1
|
Lukacher AS, O'Hara BA, Yuan W, Garabian K, Kaiserman J, MacLure E, Haley SA, Atwood WJ. The microvascular endothelium of the blood-brain barrier is highly restrictive to JC Polyomavirus neuroinvasion. Microbiol Spectr 2025; 13:e0028225. [PMID: 40130848 PMCID: PMC12054175 DOI: 10.1128/spectrum.00282-25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Accepted: 02/20/2025] [Indexed: 03/26/2025] Open
Abstract
JC Polyomavirus is the causative agent of progressive multifocal leukoencephalopathy (PML), an often-fatal demyelinating disease. Unfortunately, a diagnosis of PML occurs only after patients have suffered irreversible neuropathologies. The first step in the initiation of PML is viral entry to the brain, but the route and mechanisms responsible for neuroinvasion have not been well established. To gain a better understanding of this, we asked whether purified virus or virus associated with extracellular vesicles (EVs) could penetrate two different cell culture models of the blood-brain barrier. In one model, we used the hCMEC/D3 brain endothelial cell line, and in the other, we used pluripotent stem cells induced to a brain endothelial cell phenotype (iPSC-EC). We found that neither cell type was permissive to viral infection, but the virus bound and was internalized by both in a sialic acid-dependent manner. Despite virus internalization into these cells, very few virions or virus-associated extracellular vesicles (virus-EVs) penetrated the barriers. The small amount of virus or virus-EVs that did pass through either barrier was sufficient to establish infection in human glial cells. Our findings demonstrate that limited amounts of infectious virions and virus-associated EVs can traverse the brain microvascular endothelium and establish infection.IMPORTANCEThe human polyomavirus, JC Polyomavirus (JCPyV), causes a rapidly progressing demyelinating disease in immunocompromised or immunomodulated patients. Demyelinating lesions are often seen surrounding blood vessels in the brain. In this paper, we used two models to recapitulate a minimal blood-brain barrier and found that both were highly restrictive of virus penetration. A small amount of virus succeeded in crossing both barriers and was sufficient to establish infection of human glia. These data have direct implications for mechanisms used by JCPyV to invade the CNS and cause neurological disease.
Collapse
Affiliation(s)
- Avraham S. Lukacher
- Department of Cell Biology, Biochemistry, and Molecular Biology, Brown University, Providence, Rhode Island, USA
| | - Bethany A. O'Hara
- Department of Cell Biology, Biochemistry, and Molecular Biology, Brown University, Providence, Rhode Island, USA
| | - Wenqing Yuan
- Department of Cell Biology, Biochemistry, and Molecular Biology, Brown University, Providence, Rhode Island, USA
| | - Kaitlin Garabian
- Department of Cell Biology, Biochemistry, and Molecular Biology, Brown University, Providence, Rhode Island, USA
| | - Jacob Kaiserman
- Department of Cell Biology, Biochemistry, and Molecular Biology, Brown University, Providence, Rhode Island, USA
| | - Evan MacLure
- Department of Cell Biology, Biochemistry, and Molecular Biology, Brown University, Providence, Rhode Island, USA
| | - Sheila A. Haley
- Department of Cell Biology, Biochemistry, and Molecular Biology, Brown University, Providence, Rhode Island, USA
| | - Walter J. Atwood
- Department of Cell Biology, Biochemistry, and Molecular Biology, Brown University, Providence, Rhode Island, USA
| |
Collapse
|
2
|
Sandberg AL, Bond ACS, Bennett LJ, Craig SE, Winski DP, Kirkby LC, Kraemer AR, Kelly KG, Hess ST, Maginnis MS. GPCR Inhibitors Have Antiviral Properties against JC Polyomavirus Infection. Viruses 2024; 16:1559. [PMID: 39459893 PMCID: PMC11512265 DOI: 10.3390/v16101559] [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: 08/30/2024] [Revised: 09/24/2024] [Accepted: 09/28/2024] [Indexed: 10/28/2024] Open
Abstract
JC polyomavirus (JCPyV) infects the majority of the population and initially establishes a persistent but asymptomatic infection of the kidneys. In healthy individuals, the infection remains controlled by the host immune system, but for individuals experiencing prolonged immunosuppression, the infection can reactivate and spread to the brain, where it causes progressive multifocal leukoencephalopathy (PML), which is a fatal neurodegenerative disease. Currently, there are no approved therapies to treat PML, and affected individuals suffer rapid motor weakness and cognitive deterioration. To identify novel therapeutic treatments for JCPyV infection, receptor agonists/antagonists identified in a previously published drug screen were evaluated for their antiviral properties. Seven drugs were selected and validated using infectivity assays, and the mechanism of inhibition was further explored for G protein coupled receptor (GPCR)-associated inhibitors due to the role of the GPCR 5-hydroxytryptamine 2 receptors (5-HT2Rs) in JCPyV entry. The inhibitors cetirizine and paroxetine both reduced infection early in the JCPyV infectious cycle. Paroxetine specifically reduced viral internalization through altering the receptor density of 5-HT2CR, inhibiting β-arrestin recruitment to the receptor, and reducing MAPK signaling through ERK. These findings highlight the potential of receptor signaling and viral entry mechanisms as possible targets for antiviral drug development. Further, this research suggests that FDA-approved receptor agonists/antagonists currently used to treat other medical conditions could be repurposed into antivirals for the possible treatment of JCPyV infection and the fatal disease PML.
Collapse
Affiliation(s)
- Amanda L. Sandberg
- Department of Molecular and Biomedical Sciences, University of Maine, Orono, ME 04469, USA; (A.L.S.); (A.C.S.B.); (L.J.B.); (S.E.C.); (L.C.K.); (A.R.K.); (K.G.K.)
| | - Avery C. S. Bond
- Department of Molecular and Biomedical Sciences, University of Maine, Orono, ME 04469, USA; (A.L.S.); (A.C.S.B.); (L.J.B.); (S.E.C.); (L.C.K.); (A.R.K.); (K.G.K.)
| | - Lucas J. Bennett
- Department of Molecular and Biomedical Sciences, University of Maine, Orono, ME 04469, USA; (A.L.S.); (A.C.S.B.); (L.J.B.); (S.E.C.); (L.C.K.); (A.R.K.); (K.G.K.)
| | - Sophie E. Craig
- Department of Molecular and Biomedical Sciences, University of Maine, Orono, ME 04469, USA; (A.L.S.); (A.C.S.B.); (L.J.B.); (S.E.C.); (L.C.K.); (A.R.K.); (K.G.K.)
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME 04469, USA;
| | - David P. Winski
- Department of Physics & Astronomy, University of Maine, Orono, ME 04469, USA;
| | - Lara C. Kirkby
- Department of Molecular and Biomedical Sciences, University of Maine, Orono, ME 04469, USA; (A.L.S.); (A.C.S.B.); (L.J.B.); (S.E.C.); (L.C.K.); (A.R.K.); (K.G.K.)
| | - Abby R. Kraemer
- Department of Molecular and Biomedical Sciences, University of Maine, Orono, ME 04469, USA; (A.L.S.); (A.C.S.B.); (L.J.B.); (S.E.C.); (L.C.K.); (A.R.K.); (K.G.K.)
| | - Kristina G. Kelly
- Department of Molecular and Biomedical Sciences, University of Maine, Orono, ME 04469, USA; (A.L.S.); (A.C.S.B.); (L.J.B.); (S.E.C.); (L.C.K.); (A.R.K.); (K.G.K.)
| | - Samuel T. Hess
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME 04469, USA;
- Department of Physics & Astronomy, University of Maine, Orono, ME 04469, USA;
| | - Melissa S. Maginnis
- Department of Molecular and Biomedical Sciences, University of Maine, Orono, ME 04469, USA; (A.L.S.); (A.C.S.B.); (L.J.B.); (S.E.C.); (L.C.K.); (A.R.K.); (K.G.K.)
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME 04469, USA;
| |
Collapse
|
3
|
Kanse S, Khandelwal M, Pandey RK, Khokhar M, Desai N, Kumbhar BV. Designing a Multi-Epitope Subunit Vaccine against VP1 Major Coat Protein of JC Polyomavirus. Vaccines (Basel) 2023; 11:1182. [PMID: 37514998 PMCID: PMC10386578 DOI: 10.3390/vaccines11071182] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 06/27/2023] [Accepted: 06/28/2023] [Indexed: 07/30/2023] Open
Abstract
The JC polyomavirus virus (JCPyV) affects more than 80% of the human population in their early life stage. It mainly affects immunocompromised individuals where virus replication in oligodendrocytes and astrocytes may lead to fatal progressive multifocal encephalopathy (PML). Virus protein 1 (VP1) is one of the major structural proteins of the viral capsid, responsible for keeping the virus alive in the gastrointestinal and urinary tracts. VP1 is often targeted for antiviral drug and vaccine development. Similarly, this study implied immune-informatics and molecular modeling methods to design a multi-epitope subunit vaccine targeting JCPyV. The VP1 protein epitopic sequences, which are highly conserved, were used to build the vaccine. This designed vaccine includes two adjuvants, five HTL epitopes, five CTL epitopes, and two BCL epitopes to stimulate cellular, humoral, and innate immune responses against the JCPyV. Furthermore, molecular dynamics simulation (100 ns) studies were used to examine the interaction and stability of the vaccine protein with TLR4. Trajectory analysis showed that the vaccine and TLR4 receptor form a stable complex. Overall, this study may contribute to the path of vaccine development against JCPyV.
Collapse
Affiliation(s)
- Sukhada Kanse
- Department of Biological Sciences, Sunandan Divatia School of Science, NMIMS (Deemed to be) University, Vile Parle (West), Mumbai 400056, Maharashtra, India (N.D.)
| | - Mehak Khandelwal
- Department of Biological Sciences, Sunandan Divatia School of Science, NMIMS (Deemed to be) University, Vile Parle (West), Mumbai 400056, Maharashtra, India (N.D.)
| | - Rajan Kumar Pandey
- Department of Medical Biochemistry and Biophysics, Karolinska Institute, 17177 Stockholm, Sweden
| | - Manoj Khokhar
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS), Jodhpur 342005, Rajasthan, India
| | - Neetin Desai
- Department of Biological Sciences, Sunandan Divatia School of Science, NMIMS (Deemed to be) University, Vile Parle (West), Mumbai 400056, Maharashtra, India (N.D.)
| | - Bajarang Vasant Kumbhar
- Department of Biological Sciences, Sunandan Divatia School of Science, NMIMS (Deemed to be) University, Vile Parle (West), Mumbai 400056, Maharashtra, India (N.D.)
| |
Collapse
|
4
|
Maginnis MS. β-arrestins and G protein-coupled receptor kinases in viral entry: A graphical review. Cell Signal 2023; 102:110558. [PMID: 36509265 PMCID: PMC9811579 DOI: 10.1016/j.cellsig.2022.110558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 12/07/2022] [Accepted: 12/08/2022] [Indexed: 12/14/2022]
Abstract
Viruses rely on host-cell machinery in order to invade host cells and carry out a successful infection. G-protein coupled receptor (GPCR)-mediated signaling pathways are master regulators of cellular physiological processing and are an attractive target for viruses to rewire cells during infection. In particular, the GPCR-associated scaffolding proteins β-arrestins and GPCR signaling effectors G-protein receptor kinases (GRKs) have been identified as key cellular factors that mediate viral entry and orchestrate signaling pathways that reprogram cells for viral replication. Interestingly, a broad range of viruses have been identified to activate and/or require GPCR-mediated pathways for infection, including polyomaviruses, flaviviruses, influenza virus, and SARS-CoV-2, demonstrating that these viruses may have conserved mechanisms of host-cell invasion. Thus, GPCR-mediated pathways highlight an attractive target for the development of broad antiviral therapies.
Collapse
Affiliation(s)
- Melissa S Maginnis
- Molecular and Biomedical Sciences, The University of Maine, Orono, ME 04469, United States of America; Graduate School of Biomedical Science and Engineering, The University of Maine, Orono, ME 04469, United States of America.
| |
Collapse
|
5
|
Mehmood K, Wilczek MP, DuShane JK, Parent MT, Mayberry CL, Wallace JN, Levasseur FL, Fong TM, Hess ST, Maginnis MS. Dynamics and Patterning of 5-Hydroxytryptamine 2 Subtype Receptors in JC Polyomavirus Entry. Viruses 2022; 14:2597. [PMID: 36560603 PMCID: PMC9782046 DOI: 10.3390/v14122597] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 11/16/2022] [Accepted: 11/18/2022] [Indexed: 11/24/2022] Open
Abstract
The organization and dynamics of plasma membrane receptors are a critical link in virus-receptor interactions, which finetune signaling efficiency and determine cellular responses during infection. Characterizing the mechanisms responsible for the active rearrangement and clustering of receptors may aid in developing novel strategies for the therapeutic treatment of viruses. Virus-receptor interactions are poorly understood at the nanoscale, yet they present an attractive target for the design of drugs and for the illumination of viral infection and pathogenesis. This study utilizes super-resolution microscopy and related techniques, which surpass traditional microscopy resolution limitations, to provide both a spatial and temporal assessment of the interactions of human JC polyomavirus (JCPyV) with 5-hydroxytrypamine 2 receptors (5-HT2Rs) subtypes during viral entry. JCPyV causes asymptomatic kidney infection in the majority of the population and can cause fatal brain disease, and progressive multifocal leukoencephalopathy (PML), in immunocompromised individuals. Using Fluorescence Photoactivation Localization Microscopy (FPALM), the colocalization of JCPyV with 5-HT2 receptor subtypes (5-HT2A, 5-HT2B, and 5-HT2C) during viral attachment and viral entry was analyzed. JCPyV was found to significantly enhance the clustering of 5-HT2 receptors during entry. Cluster analysis of infected cells reveals changes in 5-HT2 receptor cluster attributes, and radial distribution function (RDF) analyses suggest a significant increase in the aggregation of JCPyV particles colocalized with 5-HT2 receptor clusters in JCPyV-infected samples. These findings provide novel insights into receptor patterning during viral entry and highlight improved technologies for the future development of therapies for JCPyV infection as well as therapies for diseases involving 5-HT2 receptors.
Collapse
Affiliation(s)
- Kashif Mehmood
- Department of Molecular and Biomedical Sciences, The University of Maine, Orono, ME 04469, USA
| | - Michael P. Wilczek
- Department of Molecular and Biomedical Sciences, The University of Maine, Orono, ME 04469, USA
| | - Jeanne K. DuShane
- Department of Molecular and Biomedical Sciences, The University of Maine, Orono, ME 04469, USA
| | - Matthew T. Parent
- Department of Physics & Astronomy, The University of Maine, Orono, ME 04469, USA
| | - Colleen L. Mayberry
- Department of Molecular and Biomedical Sciences, The University of Maine, Orono, ME 04469, USA
| | - Jaqulin N. Wallace
- Department of Physics & Astronomy, The University of Maine, Orono, ME 04469, USA
| | - Francois L. Levasseur
- Department of Molecular and Biomedical Sciences, The University of Maine, Orono, ME 04469, USA
| | - Tristan M. Fong
- Department of Molecular and Biomedical Sciences, The University of Maine, Orono, ME 04469, USA
| | - Samuel T. Hess
- Department of Physics & Astronomy, The University of Maine, Orono, ME 04469, USA
- Graduate School of Biomedical Science and Engineering, The University of Maine, Orono, ME 04469, USA
| | - Melissa S. Maginnis
- Department of Molecular and Biomedical Sciences, The University of Maine, Orono, ME 04469, USA
- Graduate School of Biomedical Science and Engineering, The University of Maine, Orono, ME 04469, USA
| |
Collapse
|
6
|
Morris-Love J, Atwood WJ. Complexities of JC Polyomavirus Receptor-Dependent and -Independent Mechanisms of Infection. Viruses 2022; 14:1130. [PMID: 35746603 PMCID: PMC9228512 DOI: 10.3390/v14061130] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 05/19/2022] [Accepted: 05/20/2022] [Indexed: 02/05/2023] Open
Abstract
JC polyomavirus (JCPyV) is a small non-enveloped virus that establishes lifelong, persistent infection in most of the adult population. Immune-competent patients are generally asymptomatic, but immune-compromised and immune-suppressed patients are at risk for the neurodegenerative disease progressive multifocal leukoencephalopathy (PML). Studies with purified JCPyV found it undergoes receptor-dependent infectious entry requiring both lactoseries tetrasaccharide C (LSTc) attachment and 5-hydroxytryptamine type 2 entry receptors. Subsequent work discovered the major targets of JCPyV infection in the central nervous system (oligodendrocytes and astrocytes) do not express the required attachment receptor at detectable levels, virus could not bind these cells in tissue sections, and viral quasi-species harboring recurrent mutations in the binding pocket for attachment. While several research groups found evidence JCPyV can use novel receptors for infection, it was also discovered that extracellular vesicles (EVs) can mediate receptor independent JCPyV infection. Recent work also found JCPyV associated EVs include both exosomes and secretory autophagosomes. EVs effectively present a means of immune evasion and increased tissue tropism that complicates viral studies and anti-viral therapeutics. This review focuses on JCPyV infection mechanisms and EV associated and outlines key areas of study necessary to understand the interplay between virus and extracellular vesicles.
Collapse
Affiliation(s)
- Jenna Morris-Love
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, RI 02912, USA;
- Pathobiology Graduate Program, Brown University, Providence, RI 02912, USA
| | - Walter J. Atwood
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, RI 02912, USA;
| |
Collapse
|
7
|
Morris‐Love J, O'Hara BA, Gee GV, Dugan AS, O'Rourke RS, Armstead BE, Assetta B, Haley SA, Atwood WJ. Biogenesis of JC polyomavirus associated extracellular vesicles. JOURNAL OF EXTRACELLULAR BIOLOGY 2022; 1:e43. [PMID: 36688929 PMCID: PMC9854252 DOI: 10.1002/jex2.43] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 04/09/2022] [Accepted: 04/13/2022] [Indexed: 01/26/2023]
Abstract
JC polyomavirus (JCPyV) is a small, non-enveloped virus that persists in the kidney in about half the adult population. In severely immune-compromised individuals JCPyV causes the neurodegenerative disease progressive multifocal leukoencephalopathy (PML) in the brain. JCPyV has been shown to infect cells by both direct and indirect mechanisms, the latter involving extracellular vesicle (EV) mediated infection. While direct mechanisms of infection are well studied indirect EV mediated mechanisms are poorly understood. Using a combination of chemical and genetic approaches we show that several overlapping intracellular pathways are responsible for the biogenesis of virus containing EV. Here we show that targeting neutral sphingomyelinase 2 (nSMase2) with the drug cambinol decreased the spread of JCPyV over several viral life cycles. Genetic depletion of nSMase2 by either shRNA or CRISPR/Cas9 reduced EV-mediated infection. Individual knockdown of seven ESCRT-related proteins including HGS, ALIX, TSG101, VPS25, VPS20, CHMP4A, and VPS4A did not significantly reduce JCPyV associated EV (JCPyV(+) EV) infectivity, whereas knockdown of the tetraspanins CD9 and CD81 or trafficking and/or secretory autophagy-related proteins RAB8A, RAB27A, and GRASP65 all significantly reduced the spread of JCPyV and decreased EV-mediated infection. These findings point to a role for exosomes and secretory autophagosomes in the biogenesis of JCPyV associated EVs with specific roles for nSMase2, CD9, CD81, RAB8A, RAB27A, and GRASP65 proteins.
Collapse
Affiliation(s)
- Jenna Morris‐Love
- Graduate Program in PathobiologyBrown UniversityProvidenceRIUSA
- Department of Molecular biologyCellular Biologyand BiochemistryBrown UniversityProvidenceRIUSA
| | - Bethany A. O'Hara
- Department of Molecular biologyCellular Biologyand BiochemistryBrown UniversityProvidenceRIUSA
| | - Gretchen V. Gee
- Department of Molecular biologyCellular Biologyand BiochemistryBrown UniversityProvidenceRIUSA
- MassBiologicsUniversity of Massachusetts Medical SchoolFall RiverMAUSA
| | - Aisling S. Dugan
- Department of BiologyAssumption UniversityWorcesterMAUSA
- Department of Molecular Microbiology and ImmunologyBrown UniversityProvidenceRIUSA
| | - Ryan S. O'Rourke
- Graduate Program in PathobiologyBrown UniversityProvidenceRIUSA
- Department of Molecular biologyCellular Biologyand BiochemistryBrown UniversityProvidenceRIUSA
| | | | - Benedetta Assetta
- Department of Molecular biologyCellular Biologyand BiochemistryBrown UniversityProvidenceRIUSA
| | - Sheila A. Haley
- Department of Molecular biologyCellular Biologyand BiochemistryBrown UniversityProvidenceRIUSA
| | - Walter J. Atwood
- Department of Molecular biologyCellular Biologyand BiochemistryBrown UniversityProvidenceRIUSA
| |
Collapse
|
8
|
Zheng HC, Xue H, Jin YZ, Jiang HM, Cui ZG. The Oncogenic Effects, Pathways, and Target Molecules of JC Polyoma Virus T Antigen in Cancer Cells. Front Oncol 2022; 12:744886. [PMID: 35350574 PMCID: PMC8958009 DOI: 10.3389/fonc.2022.744886] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 01/24/2022] [Indexed: 11/13/2022] Open
Abstract
JC polyoma virus (JCPyV) is a ubiquitous polyoma virus that infects the individual to cause progressive multifocal leukoencephalopathy and malignancies. Here, we found that T-antigen knockdown suppressed proliferation, glycolysis, mitochondrial respiration, migration, and invasion, and induced apoptosis and G2 arrest. The reverse was true for T-antigen overexpression, with overexpression of Akt, survivin, retinoblastoma protein, β-catenin, β-transducin repeat-containing protein (TRCP), and inhibitor of growth (ING)1, and the underexpression of mammalian target of rapamycin (mTOR), phosphorylated (p)-mTOR, p-p38, Cyclin D1, p21, vascular endothelial growth factor (VEGF), ING2, and ING4 in hepatocellular and pancreatic cancer cells and tissues. In lens tumor cells, T antigen transcriptionally targeted viral carcinogenesis, microRNAs in cancer, focal adhesion, p53, VEGF, phosphoinositide 3 kinase-Akt, and Forkhead box O signaling pathways, fructose and mannose metabolism, ribosome biosynthesis, and choline and pyrimidine metabolism. At a metabolomics level, it targeted protein digestion and absorption, aminoacryl-tRNA biosynthesis, biosynthesis of amino acids, and the AMPK signal pathway. At a proteomic level, it targeted ribosome biogenesis in eukaryotes, citrate cycle, carbon metabolism, protein digestion and absorption, aminoacryl-tRNA biosynthesis, extracellular-matrix-receptor interaction, and biosynthesis of amino acids. In lens tumor cells, T antigen might interact with various keratins, ribosomal proteins, apolipoproteins, G proteins, ubiquitin-related proteins, RPL19, β-catenin, β-TRCP, p53, and CCAAT-enhancer-binding proteins in lens tumor cells. T antigen induced a more aggressive phenotype in mouse and human cancer cells due to oncogene activation, inactivation of tumor suppressors, and disruption of metabolism, cell adhesion, and long noncoding RNA-microRNA-target axes.
Collapse
Affiliation(s)
- Hua-Chuan Zheng
- Department of Oncology and Experimental Center, The Affiliated Hospital of Chengde Medical University, Chengde, China
| | - Hang Xue
- Department of Oncology and Experimental Center, The Affiliated Hospital of Chengde Medical University, Chengde, China
| | - Yu-Zi Jin
- Department of Pediatrics, The Affiliated Hospital of Chengde Medical University, Chengde, China
| | - Hua-Mao Jiang
- Department of Urology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Zheng-Guo Cui
- Department of Environmental Health, University of Fukui School of Medical Science, Fukui, Japan
| |
Collapse
|
9
|
Cortés-Llanos B, Wang Y, Sims CE, Allbritton NL. A technology of a different sort: microraft arrays. LAB ON A CHIP 2021; 21:3204-3218. [PMID: 34346456 PMCID: PMC8387436 DOI: 10.1039/d1lc00506e] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
A common procedure performed throughout biomedical research is the selection and isolation of biological entities such as organelles, cells and organoids from a mixed population. In this review, we describe the development and application of microraft arrays, an analysis and isolation platform which enables a vast range of criteria and strategies to be used when separating biological entities. The microraft arrays are comprised of elastomeric microwells with detachable polymer bases (microrafts) that act as capture and culture sites as well as supporting carriers during cell isolation. The technology is elegant in its simplicity and can be implemented for samples possessing tens to millions of objects yielding a flexible platform for applications such as single-cell RNA sequencing, subcellular organelle capture and assay, high-throughput screening and development of CRISPR gene-edited cell lines, and organoid manipulation and selection. The transparent arrays are compatible with a multitude of imaging modalities enabling selection based on 2D or 3D spatial phenotypes or temporal properties. Each microraft can be individually isolated on demand with retention of high viability due to the near zero hydrodynamic stress imposed upon the cells during microraft release, capture and deposition. The platform has been utilized as a simple manual add-on to a standard microscope or incorporated into fully automated instruments that implement state-of-the-art imaging algorithms and machine learning. The vast array of selection criteria enables separations not possible with conventional sorting methods, thus garnering widespread interest in the biological and pharmaceutical sciences.
Collapse
|
10
|
GRK2 mediates β-arrestin interactions with 5-HT 2 receptors for JC polyomavirus endocytosis. J Virol 2021; 95:JVI.02139-20. [PMID: 33441347 PMCID: PMC8092707 DOI: 10.1128/jvi.02139-20] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
JC polyomavirus (JCPyV) infects the majority of the population, establishing a lifelong, asymptomatic infection in the kidney of healthy individuals. People that become severely immunocompromised may experience JCPyV reactivation, which can cause progressive multifocal leukoencephalopathy (PML), a neurodegenerative disease. Due to a lack of therapeutic options, PML results in fatality or significant debilitation among affected individuals. Cellular internalization of JCPyV is mediated by serotonin 5-hydroxytryptamine subfamily 2 receptors (5-HT2Rs) via clathrin-mediated endocytosis. The JCPyV entry process requires the clathrin-scaffolding proteins β-arrestin, adaptor protein 2 (AP2), and dynamin. Further, a β-arrestin interacting domain, the Ala-Ser-Lys (ASK) motif, within the C-terminus of 5-HT2AR is important for JCPyV internalization and infection. Interestingly, 5-HT2R subtypes A, B, and C equally support JCPyV entry and infection, and all subtypes contain an ASK motif, suggesting a conserved mechanism for viral entry. However, the role of the 5-HT2R ASK motifs and the activation of β-arrestin-associated proteins during internalization has not been fully elucidated. Through mutagenesis, the ASK motifs within 5-HT2BR and 5-HT2CR were identified as critical for JCPyV internalization and infectivity. Further, utilizing biochemical pulldown techniques, mutagenesis of the ASK motifs in 5-HT2BR and 5-HT2CR resulted in reduced β-arrestin binding. Utilizing small-molecule chemical inhibitors and RNA interference, G-protein receptor kinase 2 (GRK2) was determined to be required for JCPyV internalization and infection by mediating interactions between β-arrestin and the ASK motif of 5-HT2Rs. These findings demonstrate that GRK2 and β-arrestin interactions with 5-HT2Rs are critical for JCPyV entry by clathrin-mediated endocytosis and resultant infection.IMPORTANCE As intracellular parasites, viruses require a host cell to replicate and cause disease. Therefore, virus-host interactions contribute to viral pathogenesis. JC polyomavirus (JCPyV) infects most of the population, establishing a lifelong asymptomatic infection within the kidney. Under conditions of severe immunosuppression JCPyV may spread to the central nervous system, causing the fatal demyelinating disease progressive multifocal leukoencephalopathy (PML). Individuals living with HIV or undergoing immunomodulatory therapies are at risk for developing PML. The mechanisms of how JCPyV uses specific receptors on the surface of host cells to initiate internalization and infection is a poorly understood process. We have further identified cellular proteins involved in JCPyV internalization and infection and elucidated their specific interactions that are responsible for activation of receptors. Collectively, these findings illuminate how viruses usurp cellular receptors during infection, contributing to current development efforts for therapeutic options for the treatment or prevention of PML.
Collapse
|
11
|
Mayberry CL, Bond AC, Wilczek MP, Mehmood K, Maginnis MS. Sending mixed signals: polyomavirus entry and trafficking. Curr Opin Virol 2021; 47:95-105. [PMID: 33690104 DOI: 10.1016/j.coviro.2021.02.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/14/2021] [Accepted: 02/15/2021] [Indexed: 12/31/2022]
Abstract
Polyomaviruses are mostly non-pathogenic, yet some can cause human disease especially under conditions of immunosuppression, including JC, BK, and Merkel cell polyomaviruses. Direct interactions between viruses and the host early during infection dictate the outcome of disease, many of which remain enigmatic. However, significant work in recent years has contributed to our understanding of how this virus family establishes an infection, largely due to advances made for animal polyomaviruses murine and SV40. Here we summarize the major findings that have contributed to our understanding of polyomavirus entry, trafficking, disassembly, signaling, and immune evasion during the infectious process and highlight major unknowns in these processes that are open areas of study.
Collapse
Affiliation(s)
- Colleen L Mayberry
- Department of Molecular and Biomedical Sciences, The University of Maine, Orono, ME, USA
| | - Avery Cs Bond
- Department of Molecular and Biomedical Sciences, The University of Maine, Orono, ME, USA
| | - Michael P Wilczek
- Department of Molecular and Biomedical Sciences, The University of Maine, Orono, ME, USA
| | - Kashif Mehmood
- Department of Molecular and Biomedical Sciences, The University of Maine, Orono, ME, USA
| | - Melissa S Maginnis
- Department of Molecular and Biomedical Sciences, The University of Maine, Orono, ME, USA; Graduate School in Biomedical Sciences and Engineering, The University of Maine, Orono, ME, USA.
| |
Collapse
|
12
|
Direct Intracellular Visualization of Ebola Virus-Receptor Interaction by In Situ Proximity Ligation. mBio 2021; 12:mBio.03100-20. [PMID: 33436438 PMCID: PMC7844541 DOI: 10.1128/mbio.03100-20] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Ebola virus causes episodic but increasingly frequent outbreaks of severe disease in Middle Africa, as shown by the recently overcome second largest outbreak on record in the Democratic Republic of Congo. Despite considerable effort, FDA-approved antifiloviral therapeutics or targeted interventions are not available yet. Ebola virus (EBOV) entry into host cells comprises stepwise and extensive interactions of the sole viral surface glycoprotein (GP) with multiple host factors. During the intricate process, following virus uptake and trafficking to late endosomal/lysosomal compartments, GP is proteolytically processed to cleaved GP (GPCL) by the endosomal proteases cathepsin B and L, unmasking GP’s receptor-binding site. Engagement of GPCL with the universal filoviral intracellular receptor Niemann-Pick C1 (NPC1) eventually culminates in fusion between viral and cellular membranes, cytoplasmic escape of the viral nucleocapsid, and subsequent infection. Mechanistic delineation of the indispensable GPCL-NPC1-binding step has been severely hampered by the unavailability of a robust cell-based assay assessing interaction of GPCL with full-length endosomal NPC1. Here, we describe a novel in situ assay to monitor GPCL-NPC1 engagement in intact, infected cells. Visualization of the subcellular localization of binding complexes is based on the principle of DNA-assisted, antibody-mediated proximity ligation. Virus-receptor binding monitored by proximity ligation was contingent on GP’s proteolytic cleavage and was sensitive to perturbations in the GPCL-NPC1 interface. Our assay also specifically decoupled detection of virus-receptor binding from steps post-receptor binding, such as membrane fusion and infection. Testing of multiple FDA-approved small-molecule inhibitors revealed that drug treatments inhibited virus entry and GPCL-NPC1 recognition by distinctive mechanisms. Together, here we present a newly established proximity ligation assay, which will allow us to dissect cellular and viral requirements for filovirus-receptor binding and to delineate the mechanisms of action of inhibitors on filovirus entry in a cell-based system.
Collapse
|
13
|
Mayberry CL, Maginnis MS. Taking the Scenic Route: Polyomaviruses Utilize Multiple Pathways to Reach the Same Destination. Viruses 2020; 12:v12101168. [PMID: 33076363 PMCID: PMC7602598 DOI: 10.3390/v12101168] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 10/08/2020] [Accepted: 10/09/2020] [Indexed: 01/02/2023] Open
Abstract
Members of the Polyomaviridae family differ in their host range, pathogenesis, and disease severity. To date, some of the most studied polyomaviruses include human JC, BK, and Merkel cell polyomavirus and non-human subspecies murine and simian virus 40 (SV40) polyomavirus. Although dichotomies in host range and pathogenesis exist, overlapping features of the infectious cycle illuminate the similarities within this virus family. Of particular interest to human health, JC, BK, and Merkel cell polyomavirus have all been linked to critical, often fatal, illnesses, emphasizing the importance of understanding the underlying viral infections that result in the onset of these diseases. As there are significant overlaps in the capacity of polyomaviruses to cause disease in their respective hosts, recent advancements in characterizing the infectious life cycle of non-human murine and SV40 polyomaviruses are key to understanding diseases caused by their human counterparts. This review focuses on the molecular mechanisms by which different polyomaviruses hijack cellular processes to attach to host cells, internalize, traffic within the cytoplasm, and disassemble within the endoplasmic reticulum (ER), prior to delivery to the nucleus for viral replication. Unraveling the fundamental processes that facilitate polyomavirus infection provides deeper insight into the conserved mechanisms of the infectious process shared within this virus family, while also highlighting critical unique viral features.
Collapse
Affiliation(s)
- Colleen L. Mayberry
- Department of Molecular and Biomedical Sciences, The University of Maine, Orono, ME 04469, USA;
| | - Melissa S. Maginnis
- Department of Molecular and Biomedical Sciences, The University of Maine, Orono, ME 04469, USA;
- Graduate School in Biomedical Sciences and Engineering, The University of Maine, Orono, ME 04469, USA
- Correspondence:
| |
Collapse
|
14
|
Alstadhaug KB, Rinaldo CH, Osnes L, Sereti I, Ofte HK. Progressive multifocal leukoencephalopathy treated with interleukin-7. CLINICAL INFECTION IN PRACTICE 2020. [DOI: 10.1016/j.clinpr.2020.100049] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
15
|
Fifty Years of JC Polyomavirus: A Brief Overview and Remaining Questions. Viruses 2020; 12:v12090969. [PMID: 32882975 PMCID: PMC7552028 DOI: 10.3390/v12090969] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 08/28/2020] [Accepted: 08/30/2020] [Indexed: 12/11/2022] Open
Abstract
In the fifty years since the discovery of JC polyomavirus (JCPyV), the body of research representing our collective knowledge on this virus has grown substantially. As the causative agent of progressive multifocal leukoencephalopathy (PML), an often fatal central nervous system disease, JCPyV remains enigmatic in its ability to live a dual lifestyle. In most individuals, JCPyV reproduces benignly in renal tissues, but in a subset of immunocompromised individuals, JCPyV undergoes rearrangement and begins lytic infection of the central nervous system, subsequently becoming highly debilitating-and in many cases, deadly. Understanding the mechanisms allowing this process to occur is vital to the development of new and more effective diagnosis and treatment options for those at risk of developing PML. Here, we discuss the current state of affairs with regards to JCPyV and PML; first summarizing the history of PML as a disease and then discussing current treatment options and the viral biology of JCPyV as we understand it. We highlight the foundational research published in recent years on PML and JCPyV and attempt to outline which next steps are most necessary to reduce the disease burden of PML in populations at risk.
Collapse
|
16
|
Bouma EM, van de Pol DPI, Sanders ID, Rodenhuis-Zybert IA, Smit JM. Serotonergic Drugs Inhibit Chikungunya Virus Infection at Different Stages of the Cell Entry Pathway. J Virol 2020; 94:e00274-20. [PMID: 32321803 PMCID: PMC7307168 DOI: 10.1128/jvi.00274-20] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 04/12/2020] [Indexed: 12/15/2022] Open
Abstract
Chikungunya virus (CHIKV) is an important reemerging human pathogen transmitted by mosquitoes. The virus causes an acute febrile illness, chikungunya fever, which is characterized by headache, rash, and debilitating (poly)arthralgia that can reside for months to years after infection. Currently, effective antiviral therapies and vaccines are lacking. Due to the high morbidity and economic burden in the countries affected by CHIKV, there is a strong need for new strategies to inhibit CHIKV replication. The serotonergic drug 5-nonyloxytryptamine (5-NT) was previously identified as a potential host-directed inhibitor for CHIKV infection. In this study, we determined the mechanism of action by which the serotonin receptor agonist 5-NT controls CHIKV infection. Using time-of-addition and entry bypass assays, we found that 5-NT predominantly inhibits CHIKV in the early phases of the replication cycle, at a step prior to RNA translation and genome replication. Intriguingly, however, no effect was seen during virus-cell binding, internalization, membrane fusion and genomic RNA (gRNA) release into the cell cytosol. In addition, we show that the serotonin receptor antagonist methiothepin mesylate (MM) also has antiviral properties toward CHIKV and specifically interferes with the cell entry process and/or membrane fusion. Taken together, pharmacological targeting of 5-HT receptors may represent a potent way to limit viral spread and disease severity.IMPORTANCE The rapid spread of mosquito-borne viral diseases in humans puts a huge economic burden on developing countries. For many of these infections, including those caused by chikungunya virus (CHIKV), there are no specific treatment possibilities to alleviate disease symptoms. Understanding the virus-host interactions that are involved in the viral replication cycle is imperative for the rational design of therapeutic strategies. In this study, we discovered an antiviral compound, elucidated its mechanism of action, and propose serotonergic drugs as potential host-directed antivirals for CHIKV.
Collapse
Affiliation(s)
- Ellen M Bouma
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Denise P I van de Pol
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Ilson D Sanders
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Izabela A Rodenhuis-Zybert
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Jolanda M Smit
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
17
|
Haley SA, O'Hara BA, Atwood WJ. Adipocyte Plasma Membrane Protein (APMAP) promotes JC Virus (JCPyV) infection in human glial cells. Virology 2020; 548:17-24. [PMID: 32838939 DOI: 10.1016/j.virol.2020.06.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 06/01/2020] [Accepted: 06/01/2020] [Indexed: 11/16/2022]
Abstract
The demyelinating disease progressive multifocal leukoencephalopathy (PML) is caused by the human polyomavirus, JCPyV, under conditions of prolonged immunosuppression. Initial infection is asymptomatic, and the virus establishes lifelong persistence in the host. Following the loss of immune surveillance, the virus can traffic to the central nervous system and infect oligodendrocytes to cause demyelination and PML. The mechanisms involved in glial cell infection are not completely understood. In a screen for N-glycosylated proteins that influence JCPyV pathology, we identified Adipocyte Plasma Membrane Associated Protein (APMAP) as a host cell modulator of JCPyV infection. The removal of APMAP by small interfering siRNA as well as by CRISPR-Cas9 gene editing resulted in a significant decrease in JCPyV infection. Exogenous expression of APMAP in APMAP knockout cell lines rescued susceptibility to infection. These data suggest that virus infection of glial cells is dependent on APMAP.
Collapse
Affiliation(s)
- Sheila A Haley
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, USA.
| | - Bethany A O'Hara
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, USA
| | - Walter J Atwood
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, USA.
| |
Collapse
|
18
|
O’Hara BA, Morris-Love J, Gee GV, Haley SA, Atwood WJ. JC Virus infected choroid plexus epithelial cells produce extracellular vesicles that infect glial cells independently of the virus attachment receptor. PLoS Pathog 2020; 16:e1008371. [PMID: 32130281 PMCID: PMC7075641 DOI: 10.1371/journal.ppat.1008371] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 03/16/2020] [Accepted: 01/31/2020] [Indexed: 12/13/2022] Open
Abstract
The human polyomavirus, JCPyV, is the causative agent of progressive multifocal leukoencephalopathy (PML) in immunosuppressed and immunomodulated patients. Initial infection with JCPyV is common and the virus establishes a long-term persistent infection in the urogenital system of 50-70% of the human population worldwide. A major gap in the field is that we do not know how the virus traffics from the periphery to the brain to cause disease. Our recent discovery that human choroid plexus epithelial cells are fully susceptible to virus infection together with reports of JCPyV infection of choroid plexus in vivo has led us to hypothesize that the choroid plexus plays a fundamental role in this process. The choroid plexus is known to relay information between the blood and the brain by the release of extracellular vesicles. This is particularly important because human macroglia (oligodendrocytes and astrocytes), the major targets of virus infection in the central nervous system (CNS), do not express the known attachment receptors for the virus and do not bind virus in human tissue sections. In this report we show that JCPyV infected choroid plexus epithelial cells produce extracellular vesicles that contain JCPyV and readily transmit the infection to human glial cells. Transmission of the virus by extracellular vesicles is independent of the known virus attachment receptors and is not neutralized by antisera directed at the virus. We also show that extracellular vesicles containing virus are taken into target glial cells by both clathrin dependent endocytosis and macropinocytosis. Our data support the hypothesis that the choroid plexus plays a fundamental role in the dissemination of virus to brain parenchyma.
Collapse
Affiliation(s)
- Bethany A. O’Hara
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, Rhode Island, United States of America
| | - Jenna Morris-Love
- Graduate Program in Pathobiology, Brown University, Providence, Rhode Island, United States of America
| | - Gretchen V. Gee
- MassBiologics, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Sheila A. Haley
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, Rhode Island, United States of America
| | - Walter J. Atwood
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, Rhode Island, United States of America
| |
Collapse
|
19
|
Del Valle L, Piña-Oviedo S. Human Polyomavirus JCPyV and Its Role in Progressive Multifocal Leukoencephalopathy and Oncogenesis. Front Oncol 2019; 9:711. [PMID: 31440465 PMCID: PMC6694743 DOI: 10.3389/fonc.2019.00711] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 07/17/2019] [Indexed: 12/12/2022] Open
Abstract
The human neurotropic virus JCPyV, a member of the Polyomaviridiae family, is the opportunistic infectious agent of Progressive Multifocal Leukoencephalopathy (PML), a fatal disease seen in severe immunosuppressive conditions and, during the last decade, in patients undergoing immunotherapy. JCPyV is a ubiquitous pathogen with up to 85% of the adult population word-wide exhibiting antibodies against it. Early experiments demonstrated that direct inoculation of JCPyV into the brain of different species resulted in the development of brain tumors and other neuroectodermal-derived neoplasias. Later, several reports showed the detection of viral sequences in medulloblastomas and glial tumors, as well as expression of the viral protein T-Antigen. Few oncogenic viruses, however, have caused so much controversy regarding their role in the pathogenesis of brain tumors, but the discovery of new Polyomaviruses that cause Merkel cell carcinomas in humans and brain tumors in racoons, in addition to the role of JCPyV in colon cancer and multiple mechanistic studies have shed much needed light on the role of JCPyV in cancer. The pathways affected by the viral protein T-Antigen include cell cycle regulators, like p53 and pRb, and transcription factors that activate pro-proliferative genes, like c-Myc. In addition, infection with JCPyV causes chromosomal damage and T-Antigen inhibits homologous recombination, and activates anti-apoptotic proteins, such as Survivin. Here we review the different aspects of the biology and physiopathology of JCPyV.
Collapse
Affiliation(s)
- Luis Del Valle
- Department of Pathology and Stanley S. Scott Cancer Center, Louisiana State University Health, New Orleans, LA, United States
| | - Sergio Piña-Oviedo
- Department of Pathology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| |
Collapse
|