Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 487
Filter
1.
CNS Neurosci Ther ; 30(6): e14822, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38923860

ABSTRACT

INTRODUCTION: Despite the extensive neurological symptoms induced by COVID-19 and the identification of SARS-CoV-2 in post-mortem brain samples from COVID-19 patients months after death, the precise mechanisms of SARS-CoV-2 invasion into the central nervous system remain unclear due to the lack of research models. METHODS: We collected glioma tissue samples from glioma patients who had a recent history of COVID-19 and examined the presence of the SARS-CoV-2 spike protein. Subsequently, spatial transcriptomic analyses were conducted on normal brain tissues, glioma tissues, and glioma tissues from glioma patients with recent COVID-19 history. Additionally, single-cell sequencing data from both glioma tissues and glioma organoids were collected and analyzed. Glioma organoids were utilized to evaluate the efficacy of potential COVID-19 blocking agents. RESULTS: Glioma tissues from glioma patients with recent COVID-19 history exhibited the presence of the SARS-CoV-2 spike protein. Differences between glioma tissues from glioma patients who had a recent history of COVID-19 and healthy brain tissues primarily manifested in neuronal cells. Notably, neuronal cells within glioma tissues of COVID-19 history demonstrated heightened susceptibility to Alzheimer's disease, depression, and synaptic dysfunction, indicative of neuronal aberrations. Expressions of SARS-CoV-2 entry factors were confirmed in both glioma tissues and glioma organoids. Moreover, glioma organoids were susceptible to pseudo-SARS-CoV-2 infection and the infections could be partly blocked by the potential COVID-19 drugs. CONCLUSIONS: Gliomas had inherent traits that render them susceptible to SARS-CoV-2 infection, leading to their representability of COVID-19 neurological symptoms. This established a biological foundation for the rationality and feasibility of utilization of glioma organoids as research and blocking drug testing model in SARS-CoV-2 infection within the central nervous system.


Subject(s)
Brain Neoplasms , COVID-19 , Glioma , Organoids , SARS-CoV-2 , Humans , Glioma/pathology , Glioma/virology , COVID-19/complications , COVID-19/pathology , Organoids/virology , Brain Neoplasms/pathology , Brain Neoplasms/virology , Spike Glycoprotein, Coronavirus/metabolism , Male , Female , Middle Aged , Brain/pathology , Brain/virology , Brain/metabolism
2.
Int J Mol Sci ; 25(12)2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38928376

ABSTRACT

SARS-CoV-2 is the causative agent of the COVID-19 pandemic, the acute respiratory disease which, so far, has led to over 7 million deaths. There are several symptoms associated with SARS-CoV-2 infections which include neurological and psychiatric disorders, at least in the case of pre-Omicron variants. SARS-CoV-2 infection can also promote the onset of glioblastoma in patients without prior malignancies. In this study, we focused on the Envelope protein codified by the virus genome, which acts as viroporin and that is reported to be central for virus propagation. In particular, we characterized the electrophysiological profile of E-protein transfected U251 and HEK293 cells through the patch-clamp technique and FURA-2 measurements. Specifically, we observed an increase in the voltage-dependent (Kv) and calcium-dependent (KCa) potassium currents in HEK293 and U251 cell lines, respectively. Interestingly, in both cellular models, we observed a depolarization of the mitochondrial membrane potential in accordance with an alteration of U251 cell growth. We, therefore, investigated the transcriptional effect of E protein on the signaling pathways and found several gene alterations associated with apoptosis, cytokines and WNT pathways. The electrophysiological and transcriptional changes observed after E protein expression could explain the impact of SARS-CoV-2 infection on gliomagenesis.


Subject(s)
COVID-19 , Glioblastoma , Membrane Potential, Mitochondrial , SARS-CoV-2 , Humans , Glioblastoma/metabolism , Glioblastoma/virology , Glioblastoma/pathology , Glioblastoma/genetics , HEK293 Cells , SARS-CoV-2/physiology , COVID-19/virology , COVID-19/metabolism , Cell Line, Tumor , Coronavirus Envelope Proteins/metabolism , Coronavirus Envelope Proteins/genetics , Apoptosis , Brain Neoplasms/metabolism , Brain Neoplasms/virology , Brain Neoplasms/pathology , Brain Neoplasms/genetics
3.
Viruses ; 16(4)2024 04 03.
Article in English | MEDLINE | ID: mdl-38675903

ABSTRACT

Gliomas account for approximately 75-80% of all malignant primary tumors in the central nervous system (CNS), with glioblastoma multiforme (GBM) considered the deadliest. Despite aggressive treatment involving a combination of chemotherapy, radiotherapy, and surgical intervention, patients with GBM have limited survival rates of 2 to 5 years, accompanied by a significant decline in their quality of life. In recent years, novel management strategies have emerged, such as immunotherapy, which includes the development of vaccines or T cells with chimeric antigen receptors, and oncolytic virotherapy (OVT), wherein wild type (WT) or genetically modified viruses are utilized to selectively lyse tumor cells. In vitro and in vivo studies have shown that the Zika virus (ZIKV) can infect glioma cells and induce a robust oncolytic activity. Consequently, interest in exploring this virus as a potential oncolytic virus (OV) for high-grade gliomas has surged. Given that ZIKV actively circulates in Colombia, evaluating its neurotropic and oncolytic capabilities holds considerable national and international importance, as it may emerge as an alternative for treating highly complex gliomas. Therefore, this literature review outlines the generalities of GBM, the factors determining ZIKV's specific tropism for nervous tissue, and its oncolytic capacity. Additionally, we briefly present the progress in preclinical studies supporting the use of ZIKV as an OVT for gliomas.


Subject(s)
Brain Neoplasms , Glioma , Oncolytic Virotherapy , Oncolytic Viruses , Zika Virus , Animals , Humans , Brain Neoplasms/therapy , Brain Neoplasms/virology , Glioblastoma/therapy , Glioblastoma/virology , Glioma/therapy , Glioma/virology , Oncolytic Virotherapy/methods , Oncolytic Viruses/genetics , Oncolytic Viruses/physiology , Zika Virus/physiology , Zika Virus Infection/virology
4.
Turk Neurosurg ; 34(3): 448-452, 2024.
Article in English | MEDLINE | ID: mdl-38650560

ABSTRACT

AIM: To demonstrate if the human cytomegalovirus (HCMV) genome, that is involved in the pathogenesis of gliomas, is part of the genomic DNA of glioma cells or not. MATERIAL AND METHODS: The study included U87MG glioblastoma cell culture and tumor samples from glioma patients. The genomic DNA of tumor samples and U87MG cells were extracted and real-time quantitative PCR was used to assess the presence of the human cytomegalovirus genomic DNA. RESULTS: Consequently, HCMV positivity was not detected in the tumor and cell line genomic DNA under the aforementioned experimental conditions. CONCLUSION: We found that the genomic DNA of all the samples was negative for HCMV genomic DNA. Thus, HCMV could not be detected in human glioma tumors and we put forward that HCMV genomic DNA was not incorporated into the genomic DNA of glioma cells. Thus, total viral DNA is not involved in the pathogenesis of glioma; however, small viral particles or specific genes might be incorporated into the genomic DNA of glioma cells, leading to cancer development. This prompts further studies for verification.


Subject(s)
Brain Neoplasms , Cytomegalovirus , DNA, Viral , Genome, Viral , Glioma , Humans , Cytomegalovirus/genetics , Cytomegalovirus/isolation & purification , DNA, Viral/analysis , DNA, Viral/genetics , Glioma/virology , Glioma/genetics , Cell Line, Tumor , Brain Neoplasms/virology , Brain Neoplasms/genetics , Male , Female , Cytomegalovirus Infections/virology , Middle Aged , Real-Time Polymerase Chain Reaction , Adult
5.
J Biol Chem ; 299(2): 102836, 2023 02.
Article in English | MEDLINE | ID: mdl-36572185

ABSTRACT

Gap junctional intercellular communication (GJIC) involving astrocytes is important for proper CNS homeostasis. As determined in our previous studies, trafficking of the predominant astrocyte GJ protein, Connexin43 (Cx43), is disrupted in response to infection with a neurotropic murine ß-coronavirus (MHV-A59). However, how host factors are involved in Cx43 trafficking and the infection response is not clear. Here, we show that Cx43 retention due to MHV-A59 infection was associated with increased ER stress and reduced expression of chaperone protein ERp29. Treatment of MHV-A59-infected astrocytes with the chemical chaperone 4-sodium phenylbutyrate increased ERp29 expression, rescued Cx43 transport to the cell surface, increased GJIC, and reduced ER stress. We obtained similar results using an astrocytoma cell line (delayed brain tumor) upon MHV-A59 infection. Critically, delayed brain tumor cells transfected to express exogenous ERp29 were less susceptible to MHV-A59 infection and showed increased Cx43-mediated GJIC. Treatment with Cx43 mimetic peptides inhibited GJIC and increased viral susceptibility, demonstrating a role for intercellular communication in reducing MHV-A59 infectivity. Taken together, these results support a therapeutically targetable ERp29-dependent mechanism where ß-coronavirus infectivity is modulated by reducing ER stress and rescuing Cx43 trafficking and function.


Subject(s)
Disease Susceptibility , Endoplasmic Reticulum , Host Microbial Interactions , Molecular Chaperones , Murine hepatitis virus , Animals , Mice , Astrocytoma/pathology , Astrocytoma/virology , Brain Neoplasms/pathology , Brain Neoplasms/virology , Cell Communication , Cell Line, Tumor , Connexin 43/metabolism , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Stress , Gap Junctions/metabolism , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Murine hepatitis virus/metabolism , Protein Transport , Transfection
6.
Virol J ; 19(1): 21, 2022 01 29.
Article in English | MEDLINE | ID: mdl-35093115

ABSTRACT

BACKGROUND: Glioblastoma is one of the most serious brain cancer. Previous studies have demonstrated that PTEN function disorder affects the causing and exacerbation of glioblastoma. Newcastle disease virus (NDV) has been studied as a cancer virotherapeutics. In this study, PTEN gene was delivered to glioblastoma by recombinant NDV (rNDV) and translated into protein at the cytoplasm of the glioblastoma. METHODS: We did comparison tests PTEN protein expression efficiency and oncolytic effect depend on the PTEN gene insertion site at the between NP and P genes and the between P and M gene. PTEN protein mRNA transcription, translation in glioblastoma cell, and functional PTEN protein effect of the rNDV in vitro and in vivo test performed using western blotting, RT-qPCR, MTT assay, and Glioblastoma xenograft animal model test. RESULTS: The result of this study demonstrates that rNDV-PTEN kills glioblastoma cells and reduces cancer tissue better than rNDV without the PTEN gene. In molecular immunological and cytological assays, PTEN expression level was high at located in the between NP and P gene, and PTEN gene was successfully delivered to the glioblastoma cell using rNDV and PTEN gene translated to functional protein and inhibits hTERT and AKT gene. CONCLUSIONS: PTEN gene enhances the oncolytic effect of the rNDV. And our study demonstrated that NP and P gene site is better than P and M gene site which is commonly and conventionally used. PTEN gene containing rNDV is a good candidate virotherapeutics for glioblastoma.


Subject(s)
Brain Neoplasms , Glioblastoma , Newcastle disease virus , Oncolytic Virotherapy , PTEN Phosphohydrolase , Animals , Brain Neoplasms/genetics , Brain Neoplasms/therapy , Brain Neoplasms/virology , Cell Line, Tumor , Heterografts , Humans , Mice , Newcastle disease virus/genetics , Newcastle disease virus/metabolism , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Phosphoric Monoester Hydrolases/metabolism
7.
Sci Rep ; 11(1): 21227, 2021 10 27.
Article in English | MEDLINE | ID: mdl-34707174

ABSTRACT

Increasing studies demonstrated that oncolytic activities of oHSV-1 are limited to the capacity of virus replicating in tumors. In order to potentiate the oHSV-1 oncolytic activity and expand the application of oHSV-1 treatment in multiple types of tumors, it is critical to explore the potential factors or mechanisms mediating tumor resistance to oHSV-1 infection. Here we evaluated the levels of oHSV-1 multiplication in various tumor cell lines and showed that glioblastoma cell line A172 had the lowest virus yields but intrinsically accumulated the highest levels of Mx2 protein. Subsequently we demonstrated that genetic depletion of Mx2 specifically enhanced oHSV-1 productive replication in A172 cells through promoting the nuclear translocation of uncoated viral genomic DNA and down-regulating innate antiviral response. In the further investigation, we found that Mx2 knockdown could alter the intrinsic mRNA accumulation of diverse sets innate immune genes in A172 cells, in particular DHX36 and MyD88. Mx2 depletion led to a decrease in mRNA levels of MyD88 and DHX36 in A172 cells and MyD88/DHX36 knockdown increased virus yield in A172 cells and decreased the production of IFNα, activation of IRF3 activity and NF-κB signaling in A172 cells. This shed new lights on understanding the roles of some intrinsic antiviral genes in oHSV-1 resistance, facilitating to offer potential targets to improve oHSV-1 oncolytic efficacy and develop candidates of biomarkers to predict the efficiency of oHSV-1 multiplication in tumors.


Subject(s)
Brain Neoplasms/virology , Glioblastoma/virology , Herpesvirus 1, Human/physiology , Myxovirus Resistance Proteins/metabolism , Virus Replication , Brain Neoplasms/metabolism , Cell Line, Tumor , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Glioblastoma/metabolism , Herpesvirus 1, Human/pathogenicity , Humans , Immunity, Innate , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/metabolism , Interferon-alpha/genetics , Interferon-alpha/metabolism , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Myxovirus Resistance Proteins/genetics
8.
Iran Biomed J ; 25(6): 426-33, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34696577

ABSTRACT

Background: Glioblastoma multiforme is the most invasive and lethal form of brain cancer with unclear etiology. Our study aimed to investigate the molecular prevalence of human cytomegalovirus (HCMV) and Epstein-Barr virus (EBV) infections in patients with glioblastoma multiforme (GBM). Methods: This case-control study was conducted on 42 FFPE brain tumor samples from GBM patients and 42 brain autopsies from subjects without neurological disorders. The presence of EBV and HCMV DNA was determined, using PCR and nested-PCR assays, respectively. Results: HCMV DNA was detected in 3 out of 42 (7.1%) of GBM samples and was absent from the control group (p = 0.07). Importantly, EBV DNA was detected in 9 out of 42 (21.4%) brain tissue specimens of GBM subjects, but again in none of the control group (p = 0.001). Conclusion: Our findings indicate that infection with EBV is associated with GBM.


Subject(s)
Brain Neoplasms/complications , Cytomegalovirus Infections/epidemiology , Cytomegalovirus/isolation & purification , Epstein-Barr Virus Infections/epidemiology , Glioblastoma/complications , Herpesvirus 4, Human/isolation & purification , Adolescent , Adult , Aged , Aged, 80 and over , Brain Neoplasms/virology , Case-Control Studies , Child , Child, Preschool , Cytomegalovirus Infections/virology , Epstein-Barr Virus Infections/virology , Female , Glioblastoma/virology , Humans , Iran/epidemiology , Male , Middle Aged , Young Adult
9.
J Neurooncol ; 154(3): 375-381, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34510329

ABSTRACT

INTRODUCTION: The Coronavirus disease 2019 (COVID-19) pandemic has uprooted healthcare systems worldwide, disrupting care and increasing dependence on alternative forms of health care delivery. It is yet to be determined how the pandemic affected neuro-oncology patient outcomes, given that the majority of even "elective" neurosurgical oncology procedures are time-sensitive. This study quantifies changes in neuro-oncological care during the height of the pandemic and investigates patient outcomes in 2020 compared to a historical control. METHODS: We performed a retrospective review of patients with malignant brain tumor diagnoses who were seen at our institution between March 13 and May 1 of 2020 and 2019. Alterations in care, including shift from in-person to telehealth, delays in evaluation and intervention, and treatment modifications were evaluated. These variables were analyzed with respect to brain tumor control and mortality. RESULTS: 112 patients from 2020 to 166 patients from 2019 were included. There was no significant difference in outcomes between the cohorts, despite significantly more treatment delays (p = 0.0160) and use of telehealth (p < 0.0001) in 2020. Patients in 2020 who utilized telehealth visits had significantly more stable tumor control than those who had office visits (p = 0.0124), consistent with appropriate use of in-person visits for patients with progression. CONCLUSIONS: Our study showed that use of telehealth and selective alterations in neuro-oncological care during the COVID-19 pandemic did not lead to adverse patient outcomes. This suggests that adaptive physician-led changes were successful and may inform management during the ongoing pandemic, especially with the emergence of the Delta variant.


Subject(s)
Brain Neoplasms/epidemiology , COVID-19/complications , SARS-CoV-2/isolation & purification , Brain Neoplasms/virology , COVID-19/transmission , COVID-19/virology , Delivery of Health Care , Female , Humans , Male , Middle Aged , New York/epidemiology , Prognosis , Retrospective Studies , Telemedicine
10.
Viruses ; 13(8)2021 08 03.
Article in English | MEDLINE | ID: mdl-34452401

ABSTRACT

Human papillomavirus (HPV) positive and negative head and neck squamous cell carcinoma (HNSCC) are known to have differential phenotypes, including the incidence and location of metastases. HPV positive (HPV+) HNSCC are more likely to metastasize to distant sites, such as the lung, brain, and skin. Among these locations, metastasis to the brain is a rare event, and little is known about specific risk factors for this phenotype. In this report, we describe two patients who developed brain metastases from HNSCC. Both patient tumors had p16INK4a overexpression, suggesting these tumors were HPV+. This was confirmed after PCR, in situ hybridization, and mass spectrometry detected the presence of HPV type 16 (HPV16) DNA, RNA and protein. To further characterize the presence of HPV16, we used a target enrichment strategy on tumor DNA and RNA to isolate the viral sequences from the brain metastases. Analysis by targeted next generation sequencing revealed that both tumors had the HPV genome integrated into the host genome at known hotspots, 8q24.21 and 14q24.1. Applying a similar target enrichment strategy to a larger cohort of HPV+ HNSCC brain metastases could help to identify biomarkers that can predict metastasis and/or identify novel therapeutic options.


Subject(s)
Brain Neoplasms/virology , DNA, Viral/genetics , Human papillomavirus 16/genetics , Oropharyngeal Neoplasms/virology , Papillomavirus Infections/complications , Squamous Cell Carcinoma of Head and Neck/virology , Virus Integration/genetics , Aged , Cohort Studies , Human papillomavirus 16/pathogenicity , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Oropharyngeal Neoplasms/diagnostic imaging , Papillomavirus Infections/virology
11.
Sci Rep ; 11(1): 9642, 2021 05 05.
Article in English | MEDLINE | ID: mdl-33953301

ABSTRACT

Glioma is an aggressive primary tumor of the brain with a poorly understood etiology. We studied the association of 4 human polyomaviruses (HPyV)-JC virus (JCV), BK virus (BKV), human polyomavirus 6 (HPyV6), and Merkel cell polyomavirus (MCPyV) with glioma risk within the Cancer Prevention Study II in the US (CPS-II) and the Janus Serum Bank in Norway. Cohort participants subsequently diagnosed with glioma from the CPS-II (n = 37) and Janus Serum Bank (n = 323), a median of 6.9 and 15.4 years after blood collection, respectively, were matched to individual controls on age, sex, and date of blood draw. Serum antibodies to the major viral capsid protein (VP1) were used to establish infection history for each polyomavirus. Odds ratios (ORs) and 95% confidence intervals (CIs) were estimated using conditional logistic regression. In the Janus Serum Bank, MCPyV infection was associated with a higher risk of glioma overall (OR: 1.56; 95% CI 1.10, 2.19). A modest, nonsignificant positive association with MCPyV infection was also observed in CPS-II (OR: 1.29; 95% CI 0.54, 3.08). In both cohorts, glioma risk was not significantly related to infection with JCV, BKV or HPyV6. The present study suggests that MCPyV infection may increase glioma risk.


Subject(s)
Brain Neoplasms/virology , Glioma/virology , Polyomavirus Infections/complications , Adult , Aged , Brain Neoplasms/epidemiology , Female , Glioma/epidemiology , Humans , Incidence , Male , Middle Aged , Norway , Risk , United States , Young Adult
12.
J Immunother Cancer ; 9(5)2021 05.
Article in English | MEDLINE | ID: mdl-33963009

ABSTRACT

BACKGROUND: Pediatric high-grade gliomas (pHGGs) are among the most common and incurable malignant neoplasms of childhood. Despite aggressive, multimodal treatment, the outcome of children with high-grade gliomas has not significantly improved over the past decades, prompting the development of innovative approaches. METHODS: To develop an effective treatment, we aimed at improving the suboptimal antitumor efficacy of oncolytic adenoviruses (OAs) by testing the combination with a gene-therapy approach using a bispecific T-cell engager (BiTE) directed towards the erythropoietin-producing human hepatocellular carcinoma A2 receptor (EphA2), conveyed by a replication-incompetent adenoviral vector (EphA2 adenovirus (EAd)). The combinatorial approach was tested in vitro, in vivo and thoroughly characterized at a molecular level. RESULTS: After confirming the relevance of EphA2 as target in pHGGs, documenting a significant correlation with worse clinical outcome of the patients, we showed that the proposed strategy provides significant EphA2-BiTE amplification and enhanced tumor cell apoptosis, on coculture with T cells. Moreover, T-cell activation through an agonistic anti-CD28 antibody further increased the activation/proliferation profiles and functional response against infected tumor cells, inducing eradication of highly resistant, primary pHGG cells. The gene-expression analysis of tumor cells and T cells, after coculture, revealed the importance of both EphA2-BiTE and costimulation in the proposed system. These in vitro observations translated into significant tumor control in vivo, in both subcutaneous and a more challenging orthotopic model. CONCLUSIONS: The combination of OA and EphA2-BiTE gene therapy strongly enhances the antitumor activity of OA, inducing the eradication of highly resistant tumor cells, thus supporting the clinical translation of the approach.


Subject(s)
Adenoviridae/genetics , Antibodies, Bispecific/genetics , Brain Neoplasms/therapy , Genetic Therapy , Glioma/therapy , Oncolytic Virotherapy , Oncolytic Viruses/genetics , Receptor, EphA2/genetics , Adenoviridae/metabolism , Adenoviridae/pathogenicity , Animals , Antibodies, Bispecific/metabolism , Apoptosis , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/virology , Cell Line, Tumor , Coculture Techniques , Cytotoxicity, Immunologic , Female , Gene Expression Regulation, Neoplastic , Genetic Vectors , Glioma/genetics , Glioma/metabolism , Glioma/virology , Humans , Lymphocyte Activation , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Mice, Inbred NOD , Mice, SCID , Neoplasm Grading , Oncolytic Viruses/metabolism , Oncolytic Viruses/pathogenicity , Receptor, EphA2/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Xenograft Model Antitumor Assays
13.
Cancer Lett ; 513: 26-35, 2021 08 10.
Article in English | MEDLINE | ID: mdl-33989707

ABSTRACT

Cytomegalovirus (CMV) is widespread in humans and has been implicated in glioblastoma (GBM) and other tumors. However, the role of CMV in GBM remains poorly understood and the mechanisms involved are not well-defined. The goal of this study was to identify candidate pathways relevant to GBM that may be modulated by CMV. Analysis of RNAseq data after CMV infection of patient-derived GBM cells showed significant upregulation of GBM-associated transcripts including the MET oncogene, which is known to play a role in a subset of GBM patients. These findings were validated in vitro in both mouse and human GBM cells. Using immunostaining and RT-PCR in vivo, we confirmed c-MET upregulation in a mouse model of CMV-driven GBM progression and in human GBM. siRNA knockdown showed that MET upregulation was dependent on CMV-induced upregulation of NF-κB signaling. Finally, proneural GBM xenografts overexpressing c-MET grew much faster in vivo than controls, suggesting a mechanism by which CMV infection of tumor cells could induce a more aggressive mesenchymal phenotype. These studies implicate the CMV-induced upregulation of c-MET as a potential mechanism involved in the effects of CMV on GBM growth.


Subject(s)
Brain Neoplasms/virology , Cytomegalovirus Infections/genetics , Glioblastoma/virology , NF-kappa B/metabolism , Proto-Oncogene Proteins c-met/metabolism , Animals , Brain Neoplasms/pathology , Cytomegalovirus Infections/pathology , Glioblastoma/pathology , Humans , Mice , Up-Regulation
14.
Neuroreport ; 32(9): 771-775, 2021 06 09.
Article in English | MEDLINE | ID: mdl-33994523

ABSTRACT

Since coronavirus disease 2019 (COVID-19) swept all over the world, several studies have shown the susceptibility of a patient with cancer to COVID-19. In this case, the removed glioblastoma multiforme (GBM)-adjacent (GBM-A), GBM-peritumor and GBM-central (GBM-C) tissues from a convalescent patient of COVID-19, who also suffered from glioblastoma meanwhile, together with GBM-A and GBM tissues from a patient without COVID-19 history as negative controls, were used for RNA ISH, electron microscopy observing and immunohistochemical staining of ACE2 and the virus antigen (N protein). The results of RNA ISH, electron microscopy observing showed that SARS-CoV-2 directly infects some cells within human GBM tissues and SARS-CoV-2 in GBM-C tissue still exists even when it is cleared elsewhere. Immunohistochemical staining of ACE2 and N protein showed that the expressions of ACE2 are significantly higher in specimens, including GBM-C tissue from COVID-19 patient than other types of tissue. The unique phenomenon suggests that the surgical protection level should be upgraded even if the patient is in a convalescent period and the pharyngeal swab tests show negative results. Furthermore, more attention should be paid to confirm whether the shelter-like phenomenon happens in other malignancies due to the similar microenvironment and high expression of ACE2 in some malignancies.


Subject(s)
Brain Neoplasms/virology , COVID-19/metabolism , Coronavirus Nucleocapsid Proteins/metabolism , Glioblastoma/virology , SARS-CoV-2/metabolism , Adult , Angiotensin-Converting Enzyme 2/metabolism , Brain Neoplasms/metabolism , Brain Neoplasms/surgery , Brain Neoplasms/ultrastructure , COVID-19/virology , COVID-19 Nucleic Acid Testing , Convalescence , Glioblastoma/metabolism , Glioblastoma/surgery , Glioblastoma/ultrastructure , Humans , In Situ Hybridization , Male , Microscopy, Electron, Transmission , Phosphoproteins/metabolism , RNA, Viral/metabolism , Receptors, Coronavirus/metabolism , SARS-CoV-2/ultrastructure , Virion/ultrastructure
15.
Cancer Lett ; 509: 26-38, 2021 07 01.
Article in English | MEDLINE | ID: mdl-33819529

ABSTRACT

Oncolytic adenovirus-mediated gene therapy shows promise for cancer treatment; however, the systemic delivery of oncolytic adenovirus to tumors remains challenging. Recently, mesenchymal stem cells (MSCs) have emerged as potential vehicles for improving delivery. Yet, because the oncolytic adenovirus replicates in MSCs, balancing MSC viability with viral load is key to achieving optimal therapeutic effect. We thus developed an all-in-one Tet-on system that can regulate replication of oncolytic adenovirus. Then, we loaded the novel oncolytic adenovirus carrying interleukin (IL)-24 and/or Endostatin in human umbilical cord blood-mesenchymal stem cells (hUCB-MSCs) for glioma therapy. In vitro assays demonstrated that this novel oncolytic adenovirus could efficiently replicate and kill glioma cells while sparing normal cells. Moreover, doxycycline effectively regulated oncolytic adenovirus replication in the hUCB-MSCs. The doxycycline induction group with dual expression of IL-24 and Endostatin exhibited significantly greater antitumor effects than other groups in a xenograft model of glioma. Thus, this strategy for systemic delivery of oncolytic adenovirus with its oncolytic activity controlled by a Tet-on system is a promising method for achieving antitumor efficacy in glioma, especially for metastatic tumors.


Subject(s)
Brain Neoplasms/therapy , Cord Blood Stem Cell Transplantation , Endostatins/biosynthesis , Genetic Therapy , Glioma/therapy , Interleukins/biosynthesis , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/virology , Oncolytic Virotherapy , Oncolytic Viruses/genetics , Animals , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/virology , Cell Death , Cell Line, Tumor , Cell Movement , Cell Proliferation , Endostatins/genetics , Female , Genetic Vectors , Glioma/genetics , Glioma/metabolism , Glioma/virology , Humans , Interleukins/genetics , Mice, Inbred BALB C , Mice, Nude , Oncolytic Viruses/growth & development , Tumor Burden , Virus Replication , Xenograft Model Antitumor Assays
16.
BMC Neurol ; 21(1): 80, 2021 Feb 18.
Article in English | MEDLINE | ID: mdl-33602163

ABSTRACT

BACKGROUND: Chronic lymphocytic infiltration with pontine perivascular enhancement responsive to steroids (CLIPPERS) is a neuro-inflammatory syndrome first described in 2010. It has a relationship with lymphoproliferative disorders that has not been fully elucidated. This case represents an unusual progression of CLIPPERS to Epstein-Barr Virus (EBV)-related lymphomatoid granulomatosis (LYG). The exact connection between CLIPPERS and LYG remains poorly understood. CASE PRESENTATION: We present a case of a 75-year-old man who was diagnosed with CLIPPERS with initial response to immunosuppression but later progressed to EBV-related LYG. EBV polymerase chain reaction (PCR) was detected in his cerebrospinal fluid (CSF), and repeat imaging revealed findings that were uncharacteristic for CLIPPERS; thereby prompting a brain biopsy which led to a diagnosis of EBV-related LYG. This case highlights the following learning points: 1) CLIPPERS cases are often part of a spectrum of lymphomatous disease, 2) CLIPPERS can be associated with EBV-related lymphoproliferative disorders such as LYG, and 3) EBV detection in CSF should prompt earlier consideration for brain biopsy in patients. CONCLUSIONS: Our case highlights the difficulty in distinguishing CLIPPERS from other steroid-responsive conditions such as neoplastic and granulomatous diseases. Given the association of CLIPPERS with EBV-related LYG as demonstrated in this case, we recommend testing for EBV in CSF for all patients with suspected CLIPPERS. An early referral for brain biopsy and treatment with rituximab should be considered for patients with suspected CLIPPERS who test positive for EBV in their CSF.


Subject(s)
Brain Diseases/complications , Brain Neoplasms/complications , Epstein-Barr Virus Infections/complications , Lymphomatoid Granulomatosis/complications , Aged , Brain Diseases/virology , Brain Neoplasms/pathology , Brain Neoplasms/virology , Herpesvirus 4, Human , Humans , Lymphomatoid Granulomatosis/pathology , Lymphomatoid Granulomatosis/virology , Male , Pons/pathology , Steroids , Syndrome
17.
Viruses ; 13(2)2021 01 22.
Article in English | MEDLINE | ID: mdl-33499370

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.


Subject(s)
Antigens, Viral, Tumor/physiology , Brain Neoplasms/virology , JC Virus/pathogenicity , Leukoencephalopathy, Progressive Multifocal/virology , Animals , Antigens, Viral, Tumor/genetics , Brain/metabolism , Brain Neoplasms/pathology , Capsid Proteins/genetics , Disease Models, Animal , Humans , JC Virus/genetics , Leukoencephalopathy, Progressive Multifocal/pathology , Mice , Mice, Transgenic , Viral Proteins/genetics
18.
Asian Pac J Cancer Prev ; 22(1): 257-266, 2021 Jan 01.
Article in English | MEDLINE | ID: mdl-33507707

ABSTRACT

INTRODUCTION: Central nervous system tumors are a diverse group of tumors that account for 2% of all adult cancers and 17% of childhood malignancies. Several internal and external risk factors are involved in the development of this cancer such as viral infections. The aim of this study was to the determination of the EBV infection frequency and the expression level of miR-122 and miR-BART in CNS tumors samples. METHODS: One hundred and thirty-eight  fresh tissue sample (106 case and 32 control) was collected from CNS specimens. The presence of Epstein-Barr virus (EBV) DNA was examined by PCR assay and the expression level of miR-122 and miR-BART were evaluated by using real-time PCR assay in CNS tissue samples. RESULTS: EBV DNA was detected in 17% (18 of 106) of tumors tissue samples and 6.4% (2 of 32) of control samples. according to results, there was a significant relationship between the presence of EBV-DNA with CNS tumors. Additionally, the expression level of miR-122 was significantly downregulated in the EBV-positive sample compared to that of the EBV-negative sample. Also, the level of EBV-BART1-3p expression was significantly higher in EBV-positive tumors samples than EBV-positive normal samples. CONCLUSION: The results of this study suggest that the EBV could change the condition of cancer cells by altering the expression of miR-122 and EBV-BART1-3p and maybe contribute to the development of cancer cells. However, the role of viral infections in CNS cancer requires further studies. 
.


Subject(s)
Brain Neoplasms/pathology , Epstein-Barr Virus Infections/complications , Herpesvirus 4, Human/isolation & purification , MicroRNAs/genetics , RNA, Viral/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Brain Neoplasms/genetics , Brain Neoplasms/virology , Case-Control Studies , DNA, Viral/analysis , DNA, Viral/genetics , Epstein-Barr Virus Infections/genetics , Epstein-Barr Virus Infections/virology , Female , Follow-Up Studies , Humans , Male , Middle Aged , Prognosis , Young Adult
19.
J Neurovirol ; 27(1): 94-100, 2021 02.
Article in English | MEDLINE | ID: mdl-33405205

ABSTRACT

The human cytomegalovirus (HCMV) is a ubiquitous herpes virus which infects 40 to 99% of the population. HCMV reactivation may occur in the context of immunosuppression and can induce significant morbidities. Several cases of HCMV infections or HCMV reactivation have thus been reported in glioblastoma (GBM) patients treated with radio(chemo)therapy. With the aim to identify the main risk factors associated with HCMV reactivation, we reviewed all patients treated for a newly diagnosed GBM in our institution from October 2013 to December 2015. Age, sex, Karnofsky performance status (KPS), absolute lymphocyte count (ALC), serological HCMV status, and steroid doses were recorded at the start and 1 month after the end of radiotherapy (RT). Within the 103 patients analyzed, 34 patients (33%) had an initial negative serology for HCMV, and none of them developed a seroconversion after treatment. Among patients with positive HCMV IgG (n = 69), 16 patients (23%) developed a viremia at one point during treatment. Age (> 60 years), steroid intake, and ALC (< 1500/mm3) before RT were correlated with HCMV reactivation. HCMV viremia was associated with neurological decline 1 month after chemoradiotherapy but progression-free survival was not impacted. A shorter overall survival was seen in these patients when compared with the others, but this could be biased by the older age in this subgroup. HCMV reactivation needs to be sought in case of a neurological decline during RT especially in older patients treated with steroids and low lymphocytes counts.


Subject(s)
Brain Neoplasms/virology , Cytomegalovirus Infections/immunology , Glioblastoma/virology , Immunocompromised Host , Virus Activation/immunology , Adult , Aged , Aged, 80 and over , Chemoradiotherapy/adverse effects , Cytomegalovirus , Cytomegalovirus Infections/complications , Female , Glioblastoma/therapy , Humans , Latent Infection/immunology , Male , Middle Aged , Risk Factors
20.
Int J Clin Oncol ; 26(2): 305-315, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33118116

ABSTRACT

INTRODUCTION: The purpose of this study is to clarify the clinical features of temozolomide (TMZ)-related hepatitis B virus (HBV) reactivation and to identify HBV reactivation predictive factors. METHOD: We retrospectively reviewed the clinical course of 145 patients newly diagnosed or with recurrent malignant glioma treated with TMZ. Before treatment, we screened patients for HB surface antigen (HBsAg) positivity (HBV carrier) and HBsAg negativity. Patients were also screened for antibody for HB core antigen (anti-HBc) positivity and/or for HB surface antigen positivity (resolved HBV infection). The patients were monitored by HBV DNA, alanine, and aspartate aminotransaminase during and after the completion of TMZ. HBV carriers and those with resolved HBV infections with HBV reactivation received preemptive entecavir treatment. In those with resolved HBV infections, we analyzed clinical characters for the predictive factors for HBV reactivation. RESULTS: In one of two HBV carriers, HBV DNA turned positive 8 months after the completion of TMZ and entecavir. In four (16.7%) of 24 resolved HBV infections, HBV DNA turned detectable at completion of concomitant radiation and TMZ or during monthly TMZ. HBV DNA turned negative with entecavir in all patients without liver dysfunction. In resolved HBV infections, those with a high anti-HBc titer had significantly higher incidence of HBV reactivation than those with low anti-HBc titers (60% vs. 5.3%: p = 0.018). CONCLUSION: Screenings, monitoring, and preemptive entecavir were important for preventing TMZ-related HBV reactivations. Anti-HBc titers could be the predictive markers for HBV reactivation in the those with resolved HBV infections.


Subject(s)
Brain Neoplasms , Glioma , Hepatitis B virus , Hepatitis B , Temozolomide , Virus Activation , Antineoplastic Agents, Alkylating/adverse effects , Antineoplastic Agents, Alkylating/therapeutic use , Brain Neoplasms/drug therapy , Brain Neoplasms/virology , DNA, Viral , Glioma/drug therapy , Glioma/virology , Hepatitis B/drug therapy , Hepatitis B Antibodies/immunology , Hepatitis B virus/drug effects , Hepatitis B virus/immunology , Hepatitis B virus/physiology , Humans , Neoplasm Recurrence, Local , Retrospective Studies , Temozolomide/adverse effects , Temozolomide/therapeutic use , Virus Activation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...