1
|
Hassan Z, Kumar ND, Reggiori F, Khan G. How Viruses Hijack and Modify the Secretory Transport Pathway. Cells 2021; 10:2535. [PMID: 34685515 PMCID: PMC8534161 DOI: 10.3390/cells10102535] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 08/28/2021] [Accepted: 09/06/2021] [Indexed: 12/23/2022] Open
Abstract
Eukaryotic cells contain dynamic membrane-bound organelles that are constantly remodeled in response to physiological and environmental cues. Key organelles are the endoplasmic reticulum, the Golgi apparatus and the plasma membrane, which are interconnected by vesicular traffic through the secretory transport route. Numerous viruses, especially enveloped viruses, use and modify compartments of the secretory pathway to promote their replication, assembly and cell egression by hijacking the host cell machinery. In some cases, the subversion mechanism has been uncovered. In this review, we summarize our current understanding of how the secretory pathway is subverted and exploited by viruses belonging to Picornaviridae, Coronaviridae, Flaviviridae,Poxviridae, Parvoviridae and Herpesviridae families.
Collapse
Affiliation(s)
- Zubaida Hassan
- Department of Medical Microbiology and Immunology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 17666, United Arab Emirates;
- Department of Microbiology, School of Life Sciences, Modibbo Adama University, Yola PMB 2076, Nigeria
| | - Nilima Dinesh Kumar
- Department of Biomedical Sciences of Cells and Systems, University of Groningen, University Medical Center Groningen, 9713 AV Groningen, The Netherlands; (N.D.K.); (F.R.)
- Department of Medical Microbiology and Infection Prevention, University of Groningen, University Medical Center Groningen, 9713 AV Groningen, The Netherlands
| | - Fulvio Reggiori
- Department of Biomedical Sciences of Cells and Systems, University of Groningen, University Medical Center Groningen, 9713 AV Groningen, The Netherlands; (N.D.K.); (F.R.)
| | - Gulfaraz Khan
- Department of Medical Microbiology and Immunology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 17666, United Arab Emirates;
| |
Collapse
|
2
|
Neulinger-Muñoz M, Schaack D, Grekova SP, Bauer AS, Giese T, Salg GA, Espinet E, Leuchs B, Heller A, Nüesch JPF, Schenk M, Volkmar M, Giese NA. Human Retrotransposons and the Global Shutdown of Homeostatic Innate Immunity by Oncolytic Parvovirus H-1PV in Pancreatic Cancer. Viruses 2021; 13:v13061019. [PMID: 34071585 PMCID: PMC8228339 DOI: 10.3390/v13061019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 05/14/2021] [Accepted: 05/25/2021] [Indexed: 12/20/2022] Open
Abstract
Although the oncolytic parvovirus H-1PV has entered clinical trials, predicting therapeutic success remains challenging. We investigated whether the antiviral state in tumor cells determines the parvoviral oncolytic efficacy. The interferon/interferon-stimulated genes (IFN/ISG)-circuit and its major configurator, human endogenous retroviruses (HERVs), were evaluated using qRT-PCR, ELISA, Western blot, and RNA-Seq techniques. In pancreatic cancer cell lines, H-1PV caused a late global shutdown of innate immunity, whereby the concomitant inhibition of HERVs and IFN/ISGs was co-regulatory rather than causative. The growth-inhibitory IC50 doses correlated with the power of suppression but not with absolute ISG levels. Moreover, H-1PV was not sensitive to exogenous IFN despite upregulated antiviral ISGs. Such resistance questioned the biological necessity of the oncotropic ISG-shutdown, which instead might represent a surrogate marker for personalized oncolytic efficacy. The disabled antiviral homeostasis may modify the activity of other viruses, as demonstrated by the reemergence of endogenous AluY-retrotransposons. This way of suppression may compromise the interferogenicity of drugs having gemcitabine-like mechanisms of action. This shortcoming in immunogenic cell death induction is however amendable by immune cells which release IFN in response to H-1PV.
Collapse
Affiliation(s)
- Matthias Neulinger-Muñoz
- Department of Surgery, European Pancreas Center, University Hospital Heidelberg, 69120 Heidelberg, Germany; (M.N.-M.); (S.P.G.); (G.A.S.); (A.H.); (M.S.)
| | - Dominik Schaack
- Department of Anesthesiology, University Hospital Heidelberg, 69120 Heidelberg, Germany;
| | - Svetlana P. Grekova
- Department of Surgery, European Pancreas Center, University Hospital Heidelberg, 69120 Heidelberg, Germany; (M.N.-M.); (S.P.G.); (G.A.S.); (A.H.); (M.S.)
| | - Andrea S. Bauer
- German Cancer Research Center (DKFZ), Division of Functional Genome Analysis, 69120 Heidelberg, Germany;
| | - Thomas Giese
- Institute of Immunology and German Center for Infection Research (DZIF), Partner Site Heidelberg, University Hospital Heidelberg, 69120 Heidelberg, Germany;
| | - Gabriel A. Salg
- Department of Surgery, European Pancreas Center, University Hospital Heidelberg, 69120 Heidelberg, Germany; (M.N.-M.); (S.P.G.); (G.A.S.); (A.H.); (M.S.)
| | - Elisa Espinet
- German Cancer Research Center (DKFZ), Division of Stem Cells and Cancer, 69120 Heidelberg, Germany;
- HI-STEM—Heidelberg Institute for Stem Cell Technology and Experimental Medicine GmbH, 69120 Heidelberg, Germany
| | - Barbara Leuchs
- German Cancer Research Center (DKFZ), Division of Tumor Virology, 69120 Heidelberg, Germany;
| | - Anette Heller
- Department of Surgery, European Pancreas Center, University Hospital Heidelberg, 69120 Heidelberg, Germany; (M.N.-M.); (S.P.G.); (G.A.S.); (A.H.); (M.S.)
| | - Jürg P. F. Nüesch
- German Cancer Research Center (DKFZ), Division of Virus-Associated Carcinogenesis F170, 69120 Heidelberg, Germany;
| | - Miriam Schenk
- Department of Surgery, European Pancreas Center, University Hospital Heidelberg, 69120 Heidelberg, Germany; (M.N.-M.); (S.P.G.); (G.A.S.); (A.H.); (M.S.)
| | - Michael Volkmar
- German Cancer Research Center (DKFZ), Division of Molecular Oncology of Gastrointestinal Tumors, 69120 Heidelberg, Germany;
| | - Nathalia A. Giese
- Department of Surgery, European Pancreas Center, University Hospital Heidelberg, 69120 Heidelberg, Germany; (M.N.-M.); (S.P.G.); (G.A.S.); (A.H.); (M.S.)
- Correspondence:
| |
Collapse
|
3
|
Non-viral gene delivery of the oncotoxic protein NS1 for treatment of hepatocellular carcinoma. J Control Release 2021; 334:138-152. [PMID: 33894304 DOI: 10.1016/j.jconrel.2021.04.023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 04/10/2021] [Accepted: 04/20/2021] [Indexed: 12/12/2022]
Abstract
Hepatocellular carcinoma (HCC) is related to increasing incidence rates and poor clinical outcomes due to lack of efficient treatment options and emerging resistance mechanisms. The aim of the present study is to exploit a non-viral gene therapy enabling the expression of the parvovirus-derived oncotoxic protein NS1 in HCC. This anticancer protein interacts with different cellular kinases mediating a multimodal host-cell death. Lipoplexes (LPX) designed to deliver a DNA expression plasmid encoding NS1 are characterized using a comprehensive set of in vitro assays. The mechanisms of cell death induction are assessed and phosphoinositide-dependent kinase 1 (PDK1) is identified as a potential predictive biomarker for a NS1-LPX-based gene therapy. In an HCC xenograft mouse model, NS1-LPX therapeutic approach results in a significant reduction in tumor growth and extended survival. Data provide convincing evidence for future studies using a targeted NS1 gene therapy for PDK1 overexpressing HCC.
Collapse
|
4
|
Chen X, Qi L, Su H, He Y, Li N, Gao Q, Li H, Xu T, Lu L, Xu Z, Tang J. Prenatal hypoxia attenuated contraction of offspring coronary artery associated with decreased PKCβ Ser 660 phosphorylation and intracellular calcium. Life Sci 2020; 261:118364. [PMID: 32866516 DOI: 10.1016/j.lfs.2020.118364] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 08/19/2020] [Accepted: 08/24/2020] [Indexed: 01/15/2023]
Abstract
AIMS Prenatal hypoxia (PH) could affect peripheral vascular tone of the offspring, thus increasing the risk of cardiovascular diseases in adult. However, it's still unknown whether functions of coronary arteries (COA) in adult offspring would be influenced by PH. The present study aimed at effects of PH on vascular tone of COA and its related mechanisms. METHODS Coronary arteries of adult offspring exposed to hypoxic or normoxic circumstances during gestational day 5 to 21 were collected. Wire myograph system, whole-cell patch clamp technique, IonOptix MyoCam system, PCR, and western blot were used to detect vascular function of adult offspring COA. KEY FINDINGS PH significantly attenuated serotonin- and phorbol 12, 13-dibutyrate (PDBu)-induced constriction. Iberiotoxin potentiated PDBu-induced constriction and the effect was augmented by PH, however, no significant differences were found in whole-cell BKCa channel currents and its protein expression. Nifedipine inhibited PDBu-mediated constriction and the inhibitory effect was reduced in PH group, and whole-cell calcium channel current was decreased in offspring COA. Besides, PH reduced the capability of calcium release from the endoplasmic reticulum in COA. The phosphorylated PKCβ protein expression at Ser660 site, not Thr641 site, was significantly decreased in PH offspring. Chronic hypoxia during pregnancy attenuated PDBu-mediated constriction in offspring COA, presumably through decreased phosphorylated PKCβ at serine660 sites and decreased intracellular calcium-related weaker PKC activation. SIGNIFICANCE The findings provided new information on the influence of prenatal hypoxia on COA, and suggested potential use of PKCβ-serine660 for early prevention of coronary heart diseases in developmental origins.
Collapse
Affiliation(s)
- Xueyi Chen
- Institute for Fetology, First Hospital of Soochow University, Suzhou, Jiangsu, China; School of Preclinical Medicine, Wannan Medical College, Wuhu, Anhui, China
| | - Linglu Qi
- Institute for Fetology, First Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Hongyu Su
- Institute for Fetology, First Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Yun He
- Institute for Fetology, First Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Na Li
- Institute for Fetology, First Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Qinqin Gao
- Institute for Fetology, First Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Huan Li
- Institute for Fetology, First Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Ting Xu
- Institute for Fetology, First Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Likui Lu
- Institute for Fetology, First Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Zhice Xu
- Institute for Fetology, First Hospital of Soochow University, Suzhou, Jiangsu, China.
| | - Jiaqi Tang
- Institute for Fetology, First Hospital of Soochow University, Suzhou, Jiangsu, China.
| |
Collapse
|
5
|
Hartley A, Kavishwar G, Salvato I, Marchini A. A Roadmap for the Success of Oncolytic Parvovirus-Based Anticancer Therapies. Annu Rev Virol 2020; 7:537-557. [PMID: 32600158 DOI: 10.1146/annurev-virology-012220-023606] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Autonomous rodent protoparvoviruses (PVs) are promising anticancer agents due to their excellent safety profile, natural oncotropism, and oncosuppressive activities. Viral infection can trigger immunogenic cell death, activating the immune system against the tumor. However, the efficacy of this treatment in recent clinical trials is moderate compared with results seen in preclinical work. Various strategies have been employed to improve the anticancer activities of oncolytic PVs, including development of second-generation parvoviruses with enhanced oncolytic and immunostimulatory activities and rational combination of PVs with other therapies. Understanding the cellular factors involved in the PV life cycle is another important area of investigation. Indeed, these studies may lead to the identification of biomarkers that would allow a more personalized use of PV-based therapies. This review focuses on this work and the challenges that still need to be overcome to move PVs forward into clinical practice as an effective therapeutic option for cancer patients.
Collapse
Affiliation(s)
- Anna Hartley
- Laboratory of Oncolytic Virus Immuno-Therapeutics, German Cancer Research Center, 69120 Heidelberg, Germany;
| | - Gayatri Kavishwar
- Laboratory of Oncolytic Virus Immuno-Therapeutics, German Cancer Research Center, 69120 Heidelberg, Germany;
| | - Ilaria Salvato
- Laboratory of Oncolytic Virus Immuno-Therapeutics, Luxembourg Institute of Health, L-1526 Luxembourg, Luxembourg;
| | - Antonio Marchini
- Laboratory of Oncolytic Virus Immuno-Therapeutics, German Cancer Research Center, 69120 Heidelberg, Germany; .,Laboratory of Oncolytic Virus Immuno-Therapeutics, Luxembourg Institute of Health, L-1526 Luxembourg, Luxembourg;
| |
Collapse
|
6
|
Heckman CA, Pandey P, Cayer ML, Biswas T, Zhang Z, Boudreau NS. The tumor promoter-activated protein kinase Cs are a system for regulating filopodia. Cytoskeleton (Hoboken) 2017; 74:297-314. [PMID: 28481056 PMCID: PMC5575509 DOI: 10.1002/cm.21373] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2016] [Revised: 04/28/2017] [Accepted: 05/01/2017] [Indexed: 01/02/2023]
Abstract
Different protein kinase C (PKC) isoforms have distinct roles in regulating cell functions. The conventional (α, β, γ) and novel (δ, ɛ, η, θ) classes are targets of phorbol ester tumor promoters, which are surrogates of endogenous second messenger, diacylglycerol. The promoter-stimulated disappearance of filopodia was investigated by use of blocking peptides (BPs) that inhibit PKC maturation and/or docking. Filopodia were partially rescued by a peptide representing PKC ɛ hydrophobic sequence, but also by a myristoylated PKC α/β pseudosubstrate sequence, and an inhibitor of T-cell protein tyrosine phosphatase (TC-PTP). The ability to turn over filopodia was widely distributed among PKC isoforms. PKC α and η hydrophobic sequences enhanced filopodia in cells in the absence of tumor promoter treatment. With transcriptional knockdown of PKC α, the content of PKC ɛ predominated over other isoforms. PKC ɛ could decrease filopodia significantly in promoter-treated cells, and this was attributed to ruffling. The presence of PKC α counteracted the PKC ɛ-mediated enhancement of ruffling. The results showed that there were two mechanisms of filopodia downregulation. One operated in the steady-state and relied on PKC α and η. The other was stimulated by tumor promoters and relied on PKC ɛ. Cycles of protrusion and retraction are characteristic of filopodia and are essential for the cell to orient itself during chemotaxis and haptotaxis. By suppressing filopodia, PKC ɛ can create a long-term "memory" of an environmental signal that may act in nature as a mnemonic device to mark the direction of a repulsive signal.
Collapse
Affiliation(s)
- Carol A. Heckman
- Department of Biological SciencesBowling Green State UniversityLife Sciences Building Room 217Bowling GreenOhio43403
| | - Pratima Pandey
- Department of Biological SciencesBowling Green State UniversityLife Sciences Building Room 217Bowling GreenOhio43403
| | - Marilyn L. Cayer
- Center for Microscopy and MicroanalysisBowling Green State UniversityLife Sciences Building Room 217Bowling GreenOhio43403
| | - Tania Biswas
- Department of Biological SciencesBowling Green State UniversityLife Sciences Building Room 217Bowling GreenOhio43403
| | - Zhong‐Yin Zhang
- Department of Medicinal Chemistry and Molecular PharmacologyPurdue UniversityRobert E. Heine Pharmacy Building, Room 202A, 575 Stadium Mall DriveWest LafayetteIndiana47907
| | - Nancy S. Boudreau
- Department of Applied Statistics and Operations ResearchBowling Green State University344 Business Administration BuildingBowling GreenOhio43403
| |
Collapse
|
7
|
Angelova AL, Geletneky K, Nüesch JPF, Rommelaere J. Tumor Selectivity of Oncolytic Parvoviruses: From in vitro and Animal Models to Cancer Patients. Front Bioeng Biotechnol 2015; 3:55. [PMID: 25954743 PMCID: PMC4406089 DOI: 10.3389/fbioe.2015.00055] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 04/05/2015] [Indexed: 11/23/2022] Open
Abstract
Oncolytic virotherapy of cancer is among the innovative modalities being under development and especially promising for targeting tumors, which are resistant to conventional treatments. Presently, at least a dozen of viruses, belonging to nine different virus families, are being tested within the frames of various clinical studies in cancer patients. Continuously growing preclinical evidence showing that the autonomous rat parvovirus H-1 (H-1PV) is able to kill tumor cells that resist conventional treatments and to achieve a complete cure of various human tumors in animal models argues for its inclusion in the arsenal of oncolytic viruses with an especially promising bench to bedside translation potential. Oncolytic parvovirus safe administration to humans relies on the intrinsic preference of these agents for quickly proliferating, metabolically, and biochemically disturbed tumor versus normal cells (tumor selectivity or oncotropism). The present review summarizes and discusses (i) preclinical evidence of H-1PV innocuousness for normal cells and healthy tissues in vitro and in animals, respectively, (ii) toxicological assessments of H-1PV mono- or combined therapy in tumor-bearing virus-permissive animal models, as well as (iii) historical results of experimental infection of human cancer patients with H-1PV. Altogether, these data argue against a risk of H-1PV inducing significant toxic effects in human patients. This highly favorable safety profile allowed the translation of H-1PV preclinical research into a Phase I/IIa clinical trial being currently in progress.
Collapse
Affiliation(s)
- Assia L Angelova
- Infection and Cancer Program, Division of Tumor Virology, German Cancer Research Center (DKFZ) , Heidelberg , Germany
| | - Karsten Geletneky
- Infection and Cancer Program, Division of Tumor Virology, German Cancer Research Center (DKFZ) , Heidelberg , Germany ; Department of Neurosurgery, University of Heidelberg , Heidelberg , Germany
| | - Jürg P F Nüesch
- Infection and Cancer Program, Division of Tumor Virology, German Cancer Research Center (DKFZ) , Heidelberg , Germany
| | - Jean Rommelaere
- Infection and Cancer Program, Division of Tumor Virology, German Cancer Research Center (DKFZ) , Heidelberg , Germany
| |
Collapse
|
8
|
Geletneky K, Nüesch JP, Angelova A, Kiprianova I, Rommelaere J. Double-faceted mechanism of parvoviral oncosuppression. Curr Opin Virol 2015; 13:17-24. [PMID: 25841215 DOI: 10.1016/j.coviro.2015.03.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Revised: 03/02/2015] [Accepted: 03/16/2015] [Indexed: 11/17/2022]
Abstract
The H-1 parvovirus (H-1PV) exerts oncosuppressive action that has two components: oncotoxicity and immunostimulation. While many human tumor cells, including conventional drug-resistant ones, can be killed by H-1PV, some fail to support progeny virus production, necessary for infection propagation in neoplastic tissues. This limitation can be overcome through forced selection of H-1PV variants capable of enhanced multiplication and spreading in human tumor cells. In the context of further developing H-1PV for use in cancer therapy, arming it with immunostimulatory CpG motifs under conditions preserving replication and oncolysis enhances its action as an anticancer vaccine adjuvant. A first clinical study of H-1PV treatment in glioma patients has yielded evidence of intratumoral synthesis of the viral oncotoxic protein NS1 and immune cell infiltration.
Collapse
Affiliation(s)
- Karsten Geletneky
- German Cancer Research Center, Infection and Cancer Program, Division of Tumor Virology, 69120 Heidelberg, Germany; Department of Neurosurgery, University Hospital, 69120 Heidelberg, Germany
| | - Jürg Pf Nüesch
- German Cancer Research Center, Infection and Cancer Program, Division of Tumor Virology, 69120 Heidelberg, Germany
| | - Assia Angelova
- German Cancer Research Center, Infection and Cancer Program, Division of Tumor Virology, 69120 Heidelberg, Germany
| | - Irina Kiprianova
- German Cancer Research Center, Infection and Cancer Program, Division of Tumor Virology, 69120 Heidelberg, Germany
| | - Jean Rommelaere
- German Cancer Research Center, Infection and Cancer Program, Division of Tumor Virology, 69120 Heidelberg, Germany.
| |
Collapse
|
9
|
Bär S, Rommelaere J, Nüesch JPF. PKCη/Rdx-driven phosphorylation of PDK1: a novel mechanism promoting cancer cell survival and permissiveness for parvovirus-induced lysis. PLoS Pathog 2015; 11:e1004703. [PMID: 25742010 PMCID: PMC4351090 DOI: 10.1371/journal.ppat.1004703] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Accepted: 01/26/2015] [Indexed: 01/12/2023] Open
Abstract
The intrinsic oncotropism and oncosuppressive activities of rodent protoparvoviruses (PVs) are opening new prospects for cancer virotherapy. Virus propagation, cytolytic activity, and spread are tightly connected to activation of the PDK1 signaling cascade, which delays stress-induced cell death and sustains functioning of the parvoviral protein NS1 through PKC(η)-driven modifications. Here we reveal a new PV-induced intracellular loop-back mechanism whereby PKCη/Rdx phosphorylates mouse PDK1:S138 and activates it independently of PI3-kinase signaling. The corresponding human PDK1phosphoS135 appears as a hallmark of highly aggressive brain tumors and may contribute to the very effective targeting of human gliomas by H-1PV. Strikingly, although H-1PV does not trigger PDK1 activation in normal human cells, such cells show enhanced viral DNA amplification and NS1-induced death upon expression of a constitutively active PDK1 mimicking PDK1phosphoS135. This modification thus appears as a marker of human glioma malignant progression and sensitivity to H-1PV-induced tumor cell killing. The H-1 protoparvovirus (H-1PV) is the first replication-competent member of the Parvoviridae family to undergo a phase I/IIa clinical trial in patients suffering from glioblastoma multiforme. Although the intrinsic oncotropism and oncolytic activity of protoparvoviruses are well known, the underlying molecular mechanisms remain elusive. Here we identify a PV-induced intracellular loop-back mechanism that promotes PV replication and cytotoxicity through PI3-kinase-independent stimulation of PDK1 and of the PKC and PKB/Akt1 downstream kinases. This mechanism involves PKCη/Rdx-mediated phosphorylation of PDK1 (at S138 in mouse or S135 in human). Interestingly, this phosphorylation appears as a hallmark of highly aggressive brain tumors. Although H-1PV does not promote it in normal human cells, experimentally administered activated PDK1 variants were able to sensitize these cells to virus infection. These data lead us to propose PDK1phosphoS135 as a new candidate marker for monitoring tumor progression and responsiveness to oncolytic parvovirotherapy, particularly in the case of highly aggressive brain tumors. Furthermore, the sensitivity of PDK1phosphoS135-positive cell lines to inhibitors of PKCη/Rdx argues for considering this complex as a potential target for anticancer drug development.
Collapse
Affiliation(s)
- Séverine Bär
- Infection and Cancer Program, Tumor Virology Division (F010), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jean Rommelaere
- Infection and Cancer Program, Tumor Virology Division (F010), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jürg P. F. Nüesch
- Infection and Cancer Program, Tumor Virology Division (F010), German Cancer Research Center (DKFZ), Heidelberg, Germany
- * E-mail:
| |
Collapse
|
10
|
Gupta SK, Gandham RK, Sahoo AP, Tiwari AK. Viral genes as oncolytic agents for cancer therapy. Cell Mol Life Sci 2015; 72:1073-94. [PMID: 25408521 PMCID: PMC11113997 DOI: 10.1007/s00018-014-1782-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2014] [Revised: 10/29/2014] [Accepted: 11/13/2014] [Indexed: 12/20/2022]
Abstract
Many viruses have the ability to modulate the apoptosis, and to accomplish it; viruses encode proteins which specifically interact with the cellular signaling pathways. While some viruses encode proteins, which inhibit the apoptosis or death of the infected cells, there are viruses whose encoded proteins can kill the infected cells by multiple mechanisms, including apoptosis. A particular class of these viruses has specific gene(s) in their genomes which, upon ectopic expression, can kill the tumor cells selectively without affecting the normal cells. These genes and their encoded products have demonstrated great potential to be developed as novel anticancer therapeutic agents which can specifically target and kill the cancer cells leaving the normal cells unharmed. In this review, we will discuss about the viral genes having specific cancer cell killing properties, what is known about their functioning, signaling pathways and their therapeutic applications as anticancer agents.
Collapse
Affiliation(s)
- Shishir Kumar Gupta
- Molecular Biology Lab, Division of Veterinary Biotechnology, Indian Veterinary Research Institute, Izatnagar, Bareilly, 243122 UP India
| | - Ravi Kumar Gandham
- Molecular Biology Lab, Division of Veterinary Biotechnology, Indian Veterinary Research Institute, Izatnagar, Bareilly, 243122 UP India
| | - A. P. Sahoo
- Molecular Biology Lab, Division of Veterinary Biotechnology, Indian Veterinary Research Institute, Izatnagar, Bareilly, 243122 UP India
| | - A. K. Tiwari
- Molecular Biology Lab, Division of Veterinary Biotechnology, Indian Veterinary Research Institute, Izatnagar, Bareilly, 243122 UP India
| |
Collapse
|
11
|
Marchini A, Bonifati S, Scott EM, Angelova AL, Rommelaere J. Oncolytic parvoviruses: from basic virology to clinical applications. Virol J 2015; 12:6. [PMID: 25630937 PMCID: PMC4323056 DOI: 10.1186/s12985-014-0223-y] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Accepted: 12/03/2014] [Indexed: 12/28/2022] Open
Abstract
Accumulated evidence gathered over recent decades demonstrated that some members of the Parvoviridae family, in particular the rodent protoparvoviruses H-1PV, the minute virus of mice and LuIII have natural anticancer activity while being nonpathogenic to humans. These studies have laid the foundations for the launch of a first phase I/IIa clinical trial, in which the rat H-1 parvovirus is presently undergoing evaluation for its safety and first signs of efficacy in patients with glioblastoma multiforme. After a brief overview of the biology of parvoviruses, this review focuses on the studies which unraveled the antineoplastic properties of these agents and supported their clinical use as anticancer therapeutics. Furthermore, the development of novel parvovirus-based anticancer strategies with enhanced specificity and efficacy is discussed, in particular the development of second and third generation vectors and the combination of parvoviruses with other anticancer agents. Lastly, we address the key challenges that remain towards a more rational and efficient use of oncolytic parvoviruses in clinical settings, and discuss how a better understanding of the virus life-cycle and of the cellular factors involved in virus infection, replication and cytotoxicity may promote the further development of parvovirus-based anticancer therapies, open new prospects for treatment and hopefully improve clinical outcome.
Collapse
Affiliation(s)
- Antonio Marchini
- Infection and Cancer Program, Tumor Virology Division (F010), German Cancer Research Center (DKFZ), Im Neuenheimer Feld 242, 69120, Heidelberg, Germany.
| | - Serena Bonifati
- Infection and Cancer Program, Tumor Virology Division (F010), German Cancer Research Center (DKFZ), Im Neuenheimer Feld 242, 69120, Heidelberg, Germany.
| | - Eleanor M Scott
- Infection and Cancer Program, Tumor Virology Division (F010), German Cancer Research Center (DKFZ), Im Neuenheimer Feld 242, 69120, Heidelberg, Germany.
| | - Assia L Angelova
- Infection and Cancer Program, Tumor Virology Division (F010), German Cancer Research Center (DKFZ), Im Neuenheimer Feld 242, 69120, Heidelberg, Germany.
| | - Jean Rommelaere
- Infection and Cancer Program, Tumor Virology Division (F010), German Cancer Research Center (DKFZ), Im Neuenheimer Feld 242, 69120, Heidelberg, Germany.
| |
Collapse
|
12
|
Pal D, Basu A. The unique protein kinase Cη: implications for breast cancer (review). Int J Oncol 2014; 45:493-8. [PMID: 24841225 DOI: 10.3892/ijo.2014.2443] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Accepted: 02/18/2014] [Indexed: 11/05/2022] Open
Abstract
Deregulation of key signal transduction pathways that govern important cellular processes leads to cancer. The development of effective therapeutics for cancer warrants a comprehensive understanding of the signaling pathways that are deregulated in cancer. The protein kinase C (PKC) family has served as an attractive target for cancer therapy for decades owing to its crucial roles in several cellular processes. PKCη is a novel member of the PKC family that plays critical roles in various cellular processes such as growth, proliferation, differentiation and cell death. The regulation of PKCη appears to be unique compared to other PKC isozymes, and there are conflicting reports regarding its role in cancer. This review focuses on the unique aspects of PKCη in terms of its structure, regulation and subcellular distribution and speculates on how these features could account for its distinct functions. We have also discussed the functional implications of PKCη in cancer with particular emphasis on breast cancer.
Collapse
Affiliation(s)
- Deepanwita Pal
- Department of Molecular and Medical Genetics, University of North Texas Health Science Center and Institute for Cancer Research, Fort Worth, TX 76107, USA
| | - Alakananda Basu
- Department of Molecular and Medical Genetics, University of North Texas Health Science Center and Institute for Cancer Research, Fort Worth, TX 76107, USA
| |
Collapse
|
13
|
Nüesch JPF, Rommelaere J. Tumor suppressing properties of rodent parvovirus NS1 proteins and their derivatives. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 818:99-124. [PMID: 25001533 DOI: 10.1007/978-1-4471-6458-6_5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cancer chemotherapy with monospecific agents is often hampered by the rapid development of tumor resistance to the drug used. Therefore, combination treatments aiming at several different targets are sought. Viral regulatory proteins, modified or not, appear ideal for this purpose because of their multimodal killing action against neoplastically transformed cells. The large nonstructural protein NS1 of rodent parvoviruses is an excellent candidate for an anticancer agent, shown to interfere specifically with cancer cell growth and survival. The present review describes the structure, functions, and regulation of the multifunctional protein NS1, its specific interference with cell processes and cell protein activities, and what is known so far about the mechanisms underlying NS1 interference with cancer growth. It further outlines prospects for the development of new, multimodal cancer toxins and their potential applications.
Collapse
Affiliation(s)
- Jürg P F Nüesch
- Program "Infection and Cancer", Division Tumor Virology (F010), Deutsches Krebsforschungszentrum/German Cancer Research Center (DKFZ), Im Neuenheimer Feld 242, D-69120, Heidelberg, Germany,
| | | |
Collapse
|
14
|
Vesicular transport of progeny parvovirus particles through ER and Golgi regulates maturation and cytolysis. PLoS Pathog 2013; 9:e1003605. [PMID: 24068925 PMCID: PMC3777860 DOI: 10.1371/journal.ppat.1003605] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Accepted: 07/24/2013] [Indexed: 01/30/2023] Open
Abstract
Progeny particles of non-enveloped lytic parvoviruses were previously shown to be actively transported to the cell periphery through vesicles in a gelsolin-dependent manner. This process involves rearrangement and destruction of actin filaments, while microtubules become protected throughout the infection. Here the focus is on the intracellular egress pathway, as well as its impact on the properties and release of progeny virions. By colocalization with cellular marker proteins and specific modulation of the pathways through over-expression of variant effector genes transduced by recombinant adeno-associated virus vectors, we show that progeny PV particles become engulfed into COPII-vesicles in the endoplasmic reticulum (ER) and are transported through the Golgi to the plasma membrane. Besides known factors like sar1, sec24, rab1, the ERM family proteins, radixin and moesin play (an) essential role(s) in the formation/loading and targeting of virus-containing COPII-vesicles. These proteins also contribute to the transport through ER and Golgi of the well described analogue of cellular proteins, the secreted Gaussia luciferase in absence of virus infection. It is therefore likely that radixin and moesin also serve for a more general function in cellular exocytosis. Finally, parvovirus egress via ER and Golgi appears to be necessary for virions to gain full infectivity through post-assembly modifications (e.g. phosphorylation). While not being absolutely required for cytolysis and progeny virus release, vesicular transport of parvoviruses through ER and Golgi significantly accelerates these processes pointing to a regulatory role of this transport pathway. Previously, it was thought that non-enveloped lytic parvoviruses were released through a lytic burst of cells at the end of infection. However, recent work demonstrated that these small non-enveloped single-stranded DNA viruses are actively transported through vesicles from the nucleus, the site of replication and assembly, to the cell periphery. The current investigation demonstrates that progeny particles become engulfed into COPII-vesicles in the endoplasmic reticulum (ER) and are transported through the Golgi to the plasma membrane (PM). ERM family proteins radixin and moesin appear to play an essential role in this cellular secretion pathway. While passing through ER and Golgi cisternae, PVs maturate through post-assembly modifications, which significantly increase the infectivity of progeny virions. Finally, the vesicular transport of parvoviral particles was shown to regulate virus-induced cytolysis, thereby accelerating the further release and spread of progeny virions. As rodent PVs are currently viewed as oncolytic agents for cancer virotherapy, it is important to further investigate the mechanism of PV egress — not only to improve the spreading of these agents through the tumor mass, but also to optimize the induction of an anti-tumor immune response upon virus — induced cytolysis.
Collapse
|
15
|
Pal D, Outram SP, Basu A. Novel regulation of protein kinase C-η. Biochem Biophys Res Commun 2012; 425:836-41. [PMID: 22892130 DOI: 10.1016/j.bbrc.2012.07.163] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2012] [Accepted: 07/30/2012] [Indexed: 10/28/2022]
Abstract
Protein kinase C (PKC) is the receptor for tumor promoting phorbol esters, which are potent activators of conventional and novel PKCs, but persistent treatment with phorbol esters leads to downregulation of these PKCs. However, PKCη, a novel PKC isozyme, resists downregulation by tumor-promoting phorbol esters, but little is known about how PKCη level is regulated. Phosphorylation and dephosphorylation play an important role in regulating activity and stability of PKCs. In the present study, we have investigated the molecular mechanism of PKCη regulation. Several PKC activators, including phorbol 12,13-dibutyrate, 12-O-tetradecanoylphorbol-13-acetate and indolactam V caused upregulation of PKCη, whereas the general PKC inhibitor Gö 6983, but not the conventional PKC inhibitor Gö 6976 led to the downregulation of PKCη. Upregulation of PKCη was associated with an increase in phosphorylation of PKCη. Silencing of phosphoinositide-dependent kinase-1, which phosphorylates PKCη at the activation loop, failed to prevent PKC activator-induced upregulation of PKCη. Knockdown of PKCε but not PKCα inhibited PKC activator-induced upregulation of PKCη. Thus, our results suggest that the regulation of PKCη is unique and PKCε is required for the PKC activator-induced upregulation of PKCη.
Collapse
Affiliation(s)
- Deepanwita Pal
- Department of Molecular Biology & Immunology, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | | | | |
Collapse
|
16
|
Nüesch JPF, Lacroix J, Marchini A, Rommelaere J. Molecular pathways: rodent parvoviruses--mechanisms of oncolysis and prospects for clinical cancer treatment. Clin Cancer Res 2012; 18:3516-23. [PMID: 22566376 DOI: 10.1158/1078-0432.ccr-11-2325] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Rodent parvoviruses (PV) are recognized for their intrinsic oncotropism and oncolytic activity, which contribute to their natural oncosuppressive effects. Although PV uptake occurs in most host cells, some of the subsequent steps leading to expression and amplification of the viral genome and production of progeny particles are upregulated in malignantly transformed cells. By usurping cellular processes such as DNA replication, DNA damage response, and gene expression, and/or by interfering with cellular signaling cascades involved in cytoskeleton dynamics, vesicular integrity, cell survival, and death, PVs can induce cytostasis and cytotoxicity. Although productive PV infections normally culminate in cytolysis, virus spread to neighboring cells and secondary rounds of infection, even abortive infection or the sole expression of the PV nonstructural protein NS1, is sufficient to cause significant tumor cell death, either directly or indirectly (through activation of host immune responses). This review highlights the molecular pathways involved in tumor cell targeting by PVs and in PV-induced cell death. It concludes with a discussion of the relevance of these pathways to the application of PVs in cancer therapy, linking basic knowledge of PV-host cell interactions to preclinical assessment of PV oncosuppression.
Collapse
Affiliation(s)
- Jürg P F Nüesch
- Infection and Cancer Program, Division F010, German Cancer Research Center, Heidelberg, Germany
| | | | | | | |
Collapse
|
17
|
Argiris K, Panethymitaki C, Tavassoli M. Naturally occurring, tumor-specific, therapeutic proteins. Exp Biol Med (Maywood) 2011; 236:524-36. [PMID: 21521711 DOI: 10.1258/ebm.2011.011004] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The emerging approach to cancer treatment known as targeted therapies offers hope in improving the treatment of therapy-resistant cancers. Recent understanding of the molecular pathogenesis of cancer has led to the development of targeted novel drugs such as monoclonal antibodies, small molecule inhibitors, mimetics, antisense and small interference RNA-based strategies, among others. These compounds act on specific targets that are believed to contribute to the development and progression of cancers and resistance of tumors to conventional therapies. Delivered individually or combined with chemo- and/or radiotherapy, such novel drugs have produced significant responses in certain types of cancer. Among the most successful novel compounds are those which target tyrosine kinases (imatinib, trastuzumab, sinutinib, cetuximab). However, these compounds can cause severe side-effects as they inhibit pathways such as epidermal growth factor receptor (EGFR) or platelet-derived growth factor receptor, which are also important for normal functions in non-transformed cells. Recently, a number of proteins have been identified which show a remarkable tumor-specific cytotoxic activity. This toxicity is independent of tumor type or specific genetic changes such as p53, pRB or EGFR aberrations. These tumor-specific killer proteins are either derived from common human and animal viruses such as E1A, E4ORF4 and VP3 (apoptin) or of cellular origin, such as TRAIL (tumor necrosis factor-related apoptosis-inducing ligand) and MDA-7 (melanoma differentiation associated-7). This review aims to present a current overview of a selection of these proteins with preferential toxicity among cancer cells and will provide an insight into the possible mechanism of action, tumor specificity and their potential as novel tumor-specific cancer therapeutics.
Collapse
|
18
|
Wizla P, Begue A, Loison I, Richard A, Caillet-Fauquet P, Stéhelin D. Ectopic expression of H-1 parvovirus NS1 protein induces alterations in actin filaments and cell death in human normal MRC-5 and transformed MRC-5 SV2 cells. Arch Virol 2010; 155:771-5. [PMID: 20237811 DOI: 10.1007/s00705-010-0637-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2009] [Accepted: 02/09/2010] [Indexed: 11/28/2022]
Abstract
When grown in human cell lines, oncolytic H-1 parvovirus (H-1PV) replication preferentially occurs in transformed cells, which ultimately die upon infection. H-1PV-induced cytotoxicity is mainly due to P4 promoter-driven NS1 protein expression. Infection of untransformed cells generally does not induce deleterious effects because the P4 promoter is not activated. Here, we show that ectopic CMV-driven NS1 protein expression in normal human MRC-5 cells results in alterations of actin filaments and cell death, and both effects are prevented by a serine 473 mutation. The same substitution preserves actin filaments of transfected MRC-5 SV2 cells, that are MRC-5 transformed counterparts, but does not impair NS1-induced cytotoxicity.
Collapse
Affiliation(s)
- Pierre Wizla
- UMR 8161, CNRS, Institut de Biologie de Lille, 1 rue du Professeur Calmette, BP 447, 59021, Lille Cedex, France.
| | | | | | | | | | | |
Collapse
|
19
|
Noteborn MHM. Proteins selectively killing tumor cells. Eur J Pharmacol 2009; 625:165-73. [PMID: 19836376 DOI: 10.1016/j.ejphar.2009.06.068] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2009] [Revised: 06/24/2009] [Accepted: 06/25/2009] [Indexed: 01/04/2023]
Abstract
All human cells have a genetic program that upon activation will cause cell death, named apoptosis. Cancer cells can grow due to unbalances in proliferation, cell cycle regulation and their apoptosis machinery: genomic mutations resulting in non-functional pro-apoptosis proteins or over-expression of anti-apoptosis proteins form the basis of tumor formation. Surprisingly, lessons learned from viruses show that cancer cannot be regarded simply as the opposite of apoptosis. For instance, adenovirus can only transform cells when both its anti- and pro-apoptotic proteins are produced. Oncolytic viruses are known to replicate selectively in tumor cells resulting in cell death. Proteins derived from viruses, i.e. chicken anemia virus (CAV)-derived apoptosis-inducing protein (apoptin), adenovirus early region 4 open reading frame (E4orf4) and parvovirus-H1 derived non-structural protein 1 (NS1), the human alpha-lactalbumin made lethal to tumor cells (HAMLET), which is present in human milk or the human cytokines melanoma differentiation-associated gene-7 (mda-7) and tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) have all the ability to induce tumor-selective apoptosis. The tumor-selective apoptosis-inducing proteins seem to interact with transforming survival processes, which can become redirected by these proteins into cell death. Transformation-related processes have been identified, which seem to be crucial for the tumor-selectively killing activity of these proteins. For instance, the transformation-related protein phosphatase 2A (PP2A) plays a role in the induction of tumor-selective apoptosis. The proteins mda-7, TRAIL and HAMLET are already successfully tested in first clinical trials. Proteins harboring tumor-selective apoptosis characteristics represent, therefore, a therapeutic potential and a tool for unraveling tumor-related processes. Fundamental molecular and (pre)clinical therapeutic studies of the various tumor-selective apoptosis-inducing proteins apoptin, E4orf4, HAMLET, mda-7, NS1, TRAIL and related proteins will be discussed.
Collapse
Affiliation(s)
- Mathieu H M Noteborn
- Molecular Genetics, Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands.
| |
Collapse
|
20
|
Parvovirus H-1 induces cytopathic effects in breast carcinoma-derived cultures. Breast Cancer Res Treat 2009; 121:23-33. [PMID: 19565332 DOI: 10.1007/s10549-009-0451-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2009] [Accepted: 06/13/2009] [Indexed: 10/20/2022]
Abstract
Parvovirus H-1 (H-1 PV) preferentially replicates in malignant cells resulting in their death by cytolysis. It has often been considered a potential candidate for use in novel anticancer therapy. To evaluate its potential in a model of natural tumors, we assayed in vitro the effect exerted by H-1 PV on short-term cultures derived from breast tumor samples freshly excised from patients. Our results show that H-1 PV effectively kills tumor-derived cells, whereas normal tissue-derived cells showed no H-1 PV-induced cytopathic effects (CPE). We also determined that the H-1 PV sensitivity (up to 67% sensitive cultures) is related with the quantities of virus assayed. We further examined the expression and phosphorylation state of the parvoviral nonstructural protein 1 (NS1), known to be associated with parvoviruses-induced CPE. Both appear to be impaired in normal tissue-derived cells and resistant cultures. Finally, we show that H-1 PV sensitivity in cultures correlates significantly with higher tumor grades (Nottingham combined histologic grade 2 or 3). This report confirms that H-1 PV can efficiently induce CPE in primary breast tumor cells in vitro. It identifies tumor characteristics representing potential criteria for recruiting patients for clinical evaluation of H-1 PV antitumor effects.
Collapse
|
21
|
Ezrin-radixin-moesin family proteins are involved in parvovirus replication and spreading. J Virol 2009; 83:5854-63. [PMID: 19321616 DOI: 10.1128/jvi.00039-09] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The propagation of autonomous parvoviruses is strongly dependent on the phosphorylation of the major nonstructural protein NS1 by members of the protein kinase C (PKC) family. Minute virus of mice (MVM) replication is accompanied by changes in the overall phosphorylation pattern of NS1, which is newly modified at consensus PKC sites. These changes result, at least in part, from the ability of MVM to modulate the PDK-1/PKC pathway, leading to activation and redistribution of both PDK-1 and PKCeta. We show that proteins of the ezrin-radixin-moesin (ERM) family are essential for virus propagation and spreading through their functions as adaptors for PKCeta. MVM infection led to redistribution of radixin and moesin in the cell, resulting in increased colocalization of these proteins with PKCeta. Radixin was found to control the PKCeta-driven phosphorylation of NS1 and newly synthesized capsids in vivo. Conversely, radixin phosphorylation and activation were driven by the NS1/CKIIalpha complex. Altogether, these data argue for ERM proteins being both targets and modulators of parvovirus infection.
Collapse
|
22
|
Bär S, Daeffler L, Rommelaere J, Nüesch JPF. Vesicular egress of non-enveloped lytic parvoviruses depends on gelsolin functioning. PLoS Pathog 2008; 4:e1000126. [PMID: 18704167 PMCID: PMC2494870 DOI: 10.1371/journal.ppat.1000126] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2008] [Accepted: 07/16/2008] [Indexed: 12/02/2022] Open
Abstract
The autonomous parvovirus Minute Virus of Mice (MVM) induces specific changes in the cytoskeleton filaments of infected permissive cells, causing in particular the degradation of actin fibers and the generation of “actin patches.” This is attributed to a virus-induced imbalance between the polymerization factor N-WASP (Wiscott-Aldrich syndrome protein) and gelsolin, a multifunctional protein cleaving actin filaments. Here, the focus is on the involvement of gelsolin in parvovirus propagation and virus-induced actin processing. Gelsolin activity was knocked-down, and consequences thereof were determined for virus replication and egress and for actin network integrity. Though not required for virus replication or progeny particle assembly, gelsolin was found to control MVM (and related H1-PV) transport from the nucleus to the cell periphery and release into the culture medium. Gelsolin-dependent actin degradation and progeny virus release were both controlled by (NS1)/CKIIα, a recently identified complex between a cellular protein kinase and a MVM non-structural protein. Furthermore, the export of newly synthesized virions through the cytoplasm appeared to be mediated by (virus-modified) lysomal/late endosomal vesicles. By showing that MVM release, like entry, is guided by the cytoskeleton and mediated by vesicles, these results challenge the current view that egress of non-enveloped lytic viruses is a passive process. Rodent parvoviruses are non-enveloped lytic viruses that are thought excellent tools for a virotherapy of cancer because of their strong natural oncolytic potential and low pathogenicity in humans. Egress of non-enveloped lytic viruses is commonly thought to occur as a virus burst after cell disintegration. Indeed, we showed in the past that autonomous parvoviruses induce severe cytopathic effects to the host cell, manifested in restructuring and degradation of cytoskeletal filaments, thereby supporting such mode of virus spread. Here, we focus on the impact of virus-induced actin degradation, and particularly the functioning of the actin-severing protein gelsolin. Although not required for DNA replication or progeny particle production, gelsolin appears to facilitate a regulated virus egress from the nucleus to the cell periphery via (virus modified) lysosomal/late endosomal vesicles. These results challenge the current view that lytic virus egress is just a passive process at the end of infection and suggests that these pathogens are endowed with the ability to efficiently spread from cell to cell potentially in solid (tumor) tissue.
Collapse
Affiliation(s)
- Séverine Bär
- Program “Infection and Cancer,” Abteilung F010 and Institut National de la Santé et de la Recherche Médicale U701, Deutsches Krebsforschungszentrum, Heidelberg, Germany
| | - Laurent Daeffler
- Program “Infection and Cancer,” Abteilung F010 and Institut National de la Santé et de la Recherche Médicale U701, Deutsches Krebsforschungszentrum, Heidelberg, Germany
| | - Jean Rommelaere
- Program “Infection and Cancer,” Abteilung F010 and Institut National de la Santé et de la Recherche Médicale U701, Deutsches Krebsforschungszentrum, Heidelberg, Germany
| | - Jürg P. F. Nüesch
- Program “Infection and Cancer,” Abteilung F010 and Institut National de la Santé et de la Recherche Médicale U701, Deutsches Krebsforschungszentrum, Heidelberg, Germany
- * E-mail:
| |
Collapse
|