1
|
Schweitzer F, Letoha T, Osterhaus A, Prajeeth CK. Impact of Tick-Borne Orthoflaviviruses Infection on Compact Human Brain Endothelial Barrier. Int J Mol Sci 2025; 26:2342. [PMID: 40076965 PMCID: PMC11901142 DOI: 10.3390/ijms26052342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 02/27/2025] [Accepted: 03/04/2025] [Indexed: 03/14/2025] Open
Abstract
Tick-borne encephalitis remains a significant burden on human health in the endemic areas in Central Europe and Eastern Asia. The causative agent, tick-borne encephalitis virus (TBEV), is a neurotropic virus belonging to the genus of Orthoflavivirus. After TBEV enters the central nervous system (CNS), it mainly targets neurons, causing encephalitis and leading to life-long disabilities, coma and, in rare cases, death. The neuroinvasive mechanisms of orthoflaviviruses are poorly understood. Here we investigate the mechanism of TBEV neuroinvasion, hypothesizing that TBEV influences blood-brain barrier (BBB) properties and uses transcellular routes to cross the endothelial barrier and enter the CNS. To test this hypothesis, we employed an in vitro transwell system consisting of endothelial cell monolayers cultured on insert membranes and studied the barrier properties following inoculation to tick-borne orthoflaviviruses. It was shown that TBEV and closely related but naturally attenuated Langat virus (LGTV) crossed the intact endothelial cell monolayer without altering its barrier properties. Interestingly, transendothelial migration of TBEV was significantly affected when two cellular surface antigens, the laminin-binding protein and vimentin, were blocked with specific antibodies. Taken together, these results indicate that orthoflaviviruses use non-destructive transcellular routes through endothelial cells to establish infection within the CNS.
Collapse
Affiliation(s)
- Felix Schweitzer
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine, 30559 Hannover, Germany; (F.S.); (A.O.)
- Center for Systems Neuroscience (ZSN), 30559 Hannover, Germany
| | | | - Albert Osterhaus
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine, 30559 Hannover, Germany; (F.S.); (A.O.)
- Center for Systems Neuroscience (ZSN), 30559 Hannover, Germany
| | - Chittappen Kandiyil Prajeeth
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine, 30559 Hannover, Germany; (F.S.); (A.O.)
| |
Collapse
|
2
|
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
|
3
|
Rani A, Ergün S, Karnati S, Jha HC. Understanding the link between neurotropic viruses, BBB permeability, and MS pathogenesis. J Neurovirol 2024; 30:22-38. [PMID: 38189894 DOI: 10.1007/s13365-023-01190-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 11/04/2023] [Accepted: 12/12/2023] [Indexed: 01/09/2024]
Abstract
Neurotropic viruses can infiltrate the CNS by crossing the blood-brain barrier (BBB) through various mechanisms including paracellular, transcellular, and "Trojan horse" mechanisms during leukocyte diapedesis. These viruses belong to several families, including retroviruses; human immunodeficiency virus type 1 (HIV-1), flaviviruses; Japanese encephalitis (JEV); and herpesviruses; herpes simplex virus type 1 (HSV-1), Epstein-Barr virus (EBV), and mouse adenovirus 1 (MAV-1). For entering the brain, viral proteins act upon the tight junctions (TJs) between the brain microvascular endothelial cells (BMECs). For instance, HIV-1 proteins, such as glycoprotein 120, Nef, Vpr, and Tat, disrupt the BBB and generate a neurotoxic effect. Recombinant-Tat triggers amendments in the BBB by decreasing expression of the TJ proteins such as claudin-1, claudin-5, and zona occludens-1 (ZO-1). Thus, the breaching of BBB has been reported in myriad of neurological diseases including multiple sclerosis (MS). Neurotropic viruses also exhibit molecular mimicry with several myelin sheath proteins, i.e., antibodies against EBV nuclear antigen 1 (EBNA1) aa411-426 cross-react with MBP and EBNA1 aa385-420 was found to be associated with MS risk haplotype HLA-DRB1*150. Notably, myelin protein epitopes (PLP139-151, MOG35-55, and MBP87-99) are being used to generate model systems for MS such as experimental autoimmune encephalomyelitis (EAE) to understand the disease mechanism and therapeutics. Viruses like Theiler's murine encephalomyelitis virus (TMEV) are also commonly used to generate EAE. Altogether, this review provide insights into the viruses' association with BBB leakiness and MS along with possible mechanistic details which could potentially use for therapeutics.
Collapse
Affiliation(s)
- Annu Rani
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore, India
| | - Süleyman Ergün
- Institute of Anatomy and Cell Biology, Julius-Maximilians-University Würzburg, Würzburg, 97070, Germany
| | - Srikanth Karnati
- Institute of Anatomy and Cell Biology, Julius-Maximilians-University Würzburg, Würzburg, 97070, Germany
| | - Hem Chandra Jha
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore, India.
| |
Collapse
|
4
|
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
|
5
|
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: 0] [Impact Index Per Article: 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
|
6
|
Li J, Zheng K, Shen H, Wu H, Wan C, Zhang R, Liu Z. Calpain-2 protein influences chikungunya virus replication and regulates vimentin rearrangement caused by chikungunya virus infection. Front Microbiol 2023; 14:1229576. [PMID: 37928675 PMCID: PMC10620729 DOI: 10.3389/fmicb.2023.1229576] [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: 05/26/2023] [Accepted: 10/06/2023] [Indexed: 11/07/2023] Open
Abstract
Chikungunya fever (CHIF), a vector-borne disease transmitted mainly by Aedes albopictus and Aedes aegypti, is caused by Chikungunya virus (CHIKV) infection. To date, it is estimated that 39% of the world's population is at risk of infection for living in countries and regions where CHIKV is endemic. However, at present, the cellular receptors of CHIKV remains not clear, and there are no specific drugs and vaccines for CHIF. Here, the cytotoxicity of calpain-2 protein activity inhibitor III and specific siRNA was detected by MTT assays. The replication of CHIKV was detected by qPCR amplification and plaque assay. Western blot was used to determine the level of the calpain-2 protein and vimentin protein. Immunofluorescence was also operated for detecting the rearrangement of vimentin protein. Our results indicated that calpain-2 protein activity inhibitor III and specific siRNA might suppress CHIKV replication. Furthermore, CHIKV infection led to vimentin remodeling and formation of cage-like structures, which could be inhibited by the inhibitor III. In summary, we confirmed that calpain-2 protein influenced chikungunya virus replication and regulated vimentin rearrangement caused by chikungunya virus infection, which could be important for understanding the biological significance of CHIKV replication and the future development of antiviral strategies.
Collapse
Affiliation(s)
- Jia Li
- Department of Clinical Laboratory, Affiliated Hengyang Hospital of Southern Medical University, Hengyang Central Hospital, Hengyang, China
- Institute of Pathogenic Organisms, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
- Biosafety Level 3 Laboratory, School of Public Health, Southern Medical University, Guangzhou, China
| | - Kang Zheng
- Department of Clinical Laboratory, Affiliated Hengyang Hospital of Southern Medical University, Hengyang Central Hospital, Hengyang, China
| | - Huilong Shen
- Department of Clinical Laboratory, Affiliated Hengyang Hospital of Southern Medical University, Hengyang Central Hospital, Hengyang, China
| | - Hua Wu
- Department of Clinical Laboratory, Affiliated Hengyang Hospital of Southern Medical University, Hengyang Central Hospital, Hengyang, China
| | - Chengsong Wan
- Institute of Pathogenic Organisms, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Renli Zhang
- Biosafety Level 3 Laboratory, School of Public Health, Southern Medical University, Guangzhou, China
| | - Zhimin Liu
- Department of Clinical Laboratory, Affiliated Hengyang Hospital of Southern Medical University, Hengyang Central Hospital, Hengyang, China
| |
Collapse
|
7
|
Arrindell J, Desnues B. Vimentin: from a cytoskeletal protein to a critical modulator of immune response and a target for infection. Front Immunol 2023; 14:1224352. [PMID: 37475865 PMCID: PMC10354447 DOI: 10.3389/fimmu.2023.1224352] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 06/20/2023] [Indexed: 07/22/2023] Open
Abstract
Vimentin is an intermediate filament protein that plays a role in cell processes, including cell migration, cell shape and plasticity, or organelle anchorage. However, studies from over the last quarter-century revealed that vimentin can be expressed at the cell surface and even secreted and that its implications in cell physiology largely exceed structural and cytoskeletal functions. Consequently, vimentin contributes to several pathophysiological conditions such as cancer, autoimmune and inflammatory diseases, or infection. In this review, we aimed at covering these various roles and highlighting vimentin implications in the immune response. We also provide an overview of how some microbes including bacteria and viruses have acquired the ability to circumvent vimentin functions in order to interfere with host responses and promote their uptake, persistence, and egress from host cells. Lastly, we discuss the therapeutic approaches associated with vimentin targeting, leading to several beneficial effects such as preventing infection, limiting inflammatory responses, or the progression of cancerous events.
Collapse
Affiliation(s)
- Jeffrey Arrindell
- Aix Marseille Univ, Institut de Recherche pour le Développement (IRD), Assistance Publique-Hôpitaux de Marseille (AP-HM), Microbes Evolution Phylogeny and Infections (MEPHI), Marseille, France
- Institut Hospitalo-Universitaire (IHU)-Méditerranée Infection, Marseille, France
| | - Benoit Desnues
- Aix Marseille Univ, Institut de Recherche pour le Développement (IRD), Assistance Publique-Hôpitaux de Marseille (AP-HM), Microbes Evolution Phylogeny and Infections (MEPHI), Marseille, France
- Institut Hospitalo-Universitaire (IHU)-Méditerranée Infection, Marseille, France
| |
Collapse
|
8
|
Kurhade C, Kang S, Biering SB, Hwang S, Randall G. CAPRIN1 Is Required for Control of Viral Replication Complexes by Interferon Gamma. mBio 2023; 14:e0017223. [PMID: 37052473 PMCID: PMC10294620 DOI: 10.1128/mbio.00172-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Accepted: 03/13/2023] [Indexed: 04/14/2023] Open
Abstract
Replication complexes (RCs), formed by positive-strand (+) RNA viruses through rearrangements of host endomembranes, protect their replicating RNA from host innate immune defenses. We have shown that two evolutionarily conserved defense systems, autophagy and interferon (IFN), target viral RCs and inhibit viral replication collaboratively. However, the mechanism by which autophagy proteins target viral RCs and the role of IFN-inducible GTPases in the disruption of RCs remains poorly understood. Here, using murine norovirus (MNV) as a model (+) RNA virus, we show that the guanylate binding protein 1 (GBP1) is the human GTPase responsible for inhibiting RCs. Furthermore, we found that ATG16L1 mediates the LC3 targeting of MNV RC by binding to WIPI2B and CAPRIN1, and that IFN gamma-mediated control of MNV replication was dependent on CAPRIN1. Collectively, this study identifies a novel mechanism for the autophagy machinery-mediated recognition and inhibition of viral RCs, a hallmark of (+) RNA virus replication. IMPORTANCE Replication complexes provide a microenvironment important for (+) RNA virus replication and shield it from host immune response. Previously we have shown that interferon gamma (IFNG) disrupts the RC of MNV via evolutionarily conserved autophagy proteins and IFN-inducible GTPases. Elucidating the mechanism of targeting of viral RC by ATG16L1 and IFN-induced GTPase will pave the way for development of therapeutics targeting the viral replication complexes. Here, we have identified GBP1 as the sole GBP targeting viral RC and uncovered the novel role of CAPRIN1 in recruiting ATG16L1 to the viral RC.
Collapse
Affiliation(s)
- Chaitanya Kurhade
- Department of Microbiology, The University of Chicago, Chicago, Illinois, USA
| | - Soowon Kang
- Department of Microbiology, The University of Chicago, Chicago, Illinois, USA
| | - Scott B. Biering
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, California, USA
| | - Seungmin Hwang
- Department of Pathology, The University of Chicago, Chicago, Illinois, USA
| | - Glenn Randall
- Department of Microbiology, The University of Chicago, Chicago, Illinois, USA
| |
Collapse
|
9
|
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
|
10
|
Staruszkiewicz M, Pituch-Noworolska A, Skoczen S. Uncommon types of autoantibodies - Detection and clinical associations. Autoimmun Rev 2023; 22:103263. [PMID: 36563770 DOI: 10.1016/j.autrev.2022.103263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 12/18/2022] [Indexed: 12/24/2022]
Abstract
Immunofluorescence is a basic method for detection of autoantibodies in serum. It is used as screening for people with symptoms suggesting autoimmune process and disease. Antinuclear antibodies (ANA) assay detecting antibodies against nuclear proteins used commonly for diagnosis of systemic autoimmune disease, although antibodies against cytoplasmic components and mitotic structures are usable in clinic. The majority of ANA nuclear patterns have been comprehensively studied with increasing data. However, the cytoplasmic and mitotic patterns are underestimated and still require further assessment. In this review the clinical associations and significance of uncommon types of autoantibodies are presented and discussed.
Collapse
Affiliation(s)
| | | | - Szymon Skoczen
- Department of Paediatric Oncology and Haematology, Jagiellonian University, Medical College, Krakow, Poland; Department of Oncology and Haematology, University Children's Hospital, Krakow, Poland.
| |
Collapse
|
11
|
Harris J, Borg NA. The multifaceted roles of NLRP3-modulating proteins in virus infection. Front Immunol 2022; 13:987453. [PMID: 36110852 PMCID: PMC9468583 DOI: 10.3389/fimmu.2022.987453] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 08/11/2022] [Indexed: 12/14/2022] Open
Abstract
The innate immune response to viruses is critical for the correct establishment of protective adaptive immunity. Amongst the many pathways involved, the NLRP3 [nucleotide-binding oligomerisation domain (NOD)-like receptor protein 3 (NLRP3)] inflammasome has received considerable attention, particularly in the context of immunity and pathogenesis during infection with influenza A (IAV) and SARS-CoV-2, the causative agent of COVID-19. Activation of the NLRP3 inflammasome results in the secretion of the proinflammatory cytokines IL-1β and IL-18, commonly coupled with pyroptotic cell death. While this mechanism is protective and key to host defense, aberrant NLRP3 inflammasome activation causes a hyperinflammatory response and excessive release of cytokines, both locally and systemically. Here, we discuss key molecules in the NLRP3 pathway that have also been shown to have significant roles in innate and adaptive immunity to viruses, including DEAD box helicase X-linked (DDX3X), vimentin and macrophage migration inhibitory factor (MIF). We also discuss the clinical opportunities to suppress NLRP3-mediated inflammation and reduce disease severity.
Collapse
Affiliation(s)
- James Harris
- Cell Biology Assays Team, Biomedical Manufacturing, Commonwealth Scientific and Industrial Research Organisation (CSIRO), Clayton, VIC, Australia
- Centre for Inflammatory diseases, Department of Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, VIC, Australia
| | - Natalie A. Borg
- Immunity and Immune Evasion Laboratory, Chronic Infectious and Inflammatory Diseases Research, School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
| |
Collapse
|
12
|
Thalla DG, Rajwar AC, Laurent AM, Becher JE, Kainka L, Lautenschläger F. Extracellular vimentin is expressed at the rear of activated macrophage-like cells: Potential role in enhancement of migration and phagocytosis. Front Cell Dev Biol 2022; 10:891281. [PMID: 35923851 PMCID: PMC9340215 DOI: 10.3389/fcell.2022.891281] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 06/27/2022] [Indexed: 11/15/2022] Open
Abstract
Macrophages have a vital role in the immune system through elimination of cell debris and microorganisms by phagocytosis. The activation of macrophages by tumour necrosis factor-α induces expression of extracellular cell-surface vimentin and promotes release of this vimentin into the extracellular environment. Vimentin is a cytoskeletal protein that is primarily located in the cytoplasm of cells. However, under circumstances like injury, stress, senescence and activation, vimentin can be expressed on the extracellular cell surface, or it can be released into the extracellular space. The characteristics of this extracellular vimentin, and its implications for the functional role of macrophages and the mechanism of secretion remain unclear. Here, we demonstrate that vimentin is released mainly from the back of macrophage-like cells. This polarisation is strongly enhanced upon macrophage activation. One-dimensional patterned lines showed that extracellular cell-surface vimentin is localised primarily at the back of activated macrophage-like cells. Through two-dimensional migration and phagocytosis assays, we show that this extracellular vimentin enhances migration and phagocytosis of macrophage-like cells. We further show that this extracellular vimentin forms agglomerates on the cell surface, in contrast to its intracellular filamentous form, and that it is released into the extracellular space in the form of small fragments. Taken together, we provide new insights into the release of extracellular cell-surface vimentin and its implications for macrophage functionality.
Collapse
Affiliation(s)
| | | | | | | | - Lucina Kainka
- Experimental Physics, Saarland University, Saarbrücken, Germany
| | - Franziska Lautenschläger
- Experimental Physics, Saarland University, Saarbrücken, Germany
- Centre for Biophysics, Saarland University, Saarbrücken, Germany
| |
Collapse
|
13
|
Li J, Jia H, Tian M, Wu N, Yang X, Qi J, Ren W, Li F, Bian H. SARS-CoV-2 and Emerging Variants: Unmasking Structure, Function, Infection, and Immune Escape Mechanisms. Front Cell Infect Microbiol 2022; 12:869832. [PMID: 35646741 PMCID: PMC9134119 DOI: 10.3389/fcimb.2022.869832] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Accepted: 04/06/2022] [Indexed: 12/24/2022] Open
Abstract
As of April 1, 2022, over 468 million COVID-19 cases and over 6 million deaths have been confirmed globally. Unlike the common coronavirus, SARS-CoV-2 has highly contagious and attracted a high level of concern worldwide. Through the analysis of SARS-CoV-2 structural, non-structural, and accessory proteins, we can gain a deeper understanding of structure-function relationships, viral infection mechanisms, and viable strategies for antiviral therapy. Angiotensin-converting enzyme 2 (ACE2) is the first widely acknowledged SARS-CoV-2 receptor, but researches have shown that there are additional co-receptors that can facilitate the entry of SARS-CoV-2 to infect humans. We have performed an in-depth review of published papers, searching for co-receptors or other auxiliary membrane proteins that enhance viral infection, and analyzing pertinent pathogenic mechanisms. The genome, and especially the spike gene, undergoes mutations at an abnormally high frequency during virus replication and/or when it is transmitted from one individual to another. We summarized the main mutant strains currently circulating global, and elaborated the structural feature for increased infectivity and immune evasion of variants. Meanwhile, the principal purpose of the review is to update information on the COVID-19 outbreak. Many countries have novel findings on the early stage of the epidemic, and accruing evidence has rewritten the timeline of the outbreak, triggering new thinking about the origin and spread of COVID-19. It is anticipated that this can provide further insights for future research and global epidemic prevention and control.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Feifei Li
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Hongjun Bian
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| |
Collapse
|
14
|
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
|
15
|
Yu J, Li X, Zhou D, Liu X, He X, Huang SH, Wu Q, Zhu L, Yu L, Yao J, Zhang B, Zhao W. Vimentin Inhibits Dengue Virus Type 2 Invasion of the Blood-Brain Barrier. Front Cell Infect Microbiol 2022; 12:868407. [PMID: 35433510 PMCID: PMC9005901 DOI: 10.3389/fcimb.2022.868407] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 03/03/2022] [Indexed: 12/28/2022] Open
Abstract
Dengue virus (DENV) causes dengue fever, which is prevalent in the tropical and subtropical regions, and in recent years, has resulted in several major epidemics. Vimentin, a cytoskeletal component involved in DENV infection, is significantly reorganized during infection. However, the mechanism underlying the association between DENV infection and vimentin is still poorly understood. We generated vimentin-knockout (Vim-KO) human brain microvascular endothelial cells (HBMECs) and a Vim-KO SV129 suckling mouse model, combining the dynamic vimentin changes observed in vitro and differences in disease course in vivo, to clarify the role of vimentin in DENV-2 infection. We found that the phosphorylation and solubility of vimentin changed dynamically during DENV-2 infection of HBMECs, suggesting the regulation of vimentin by DENV-2 infection. The similar trends observed in the phosphorylation and solubility of vimentin showed that these characteristics are related. Compared with that in control cells, the DENV-2 viral load was significantly increased in Vim-KO HBMECs, and after DENV-2 infection, Vim-KO SV129 mice displayed more severe disease signs than wild-type SV129 mice, as well as higher viral loads in their serum and brain tissue, demonstrating that vimentin can inhibit DENV-2 infection. Moreover, Vim-KO SV129 mice had more disordered cerebral cortical nerve cells, confirming that Vim-KO mice were more susceptible to DENV-2 infection, which causes severe brain damage. The findings of our study help clarify the mechanism by which vimentin inhibits DENV-2 infection and provides guidance for antiviral treatment strategies for DENV infections.
Collapse
Affiliation(s)
- Jianhai Yu
- Biological Safety Laboratory of Level 3 (BSL-3) Laboratory (Guangdong), Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Xujuan Li
- Biological Safety Laboratory of Level 3 (BSL-3) Laboratory (Guangdong), Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Dongrui Zhou
- Biological Safety Laboratory of Level 3 (BSL-3) Laboratory (Guangdong), Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Xuling Liu
- Biological Safety Laboratory of Level 3 (BSL-3) Laboratory (Guangdong), Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Xiaoen He
- Biological Safety Laboratory of Level 3 (BSL-3) Laboratory (Guangdong), Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Sheng-He Huang
- Saban Research Institute of Children’s Hospital Los Angeles, Department of Pediatrics, University of Southern California, Los Angeles, CA, United States
| | - Qinghua Wu
- Biological Safety Laboratory of Level 3 (BSL-3) Laboratory (Guangdong), Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Li Zhu
- Biological Safety Laboratory of Level 3 (BSL-3) Laboratory (Guangdong), Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Linzhong Yu
- Department of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Jinxiu Yao
- Department of Laboratory, People's Hospital of Yangjiang, Yangjiang, China
| | - Bao Zhang
- Biological Safety Laboratory of Level 3 (BSL-3) Laboratory (Guangdong), Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
- *Correspondence: Wei Zhao, ; Bao Zhang,
| | - Wei Zhao
- Biological Safety Laboratory of Level 3 (BSL-3) Laboratory (Guangdong), Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
- *Correspondence: Wei Zhao, ; Bao Zhang,
| |
Collapse
|
16
|
Amraei R, Xia C, Olejnik J, White MR, Napoleon MA, Lotfollahzadeh S, Hauser BM, Schmidt AG, Chitalia V, Mühlberger E, Costello CE, Rahimi N. Extracellular vimentin is an attachment factor that facilitates SARS-CoV-2 entry into human endothelial cells. Proc Natl Acad Sci U S A 2022; 119:2113874119. [PMID: 35078919 PMCID: PMC8833221 DOI: 10.1073/pnas.2113874119] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/18/2021] [Indexed: 02/07/2023] Open
Abstract
SARS-CoV-2 entry into host cells is a crucial step for virus tropism, transmission, and pathogenesis. Angiotensin-converting enzyme 2 (ACE2) has been identified as the primary entry receptor for SARS-CoV-2; however, the possible involvement of other cellular components in the viral entry has not yet been fully elucidated. Here we describe the identification of vimentin (VIM), an intermediate filament protein widely expressed in cells of mesenchymal origin, as an important attachment factor for SARS-CoV-2 on human endothelial cells. Using liquid chromatography-tandem mass spectrometry, we identified VIM as a protein that binds to the SARS-CoV-2 spike (S) protein. We showed that the S-protein receptor binding domain (RBD) is sufficient for S-protein interaction with VIM. Further analysis revealed that extracellular VIM binds to SARS-CoV-2 S-protein and facilitates SARS-CoV-2 infection, as determined by entry assays performed with pseudotyped viruses expressing S and with infectious SARS-CoV-2. Coexpression of VIM with ACE2 increased SARS-CoV-2 entry in HEK-293 cells, and shRNA-mediated knockdown of VIM significantly reduced SARS-CoV-2 infection of human endothelial cells. Moreover, incubation of A549 cells expressing ACE2 with purified VIM increased pseudotyped SARS-CoV-2-S entry. CR3022 antibody, which recognizes a distinct epitope on SARS-CoV-2-S-RBD without interfering with the binding of the spike with ACE2, inhibited the binding of VIM with CoV-2 S-RBD, and neutralized viral entry in human endothelial cells, suggesting a key role for VIM in SARS-CoV-2 infection of endothelial cells. This work provides insight into the pathogenesis of COVID-19 linked to the vascular system, with implications for the development of therapeutics and vaccines.
Collapse
Affiliation(s)
- Razie Amraei
- Department of Pathology, Boston University School of Medicine, Boston, MA 02118
| | - Chaoshuang Xia
- Center for Biomedical Mass Spectrometry, Boston University School of Medicine, Boston, MA 02118
| | - Judith Olejnik
- Department of Microbiology, Boston University School of Medicine, Boston, MA 02118
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA 02118
| | - Mitchell R White
- Department of Microbiology, Boston University School of Medicine, Boston, MA 02118
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA 02118
| | - Marc A Napoleon
- Renal Section, Department of Medicine, Boston University Medical Center, Boston, MA 02118
| | - Saran Lotfollahzadeh
- Renal Section, Department of Medicine, Boston University Medical Center, Boston, MA 02118
| | - Blake M Hauser
- Ragon Institute of Massachusetts General Hospital (MGH), Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139
- Department of Microbiology, Harvard Medical School, Boston, MA 02115
| | - Aaron G Schmidt
- Ragon Institute of Massachusetts General Hospital (MGH), Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139
- Department of Microbiology, Harvard Medical School, Boston, MA 02115
| | - Vipul Chitalia
- Renal Section, Department of Medicine, Boston University Medical Center, Boston, MA 02118
- Veterans Affairs Boston Healthcare System, Boston, MA 02118
- Institute of Medical Engineering and Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Elke Mühlberger
- Department of Microbiology, Boston University School of Medicine, Boston, MA 02118
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA 02118
| | - Catherine E Costello
- Center for Biomedical Mass Spectrometry, Boston University School of Medicine, Boston, MA 02118;
| | - Nader Rahimi
- Department of Pathology, Boston University School of Medicine, Boston, MA 02118;
| |
Collapse
|
17
|
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]
|
18
|
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
|
19
|
Utomo DIS, Pambudi S, Park EY. Humoral immune response induced with dengue virus-like particles serotypes 1 and 4 produced in silkworm. AMB Express 2022; 12:8. [PMID: 35102445 PMCID: PMC8802989 DOI: 10.1186/s13568-022-01353-6] [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: 11/16/2021] [Accepted: 01/22/2022] [Indexed: 11/12/2022] Open
Abstract
Dengue is an arboviral disease, which threatens almost half the global population, and has emerged as the most significant of current global public health challenges. In this study, we prepared dengue virus-like particles (DENV-LPs) consisting of Capsid-premembrane-envelope (CprME) and premembrane-envelope (prME) polypeptides from serotype 1 and 4, which were expressed in the silkworms using Bombyx mori nucleopolyhedrovirus (BmNPV) bacmid. 1CprME, 1prME, 4CprME, and 4prME expressed proteins in hemolymph, and the molecular weight of the purified proteins was 55 kDa, respectively. The purified polypeptides formed spherical Dengue virus-like particles (DENV-LPs) with ~ 30–55 nm in diameter. The immunoelectron microscopy (IEM) images revealed antigens to the surface of a lipid bilayer of DENV-LPs. The heparin-binding assay shows a positive relationship between absorbance and E protein domain III (EDIII) quantity, which is supported by the isothermal titration calorimetry assay. This indicates a moderate binding affinity between heparin and DENV-LP. The high correlation between patient sera and DENV-LP reactivities revealed that these DENV-LPs shared similar epitopes with the natural dengue virus. IgG elicitation studies in mice have demonstrated that DENV-LPs/CPrMEs elicit a stronger immune response than DENV-LP/prMEs, which lends credence to this claim. Dengue virus-like particles for serotype 1 and serotype 4 (DENV-LPs/1 and DENV-LPs/4) were produced in silkworm. Heparin-binding assay by ELISA and ITC showed that DENV-LPs/1 and DENV-LPs/4 contain Envelope Domain III. DENV-LPs/1 and DENV-LPs/4 showed affinity to sera from human dengue patients and immunized mice.
Collapse
|
20
|
de França NR, Ménard HA, Lora M, Zhou Z, Rauch J, Hitchon C, Andrade LEC, Colmegna I. Characterization and use of the ECV304 autoantigenic citrullinome to understand anti-citrullinated protein/peptide autoantibodies in rheumatoid arthritis. Arthritis Res Ther 2022; 24:23. [PMID: 35027076 PMCID: PMC8756661 DOI: 10.1186/s13075-021-02698-2] [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: 08/27/2021] [Accepted: 12/08/2021] [Indexed: 11/12/2022] Open
Abstract
Background Anti-citrullinated protein antibodies (ACPAs) are highly specific for rheumatoid arthritis (RA). In vivo, ACPAs target peptidyl-citrulline epitopes (cit-) in a variety of proteins (cit-prot-ACPAs) and derived peptides (cit-pept-ACPAs) generated via the peptidylarginine deiminase (PAD) isoenzymes. We aimed to identify a cell line with self-citrullination capacity, to describe its autoantigenic citrullinome, and to test it as a source of autocitrullinated proteins and peptides. Methods Human cell lines were screened for cit-proteins by Western blot. PAD isoenzymes were identified by RT-PCR. Autocitrullination of ECV304 was optimized, and the ECV304 autocitrullinomes immunoprecipitated by sera from three RA patients were characterized by mass spectrometry. Cit-pept-ACPAs were detected using anti-CCP2 ELISA and cit-prot-ACPAs, by an auto-cit-prot-ECV304 ELISA. Sera from 177 RA patients, 59 non-RA rheumatic disease patients and 25 non-disease controls were tested. Results Of the seven cell lines studied, only ECV304 simultaneously overexpressed PAD2 and PAD3 and its extracts reproducibly autocitrullinated self and non-self-proteins. Proteomic analysis of the cit-ECV304 products immunoprecipitated by RA sera, identified novel cit-targets: calreticulin, profilin 1, vinculin, new 14–3-3 protein family members, chaperones, and mitochondrial enzymes. The auto-cit-prot-ECV304 ELISA had a sensitivity of 50% and a specificity of 95% for RA diagnosis. Conclusions ECV304 cells overexpress two of the PAD isoenzymes capable of citrullinating self-proteins. These autocitrullinated cells constitute a basic and clinical research tool that enable the detection of cit-prot-ACPAs with high diagnostic specificity and allow the identification of the specific cit-proteins targeted by individual RA sera. Supplementary Information The online version contains supplementary material available at 10.1186/s13075-021-02698-2.
Collapse
Affiliation(s)
- Natalia Regine de França
- Division of Rheumatology, Department of Medicine, McGill University, The Research Institute of the McGill University Health Centre, 1001 Décarie Boulevard, Montréal, QC, H4A 3J1, Canada.,Division of Rheumatology, Paulista School of Medicine, Federal University of Sao Paulo, Sao Paulo, Brazil
| | - Henri André Ménard
- Division of Rheumatology, Department of Medicine, McGill University, The Research Institute of the McGill University Health Centre, 1001 Décarie Boulevard, Montréal, QC, H4A 3J1, Canada
| | - Maximilien Lora
- Division of Rheumatology, Department of Medicine, McGill University, The Research Institute of the McGill University Health Centre, 1001 Décarie Boulevard, Montréal, QC, H4A 3J1, Canada
| | - Zhijie Zhou
- Division of Rheumatology, Department of Medicine, McGill University, The Research Institute of the McGill University Health Centre, 1001 Décarie Boulevard, Montréal, QC, H4A 3J1, Canada
| | - Joyce Rauch
- Division of Rheumatology, Department of Medicine, McGill University, The Research Institute of the McGill University Health Centre, 1001 Décarie Boulevard, Montréal, QC, H4A 3J1, Canada
| | - Carol Hitchon
- Section of Rheumatology, Department of Medicine, University of Manitoba, Winnipeg, MB, Canada
| | | | - Inés Colmegna
- Division of Rheumatology, Department of Medicine, McGill University, The Research Institute of the McGill University Health Centre, 1001 Décarie Boulevard, Montréal, QC, H4A 3J1, Canada.
| |
Collapse
|
21
|
Ivanušec A, Šribar J, Križaj I. Secreted Phospholipases A 2 - not just Enzymes: Revisited. Int J Biol Sci 2022; 18:873-888. [PMID: 35002531 PMCID: PMC8741859 DOI: 10.7150/ijbs.68093] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 12/02/2021] [Indexed: 12/18/2022] Open
Abstract
Secreted phospholipases A2 (sPLA2s) participate in a very broad spectrum of biological processes through their enzymatic activity and as ligands for membrane and soluble receptors. The physiological roles of sPLA2s as enzymes have been very well described, while their functions as ligands are still poorly known. Since the last overview of sPLA2-binding proteins (sPLA2-BPs) 10 years ago, several important discoveries have occurred in this area. New and more sensitive analytical tools have enabled the discovery of additional sPLA2-BPs, which are presented and critically discussed here. The structural diversity of sPLA2-BPs reveals sPLA2s as very promiscuous proteins, and we offer some structural explanations for this nature that makes these proteins evolutionarily highly advantageous. Three areas of physiological engagement of sPLA2-BPs have appeared most clearly: cellular transport and signalling, and regulation of the enzymatic activity of sPLA2s. Due to the multifunctionality of sPLA2s, they appear to be exceptional pharmacological targets. We reveal the potential to exploit interactions of sPLA2s with other proteins in medical terms, for the development of original diagnostic and therapeutic procedures. We conclude this survey by suggesting the priority questions that need to be answered.
Collapse
Affiliation(s)
- Adrijan Ivanušec
- Department of Molecular and Biomedical Sciences, Jožef Stefan Institute, Jamova 39, 1000 Ljubljana, Slovenia.,Faculty of Medicine, University of Ljubljana, Vrazov trg 2, 1000 Ljubljana, Slovenia
| | - Jernej Šribar
- Department of Molecular and Biomedical Sciences, Jožef Stefan Institute, Jamova 39, 1000 Ljubljana, Slovenia
| | - Igor Križaj
- Department of Molecular and Biomedical Sciences, Jožef Stefan Institute, Jamova 39, 1000 Ljubljana, Slovenia
| |
Collapse
|
22
|
Carse S, Lang D, Katz AA, Schäfer G. Exogenous Vimentin Supplementation Transiently Affects Early Steps during HPV16 Pseudovirus Infection. Viruses 2021; 13:v13122471. [PMID: 34960740 PMCID: PMC8703489 DOI: 10.3390/v13122471] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 12/07/2021] [Accepted: 12/08/2021] [Indexed: 11/23/2022] Open
Abstract
Understanding and modulating the early steps in oncogenic Human Papillomavirus (HPV) infection has great cancer-preventative potential, as this virus is the etiological agent of virtually all cervical cancer cases and is associated with many other anogenital and oropharyngeal cancers. Previous work from our laboratory has identified cell-surface-expressed vimentin as a novel HPV16 pseudovirus (HPV16-PsVs)-binding molecule modulating its infectious potential. To further explore its mode of inhibiting HPV16-PsVs internalisation, we supplemented it with exogenous recombinant human vimentin and show that only the globular form of the molecule (as opposed to the filamentous form) inhibited HPV16-PsVs internalisation in vitro. Further, this inhibitory effect was only transient and not sustained over prolonged incubation times, as demonstrated in vitro and in vivo, possibly due to full-entry molecule engagement by the virions once saturation levels have been reached. The vimentin-mediated delay of HPV16-PsVs internalisation could be narrowed down to affecting multiple steps during the virus’ interaction with the host cell and was found to affect both heparan sulphate proteoglycan (HSPG) binding as well as the subsequent entry receptor complex engagement. Interestingly, decreased pseudovirus internalisation (but not infection) in the presence of vimentin was also demonstrated for oncogenic HPV types 18, 31 and 45. Together, these data demonstrate the potential of vimentin as a modulator of HPV infection which can be used as a tool to study early mechanisms in infectious internalisation. However, further refinement is needed with regard to vimentin’s stabilisation and formulation before its development as an alternative prophylactic means.
Collapse
Affiliation(s)
- Sinead Carse
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town 7925, South Africa;
- Faculty of Health Sciences, Institute of Infectious Disease and Molecular Medicine (IDM), University of Cape Town, Cape Town 7925, South Africa;
- Department of Integrative Biomedical Sciences, Division of Medical Biochemistry and Structural Biology, Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa
| | - Dirk Lang
- Department of Human Biology, Division of Cell Biology, Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa;
| | - Arieh A. Katz
- Faculty of Health Sciences, Institute of Infectious Disease and Molecular Medicine (IDM), University of Cape Town, Cape Town 7925, South Africa;
- Department of Integrative Biomedical Sciences, Division of Medical Biochemistry and Structural Biology, Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa
- SA-MRC-UCT Gynaecological Cancer Research Centre, University of Cape Town, Cape Town 7925, South Africa
| | - Georgia Schäfer
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town 7925, South Africa;
- Faculty of Health Sciences, Institute of Infectious Disease and Molecular Medicine (IDM), University of Cape Town, Cape Town 7925, South Africa;
- Department of Integrative Biomedical Sciences, Division of Medical Biochemistry and Structural Biology, Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa
- Correspondence: ; Tel.: +27-21-404-7688
| |
Collapse
|
23
|
Vimentin Regulates Chemokine Expression and NOD2 Activation in Brain Endothelium during Group B Streptococcal Infection. Infect Immun 2021; 89:e0034021. [PMID: 34491787 DOI: 10.1128/iai.00340-21] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Streptococcus agalactiae (group B Streptococcus, or GBS) is an opportunistic pathogen capable of causing invasive disease in susceptible individuals, including the newborn. Currently, GBS is the leading cause of meningitis in the neonatal period. We have recently shown that GBS interacts directly with host type III intermediate filament vimentin to gain access to the central nervous system. This results in characteristic meningeal inflammation and disease progression; however, the specific role of vimentin in the inflammatory process is unknown. Here, we investigate the contribution of vimentin to the pathogenesis of GBS meningitis. We show that a CRISPR-targeted deletion of vimentin in human cerebral microvascular endothelial cells (hCMEC) reduced GBS induction of neutrophil attractants interleukin-8 (IL-8) and CXCL-1 as well as NF-κB activation. We further show that inhibition of vimentin localization also prevented similar chemokine activation by GBS. One known chemokine regulator is the nucleotide-binding oligomerization domain containing protein 2 (NOD2), which is known to interact directly with vimentin. Thus, we hypothesized that NOD2 would also promote GBS chemokine induction. We show that GBS infection induced NOD2 transcription in hCMEC comparably to the muramyl dipeptide (MDP) NOD2 agonist, and the chemokine induction was reduced in the presence of a NOD2 inhibitor. Using a mouse model of GBS meningitis, we also observed increased NOD2 transcript and NOD2 activation in brain tissue of infected mice. Lastly, we show that NOD2-mediated IL-8 and CXCL1 induction required vimentin, further indicating the importance of vimentin in mediating inflammatory responses in brain endothelium.
Collapse
|
24
|
Korolowicz KE, Suresh M, Li B, Huang X, Yon C, Kallakury BV, Lee KP, Park S, Kim YW, Menne S. Combination Treatment with the Vimentin-Targeting Antibody hzVSF and Tenofovir Suppresses Woodchuck Hepatitis Virus Infection in Woodchucks. Cells 2021; 10:2321. [PMID: 34571970 PMCID: PMC8466705 DOI: 10.3390/cells10092321] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 08/24/2021] [Accepted: 08/27/2021] [Indexed: 02/07/2023] Open
Abstract
Current treatment options for patients infected with hepatitis B virus (HBV) are suboptimal, because the approved drugs rarely induce cure due to the persistence of the viral DNA genome in the nucleus of infected hepatocytes, and are associated with either severe side effects (pegylated interferon-alpha) or require life-long administration (nucleos(t)ide analogs). We report here the evaluation of the safety and therapeutic efficacy of a novel, humanized antibody (hzVSF) in the woodchuck model of HBV infection. hzVSF has been shown to act as a viral entry inhibitor, most likely by suppressing vimentin-mediated endocytosis of virions. Targeting the increased vimentin expression on liver cells by hzVSF after infection with HBV or woodchuck hepatitis virus (WHV) was demonstrated initially. Thereafter, hzVSF safety was assessed in eight woodchucks naïve for WHV infection. Antiviral efficacy of hzVSF was evaluated subsequently in 24 chronic WHV carrier woodchucks by monotreatment with three ascending doses and in combination with tenofovir alafenamide fumarate (TAF). Consistent with the proposed blocking of WHV reinfection, intravenous hzVSF administration for 12 weeks resulted in a modest but transient reduction of viral replication and associated liver inflammation. In combination with oral TAF dosing, the antiviral effect of hzVSF was enhanced and sustained in half of the woodchucks with an antibody response to viral proteins. Thus, hzVSF safely but modestly alters chronic WHV infection in woodchucks; however, as a combination partner to TAF, its antiviral efficacy is markedly increased. The results of this preclinical study support future evaluation of this novel anti-HBV drug in patients.
Collapse
Affiliation(s)
- Kyle E. Korolowicz
- Department of Microbiology & Immunology, Georgetown University Medical Center, Washington, DC 20057, USA; (K.E.K.); (M.S.); (B.L.); (X.H.); (C.Y.)
| | - Manasa Suresh
- Department of Microbiology & Immunology, Georgetown University Medical Center, Washington, DC 20057, USA; (K.E.K.); (M.S.); (B.L.); (X.H.); (C.Y.)
| | - Bin Li
- Department of Microbiology & Immunology, Georgetown University Medical Center, Washington, DC 20057, USA; (K.E.K.); (M.S.); (B.L.); (X.H.); (C.Y.)
| | - Xu Huang
- Department of Microbiology & Immunology, Georgetown University Medical Center, Washington, DC 20057, USA; (K.E.K.); (M.S.); (B.L.); (X.H.); (C.Y.)
| | - Changsuek Yon
- Department of Microbiology & Immunology, Georgetown University Medical Center, Washington, DC 20057, USA; (K.E.K.); (M.S.); (B.L.); (X.H.); (C.Y.)
| | - Bhaskar V. Kallakury
- Department of Pathology, Georgetown University Medical Center, Washington, DC 20057, USA;
| | - Kyoung-pil Lee
- ImmuneMed, Inc., Chuncheon BioTown, Soyanggang ro 32, Chuncheon-si 24232, Gangwon-do, Korea; (K.-p.L.); (S.P.); (Y.-W.K.)
| | - Sungman Park
- ImmuneMed, Inc., Chuncheon BioTown, Soyanggang ro 32, Chuncheon-si 24232, Gangwon-do, Korea; (K.-p.L.); (S.P.); (Y.-W.K.)
| | - Yoon-Won Kim
- ImmuneMed, Inc., Chuncheon BioTown, Soyanggang ro 32, Chuncheon-si 24232, Gangwon-do, Korea; (K.-p.L.); (S.P.); (Y.-W.K.)
| | - Stephan Menne
- Department of Microbiology & Immunology, Georgetown University Medical Center, Washington, DC 20057, USA; (K.E.K.); (M.S.); (B.L.); (X.H.); (C.Y.)
| |
Collapse
|
25
|
Vimentin as a Cap of Invisibility: Proposed Role of Vimentin in Rabbit Hemorrhagic Disease Virus (RHDV) Infection. Viruses 2021; 13:v13071416. [PMID: 34372621 PMCID: PMC8310380 DOI: 10.3390/v13071416] [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: 06/25/2021] [Revised: 07/15/2021] [Accepted: 07/19/2021] [Indexed: 11/24/2022] Open
Abstract
Vimentin is an intermediate filament, a cytoskeleton protein expressed mainly in cells of mesenchymal origin. Increasing evidence indicates that vimentin could play a key role in viral infections. Therefore, changes in tissue and extracellular vimentin expression and associated signal trails may determine/protect the fate of cells and the progression of disease caused by viral infection. Rabbit hemorrhagic disease virus (RHDV), genotype GI.1, is an etiological agent that causes a severe and highly lethal disease—RHD (rabbit hemorrhagic disease). This article evaluates the gene and protein expression of vimentin in the tissues (liver, lungs, spleen, and kidneys) and serum of rabbits experimentally infected with two RHDV variants (GI.1a). The VIM mRNA expression levels in the tissues were determined using reverse transcription quantitative real-time PCR (RT-qPCR). In addition, the amount of vimentin protein in the serum was analyzed by an ELISA test. We observed significantly elevated expression levels of VIM mRNA and protein in the liver and kidney tissues of infected rather than healthy rabbits. In addition, VIM mRNA expression was increased in the lung tissues; meanwhile, we observed only protein-enhanced vimentin in the spleen. The obtained results are significant and promising, as they indicate the role of vimentin in RHDV infection and the course of RHD. The role of vimentin in RHDV infection could potentially rely on the one hand, on creating a cap of invisibility against the intracellular viral spread, or, on the other hand, after the damage of cells, vimentin could act as a signal of tissue damage.
Collapse
|
26
|
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
|
27
|
Wang P, Liu X, Li Q, Wang J, Ruan W. Proteomic analyses identify intracellular targets for Japanese encephalitis virus nonstructural protein 1 (NS1). Virus Res 2021; 302:198495. [PMID: 34175344 DOI: 10.1016/j.virusres.2021.198495] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 06/15/2021] [Accepted: 06/16/2021] [Indexed: 11/25/2022]
Abstract
Japanese encephalitis is a zoonotic disease caused by Japanese encephalitis virus (JEV). JEV nonstructural protein 1 (NS1) is involved in many crucial biological events during viral infection and immune suppression. To investigate the role of JEV NS1 in virus-infected cells, the molecules with which it interacts intracellularly were screened with a pull-down assay and liquid chromatography-tandem mass spectrometry (LC-MS/MS). The interaction between heterogeneous nuclear ribonucleoprotein K (hnRNP K), vimentin and NS1 were verified with coimmunoprecipitation and confocal assays. Our results show that JEV NS1 interacts with vimentin, hnRNP K and colocalizes with cellular vimentin and hnRNP K. Furthermore, our results demonstrate that the expression of vimentin and hnRNP K were up-regulated in both NS1-transfected and JEV-infected cells. Knocking down vimentin and hnRNP K reduced viral replication while conversely, over-expression of vimentin and hnRNP K improved viral replication, suggesting an important role for this protein in the viral life cycle. Also, We found that vimentin also interacted with hnRNP K after overexpression of NS1 or JEV infection. These findings provide insight into the molecular mechanism of JEV replication and highlight the key role the NS1 in JEV infection.
Collapse
Affiliation(s)
- Peng Wang
- Beijing Key Laboratory of Traditional Chinese Veterinary Medicine, College of Animal Science and Technology, Beijing University of Agriculture, Beijing, 102206, China
| | - Xinze Liu
- Beijing Key Laboratory of Traditional Chinese Veterinary Medicine, College of Animal Science and Technology, Beijing University of Agriculture, Beijing, 102206, China
| | - Qi Li
- Beijing Key Laboratory of Traditional Chinese Veterinary Medicine, College of Animal Science and Technology, Beijing University of Agriculture, Beijing, 102206, China
| | - Jue Wang
- Beijing Key Laboratory of Traditional Chinese Veterinary Medicine, College of Animal Science and Technology, Beijing University of Agriculture, Beijing, 102206, China
| | - Wenke Ruan
- Beijing Key Laboratory of Traditional Chinese Veterinary Medicine, College of Animal Science and Technology, Beijing University of Agriculture, Beijing, 102206, China.
| |
Collapse
|
28
|
Comprehensive interactome analysis of the spike protein of swine acute diarrhea syndrome coronavirus. BIOSAFETY AND HEALTH 2021; 3:156-163. [PMID: 34027383 PMCID: PMC8127515 DOI: 10.1016/j.bsheal.2021.05.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 05/12/2021] [Accepted: 05/13/2021] [Indexed: 12/01/2022] Open
Abstract
Swine acute diarrhea syndrome coronavirus (SADS-CoV) is a recently discovered coronavirus that causes severe and acute diarrhea and rapid weight loss in piglets. SADS-CoV was reported to be capable of infecting cell lines derived from diverse species, including bats, mice, hamsters, rats, chickens, pigs, nonhuman primates, and humans, implying its high risk of cross-species infection. However, its receptor is still unknown. In this study, the receptor-binding domain of the SADS-CoV spike (S) protein was purified and then subjected to affinity purification (AP)-coupled mass spectrometry (MS)-based proteomic analysis to identify the interactors of the SADS-CoV S protein. Forty-three host proteins were identified, and a Gene Ontology analysis indicated that these interactors can be grouped into categories such as “cell-cell adhesion”, “translation” “viral transcription”, suggesting that these processes may participate in the SADS-CoV life cycles. RNA interference-based screening of these interactors indicated that PPIB and vimentin can affect SADS-CoV replication. Our study provides an overarching view into the host interactome of the SADS-CoV S protein and highlights potential targets for the development of therapeutics against SADS-CoV.
Collapse
|
29
|
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
|
30
|
Zhang Y, Wen Z, Shi X, Liu YJ, Eriksson JE, Jiu Y. The diverse roles and dynamic rearrangement of vimentin during viral infection. J Cell Sci 2020; 134:134/5/jcs250597. [PMID: 33154171 DOI: 10.1242/jcs.250597] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Epidemics caused by viral infections pose a significant global threat. Cytoskeletal vimentin is a major intermediate filament (IF) protein, and is involved in numerous functions, including cell signaling, epithelial-mesenchymal transition, intracellular organization and cell migration. Vimentin has important roles for the life cycle of particular viruses; it can act as a co-receptor to enable effective virus invasion and guide efficient transport of the virus to the replication site. Furthermore, vimentin has been shown to rearrange into cage-like structures that facilitate virus replication, and to recruit viral components to the location of assembly and egress. Surprisingly, vimentin can also inhibit virus entry or egress, as well as participate in host-cell defense. Although vimentin can facilitate viral infection, how this function is regulated is still poorly understood. In particular, information is lacking on its interaction sites, regulation of expression, post-translational modifications and cooperation with other host factors. This Review recapitulates the different functions of vimentin in the virus life cycle and discusses how they influence host-cell tropism, virulence of the pathogens and the consequent pathological outcomes. These insights into vimentin-virus interactions emphasize the importance of cytoskeletal functions in viral cell biology and their potential for the identification of novel antiviral targets.
Collapse
Affiliation(s)
- Yue Zhang
- The Center for Microbes, Development and Health, Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai 200031, China.,University of Chinese Academy of Sciences, Yuquan Road No. 19(A), Shijingshan District, Beijing 100049, China
| | - Zeyu Wen
- The Center for Microbes, Development and Health, Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai 200031, China.,University of Chinese Academy of Sciences, Yuquan Road No. 19(A), Shijingshan District, Beijing 100049, China
| | - Xuemeng Shi
- The Center for Microbes, Development and Health, Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yan-Jun Liu
- Shanghai Institute of Cardiovascular Diseases, and Institutes of Biomedical Sciences, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - John E Eriksson
- Cell Biology, Biosciences, Faculty of Science and Engineering, Åbo Akademi University, Turku FI-20520, Finland .,Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku FI-20520, Finland
| | - Yaming Jiu
- The Center for Microbes, Development and Health, Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai 200031, China .,University of Chinese Academy of Sciences, Yuquan Road No. 19(A), Shijingshan District, Beijing 100049, China
| |
Collapse
|
31
|
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: 51] [Impact Index Per Article: 10.2] [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
|
32
|
Li Z, Paulin D, Lacolley P, Coletti D, Agbulut O. Vimentin as a target for the treatment of COVID-19. BMJ Open Respir Res 2020; 7:7/1/e000623. [PMID: 32913008 PMCID: PMC7482103 DOI: 10.1136/bmjresp-2020-000623] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 08/24/2020] [Accepted: 08/25/2020] [Indexed: 12/18/2022] Open
Abstract
We and others propose vimentin as a possible cellular target for the treatment of COVID-19. This innovative idea is so recent that it requires further attention and debate. The significant role played by vimentin in virus-induced infection however is well established: (1) vimentin has been reported as a co-receptor and/or attachment site for SARS-CoV; (2) vimentin is involved in viral replication in cells; (3) vimentin plays a fundamental role in both the viral infection and the consequent explosive immune-inflammatory response and (4) a lower vimentin expression is associated with the inhibition of epithelial to mesenchymal transition and fibrosis. Moreover, the absence of vimentin in mice makes them resistant to lung injury. Since vimentin has a twofold role in the disease, not only being involved in the viral infection but also in the associated life-threatening lung inflammation, the use of vimentin-targeted drugs may offer a synergistic advantage as compared with other treatments not targeting vimentin. Consequently, we speculate here that drugs which decrease the expression of vimentin can be used for the treatment of patients with COVID-19 and advise that several Food and Drug Administration-approved drugs be immediately tested in clinical trials against SARS-CoV-2, thus broadening therapeutic options for this type of viral infection.
Collapse
Affiliation(s)
- Zhenlin Li
- Biological Adaptation and Ageing, CNRS UMR 8256, Inserm U1164, Sorbonne Université, Institut de Biologie Paris-Seine, Paris, France
| | - Denise Paulin
- Biological Adaptation and Ageing, CNRS UMR 8256, Inserm U1164, Sorbonne Université, Institut de Biologie Paris-Seine, Paris, France
| | - Patrick Lacolley
- Inserm, UMR_S 1116, DCAC, Université de Lorraine, Nancy, Lorraine, France
| | - Dario Coletti
- Biological Adaptation and Ageing, CNRS UMR 8256, Inserm U1164, Sorbonne Université, Institut de Biologie Paris-Seine, Paris, France.,Department of Anatomy, Histology, Forensic Medicine & Orthopedics, Histology & Medical Embryology Section, Sapienza University of Rome, Roma, Lazio, Italy
| | - Onnik Agbulut
- Biological Adaptation and Ageing, CNRS UMR 8256, Inserm U1164, Sorbonne Université, Institut de Biologie Paris-Seine, Paris, France
| |
Collapse
|
33
|
Utomo DIS, Pambudi S, Sjatha F, Kato T, Park EY. Production of dengue virus-like particles serotype-3 in silkworm larvae and their ability to elicit a humoral immune response in mice. AMB Express 2020; 10:147. [PMID: 32804287 PMCID: PMC7431508 DOI: 10.1186/s13568-020-01087-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 08/11/2020] [Indexed: 02/08/2023] Open
Abstract
To develop monovalent dengue virus-like particle for serotype 3 (DENV-LP/3), we prepared and expressed two structural polyprotein constructs using silkworm and Bm5 cells: DENV-3 Capsid-premembrane-envelope (DENV-3CprME) and premembrane-envelope (DENV-3prME). The expressed PA-tagged 3CprME and 3prME polypeptides were partially purified by PA-tag affinity chromatography and had molecular weights of 85 and 75 kDa, respectively. Expressed proteins were separately verified using the following primary antibodies: the anti-PA tag antibody, DENV premembrane polyclonal antibody, and DENV envelope polyclonal antibody. Transmission electron microscopy revealed that these DENV-3CprME and 3prME formed rough, spherical DENV-LPs (DENV-LP/3CprME and DENV-LP/3prME), respectively, with a diameter of 30–55 nm. The heparin-binding assay demonstrated that these DENV-LPs contained the envelope protein domain III on their surfaces. Both DENV-LPs showed an affinity to sera from human dengue patients and immunized mice. Immunization of mice with DENV-LP/3prME significantly induced the level of antibodies compared with DENV-LP/3CprME. These results indicate that DENV-LP/3prME is suitable as a vaccine candidate compared with DENV-LP/3CprME.
Collapse
|
34
|
Li M, Ramage H, Cherry S. Deciphering flavivirus-host interactions using quantitative proteomics. Curr Opin Immunol 2020; 66:90-97. [PMID: 32682290 DOI: 10.1016/j.coi.2020.06.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 05/13/2020] [Accepted: 06/16/2020] [Indexed: 01/09/2023]
Abstract
Flaviviruses are a group of important emerging and re-emerging human pathogens that cause worldwide epidemics with thousands of deaths annually. Flaviviruses are small, enveloped, positive-sense, single-stranded RNA viruses that are obligate intracellular pathogens, relying heavily on host cell machinery for productive replication. Proteomic approaches have become an increasingly powerful tool to investigate the mechanisms by which viruses interact with host proteins and manipulate cellular processes to promote infection. Here, we review recent advances in employing quantitative proteomics techniques to improve our understanding of the complex interplay between flaviviruses and host cells. We describe new findings on our understanding of how flaviviruses impact protein-protein interactions, protein-RNA interactions, protein abundance, and post-translational modifications to modulate viral infection.
Collapse
Affiliation(s)
- Minghua Li
- Department of Microbiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Holly Ramage
- Department of Microbiology, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Sara Cherry
- Department of Microbiology, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
35
|
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: 116] [Impact Index Per Article: 23.2] [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
|
36
|
Kavathekar VK, Dhanavade MJ, Sonawane KD, Balakrishnan A. Role of cell surface vimentin in Chandipura virus replication in Neuro-2a cells. Virus Res 2020; 285:198014. [PMID: 32418904 PMCID: PMC7270567 DOI: 10.1016/j.virusres.2020.198014] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 05/03/2020] [Accepted: 05/04/2020] [Indexed: 01/31/2023]
Abstract
Study of virus host interaction helps to understand the mechanism of virus life cycle. Chandipura virus is associated with the encephalitis among the children’s in India. Chandipura virus co-localizes with surface vimentin on Neuro-2a cells. Surface vimentin on Neuro-2a cells is involved in interaction with Chandipura virus.
The neurotropic behavior of Chandipura virus (CHPV) is partly understood in experimental animals. Under in vitro conditions, neuronal cells could be a useful tool to study the CHPV interaction with neuronal proteins. The information gathered from such studies will help to design the new therapeutics for CHPV infection. This study identified the surface vimentin protein involved in adsorption of CHPV on Neuro-2a cell line (mouse neuroblastoma cells). The decrease in CHPV infectivity to Neuro-2a cells was observed in the presence of recombinant vimentin or anti-vimentin antibody. Vimentin mRNA expression remains unaltered in CHPV infected Neuro-2a cells. Furthermore, in silico analysis predicted the residues in vimentin and CHPV glycoprotein (G); probably involved in cell-virus interactions. Overall, we conclude that surface vimentin in Neuro-2a cells interact with CHPV and facilitate the binding of CHPV to the cells; it could be acting as a co-receptor for the CHPV. Further investigation is necessary to confirm the exact role of vimentin in CHPV infection in neuronal cells.
Collapse
Affiliation(s)
- Vishal K Kavathekar
- National Institute of Virology, Kerala Unit, TDMC Hospital complex, Vandanam, Alappuzha, Kerala, 688005, India
| | - Maruti J Dhanavade
- Deartment of Biochemistry, Shivaji University, Vidyanagari, Kolhapur, Maharashtra, 416004, India
| | - Kailas D Sonawane
- Deartment of Biochemistry, Shivaji University, Vidyanagari, Kolhapur, Maharashtra, 416004, India
| | - Anukumar Balakrishnan
- National Institute of Virology, Kerala Unit, TDMC Hospital complex, Vandanam, Alappuzha, Kerala, 688005, India.
| |
Collapse
|
37
|
Hwang B, Ise H. Multimeric conformation of type III intermediate filaments but not the filamentous conformation exhibits high affinity to lipid bilayers. Genes Cells 2020; 25:413-426. [DOI: 10.1111/gtc.12768] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 03/19/2020] [Accepted: 03/19/2020] [Indexed: 01/16/2023]
Affiliation(s)
- Beomju Hwang
- Graduate School of Engineering Kyushu University Fukuoka Japan
| | - Hirohiko Ise
- Institute for Materials Chemistry and Engineering Kyushu University Fukuoka Japan
| |
Collapse
|
38
|
Wang W, Sun J, Wang N, Sun Z, Ma Q, Li J, Zhang M, Xu J. Enterovirus A71 capsid protein VP1 increases blood-brain barrier permeability and virus receptor vimentin on the brain endothelial cells. J Neurovirol 2020; 26:84-94. [PMID: 31512144 PMCID: PMC7040057 DOI: 10.1007/s13365-019-00800-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Revised: 08/01/2019] [Accepted: 08/25/2019] [Indexed: 12/22/2022]
Abstract
Enterovirus A71 (EV-A71) is the major cause of severe hand-foot-and-mouth diseases (HFMD), especially encephalitis and other nervous system diseases. EV-A71 capsid protein VP1 mediates virus attachment and is the important virulence factor in the EV-A71pathogenesis. In this study, we explored the roles of VP1 in the permeability of blood-brain barrier (BBB). Sera albumin, Evans blue, and dextran leaked into brain parenchyma of the 1-week-old C57BL/6J mice intracranially injected with VP1 recombinant protein. VP1 also increased the permeability of the brain endothelial cells monolayer, an in vitro BBB model. Tight junction protein claudin-5 was reduced in the brain tissues or brain endothelial cells treated with VP1. In contrast, VP1 increased the expression of virus receptor vimentin, which could be blocked with VP1 neutralization antibody. Vimentin expression in the VP1-treated brain endothelial cells was regulated by TGF-β/Smad-3 and NF-κB signal pathways. Moreover, vimentin over-expression was accompanied with compromised BBB. From these studies, we conclude that EV-A71 virus capsid protein VP1 disrupted BBB and increased virus receptor vimentin, which both may contribute to the virus entrance into brain and EV-A71 CNS infection.
Collapse
Affiliation(s)
- Wenjing Wang
- Department of Infectious Disease, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
- Department of Infectious Disease, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210029, China
| | - Jiandong Sun
- Department of Infectious Disease, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210029, China
| | - Nan Wang
- Department of Respiratory Medicine, People's Hospital of Gaochun, Nanjing, 211300, China
| | - Zhixiao Sun
- Department of Respiratory Medicine, People's Hospital of Gaochun, Nanjing, 211300, China
| | - Qiyun Ma
- Department of Respiratory Medicine, People's Hospital of Gaochun, Nanjing, 211300, China
| | - Jun Li
- Department of Infectious Disease, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| | - Mingshun Zhang
- Key Lab of Antibody Technique of Health Ministry, Nanjing Medical University, Nanjing, 210016, China.
- Department of Immunology, Nanjing Medical University, Nanjing, 210016, China.
| | - Juan Xu
- Department of Immunology, Nanjing Medical University, Nanjing, 210016, China.
| |
Collapse
|
39
|
Begum F, Das S, Mukherjee D, Mal S, Ray U. Insight into the Tropism of Dengue Virus in Humans. Viruses 2019; 11:v11121136. [PMID: 31835302 PMCID: PMC6950149 DOI: 10.3390/v11121136] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 11/01/2019] [Accepted: 11/04/2019] [Indexed: 12/13/2022] Open
Abstract
In tropical and subtropical zones, arboviruses are among the major threats to human life, affecting a large number of populations with serious diseases. Worldwide, over three hundred million people are infected with dengue virus (DENV) every year as per the World Health Organization (WHO). DENV-mediated disease severity ranges from a mild fever to hemorrhagic fever and shock syndrome. Patients suffering from severe infection might experience multi-organ failure, cardiomyopathy and even encephalopathy, further complicating the disease pathogenesis. In life-threatening cases, DENV has been reported to affect almost all organs of the human body. In this review, we discuss the organ tropism of DENV in humans in depth as detected in various autopsy studies. Keeping in mind the fact that there is currently no DENV-specific antiviral, it is of utmost importance to achieve a vivid picture of the susceptible cells in humans which might help in designing antivirals against DENV, especially targeting those tissues in which infection might lead to life-threatening conditions.
Collapse
Affiliation(s)
- Feroza Begum
- CSIR-Indian Institute of Chemical Biology, 4 Raja S.C. Mullick Road, Jadavpur, Kolkata700032, India; (F.B.); (S.D.); (D.M.); (S.M.)
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Sandeepan Das
- CSIR-Indian Institute of Chemical Biology, 4 Raja S.C. Mullick Road, Jadavpur, Kolkata700032, India; (F.B.); (S.D.); (D.M.); (S.M.)
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Debica Mukherjee
- CSIR-Indian Institute of Chemical Biology, 4 Raja S.C. Mullick Road, Jadavpur, Kolkata700032, India; (F.B.); (S.D.); (D.M.); (S.M.)
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Sweety Mal
- CSIR-Indian Institute of Chemical Biology, 4 Raja S.C. Mullick Road, Jadavpur, Kolkata700032, India; (F.B.); (S.D.); (D.M.); (S.M.)
| | - Upasana Ray
- CSIR-Indian Institute of Chemical Biology, 4 Raja S.C. Mullick Road, Jadavpur, Kolkata700032, India; (F.B.); (S.D.); (D.M.); (S.M.)
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
- Correspondence: ; Tel.: +91-978-187-8333
| |
Collapse
|
40
|
Stamova B, Ander BP, Jickling G, Hamade F, Durocher M, Zhan X, Liu DZ, Cheng X, Hull H, Yee A, Ng K, Shroff N, Sharp FR. The intracerebral hemorrhage blood transcriptome in humans differs from the ischemic stroke and vascular risk factor control blood transcriptomes. J Cereb Blood Flow Metab 2019; 39:1818-1835. [PMID: 29651892 PMCID: PMC6727143 DOI: 10.1177/0271678x18769513] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Understanding how the blood transcriptome of human intracerebral hemorrhage (ICH) differs from ischemic stroke (IS) and matched controls (CTRL) will improve understanding of immune and coagulation pathways in both disorders. This study examined RNA from 99 human whole-blood samples using GeneChip® HTA 2.0 arrays to assess differentially expressed transcripts of alternatively spliced genes between ICH, IS and CTRL. We used a mixed regression model with FDR-corrected p(Dx) < 0.2 and p < 0.005 and |FC| > 1.2 for individual comparisons. For time-dependent analyses, subjects were divided into four time-points: 0(CTRL), <24 h, 24-48 h, >48 h; 489 transcripts were differentially expressed between ICH and CTRL, and 63 between IS and CTRL. ICH had differentially expressed T-cell receptor and CD36 genes, and iNOS, TLR, macrophage, and T-helper pathways. IS had more non-coding RNA. ICH and IS both had angiogenesis, CTLA4 in T lymphocytes, CD28 in T helper cells, NFAT regulation of immune response, and glucocorticoid receptor signaling pathways. Self-organizing maps revealed 4357 transcripts changing expression over time in ICH, and 1136 in IS. Understanding ICH and IS transcriptomes will be useful for biomarker development, treatment and prevention strategies, and for evaluating how well animal models recapitulate human ICH and IS.
Collapse
Affiliation(s)
- Boryana Stamova
- 1 Department of Neurology, School of Medicine, University of California at Davis, Sacramento, CA, USA
| | - Bradley P Ander
- 1 Department of Neurology, School of Medicine, University of California at Davis, Sacramento, CA, USA
| | - Glen Jickling
- 1 Department of Neurology, School of Medicine, University of California at Davis, Sacramento, CA, USA.,2 Department of Medicine, University of Alberta, Edmonton, Canada
| | - Farah Hamade
- 1 Department of Neurology, School of Medicine, University of California at Davis, Sacramento, CA, USA
| | - Marc Durocher
- 1 Department of Neurology, School of Medicine, University of California at Davis, Sacramento, CA, USA
| | - Xinhua Zhan
- 1 Department of Neurology, School of Medicine, University of California at Davis, Sacramento, CA, USA
| | - Da Zhi Liu
- 1 Department of Neurology, School of Medicine, University of California at Davis, Sacramento, CA, USA
| | - Xiyuan Cheng
- 1 Department of Neurology, School of Medicine, University of California at Davis, Sacramento, CA, USA
| | - Heather Hull
- 1 Department of Neurology, School of Medicine, University of California at Davis, Sacramento, CA, USA
| | - Alan Yee
- 1 Department of Neurology, School of Medicine, University of California at Davis, Sacramento, CA, USA
| | - Kwan Ng
- 1 Department of Neurology, School of Medicine, University of California at Davis, Sacramento, CA, USA
| | - Natasha Shroff
- 1 Department of Neurology, School of Medicine, University of California at Davis, Sacramento, CA, USA
| | - Frank R Sharp
- 1 Department of Neurology, School of Medicine, University of California at Davis, Sacramento, CA, USA
| |
Collapse
|
41
|
Calderón-Peláez MA, Velandia-Romero ML, Bastidas-Legarda LY, Beltrán EO, Camacho-Ortega SJ, Castellanos JE. Dengue Virus Infection of Blood-Brain Barrier Cells: Consequences of Severe Disease. Front Microbiol 2019; 10:1435. [PMID: 31293558 PMCID: PMC6606788 DOI: 10.3389/fmicb.2019.01435] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 06/06/2019] [Indexed: 01/10/2023] Open
Abstract
More than 500 million people worldwide are infected each year by any of the four-dengue virus (DENV) serotypes. The clinical spectrum caused during these infections is wide and some patients may develop neurological alterations during or after the infection, which could be explained by the cryptic neurotropic and neurovirulent features of flaviviruses like DENV. Using in vivo and in vitro models, researchers have demonstrated that DENV can affect the cells from the blood-brain barrier (BBB) in several ways, which could result in brain tissue damage, neuronal loss, glial activation, tissue inflammation and hemorrhages. The latter suggests that BBB may be compromised during infection; however, it is not clear whether the damage is due to the infection per se or to the local and/or systemic inflammatory response established or activated by the BBB cells. Similarly, the kinetics and cascade of events that trigger tissue damage, and the cells that initiate it, are unknown. This review presents evidence of the BBB cell infection with DENV and the response established toward it by these cells; it also describes the consequences of this response on the nervous tissue, compares these evidence with the one reported with neurotropic viruses of the Flaviviridae family, and shows the complexity and unpredictability of dengue and the neurological alterations induced by it. Clinical evidence and in vitro and in vivo models suggest that this virus uses the bloodstream to enter nerve tissue where it infects the different cells of the neurovascular unit. Each of the cell populations respond individually and collectively and control infection and inflammation, in other cases this response exacerbates the damage leaving irreversible sequelae or causing death. This information will allow us to understand more about the complex disease known as dengue, and its impact on a specialized and delicate tissue like is the nervous tissue.
Collapse
|
42
|
The Group B Streptococcal surface antigen I/II protein, BspC, interacts with host vimentin to promote adherence to brain endothelium and inflammation during the pathogenesis of meningitis. PLoS Pathog 2019; 15:e1007848. [PMID: 31181121 PMCID: PMC6586375 DOI: 10.1371/journal.ppat.1007848] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 06/20/2019] [Accepted: 05/16/2019] [Indexed: 12/29/2022] Open
Abstract
Streptococcus agalactiae (Group B Streptococcus, GBS) normally colonizes healthy adults but can cause invasive disease, such as meningitis, in the newborn. To gain access to the central nervous system, GBS must interact with and penetrate brain or meningeal blood vessels; however, the exact mechanisms are still being elucidated. Here, we investigate the contribution of BspC, an antigen I/II family adhesin, to the pathogenesis of GBS meningitis. Disruption of the bspC gene reduced GBS adherence to human cerebral microvascular endothelial cells (hCMEC), while heterologous expression of BspC in non-adherent Lactococcus lactis conferred bacterial attachment. In a murine model of hematogenous meningitis, mice infected with ΔbspC mutants exhibited lower mortality as well as decreased brain bacterial counts and inflammatory infiltrate compared to mice infected with WT GBS strains. Further, BspC was both necessary and sufficient to induce neutrophil chemokine expression. We determined that BspC interacts with the host cytoskeleton component vimentin and confirmed this interaction using a bacterial two-hybrid assay, microscale thermophoresis, immunofluorescent staining, and imaging flow cytometry. Vimentin null mice were protected from WT GBS infection and also exhibited less inflammatory cytokine production in brain tissue. These results suggest that BspC and the vimentin interaction is critical for the pathogenesis of GBS meningitis. Group B Streptococcus (GBS) typically colonizes healthy adults but can cause severe disease in immune-compromised individuals, including newborns. Despite wide-spread intrapartum antibiotic prophylaxis given to pregnant women, GBS remains a leading cause of neonatal meningitis. To cause meningitis, GBS must interact with and penetrate the blood-brain barrier (BBB), which separates bacteria and immune cells in the blood from the brain. In order to develop targeted therapies to treat GBS meningitis, it is important to understand the mechanisms of BBB crossing. Here, we describe the role of the GBS surface factor, BspC, in promoting meningitis and discover the host ligand for BspC, vimentin, which is an intermediate filament protein that is constitutively expressed by endothelial cells. We determined that BspC interacts with the C-terminal domain of cell-surface vimentin to promote bacterial attachment to brain endothelial cells and that purified BspC protein can induce immune signaling pathways. In a mouse model of hematogenous meningitis, we observed that a GBS mutant lacking BspC was less virulent compared to WT GBS and resulted in less inflammatory disease. We also observed that mice lacking vimentin were protected from GBS infection. These results reveal the importance of the BspC-vimentin interaction in the progression of GBS meningitis disease.
Collapse
|
43
|
Sabharwal P, Amritha CK, Sushmitha C, Natraj U, Savithri HS. Intracellular trafficking and endocytic uptake pathway of Pepper vein banding virus-like particles in epithelial cells. Nanomedicine (Lond) 2019; 14:1247-1265. [PMID: 31084385 DOI: 10.2217/nnm-2018-0405] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Aim: Plant virus-like particles (VLPs) have emerged as a novel platform for delivery of drugs/antibodies. The aim of the present investigation is to establish the entry mechanism of flexuous rod-shaped virus particles into mammalian cells. Methods: Far-Western blot analysis, pull-down and ELISA were used to characterize vimentin and Hsp60 interaction with VLPs. The mode/kinetics of internalization of VLPs was deciphered using pharmacological inhibitors/endosomal markers. Results & discussion: The flexuous rod-shaped VLPs of Pepper vein banding virus (PVBV) enter HeLa and HepG2 cells via cell-surface proteins: vimentin and Hsp60, respectively. VLPs internalize via different modes of endocytosis in HeLa, HepG2 cells and are biodegradable. Vimentin and Hsp60 could be potential epithelial ligands that facilitate targeting of nanoparticles to tumor cells.
Collapse
Affiliation(s)
- Pallavi Sabharwal
- Department of Biochemistry, Indian Institute of Science, Bangalore 560012, India
| | | | - Cheekati Sushmitha
- Department of Biochemistry, Indian Institute of Science, Bangalore 560012, India
| | - Usha Natraj
- Department of Biochemistry, Indian Institute of Science, Bangalore 560012, India
| | | |
Collapse
|
44
|
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
|
45
|
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: 178] [Impact Index Per Article: 25.4] [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
|
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: 29] [Impact Index Per Article: 4.1] [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
|
Yang Y, Yang J, Rao X. What role does superficial vimentin have during DENV-2 infection? Future Virol 2018. [DOI: 10.2217/fvl-2017-0007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Affiliation(s)
- Yi Yang
- Department of Microbiology, College of Basic Medical Sciences, Third Military Medical University, Chongqing 400038, People's Republic of China
| | - Jie Yang
- Department of Microbiology, College of Basic Medical Sciences, Third Military Medical University, Chongqing 400038, People's Republic of China
| | - Xiancai Rao
- Department of Microbiology, College of Basic Medical Sciences, Third Military Medical University, Chongqing 400038, People's Republic of China
| |
Collapse
|
48
|
Wang ZJ, Xu CM, Song ZB, Wang M, Liu QY, Jiang P, Li YF, Bai J, Wang XW. Vimentin modulates infectious porcine circovirus type 2 in PK-15 cells. Virus Res 2017; 243:110-118. [PMID: 29079448 PMCID: PMC7114564 DOI: 10.1016/j.virusres.2017.10.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2017] [Revised: 10/15/2017] [Accepted: 10/17/2017] [Indexed: 01/17/2023]
Abstract
Porcine circovirus type 2 (PCV2) is the pathogen that causes postweaning multisystemic wasting syndrome, which leads to significant economic losses for swine farms worldwide. However, the infection mechanism of PCV2 is not completely understood yet. Vimentin is a part of the cytoskeleton network and plays an important role in several virus infections. It is not clear whether vimentin has a role in PCV2 infection nor how it affects PCV2 infection. In this study, the function of vimentin in PK-15 cells infected with PCV2 has been elucidated. We found that vimentin had a restrictive effect on the replication of PCV2 in PK-15 cells. Overexpression of vimentin by transferred pCAGGS-vimentin and down-regulation by the respective scrambled small interfering RNA showed that vimentin restricted the replication and virion production of PCV2. A special interaction between vimentin and PCV2 Cap protein was observed using laser confocal microscopy and immunoprecipitation assay. Moreover, overexpression of vimentin could decrease NF-κB activity and increase PCV2-induced caspase-3 activity in PK-15 cells. These data suggest that vimentin is involved in the replication of PCV2 and has a restrictive effect on it, which is helpful in the study of the replication mechanism of PCV2.
Collapse
Affiliation(s)
- Zhi-Jian Wang
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Chang-Meng Xu
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Zhong-Bao Song
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Mi Wang
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Qian-Yu Liu
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Ping Jiang
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Yu-Feng Li
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Juan Bai
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Xian-Wei Wang
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China.
| |
Collapse
|
49
|
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: 10] [Impact Index Per Article: 1.3] [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
|