
Collapse
Affiliation(s)
- Zhiyou Yang
- Division of Neuromedical Science, Department of Bioscience, Institute of Natural Medicine, University of ToyamaToyama, Japan
| | - Tomoharu Kuboyama
- Division of Neuromedical Science, Department of Bioscience, Institute of Natural Medicine, University of ToyamaToyama, Japan
| | - Chihiro Tohda
- Division of Neuromedical Science, Department of Bioscience, Institute of Natural Medicine, University of ToyamaToyama, Japan
| |
Collapse
|
50
|
Ding Y, Li J, Liu Z, Liu H, Li H, Li Z. IGF-1 potentiates sensory innervation signalling by modulating the mitochondrial fission/fusion balance. Sci Rep 2017; 7:43949. [PMID: 28276453 PMCID: PMC5343424 DOI: 10.1038/srep43949] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 01/31/2017] [Indexed: 01/07/2023] Open
Abstract
Restoring the contractile function of long-term denervated skeletal muscle (SKM) cells is difficult due to the long period of denervation, which causes a loss of contractility. Although sensory innervation is considered a promising protective approach, its effect is still restricted. In this study, we introduced insulin-like growth factor-1 (IGF-1) as an efficient protective agent and observed that IGF-1 potentiated the effects of sensory protection by preventing denervated muscle atrophy and improving the condition of denervated muscle cells in vivo and in vitro. IGF-1-induced Akt phosphorylation suppressed the mitochondrial outer-membrane protein Mul1 expression, which is a key step on preserving contractile property of sensory innervated SKM cells. Mul1 overexpression interfered with the balance between mitochondrial fusion and fission and was a key node for blocking the effects of IGF-1 that preserved the contractility of sensory-innervated SKM cells. Activation of AMP-activated protein kinase α (AMPKα), a mitochondrial downstream target, could block the effects of IGF-1. These data provide novel evidence that might be applied when searching for new approaches to improve the functional condition of long-term denervated SKM cells by increasing sensory protection using the IGF-1 signalling system to modulate the balance between mitochondrial fusion and fission.
Collapse
Affiliation(s)
- Yuan Ding
- Department of Anatomy, Shandong University School of Medicine, Jinan 250012, China
| | - Jianmin Li
- Department of Orthopaedics, Shandong University Qilu Hospital, Jinan 250012, China
| | - Zhen Liu
- Department of Anatomy, Shandong University School of Medicine, Jinan 250012, China
| | - Huaxiang Liu
- Department of Rheumatology, Shandong University Qilu Hospital, Jinan 250012, China
| | - Hao Li
- Department of Orthopaedics, Shandong University Qilu Hospital, Jinan 250012, China
| | - Zhenzhong Li
- Department of Anatomy, Shandong University School of Medicine, Jinan 250012, China
| |
Collapse
|