Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
J Pediatr Hematol Oncol ; 43(7): e972-e974, 2021 10 01.
Article in English | MEDLINE | ID: mdl-33235157

ABSTRACT

COVID-19, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is responsible for a global pandemic that can cause severe infections in children, especially those with comorbid conditions. Here, we report a case of a child with a newly diagnosed medulloblastoma, Fanconi Anemia, and SARS-CoV-2 infection. Through multidisciplinary care coordination and meticulous planning, we were able to safely initiate this patient's oncology care and implement a long-term model to address the patient's care. This approach could be replicated with any newly diagnosed pediatric patient that requires monitoring for signs of COVID-19 with concurrent oncology care.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , COVID-19/complications , Fanconi Anemia/drug therapy , Medulloblastoma/drug therapy , SARS-CoV-2/isolation & purification , COVID-19/transmission , COVID-19/virology , Child, Preschool , Fanconi Anemia/diagnosis , Fanconi Anemia/virology , Female , Humans , Medulloblastoma/diagnosis , Medulloblastoma/virology , Prognosis
2.
Sci Rep ; 8(1): 16595, 2018 11 09.
Article in English | MEDLINE | ID: mdl-30413749

ABSTRACT

Japanese encephalitis virus (JEV) is a member of neurotropic flaviviruses transmitted by mosquito bites, causing severe central nervous system disorders. Current JEV genotype III vaccines have a low protection against genotype I isolates in the risk zone. The lead compound CW-33, ethyl 2-(3',5'-dimethylanilino)-4-oxo-4,5-dihydrofuran-3-carboxylate, demonstrates the antiviral activity against JEV with an IC50 values of 38.5 µM for virus yield reduction (Int J Mol Sci 2016,17: E1386). This study synthesized fourteen CW-33 analogues containing a fluoro atom or one methoxy group at the C-2, C-3, or C-4 of anilino ring, and then evaluated for their antiviral activity and mechanism. Among 6 amalogues, CW-33A (ethyl 2-(2-fluoroanilino)-4-oxo- 4,5-dihydrofuran-3-carboxylate), and CW-33D (ethyl 2-(3-methoxyanilino)-4-oxo- 4,5-dihydrofuran-3-carboxylate exhibited antiviral potentials in viral cytopathic effect (CPE) inhibition. CW-33A significantly suppressed the viral protein expression, genome synthesis and intracellular JEV particle production, showing a higher inhibitory effect on JEV yield than CW-33 and CW-33D. The study demonstrated that a mono-fluoro substitution on at the C-2 anilino ring of CW-33 improved the antiviral activity JEV, revealing the structure-activity relationship for developing novel agents against JEV infection.


Subject(s)
Aniline Compounds/pharmacology , Antiviral Agents/pharmacology , Cytopathogenic Effect, Viral/drug effects , Encephalitis, Japanese/drug therapy , Furans/pharmacology , Medulloblastoma/drug therapy , Viral Proteins/genetics , Virus Replication/drug effects , Aniline Compounds/chemistry , Antiviral Agents/chemistry , Cerebellar Neoplasms/drug therapy , Cerebellar Neoplasms/virology , Encephalitis Virus, Japanese/drug effects , Encephalitis, Japanese/complications , Encephalitis, Japanese/virology , Furans/chemistry , Genome, Viral , Genotype , Humans , Medulloblastoma/virology , Molecular Structure
3.
Neuro Oncol ; 20(12): 1606-1615, 2018 11 12.
Article in English | MEDLINE | ID: mdl-29912438

ABSTRACT

Background: Oncolytic measles virus (MV) is effective in xenograft models of many tumor types in immune-compromised mice. However, no murine cell line exists that is tumorigenic, grows in immune-competent mice, and is killed by MV. The lack of such a model prevents an examination of the effect of the immune system on MV oncotherapy. Methods: Cerebellar stem cells from human CD46-transgenic immunocompetent mice were transduced to express Sendai virus C-protein, murine C-Myc, and Gfi1b proteins. The resultant cells were injected into the brain of NSG mice, and a cell line, called CSCG, was prepared from the resulting tumor. Results: CSCG cells are highly proliferative, and express stem cell markers. These cells are permissive for replication of MV and are killed by the virus in a dose- and time-dependent manner. CSCG cells form aggressive tumors that morphologically resemble medulloblastoma when injected into the brains of immune-competent mice. On the molecular level, CSCG tumors overexpress natriuretic peptide receptor 3 and gamma-aminobutyric acid type A receptor alpha 5, markers of Group 3 medulloblastoma. A single intratumoral injection of MV‒green fluorescent protein resulted in complete tumor regression and prolonged survival of animals compared with treatments with phosphate buffered saline (P = 0.0018) or heat-inactivated MV (P = 0.0027). Conclusions: This immune-competent model provides the first platform to test therapeutic regimens of oncolytic MV for Group 3 medulloblastoma in the presence of anti-measles immunity. The strategy presented here can be used to make MV-sensitive murine models of any human tumor for which the driving mutations are known.


Subject(s)
Cerebellar Neoplasms/therapy , Disease Models, Animal , Immunocompetence , Measles virus/genetics , Medulloblastoma/therapy , Oncolytic Virotherapy , Animals , Cerebellar Neoplasms/immunology , Cerebellar Neoplasms/metabolism , Cerebellar Neoplasms/virology , Humans , Measles/virology , Medulloblastoma/immunology , Medulloblastoma/metabolism , Medulloblastoma/virology , Membrane Cofactor Protein/genetics , Membrane Cofactor Protein/metabolism , Mice , Mice, Transgenic , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Tumor Cells, Cultured , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Replication , Xenograft Model Antitumor Assays
4.
Int J Oncol ; 52(4): 1317-1327, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29484388

ABSTRACT

Among all brain tumors diagnosed in children, medulloblastomas (MBs) are associated with a poor prognosis. The etiology of MB is not fully understood, yet the impact of epigenetic alterations of oncogenes has previously been established. During the past decade, the human cytomegalovirus (HCMV) has been detected in several types of cancer, including MB. Since DNA methylation occurs in the cell nucleus and this is considered a host defence response, we studied the impact of HCMV infection on DNA methyltransferase (DNMT­1) in MB (D324) cells, human umbilical vein endothelial cells (HUVECs) as well as in MB tissue sections. We hypothesized that infection and DNMT­1 intracellular localization are linked. Uninfected and HCMV­infected D324 cells and HUVECs were analyzed for HCMV immediate early (HCMV­IE) protein, HCMV­glycoprotein B (HCMV­gB) and DNMT­1 using immunofluorescence staining and quantitative ELISA. DNMT­1 localized to the nucleus of uninfected and HCMV­IE- expressing D324 cells and HUVECs, but accumulated in the extra nuclear space in all HCMV­gB-positive cells. Inhibition of HCMV late protein expression by Cymevene® (ganciclovir) prevented the cytoplasmic localization of DNMT­1. Treatment of HCMV­ infected D324 cells and HUVECs with the methylation inhibitor 5-Azacytidine (5AZA), significantly increased HCMV­IE and HCMV­gB gene transcription and protein expression. Immunohistochemical staining of DNMT­1 and HCMV proteins in MB cancer tissue sections revealed both nuclear and cytoplasmic DNMT­1 localization. In conclusion, DNMT­1 resides in the cytoplasm of HCMV­gB-expressing HUVECs and D324 cells. Increased viral protein synthesis in 5AZA-treated cells suggests that HCMV replication may benefit from a DNA methyltransferase-free cellular environment. Our findings emphasize the importance of assessing potential viral activation in the treatment of MB patients with epigenetic drugs.


Subject(s)
Cerebellar Neoplasms/virology , Cytomegalovirus Infections/complications , DNA (Cytosine-5-)-Methyltransferase 1/biosynthesis , Human Umbilical Vein Endothelial Cells/virology , Medulloblastoma/virology , Virus Replication/drug effects , Antimetabolites, Antineoplastic/pharmacology , Azacitidine/pharmacology , Cytomegalovirus , Cytoplasm/metabolism , Humans , Viral Proteins , Virus Activation/drug effects , Virus Replication/physiology
5.
Cancer Lett ; 408: 55-59, 2017 11 01.
Article in English | MEDLINE | ID: mdl-28844716

ABSTRACT

Medulloblastoma is the most common solid tumor among children. Current therapeutic strategies for this malignancy include surgical resection, radiation therapy and chemotherapy. However, these treatments are accompanied with serious side effects such as neurological complications and psychosocial problems, due to the severity of treatment on the developing nervous system. To solve this problem, novel therapeutic approaches are currently being investigated. One of them is targeting human cytomegalovirus in medulloblastoma cancer cells. However, this approach is still under debate, since the presence of cytomegalovirus in medulloblastomas remains controversial. In this review, we discuss the current controversies on the role of cytomegalovirus in medulloblastoma oncogenesis and the potential of cytomegalovirus as a novel (immuno)therapeutic target.


Subject(s)
Cerebellar Neoplasms/therapy , Cytomegalovirus/isolation & purification , Immunotherapy , Medulloblastoma/therapy , Oncogenes , Animals , Cerebellar Neoplasms/immunology , Cerebellar Neoplasms/virology , Humans , Medulloblastoma/immunology , Medulloblastoma/virology
7.
Virus Genes ; 52(3): 309-16, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26924586

ABSTRACT

A short upstream open reading frame (uORF) was recently identified in the 5' untranslated region of some tick-borne encephalitis virus (TBEV) strains. However, it is not known if the peptide encoded by TBEV uORF (TuORF) is expressed in infected cells. Here we show that TuORF forms three phylogenetically separated clades which are typical of European, Siberian, and Far-Eastern TBEV subtypes. Analysis of selection pressure acting on the TuORF area showed that it is under positive selection pressure. Theoretically, TuORF may code for a short hydrophobic peptide embedded in a biological membrane. However, expression of TuORF was detectable neither by immunoblotting in tick and mammalian cell lines infected with TBEV nor by immunofluorescence in TBEV-infected mammalian cell lines. These results support the idea that TuORF is not expressed in TBEV-infected cell or expressed in undetectably low concentrations. Therefore we can assume that TuORF has either minor or no biological role in the TBEV life cycle.


Subject(s)
Encephalitis Viruses, Tick-Borne/genetics , Encephalitis, Tick-Borne/virology , Genome, Viral , Open Reading Frames , Peptide Biosynthesis/genetics , Animals , Cell Line , Glioblastoma/virology , Humans , Ixodes/virology , Medulloblastoma/virology , Mutation , Neuroblastoma/virology , Peptide Biosynthesis/immunology , Phylogeny
8.
PLoS One ; 9(9): e108861, 2014.
Article in English | MEDLINE | ID: mdl-25268364

ABSTRACT

Human cytomegalovirus (HCMV) has been indicated being a significant oncomodulator. Recent reports have suggested that an antiviral treatment alters the outcome of a glioblastoma. We analysed the performance of commercial HCMV-antibodies applying the immunohistochemical (IHC) methods on brain sample obtained from a subject with a verified HCMV infection, on samples obtained from 14 control subjects, and on a tissue microarray block containing cores of various brain tumours. Based on these trials, we selected the best performing antibody and analysed a cohort of 417 extra- and intra-axial brain tumours such as gliomas, medulloblastomas, primary diffuse large B-cell lymphomas, and meningiomas. HCMV protein pp65 immunoreactivity was observed in all types of tumours analysed, and the IHC expression did not depend on the patient's age, gender, tumour type, or grade. The labelling pattern observed in the tumours differed from the labelling pattern observed in the tissue with an active HCMV infection. The HCMV protein was expressed in up to 90% of all the tumours investigated. Our results are in accordance with previous reports regarding the HCMV protein expression in glioblastomas and medulloblastomas. In addition, the HCMV protein expression was seen in primary brain lymphomas, low-grade gliomas, and in meningiomas. Our results indicate that the HCMV protein pp65 expression is common in intra- and extra-axial brain tumours. Thus, the assessment of the HCMV expression in tumours of various origins and pathologically altered tissue in conditions such as inflammation, infection, and even degeneration should certainly be facilitated.


Subject(s)
Brain Neoplasms/virology , Cytomegalovirus/metabolism , Phosphoproteins/analysis , Viral Matrix Proteins/analysis , Adolescent , Adult , Aged , Aged, 80 and over , Brain Neoplasms/pathology , Cohort Studies , Cytomegalovirus/isolation & purification , Female , Glioblastoma/pathology , Glioblastoma/virology , Humans , Immunohistochemistry , Lymphoma/pathology , Lymphoma/virology , Male , Medulloblastoma/pathology , Medulloblastoma/virology , Meningioma/pathology , Meningioma/virology , Middle Aged , Neoplasm Grading , Young Adult
9.
Cancer Med ; 3(4): 1038-40, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24740962

ABSTRACT

Wolmer-Solberg et al., reported that six human neuroblastoma cell lines and the vast majority of clinical neuroblastoma samples contained HCMV DNA and expressed HCMV proteins. We could not replicate the data and therefore remain skeptical towards the prevalence of HCMV DNA in neuroblastomas.


Subject(s)
Brain Neoplasms/virology , Cytomegalovirus/isolation & purification , Medulloblastoma/virology , Animals , Female , Humans , Male
10.
BMC Cancer ; 12: 508, 2012 Nov 07.
Article in English | MEDLINE | ID: mdl-23134812

ABSTRACT

BACKGROUND: Medulloblastoma is the most common malignant brain tumor of childhood. Although the clinical outcome for medulloblastoma patients has improved significantly, children afflicted with the disease frequently suffer from debilitating side effects related to the aggressive nature of currently available therapy. Alternative means for treating medulloblastoma are desperately needed. We have previously shown that oncolytic measles virus (MV) can selectively target and destroy medulloblastoma tumor cells in localized and disseminated models of the disease. MV-NIS, an oncolytic measles virus that encodes the human thyroidal sodium iodide symporter (NIS), has the potential to deliver targeted radiotherapy to the tumor site and promote a localized bystander effect above and beyond that achieved by MV alone. METHODS: We evaluated the efficacy of MV-NIS against medulloblastoma cells in vitro and examined their ability to incorporate radioiodine at various timepoints, finding peak uptake at 48 hours post infection. The effects of MV-NIS were also evaluated in mouse xenograft models of localized and disseminated medulloblastoma. Athymic nude mice were injected with D283med-Luc medulloblastoma cells in the caudate putamen (localized disease) or right lateral ventricle (disseminated disease) and subsequently treated with MV-NIS. Subsets of these mice were given a dose of 131I at 24, 48 or 72 hours later. RESULTS: MV-NIS treatment, both by itself and in combination with 131I, elicited tumor stabilization and regression in the treated mice and significantly extended their survival times. Mice given 131I were found to concentrate radioiodine at the site of their tumor implantations. In addition, mice with localized tumors that were given 131I either 24 or 48 hours after MV-NIS treatment exhibited a significant survival advantage over mice given MV-NIS alone. CONCLUSIONS: These data suggest MV-NIS plus radioiodine may be a potentially useful therapy for the treatment of medulloblastoma.


Subject(s)
Iodine Radioisotopes/therapeutic use , Measles virus/physiology , Medulloblastoma/therapy , Oncolytic Viruses/physiology , Symporters/physiology , Animals , Biological Transport/physiology , Bystander Effect/radiation effects , Cell Line, Tumor , Chlorocebus aethiops , Combined Modality Therapy , Giant Cells/metabolism , Giant Cells/radiation effects , Giant Cells/virology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Iodine Radioisotopes/pharmacokinetics , Luminescent Measurements/methods , Measles virus/genetics , Medulloblastoma/pathology , Medulloblastoma/virology , Mice , Mice, Nude , Oncolytic Viruses/genetics , Survival Analysis , Symporters/genetics , Thyroid Gland/metabolism , Treatment Outcome , Vero Cells , Xenograft Model Antitumor Assays
11.
PLoS One ; 7(4): e35054, 2012.
Article in English | MEDLINE | ID: mdl-22496891

ABSTRACT

Recent studies have reported the detection of the human neurotropic virus, JCV, in a significant population of brain tumors, including medulloblastomas. Accordingly, expression of the JCV early protein, T-antigen, which has transforming activity in cell culture and in transgenic mice, results in the development of a broad range of tumors of neural crest and glial origin. Evidently, the association of T-antigen with a range of tumor-suppressor proteins, including p53 and pRb, and signaling molecules, such as ß-catenin and IRS-1, plays a role in the oncogenic function of JCV T-antigen. We demonstrate that T-antigen expression is suppressed by glucose deprivation in medulloblastoma cells and in glioblastoma xenografts that both endogenously express T-antigen. Mechanistic studies indicate that glucose deprivation-mediated suppression of T-antigen is partly influenced by 5'-activated AMP kinase (AMPK), an important sensor of the AMP/ATP ratio in cells. In addition, glucose deprivation-induced cell cycle arrest in the G1 phase is blocked with AMPK inhibition, which also prevents T-antigen downregulation. Furthermore, T-antigen prevents G1 arrest and sustains cells in the G2 phase during glucose deprivation. On a functional level, T-antigen downregulation is partially dependent on reactive oxygen species (ROS) production during glucose deprivation, and T-antigen prevents ROS induction, loss of ATP production, and cytotoxicity induced by glucose deprivation. Additionally, we have found that T-antigen is downregulated by the glycolytic inhibitor, 2-deoxy-D-glucose (2-DG), and the pentose phosphate inhibitors, 6-aminonicotinamide and oxythiamine, and that T-antigen modulates expression of the glycolytic enzyme, hexokinase 2 (HK2), and the pentose phosphate enzyme, transaldolase-1 (TALDO1), indicating a potential link between T-antigen and metabolic regulation. These studies point to the possible involvement of JCV T-antigen in medulloblastoma proliferation and the metabolic phenotype and may enhance our understanding of the role of viral proteins in glycolytic tumor metabolism, thus providing useful targets for the treatment of virus-induced tumors.


Subject(s)
Antigens, Viral, Tumor/metabolism , Brain Neoplasms/metabolism , Glioblastoma/metabolism , Glucose/metabolism , JC Virus/metabolism , Medulloblastoma/metabolism , 6-Aminonicotinamide/pharmacology , Animals , Antigens, Viral, Tumor/analysis , Antimetabolites/pharmacology , Brain Neoplasms/drug therapy , Brain Neoplasms/virology , Cell Cycle Checkpoints/drug effects , Cricetinae , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/metabolism , Deoxyglucose/pharmacology , Down-Regulation , Glioblastoma/drug therapy , Glioblastoma/virology , Glycolysis/drug effects , Hexokinase/biosynthesis , Humans , JC Virus/drug effects , Medulloblastoma/drug therapy , Medulloblastoma/virology , Metabolic Networks and Pathways/drug effects , Mice , Oxythiamine/pharmacology , Pentose Phosphate Pathway/drug effects , Reactive Oxygen Species/metabolism , Transaldolase/biosynthesis
12.
J Clin Invest ; 121(10): 3831-3, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21968105

ABSTRACT

Medulloblastoma is the most common malignant brain tumor in children. Overall survival rates have improved in recent years as a result of risk-stratified treatment regimens. However, medulloblastoma remains associated with substantial mortality, and survivors often experience debilitating neurological, endocrinological, and social sequelae as a result of treatment. Targeted and less toxic therapeutic strategies are therefore needed. In this issue of the JCI, Baryawno et al. report their findings that a large percentage of primary medulloblastomas and medulloblastoma cell lines are infected with human cytomegalovirus (HCMV) and suggest that targeting this virus could provide a new way to treat individuals with medulloblastoma.


Subject(s)
Brain Neoplasms/virology , Cytomegalovirus/isolation & purification , Medulloblastoma/virology , Animals , Female , Humans , Male
13.
J Clin Invest ; 121(10): 4043-55, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21946257

ABSTRACT

Medulloblastomas are the most common malignant brain tumors in children. They express high levels of COX-2 and produce PGE2, which stimulates tumor cell proliferation. Human cytomegalovirus (HCMV) is prevalent in the human population and encodes proteins that provide immune evasion strategies and promote oncogenic transformation and oncomodulation. In particular, HCMV induces COX-2 expression; STAT3 phosphorylation; production of PGE2, vascular endothelial growth factor, and IL-6; and tumor formation in vivo. Here, we show that a large proportion of primary medulloblastomas and medulloblastoma cell lines are infected with HCMV and that COX-2 expression, along with PGE2 levels, in tumors is directly modulated by the virus. Our analysis indicated that both HCMV immediate-early proteins and late proteins are expressed in the majority of primary medulloblastomas. Remarkably, all of the human medulloblastoma cell lines that we analyzed contained HCMV DNA and RNA and expressed HCMV proteins at various levels in vitro. When engrafted into immunocompromised mice, human medulloblastoma cells induced expression of HCMV proteins. HCMV and COX-2 expression correlated in primary tumors, cell lines, and medulloblastoma xenografts. The antiviral drug valganciclovir and the specific COX-2 inhibitor celecoxib prevented HCMV replication in vitro and inhibited PGE2 production and reduced medulloblastoma tumor cell growth both in vitro and in vivo. Ganciclovir did not affect the growth of HCMV-negative tumor cell lines. These findings imply an important role for HCMV in medulloblastoma and suggest HCMV as a novel therapeutic target for this tumor.


Subject(s)
Brain Neoplasms/virology , Cytomegalovirus/isolation & purification , Medulloblastoma/virology , Adult , Animals , Antiviral Agents/pharmacology , Brain Neoplasms/drug therapy , Celecoxib , Cell Line, Tumor , Child , Child, Preschool , Cyclooxygenase 2 Inhibitors/pharmacology , Cytomegalovirus/drug effects , Cytomegalovirus/pathogenicity , Cytomegalovirus/physiology , Dinoprostone/biosynthesis , Female , Ganciclovir/analogs & derivatives , Ganciclovir/pharmacology , Humans , Infant , Male , Medulloblastoma/drug therapy , Mice , Mice, Nude , Middle Aged , Pyrazoles/pharmacology , Sulfonamides/pharmacology , Valganciclovir , Virus Replication/drug effects , Xenograft Model Antitumor Assays , Young Adult
14.
J Med Virol ; 83(9): 1608-13, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21739452

ABSTRACT

A total of 111 fresh brain biopsies from patients with primary brain tumors were examined for JC polyomavirus sequences from the Large T antigen encoding region (LT) and the viral non-coding control region (NCCR). SYBR Green and TaqMan real-time polymerase chain reaction assays were used. In the glioblastoma group of 39 patients 48.7% were positive for LT sequences. Among the astrocytoma group (19 patients) and the oligodendroglioma group (12 patients) 31.6% and 33.3% were also positive. The prevalence of LT genomic sequences among the other groups was as follows: in 2 out of 3 oligoastrocytomas; in 3 out 5 gangliogliomas; in 2 out of 5 meduloblastomas; in 1 out 3 pineocytomas; and in none of the tested 5 ependimomas. All positive samples had a late threshold cycle that varied from 36 to 49, indicative of very low starting viral number. Only 21 of all the 111 samples were positive for NCCR. Low copy number in range of 10-1,000 was present. Notably, only 8 of all NCCR positive specimens were also LT positive. It might be suggested that the disproportion between the results for LT and NCCR is either due to clonally integrated LT fragments, with loss of genetic material, or changes in the NCCR. The latter would alter the productive course of the infection and may establish a premise for continuous interaction of viral regulatory proteins with cell molecules that are responsible for the control of the cell cycle. This may lead subsequently to malignant transformation.


Subject(s)
Antigens, Polyomavirus Transforming/genetics , Brain Neoplasms/virology , JC Virus/genetics , RNA, Untranslated/genetics , Adolescent , Adult , Aged , Astrocytoma/virology , Base Sequence , Benzothiazoles , Bulgaria , Child , Child, Preschool , DNA, Viral/analysis , Diamines , Ependymoma/virology , Female , Ganglioglioma/virology , Glioblastoma/virology , Humans , Male , Medulloblastoma/virology , Middle Aged , Molecular Sequence Data , Oligodendroglioma/virology , Organic Chemicals , Pinealoma/virology , Polymerase Chain Reaction , Quinolines , Sequence Analysis, DNA
15.
Neuro Oncol ; 12(10): 1034-42, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20494960

ABSTRACT

Although treatment of medulloblastoma has improved, at least 30% of patients with this tumor die of progressive disease. Unfortunately, many of the children who survive suffer long-term treatment-related morbidity. Previous studies have demonstrated the efficacy of using oncolytic viruses to eradicate brain tumors. The objective of this study was to test the efficacy of measles virus in treating medulloblastoma. To determine whether medulloblastoma cells are susceptible, 5 different human medulloblastoma cell lines were analyzed for the expression of the measles virus receptor CD46. Fluorescence-activated cell-sorting analysis confirmed expression of CD46 on all cell lines tested, with UW288-1 having the most prominent expression and D283med displaying the lowest expression. CD46 expression was also demonstrated, using immunohistochemistry, in 13 of 13 medulloblastoma tissue specimens. All 5 medulloblastoma cell lines were examined for their susceptibility to measles virus killing in vitro. A measles virus containing the green fluorescent protein (GFP) gene as a marker for infection (MV-GFP) was used. All cell lines exhibited significant killing when infected with MV-GFP, all formed syncytia with infection, all showed fluorescence, and all allowed viral replicaton after infection. In an intracerebral murine xenograft model, a statistically significant increase in survival was seen in animals treated with the active measles virus compared with those treated with inactivated virus. These data demonstrate that medulloblastoma is susceptible to measles virus killing and that the virus may have a role in treating this tumor in the clinical setting.


Subject(s)
Antineoplastic Agents/therapeutic use , Cerebellar Neoplasms/therapy , Medulloblastoma/therapy , Oncolytic Virotherapy/methods , Animals , Cell Line, Tumor , Cell Separation , Cerebellar Neoplasms/virology , Flow Cytometry , Humans , Immunohistochemistry , Measles virus , Medulloblastoma/virology , Membrane Cofactor Protein/biosynthesis , Mice , Mice, Nude , Xenograft Model Antitumor Assays
16.
J Cell Biochem ; 110(1): 182-90, 2010 May.
Article in English | MEDLINE | ID: mdl-20336668

ABSTRACT

The formation and progression of mudulloblastoma (MB) is poorly understood. However, somatic inactivation of pRb/p105, in combination with a somatic or a germ-line TP53 inactivation, leads to MB in a mouse model. Presently, there is no specific evidence of pathway/s alterations for the other two members of the retinoblastoma family, pRb2/p130 and/or p107 in MB. JC virus (JCV) is a human polyomavirus. Although there is no firm evidence that this virus plays a causal role in human neoplasia, it has been clearly proven that JCV is highly oncogenic when injected into the brain of experimental animals. The mechanism of JCV-induced tumorigenesis is not entirely clear. However, several studies relate the oncogenic properties of JCV mainly to its early protein large T-antigen (T-Ag), which is able to bind and inactivate both TP53 and Rb family proteins. Here, we compared the protein expression profiles of p53, p73, pRb family proteins, and PCNA, as main regulators of cell proliferation and death, in different cell lines of mouse primitive neuroectodermal tumors (PNET), either T-Ag-positive or -negative, and in human MB cell lines. Our goal was to determine if changes in the relative expression of these regulators could trigger molecular perturbations underlying MB pathogenesis in mouse and human cells. Our results support that the presence of JCV T-Ag may interfere with the expression of pRb family proteins, specific p73 isoforms, and p53. In turn, this "perturbation" may trigger a network of signals strictly connected with survival and apoptosis.


Subject(s)
Antigens, Viral, Tumor/immunology , DNA-Binding Proteins/metabolism , JC Virus/immunology , Medulloblastoma/metabolism , Nuclear Proteins/metabolism , Retinoblastoma Protein/metabolism , Signal Transduction , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Cell Line, Tumor , Child, Preschool , Fibroblasts/metabolism , Gene Expression Profiling , Humans , Male , Medulloblastoma/virology , Mice , Neoplasm Proteins/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Tumor Protein p73
17.
Antiviral Res ; 85(3): 504-11, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20035788

ABSTRACT

IFN-stimulated gene 15 (ISG15), an ubiquitin-like protein, is rapidly induced by IFN-alpha/beta, and ISG15 conjugation is associated with the antiviral immune response. Japanese encephalitis virus (JEV), a mosquito-borne neurotropic flavivirus, causes severe central nervous system diseases. We investigated the potential anti-JEV effect of ISG15 over-expression. ISG15 over-expression in human medulloblastoma cells significantly reduced the JEV-induced cytopathic effect and inhibited JEV replication by reducing the viral titers and genomes (p<0.05, Student's t-test); it also increased activation of the interferon stimulatory response element (ISRE)-luciferase cis-acting reporter in JEV-infected cells (p<0.05, Chi-square test). Furthermore, Western blotting revealed that ISG15 over-expression increased phosphorylation of IRF-3 (Ser396), JAK2 (Tyr1007/1008) and STAT1 (Tyr701 and Ser727) in JEV-infected cells (P<0.05, Chi-square test). Confocal imaging indicated that nucleus translocation of transcription factor STAT1 occurred in ISG15-over-expressing cells but not in vector control cells post-JEV infection. ISG15 over-expression activated the expression of STAT1-dependent genes including IRF-3, IFN-beta, IL-8, PKR and OAS before and post-JEV infection (p=0.063, Student's t-test). The results enabled elucidation of the molecular mechanism of ISG15 over-expression against JEV, which will be useful for developing a novel treatment to combat JEV infection.


Subject(s)
Cytokines/biosynthesis , Cytokines/immunology , Encephalitis Virus, Japanese/growth & development , Encephalitis Virus, Japanese/immunology , Medulloblastoma/virology , Ubiquitins/biosynthesis , Ubiquitins/immunology , Cell Line, Tumor , Cytopathogenic Effect, Viral , Gene Expression , Humans , Signal Transduction , Viral Load
18.
Indian J Pathol Microbiol ; 52(1): 42-5, 2009.
Article in English | MEDLINE | ID: mdl-19136778

ABSTRACT

BACKGROUND: The human polyoma virus, also known as the JC virus (JCV), replicates predominantly in the oligodendrocytes, the myelin producing cells in the central nervous system and results in the fatal demyelinating disease progressive multifocal leukoencephalopathy (PML) especially in immunosuppressed patients with AIDS. Several investigators have also documented the presence of the viral genome and early and late antigens in a variety of brain tumors particularly in medulloblastomas, gliomas and ependymomas. Reports also indicate the presence of JCV in patients with colon cancer. The T antigen of JCV has been postulated to have oncogenic potential as substantiated by animal experiments. Although JCV infects 80% of the population, there are scant epidemiological studies regarding JCV from India. There are also reports of the low prevalence of PML in patients with AIDS from India and Africa. AIM: This study was undertaken to investigate if Indian children with medulloblastomas also show evidence of JCV. METHODS: Twenty-two consecutive cases of medulloblastomas were investigated for the presence of T antigen and agnoprotein of JCV in biopsy specimens by immunohistochemistry. Antibodies to the agnoprotein antigen raised in rabbits and a monoclonal antibody against SV40 T antigen raised in mice that cross-reacts with JCV T antigen were used. RESULTS: Out of 22 patients, 4 had desmoplastic tumors while the rest had classical tumors. All children were below the age of 10. Results indicate that while PML tissues showed consistent immunostaining both with antibody to T antigen and agnoprotein antibody, none of the tumors showed any positive staining for JC viral antigens. CONCLUSION: JCV antigens could not be detected by immunohistochemistry in the tumor tissues of Indian children with medulloblastomas.


Subject(s)
Antigens, Polyomavirus Transforming/analysis , JC Virus/chemistry , Medulloblastoma/virology , Viral Regulatory and Accessory Proteins/analysis , Animals , Antibodies, Monoclonal , Antibodies, Viral , Biopsy , Brain/pathology , Child , Child, Preschool , Humans , Immunohistochemistry , India , Mice , Rabbits
19.
Cancer Res ; 67(18): 8818-27, 2007 Sep 15.
Article in English | MEDLINE | ID: mdl-17875723

ABSTRACT

We have shown previously the oncolytic potential of myxoma virus in a murine xenograft model of human glioma. Here, we show that myxoma virus used alone or in combination with rapamycin is effective and safe when used in experimental models of medulloblastoma in vitro and in vivo. Nine of 10 medulloblastoma cell lines tested were susceptible to lethal myxoma virus infection, and pretreatment of cells with rapamycin increased the extent of in vitro oncolysis. Intratumoral injection of live myxoma virus when compared with control inactivated virus prolonged survival in D341 and Daoy orthotopic human medulloblastoma xenograft mouse models [D341 median survival: 21 versus 12.5 days; P = 0.0008; Daoy median survival: not reached (three of five mice apparently "cured" after 223 days) versus 75 days; P = 0.0021]. Rapamycin increased the extent of viral oncolysis, "curing" most Daoy tumor-bearing mice and reducing or eliminating spinal cord and ventricle metastases. Rapamycin enhanced tumor-specific myxoma virus replication in vivo and prolonged survival of D341 tumor-bearing mice (median survival of mice treated with live virus (LV) and rapamycin, versus LV alone, versus rapamycin alone, versus inactivated virus: 25 days versus 19, 13, and 11 days, respectively; P < 0.0001). Rapamycin increased the levels of constitutively activated Akt in Daoy and D341 cells, which may explain its ability to enhance myxoma virus oncolysis. These observations suggest that myxoma virus may be an effective oncolytic agent against medulloblastoma and that combination therapy with signaling inhibitors that modulate activity of the phosphatidylinositol 3-kinase/Akt pathway will further enhance the oncolytic potential of myxoma virus.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Medulloblastoma/therapy , Myxoma virus/physiology , Oncolytic Virotherapy/methods , Sirolimus/pharmacology , Animals , Combined Modality Therapy , Enzyme Activation/drug effects , Humans , Injections, Intralesional , Medulloblastoma/drug therapy , Medulloblastoma/virology , Mice , Mice, Nude , NIH 3T3 Cells , Neoplasm Metastasis , Oncogene Protein v-akt/metabolism , Virus Replication/drug effects , Xenograft Model Antitumor Assays
20.
Neuropathol Appl Neurobiol ; 32(2): 131-40, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16599942

ABSTRACT

JC virus (JCV), the agent of progressive multifocal leucoencephalopathy (PML), exerts an oncogenic effect in several laboratory animal models. Moreover, JCV genomic DNA and early viral protein T-antigen have been detected in various types of human central nervous system (CNS) neoplasms. To further explore this association we have studied paraffin-embedded brain biopsy tissue from 60 neoplasms (55 gliomas and five medulloblastomas) and 15 reactive gliosis cases for the presence of JCV DNA sequences and proteins. Four post mortem cases of HIV-associated PML were used as positive controls. Samples were assessed by polymerase chain reaction (PCR) amplification of early (large T antigen) and late (virion protein 3) sequences and immunohistochemistry (IHC) with both PAb 2024 and anti-SV40 large T antigen monoclonal antibodies. Five cases (three neoplasms and two reactive gliosis instances) showed low viral DNA levels when PCR-tested for VP3 or large T, while no case was immunoreactive for any of the two antibodies used. The four PML cases yielded positive results with both PCR and IHC. Additionally, IHC with both antibodies was applied to a tissue micro-array including 109 CNS tumours and 21 reactive gliosis samples. No immunoreactivity was detected in any of these tissue micro-array samples. The rarity of JCV DNA sequences and early proteins in our brain tumours enriches the controversy over the role of JCV in human neurooncogenesis, whose clarification is in need of further molecular and epidemiologic studies.


Subject(s)
Brain Neoplasms/virology , DNA, Viral/isolation & purification , Glioma/virology , JC Virus/genetics , Medulloblastoma/virology , Adult , Animals , Antigens, Viral, Tumor/isolation & purification , Cell Transformation, Neoplastic , Child , Female , Humans , Immunohistochemistry , Male , Middle Aged , Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...