1
|
Sun M, Manson ML, Guo T, de Lange ECM. CNS Viral Infections-What to Consider for Improving Drug Treatment: A Plea for Using Mathematical Modeling Approaches. CNS Drugs 2024; 38:349-373. [PMID: 38580795 PMCID: PMC11026214 DOI: 10.1007/s40263-024-01082-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/10/2024] [Indexed: 04/07/2024]
Abstract
Neurotropic viruses may cause meningitis, myelitis, encephalitis, or meningoencephalitis. These inflammatory conditions of the central nervous system (CNS) may have serious and devastating consequences if not treated adequately. In this review, we first summarize how neurotropic viruses can enter the CNS by (1) crossing the blood-brain barrier or blood-cerebrospinal fluid barrier; (2) invading the nose via the olfactory route; or (3) invading the peripheral nervous system. Neurotropic viruses may then enter the intracellular space of brain cells via endocytosis and/or membrane fusion. Antiviral drugs are currently used for different viral CNS infections, even though their use and dosing regimens within the CNS, with the exception of acyclovir, are minimally supported by clinical evidence. We therefore provide considerations to optimize drug treatment(s) for these neurotropic viruses. Antiviral drugs should cross the blood-brain barrier/blood cerebrospinal fluid barrier and pass the brain cellular membrane to inhibit these viruses inside the brain cells. Some antiviral drugs may also require intracellular conversion into their active metabolite(s). This illustrates the need to better understand these mechanisms because these processes dictate drug exposure within the CNS that ultimately determine the success of antiviral drugs for CNS infections. Finally, we discuss mathematical model-based approaches for optimizing antiviral treatments. Thereby emphasizing the potential of CNS physiologically based pharmacokinetic models because direct measurement of brain intracellular exposure in living humans faces ethical restrictions. Existing physiologically based pharmacokinetic models combined with in vitro pharmacokinetic/pharmacodynamic information can be used to predict drug exposure and evaluate efficacy of antiviral drugs within the CNS, to ultimately optimize the treatments of CNS viral infections.
Collapse
Affiliation(s)
- Ming Sun
- Division of Systems Pharmacology and Pharmacy, Leiden Academic Center for Drug Research, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Martijn L Manson
- Division of Systems Pharmacology and Pharmacy, Leiden Academic Center for Drug Research, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Tingjie Guo
- Division of Systems Pharmacology and Pharmacy, Leiden Academic Center for Drug Research, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Elizabeth C M de Lange
- Division of Systems Pharmacology and Pharmacy, Leiden Academic Center for Drug Research, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands.
| |
Collapse
|
2
|
Zhu X, Fan C, Xiong Z, Chen M, Li Z, Tao T, Liu X. Development and application of oncolytic viruses as the nemesis of tumor cells. Front Microbiol 2023; 14:1188526. [PMID: 37440883 PMCID: PMC10335770 DOI: 10.3389/fmicb.2023.1188526] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 05/18/2023] [Indexed: 07/15/2023] Open
Abstract
Viruses and tumors are two pathologies that negatively impact human health, but what occurs when a virus encounters a tumor? A global consensus among cancer patients suggests that surgical resection, chemotherapy, radiotherapy, and other methods are the primary means to combat cancer. However, with the innovation and development of biomedical technology, tumor biotherapy (immunotherapy, molecular targeted therapy, gene therapy, oncolytic virus therapy, etc.) has emerged as an alternative treatment for malignant tumors. Oncolytic viruses possess numerous anti-tumor properties, such as directly lysing tumor cells, activating anti-tumor immune responses, and improving the tumor microenvironment. Compared to traditional immunotherapy, oncolytic virus therapy offers advantages including high killing efficiency, precise targeting, and minimal side effects. Although oncolytic virus (OV) therapy was introduced as a novel approach to tumor treatment in the 19th century, its efficacy was suboptimal, limiting its widespread application. However, since the U.S. Food and Drug Administration (FDA) approved the first OV therapy drug, T-VEC, in 2015, interest in OV has grown significantly. In recent years, oncolytic virus therapy has shown increasingly promising application prospects and has become a major research focus in the field of cancer treatment. This article reviews the development, classification, and research progress of oncolytic viruses, as well as their mechanisms of action, therapeutic methods, and routes of administration.
Collapse
Affiliation(s)
- Xiao Zhu
- Zhejiang Provincial People's Hospital Affiliated to Hangzhou Medical College, Hangzhou Medical College, Hangzhou, China
- The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China
- Department of Biological and Chemical Sciences, New York Institute of Technology—Manhattan Campus, New York, NY, United States
| | - Chenyang Fan
- Department of Clinical Medicine, Medicine and Technology, School of Zunyi Medical University, Zunyi, China
| | - Zhuolong Xiong
- The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China
| | - Mingwei Chen
- The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China
| | - Zesong Li
- Guangdong Provincial Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Key Laboratory of Genitourinary Tumor, Department of Urology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital(Shenzhen Institute of Translational Medicine), Shenzhen, China
| | - Tao Tao
- Department of Gastroenterology, Zibo Central Hospital, Zibo, China
| | - Xiuqing Liu
- Department of Clinical Laboratory, Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, China
| |
Collapse
|
3
|
Yusni R, Mariya S, Saepuloh U, Mariya SS, Darusman HS. Kidney cell culture Macaca fascicularis as a candidate for vaccine development and in vitro model. J Med Primatol 2023. [PMID: 37296521 DOI: 10.1111/jmp.12655] [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: 08/31/2022] [Revised: 05/08/2023] [Accepted: 05/25/2023] [Indexed: 06/12/2023]
Abstract
BACKGROUND Cell culture is the proliferation of a cell population in vitro by isolating from the original tissue or growing from existing ones. One essential source is the monkey kidney cell cultures which have an essential role in biomedical study. This is due to the significant homology between the human and macaque genomes making these useful for cultivating human viruses, especially enteroviruses, and growing vaccines. METHODS This study developed cell cultures derived from the kidney of Macaca fascicularis (Mf) and validated its gene expression. RESULTS The primary cultures were successfully subcultured up to six passages, grew as monolayers, and exhibited epithelial-like morphology. The cultured cells remained heterogeneous in phenotype and they expressed CD155 and CD46 as viral receptors, cell morphology (CD24, endosialin, and vWF), proliferation, also apoptosis markers (Ki67 and p53). CONCLUSIONS These results indicated that the cell cultures can be used as in vitro model cells for vaccine development and bioactive compound.
Collapse
Affiliation(s)
- Rahmat Yusni
- Biotechnology Graduate School of Bogor Agricultural University, Bogor, Indonesia
| | - Silmi Mariya
- Primate Research Center Bogor Agricultural University, Bogor, Indonesia
| | - Uus Saepuloh
- Primate Research Center Bogor Agricultural University, Bogor, Indonesia
| | - Sela S Mariya
- Primate Research Center Bogor Agricultural University, Bogor, Indonesia
- Center for Biomedical Research, National Research and Innovation Agency of Indonesia, Cibinong Sciences center, Bogor, Indonesia
| | - Huda S Darusman
- Biotechnology Graduate School of Bogor Agricultural University, Bogor, Indonesia
- Primate Research Center Bogor Agricultural University, Bogor, Indonesia
- Primatology Graduate School of Bogor Agricultural University, Bogor, Indonesia
- Faculty of Veterinary Medicine Bogor Agricultural University, Bogor, Indonesia
| |
Collapse
|
4
|
CD155 is a putative therapeutic target in medulloblastoma. Clin Transl Oncol 2023; 25:696-705. [PMID: 36301489 DOI: 10.1007/s12094-022-02975-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 10/05/2022] [Indexed: 10/31/2022]
Abstract
BACKGROUND Medulloblastoma is the most common pediatric malignant brain tumor, consisting of four molecular subgroups (WNT, SHH, Group 3, Group 4) and 12 subtypes. Expression of the cell surface poliovirus receptor (PVR), CD155, is necessary for entry of the viral immunotherapeutic agent, PVSRIPO, a polio:rhinovirus chimera. CD155, physiologically expressed in the mononuclear phagocytic system, is widely expressed ectopically in solid tumors. The objective of this study is to elucidate CD155 expression as both a receptor for PVSRIPO and a therapeutic target in medulloblastoma. METHODS PVR mRNA expression was determined in several patient cohorts and human medulloblastoma cell lines. Patient samples were also analyzed for CD155 expression using immunohistochemistry and cell lines were analyzed using Western Blots. CD155 was blocked using a monoclonal antibody and cell viability, invasion, and migration were assessed. RESULTS AND DISCUSSION PVR mRNA expression was highest in the WNT subgroup and lowest in Group 4. PVR expression in the subgroups of medulloblastoma were similar to other pediatric brain and non-brain tumors. PVR expression was largely not associated with subgroup or subtype. Neither PVR protein expression intensity nor frequency were associated with overall survival. PVR expression was elevated in Group 3 patients with metastases but there was no difference in paired primary and metastatic medulloblastoma. Blocking PVR resulted in dose-dependent cell death, decreased invasion in vitro, and modestly inhibited cell migration. CONCLUSIONS CD155 is expressed across medulloblastoma subgroups and subtypes. Blocking CD155 results in cell death and decreased cellular invasion. This study provides rationale for CD155-targeting agents including PVSRIPO and antibody-mediated blockade of CD155.
Collapse
|
5
|
Muacevic A, Adler JR, Bisen YT, Iratwar S, Kesharwani A, Vardhan S, Singh A. Emerging Recombinant Oncolytic Poliovirus Therapies Against Malignant Glioma: A Review. Cureus 2023; 15:e34028. [PMID: 36814733 PMCID: PMC9939956 DOI: 10.7759/cureus.34028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Accepted: 01/21/2023] [Indexed: 01/22/2023] Open
Abstract
Glioblastoma multiforme (GBM) is a fourth-grade malignant glioma that continues to be the main contributor to primary malignant brain tumour-related death in humans. The most prevalent primary brain tumours are gliomas. The most dangerous of these neoplasms, GBM, has been shown to be one of the most lethal and refractory tumours. For those who have been diagnosed with GBM, the median time to progression, as determined by magnetic resonance imaging, is roughly six months, and the median survival is approximately one year. GBM is challenging to manage with old treatments like chemotherapy, tumour debulking, and radiation therapy. Treatment outcomes are poor, and due to this effect, the treatment is not up to the mark. GBM also shows diagnostic complexity due to limitations in the use of specific targeted therapies. The treatment protocol followed currently has an entire focus on safe resection and radiotherapy. Protein synthesis is not tightly regulated physiologically in malignant cells, which promotes unchecked growth and proliferation. An innovative, experimental technique for treating cancer uses polioviruses that have been genetically altered to target a fascinating aberration of translation regulation in cancer. This approach enables precise and effective cancer cell targeting based on the convergence of numerous variables. Oncolytic viruses have revolutionised cancer treatment. However, their effectiveness in glioblastoma remains restricted, necessitating more improvement. Oncolytic poliovirus has shown great potential in the treatment of GBM. Factors like the blood-brain barrier, immunosuppressive tumour microenvironment (TME), and tumour heterogeneity make treatment for malignant gliomas ineffective. In this review, we have focused on oncolytic viruses, specifically oncolytic poliovirus, and we explore malignant glioma treatments. We have also discussed currently available conventional treatment options for malignant glioma and other brain tumours.
Collapse
|
6
|
Annese T, Tamma R, Ribatti D. Update in TIGIT Immune-Checkpoint Role in Cancer. Front Oncol 2022; 12:871085. [PMID: 35656508 PMCID: PMC9152184 DOI: 10.3389/fonc.2022.871085] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 04/14/2022] [Indexed: 12/12/2022] Open
Abstract
The in-depth characterization of cross-talk between tumor cells and T cells in solid and hematological malignancies will have to be considered to develop new therapeutical strategies concerning the reactivation and maintenance of patient-specific antitumor responses within the patient tumor microenvironment. Activation of immune cells depends on a delicate balance between activating and inhibitory signals mediated by different receptors. T cell immunoreceptor with immunoglobulin and ITIM domain (TIGIT) is an inhibitory receptor expressed by regulatory T cells (Tregs), activated T cells, and natural killer (NK) cells. TIGIT pathway regulates T cell-mediated tumor recognition in vivo and in vitro and represents an exciting target for checkpoint blockade immunotherapy. TIGIT blockade as monotherapy or in combination with other inhibitor receptors or drugs is emerging in clinical trials in patients with cancer. The purpose of this review is to update the role of TIGIT in cancer progression, looking at TIGIT pathways that are often upregulated in immune cells and at possible therapeutic strategies to avoid tumor aggressiveness, drug resistance, and treatment side effects. However, in the first part, we overviewed the role of immune checkpoints in immunoediting, the TIGIT structure and ligands, and summarized the key immune cells that express TIGIT.
Collapse
Affiliation(s)
- Tiziana Annese
- Department of Medicine and Surgery, Libera Università del Mediterraneo (LUM) Giuseppe Degennaro University, Bari, Italy.,Department of Basic Medical Sciences, Neurosciences and Sensory Organs, Section of Human Anatomy and Histology, University of Bari Medical School, Bari, Italy
| | - Roberto Tamma
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, Section of Human Anatomy and Histology, University of Bari Medical School, Bari, Italy
| | - Domenico Ribatti
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, Section of Human Anatomy and Histology, University of Bari Medical School, Bari, Italy
| |
Collapse
|
7
|
Löscher W, Howe CL. Molecular Mechanisms in the Genesis of Seizures and Epilepsy Associated With Viral Infection. Front Mol Neurosci 2022; 15:870868. [PMID: 35615063 PMCID: PMC9125338 DOI: 10.3389/fnmol.2022.870868] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 04/05/2022] [Indexed: 12/16/2022] Open
Abstract
Seizures are a common presenting symptom during viral infections of the central nervous system (CNS) and can occur during the initial phase of infection ("early" or acute symptomatic seizures), after recovery ("late" or spontaneous seizures, indicating the development of acquired epilepsy), or both. The development of acute and delayed seizures may have shared as well as unique pathogenic mechanisms and prognostic implications. Based on an extensive review of the literature, we present an overview of viruses that are associated with early and late seizures in humans. We then describe potential pathophysiologic mechanisms underlying ictogenesis and epileptogenesis, including routes of neuroinvasion, viral control and clearance, systemic inflammation, alterations of the blood-brain barrier, neuroinflammation, and inflammation-induced molecular reorganization of synapses and neural circuits. We provide clinical and animal model findings to highlight commonalities and differences in these processes across various neurotropic or neuropathogenic viruses, including herpesviruses, SARS-CoV-2, flaviviruses, and picornaviruses. In addition, we extensively review the literature regarding Theiler's murine encephalomyelitis virus (TMEV). This picornavirus, although not pathogenic for humans, is possibly the best-characterized model for understanding the molecular mechanisms that drive seizures, epilepsy, and hippocampal damage during viral infection. An enhanced understanding of these mechanisms derived from the TMEV model may lead to novel therapeutic interventions that interfere with ictogenesis and epileptogenesis, even within non-infectious contexts.
Collapse
Affiliation(s)
- Wolfgang Löscher
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine, Hannover, Germany
- Center for Systems Neuroscience, Hannover, Germany
| | - Charles L. Howe
- Division of Experimental Neurology, Department of Neurology, Mayo Clinic, Rochester, MN, United States
- Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
8
|
Nandi SS, Gohil T, Sawant SA, Lambe UP, Ghosh S, Jana S. CD155: A Key Receptor Playing Diversified Roles. Curr Mol Med 2021; 22:594-607. [PMID: 34514998 DOI: 10.2174/1566524021666210910112906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 06/16/2021] [Accepted: 06/20/2021] [Indexed: 11/22/2022]
Abstract
Cluster of differentiation (CD155), formerly identified as poliovirus receptor (PVR) and later as immunoglobulin molecule involved in cell adhesion, proliferation, invasion and migration. It is a surface protein expressed mostly on normal and transformed malignant cells. The expression of the receptor varies based on the origin of tissue. The expression of the protein is determined by factors involved in sonic hedgehog pathway, Ras-MEK-ERK pathway and during stress conditions like DNA damage response. The protein uses alternate splicing mechanism, producing four isoforms - two being soluble (CD155β and CD155γ) and two being transmembrane protein (CD155α and CD155δ). Apart from being a viral receptor, researchers have identified CD155 having important roles in cancer research and cell signaling field. The receptor is recognized as biomarker for identifying cancerous tissue. The receptor interacts with molecules involved in cells defense mechanism. The immune-surveillance role of CD155 is being deciphered to understand the mechanistic approach it utilizes as onco-immunologic molecule. CD155 is a non-MHC-I ligand which helps in identifying non-self to NK cells via an inhibitory TIGIT ligand. The TIGIT-CD155 pathway is a novel MHC-I-independent education mechanism for cell tolerance and activation of NK cell. The receptor also has a role in metastasis of cancer and trans endothelial mechanism. In this review, authors discuss the virus-host interaction that occurs via single transmembrane receptor, the poliovirus infection pathway, which is being exploited as therapeutic pathway. The oncolytic virotherapy is now promising way for curing cancer.
Collapse
Affiliation(s)
- Shyam Sundar Nandi
- National Institute of Virology, (Mumbai unit), (Formerly Enterovirus Research Centre). Haffkine Institute Compound, Indian Council of Medical Research, A. D. Marg, Parel. Mumbai-12. India
| | - Trupti Gohil
- National Institute of Virology, (Mumbai unit), (Formerly Enterovirus Research Centre). Haffkine Institute Compound, Indian Council of Medical Research, A. D. Marg, Parel. Mumbai-12. India
| | - Sonali Ankush Sawant
- National Institute of Virology, (Mumbai unit), (Formerly Enterovirus Research Centre). Haffkine Institute Compound, Indian Council of Medical Research, A. D. Marg, Parel. Mumbai-12. India
| | - Upendra Pradeep Lambe
- National Institute of Virology, (Mumbai unit), (Formerly Enterovirus Research Centre). Haffkine Institute Compound, Indian Council of Medical Research, A. D. Marg, Parel. Mumbai-12. India
| | - Sudip Ghosh
- Molecular Biology Division, ICMR-National Institute of Nutrition, Jamai-Osmania PO, Hyderabad. India
| | - Snehasis Jana
- Trivedi Science Research Laboratory Pvt Ltd., Thane-West, Maharashtra-400604. India
| |
Collapse
|
9
|
Johnston RJ, Lee PS, Strop P, Smyth MJ. Cancer Immunotherapy and the Nectin Family. ANNUAL REVIEW OF CANCER BIOLOGY-SERIES 2021. [DOI: 10.1146/annurev-cancerbio-060920-084910] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
It is increasingly clear that the nectin family and its immunoreceptors shape the immune response to cancer through several pathways. Yet, even as antibodies against TIGIT, CD96, and CD112R advance into clinical development, biological and therapeutic questions remain unanswered. Here, we review recent progress, prospects, and challenges to understanding and tapping this family in cancer immunotherapy.
Collapse
Affiliation(s)
- Robert J. Johnston
- Oncology Discovery, Bristol Myers Squibb, Redwood City, California 94063, USA
| | - Peter S. Lee
- Discovery Biotherapeutics, Bristol Myers Squibb, Redwood City, California 94063, USA;,
| | - Pavel Strop
- Discovery Biotherapeutics, Bristol Myers Squibb, Redwood City, California 94063, USA;,
| | - Mark J. Smyth
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland 4006, Australia
| |
Collapse
|
10
|
McKay ZP, Brown MC, Gromeier M. Aryl Hydrocarbon Receptor Signaling Controls CD155 Expression on Macrophages and Mediates Tumor Immunosuppression. THE JOURNAL OF IMMUNOLOGY 2021; 206:1385-1394. [PMID: 33504618 DOI: 10.4049/jimmunol.2000792] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 01/05/2021] [Indexed: 01/02/2023]
Abstract
Crosstalk between costimulatory and coinhibitory ligands are a prominent node of immune cell regulation. Mounting evidence points toward a critical role for CD155, the poliovirus receptor, in suppressing T cell function, particularly in cancer. However, relative to other known costimulatory/coinhibitory ligands (e.g., CD86, CD80, PD-L1), the physiological functions of CD155 and the mechanisms controlling its expression remain unclear. We discovered that CD155 expression is coregulated with PD-L1 on tumor-associated macrophages, is transcriptionally regulated by persistently active aryl hydrocarbon receptor (AhR), and can be targeted for suppression via AhR inhibition in vivo. Therapeutic inhibition of AhR reversed tumor immunosuppression in an immune competent murine tumor model, and markers of AhR activity were highly correlated with tumor-associated macrophage markers in human glioblastomas. Thus, CD155 functions within a broader, AhR-controlled macrophage activation phenotype that can be targeted to reverse tumor immunosuppression.
Collapse
Affiliation(s)
- Zachary P McKay
- Department of Neurosurgery, Duke University Medical School, Durham, NC 27710
| | - Michael C Brown
- Department of Neurosurgery, Duke University Medical School, Durham, NC 27710
| | - Matthias Gromeier
- Department of Neurosurgery, Duke University Medical School, Durham, NC 27710
| |
Collapse
|
11
|
Abstract
Checkpoint inhibitors have become an efficient way to treat cancers. Indeed, anti-CTLA-4, anti-PD1, and anti-PDL-1 antibodies are now used as therapies for cancers. However, while these therapies are very efficient in certain tumors, they remain poorly efficient in others. This might be explained by the immune infiltrate, the expression of target molecules, and the influence of the tumor microenvironment. It is therefore critical to identify checkpoint antigens that represent alternative targets for immunotherapies. PVR-like molecules play regulatory roles in immune cell functions. These proteins are expressed by different cell types and have been shown to be upregulated in various malignancies. PVR and Nectin-2 are expressed by tumor cells as well as myeloid cells, while TIGIT, CD96, and DNAM-1 are expressed on effector lymphoid cells. PVR is able to bind DNAM-1, CD96, and TIGIT, which results in two distinct profiles of effector cell activation. Indeed, while binding to DNAM-1 induces the release of cytokines and cytotoxicity of cytotoxic effector cells, binding TIGIT induces an immunosuppressive and non-cytotoxic profile. PVR is also able to bind CD96, which induces an immunosuppressive response in murine models. Unfortunately, in humans, results remain contradictory, and this interaction might induce the activation or the suppression of the immune response. Similarly, Nectin-2 was shown to bind TIGIT and to induce regulatory profiles in effectors cells such as NK and T cells. Therefore, these data highlight the potential of each of the molecules of the “PVR–TIGIT axis” as a potential target for immune checkpoint therapy. However, many questions remain to be answered to fully understand the mechanisms of this synapse, in particular for human CD96 and Nectin-2, which are still understudied. Here, we review the recent advances in “PVR–TIGIT axis” research and discuss the potential of targeting this axis by checkpoint immunotherapies.
Collapse
Affiliation(s)
- Laurent Gorvel
- Cancer Research Center of Marseille, INSERM U1068, CNRS U7258, Aix Marseille Université, Institut Paoli - Calmettes, Marseille, France
| | - Daniel Olive
- Cancer Research Center of Marseille, INSERM U1068, CNRS U7258, Aix Marseille Université, Institut Paoli - Calmettes, Marseille, France
| |
Collapse
|
12
|
Kučan Brlić P, Lenac Roviš T, Cinamon G, Tsukerman P, Mandelboim O, Jonjić S. Targeting PVR (CD155) and its receptors in anti-tumor therapy. Cell Mol Immunol 2019; 16:40-52. [PMID: 30275538 PMCID: PMC6318332 DOI: 10.1038/s41423-018-0168-y] [Citation(s) in RCA: 118] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 08/20/2018] [Indexed: 12/22/2022] Open
Abstract
Poliovirus receptor (PVR, CD155) has recently been gaining scientific interest as a therapeutic target in the field of tumor immunology due to its prominent endogenous and immune functions. In contrast to healthy tissues, PVR is expressed at high levels in several human malignancies and seems to have protumorigenic and therapeutically attractive properties that are currently being investigated in the field of recombinant oncolytic virotherapy. More intriguingly, PVR participates in a considerable number of immunoregulatory functions through its interactions with activating and inhibitory immune cell receptors. These functions are often modified in the tumor microenvironment, contributing to tumor immunosuppression. Indeed, increasing evidence supports the rationale for developing strategies targeting these interactions, either in terms of checkpoint therapy (i.e., targeting inhibitory receptors) or in adoptive cell therapy, which targets PVR as a tumor marker.
Collapse
Affiliation(s)
- Paola Kučan Brlić
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51 000, Rijeka, Croatia.
| | - Tihana Lenac Roviš
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51 000, Rijeka, Croatia
| | - Guy Cinamon
- Nectin Therapeutics Ltd., Hi-Tech Campus Givat Ram, POB 39135, 91390, Jerusalem, Israel
| | - Pini Tsukerman
- Nectin Therapeutics Ltd., Hi-Tech Campus Givat Ram, POB 39135, 91390, Jerusalem, Israel
| | - Ofer Mandelboim
- The Lautenberg Center for General and Tumor Immunology, The Faculty of Medicine, IMRIC, The Hebrew University Medical School, Jerusalem, Israel
| | - Stipan Jonjić
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51 000, Rijeka, Croatia.
| |
Collapse
|
13
|
Zhand S, Hosseini SM, Tabarraei A, Saeidi M, Jazi MS, Kalani MR, Moradi A. Oral poliovirus vaccine-induced programmed cell death involves both intrinsic and extrinsic pathways in human colorectal cancer cells. Oncolytic Virother 2018; 7:95-105. [PMID: 30464928 PMCID: PMC6214410 DOI: 10.2147/ov.s177260] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
PURPOSE Colorectal cancer (CRC) is one of the most common causes of cancer death throughout the world. Replication-competent viruses, which are naturally able to infect and lyse tumor cells, seem to be promising in this field. The aim of this study was to evaluate the potential of oral poliovirus vaccine (OPV) on human CRC cells and elucidate the mechanism of apoptosis induction. MATERIALS AND METHODS Protein and gene expression of poliovirus (PV) receptor (CD155) on four human CRC cell lines including HCT116, SW480, HT-29, and Caco-2 and normal fetal human colon (FHC) cell line as a control were examined by flow cytometry and SYBR Green Real-Time PCR, respectively. Cytotoxicity of OPV on indicated cell lines was tested using MTT assay. The ability of OPV on apoptosis induction for both intrinsic and extrinsic pathways was examined using caspase-8 and caspase-9 colorimetric assay kits. The PV propagation in mentioned cell lines was investigated, and the quantity of viral yields (cells associated and extracellular) was determined using TaqMan PCR. RESULTS CD155 mRNA and protein were expressed significantly higher in studied CRC cell lines rather than the normal cell line (P=0). OPV induced cell death in a time- and dose-dependent manner in human CRC cells. Apoptosis through both extrinsic and intrinsic pathways was detected in CRC cells with the minimum level found in FHC. PV viral load was significantly correlated with apoptosis via extrinsic (R=0.945, P=0.0001) and intrinsic (R=0.756, P=0.001) pathways. CONCLUSION This study suggests that OPV has potential for clinical treatment of CRC. However further studies in animal models (tumor xenografts) are needed to be certain that it is qualified enough for treatment of CRC.
Collapse
Affiliation(s)
- Sareh Zhand
- Department of Microbiology, Faculty of Biological Sciences and Technology, Shahid Beheshti University, Tehran, Iran
| | - Seyed Masoud Hosseini
- Department of Microbiology, Faculty of Biological Sciences and Technology, Shahid Beheshti University, Tehran, Iran
| | - Alijan Tabarraei
- Department of Microbiology, School of Medicine, Golestan University of Medical Sciences, Gorgan, Iran,
| | - Mohsen Saeidi
- Department of Microbiology, School of Medicine, Golestan University of Medical Sciences, Gorgan, Iran,
| | - Marie Saghaeian Jazi
- Metabolic Disorders Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Mohamad Reza Kalani
- Department of Biochemistry, School of Molecular and Cell Biology, University of Illinois at Urbana Champaign, Urbana, IL, USA
| | - Abdolvahab Moradi
- Department of Microbiology, School of Medicine, Golestan University of Medical Sciences, Gorgan, Iran,
| |
Collapse
|
14
|
TIGIT-Fc alleviates acute graft-versus-host disease by suppressing CTL activation via promoting the generation of immunoregulatory dendritic cells. Biochim Biophys Acta Mol Basis Dis 2018; 1864:3085-3098. [PMID: 29960041 DOI: 10.1016/j.bbadis.2018.06.022] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 06/25/2018] [Accepted: 06/26/2018] [Indexed: 02/07/2023]
Abstract
Graft-versus-host disease (GVHD) is the most common complication and major limitation of allogeneic hematopoietic stem cell transplantation. The CD226/TIGIT-CD155 signal is critical for the cross-talk between T cells and dendritic cells (DCs). Studies have shown that blockade of the CD226-CD155 interaction, using an anti-CD226 antibody, can significantly ameliorate GVHD. It has also been reported that a TIGIT-Fc fusion protein exerts immunosuppressive effects by binding to CD155 on DCs. Here, we used a mouse allogeneic acute GVHD model to explore the therapeutic potential and mechanism of action of TIGIT-Fc. C57/BL6 and Balb/c mice were used as hematopoietic cell graft donors and recipients, respectively. In the TIGIT-Fc-treated mice, GVHD symptom occurrence and mortality were delayed compared to that in isotype control group mice. Histopathological analyses revealed that following TIGIT-Fc treatment, liver and small intestine tissue damage was reduced with minimal lymphocytic infiltration. The percentage of CD8+IFN-γ+ and CD8+ granzyme B+ cells significantly decreased in the TIGIT-Fc group. Moreover, treatment with TIGIT-Fc, even after the onset of GVHD, ameliorated symptoms and prolonged survival. TIGIT-Fc also inhibited CD8+ T cell activation in vitro; this was dependent on the presence of CD155 on bone marrow-derived dendritic cells (BMDCs) and on IL-10 production. In addition, TIGIT-CD155 ligation triggered both Erk phosphorylation and STAT3 nuclear translocation. These data indicate that TIGIT plays an important role in the development of GVHD and is an ideal molecular target to treat acute GVHD.
Collapse
|
15
|
Celeste DB, Miller MS. Reviewing the evidence for viruses as environmental risk factors for ALS: A new perspective. Cytokine 2018; 108:173-178. [PMID: 29684753 DOI: 10.1016/j.cyto.2018.04.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 04/05/2018] [Accepted: 04/07/2018] [Indexed: 12/11/2022]
Abstract
Amyotrophic lateral sclerosis is a devastating neurodegenerative disease whose etiology remains poorly understood. Since the genetic basis of disease is known in only a small subset of cases, there has been substantial interest in determining whether environmental factors act as triggers of ALS. Viruses have received longstanding attention as potential ALS triggers. Yet, existing studies have not provided a compelling case for causation. This review summarizes the evidence supporting a link between viral infection and motor neuron disease, with a focus on ALS. Limitations of prior studies are discussed and contextualized, and recent work that has provided stronger mechanistic evidence for viruses in disease pathogenesis is highlighted. Finally, we offer a new perspective on the association of viruses with ALS, and underscore the need for multidisciplinary approaches bridging neurology and infectious diseases research to move the field forward in the future.
Collapse
Affiliation(s)
- Daniel B Celeste
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster Immunology Research Centre, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
| | - Matthew S Miller
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster Immunology Research Centre, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada.
| |
Collapse
|
16
|
|
17
|
Masemann D, Boergeling Y, Ludwig S. Employing RNA viruses to fight cancer: novel insights into oncolytic virotherapy. Biol Chem 2017; 398:891-909. [DOI: 10.1515/hsz-2017-0103] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 04/08/2017] [Indexed: 12/13/2022]
Abstract
Abstract
Within recent decades, viruses that specifically target tumor cells have emerged as novel therapeutic agents against cancer. These viruses do not only act via their cell-lytic properties, but also harbor immunostimulatory features to re-direct the tumor microenvironment and stimulate tumor-directed immune responses. Furthermore, oncolytic viruses are considered to be superior to classical cancer therapies due to higher selectivity towards tumor cell destruction and, consequently, less collateral damage of non-transformed healthy tissue. In particular, the field of oncolytic RNA viruses is rapidly developing since these agents possess alternative tumor-targeting strategies compared to established oncolytic DNA viruses. Thus, oncolytic RNA viruses have broadened the field of virotherapy facilitating new strategies to fight cancer. In addition to several naturally occurring oncolytic viruses, genetically modified RNA viruses that are armed to express foreign factors such as immunostimulatory molecules have been successfully tested in early clinical trials showing promising efficacy. This review aims to provide an overview of the most promising RNA viruses in clinical development, to summarize the current knowledge of clinical trials using these viral agents, and to discuss the main issues as well as future perspectives of clinical approaches using oncolytic RNA viruses.
Collapse
|
18
|
Anastasina M, Domanska A, Palm K, Butcher S. Human picornaviruses associated with neurological diseases and their neutralization by antibodies. J Gen Virol 2017. [PMID: 28631594 DOI: 10.1099/jgv.0.000780] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Picornaviruses are the most commonly encountered infectious agents in mankind. They typically cause mild infections of the gastrointestinal or respiratory tract, but sometimes also invade the central nervous system. There, they can cause severe diseases with long-term sequelae and even be lethal. The most infamous picornavirus is poliovirus, for which significant epidemics of poliomyelitis were reported from the end of the nineteenth century. A successful vaccination campaign has brought poliovirus close to eradication, but neurological diseases caused by other picornaviruses have increasingly been reported since the late 1990s. In this review we focus on enterovirus 71, coxsackievirus A16, enterovirus 68 and human parechovirus 3, which have recently drawn attention because of their links to severe neurological diseases. We discuss the clinical relevance of these viruses and the primary role of humoral immunity in controlling them, and summarize current knowledge on the neutralization of such viruses by antibodies.
Collapse
Affiliation(s)
- Maria Anastasina
- Institute of Biotechnology and Department of Biosciences, University of Helsinki, Viikinkaari 1, 00790 Helsinki, Finland.,Protobios LLC, Mäealuse 4, 12618 Tallinn, Estonia
| | - Aušra Domanska
- Institute of Biotechnology and Department of Biosciences, University of Helsinki, Viikinkaari 1, 00790 Helsinki, Finland
| | - Kaia Palm
- Protobios LLC, Mäealuse 4, 12618 Tallinn, Estonia.,Institute of Chemistry and Biotechnology, Tallinn University of Technology, Akadeemia tee 15, 12618 Tallinn, Estonia
| | - Sarah Butcher
- Institute of Biotechnology and Department of Biosciences, University of Helsinki, Viikinkaari 1, 00790 Helsinki, Finland
| |
Collapse
|
19
|
He Y, Peng H, Sun R, Wei H, Ljunggren HG, Yokoyama WM, Tian Z. Contribution of inhibitory receptor TIGIT to NK cell education. J Autoimmun 2017; 81:1-12. [PMID: 28438433 DOI: 10.1016/j.jaut.2017.04.001] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 04/01/2017] [Accepted: 04/03/2017] [Indexed: 01/20/2023]
Abstract
Engagement of inhibitory receptors by cognate host MHC-I molecules triggers NK cell education, resulting in functional maturation and allowing NK cells to sense missing-self. However, NK cells also express inhibitory receptors for non-MHC-I ligands and their role in NK cell education is poorly understood. TIGIT is a recently identified inhibitory receptor that recognizes a non-MHC-I ligand CD155. Here, we demonstrated that TIGIT+ NK cells from wild-type mice exerted augmented responsiveness to various stimuli, including targets that lacked expression of CD155 ligand. TIGIT+ NK cells derived from CD155-deficient hosts, however, exhibited functional impairment, indicating that the engagement of TIGIT receptor by host CD155 promoted NK cell functional maturation. Furthermore, TIGIT deficiency impaired NK cell-mediated missing-self recognition and rejection of CD155- targets, such as allogenic splenocytes and certain tumor cells, in an MHC-I-independent and CD226-unrelated manner. Thus, TIGIT-CD155 pathway is also involved in the acquisition of optimal NK cell effector function, representing a novel MHC-I-independent education mechanism for NK cell tolerance and activation.
Collapse
Affiliation(s)
- Yuke He
- Institute of Immunology, Key Laboratory of Innate Immunity and Chronic Disease of Chinese Academy of Science, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, 230027, China
| | - Hui Peng
- Institute of Immunology, Key Laboratory of Innate Immunity and Chronic Disease of Chinese Academy of Science, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, 230027, China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Rui Sun
- Institute of Immunology, Key Laboratory of Innate Immunity and Chronic Disease of Chinese Academy of Science, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, 230027, China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, China.
| | - Haiming Wei
- Institute of Immunology, Key Laboratory of Innate Immunity and Chronic Disease of Chinese Academy of Science, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, 230027, China
| | - Hans-Gustaf Ljunggren
- Center for Infectious Medicine, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, 16451, Sweden
| | - Wayne M Yokoyama
- Howard Hughes Medical Institute, Washington University School of Medicine, St. Louis, Missouri, 63123, USA
| | - Zhigang Tian
- Institute of Immunology, Key Laboratory of Innate Immunity and Chronic Disease of Chinese Academy of Science, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, 230027, China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, China.
| |
Collapse
|
20
|
Gardeck AM, Sheehan J, Low WC. Immune and viral therapies for malignant primary brain tumors. Expert Opin Biol Ther 2017; 17:457-474. [DOI: 10.1080/14712598.2017.1296132] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Andrew M. Gardeck
- Departments of Neurosurgery, University of Minnesota, Minneapolis, MN, USA
| | - Jordan Sheehan
- Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA
| | - Walter C. Low
- Departments of Neurosurgery, University of Minnesota, Minneapolis, MN, USA
- Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN, USA
- Microbiology, Immunology, and Cancer Biology Graduate Program, University of Minnesota, Minneapolis, MN, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
21
|
Neurotropic Enterovirus Infections in the Central Nervous System. Viruses 2015; 7:6051-66. [PMID: 26610549 PMCID: PMC4664993 DOI: 10.3390/v7112920] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 11/06/2015] [Accepted: 11/13/2015] [Indexed: 02/03/2023] Open
Abstract
Enteroviruses are a group of positive-sense single stranded viruses that belong to the Picornaviridae family. Most enteroviruses infect humans from the gastrointestinal tract and cause mild symptoms. However, several enteroviruses can invade the central nervous system (CNS) and result in various neurological symptoms that are correlated to mortality associated with enteroviral infections. In recent years, large outbreaks of enteroviruses occurred worldwide. Therefore, these neurotropic enteroviruses have been deemed as re-emerging pathogens. Although these viruses are becoming large threats to public health, our understanding of these viruses, especially for non-polio enteroviruses, is limited. In this article, we review recent advances in the trafficking of these pathogens from the peripheral to the central nervous system, compare their cell tropism, and discuss the effects of viral infections in their host neuronal cells.
Collapse
|
22
|
Muehlenbachs A, Bhatnagar J, Zaki SR. Tissue tropism, pathology and pathogenesis of enterovirus infection. J Pathol 2015; 235:217-28. [PMID: 25211036 DOI: 10.1002/path.4438] [Citation(s) in RCA: 115] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Revised: 09/08/2014] [Accepted: 09/09/2014] [Indexed: 02/04/2023]
Abstract
Enteroviruses are very common and cause infections with a diverse array of clinical features. Enteroviruses are most frequently considered by practising pathologists in cases of aseptic meningitis, encephalitis, myocarditis and disseminated infections in neonates and infants. Congenital infections have been reported and transplacental transmission is thought to occur. Although skin biopsies during hand, foot and mouth disease are infrequently obtained, characteristic dermatopathological findings can be seen. Enteroviruses have been implicated in lower respiratory tract infections. This review highlights histopathological features of enterovirus infection and discusses diagnostic modalities for formalin-fixed paraffin-embedded tissues and their associated pitfalls. Immunohistochemistry can detect enterovirus antigen within cells of affected tissues; however, assays can be non-specific and detect other viruses. Molecular methods are increasingly relied upon but, due to the high frequency of asymptomatic enteroviral infections, clinical-pathological correlation is needed to determine significance. Of note, diagnostic assays on central nervous system or cardiac tissues from immunocompetent patients with prolonged disease courses are most often negative. Histopathological, immunohistochemical and molecular studies performed on clinical specimens also provide insight into enteroviral tissue tropism and pathogenesis.
Collapse
Affiliation(s)
- Atis Muehlenbachs
- Infectious Diseases Pathology Branch, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | | | | |
Collapse
|
23
|
Brown MC, Dobrikova EY, Dobrikov MI, Walton RW, Gemberling SL, Nair SK, Desjardins A, Sampson JH, Friedman HS, Friedman AH, Tyler DS, Bigner DD, Gromeier M. Oncolytic polio virotherapy of cancer. Cancer 2014; 120:3277-86. [PMID: 24939611 DOI: 10.1002/cncr.28862] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Accepted: 05/13/2014] [Indexed: 01/23/2023]
Abstract
Recently, the century-old idea of targeting cancer with viruses (oncolytic viruses) has come of age, and promise has been documented in early stage and several late-stage clinical trials in a variety of cancers. Although originally prized for their direct tumor cytotoxicity (oncolytic virotherapy), recently, the proinflammatory and immunogenic effects of viral tumor infection (oncolytic immunotherapy) have come into focus. Indeed, a capacity for eliciting broad, sustained antineoplastic effects stemming from combined direct viral cytotoxicity, innate antiviral activation, stromal proinflammatory stimulation, and recruitment of adaptive immune effector responses is the greatest asset of oncolytic viruses. However, it also is the source for enormous mechanistic complexity that must be considered for successful clinical translation. Because of fundamentally different relationships with their hosts (malignant or not), diverse replication strategies, and distinct modes of tumor cytotoxicity/killing, oncolytic viruses should not be referred to collectively. These agents must be evaluated based on their individual merits. In this review, the authors highlight key mechanistic principles of cancer treatment with the polio:rhinovirus chimera PVSRIPO and their implications for oncolytic immunotherapy in the clinic.
Collapse
Affiliation(s)
- Michael C Brown
- Department of Surgery, Duke Cancer Institute, Duke University Medical Center, Durham, North Carolina; Division of Neurosurgery Duke Cancer Institute, Duke University Medical Center, Durham, North Carolina; Department of Molecular Genetics and Microbiology, Duke Cancer Institute, Duke University Medical Center, Durham, North Carolina
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Kureha F, Satomi-Kobayashi S, Kubo Y, Kinugasa M, Ishida T, Takai Y, Hirata KI, Rikitake Y. Nectin-Like Molecule-5 Regulates Intimal Thickening After Carotid Artery Ligation in Mice. Arterioscler Thromb Vasc Biol 2013; 33:1206-11. [DOI: 10.1161/atvbaha.113.301425] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Affiliation(s)
- Fumie Kureha
- From the Division of Cardiovascular Medicine, Department of Internal Medicine (F.K., S.S.-K., M.K., T.I., K.-I.H., Y.R.), Division of Signal Transduction (Y.K., Y.R.), and Division of Molecular and Cellular Biology (Y.T., Y.R.), Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Seimi Satomi-Kobayashi
- From the Division of Cardiovascular Medicine, Department of Internal Medicine (F.K., S.S.-K., M.K., T.I., K.-I.H., Y.R.), Division of Signal Transduction (Y.K., Y.R.), and Division of Molecular and Cellular Biology (Y.T., Y.R.), Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yoshiki Kubo
- From the Division of Cardiovascular Medicine, Department of Internal Medicine (F.K., S.S.-K., M.K., T.I., K.-I.H., Y.R.), Division of Signal Transduction (Y.K., Y.R.), and Division of Molecular and Cellular Biology (Y.T., Y.R.), Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Mitsuo Kinugasa
- From the Division of Cardiovascular Medicine, Department of Internal Medicine (F.K., S.S.-K., M.K., T.I., K.-I.H., Y.R.), Division of Signal Transduction (Y.K., Y.R.), and Division of Molecular and Cellular Biology (Y.T., Y.R.), Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Tatsuro Ishida
- From the Division of Cardiovascular Medicine, Department of Internal Medicine (F.K., S.S.-K., M.K., T.I., K.-I.H., Y.R.), Division of Signal Transduction (Y.K., Y.R.), and Division of Molecular and Cellular Biology (Y.T., Y.R.), Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yoshimi Takai
- From the Division of Cardiovascular Medicine, Department of Internal Medicine (F.K., S.S.-K., M.K., T.I., K.-I.H., Y.R.), Division of Signal Transduction (Y.K., Y.R.), and Division of Molecular and Cellular Biology (Y.T., Y.R.), Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Ken-ichi Hirata
- From the Division of Cardiovascular Medicine, Department of Internal Medicine (F.K., S.S.-K., M.K., T.I., K.-I.H., Y.R.), Division of Signal Transduction (Y.K., Y.R.), and Division of Molecular and Cellular Biology (Y.T., Y.R.), Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yoshiyuki Rikitake
- From the Division of Cardiovascular Medicine, Department of Internal Medicine (F.K., S.S.-K., M.K., T.I., K.-I.H., Y.R.), Division of Signal Transduction (Y.K., Y.R.), and Division of Molecular and Cellular Biology (Y.T., Y.R.), Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| |
Collapse
|
25
|
Chumakov PM, Morozova VV, Babkin IV, Baikov IK, Netesov SV, Tikunova NV. Oncolytic enteroviruses. Mol Biol 2012. [DOI: 10.1134/s0026893312050032] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
26
|
Goetz C, Dobrikova E, Shveygert M, Dobrikov M, Gromeier M. Oncolytic poliovirus against malignant glioma. Future Virol 2011; 6:1045-1058. [PMID: 21984883 DOI: 10.2217/fvl.11.76] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
In cancerous cells, physiologically tight regulation of protein synthesis is lost, contributing to uncontrolled growth and proliferation. We describe a novel experimental cancer therapy approach based on genetically recombinant poliovirus that targets an intriguing aberration of translation control in malignancy. This strategy is based on the confluence of several factors enabling specific and efficacious cancer cell targeting. Poliovirus naturally targets the vast majority of ectodermal/neuroectodermal cancers expressing its cellular receptor. Evidence from glioblastoma patients suggests that the poliovirus receptor is ectopically upregulated on tumor cells and may be associated with stem cell-like cancer cell populations and proliferating tumor vasculature. We exploit poliovirus' reliance on an unorthodox mechanism of protein synthesis initiation to selectively drive viral translation, propagation and cytotoxicity in glioblastoma. PVSRIPO, a prototype nonpathogenic poliovirus recombinant, is scheduled to enter clinical investigation against glioblastoma.
Collapse
Affiliation(s)
- Christian Goetz
- Division of Neurosurgery, Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | | | |
Collapse
|
27
|
Rhoades RE, Tabor-Godwin JM, Tsueng G, Feuer R. Enterovirus infections of the central nervous system. Virology 2011; 411:288-305. [PMID: 21251690 PMCID: PMC3060663 DOI: 10.1016/j.virol.2010.12.014] [Citation(s) in RCA: 148] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2010] [Accepted: 12/08/2010] [Indexed: 12/15/2022]
Abstract
Enteroviruses (EV) frequently infect the central nervous system (CNS) and induce neurological diseases. Although the CNS is composed of many different cell types, the spectrum of tropism for each EV is considerable. These viruses have the ability to completely shut down host translational machinery and are considered highly cytolytic, thereby causing cytopathic effects. Hence, CNS dysfunction following EV infection of neuronal or glial cells might be expected. Perhaps unexpectedly given their cytolytic nature, EVs may establish a persistent infection within the CNS, and the lasting effects on the host might be significant with unanticipated consequences. This review will describe the clinical aspects of EV-mediated disease, mechanisms of disease, determinants of tropism, immune activation within the CNS, and potential treatment regimes.
Collapse
Affiliation(s)
| | | | | | - Ralph Feuer
- Corresponding author. Cell & Molecular Biology Joint Doctoral Program, Department of Biology, San Diego State University, 5500 Campanile Drive; San Diego, CA 92182-4614, USA. Fax: +1 619 594 0777.
| |
Collapse
|
28
|
Goetz C, Gromeier M. Preparing an oncolytic poliovirus recombinant for clinical application against glioblastoma multiforme. Cytokine Growth Factor Rev 2010; 21:197-203. [PMID: 20299272 DOI: 10.1016/j.cytogfr.2010.02.005] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
PVS-RIPO is a genetically recombinant, non-pathogenic poliovirus chimera with a tumor-specific conditional replication phenotype. Consisting of the genome of the live attenuated poliovirus type 1 (Sabin) vaccine with its cognate IRES element replaced with that of human rhinovirus type 2, PVS-RIPO displays an inability to translate its genome in untransformed neuronal cells, but effectively does so in cells originating from primary tumors in the central nervous system or other cancers. Hence, PVS-RIPO unleashes potent cytotoxic effects on infected cancer cells and produces sustained anti-tumoral responses in animal tumor models. PVS-RIPO presents a novel approach to the treatment of patients with glioblastoma multiforme, based on conditions favoring an unconventional viral translation initiation mechanism in cancerous cells. In this review we summarize advances in the understanding of major molecular determinants of PVS-RIPO oncolytic efficacy and safety and discuss their implications for upcoming clinical investigations.
Collapse
Affiliation(s)
- Christian Goetz
- Division of Neurosurgery, Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA
| | | |
Collapse
|
29
|
Viren mit einzelsträngigem RNA-Genom in Plusstrangorientierung. MOLEKULARE VIROLOGIE 2010. [PMCID: PMC7120496 DOI: 10.1007/978-3-8274-2241-5_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Heute sind acht Virusfamilien bekannt, deren Vertreter eine einzelsträngige RNA in Plusstrangorientierung besitzen: Die Picornaviridae, Caliciviridae, Astroviridae und Hepeviren verfügen über Capside, die keine Hüllmembran aufweisen, wohingegen die Flaviviridae, Togaviridae, Arteriviridae und Coronaviridae durch membranumhüllte Partikel gekennzeichnet sind. Allen gemeinsam ist, dass sie ihre Genome als mRNA verwenden und davon ein oder mehrere Polyproteine synthetisieren, die im weiteren Verlauf durch virale oder auch zelluläre Proteasen in Einzelkomponenten gespalten werden. Die Viren verfügen über eine RNA-abhängige RNA-Polymerase, welche die Plusstrang-RNA sowie die als Zwischenprodukte der Replikation auftretenden Negativstränge übersetzt; dabei gehen die neuen genomischen RNA-Moleküle aus dem zweiten Transkriptionsschritt hervor. Die Einteilung in die unterschiedlichen Familien richtet sich nach Zahl, Größe, Lage und Orientierung der Virusgene auf der RNA, nach der Anzahl der unterschiedlichen Polyproteine, die während der Infektion synthetisiert werden, und nach dem Vorhandensein einer Hüllmembran als Teil der Virionen.
Collapse
|
30
|
Abstract
The picornavirus family consists of a large number of small RNA viruses, many of which are significant pathogens of humans and livestock. They are amongst the simplest of vertebrate viruses comprising a single stranded positive sense RNA genome within a T = 1 (quasi T = 3) icosahedral protein capsid of approximately 30 nm diameter. The structures of a number of picornaviruses have been determined at close to atomic resolution by X-ray crystallography. The structures of cell entry intermediate particles and complexes of virus particles with receptor molecules or antibodies have also been obtained by X-ray crystallography or at a lower resolution by cryo-electron microscopy. Many of the receptors used by different picornaviruses have been identified, and it is becoming increasingly apparent that many use co-receptors and alternative receptors to bind to and infect cells. However, the mechanisms by which these viruses release their genomes and transport them across a cellular membrane to gain access to the cytoplasm are still poorly understood. Indeed, detailed studies of cell entry mechanisms have been made only on a few members of the family, and it is yet to be established how broadly the results of these are applicable across the full spectrum of picornaviruses. Working models of the cell entry process are being developed for the best studied picornaviruses, the enteroviruses. These viruses maintain particle integrity throughout the infection process and function as genome delivery modules. However, there is currently no model to explain how viruses such as cardio- and aphthoviruses that appear to simply dissociate into subunits during uncoating deliver their genomes into the cytoplasm.
Collapse
Affiliation(s)
- Tobias J. Tuthill
- Faculty of Biological Sciences, Institute for Molecular and Cellular Biology, Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, West Yorkshire LS2 9JT, UK, Institute for Animal Health, Pirbright, Surrey GU24 ONF, UK,
| | - Elisabetta Groppelli
- Faculty of Biological Sciences Institute for Molecular and Cellular Biology, Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, West Yorkshire LS2 9JT UK
| | - James M. Hogle
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA,
| | - David J. Rowlands
- Faculty of Biological Sciences Institute for Molecular and Cellular Biology, Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, West Yorkshire LS2 9JT UK
| |
Collapse
|
31
|
Merrill MK, Gromeier M. The double-stranded RNA binding protein 76:NF45 heterodimer inhibits translation initiation at the rhinovirus type 2 internal ribosome entry site. J Virol 2006; 80:6936-42. [PMID: 16809299 PMCID: PMC1489066 DOI: 10.1128/jvi.00243-06] [Citation(s) in RCA: 91] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Poliovirus (PV) plus-strand RNA genomes initiate translation in a cap-independent manner via an internal ribosome entry site (IRES) in their 5' untranslated region. Viral translation is codetermined by cellular IRES trans-acting factors, which can influence viral propagation in a cell-type-specific manner. Engineering of a poliovirus recombinant devoid of neuropathogenic properties but highly lytic in malignant glioma cells was accomplished by exchange of the cognate poliovirus IRES with its counterpart from human rhinovirus type 2 (HRV2), generating PV-RIPO. Neuroblast:glioma heterokaryon analyses revealed that loss of neurovirulence is due to trans-dominant repression of PV-RIPO propagation in neuronal cells. The double-stranded RNA binding protein 76 (DRBP76) was previously identified to bind to the HRV2 IRES in neuronal cells and to inhibit PV-RIPO translation and propagation (M. Merrill, E. Dobrikova, and M. Gromeier, J. Virol. 80:3347-3356, 2006). The results of size exclusion chromatography indicate that DRBP76 heterodimerizes with nuclear factor of activated T cells, 45 kDa (NF45), in neuronal but not in glioma cells. The DRBP76:NF45 heterodimer binds to the HRV2 IRES in neuronal but not in glioma cells. Ribosomal profile analyses show that the heterodimer preferentially associates with the translation apparatus in neuronal cells and arrests translation at the HRV2 IRES, preventing PV-RIPO RNA assembly into polysomes. Results of this study suggest that the DRBP76:NF45 heterodimer selectively blocks HRV2 IRES-driven translation initiation in neuron-derived cells.
Collapse
Affiliation(s)
- Melinda K Merrill
- Department of Molecular Genetics & Microbiology, Duke University Medical Center, Box 3020, Durham, NC 27710, USA
| | | |
Collapse
|
32
|
Merrill MK, Dobrikova EY, Gromeier M. Cell-type-specific repression of internal ribosome entry site activity by double-stranded RNA-binding protein 76. J Virol 2006; 80:3147-56. [PMID: 16537583 PMCID: PMC1440377 DOI: 10.1128/jvi.80.7.3147-3156.2006] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Translation of picornavirus plus-strand RNA genomes occurs via internal ribosomal entry at highly structured 5' untranslated regions. In addition to canonical translation factors, translation rate is likely influenced by supplementary host and viral trans-acting factors. We previously reported that insertion of a heterologous human rhinovirus type 2 internal ribosomal entry site (IRES) into the poliovirus (PV) genome, generating the chimeric virus PV-RIPO, selectively abrogates viral translation and propagation in neurons, which eliminate poliovirus's signature neuropathogenicity. While severely deficient in cells of neuronal lineage, the rhinovirus IRES promotes efficient propagation of PV-RIPO in cancer cells. Tumor-specific IRES function can be therapeutically exploited to direct viral cytotoxicity to cancer cells. Neuron-glioma heterokaryon analysis implicates neuronal trans-dominant inhibition in this effect, suggesting that host trans-acting factors repress IRES function in a cell-type-specific manner. We identified a set of proteins from neuronal cells with affinity for the rhinovirus IRES, including double-stranded RNA-binding protein 76 (DRBP76). DRBP76 associates with the IRES in neuronal but not in malignant glioma cells. Moreover, DRBP76 depletion in neuronal cells enhances rhinovirus IRES-driven translation and virus propagation. Our observations suggest that cell-type-specific association of DRBP76 with the rhinovirus IRES represses PV-RIPO translation and propagation in neuronal cells.
Collapse
Affiliation(s)
- Melinda K Merrill
- Department of Molecular Genetics & Microbiology, Duke University Medical Center, Durham, NC 27710, USA
| | | | | |
Collapse
|
33
|
Abstract
The hedgehog (Hh) family of genes, sonic hedgehog (Shh), Indian hedgehog (Ihh), and desert hedgehog (Dhh) encode signaling molecules that regulate multiple functions during organ development and in adult tissues. Altered hedgehog signaling has been implicated in disturbed organ development as well as in different degenerative and neoplastic human diseases. Hedgehog signaling plays an important role in determination the fate of the mesoderm of the gut tube, as well as in early pancreatic development, and islet cell function. Recently, it has been shown that deregulation of hedgehog signaling molecules contributes to the pathogenesis and progression of pancreatic cancer and of chronic pancreatitis. Inhibition of hedgehog signaling using hedgehog antagonists reduces pancreatic cancer cell growth in vitro and in vivo, thus holding promise of novel agents in the treatment of this devastating disease. In this review, we discuss the role of hedgehog signaling during pancreatic development, its role in the pathogenesis of both chronic pancreatitis and pancreatic cancer, and lastly, the implications of this newly available information with regards to treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Hany Kayed
- Department of General Surgery, University of Heidelberg, Heidelberg, Germany
| | | | | | | | | | | |
Collapse
|
34
|
Sloan KE, Stewart JK, Treloar AF, Matthews RT, Jay DG. CD155/PVR enhances glioma cell dispersal by regulating adhesion signaling and focal adhesion dynamics. Cancer Res 2006; 65:10930-7. [PMID: 16322240 DOI: 10.1158/0008-5472.can-05-1890] [Citation(s) in RCA: 93] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
We recently identified the immunoglobulin-CAM CD155/PVR (the poliovirus receptor) as a regulator of cancer invasiveness and glioma migration, but the mechanism through which CD155/PVR controls these processes is unknown. Here, we show that expression of CD155/PVR in rat glioma cells that normally lack this protein enhances their dispersal both in vitro and on primary brain tissue. CD155/PVR expression also reduced substrate adhesion, cell spreading, focal adhesion density, and the number of actin stress fibers in a substrate-dependent manner. Furthermore, we found that expression of CD155/PVR increased Src/focal adhesion kinase signaling in a substrate-dependent manner, enhancing the adhesion-induced activation of paxillin and p130Cas in cells adhering to vitronectin. Conversely, depletion of endogenous CD155/PVR from human glioma cells inhibited their migration, increased cell spreading, and down-regulated the same signaling pathway. These findings implicate CD155/PVR as a regulator of adhesion signaling and suggest a pathway through which glioma and other cancer cells may acquire a dispersive phenotype.
Collapse
Affiliation(s)
- Kevin E Sloan
- Department of Physiology, Tufts University School of Medicine, Boston, Massachusetts 02111, USA
| | | | | | | | | |
Collapse
|
35
|
Siafakas N, Papaventsis D, Levidiotou-Stefanou S, Vamvakopoulos NC, Markoulatos P. Classification and Structure of Echovirus 5′-UTR Sequences. Virus Genes 2005; 31:293-306. [PMID: 16175335 DOI: 10.1007/s11262-005-3244-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2005] [Revised: 02/15/2005] [Accepted: 04/21/2005] [Indexed: 11/25/2022]
Abstract
Enteroviruses are classified into two genetic clusters on the basis of 5'-UTR and all echoviruses (ECV) are classified together with coxsackie B viruses (CBV), coxsackie A viruses (CAV) types 2-10, 12, 14 and 16, and enteroviruses (EV) 68, 69, 71 and 73. During the present study, 5'-UTR-derived sequences constituting the largest part of the Internal Ribosome Entry Site (IRES) of ECVs were studied with respect to their possible secondary structures, which were predicted following the phenomenon of "covariance", i.e. the existence of evolutionary pressure in favour of structural conservation in the light of nucleotide sequence variability. In this and previous studies, no correlation between overall 5'-UTR identity and the currently recognised Human Enterovirus species was found, implying that notwithstanding their divergent protein-encoding regions, these species are free to exchange 5'-UTRs by recombination. Secondary structure features which are known to be highly conserved amongst enteroviruses and specifically the GNRA tetraloop in secondary structure domain IV, involved in long-term tertiary interactions and loop B in secondary structure domain V with an as yet unknown function were also conserved in ECVs. In contrast, the C(NANCCA)G motif, which is considered to be important in virus transcription and translation, was not conserved in all ECVs and sequence patterns observed in other enterovirus groups and rhinoviruses were recorded.
Collapse
Affiliation(s)
- Nikolaos Siafakas
- Department of Biochemistry and Biotechnology, University of Thessaly, 26 Ploutonos and Aeolou str, 41221, Larissa, Greece
| | | | | | | | | |
Collapse
|
36
|
Ochiai H, Moore SA, Archer GE, Okamura T, Chewning TA, Marks JR, Sampson JH, Gromeier M. Treatment of intracerebral neoplasia and neoplastic meningitis with regional delivery of oncolytic recombinant poliovirus. Clin Cancer Res 2005; 10:4831-8. [PMID: 15269159 DOI: 10.1158/1078-0432.ccr-03-0694] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Spread to the central nervous system (CNS) and the leptomeninges is a frequent complication of systemic cancers that is associated with serious morbidity and high mortality. We have evaluated a novel therapeutic approach against CNS complications of breast cancer based on the human neuropathogen poliovirus (PV). EXPERIMENTAL DESIGN Susceptibility to PV infection and ensuing rapid cell lysis is mediated by the cellular receptor of PV, CD155. We evaluated CD155 expression in several human breast tumor tissue specimens and cultured breast cancer cell lines. In addition, we tested an oncolytic PV recombinant for efficacy in xenotransplantation models of neoplastic meningitis and cerebral metastasis secondary to breast cancer. RESULTS We observed that breast cancer tissues and cell lines derived thereof express CD155 at levels mediating exquisite sensitivity toward PV-induced oncolysis in the latter. An association with the immunoglobulin superfamily molecule CD155 renders breast cancer a likely target for oncolytic PV recombinants. This assumption was confirmed in xenotransplantation models for neoplastic meningitis or solitary cerebral metastasis, where local virus treatment dramatically improved survival. CONCLUSIONS Our findings suggest oncolytic PV recombinants as a viable treatment option for CNS complications of breast cancer.
Collapse
Affiliation(s)
- Hidenobu Ochiai
- Department of Surgery, Duke University Medical Center, Durham, North Carolina 27710, USA
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Blondel B, Colbère-Garapin F, Couderc T, Wirotius A, Guivel-Benhassine F. Poliovirus, pathogenesis of poliomyelitis, and apoptosis. Curr Top Microbiol Immunol 2005; 289:25-56. [PMID: 15791950 DOI: 10.1007/3-540-27320-4_2] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
Poliovirus (PV) is the causal agent of paralytic poliomyelitis, an acute disease of the central nervous system (CNS) resulting in flaccid paralysis. The development of new animal and cell models has allowed the key steps of the pathogenesis of poliomyelitis to be investigated at the molecular level. In particular, it has been shown that PV-induced apoptosis is an important component of the tissue injury in the CNS of infected mice, which leads to paralysis. In this review the molecular biology of PV and the pathogenesis of poliomyelitis are briefly described, and then several models of PV-induced apoptosis are considered; the role of the cellular receptor of PV, CD155, in the modulation of apoptosis is also addressed.
Collapse
Affiliation(s)
- B Blondel
- Laboratoire des Virus Entérotropes et Stratégies Antivirales, Institut Pasteur, 75724 Paris Cedex 15, France.
| | | | | | | | | |
Collapse
|
38
|
Okabe N, Ozaki-Kuroda K, Nakanishi H, Shimizu K, Takai Y. Expression patterns of nectins and afadin during epithelial remodeling in the mouse embryo. Dev Dyn 2004; 230:174-86. [PMID: 15108322 DOI: 10.1002/dvdy.20033] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Cell-cell adhesion plays key roles in tissue morphogenesis and organogenesis. Nectins are Ca2+-independent immunoglobulin-like cell adhesion molecules connected to the actin cytoskeleton through afadin. Nectins play roles in a variety of cell-cell junctions in cooperation with or independently of cadherins. Here, we examined the cellular localization of nectins and afadin throughout primitive streak, neural plate, and early organogenesis stages of mouse development. Nectin and afadin localization coincided with a honeycomb-shaped meshwork of actin filaments at adherens junctions of polarized epithelia, including neuroepithelium, epithelial somites, and facial primordia. As organogenesis progressed, nectin-2 expression was maintained in general columnar epithelia, whereas nectin-1 and -3 became highly concentrated at sites of neural morphogenesis. Moreover, nectin-1 was highly expressed in keratinocytes of the skin, developing hair follicles, and epithelium of developing teeth. These results suggest that nectins and afadin are involved in dynamic epithelial remodeling during mouse development.
Collapse
Affiliation(s)
- Noriko Okabe
- Department of Molecular Biology and Biochemistry, Osaka University Graduate School of Medicine/Faculty of Medicine, Suita, Osaka, Japan
| | | | | | | | | |
Collapse
|
39
|
CD155/PVR plays a key role in cell motility during tumor cell invasion and migration. BMC Cancer 2004; 4:73. [PMID: 15471548 PMCID: PMC524493 DOI: 10.1186/1471-2407-4-73] [Citation(s) in RCA: 187] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2004] [Accepted: 10/07/2004] [Indexed: 02/01/2023] Open
Abstract
Background Invasion is an important early step of cancer metastasis that is not well understood. Developing therapeutics to limit metastasis requires the identification and validation of candidate proteins necessary for invasion and migration. Methods We developed a functional proteomic screen to identify mediators of tumor cell invasion. This screen couples Fluorophore Assisted Light Inactivation (FALI) to a scFv antibody library to systematically inactivate surface proteins expressed by human fibrosarcoma cells followed by a high-throughput assessment of transwell invasion. Results Using this screen, we have identified CD155 (the poliovirus receptor) as a mediator of tumor cell invasion through its role in migration. Knockdown of CD155 by FALI or by RNAi resulted in a significant decrease in transwell migration of HT1080 fibrosarcoma cells towards a serum chemoattractant. CD155 was found to be highly expressed in multiple cancer cell lines and primary tumors including glioblastoma (GBM). Knockdown of CD155 also decreased migration of U87MG GBM cells. CD155 is recruited to the leading edge of migrating cells where it colocalizes with actin and αv-integrin, known mediators of motility and adhesion. Knockdown of CD155 also altered cellular morphology, resulting in cells that were larger and more elongated than controls when plated on a Matrigel substrate. Conclusion These results implicate a role for CD155 in mediating tumor cell invasion and migration and suggest that CD155 may contribute to tumorigenesis.
Collapse
|
40
|
Merrill MK, Bernhardt G, Sampson JH, Wikstrand CJ, Bigner DD, Gromeier M. Poliovirus receptor CD155-targeted oncolysis of glioma. Neuro Oncol 2004; 6:208-17. [PMID: 15279713 PMCID: PMC1871993 DOI: 10.1215/s1152851703000577] [Citation(s) in RCA: 97] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Cell adhesion molecules of the immunoglobulin superfamily are aberrantly expressed in malignant glioma. Amongst these, the human poliovirus receptor CD155 provides a molecular target for therapeutic intervention with oncolytic poliovirus recombinants. Poliovirus has been genetically modified through insertion of regulatory sequences derived from human rhinovirus type 2 to selectively replicate within and destroy cancerous cells. Efficacious oncolysis mediated by poliovirus derivatives depends on the presence of CD155 in targeted tumors. To prepare oncolytic polioviruses for clinical application, we have developed a series of assays in high-grade malignant glioma (HGL) to characterize CD155 expression levels and susceptibility to oncolytic poliovirus recombinants. Analysis of 6 HGL cases indicates that CD155 is expressed in these tumors and in primary cell lines derived from these tumors. Upregulation of the molecular target CD155 rendered explant cultures of all studied tumors highly susceptible to a prototype oncolytic poliovirus recombinant. Our observations support the clinical application of such agents against HGL.
Collapse
Affiliation(s)
| | | | | | | | | | - Matthias Gromeier
- Address correspondence to Matthias Gromeier, Department of Molecular Genetics and Microbiology, Duke University Medical Center, Box 3020, Durham, NC 27710 (
)
| |
Collapse
|
41
|
Oda T, Ohka S, Nomoto A. Ligand stimulation of CD155alpha inhibits cell adhesion and enhances cell migration in fibroblasts. Biochem Biophys Res Commun 2004; 319:1253-64. [PMID: 15194502 DOI: 10.1016/j.bbrc.2004.05.111] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2004] [Indexed: 11/17/2022]
Abstract
CD155 (poliovirus receptor) localizes in cell-matrix adhesions and cell-cell junctions, but its role in the regulation of cell adhesion and cell motility has not been investigated. We identified a conserved immunoreceptor tyrosine-based inhibitory motif (ITIM) in the cytoplasmic domain of human CD155alpha. The ITIM was tyrosine-phosphorylated upon binding of anti-CD155 monoclonal antibody D171, poliovirus, and DNAM-1 (CD226) to human CD155alpha, and recruited SH2-domain-containing tyrosine phosphatase-2 (SHP-2). After CD155alpha stimulation with its ligands, cell adhesion was inhibited and cell motility was enhanced, effects that were associated with the phosphorylation of ITIM by Src kinases and accompanied by dephosphorylation of focal adhesion kinase and paxillin. These effects were abolished by introducing a point-mutation in Y398F into the ITIM of CD155alpha and by coexpression of a dominant negative SHP-2 mutant with CD155alpha. These results suggest that CD155alpha plays a role in the regulation of cell adhesion and cell motility.
Collapse
Affiliation(s)
- Toshiyuki Oda
- Department of Microbiology, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | | | | |
Collapse
|
42
|
Saunderson R, Yu B, Trent RJ, Pamphlett R. A polymorphism in the poliovirus receptor gene differs in motor neuron disease. Neuroreport 2004; 15:383-6. [PMID: 15076773 DOI: 10.1097/00001756-200402090-00034] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Poliovirus has been implicated in the etiology of sporadic motor neuron disease. DNA polymorphisms in the poliovirus receptor gene (PVR) are associated with persistent poliovirus infection in cell culture. PVR DNA polymorphisms were therefore studied in 110 cases of amyotrophic lateral sclerosis, 30 cases of progressive muscular atrophy (a disorder of lower motor neurons) and 280 normal controls. In exon 2 of PVR the heterozygous Ala67Thr change was detected in 20.0% of progressive muscular atrophy, 11.8% of amyotrophic lateral sclerosis and 6.8% of control subjects. The frequency of the polymorphism was significantly higher in patients with progressive muscular atrophy than in controls. Differences in the poliovirus receptor gene may result in slowly progressive viral cytopathic effects that lead to lower motor neuron forms of motor neuron disease.
Collapse
Affiliation(s)
- Rebecca Saunderson
- Department of Pathology, D06, Central Clinical School, University of Sydney, New South Wales 2006, Australia
| | | | | | | |
Collapse
|
43
|
Blondel B, Couderc T, Simonin Y, Gosselin AS, Guivel-Benhassine F. Poliovirus and Apoptosis. VIRUSES AND APOPTOSIS 2004; 36:151-69. [PMID: 15171611 DOI: 10.1007/978-3-540-74264-7_8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- B Blondel
- Unité de Neurovirologie et Régénération du Système Nerveux, Institut Pasteur, 75724 Paris cedex 15, France
| | | | | | | | | |
Collapse
|
44
|
Jackson CA, Messinger J, Palmer MT, Peduzzi JD, Morrow CD. Gene expression in the muscle and central nervous system following intramuscular inoculation of encapsidated or naked poliovirus replicons. Virology 2003; 314:45-61. [PMID: 14517059 DOI: 10.1016/s0042-6822(03)00385-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The spread of intramuscularly inoculated poliovirus to the central nervous system (CNS) has been documented in humans, monkeys, and mice transgenic for the human poliovirus receptor. Poliovirus spread is thought to be due to infection of the peripheral nerve and retrograde transport of poliovirus through the axon to the neuron cell body, where final virus uncoating occurs and translation/replication ensues. In previous studies, we have shown that polio-based vectors (replicons) can be used for gene delivery to motor neurons of the CNS. Using a replicon that encodes green fluorescent protein (GFP), we found that following intrathecal inoculation, GFP expression was confined to motorneurons of the spinal cord. To further characterize the gene expression of poliovirus in the periphery and CNS, we have intramuscularly inoculated transgenic mice with poliovirus replicons encoding GFP. Expression of GFP was demonstrated in the muscle, sciatic nerve, dorsal root ganglion, and the ventral horn motorneurons following intramuscular inoculation. There was no evidence of paralysis or behavioral abnormalities in the mice following intramuscular inoculation of the replicon encoding GFP. Injection of replicon RNA alone (naked RNA) into the muscle of transgenic mice or rats, which do not express the poliovirus receptor, also resulted in expression of GFP in the muscle, sciatic nerve, dorsal root ganglion, and ventral horn motorneurons, indicating that transport of the replicon RNA from the periphery to CNS had occurred. GFP expression was found in the muscles and sciatic nerve as early as 6 h after injection of replicons or replicon RNA, even after sciatic nerve section. Analysis at longer times postinjection revealed GFP expression similar to 6 h levels in the cut sciatic nerves and robust expression in the nerves of uncut animals. The infection and expression of GFP in the CNS following intramuscular inoculation of encapsidated replicons encoding GFP occurred in juvenile or adult animals. The expression of GFP in the CNS of juvenile animals was more intense and lasted for up to 5 weeks, in contrast to the duration of expression of approximately 96 h for adult animals. The results of these studies establish that poliovirus replicon RNA is expressed locally within the sciatic nerve and transported from the periphery to the CNS via axonal transport and support the potential of replicons for gene delivery to the CNS.
Collapse
Affiliation(s)
- Cheryl A Jackson
- Department of Cell Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | | | | | | | |
Collapse
|
45
|
He Y, Mueller S, Chipman PR, Bator CM, Peng X, Bowman VD, Mukhopadhyay S, Wimmer E, Kuhn RJ, Rossmann MG. Complexes of poliovirus serotypes with their common cellular receptor, CD155. J Virol 2003; 77:4827-35. [PMID: 12663789 PMCID: PMC152153 DOI: 10.1128/jvi.77.8.4827-4835.2003] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Structures of all three poliovirus (PV) serotypes (PV1, PV2, and PV3) complexed with their cellular receptor, PV receptor (PVR or CD155), were determined by cryoelectron microscopy. Both glycosylated and fully deglycosylated CD155 exhibited similar binding sites and orientations in the viral canyon for all three PV serotypes, showing that all three serotypes use a common mechanism for cell entry. Difference maps between the glycosylated and deglycosylated CD155 complexes determined the sites of the carbohydrate moieties that, in turn, helped to verify the position of the receptor relative to the viral surface. The proximity of the CD155 carbohydrate site at Asn105 to the viral surface in the receptor-virus complex suggests that it might interfere with receptor docking, an observation consistent with the properties of mutant CD155. The footprints of CD155 on PV surfaces indicate that the south rim of the canyon dominates the virus-receptor interactions and may correspond to the initial CD155 binding state of the receptor-mediated viral uncoating. In contrast, the interaction of CD155 with the north rim of the canyon, especially the region immediately outside the viral hydrophobic pocket that normally binds a cellular "pocket factor," may be critical for the release of the pocket factor, decreasing the virus stability and hence initiating uncoating. The large area of the CD155 footprint on the PV surface, in comparison with other picornavirus-receptor interactions, could be a potential limitation on the viability of PV escape mutants from antibody neutralization. Many of these are likely to have lost their ability to bind CD155, resulting in there being only three PV serotypes.
Collapse
Affiliation(s)
- Yongning He
- Department of Biological Sciences, Purdue University, West Lafayette, Indiana 47907,USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Gosselin AS, Simonin Y, Guivel-Benhassine F, Rincheval V, Vayssière JL, Mignotte B, Colbère-Garapin F, Couderc T, Blondel B. Poliovirus-induced apoptosis is reduced in cells expressing a mutant CD155 selected during persistent poliovirus infection in neuroblastoma cells. J Virol 2003; 77:790-8. [PMID: 12477887 PMCID: PMC140570 DOI: 10.1128/jvi.77.1.790-798.2003] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Poliovirus (PV) can establish persistent infections in human neuroblastoma IMR-32 cells. We previously showed that during persistent infection, specific mutations were selected in the first extracellular domain of the PV receptor (CD155) of these cells (N. Pavio, T. Couderc, S. Girard, J. Y. Sgro, B. Blondel, and F. Colbère-Garapin, Virology 274:331-342, 2000). These mutations included the Ala 67 --> Thr substitution, corresponding to a previously described allelic form of the PV receptor. The mutated CD155(Thr67) and the nonmutated IMR-32 CD155 (CD155(IMR)) were expressed independently in murine LM cells lacking the CD155 gene. Following infection of the cells with PV, we analyzed the death of cells expressing these two forms of CD155. Levels of DNA fragmentation, caspase activity, and cytochrome c release were lower in LM-CD155(Thr67) cells than in LM-CD155(IMR) cells. Thus, the level of apoptosis was lower in cells expressing mutated CD155 selected during persistent PV infection in IMR-32 than in cells expressing the wild-type receptor.
Collapse
Affiliation(s)
- Anne-Sophie Gosselin
- Unité de Neurovirologie et Régénération du Système Nerveux, Institut Pasteur, 75724 Paris cedex 15, France
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Ditunno JF, Herbison GJ. Franklin D. Roosevelt: diagnosis, clinical course, and rehabilitation from poliomyelitis. Am J Phys Med Rehabil 2002; 81:557-66. [PMID: 12172063 DOI: 10.1097/00002060-200208000-00001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
In this case report, we examine Franklin Delano Roosevelt's prognosis, clinical course, and rehabilitation from poliomyelitis, and we examine the criticisms of errors in his diagnosis and management on the basis of current knowledge of the pathophysiology of poliomyelitis. Medical and historical records reveal the onset of severe paralysis, which progressed over several days, with minimal improvement in hips and lower limbs, but recovery of facial, upper limb, and upper trunk muscles. There is no scientific basis for assertions of mismanagement that led to more severe paralysis; the paralysis was most likely caused by strenuous activities in the preparalytic phase of his illness.
Collapse
Affiliation(s)
- John F Ditunno
- Department of Rehabilitation Medicine, Jefferson Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, USA
| | | |
Collapse
|
48
|
Solecki DJ, Gromeier M, Mueller S, Bernhardt G, Wimmer E. Expression of the human poliovirus receptor/CD155 gene is activated by sonic hedgehog. J Biol Chem 2002; 277:25697-702. [PMID: 11983699 DOI: 10.1074/jbc.m201378200] [Citation(s) in RCA: 63] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The human poliovirus receptor/CD155 is a transmembrane glycoprotein belonging to the immunoglobulin superfamily. The ectodomain of CD155 mediates cell attachment to the extracellular matrix molecule vitronectin, while its intracellular domain interacts with the dynein light chain Tctex-1. CD155 is a primate-restricted gene that is expressed during development in mesenchymal tissues and ventrally derived structures within the CNS. Its function in adults is as yet unknown, but significantly, CD155 is aberrantly expressed in neuroectodermal tumors. We show that the expression of CD155 mRNA is up-regulated when human Ntera2 cells are treated with purified Sonic hedgehog (Shh) protein. Reporter gene expression driven by the CD155 core promoter is activated by Shh in transient co-transfection assays. Analysis of the CD155 core promoter indicates that an intact GLI binding site is required for Shh activation. In addition, overexpression of Gli1 or Gli3 potently activates reporter gene expression driven by the CD155 core promoter. These data identify the CD155 gene as a transcriptional target of Shh, a finding that has significance for the normal function of CD155 during development and the expression of CD155 in neuroectodermal tumors.
Collapse
Affiliation(s)
- David J Solecki
- Department of Molecular Genetics and Microbiology, School of Medicine, State University of New York at Stony Brook, 11794, USA.
| | | | | | | | | |
Collapse
|
49
|
Harvala H, Kalimo H, Dahllund L, Santti J, Hughes P, Hyypiä T, Stanway G. Mapping of tissue tropism determinants in coxsackievirus genomes. J Gen Virol 2002; 83:1697-1706. [PMID: 12075089 DOI: 10.1099/0022-1317-83-7-1697] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Genomic regions responsible for the different tissue tropisms of coxsackievirus A9 (CAV9) and coxsackievirus B3 (CBV3) in newborn mice were investigated using recombinant viruses. Infectious cDNA clones of CAV9, a virus known to infect striated muscle, and CBV3, affecting the central nervous system, pancreas, liver, brown fat and striated muscle, were used to generate chimeric viruses. In situ hybridization analysis of different tissues from mice infected with the recombinant viruses, constructed by exchanging the 5' non-coding region (5'NCR), structural and non-structural genes, demonstrated that the pancreo- and liver tropism map predominantly to CBV3 sequences within the capsid genes, evidently due to receptor recognition. Although the major neurotropism determinant in the CBV3 genome was in the capsid region, viruses containing the CAV9 capsid were also able to initiate infection in the central nervous system provided they contained the CBV3 5'NCR. The presence of the 5'NCR of CAV9 clearly enhanced muscle tissue tropism.
Collapse
Affiliation(s)
- Heli Harvala
- Department of Virology and MediCity Research Laboratory, University of Turku, Kiinamyllynkatu 13, FIN-20520 Turku, Finland1
| | - Hannu Kalimo
- Department of Pathology, University of Turku and Turku University Hospital, FIN-20520 Turku, Finland2
| | - Leif Dahllund
- Department of Virology and MediCity Research Laboratory, University of Turku, Kiinamyllynkatu 13, FIN-20520 Turku, Finland1
| | - Juhana Santti
- Department of Virology and MediCity Research Laboratory, University of Turku, Kiinamyllynkatu 13, FIN-20520 Turku, Finland1
| | - Pamela Hughes
- Department of Biological Sciences, University of Essex, Colchester CO4 3SQ, UK3
| | - Timo Hyypiä
- Department of Virology, Haartman Institute, PO Box 21, FIN-00014 Helsinki, Finland4
- Department of Virology and MediCity Research Laboratory, University of Turku, Kiinamyllynkatu 13, FIN-20520 Turku, Finland1
| | - Glyn Stanway
- Department of Biological Sciences, University of Essex, Colchester CO4 3SQ, UK3
| |
Collapse
|
50
|
Mueller S, Cao X, Welker R, Wimmer E. Interaction of the poliovirus receptor CD155 with the dynein light chain Tctex-1 and its implication for poliovirus pathogenesis. J Biol Chem 2002; 277:7897-904. [PMID: 11751937 DOI: 10.1074/jbc.m111937200] [Citation(s) in RCA: 114] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The cellular receptor for poliovirus CD155 (or PVR) is the founding member of a new class of membrane-associated immunoglobulin-like proteins, which include the mouse tumor-associated antigen E4 (Tage4) and three proteins termed "nectins." Using a yeast two-hybrid screen we have discovered that the cytoplasmic domain of CD155 associates strongly and specifically with Tctex-1, a light chain of the dynein motor complex, the latter representing the major driving force for retrograde transport of endocytic vesicles and membranous organelles. We confirmed the interaction biochemically and by co-immunoprecipitation, and we mapped the Tctex-1 binding site to a SKCSR motif within the juxtamembrane region of CD155. Tctex-1 immunoreactivity was detected in mouse sciatic nerve and spinal cord (two tissues of central importance for poliovirus pathogenesis) in punctate, possibly vesicular, patterns. We propose that the cytoplasmic domain may target CD155-containing endocytic vesicles to the microtubular network. Neurotropic viruses like poliovirus, herpesvirus, rabies virus, and pseudorabies virus all utilize neuronal retrograde transport to invade the central nervous system. Association with Tctex-1 and, hence, with the dynein motor complex may offer an explanation for how poliovirus hijacks the cellular transport machinery to retrogradely ascend along the axon to the neuronal cell body.
Collapse
Affiliation(s)
- Steffen Mueller
- Department of Molecular Genetics and Microbiology, State University of New York, Stony Brook, New York 11794, USA
| | | | | | | |
Collapse
|