1
|
Mella C, Tsarouhas P, Brockwell M, Ball HC. The Role of Chronic Inflammation in Pediatric Cancer. Cancers (Basel) 2025; 17:154. [PMID: 39796780 PMCID: PMC11719864 DOI: 10.3390/cancers17010154] [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: 12/04/2024] [Revised: 12/31/2024] [Accepted: 01/01/2025] [Indexed: 01/13/2025] Open
Abstract
Inflammation plays a crucial role in wound healing and the host immune response following pathogenic invasion. However, unresolved chronic inflammation can result in tissue fibrosis and genetic alterations that contribute to the pathogenesis of human diseases such as cancer. Recent scientific advancements exploring the underlying mechanisms of malignant cellular transformations and cancer progression have exposed significant disparities between pediatric and adult-onset cancers. For instance, pediatric cancers tend to have lower mutational burdens and arise in actively developing tissues, where cell-cycle dysregulation leads to gene, chromosomal, and fusion gene development not seen in adult-onset counterparts. As such, scientific findings in adult cancers cannot be directly applied to pediatric cancers, where unique mutations and inherent etiologies remain poorly understood. Here, we review the role of chronic inflammation in processes of genetic and chromosomal instability, the tumor microenvironment, and immune response that result in pediatric tumorigenesis transformation and explore current and developing therapeutic interventions to maintain and/or restore inflammatory homeostasis.
Collapse
Affiliation(s)
- Christine Mella
- Division of Hematology Oncology, Akron Children’s Hospital, One Perkins Square, Akron, OH 44308, USA;
| | - Panogiotis Tsarouhas
- Department of Biology, The University of Akron, 302 Buchtel Common, Akron, OH 44325, USA;
| | - Maximillian Brockwell
- College of Medicine, Northeast Ohio Medical University, 4029 State Route 44, Rootstown, OH 44272, USA;
| | - Hope C. Ball
- Division of Hematology Oncology, Akron Children’s Hospital, One Perkins Square, Akron, OH 44308, USA;
- College of Medicine, Northeast Ohio Medical University, 4029 State Route 44, Rootstown, OH 44272, USA;
- Rebecca D. Considine Research Institute, Akron Children’s Hospital, One Perkins Square, Akron, OH 44308, USA
| |
Collapse
|
2
|
Butic AB, Spencer SA, Shaheen SK, Lukacher AE. Polyomavirus Wakes Up and Chooses Neurovirulence. Viruses 2023; 15:2112. [PMID: 37896889 PMCID: PMC10612099 DOI: 10.3390/v15102112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 10/13/2023] [Accepted: 10/16/2023] [Indexed: 10/29/2023] Open
Abstract
JC polyomavirus (JCPyV) is a human-specific polyomavirus that establishes a silent lifelong infection in multiple peripheral organs, predominantly those of the urinary tract, of immunocompetent individuals. In immunocompromised settings, however, JCPyV can infiltrate the central nervous system (CNS), where it causes several encephalopathies of high morbidity and mortality. JCPyV-induced progressive multifocal leukoencephalopathy (PML), a devastating demyelinating brain disease, was an AIDS-defining illness before antiretroviral therapy that has "reemerged" as a complication of immunomodulating and chemotherapeutic agents. No effective anti-polyomavirus therapeutics are currently available. How depressed immune status sets the stage for JCPyV resurgence in the urinary tract, how the virus evades pre-existing antiviral antibodies to become viremic, and where/how it enters the CNS are incompletely understood. Addressing these questions requires a tractable animal model of JCPyV CNS infection. Although no animal model can replicate all aspects of any human disease, mouse polyomavirus (MuPyV) in mice and JCPyV in humans share key features of peripheral and CNS infection and antiviral immunity. In this review, we discuss the evidence suggesting how JCPyV migrates from the periphery to the CNS, innate and adaptive immune responses to polyomavirus infection, and how the MuPyV-mouse model provides insights into the pathogenesis of JCPyV CNS disease.
Collapse
Affiliation(s)
| | | | | | - Aron E. Lukacher
- Department of Microbiology and Immunology, Penn State College of Medicine, Hershey, PA 17033, USA; (A.B.B.); (S.A.S.); (S.K.S.)
| |
Collapse
|
3
|
Karimi AA, Tarharoudi R, Kianmehr Z, Sakhaee F, Jamnani FR, Siadat SD, Fateh A. Traces of JC polyomavirus in papillary thyroid cancer: a comprehensive study in Iran. Virol J 2022; 19:153. [PMID: 36163265 PMCID: PMC9513940 DOI: 10.1186/s12985-022-01881-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 09/16/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND JC polyomavirus (JCPyV) is known to induce solid tumors such as astrocytomas, glioblastomas, and neuroblastomas in experimental animals, and recent studies have shown that the virus may be correlated with carcinogenesis. This study aimed to evaluate the impact of JCPyV on the progression of papillary thyroid cancer (PTC). METHODS A total of 1057 samples, including 645 paraffin-embedded PTC biopsy samples (PEBS) and 412 fresh biopsy samples (FBS), and 1057 adjacent non-cancerous samples were evaluated for the presence of JCPyV DNA and RNA. RESULTS We observed that 10.8% (114/1057) samples, including 17.5% (72/412) FBS and 6.5% (42/645) PEBS were positive for the JCPyV DNA. Among the JCPyV-positive samples, the mean JCPyV copy number was lower in patients with PEBS (0.3 × 10-4 ± 0.1 × 10-4 copies/cell) compared to FBS (1.8 × 10-1 ± 0.4 × 10-1 copies/cell) and non-PTC normal samples (0.2 × 10-5 ± 0.01 × 10-5 copies/cell), with a statistically significant difference (P < 0.001). The LT-Ag RNA expression was lower in PEBS than in FBS, while no VP1 gene transcript expression was found. CONCLUSIONS Although our results confirmed the presence of JCPyV in some Iranian patients with PTC, more research is needed to verify these results.
Collapse
Affiliation(s)
- Amir Ali Karimi
- Department of Biotechnology, Faculty of Biological Science, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Rahil Tarharoudi
- Department of Molecular and Cellular Biology, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Zahra Kianmehr
- Department of Biochemistry, Faculty of Biological Science, Islamic Azad University, North Tehran Branch, Tehran, Iran
| | - Fatemeh Sakhaee
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran
| | - Fatemeh Rahimi Jamnani
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran
- Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran
| | - Seyed Davar Siadat
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran
- Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran
| | - Abolfazl Fateh
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran.
- Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
4
|
Del Valle L, Khalili K. Induction of Brain Tumors by the Archetype Strain of Human Neurotropic JCPyV in a Transgenic Mouse Model. Viruses 2021; 13:v13020162. [PMID: 33499370 PMCID: PMC7911272 DOI: 10.3390/v13020162] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 01/14/2021] [Accepted: 01/18/2021] [Indexed: 12/18/2022] Open
Abstract
JC Virus (JCPyV), a member of the Polyomaviridiæ family, is a human neurotropic virus with world-wide distribution. JCPyV is the established opportunistic infectious agent of progressive multifocal leukoencephalopathy, a fatal demyelinating disease, which results from the cytolytic infection of oligodendrocytes. Mutations in the regulatory region of JCPyV determine the different viral strains. Mad-1 the strain associated with PML contains two 98 base pair repeats, whereas the archetype strain (CY), which is the transmissible form of JCPyV, contains only one 98 tandem with two insertions of 62 and 23 base pairs respectively. The oncogenicity of JCPyV has been suspected since direct inoculation into the brain of rodents and primates resulted in the development of brain tumors and has been attributed to the viral protein, T-Antigen. To further understand the oncogenicity of JCPyV, a transgenic mouse colony containing the early region of the archetype strain (CY), under the regulation of its own promoter was generated. These transgenic animals developed tumors of neural crest origin, including: primitive neuroectodermal tumors, medulloblastomas, adrenal neuroblastomas, pituitary tumors, malignant peripheral nerve sheath tumors, and glioblastomas. Neoplastic cells from all different phenotypes express T-Antigen. The close parallels between the tumors developed by these transgenic animals and human CNS tumors make this animal model an excellent tool for the study of viral oncogenesis.
Collapse
Affiliation(s)
- Luis Del Valle
- Neurological Cancer Research, Stanley S. Scott Cancer Center, Departments of Medicine and Pathology, Louisiana State University Health, New Orleans, LA 70112, USA
- Correspondence: (L.D.V.); (K.K.)
| | - Kamel Khalili
- Department of Neurosciences and Center for Neurovirology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
- Correspondence: (L.D.V.); (K.K.)
| |
Collapse
|
5
|
Saxena R, Saribas S, Jadiya P, Tomar D, Kaminski R, Elrod JW, Safak M. Human neurotropic polyomavirus, JC virus, agnoprotein targets mitochondrion and modulates its functions. Virology 2021; 553:135-153. [PMID: 33278736 PMCID: PMC7847276 DOI: 10.1016/j.virol.2020.11.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 11/12/2020] [Indexed: 01/18/2023]
Abstract
JC virus encodes an important regulatory protein, known as Agnoprotein (Agno). We have recently reported Agno's first protein-interactome with its cellular partners revealing that it targets various cellular networks and organelles, including mitochondria. Here, we report further characterization of the functional consequences of its mitochondrial targeting and demonstrated its co-localization with the mitochondrial networks and with the mitochondrial outer membrane. The mitochondrial targeting sequence (MTS) of Agno and its dimerization domain together play major roles in this targeting. Data also showed alterations in various mitochondrial functions in Agno-positive cells; including a significant reduction in mitochondrial membrane potential, respiration rates and ATP production. In contrast, a substantial increase in ROS production and Ca2+ uptake by the mitochondria were also observed. Finally, findings also revealed a significant decrease in viral replication when Agno MTS was deleted, highlighting a role for MTS in the function of Agno during the viral life cycle.
Collapse
Affiliation(s)
- Reshu Saxena
- Department of Neuroscience, Laboratory of Molecular Neurovirology, MERB-757, Lewis Katz School of Medicine at Temple University, 3500 N. Broad Street, Philadelphia, PA, 19140, USA
| | - Sami Saribas
- Department of Neuroscience, Laboratory of Molecular Neurovirology, MERB-757, Lewis Katz School of Medicine at Temple University, 3500 N. Broad Street, Philadelphia, PA, 19140, USA
| | - Pooja Jadiya
- Center for Translational Medicine, Lewis Katz School of Medicine at Temple University, USA
| | - Dhanendra Tomar
- Center for Translational Medicine, Lewis Katz School of Medicine at Temple University, USA
| | - Rafal Kaminski
- Department of Neuroscience, Laboratory of Molecular Neurovirology, MERB-757, Lewis Katz School of Medicine at Temple University, 3500 N. Broad Street, Philadelphia, PA, 19140, USA
| | - John W Elrod
- Center for Translational Medicine, Lewis Katz School of Medicine at Temple University, USA
| | - Mahmut Safak
- Department of Neuroscience, Laboratory of Molecular Neurovirology, MERB-757, Lewis Katz School of Medicine at Temple University, 3500 N. Broad Street, Philadelphia, PA, 19140, USA.
| |
Collapse
|
6
|
Ahye N, Bellizzi A, May D, Wollebo HS. The Role of the JC Virus in Central Nervous System Tumorigenesis. Int J Mol Sci 2020; 21:ijms21176236. [PMID: 32872288 PMCID: PMC7503523 DOI: 10.3390/ijms21176236] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 08/22/2020] [Accepted: 08/24/2020] [Indexed: 12/14/2022] Open
Abstract
Cancer is the second leading cause of mortality worldwide. The study of DNA tumor-inducing viruses and their oncoproteins as a causative agent in cancer initiation and tumor progression has greatly enhanced our understanding of cancer cell biology. The initiation of oncogenesis is a complex process. Specific gene mutations cause functional changes in the cell that ultimately result in the inability to regulate cell differentiation and proliferation effectively. The human neurotropic Polyomavirus JC (JCV) belongs to the family Polyomaviridae and it is the causative agent of progressive multifocal leukoencephalopathy (PML), which is a fatal neurodegenerative disease in an immunosuppressed state. Sero-epidemiological studies have indicated JCV infection is prevalent in the population (85%) and that initial infection usually occurs during childhood. The JC virus has small circular, double-stranded DNA that includes coding sequences for viral early and late proteins. Persistence of the virus in the brain and other tissues, as well as its potential to transform cells, has made it a subject of study for its role in brain tumor development. Earlier observation of malignant astrocytes and oligodendrocytes in PML, as well as glioblastoma formation in non-human primates inoculated with JCV, led to the hypothesis that JCV plays a role in central nervous system (CNS) tumorigenesis. Some studies have reported the presence of both JC viral DNA and its proteins in several primary brain tumor specimens. The discovery of new Polyomaviruses such as the Merkel cell Polyomavirus, which is associated with Merkel cell carcinomas in humans, ignited our interest in the role of the JC virus in CNS tumors. The current evidence known about JCV and its effects, which are sufficient to produce tumors in animal models, suggest it can be a causative factor in central nervous system tumorigenesis. However, there is no clear association between JCV presence in CNS and its ability to initiate CNS cancer and tumor formation in humans. In this review, we will discuss the correlation between JCV and tumorigenesis of CNS in animal models, and we will give an overview of the current evidence for the JC virus’s role in brain tumor formation.
Collapse
|
7
|
Mazzoni E, Pellegrinelli E, Mazziotta C, Lanzillotti C, Rotondo JC, Bononi I, Iaquinta MR, Manfrini M, Vesce F, Tognon M, Martini F. Mother-to-child transmission of oncogenic polyomaviruses BKPyV, JCPyV and SV40. J Infect 2020; 80:563-570. [PMID: 32097686 DOI: 10.1016/j.jinf.2020.02.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 01/29/2020] [Accepted: 02/14/2020] [Indexed: 12/12/2022]
Abstract
OBJECTIVES Polyomavirus (PyV) infections have been associated with different diseases. BK (BKPyV), JC (JCPyV) and simian virus 40 (SV40) are the three main PyVs whose primary infection occurs early in life. Their vertical transmission was investigated in this study. METHODS PyV sequences were analyzed by the digital droplet PCR in blood, serum, placenta, amniotic fluid, vaginal smear from two independent cohorts of pregnant females and umbilical cord blood (UCB) samples. IgG antibodies against the three PyVs were investigated by indirect E.L.I.S.As with viral mimotopes. RESULTS DNAs from blood, vaginal smear and placenta tested BKPyV-, JCPyV- and SV40-positive with a distinct prevalence, while amniotic fluids were all PyVs-negative. A prevalence of 3%, 7%, and 3% for BKPyV, JCPyV and SV40 DNA sequences, respectively, was obtained in UCBs. Serum IgG antibodies from pregnant females reached an overall prevalence of 62%, 42% and 17% for BKPyV, JCPyV and SV40, respectively. Sera from newborns (UCB) tested IgG-positive with a prevalence of 10% for BKPyV/JCPyV and 3% for SV40. CONCLUSIONS In this investigation, PyV vertical transmission was revealed by detecting PyV DNA sequences and IgG antibodies in samples from females and their offspring suggesting a potential risk of diseases in newborns.
Collapse
Affiliation(s)
- Elisa Mazzoni
- Laboratories of Cell Biology and Molecular Genetics, Section of Pathology, Oncology and Experimental Biology, Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 64/b, Fossato di Mortara Street, Ferrara 44121, Italy
| | - Elena Pellegrinelli
- Laboratories of Cell Biology and Molecular Genetics, Section of Pathology, Oncology and Experimental Biology, Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 64/b, Fossato di Mortara Street, Ferrara 44121, Italy
| | - Chiara Mazziotta
- Laboratories of Cell Biology and Molecular Genetics, Section of Pathology, Oncology and Experimental Biology, Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 64/b, Fossato di Mortara Street, Ferrara 44121, Italy
| | - Carmen Lanzillotti
- Laboratories of Cell Biology and Molecular Genetics, Section of Pathology, Oncology and Experimental Biology, Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 64/b, Fossato di Mortara Street, Ferrara 44121, Italy
| | - John Charles Rotondo
- Laboratories of Cell Biology and Molecular Genetics, Section of Pathology, Oncology and Experimental Biology, Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 64/b, Fossato di Mortara Street, Ferrara 44121, Italy
| | - Ilaria Bononi
- Laboratories of Cell Biology and Molecular Genetics, Section of Pathology, Oncology and Experimental Biology, Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 64/b, Fossato di Mortara Street, Ferrara 44121, Italy
| | - Maria Rosa Iaquinta
- Laboratories of Cell Biology and Molecular Genetics, Section of Pathology, Oncology and Experimental Biology, Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 64/b, Fossato di Mortara Street, Ferrara 44121, Italy
| | - Marco Manfrini
- Laboratories of Cell Biology and Molecular Genetics, Section of Pathology, Oncology and Experimental Biology, Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 64/b, Fossato di Mortara Street, Ferrara 44121, Italy; Biostatistic Unit, GVM Care & Research, Maria Cecilia Hospital, Cotignola, Italy
| | - Fortunato Vesce
- Section of Gynecology and Obstetrics, Department of Morphology, Surgery and Experimental Medicine, School of Medicine, University of Ferrara, Ferrara, Italy
| | - Mauro Tognon
- Laboratories of Cell Biology and Molecular Genetics, Section of Pathology, Oncology and Experimental Biology, Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 64/b, Fossato di Mortara Street, Ferrara 44121, Italy.
| | - Fernanda Martini
- Laboratories of Cell Biology and Molecular Genetics, Section of Pathology, Oncology and Experimental Biology, Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 64/b, Fossato di Mortara Street, Ferrara 44121, Italy.
| |
Collapse
|
8
|
Saribas AS, Datta PK, Safak M. A comprehensive proteomics analysis of JC virus Agnoprotein-interacting proteins: Agnoprotein primarily targets the host proteins with coiled-coil motifs. Virology 2019; 540:104-118. [PMID: 31765920 DOI: 10.1016/j.virol.2019.10.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 10/13/2019] [Accepted: 10/16/2019] [Indexed: 11/29/2022]
Abstract
JC virus (JCV) Agnoprotein (Agno) plays critical roles in successful completion of the viral replication cycle. Understanding its regulatory roles requires a complete map of JCV-host protein interactions. Here, we report the first Agno interactome with host cellular targets utilizing "Two-Strep-Tag" affinity purification system coupled with mass spectroscopy (AP/MS). Proteomics data revealed that Agno primarily targets 501 cellular proteins, most of which contain "coiled-coil" motifs. Agno-host interactions occur in several cellular networks including those involved in protein synthesis and degradation; and cellular transport; and in organelles, including mitochondria, nucleus and ER-Golgi network. Among the Agno interactions, Rab11B, Importin and Crm-1 were first validated biochemically and further characterization was done for Crm-1, using a HIV-1 Rev-M10-like Agno mutant (L33D + E34L), revealing the critical roles of L33 and E34 residues in Crm-1 interaction. This comprehensive proteomics data provides new foundations to unravel the critical regulatory roles of Agno during the JCV life cycle.
Collapse
Affiliation(s)
- A Sami Saribas
- Department of Neuroscience, Laboratory of Molecular Neurovirology, Lewis Katz School of Medicine at Temple University, 3500 N. Broad Street, Philadelphia, PA, 19140, USA
| | - Prasun K Datta
- Department of Neuroscience, Laboratory of Molecular Neurovirology, Lewis Katz School of Medicine at Temple University, 3500 N. Broad Street, Philadelphia, PA, 19140, USA
| | - Mahmut Safak
- Department of Neuroscience, Laboratory of Molecular Neurovirology, Lewis Katz School of Medicine at Temple University, 3500 N. Broad Street, Philadelphia, PA, 19140, USA.
| |
Collapse
|
9
|
Del Valle L, Piña-Oviedo S. Human Polyomavirus JCPyV and Its Role in Progressive Multifocal Leukoencephalopathy and Oncogenesis. Front Oncol 2019; 9:711. [PMID: 31440465 PMCID: PMC6694743 DOI: 10.3389/fonc.2019.00711] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 07/17/2019] [Indexed: 12/12/2022] Open
Abstract
The human neurotropic virus JCPyV, a member of the Polyomaviridiae family, is the opportunistic infectious agent of Progressive Multifocal Leukoencephalopathy (PML), a fatal disease seen in severe immunosuppressive conditions and, during the last decade, in patients undergoing immunotherapy. JCPyV is a ubiquitous pathogen with up to 85% of the adult population word-wide exhibiting antibodies against it. Early experiments demonstrated that direct inoculation of JCPyV into the brain of different species resulted in the development of brain tumors and other neuroectodermal-derived neoplasias. Later, several reports showed the detection of viral sequences in medulloblastomas and glial tumors, as well as expression of the viral protein T-Antigen. Few oncogenic viruses, however, have caused so much controversy regarding their role in the pathogenesis of brain tumors, but the discovery of new Polyomaviruses that cause Merkel cell carcinomas in humans and brain tumors in racoons, in addition to the role of JCPyV in colon cancer and multiple mechanistic studies have shed much needed light on the role of JCPyV in cancer. The pathways affected by the viral protein T-Antigen include cell cycle regulators, like p53 and pRb, and transcription factors that activate pro-proliferative genes, like c-Myc. In addition, infection with JCPyV causes chromosomal damage and T-Antigen inhibits homologous recombination, and activates anti-apoptotic proteins, such as Survivin. Here we review the different aspects of the biology and physiopathology of JCPyV.
Collapse
Affiliation(s)
- Luis Del Valle
- Department of Pathology and Stanley S. Scott Cancer Center, Louisiana State University Health, New Orleans, LA, United States
| | - Sergio Piña-Oviedo
- Department of Pathology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| |
Collapse
|
10
|
Saribas AS, Coric P, Bouaziz S, Safak M. Expression of novel proteins by polyomaviruses and recent advances in the structural and functional features of agnoprotein of JC virus, BK virus, and simian virus 40. J Cell Physiol 2018; 234:8295-8315. [PMID: 30390301 DOI: 10.1002/jcp.27715] [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: 08/15/2018] [Accepted: 10/18/2018] [Indexed: 12/30/2022]
Abstract
Polyomavirus family consists of a highly diverse group of small DNA viruses. The founding family member (MPyV) was first discovered in the newborn mouse in the late 1950s, which induces solid tumors in a wide variety of tissue types that are the epithelial and mesenchymal origin. Later, other family members were also isolated from a number of mammalian, avian and fish species. Some of these viruses significantly contributed to our current understanding of the fundamentals of modern biology such as transcription, replication, splicing, RNA editing, and cell transformation. After the discovery of first two human polyomaviruses (JC virus [JCV] and BK virus [BKV]) in the early 1970s, there has been a rapid expansion in the number of human polyomaviruses in recent years due to the availability of the new technologies and brought the present number to 14. Some of the human polyomaviruses cause considerably serious human diseases, including progressive multifocal leukoencephalopathy, polyomavirus-associated nephropathy, Merkel cell carcinoma, and trichodysplasia spinulosa. Emerging evidence suggests that the expression of the polyomavirus genome is more complex than previously thought. In addition to encoding universally expressed regulatory and structural proteins (LT-Ag, Sm t-Ag, VP1, VP2, and VP3), some polyomaviruses express additional virus-specific regulatory proteins and microRNAs. This review summarizes the recent advances in polyomavirus genome expression with respect to the new viral proteins and microRNAs other than the universally expressed ones. In addition, a special emphasis is devoted to the recent structural and functional discoveries in the field of polyomavirus agnoprotein which is expressed only by JCV, BKV, and simian virus 40 genomes.
Collapse
Affiliation(s)
- A Sami Saribas
- Laboratory of Molecular Neurovirology, Department of Neuroscience, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Pascale Coric
- Laboratoire de Cristallographie et RMN Biologiques, Université Paris Descartes, Sorbonne Paris Cité, UMR 8015 CNRS, Paris, France
| | - Serge Bouaziz
- Laboratoire de Cristallographie et RMN Biologiques, Université Paris Descartes, Sorbonne Paris Cité, UMR 8015 CNRS, Paris, France
| | - Mahmut Safak
- Laboratory of Molecular Neurovirology, Department of Neuroscience, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| |
Collapse
|
11
|
Craigie M, Cicalese S, Sariyer IK. Neuroimmune Regulation of JC Virus by Intracellular and Extracellular Agnoprotein. J Neuroimmune Pharmacol 2017; 13:126-142. [PMID: 29159704 DOI: 10.1007/s11481-017-9770-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 11/10/2017] [Indexed: 12/12/2022]
Abstract
JC virus (JCV) is a human polyomavirus and the etiologic agent of the demyelinating disease progressive multifocal leukoencephalopathy (PML). PML is observed in patients with underlying immunocompromising conditions, suggesting that neuro-immune interactions between peripheral immune cells and neuro-glia play an important role in controlling viral reactivation in the brain. There is little known about the immunobiology of JCV reactivation in glial cells and the role of immune, glial, and viral players in this regulation. We have previously showed that agnoprotein, a small JCV regulatory protein, is released from infected cells and internalized by neighboring bystander cells. Here we have investigated the possible role of extracellular and intracellular agnoprotein in the neuroimmune response to JC virus. Our findings suggest that glial cells exposed to agnoprotein secrete significantly less GM-CSF, which is mediated by agnoprotein induced suppression of GM-CSF transcription. Likewise, monocytes treated with agnoprotein showed altered differentiation and maturation. In addition, monocytes and microglial cells exposed to agnoprotein showed a significant reduction in their phagocytic activities. Moreover, when an in vitro blood-brain barrier model was used, agnoprotein treatment resulted in decreased monocyte migration through the endothelial cell layer in response to activated astrocytes. All together, these results have revealed a novel immunomodulatory function of agnoprotein during JCV infection within theCNS and open a new avenue of research to better understand the mechanisms associated with JCV reactivation in patients who are at risk of developing PML.
Collapse
Affiliation(s)
- Michael Craigie
- Department of Neuroscience and Center for Neurovirology, Temple University Lewis Katz School of Medicine, 3500 N. Broad Street, Philadelphia, PA, 19140, USA
| | - Stephanie Cicalese
- Department of Neuroscience and Center for Neurovirology, Temple University Lewis Katz School of Medicine, 3500 N. Broad Street, Philadelphia, PA, 19140, USA
| | - Ilker Kudret Sariyer
- Department of Neuroscience and Center for Neurovirology, Temple University Lewis Katz School of Medicine, 3500 N. Broad Street, Philadelphia, PA, 19140, USA.
| |
Collapse
|
12
|
Saribas AS, White MK, Safak M. Structure-based release analysis of the JC virus agnoprotein regions: A role for the hydrophilic surface of the major alpha helix domain in release. J Cell Physiol 2017; 233:2343-2359. [PMID: 28722139 DOI: 10.1002/jcp.26106] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 06/15/2017] [Indexed: 12/17/2022]
Abstract
Agnoprotein (Agno) is an important regulatory protein of JC virus (JCV), BK virus (BKV) and simian virus 40 (SV40) and these viruses are unable to replicate efficiently in the absence of this protein. Recent 3D-NMR structural data revealed that Agno contains two alpha-helices (a minor and a major) while the rest of the protein adopts an unstructured conformation (Coric et al., 2017, J Cell Biochem). Previously, release of the JCV Agno from the Agno-positive cells was reported. Here, we have further mapped the regions of Agno responsible for its release by a structure-based systematic mutagenesis approach. Results revealed that amino acid residues (Lys22, Lys23, Phe31, Glu34, and Asp38) located either on or adjacent to the hydrophilic surface of the major alpha-helix domain of Agno play critical roles in release. Additionally, Agno was shown to strongly interact with unidentified components of the cell surface when cells are treated with Agno, suggesting additional novel roles for Agno during the viral infection cycle.
Collapse
Affiliation(s)
- A Sami Saribas
- Laboratory of Molecular Neurovirology, Department of Neuroscience, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Martyn K White
- Laboratory of Molecular Neurovirology, Department of Neuroscience, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Mahmut Safak
- Laboratory of Molecular Neurovirology, Department of Neuroscience, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| |
Collapse
|
13
|
Mouh FZ, Mzibri ME, Slaoui M, Amrani M. Recent Progress in Triple Negative Breast Cancer Research. Asian Pac J Cancer Prev 2017; 17:1595-608. [PMID: 27221827 DOI: 10.7314/apjcp.2016.17.4.1595] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is defined as a type of breast carcinoma that is negative for expression of oestrogene and progesterone hormone receptors (ER, PR) and HER2. This form of breast cancer is marked by its aggressiveness, low survival rate and lack of specific therapies. Recently, important molecular characteristics of TNBC have been highlighted and led to the identification of some biomarkers that could be used in diagnosis, as therapeutic targets or to assess the prognosis. In this review, we summarize recent progress in TNBC research focusing on the genetic and epigenetic alterations of TNBC and the potential use of these biomarkers in the targeted therapy for better management of TNBC.
Collapse
Affiliation(s)
- Fatima Zahra Mouh
- Equipe deRecherche ONCOGYMA, University of Mohamed V, Faculty of Medicine and Pharmacy of Rabat Morocco E-mail :
| | | | | | | |
Collapse
|
14
|
Trofe J, Gordon J, Roy-Chaudhury P, Koralnik IJ, Atwood WJ, Alloway RR, Khalili K, Woodle ES. Polyomavirus Nephropathy in Kidney Transplantation. Prog Transplant 2016; 14:130-40; quiz 141-2. [PMID: 15264457 DOI: 10.1177/152692480401400207] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Polyomavirus nephropathy has become an important complication in kidney transplantation, with a prevalence of 1% to 8%. Unfortunately, the risk factors for polyomavirus nephropathy and renal allograft loss are not well defined. The definitive diagnosis is made through assessment of a kidney transplant biopsy. Recently, noninvasive urine and serum markers have been used to assist in polyomavirus nephropathy diagnosis and monitoring. Primary treatment is immunosuppression reduction, but must be balanced with the risks of rejection. No antiviral treatments for polyomavirus nephropathy have been approved by the Food and Drug Administration. Although cidofovir has shown in vitro activity against murine polyomaviruses, and has been effective in some patients, it is associated with significant nephrotoxicity. Graft loss due to polyomavirus nephropathy should not be a contraindication to retransplantation; however, experience is limited. This review presents potential risk factors, screening, diagnostic and monitoring methods, therapeutic management, and retransplantation experience for polyomavirus nephropathy.
Collapse
Affiliation(s)
- Jennifer Trofe
- University of Cincinnati, Division of Transplantation, Ohio, USA
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Saribas AS, Coric P, Hamazaspyan A, Davis W, Axman R, White MK, Abou-Gharbia M, Childers W, Condra JH, Bouaziz S, Safak M. Emerging From the Unknown: Structural and Functional Features of Agnoprotein of Polyomaviruses. J Cell Physiol 2016; 231:2115-27. [PMID: 26831433 DOI: 10.1002/jcp.25329] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 02/01/2016] [Indexed: 12/15/2022]
Abstract
Agnoprotein is an important regulatory protein of polyomaviruses, including JCV, BKV, and SV40. In the absence of its expression, these viruses are unable to sustain their productive life cycle. It is a highly basic phosphoprotein that localizes mostly to the perinuclear area of infected cells, although a small amount of the protein is also found in nucleus. Much has been learned about the structure and function of this important regulatory protein in recent years. It forms highly stable dimers/oligomers in vitro and in vivo through its Leu/Ile/Phe-rich domain. Structural NMR studies revealed that this domain adopts an alpha-helix conformation and plays a critical role in the stability of the protein. It associates with cellular proteins, including YB-1, p53, Ku70, FEZ1, HP1α, PP2A, AP-3, PCNA, and α-SNAP; and viral proteins, including small t antigen, large T antigen, HIV-1 Tat, and JCV VP1; and significantly contributes the viral transcription and replication. This review summarizes the recent advances in the structural and functional properties of this important regulatory protein. J. Cell. Physiol. 231: 2115-2127, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- A Sami Saribas
- Department of Neuroscience, Laboratory of Molecular Neurovirology, MERB-757, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania
| | - Pascale Coric
- Université Paris Descartes, Sorbonne Paris Cité, Laboratoire de Cristallographie et RMN Biologiques, 4 av. de l'Observatoire, Paris, France
| | - Anahit Hamazaspyan
- Department of Neuroscience, Laboratory of Molecular Neurovirology, MERB-757, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania
| | - William Davis
- Department of Neuroscience, Laboratory of Molecular Neurovirology, MERB-757, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania
| | - Rachel Axman
- Department of Neuroscience, Laboratory of Molecular Neurovirology, MERB-757, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania
| | - Martyn K White
- Department of Neuroscience, Laboratory of Molecular Neurovirology, MERB-757, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania
| | - Magid Abou-Gharbia
- Moulder Center for Drug Discovery Research, Temple University School of Pharmacy, Philadelphia, Pennsylvania
| | - Wayne Childers
- Moulder Center for Drug Discovery Research, Temple University School of Pharmacy, Philadelphia, Pennsylvania
| | - Jon H Condra
- Moulder Center for Drug Discovery Research, Temple University School of Pharmacy, Philadelphia, Pennsylvania
| | - Serge Bouaziz
- Université Paris Descartes, Sorbonne Paris Cité, Laboratoire de Cristallographie et RMN Biologiques, 4 av. de l'Observatoire, Paris, France
| | - Mahmut Safak
- Department of Neuroscience, Laboratory of Molecular Neurovirology, MERB-757, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania
| |
Collapse
|
16
|
Sinagra E, Raimondo D, Gallo E, Stella M, Cottone M, Orlando A, Rossi F, Orlando E, Messina M, Tomasello G, Lo Monte AI, La Rocca E, Rizzo AG. Could JC virus provoke metastasis in colon cancer? World J Gastroenterol 2014; 20:15745-15749. [PMID: 25400458 PMCID: PMC4229539 DOI: 10.3748/wjg.v20.i42.15745] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Revised: 05/05/2014] [Accepted: 05/19/2014] [Indexed: 02/06/2023] Open
Abstract
AIM To evaluate the prevalence of John Cunningham virus (JC virus) in a small cohort of patients with colon cancer and to assess its presence in hepatic metastasis. METHODS Nineteen consecutive patients with histologically diagnosed colon cancer were included in our study, together with ten subjects affected by histologically and serologically diagnosed hepatitis C virus infection. In the patients included in the colon cancer group, JC virus was searched for in the surgical specimen; in the control group, JC virus was searched for in the hepatic biopsy. The difference in the prevalence of JC virus in the hepatic biopsy between the two groups was assessed through the χ(2) test. RESULTS Four out of 19 patients with colon cancer had a positive polymerase chain reaction (PCR) test for JC virus, and four had liver metastasis. Among the patients with liver metastasis, three out of four had a positive PCR test for JC virus in the surgical specimen and in the liver biopsy; the only patient with liver metastasis with a negative test for JC virus also presented a negative test for JC virus in the surgical specimen. In the control group of patients with hepatitis C infection, none of the ten patients presented JC virus infection in the hepatic biopsy. The difference between the two groups regarding JC virus infection was statistically significant (χ(2) = 9.55, P = 0.002). CONCLUSION JC virus may play a broader role than previously thought, and may be mechanistically involved in the late stages of these tumors.
Collapse
|
17
|
White MK, Kaminski R, Khalili K, Wollebo HS. Rad51 activates polyomavirus JC early transcription. PLoS One 2014; 9:e110122. [PMID: 25310191 PMCID: PMC4195707 DOI: 10.1371/journal.pone.0110122] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Accepted: 09/15/2014] [Indexed: 11/18/2022] Open
Abstract
The human neurotropic polyomavirus JC (JCV) causes the fatal CNS demyelinating disease progressive multifocal leukoencephalopathy (PML). JCV infection is very common and after primary infection, the virus is able to persist in an asymptomatic state. Rarely, and usually only under conditions of immune impairment, JCV re-emerges to actively replicate in the astrocytes and oligodendrocytes of the brain causing PML. The regulatory events involved in the reactivation of active viral replication in PML are not well understood but previous studies have implicated the transcription factor NF-κB acting at a well-characterized site in the JCV noncoding control region (NCCR). NF-κB in turn is regulated in a number of ways including activation by cytokines such as TNF-α, interactions with other transcription factors and epigenetic events involving protein acetylation – all of which can regulate the transcriptional activity of JCV. Active JCV infection is marked by the occurrence of rapid and extensive DNA damage in the host cell and the induction of the expression of cellular proteins involved in DNA repair including Rad51, a major component of the homologous recombination-directed double-strand break DNA repair machinery. Here we show that increased Rad51 expression activates the JCV early promoter. This activation is co-operative with the stimulation caused by NF-κB p65, abrogated by mutation of the NF-κB binding site or siRNA to NFκB p65 and enhanced by the histone deacetylase inhibitor sodium butyrate. These data indicate that the induction of Rad51 resulting from infection with JCV acts through NF-κB via its binding site to stimulate JCV early transcription. We suggest that this provides a novel positive feedback mechanism to enhance viral gene expression during the early stage of JCV infection.
Collapse
Affiliation(s)
- Martyn K. White
- Center for Neurovirology, Department of Neuroscience, Temple University School of Medicine, Philadelphia, Pennsylvania, United States of America
- * E-mail: (MKW); (KK)
| | - Rafal Kaminski
- Center for Neurovirology, Department of Neuroscience, Temple University School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Kamel Khalili
- Center for Neurovirology, Department of Neuroscience, Temple University School of Medicine, Philadelphia, Pennsylvania, United States of America
- * E-mail: (MKW); (KK)
| | - Hassen S. Wollebo
- Center for Neurovirology, Department of Neuroscience, Temple University School of Medicine, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
18
|
Activation of c-Myc and Cyclin D1 by JCV T-Antigen and β-catenin in colon cancer. PLoS One 2014; 9:e106257. [PMID: 25229241 PMCID: PMC4167695 DOI: 10.1371/journal.pone.0106257] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2014] [Accepted: 07/30/2014] [Indexed: 12/17/2022] Open
Abstract
During the last decade, mounting evidence has implicated the human neurotropic virus JC virus in the pathology of colon cancer. However, the mechanisms of JC virus-mediated oncogenesis are still not fully determined. One candidate to mediate these effects is the viral early transcriptional product T-Antigen, which has the ability to inactivate cell cycle regulatory proteins such as p53. In medulloblastomas, T-Antigen has been shown to bind the Wnt signaling pathway protein β-catenin; however, the effects of this interaction on downstream cell cycle regulatory proteins remain unknown. In light of these observations, we investigated the association of T-Antigen and nuclear β-catenin in colon cancer cases and the effects of this complex in the activation of the transcription and cell cycle regulators c-Myc and Cyclin D1 in vitro. Gene amplification demonstrated the presence of viral sequences in 82.4% of cases and we detected expression of T-Antigen in 64.6% of cases by immunohistochemistry. Further, we found that T-Antigen and β-catenin co-localized in the nuclei of tumor cells and we confirmed the physical binding between these two proteins in vitro. The nuclear presence of T-Antigen and β-catenin resulted in the significant enhancement of TCF-dependent promoter activity and activation of the β-catenin downstream targets, c-Myc and Cyclin D1. These observations provide further evidence for a role of JCV T-Antigen in the dysregulation of the Wnt signaling pathway and in the pathogenesis of colon cancer.
Collapse
|
19
|
Nuclear magnetic resonance structure revealed that the human polyomavirus JC virus agnoprotein contains an α-helix encompassing the Leu/Ile/Phe-rich domain. J Virol 2014; 88:6556-75. [PMID: 24672035 DOI: 10.1128/jvi.00146-14] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
UNLABELLED Agnoprotein is a small multifunctional regulatory protein required for sustaining the productive replication of JC virus (JCV). It is a mostly cytoplasmic protein localizing in the perinuclear area and forms highly stable dimers/oligomers through a Leu/Ile/Phe-rich domain. There have been no three-dimensional structural data available for agnoprotein due to difficulties associated with the dynamic conversion from monomers to oligomers. Here, we report the first nuclear magnetic resonance (NMR) structure of a synthetic agnoprotein peptide spanning amino acids Thr17 to Glu55 where Lys23 to Phe39 encompassing the Leu/Ile/Phe-rich domain forms an amphipathic α-helix. On the basis of these structural data, a number of Ala substitution mutations were made to investigate the role of the α-helix in the structure and function of agnoprotein. Single L29A and L36A mutations exhibited a significant negative effect on both protein stability and viral replication, whereas the L32A mutation did not. In addition, the L29A mutant displayed a highly nuclear localization pattern, in contrast to the pattern for the wild type (WT). Interestingly, a triple mutant, the L29A+L32A+L36A mutant, yielded no detectable agnoprotein expression, and the replication of this JCV mutant was significantly reduced, suggesting that Leu29 and Leu36 are located at the dimer interface, contributing to the structure and stability of agnoprotein. Two other single mutations, L33A and E34A, did not perturb agnoprotein stability as drastically as that observed with the L29A and L36A mutations, but they negatively affected viral replication, suggesting that the role of these residues is functional rather than structural. Thus, the agnoprotein dimerization domain can be targeted for the development of novel drugs active against JCV infection. IMPORTANCE Agnoprotein is a small regulatory protein of JC virus (JCV) and is required for the successful completion of the viral replication cycle. It forms highly stable dimers and oligomers through its hydrophobic (Leu/Ile/Phe-rich) domain, which has been shown to play essential roles in the stability and function of the protein. In this work, the Leu/Ile/Phe-rich domain has been further characterized by NMR studies using an agnoprotein peptide spanning amino acids T17 to Q54. Those studies revealed that the dimerization domain of the protein forms an amphipathic α-helix. Subsequent NMR structure-based mutational analysis of the region highlighted the critical importance of certain amino acids within the α-helix for the stability and function of agnoprotein. In conclusion, this study provides a solid foundation for developing effective therapeutic approaches against the dimerization domain of the protein to inhibit its critical roles in JCV infection.
Collapse
|
20
|
Saribas AS, Mun S, Johnson J, El-Hajmoussa M, White MK, Safak M. Human polyoma JC virus minor capsid proteins, VP2 and VP3, enhance large T antigen binding to the origin of viral DNA replication: evidence for their involvement in regulation of the viral DNA replication. Virology 2013; 449:1-16. [PMID: 24418532 DOI: 10.1016/j.virol.2013.10.031] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2013] [Revised: 07/08/2013] [Accepted: 10/22/2013] [Indexed: 10/26/2022]
Abstract
JC virus (JCV) lytically infects the oligodendrocytes in the central nervous system in a subset of immunocompromized patients and causes the demyelinating disease, progressive multifocal leukoencephalopathy. JCV replicates and assembles into infectious virions in the nucleus. However, understanding the molecular mechanisms of its virion biogenesis remains elusive. In this report, we have attempted to shed more light on this process by investigating molecular interactions between large T antigen (LT-Ag), Hsp70 and minor capsid proteins, VP2/VP3. We demonstrated that Hsp70 interacts with VP2/VP3 and LT-Ag; and accumulates heavily in the nucleus of the infected cells. We also showed that VP2/VP3 associates with LT-Ag through their DNA binding domains resulting in enhancement in LT-Ag DNA binding to Ori and induction in viral DNA replication. Altogether, our results suggest that VP2/VP3 and Hsp70 actively participate in JCV DNA replication and may play critical roles in coupling of viral DNA replication to virion encapsidation.
Collapse
Affiliation(s)
- A Sami Saribas
- Department of Neuroscience, Laboratory of Molecular Neurovirology, MERB-757, Temple University School of Medicine, 3500 N. Broad Street, Philadelphia, PA 19140, United States
| | - Sarah Mun
- Department of Neuroscience, Laboratory of Molecular Neurovirology, MERB-757, Temple University School of Medicine, 3500 N. Broad Street, Philadelphia, PA 19140, United States
| | - Jaslyn Johnson
- Department of Neuroscience, Laboratory of Molecular Neurovirology, MERB-757, Temple University School of Medicine, 3500 N. Broad Street, Philadelphia, PA 19140, United States
| | - Mohammad El-Hajmoussa
- Department of Neuroscience, Laboratory of Molecular Neurovirology, MERB-757, Temple University School of Medicine, 3500 N. Broad Street, Philadelphia, PA 19140, United States
| | - Martyn K White
- Department of Neuroscience, Laboratory of Molecular Neurovirology, MERB-757, Temple University School of Medicine, 3500 N. Broad Street, Philadelphia, PA 19140, United States
| | - Mahmut Safak
- Department of Neuroscience, Laboratory of Molecular Neurovirology, MERB-757, Temple University School of Medicine, 3500 N. Broad Street, Philadelphia, PA 19140, United States.
| |
Collapse
|
21
|
Wilk A, Waligórski P, Lassak A, Vashistha H, Lirette D, Tate D, Zea AH, Koochekpour S, Rodriguez P, Meggs LG, Estrada JJ, Ochoa A, Reiss K. Polycyclic aromatic hydrocarbons-induced ROS accumulation enhances mutagenic potential of T-antigen from human polyomavirus JC. J Cell Physiol 2013; 228:2127-38. [PMID: 23558788 DOI: 10.1002/jcp.24375] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2013] [Accepted: 03/28/2013] [Indexed: 01/28/2023]
Abstract
Polycyclic aromatic hydrocarbons (PAHs) are the products of incomplete combustion of organic materials, which are present in cigarette smoke, deep-fried food, and in natural crude oil. Since PAH-metabolites form DNA adducts and cause oxidative DNA damage, we asked if these environmental carcinogens could affect transforming potential of the human Polyomavirus JC oncoprotein, T-antigen (JCV T-antigen). We extracted DMSO soluble PAHs from Deepwater Horizon oil spill in the Gulf of Mexico (oil-PAHs), and detected several carcinogenic PAHs. The oil-PAHs were tested in exponentially growing cultures of normal mouse fibroblasts (R508), and in R508 stably expressing JCV T-antigen (R508/T). The oil-PAHs were cytotoxic only at relatively high doses (1:50-1:100 dilution), and at 1:500 dilution the growth and cell survival rates were practically unaffected. This non-toxic dose triggered however, a significant accumulation of reactive oxygen species (ROS), caused oxidative DNA damage and the formation of DNA double strand breaks (DSBs). Although oil-PAHs induced similar levels of DNA damage in R508 and R508/T cells, only T-antigen expressing cells demonstrated inhibition of high fidelity DNA repair by homologous recombination (HRR). In contrast, low-fidelity repair by non-homologous end joining (NHEJ) was unaffected. This potential mutagenic shift between DNA repair mechanisms was accompanied by a significant increase in clonal growth of R508/T cells chronically exposed to low doses of the oil-PAHs. Our results indicate for the first time carcinogenic synergy in which oil-PAHs trigger oxidative DNA damage and JCV T-antigen compromises DNA repair fidelity.
Collapse
Affiliation(s)
- Anna Wilk
- Neurological Cancer Research at Stanley S Scott Cancer Center, New Orleans, Louisiana, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Giannitti F, Higgins RJ, Pesavento PA, Cruz FD, Clifford DL, Piazza M, Struckhoff AP, Valle LD, Bollen AW, Puschner B, Kerr E, Gelberg H, Mete A, McGraw S, Woods LW. Temporal and Geographic Clustering of Polyomavirus-Associated Olfactory Tumors in 10 Free-Ranging Raccoons (Procyon lotor). Vet Pathol 2013; 51:832-45. [DOI: 10.1177/0300985813502817] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Reports of primary nervous system tumors in wild raccoons are extremely rare. Olfactory tumors were diagnosed postmortem in 9 free-ranging raccoons from 4 contiguous counties in California and 1 raccoon from Oregon within a 26-month period between 2010 and 2012. We describe the geographic and temporal features of these 10 cases, including the laboratory diagnostic investigations and the neuropathologic, immunohistochemical, and ultrastructural characteristics of these tumors in the affected animals. All 9 raccoons from California were found within a localized geographic region of the San Francisco Bay Area (within a 44.13-km radius). The tight temporal and geographic clustering and consistent anatomic location in the olfactory system of tumor types not previously described in raccoons (malignant peripheral nerve sheath tumors and undifferentiated sarcomas) strongly suggest either a common cause or a precipitating factor leading to induction or potentiation of neuro-oncogenesis and so prompted an extensive diagnostic investigation to explore possible oncogenic infectious and/or toxic causes. By a consensus polymerase chain reaction strategy, a novel, recently reported polyomavirus called raccoon polyomavirus was identified in all 10 tumors but not in the normal brain tissue from the affected animals, suggesting that the virus might play a role in neuro-oncogenesis. In addition, expression of the viral protein T antigen was detected in all tumors containing the viral sequences. We discuss the potential role of raccoon polyomavirus as an oncogenic virus.
Collapse
Affiliation(s)
- F. Giannitti
- California Animal Health and Food Safety Laboratory, School of Veterinary Medicine, University of California, Davis, CA, USA
| | - R. J. Higgins
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California, Davis, CA, USA
| | - P. A. Pesavento
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California, Davis, CA, USA
| | - F. Dela Cruz
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California, Davis, CA, USA
| | - D. L. Clifford
- Wildlife Investigations Laboratory, California Department of Fish and Wildlife, Rancho Cordova, CA, USA
| | | | - A. Parker Struckhoff
- Departments of Medicine and Pathology, Stanley S. Scott Cancer Center, School of Medicine, Louisiana State University, New Orleans, LA, USA
| | - L. Del Valle
- Departments of Medicine and Pathology, Stanley S. Scott Cancer Center, School of Medicine, Louisiana State University, New Orleans, LA, USA
| | - A. W. Bollen
- School of Medicine, University of California, San Francisco, CA, USA
| | - B. Puschner
- California Animal Health and Food Safety Laboratory, School of Veterinary Medicine, University of California, Davis, CA, USA
| | - E. Kerr
- Natural Resources DNA Profiling and Forensic Centre, DNA Building, Trent University, Peterborough, Ontario, Canada
| | - H. Gelberg
- Department of Biomedical Sciences and the Veterinary Diagnostic Laboratory, College of Veterinary Medicine, Oregon State University, Corvallis, OR, USA
| | - A. Mete
- California Animal Health and Food Safety Laboratory, School of Veterinary Medicine, University of California, Davis, CA, USA
| | - S. McGraw
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California, Davis, CA, USA
| | - L. W. Woods
- California Animal Health and Food Safety Laboratory, School of Veterinary Medicine, University of California, Davis, CA, USA
| |
Collapse
|
23
|
Sami Saribas A, Abou-Gharbia M, Childers W, Sariyer IK, White MK, Safak M. Essential roles of Leu/Ile/Phe-rich domain of JC virus agnoprotein in dimer/oligomer formation, protein stability and splicing of viral transcripts. Virology 2013; 443:161-76. [PMID: 23747198 DOI: 10.1016/j.virol.2013.05.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2013] [Revised: 03/21/2013] [Accepted: 05/03/2013] [Indexed: 10/26/2022]
Abstract
Agnoprotein is one of the key regulatory proteins of polyomaviruses, including JCV, BKV and SV40 and is required for a productive viral life cycle. We have recently reported that agnoprotein forms stable dimer/oligomers mediated by a predicted amphipathic α-helix, spanning amino acids (aa), 17 to 42. Deletion of the α-helix renders a replication incompetent virus. Here, we have further characterized this region by a systematic deletion and substitution mutagenesis and demonstrated that a Leu/Ile/Phe-rich domain, (spanning aa 28-39) within α-helix is indispensable for agnoprotein structure and function. Deletion of aa 30-37 severely affects the dimer/oligomer formation and stable expression of the protein. Mutagenesis data also indicate that the residues, 34-36, may be involved in regulation of the splicing events of JCV transcripts. Collectively, these data suggest that the Leu/Ile/Phe-rich domain plays critical roles in agnoprotein function and thus represents a potential target for developing novel therapeutics against JCV infections.
Collapse
Affiliation(s)
- A Sami Saribas
- Department of Neuroscience, Laboratory of Molecular Neurovirology, MERB-757, Temple University School of Medicine, 3500 N. Broad Street, Philadelphia, PA 19140, United States
| | | | | | | | | | | |
Collapse
|
24
|
Matalka I, Swedan S, Khabaz MN, Barahmeh M. JC virus in colorectal cancer: where do we stand? Future Virol 2013. [DOI: 10.2217/fvl.13.36] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
AIm: Colorectal cancer (CRC) is a major cause of morbidity and mortality worldwide. In 2008, CRC was ranked as the second most common cancer among all newly diagnosed cancers in Jordan. It was recently suggested that the JC virus (JCV) could be linked to the development of CRC. However, this topic remains highly controversial. JCV is a common human polyomavirus that first infects individuals during early childhood. Infection with JCV can occur due to intake of contaminated water and food. The vast majority of those infected have no symptoms; however, severe immunosuppression can trigger reactivation of the virus. The oncogenicity of JCV was suggested to be related to its large T-antigen (T-Ag). This study aimed to investigate the incidence of JCV in CRC tissue of patients from northern Jordan. Materials & methods: Paraffin-embedded tissues from cases previously diagnosed with CRC were used. Immunohistochemistry and PCR techniques were used to detect the presence of JCV. Results: In total, 15.6% of CRC samples were positive for JCV T-Ag protein, whereas none of the noncancer control samples were positive. In total, 43.5% of CRC samples contained the JCV T-Ag gene compared with 16.7% within control samples. All CRC samples expressing the JCV T-Ag protein harbored the JCV T-Ag gene. However, not all samples that harbored the T-Ag gene expressed the T-Ag protein. Conclusion: Our results are consistent with recently published data and raise the possibility of an association between JCV infection and CRC. These findings deserve further investigation in larger-scale studies in order to elucidate the possible molecular carcinogenesis pathways in JCV-infected colorectal neoplasms.
Collapse
Affiliation(s)
- Ismail Matalka
- Department of Pathology & Laboratory Medicine, School of Medicine, Jordan University of Science & Technology, Irbid 22110, PO Box 3030, Jordan
| | - Samer Swedan
- Department of Medical Laboratory Sciences, Faculty of Allied Medical Sciences, Jordan University of Science & Technology, Irbid 22110, PO Box 3030, Jordan
| | - Mohamad Nidal Khabaz
- Department of Pathology, King Abdulaziz University, Jeddah 21589, PO Box 80200, Kingdom of Saudi Arabia
| | - Myassar Barahmeh
- Department of Medical Laboratory Sciences, Faculty of Allied Medical Sciences, Jordan University of Science & Technology, Irbid 22110, PO Box 3030, Jordan
| |
Collapse
|
25
|
Molecular biology, epidemiology, and pathogenesis of progressive multifocal leukoencephalopathy, the JC virus-induced demyelinating disease of the human brain. Clin Microbiol Rev 2012; 25:471-506. [PMID: 22763635 DOI: 10.1128/cmr.05031-11] [Citation(s) in RCA: 299] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Progressive multifocal leukoencephalopathy (PML) is a debilitating and frequently fatal central nervous system (CNS) demyelinating disease caused by JC virus (JCV), for which there is currently no effective treatment. Lytic infection of oligodendrocytes in the brain leads to their eventual destruction and progressive demyelination, resulting in multiple foci of lesions in the white matter of the brain. Before the mid-1980s, PML was a relatively rare disease, reported to occur primarily in those with underlying neoplastic conditions affecting immune function and, more rarely, in allograft recipients receiving immunosuppressive drugs. However, with the onset of the AIDS pandemic, the incidence of PML has increased dramatically. Approximately 3 to 5% of HIV-infected individuals will develop PML, which is classified as an AIDS-defining illness. In addition, the recent advent of humanized monoclonal antibody therapy for the treatment of autoimmune inflammatory diseases such as multiple sclerosis (MS) and Crohn's disease has also led to an increased risk of PML as a side effect of immunotherapy. Thus, the study of JCV and the elucidation of the underlying causes of PML are important and active areas of research that may lead to new insights into immune function and host antiviral defense, as well as to potential new therapies.
Collapse
|
26
|
JC virus agnoprotein enhances large T antigen binding to the origin of viral DNA replication: evidence for its involvement in viral DNA replication. Virology 2012; 433:12-26. [PMID: 22840425 DOI: 10.1016/j.virol.2012.06.017] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2012] [Revised: 05/25/2012] [Accepted: 06/11/2012] [Indexed: 10/28/2022]
Abstract
Agnoprotein is required for the successful completion of the JC virus (JCV) life cycle and was previously shown to interact with JCV large T-antigen (LT-Ag). Here, we further characterized agnoprotein's involvement in viral DNA replication. Agnoprotein enhances the DNA binding activity of LT-Ag to the viral origin (Ori) without directly interacting with DNA. The predicted amphipathic α-helix of agnoprotein plays a major role in this enhancement. All three phenylalanine (Phe) residues of agnoprotein localize to this α-helix and Phe residues in general are known to play critical roles in protein-protein interaction, protein folding and stability. The functional relevance of all Phe residues was investigated by mutagenesis. When all were mutated to alanine (Ala), the mutant virus (F31AF35AF39A) replicated significantly less efficiently than each individual Phe mutant virus alone, indicating the importance of Phe residues for agnoprotein function. Collectively, these studies indicate a close involvement of agnoprotein in viral DNA replication.
Collapse
|
27
|
Wollebo HS, Melis S, Khalili K, Safak M, White MK. Cooperative roles of NF-κB and NFAT4 in polyomavirus JC regulation at the KB control element. Virology 2012; 432:146-54. [PMID: 22749879 DOI: 10.1016/j.virol.2012.06.010] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2011] [Revised: 03/15/2012] [Accepted: 06/05/2012] [Indexed: 11/29/2022]
Abstract
The human polyomavirus JC (JCV) is the causative agent of the CNS demyelinating disease progressive multifocal leukoencephalopathy (PML). Infection by JCV is extremely common and after primary infection, JCV persists in a latent state. However, PML is a very rare disease suggesting that the virus is tightly regulated. Previously, we showed that NF-κB and C/EBPβ regulate the JCV early and late promoters via a DNA control element, KB, which also mediates the stimulatory effects of proinflammatory cytokines such as TNF-α on JCV gene expression. Other studies have implicated NFAT4 in JCV regulation. We now report that NFAT4 and NF-κB interact at the KB element to co-operatively activate both JCV early and late transcription and viral DNA replication. This interplay is inhibited by C/EBPβ and by agents that block the calcineurin/NFAT signaling pathway. The importance of these events in the regulation of JCV latency and reactivation is discussed.
Collapse
Affiliation(s)
- Hassen S Wollebo
- Department of Neuroscience, Center for Neurovirology, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | | | | | | | | |
Collapse
|
28
|
Hachana M, Amara K, Ziadi S, Gacem RB, Korbi S, Trimeche M. Investigation of human JC and BK polyomaviruses in breast carcinomas. Breast Cancer Res Treat 2012; 133:969-77. [PMID: 22108781 DOI: 10.1007/s10549-011-1876-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2011] [Accepted: 11/04/2011] [Indexed: 11/30/2022]
Abstract
We have previously showed the presence of the simian virus 40 (SV40) and the mouse mammary tumor virus (MMTV)-like in a significant proportions of Tunisian breast carcinomas. However, to date there are no published studies concerning evaluation of the possible implication of the human polyomaviruses JC (JCV) and BK (BKV) in breast carcinomas. The presence of JCV and BKV DNA was investigated by PCR in a 123 primary breast carcinomas and matched adjacent non-tumor breast tissues. The results were correlated to clinicopathological and virological parameters. JCV T-antigen DNA was detected in 23% of breast carcinoma cases; however, all cases were negative for BKV. JCV T antigen PCR products were further confirmed as authentic JCV genome by direct sequencing. JCV was found in invasive ductal carcinomas (28/112 cases) but not in invasive lobular carcinomas (0/5) or medullary carcinomas (0/6). JCV DNA presence correlates inversely with the expression of estrogen (P = 0.022) and progesterone (P = 0.008) receptors. JCV DNA presence correlates also with "triple negative" phenotype (P = 0.021). With regard to virological data, a trend toward an inverse correlation was noted between the presence of JCV and SV40 (P = 0.06). Moreover, significant correlation was found between multiple viral infection (JCV, and/or SV40, and/or MMTV-like in the same tumor) and "triple negative" phenotype (P = 0.001) and also with p53 accumulation (P = 0.028). To the best of our knowledge, this is the first study demonstrating the presence of JCV in a subset of breast carcinomas. Also our results suggest that "triple negative" breast carcinomas are viral-related tumors.
Collapse
Affiliation(s)
- Mohamed Hachana
- Department of Pathology, Farhat Hached Hospital, 4000 Sousse, Tunisia
| | | | | | | | | | | |
Collapse
|
29
|
Morgenstern DA, Anderson J. Inflammation: what role in pediatric cancer? Pediatr Blood Cancer 2012; 58:659-64. [PMID: 22162439 DOI: 10.1002/pbc.24008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2011] [Accepted: 10/24/2011] [Indexed: 01/22/2023]
Abstract
There is growing evidence for the importance of chronic inflammation in the pathogenesis of adult cancers and for an ongoing role of the inflammatory response in tumor growth and metastasis. Here, we examine how these processes relate to pediatric malignancies. While it is unlikely that chronic inflammation plays a significant role in driving malignant progression in childhood tumors that typically have developmental origins, the inflammatory response does appear to play an important role in the development and progression of many types of childhood cancer. An enhanced understanding of these processes will be of critical importance in developing novel therapeutic strategies.
Collapse
Affiliation(s)
- Daniel A Morgenstern
- Molecular Haematology and Cancer Biology Unit, Institute of Child Health, London, UK.
| | | |
Collapse
|
30
|
Dang X, Wüthrich C, Gordon J, Sawa H, Koralnik IJ. JC virus encephalopathy is associated with a novel agnoprotein-deletion JCV variant. PLoS One 2012; 7:e35793. [PMID: 22536439 PMCID: PMC3334910 DOI: 10.1371/journal.pone.0035793] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2012] [Accepted: 03/22/2012] [Indexed: 11/19/2022] Open
Abstract
JC virus encephalopathy (JCVE) is a newly described gray matter disease of the brain caused by productive infection of cortical pyramidal neurons. We characterized the full length sequence of JCV isolated from the brain of a JCVE patient, analyzed its distribution in various compartments by PCR, and determined viral gene expression in the brain by immunohistochemistry(IHC). We identified a novel JCV variant, JCVCPN1, with a unique 143 bp deletion in the Agno gene encoding a truncated 10 amino acid peptide, and harboring an archetype-like regulatory region. This variant lacked one of three nuclear protein binding regions in the Agno gene. It was predominant in the brain, where it coexisted with an Agno-intact wild-type strain. Double immunostaining with anti-Agno and anti- VP1 antibodies demonstrated that the truncated JCVCPN1 Agno peptide was present in the majority of cortical cells productively infected with JCV. We then screened 68 DNA samples from 8 brain, 30 CSF and 30 PBMC samples of PML patients, HIV+ and HIV- control subjects. Another JCVCPN strain with a different pattern of Agno-deletion was found in the CSF of an HIV+/PML patient, where it also coexisted with wild-type, Agno-intact JCV. These findings suggest that the novel tropism for cortical pyramidal neurons of JCVCPN1, may be associated with the Agno deletion. Productive and lytic infection of these cells, resulting in fulminant JCV encephalopathy and death may have been facilitated by the co-infection with a wild- type strain of JCV.
Collapse
Affiliation(s)
- Xin Dang
- Division of Neurovirology, Department of Neurology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States of America
- Division of Viral Pathogenesis, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States of America
| | - Christian Wüthrich
- Division of Neurovirology, Department of Neurology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States of America
- Division of Viral Pathogenesis, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States of America
| | - Jennifer Gordon
- Department of Neuroscience and Center for Neurovirology, Temple University School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Hirofumi Sawa
- Department of Molecular Pathobiology, Research Center for Zoonosis Control, Global COE Program for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Igor J. Koralnik
- Division of Neurovirology, Department of Neurology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States of America
- Division of Viral Pathogenesis, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
31
|
Human polyomavirus JC small regulatory agnoprotein forms highly stable dimers and oligomers: implications for their roles in agnoprotein function. Virology 2011; 420:51-65. [PMID: 21920573 DOI: 10.1016/j.virol.2011.08.015] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2011] [Revised: 08/09/2011] [Accepted: 08/19/2011] [Indexed: 11/22/2022]
Abstract
JC virus (JCV) encodes a small basic phosphoprotein from the late coding region called agnoprotein, which has been shown to play important regulatory roles in the viral replication cycle. In this study, we report that agnoprotein forms highly stable dimers and higher order oligomer complexes. This was confirmed by immunoblotting and mass spectrometry studies. These complexes are extremely resistant to strong denaturing agents, including urea and SDS. Central portion of the protein, amino acids spanning from 17 to 42 is important for dimer/oligomer formation. Removal of 17 to 42 aa region from the viral background severely affected the efficiency of the JCV replication. Extracts prepared from JCV-infected cells showed a double banding pattern for agnoprotein in vivo. Collectively, these findings suggest that agnoprotein forms functionally active homodimer/oligomer complexes and these may be important for its function during viral propagation and thus for the progression of PML.
Collapse
|
32
|
Wollebo HS, Safak M, Valle LD, Khalili K, White MK. Role for tumor necrosis factor-α in JC virus reactivation and progressive multifocal leukoencephalopathy. J Neuroimmunol 2011; 233:46-53. [PMID: 21185609 PMCID: PMC3074035 DOI: 10.1016/j.jneuroim.2010.11.013] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2010] [Revised: 11/05/2010] [Accepted: 11/26/2010] [Indexed: 11/15/2022]
Abstract
JCV causes the CNS demyelinating disease progressive multifocal leukoencephalopathy (PML). After primary infection, JCV persists in a latent state, where viral protein expression and replication are not detectable. NF-κB and C/EBPβ regulate the JCV promoter via a control element, κB, suggesting proinflammatory cytokines may reactivate JCV to cause PML, e.g., in HIV-1/AIDS. Since HIV-1 induces cytokines in brain, including TNF-α, we examined a role for TNF-α in JCV regulation. TNF-α stimulated both early and late JCV transcription. Further, the κB element conferred TNF-α response to a heterologous promoter. Immunohistochemistry of HIV+/PML revealed robust labeling for TNF-α and TNFR-1. These data suggest TNF-α stimulation of κB may contribute to JCV reactivation in HIV+/PML.
Collapse
Affiliation(s)
- Hassen S. Wollebo
- Center for Neurovirology, Department of Neuroscience, Temple University School of Medicine, 3500 N. Broad Street, Philadelphia, PA 19140, USA
| | - Mahmut Safak
- Center for Neurovirology, Department of Neuroscience, Temple University School of Medicine, 3500 N. Broad Street, Philadelphia, PA 19140, USA
| | | | - Kamel Khalili
- Center for Neurovirology, Department of Neuroscience, Temple University School of Medicine, 3500 N. Broad Street, Philadelphia, PA 19140, USA
| | - Martyn K. White
- Center for Neurovirology, Department of Neuroscience, Temple University School of Medicine, 3500 N. Broad Street, Philadelphia, PA 19140, USA
| |
Collapse
|
33
|
Zhang B, Izadjoo M, Horkayne-Szakaly I, Morrison A, Wear DJ. Medulloblastoma and Brucellosis - Molecular Evidence of Brucella sp in Association with Central Nervous System Cancer. J Cancer 2011; 2:136-41. [PMID: 21475718 PMCID: PMC3053536 DOI: 10.7150/jca.2.136] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2011] [Accepted: 03/08/2011] [Indexed: 11/23/2022] Open
Abstract
Neurobrucellosis has been reported to cause lesions in a number of different locations in the central nervous system. Histologically or radiologically, these lesions were consistent with an infection. In response to parents who believed their child's brain tumor, histologically typical of medulloblastoma, was in reality neurobrucellosis, formalin-fixed paraffin-embedded tumor tissue from the medulloblastoma was sectioned, DNA extracted, and tested by polymerase chain reaction (PCR). Specific primer/probe sets, designed in our laboratory to target Brucella species, B. melitensis, B. abortus and B. suis, and designated OMP31, B-m, B-a and B-s, respectively, were used in TaqMan real-time PCR to amplify those gene targets in two separate blocks of the child's tumor. Sections from two blocks were positive only for Brucella species. Although the patient grew up in a European country known to harbor brucella in foods, such as unpasturized milk and cheese, the patient was seronegative for B. mellitensis, B. suis, and B. abortus. In an effort to test whether a relationship existed between the presence of brucella and medulloblastoma, 20 medulloblastomas were retrieved from the tissue repository of the AFIP. The above four primer/probe sets were again used to amplify brucella DNA. Five of 20 tumors (25%) contained Brucella species DNA by the OMP31 primer/probe set. None of the 20 medulloblastomas had specific sequences for B. mellitensis, B. suis, or B. abortus. Is chronic brucellosis similar to other infectious agents such as helicobacter that is associated with tumor formation?
Collapse
Affiliation(s)
- Binxue Zhang
- 1. Division of Wound and Translational Research, Department of Environmental and Infectious Disease Sciences, Armed Forces Institute of Pathology, Washington, DC 20306-6000, USA
| | | | | | | | | |
Collapse
|
34
|
Primaquine-NSAID twin drugs: Synthesis, radical scavenging, antioxidant and Fe2+ chelating activity. ACTA PHARMACEUTICA (ZAGREB, CROATIA) 2010; 60:325-37. [PMID: 21134866 DOI: 10.2478/v10007-010-0024-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Novel primaquine conjugates with non-steroidal anti-inlammatory drugs (PQ-NSAIDs, 4a-h) were prepared, fully chemically characterized and screened for radical scavenging and antioxidant activities. The synthetic procedure leading to twin drugs 4a-h involved two steps: i) preparation of NSAID benzotriazolides 3a-h from the corresponding NSAID (ibuprofen, ketoprofen, fenoprofen, ketoprofen hydroxy and methylene analogues, diclofenac or indomethacin) and benzotriazole carboxylic acid chloride (BtCOCl, 1), ii) reaction of intermediates 3a-h with PQ. The prepared PQ-NSAIDs exerted moderate activities in the DPPH free radical test and β-carotene-linoleic acid assay. Moreover, ketoprofen derivatives 4d and 4b demonstrated a notable Fe2+ chelating ability as well. On the other hand, negligible antiproliferative and antituberculotic effects of conjugates 4a-h were observed.
Collapse
|
35
|
Shishido-Hara Y. Progressive multifocal leukoencephalopathy and promyelocytic leukemia nuclear bodies: a review of clinical, neuropathological, and virological aspects of JC virus-induced demyelinating disease. Acta Neuropathol 2010; 120:403-17. [PMID: 20464404 PMCID: PMC2910879 DOI: 10.1007/s00401-010-0694-x] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2010] [Revised: 04/11/2010] [Accepted: 05/02/2010] [Indexed: 12/01/2022]
Abstract
Progressive multifocal leukoencephalopathy is a fatal viral-induced demyelinating disease that was once rare but has become more prevalent today. Over the past decades, much has been learned about the disease from molecular study of the etiological agent of the disease, JC virus. Recently, promyelocytic leukemia nuclear bodies (PML-NBs), punctuate structures for important nuclear functions in eukaryotic cells, were identified as an intranuclear target of JC virus infection. Neuropathologically, JC virus-infected glial cells display diffuse amphophilic viral inclusions by hematoxylin–eosin staining (full inclusions), a diagnostic hallmark of this disease. Recent results using immunohistochemistry, however, revealed the presence of punctate viral inclusions preferentially located along the inner nuclear periphery (dot-shaped inclusions). Dot-shaped inclusions reflect the accumulation of viral progeny at PML-NBs, which may be disrupted after viral replication. Structural changes to PML-NBs have been reported for a variety of human diseases, including cancers and neurodegenerative disorders. Thus, PML-NBs may provide clues to the further pathogenesis of JC virus-induced demyelinating disease. Here, we review what we have learned since the disease entity establishment, including a look at recent progress in understanding the relationship between JC virus, etiology and PML-NBs.
Collapse
Affiliation(s)
- Yukiko Shishido-Hara
- Department of Pathology, Kyorin University School of Medicine, Mitaka, Tokyo 181-8611, Japan.
| |
Collapse
|
36
|
Abstract
Progressive multifocal encephalopathy (PML) is a fatal demyelinating disease of the central nervous system (CNS), caused by the lytic infection of oligodendrocytes by a human polyomavirus, JC virus (JCV). PML is rare disease but mostly develops in patients with underlying immunosuppressive conditions, including Hodgkin's lymphoma, lymphoproliferative diseases, in those undergoing antineoplastic therapy and AIDS. However, consistent with the occurrence of PML under immunocompromised conditions, this disease seems to be also steadily increasing among autoimmune disease patients (multiple sclerosis and Crohn's disease), who are treated with antibody-based regimens (natalizumab, efalizumab and rituximab). This unexpected occurrence of the disease among such a patient population reconfirms the existence of a strong link between the underlying immunosuppressive conditions and development of PML. These recent observations have generated a new interest among investigators to further examine the unique biology of JCV.
Collapse
Affiliation(s)
- A Sami Saribas
- Department of Neuroscience and Center for Neurovirology, Temple University School of Medicine, Education & Research Building (MERB-757), 3500 North Broad Street-7th floor, Philadelphia, PA 19140-5104
| | - Ahmet Ozdemir
- Department of Neuroscience and Center for Neurovirology, Temple University School of Medicine, Education & Research Building (MERB-757), 3500 North Broad Street-7th floor, Philadelphia, PA 19140-5104
| | - Cathy Lam
- Department of Neuroscience and Center for Neurovirology, Temple University School of Medicine, Education & Research Building (MERB-757), 3500 North Broad Street-7th floor, Philadelphia, PA 19140-5104
| | - Mahmut Safak
- Department of Neuroscience and Center for Neurovirology, Temple University School of Medicine, Education & Research Building (MERB-757), 3500 North Broad Street-7th floor, Philadelphia, PA 19140-5104
| |
Collapse
|
37
|
Del Valle L, Khalili K. Detection of human polyomavirus proteins, T-antigen and agnoprotein, in human tumor tissue arrays. J Med Virol 2010; 82:806-11. [PMID: 20336718 PMCID: PMC2861297 DOI: 10.1002/jmv.21514] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Expression of the human polyomavirus JCV genome in several experimental animals induces a variety of neural origin tumors. The viral proteins, T-antigen and Agnoprotein, contribute to the oncogenesis of JCV by associating with several tumor suppressor proteins and dysregulating signaling pathways, which results in uncontrolled cell proliferation. In addition, T-antigen and Agnoprotein have been associated with DNA damage and interfering with DNA repair mechanisms. In this study, we have utilized commercially available tissue arrays of human tumors of various origins and demonstrated the expression of both T-antigen and Agnoprotein in some, but not all, tumors of neural and non-neural origin. Most notably, more than 40% of human glioblastomas and greater than 30% of colon adenocarcinomas express viral proteins. The detection of viral transforming proteins, T-antigen and Agnoprotein in the absence of viral capsid proteins suggests a role for JCV in the development and/or progression of human tumors. These results invite further large-scale investigation on the role of polyomaviruses, particularly JCV in the pathogenesis of human cancer.
Collapse
Affiliation(s)
- Luis Del Valle
- Department of Neuroscience, Center for Neurovirology, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Kamel Khalili
- Department of Neuroscience, Center for Neurovirology, Temple University School of Medicine, Philadelphia, Pennsylvania
| |
Collapse
|
38
|
Amirian E, Scheurer ME, Bondy ML. The association between birth order, sibship size and glioma development in adulthood. Int J Cancer 2010; 126:2752-6. [PMID: 19830691 DOI: 10.1002/ijc.24962] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The etiology of brain tumors is still largely unknown. Previous research indicates that infectious agents and immunological characteristics may influence adult glioma risk. The purpose of our study was to evaluate the effects of birth order and sibship size (total number of siblings), as indicators of the timing and frequency of early life infections, on adult glioma risk using a population of 489 cases and 540 cancer-free controls from the Harris County Brain Tumor Study. Odds ratios for birth order and sibship size were calculated separately from multivariable logistic regression models, adjusting for sex, family history of cancer, education, and age. Each one-unit increase in birth order confers a 13% decreased risk of glioma development in adulthood (OR = 0.87, 95% CI = 0.79-0.97). However, sibship size was not significantly associated with adult glioma status (OR = 0.97, 95% CI = 0.91-1.04). Our study indicates that individuals who were more likely to develop common childhood infections at an earlier age (those with a higher birth order) may be more protected against developing glioma in adulthood. More biological and epidemiological research is warranted to clarify the exact mechanisms through which the timing of common childhood infections and the course of early life immune development affect gliomagenesis.
Collapse
Affiliation(s)
- E Amirian
- Department of Epidemiology, University of Texas M. D. Anderson Cancer Center, Houston, TX, USA
| | | | | |
Collapse
|
39
|
Chronic viral infection and primary central nervous system malignancy. J Neuroimmune Pharmacol 2010; 5:387-403. [PMID: 20387126 PMCID: PMC2914282 DOI: 10.1007/s11481-010-9204-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2009] [Accepted: 03/05/2010] [Indexed: 01/08/2023]
Abstract
Primary central nervous system (CNS) tumors cause significant morbidity and mortality in both adults and children. While some of the genetic and molecular mechanisms of neuro-oncogenesis are known, much less is known about possible epigenetic contributions to disease pathophysiology. Over the last several decades, chronic viral infections have been associated with a number of human malignancies. In primary CNS malignancies, two families of viruses, namely polyomavirus and herpesvirus, have been detected with varied frequencies in a number of pediatric and adult histological tumor subtypes. However, establishing a link between chronic viral infection and primary CNS malignancy has been an area of considerable controversy, due in part to variations in detection frequencies and methodologies used among researchers. Since a latent viral neurotropism can be seen with a variety of viruses and a widespread seropositivity exists among the population, it has been difficult to establish an association between viral infection and CNS malignancy based on epidemiology alone. While direct evidence of a role of viruses in neuro-oncogenesis in humans is lacking, a more plausible hypothesis of neuro-oncomodulation has been proposed. The overall goals of this review are to summarize the many human investigations that have studied viral infection in primary CNS tumors, discuss potential neuro-oncomodulatory mechanisms of viral-associated CNS disease and propose future research directions to establish a more firm association between chronic viral infections and primary CNS malignancies.
Collapse
|
40
|
Valle LD, Piña-Oviedo S, Perez-Liz G, Augelli BJ, Azizi SA, Khalili K, Gordon J, Krynska B. Bone marrow-derived mesenchymal stem cells undergo JCV T-antigen mediated transformation and generate tumors with neuroectodermal characteristics. Cancer Biol Ther 2010; 9:286-94. [PMID: 20190567 PMCID: PMC2921558 DOI: 10.4161/cbt.9.4.10653] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
There is now accumulating evidence showing that some tumors may arise from transformed stem cells. In this study we demonstrate that adult bone marrow- derived mesenchymal stem cells (MSCs) undergo neoplastic transformation induced by the human polyomavirus JCV, early protein, T-antigen, and are tumorigenic when transplanted into the flanks of Nude mice as compared to non-transformed MSCs. Histologically, the tumors are heterogeneous with mesenchymal and neural crest characteristics as evidenced by expression of the neural crest markers p75, SOX-10, and S-100, with populations of tumor cells exhibiting characteristics of primitive neuroectodermal cells. In addition, a subset of T-antigen positive tumor cells exhibit a high proliferation index as detected by Ki-67 labeling, and co-express CD133, a marker which is expressed on cancer stem cells. These results show that tumors with neuroectodermal characteristics may arise from transformation of MSCs, a globally accessible adult stem cell with multipotent differentiation capacity. In light of earlier reports on the association of JCV with a broad variety of human tumors, our data suggests that T-antigen transformation of adult stem cells with a multipotent capacity can serve as a possible common origin for some of these cancers, and offers a novel model for oncogenesis.
Collapse
Affiliation(s)
- Luis Del Valle
- Department of Neuroscience, Temple University School of Medicine, Philadelphia, PA USA
- Department of Pathology, Temple University School of Medicine, Philadelphia, PA USA
| | - Sergio Piña-Oviedo
- Department of Neuroscience, Temple University School of Medicine, Philadelphia, PA USA
| | - Georgina Perez-Liz
- Department of Neuroscience, Temple University School of Medicine, Philadelphia, PA USA
| | - Brian J. Augelli
- Department of Neurology, Temple University School of Medicine, Philadelphia, PA USA
| | - S. Ausim Azizi
- Department of Neurology, Temple University School of Medicine, Philadelphia, PA USA
| | - Kamel Khalili
- Department of Neuroscience, Temple University School of Medicine, Philadelphia, PA USA
| | - Jennifer Gordon
- Department of Neuroscience, Temple University School of Medicine, Philadelphia, PA USA
| | - Barbara Krynska
- Department of Neurology, Temple University School of Medicine, Philadelphia, PA USA
| |
Collapse
|
41
|
Gualco E, Urbanska K, Perez-Liz G, Sweet T, Peruzzi F, Reiss K, Del Valle L. IGF-IR-dependent expression of Survivin is required for T-antigen-mediated protection from apoptosis and proliferation of neural progenitors. Cell Death Differ 2009; 17:439-51. [PMID: 19834489 PMCID: PMC2822053 DOI: 10.1038/cdd.2009.146] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The Insulin-like Growth Factor-1 Receptor (IGF-IR) and the human polyomavirus JCV protein, T-Antigen cooperate in the transformation of neuronal precursors in the cerebellum, which may be a contributing factor in the development of brain tumors. Since it is not clear why T-Antigen requires IGF-IR for transformation, we investigated this process in neural progenitors from IGF-IR knockout embryos (ko-IGF-IR) and from their wild type non-transgenic littermates (wt-IGF-IR). In contrast to wt-IGF-IR, the brain and dorsal root ganglia of ko-IGF-IR embryos showed low levels of the anti-apoptotic protein Survivin, accompanied by elevated numbers of apoptotic neurons and an earlier differentiation phenotype. In wt-IGF-IR neural progenitors in vitro, induction of T-Antigen expression tripled the expression of Survivin, and accelerated cell proliferation. In ko-IGF-IR progenitors induction of T-Antigen failed to increase Survivin, resulting in massive apoptosis. Importantly, ectopic expression of Survivin protected ko-IGF-IR progenitor cells from apoptosis and siRNA inhibition of Survivin activated apoptosis in wt-IGF-IR progenitors expressing T-Antigen. Our results indicate that reactivation of the anti-apoptotic Survivin may be a critical step in JCV T-Antigen induced transformation, which in neural progenitors requires IGF-IR.
Collapse
Affiliation(s)
- E Gualco
- Department of Neuroscience, Center for Neurovirology, Temple University School of Medicine, Philadelphia, PA, USA
| | | | | | | | | | | | | |
Collapse
|
42
|
Modulation of JC virus transcription by C/EBPbeta. Virus Res 2009; 146:97-106. [PMID: 19747512 DOI: 10.1016/j.virusres.2009.09.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2009] [Revised: 09/03/2009] [Accepted: 09/03/2009] [Indexed: 11/21/2022]
Abstract
The polyomavirus JC (JCV) causes the demyelinating disease progressive multifocal leukoencephalopathy (PML). Infection by JCV is very common in childhood after which the virus enters a latent state, which is poorly understood. Under conditions of severe immunosuppression, especially AIDS, JCV may reactivate to cause PML. Expression of JC viral proteins is regulated by the JCV non-coding control region (NCCR), which contains an NF-kappaB binding site previously shown to activate transcription. We now report that C/EBPbeta inhibits basal and NF-kappaB-stimulated JCV transcription via the same site. Gel shift analysis showed C/EBPbeta bound to this region in vitro and ChIP assays confirmed this binding in vivo. Further, a ternary complex of NF-kappaB/p65, C/EBPbeta-LIP and JCV DNA could be detected in co-immunoprecipitation experiments. Mutagenesis analysis of the JCV NCCR indicated p65 and C/EBPbeta-LIP bound to adjacent but distinct sites and that both sites regulate basal and p65-stimulated transcription. Thus C/EBPbeta negatively regulates JCV, which together with NF-kappaB activation, may control the balance between JCV latency and activation leading to PML. This balance may be regulated by proinflammatory cytokines in the brain.
Collapse
|
43
|
Maginnis MS, Atwood WJ. JC virus: an oncogenic virus in animals and humans? Semin Cancer Biol 2009; 19:261-9. [PMID: 19505654 PMCID: PMC2694964 DOI: 10.1016/j.semcancer.2009.02.013] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2008] [Revised: 02/09/2009] [Accepted: 02/12/2009] [Indexed: 12/12/2022]
Abstract
JC virus (JCV) is a human polyomavirus of the Polyomaviridae family, which also includes BK virus and simian vacuolating virus 40 (SV40). JC virus was first isolated in 1971 from the brain of a patient with Progressive Multifocal Leukoencephalopathy (PML). Like other polyomaviruses, JCV has a restricted host range. The virus infects the majority of the human population with seroconversion occurring during adolescence. JCV has a limited and specific tissue tropism infecting the kidney and oligodendrocytes and astrocytes in the central nervous system (CNS). Initial JCV infection is generally asymptomatic in immunocompetent hosts, and it establishes a persistent infection in the kidney and possibly bone marrow. In immunocompromised individuals JCV can cause a lytic infection in the CNS and lead to development of the fatal, demyelinating disease PML. The name polyoma is derived from the Greek terms: poly, meaning many, and oma, meaning tumors, owing to the capacity of this group of viruses to cause tumors. JCV inoculation of small animal models and non-human primates, which are not permissive to a productive JCV infection, leads to tumor formation. Given the ubiquitous nature of the virus and its strong association with cancer in animal models, it is hypothesized that JCV plays a role in human cancers. However, the role for JCV in human cancers and tumor formation is not clear. Some researchers have reported an association of JCV with human cancers including brain tumors, colorectal cancers, and cancers of the gastrointestinal tract, while other groups report no correlation. Here, we review the role of JCV in cancers in animal models and present the findings on JCV in human cancers.
Collapse
Affiliation(s)
- Melissa S. Maginnis
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, 70 Ship Street Box G-E4 Providence, RI 02903
| | - Walter J. Atwood
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, 70 Ship Street Box G-E4 Providence, RI 02903
| |
Collapse
|
44
|
Basile A, Darbinian N, Kaminski R, White MK, Gentilella A, Turco MC, Khalili K. Evidence for modulation of BAG3 by polyomavirus JC early protein. J Gen Virol 2009; 90:1629-1640. [PMID: 19282432 PMCID: PMC2811539 DOI: 10.1099/vir.0.008722-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2008] [Accepted: 03/08/2009] [Indexed: 12/13/2022] Open
Abstract
Polyomavirus JC (JCV) infects oligodendrocytes and astrocytes in the brain and is the cause of the demyelinating disease progressive multifocal leukoencephalopathy (PML). In cell culture, JCV infection is characterized by severe damage to cellular DNA, which begins early in infection, and a viral cytopathic effect, which is observed late in infection. Nevertheless, these JCV-infected cells show a low level of apoptosis, at both the early and late stages of infection. This suggests that there is conflicting interplay between viral anti-apoptotic pathways that seek to optimize virus production, e.g. through T antigen (T-Ag)-p53 interaction, and cellular pro-apoptotic pathways that seek to eliminate virally infected cells. The apoptosis regulatory protein BAG3 is a member of the human Bcl-2-associated athanogene (BAG) family of proteins, which function as molecular co-chaperones through their interaction with Hsc70/Hsp70 and function in the regulation of the cellular stress response, proliferation and apoptosis. This study showed that BAG3 protein is downregulated upon JCV infection and that this effect is mediated by JCV T-Ag via repression of the BAG3 promoter. The site of action of T-Ag was mapped to an AP2 site in the BAG3 promoter, and gel shift and chromatin immunoprecipitation assays showed that T-Ag inhibited AP2 binding to this site, resulting in downregulation of BAG3 promoter expression. Using BAG3 and T-Ag expression and BAG3 siRNA, it was found that BAG3 and T-Ag had antagonistic effects on the induction of apoptosis, being anti-apoptotic and pro-apoptotic, respectively. The significance of these interactions to the JCV life cycle is discussed.
Collapse
Affiliation(s)
- Anna Basile
- Department of Neuroscience, Center for Neurovirology, Temple University School of Medicine, 1900 North 12th Street, Philadelphia, PA 19122, USA
- Department of Pharmaceutical Sciences, University of Salerno, via ponte don Melillo, 84084 Fisciano, Italy
| | - Nune Darbinian
- Department of Neuroscience, Center for Neurovirology, Temple University School of Medicine, 1900 North 12th Street, Philadelphia, PA 19122, USA
| | - Rafal Kaminski
- Department of Neuroscience, Center for Neurovirology, Temple University School of Medicine, 1900 North 12th Street, Philadelphia, PA 19122, USA
| | - Martyn K. White
- Department of Neuroscience, Center for Neurovirology, Temple University School of Medicine, 1900 North 12th Street, Philadelphia, PA 19122, USA
| | - Antonio Gentilella
- Department of Neuroscience, Center for Neurovirology, Temple University School of Medicine, 1900 North 12th Street, Philadelphia, PA 19122, USA
| | - Maria Caterina Turco
- Department of Pharmaceutical Sciences, University of Salerno, via ponte don Melillo, 84084 Fisciano, Italy
| | - Kamel Khalili
- Department of Neuroscience, Center for Neurovirology, Temple University School of Medicine, 1900 North 12th Street, Philadelphia, PA 19122, USA
| |
Collapse
|
45
|
Sariyer IK, Safak M, Gordon J, Khalili K. Generation and characterization of JCV permissive hybrid cell lines. J Virol Methods 2009; 159:122-6. [PMID: 19442856 PMCID: PMC2692597 DOI: 10.1016/j.jviromet.2009.02.023] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2008] [Revised: 02/10/2009] [Accepted: 02/19/2009] [Indexed: 11/30/2022]
Abstract
JC virus (JCV) is a human neurotropic polyomavirus whose replication in the central nervous system induces the fatal demyelinating disease, progressive multifocal leukoencephalopathy (PML). JCV particles have been detected primarily in oligodendrocytes and astrocytes of the brains of patients with PML and in the laboratory its propagation is limited to primary cultures of human fetal glial cells. In this short communication, the development of a new cell culture system is described through the fusion of primary human fetal astrocytes with the human glioblastoma cell line, U-87MG. The new hybrid cell line obtained from this fusion has the capacity to support efficiently expression of JCV and replication of viral DNA in vitro up to 16 passages. This cell line can serve as a reliable culture system to study the biology of JCV host-cell interaction, determine the mechanisms involved in cell type specific replication of JCV, and provide a convenient cell culture system for high throughput screening of anti-viral agents.
Collapse
Affiliation(s)
- Ilker K. Sariyer
- Department of Neuroscience, Center for Neurovirology, Temple University School of Medicine, 1900 North 12th Street, 015-96, Room 203, Philadelphia, PA 19122 USA
| | - Mahmut Safak
- Department of Neuroscience, Center for Neurovirology, Temple University School of Medicine, 1900 North 12th Street, 015-96, Room 203, Philadelphia, PA 19122 USA
| | - Jennifer Gordon
- Department of Neuroscience, Center for Neurovirology, Temple University School of Medicine, 1900 North 12th Street, 015-96, Room 203, Philadelphia, PA 19122 USA
| | - Kamel Khalili
- Department of Neuroscience, Center for Neurovirology, Temple University School of Medicine, 1900 North 12th Street, 015-96, Room 203, Philadelphia, PA 19122 USA
| |
Collapse
|
46
|
Perez-Liz G, Del Valle L, Gentilella A, Croul S, Khalili K. Detection of JC virus DNA fragments but not proteins in normal brain tissue. Ann Neurol 2008; 64:379-87. [PMID: 18688812 PMCID: PMC2632778 DOI: 10.1002/ana.21443] [Citation(s) in RCA: 99] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Progressive multifocal leukoencephalopathy (PML) is a fatal demyelinating disease of the white matter affecting immunocompromised patients that results from the cytolytic destruction of glial cells by the human neurotropic JC virus (JCV). According to one model, during the course of immunosuppression, JCV departs from its latent state in the kidney and after entering the brain, productively infects and destroys oligodendrocytes. The goal of this study was to test the hypothesis that JCV may reside in a latent state in a specific region of the brains of immunocompetent (non-PML) individuals without any neurological conditions. METHODS Gene amplification was performed together with immunohistochemistry to examine the presence of JCV DNA sequences and expression of its genome in five distinct regions of the brain from seven immunocompetent non-PML individuals. RESULTS Although no viral proteins were expressed in any of these cases, fragments of the viral DNA were present in various regions of normal brain. Laser-capture microdissection showed the presence of JCV DNA in oligodendrocytes and astrocytes, but not in neurons. INTERPRETATION The detection of fragments of viral DNA in non-PML brain suggests that JCV has full access to all regions of the brain in immunocompetent individuals. Thus, should the immune system become impaired, the passing and/or the resident virus may gain the opportunity to express its genome and initiate its lytic cycle in oligodendrocytes. The brain as a site of JCV latency is a possibility.
Collapse
Affiliation(s)
- Georgina Perez-Liz
- Department of Neuroscience, Center for Neurovirology, Temple University School of Medicine, Philadelphia, PA 19122, USA
| | | | | | | | | |
Collapse
|
47
|
Promyelocytic Leukemia Nuclear Bodies Provide a Scaffold for Human Polyomavirus JC Replication and Are Disrupted After Development of Viral Inclusions in Progressive Multifocal Leukoencephalopathy. J Neuropathol Exp Neurol 2008; 67:299-308. [DOI: 10.1097/nen.0b013e31816a1dd3] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
48
|
Abstract
BACKGROUND JC virus (JCV) has been implicated in the pathogenesis of colorectal cancer; however, its role in premalignant lesions is unknown. The hypothesis that JCV DNA sequences and T-antigen (T-Ag) expression may be present in adenomatous polyps of the colon was tested. Furthermore, an association between JCV and microsatellite instability (MSI) was also sought in these lesions. METHODS DNA was extracted from 74 paraffin-embedded adenomatous polyps. JCV gene sequences were amplified by polymerase chain reaction (PCR), and the specificity confirmed by DNA sequencing. Immunohistochemical staining was performed to localize T-Ag expression in the adenomas using a monoclonal antibody. For microsatellite instability analysis, 5 mononucleotide repeat markers (BAT-25, BAT-26, NR-21, NR-24, and NR-27) were coamplified in a pentaplex PCR and analyzed for deletion mutations. RESULTS JCV T-Ag sequences were found in 82% (61 of 74) of adenomas, and T-Ag protein was expressed in 16% (12 of 74) of these polyps. The T-Ag staining was localized exclusively in the nuclei of adenoma cells, but never in the cytoplasm or the adjacent nonneoplastic cells. The prevalence of MSI-H and non-MSI-H (MSI-L/MSS) in adenomatous polyps was 9.5% (7 of 74) and 90.5% (67 of 74), respectively. Among the 61 adenomas that harbored JCV sequences, 8% (5 of 61) were MSI-H, and similarly among 12 adenomatous polyps expressing T-Ag protein 8% (1 of 12) of the adenomatous polyps were MSI-H. CONCLUSIONS JCV T-Ag DNA sequences are frequently present in adenomatous polyps of the colon, and T-Ag is expressed specifically in the nuclei of these premalignant lesions. This study indicates that JCV T-Ag is present in the early stage of colonic carcinogenesis. Future studies will be required to determine the molecular mechanism of carcinogenesis in these JCV-infected lesions.
Collapse
Affiliation(s)
- Woon-Tae Jung
- Division of Gastroenterology, Department of Internal Medicine, the Charles A Sammons Cancer Center, and the Baylor Research Institute, Baylor University Medical Center, Dallas, Texas
- Department of Internal Medicine, College of Medicine, Institute of Health Sciences, Gyeong-sang National University, Jinju, South Korea
| | - Mei-Shu Li
- Division of Gastroenterology, Department of Internal Medicine, the Charles A Sammons Cancer Center, and the Baylor Research Institute, Baylor University Medical Center, Dallas, Texas
| | - Ajay Goel
- Division of Gastroenterology, Department of Internal Medicine, the Charles A Sammons Cancer Center, and the Baylor Research Institute, Baylor University Medical Center, Dallas, Texas
| | - C. Richard Boland
- Division of Gastroenterology, Department of Internal Medicine, the Charles A Sammons Cancer Center, and the Baylor Research Institute, Baylor University Medical Center, Dallas, Texas
| |
Collapse
|
49
|
Kutsuna T, Zheng H, Abdel-Aziz HO, Murai Y, Tsuneyama K, Furuta I, Takano Y. High JC virus load in tongue carcinomas may be a risk factor for tongue tumorigenesis. Virchows Arch 2008; 452:405-10. [PMID: 18283491 PMCID: PMC2668633 DOI: 10.1007/s00428-007-0534-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2007] [Revised: 10/17/2007] [Accepted: 10/21/2007] [Indexed: 11/26/2022]
Abstract
The John Cunningham virus (JCV) asymptomatically infects a large proportion (approximately 90%) of the population worldwide but may be activated in immunodeficient patients, resulting in progressive multifocal leukoencephalopathy. Recent reports demonstrated its oncogenic role in malignancies. In this paper, the presence of JCV-targeting T antigen was investigated in tongue carcinoma (TC, n = 39), dysplastic tongue epithelium (DTE, n = 15) and glossitis (n = 15) using real-time polymerase chain reaction (PCR) and in situ PCR and immunohistochemistry, and JCV copies were analyzed with the clinicopathological parameters of TCs. The results demonstrated that glossitis and DTEs had significantly lower copies of JCV (410.5 +/- 44.3 and 658.3 +/- 53.3 copies/mug DNA respectively) than TCs (981.5 +/- 14.0, p < 0.05). When they were divided into three groups with 0-200 copies/mug DNA (low), 201-1,000 (moderate) and more than 1001 (high), TCs showed 3 (7.6%) in the low group, 21 (53.8%) in the moderate group and 15 (38.4%) in the high group and glossitis showed 11 (73.3%) in the low group, 0 (0%) in the moderate group and 4 (26.6%) in the high group. The DTEs occupied an intermediate position between them (p < 0.001). In situ PCR demonstrated that the nuclei of TC and DTE cells are sporadically T-antigen positive but not in nasal turbinate epithelial cells. Immunohistochemistry for T-antigen protein revealed four positive cases only in TCs. The existence of JCV T-antigen DNA was not associated with the clinicopathological variables of TCs. In conclusion, the presence of JCV may be a risk factor of tongue carcinogenesis.
Collapse
Affiliation(s)
- Tomohiko Kutsuna
- Department of Diagnostic Pathology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama, 930-0194 Japan
- Department of Oral and Maxillofacial Surgery, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Huachuan Zheng
- Department of Diagnostic Pathology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama, 930-0194 Japan
- Department of Biochemistry, College of Basic Medicine, China Medical University, Shenyang, China
| | - Hekmat Osman Abdel-Aziz
- Department of Diagnostic Pathology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama, 930-0194 Japan
| | - Yoshihiro Murai
- Department of Diagnostic Pathology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama, 930-0194 Japan
| | - Koichi Tsuneyama
- Department of Diagnostic Pathology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama, 930-0194 Japan
| | - Isao Furuta
- Department of Oral and Maxillofacial Surgery, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Yasuo Takano
- Department of Diagnostic Pathology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama, 930-0194 Japan
| |
Collapse
|
50
|
Detection of the JC virus genome in lung cancers: possible role of the T-antigen in lung oncogenesis. Appl Immunohistochem Mol Morphol 2008; 15:394-400. [PMID: 18091381 DOI: 10.1097/01.pai.0000213126.96590.64] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The JC virus (JCV) infects a large proportion of the population worldwide and 80% to 90% of adults are seropositive and it may be activated in immunodeficient patients, resulting in progressive multifocal leukoencephalopathy. Recent reports described the possibility of its oncogenetic role in several malignancies. To clarify whether JCV might have a potential role in the genesis of lung cancers, we investigated the presence of its genome in 62 tumors, along with 23 samples of normal lung tissue, targeting the T-antigen, VP, and Agnoprotein by nested polymerase chain reaction/Southern blotting followed by direct DNA sequencing. Immunohistochemistry was performed to assess links between p53 and beta-catenin in lung cancers and the presence of T-antigen. The T-antigen was detected in 25 of 62 lung cancers but only 4 of 23 normal lung samples (P=0.048). In total, the JCV genome was present in 33 of the lung cancers and 10 of the normal samples. Furthermore, T-antigen was found in cancer cells in metastatic lymph nodes in 3 of 4 cases (P=0.042) and was more frequently detected in adenocarcinomas than in squamous cell carcinomas (P=0.038). Immunohistochemistry showed significant correlations between T-antigen and p53 (P=0.022) and also nuclear detection of beta-catenin (P=0.021). It is concluded that the JCV genome might be present in cancer cells in approximately half of all Japanese lung cancer cases, and that the T-antigen may play a role in oncogenesis of lung cancers through inactivation of p53 and dysregulation of the Wnt signaling pathway.
Collapse
|