Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 215
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.
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
3.
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
4.
Int J Mol Sci ; 22(24)2021 Dec 07.
Article in English | MEDLINE | ID: mdl-34947988

ABSTRACT

Recently, we have shown the molecular basis for lactate sensing by cervical epithelial cells resulting in enhanced DNA repair processes through DNA-PKcs regulation. Interestingly, DNA-PKcs is indispensable for proper retroviral DNA integration in the cell host genome. According to recent findings, the mucosal epithelium can be efficiently transduced by retroviruses and play a pivotal role in regulating viral release by cervical epithelial cells. This study examined the effects of lactate on lentiviral transduction in cervical cancer cells (HeLa, CaSki, and C33A) and model glioma cell lines (DNA-PKcs proficient and deficient). Our study showed that L- and D-lactate enhanced DNA-PKcs presence in nuclear compartments by between 38 and 63%, which corresponded with decreased lentiviral transduction rates by between 15 and 36%. Changes in DNA-PKcs expression or its inhibition with NU7441 also greatly affected lentiviral transduction efficacy. The stimulation of cells with either HCA1 agonist 3,5-DHBA or HDAC inhibitor sodium butyrate mimicked, in part, the effects of L-lactate. The inhibition of lactate flux by BAY-8002 enhanced DNA-PKcs nuclear localization which translated into diminished lentiviral transduction efficacy. Our study suggests that L- and D-lactate present in the uterine cervix may play a role in the mitigation of viral integration in cervical epithelium and, thus, restrict the viral oncogenic and/or cytopathic potential.


Subject(s)
DNA-Activated Protein Kinase/metabolism , Glioma/virology , Lactic Acid/pharmacology , Lentivirus/physiology , Uterine Cervical Neoplasms/virology , Benzoates/pharmacology , Butyric Acid/pharmacology , Cell Line, Tumor , Cell Nucleus/metabolism , Chromones/pharmacology , Female , Glioma/metabolism , HeLa Cells , Humans , Lentivirus/drug effects , Morpholines/pharmacology , Transduction, Genetic , Uterine Cervical Neoplasms/metabolism
5.
Sci Rep ; 11(1): 16088, 2021 08 09.
Article in English | MEDLINE | ID: mdl-34373477

ABSTRACT

Two-cycle cesium chloride (2 × CsCl) gradient ultracentrifugation is a conventional approach for purifying recombinant adenoviruses (rAds) for research purposes (gene therapy, vaccines, and oncolytic vectors). However, rAds containing the RGD-4C peptide in the HI loop of the fiber knob domain tend to aggregate during 2 × CsCl gradient ultracentrifugation resulting in a low infectious titer yield or even purification failure. An iodixanol-based purification method preventing aggregation of the RGD4C-modified rAds has been proposed. However, the reason explaining aggregation of the RGD4C-modified rAds during 2 × CsCl but not iodixanol gradient ultracentrifugation has not been revealed. In the present study, we showed that rAds with the RGD-4C peptide in the HI loop but not at the C-terminus of the fiber knob domain were prone to aggregate during 2 × CsCl but not iodixanol gradient ultracentrifugation. The cysteine residues with free thiol groups after the RGD motif within the inserted RGD-4C peptide were responsible for formation of the interparticle disulfide bonds under atmospheric oxygen and aggregation of Ad5-delta-24-RGD4C-based rAds during 2 × CsCl gradient ultracentrifugation, which could be prevented using iodixanol gradient ultracentrifugation, most likely due to antioxidant properties of iodixanol. A cysteine-to-glycine substitution of the cysteine residues with free thiol groups (RGD-2C2G) prevented aggregation during 2 × CsCl gradient purification but in coxsackie and adenovirus receptor (CAR)-low/negative cancer cell lines of human and rodent origin, this reduced cytolytic efficacy to the levels observed for a fiber non-modified control vector. However, both Ad5-delta-24-RGD4C and Ad5-delta-24-RGD2C2G were equally effective in the murine immunocompetent CT-2A glioma model due to a primary role of antitumor immune responses in the therapeutic efficacy of oncolytic virotherapy.


Subject(s)
Adenoviridae/isolation & purification , Cesium/chemistry , Chlorides/chemistry , Genetic Vectors/genetics , A549 Cells , Adenoviridae Infections/therapy , Animals , Antioxidants/chemistry , Cell Line , Cell Line, Tumor , Coxsackie and Adenovirus Receptor-Like Membrane Protein/genetics , Glioma/therapy , Glioma/virology , HEK293 Cells , Humans , Mice , Oligopeptides/genetics , Oncolytic Virotherapy/methods , Rats , Triiodobenzoic Acids/chemistry , Ultracentrifugation/methods
6.
Sci Rep ; 11(1): 15384, 2021 07 28.
Article in English | MEDLINE | ID: mdl-34321569

ABSTRACT

Determination of treatment response to immunotherapy in glioblastoma multiforme (GBM) is a process which can take months. Detection of CD8+ T cell recruitment to the tumor with a noninvasive imaging modality such as positron emission tomography (PET) may allow for tumor characterization and early evaluation of therapeutic response to immunotherapy. In this study, we utilized 89Zr-labeled anti-CD8 cys-diabody-PET to provide proof-of-concept to detect CD8+ T cell immune response to oncolytic herpes simplex virus (oHSV) M002 immunotherapy in a syngeneic GBM model. Immunocompetent mice (n = 16) were implanted intracranially with GSC005 GBM tumors, and treated with intratumoral injection of oHSV M002 or saline control. An additional non-tumor bearing cohort (n = 4) receiving oHSV M002 treatment was also evaluated. Mice were injected with 89Zr-labeled anti-CD8 cys-diabody seven days post oHSV administration and imaged with a preclinical PET scanner. Standardized uptake value (SUV) was quantified. Ex vivo tissue analyses included autoradiography and immunohistochemistry. PET imaging showed significantly higher SUV in tumors which had been treated with M002 compared to those without M002 treatment (p = 0.0207) and the non-tumor bearing M002 treated group (p = 0.0021). Accumulation in target areas, especially the spleen, was significantly reduced by blocking with the non-labeled diabody (p < 0.001). Radioactive probe accumulation in brains was consistent with CD8+ cell trafficking patterns after oHSV treatment. This PET imaging strategy could aid in distinguishing responders from non-responders during immunotherapy of GBM.


Subject(s)
CD8 Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , Glioma/therapy , Oncolytic Virotherapy/methods , Animals , CD8 Antigens/antagonists & inhibitors , CD8 Antigens/isolation & purification , CD8-Positive T-Lymphocytes/virology , Cell Line, Tumor , Disease Models, Animal , Glioma/diagnostic imaging , Glioma/immunology , Glioma/virology , Humans , Mice , Radioisotopes/pharmacology , Simplexvirus/genetics , Tomography, X-Ray Computed , Zirconium/pharmacology
7.
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
8.
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
9.
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
10.
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
11.
Cancer Invest ; 38(7): 394-405, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32643440

ABSTRACT

The study investigated the human cytomegalovirus (HCMV) and human papillomavirus (HPV) in gliomas. A retrospective study was conducted on 112 samples. HCMV was investigated by PCR, in situ hybridization (ISH) and immunohistochemistry. HPV was tested by PCR and DNA ISH. HCMV was identified in 60 gliomas, including 55 GBM. However, RNA ISH and immunohistochemistry failed to detect HCMV positivity. HPV was identified in 44 GBM. No significant relationship was identified between HCMV and HPV and tumour characteristics (p > 0.05). Our findings support the HCMV and HPV presence in gliomas. Further assays are required to more explore the potential efficient antiviral management.


Subject(s)
Brain Neoplasms/virology , Cytomegalovirus/isolation & purification , Glioma/virology , Papillomaviridae/isolation & purification , Adult , Aged , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Cytomegalovirus/genetics , Cytomegalovirus Infections/virology , Female , Glioma/mortality , Glioma/pathology , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Male , Middle Aged , Papillomaviridae/genetics , Papillomavirus Infections/virology , Polymerase Chain Reaction , Retrospective Studies , Tunisia , Young Adult
13.
BMC Cancer ; 20(1): 549, 2020 Jun 12.
Article in English | MEDLINE | ID: mdl-32532243

ABSTRACT

BACKGROUND: Glioma is the most common primary brain tumor, occurring due to the carcinogenesis of glial cells in the brain and spinal cord. Many aspects of the mechanism of its tumorigenesis remain unknown. The relationship between viral infection and glioma is one of the most important research aspects in this field. Currently, there is a lack of systematic reviews and meta-analyses to evaluate the effect of viral infection on the prognosis of glioma patients. The purpose of this study was to evaluate the relationship between viral infection and the prognosis of glioma patients, aimed at evaluating the prognostic value of the detection of viral infection. METHODS: Through careful and comprehensive retrieval of results from the PubMed, Embase, and Cochrane databases, eligible articles were selected strictly according to the inclusion and exclusion criteria. The regional sources, detection methods, detection indicators, patient survival, and other data from the samples in the papers were extracted, and the integrated analysis was conducted using Stata 15.1. We conducted a subgroup analysis of the relationship between the degree of infection and prognosis in cytomegalovirus (CMV) patients. RESULTS: A total of 11 studies were included in the analysis. Among them, 7 studies involved the relationship between CMV infection and the prognosis of patients with glioma, 2 studies involved human papillomavirus (HPV), 2 studies involved human herpesvirus-6 (HHV-6), and one study involved simian virus 40 (SV40), woolly monkey sarcoma virus (WMSV) and human endogenous retrovirus K113 (HERV-K113). In the CMV study, the pooled Hazard ratio (HR) of Overall survival (OS) was 1.024 (CI: 0.698-1.501), with a P value of 0.905. The pooled HR of Progression free survival (PFS) was 1.067 (CI: 0.770-1.478), with a P value of 0.697. The pooled HR value of low-degree infection versus high-degree infection was 1.476 (CI: 0.799-2.727), with a P value of 0.213. In the HPV study, the pooled HR of OS was 1.467 (CI: 0.552-3.901), with a P value of 0.443. CONCLUSION: CMV infection has no significant effect on the prognosis of glioma patients. Using the IEA as the detection index, the degree of CMV infection was found to have a significant impact on the prognosis of glioma patients; it was not found to possess a significant prognostic value after the integration of different indicators. Neither HPV nor HHV-6 infection has a significant effect on the prognosis of glioma patients. SV40 and WMSV infection are associated with poor prognosis in patients with low-grade glioma. TRIAL REGISTRATION: This meta-analysis registered in https://www.crd.york.ac.uk/PROSPERO/, PROSPERO ID: CRD42019127648.


Subject(s)
Glioma/mortality , Virus Diseases/epidemiology , Glioma/virology , Humans , Prognosis , Progression-Free Survival , Risk Factors , Virus Diseases/complications , Virus Diseases/virology
14.
Avian Pathol ; 49(5): 440-447, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32301629

ABSTRACT

ABSTRACT The prototype fowl glioma-inducing virus (FGVp) causes fowl glioma and cerebellar hypoplasia in chickens. In this study, we investigated whether a strain of avian leukosis virus (ALV), associated with avian osteopetrosis and mesenchymal neoplasms, is able to induce fowl glioma. We encountered avian osteopetrosis and mesenchymal neoplasms, including myxosarcoma and rhabdomyosarcoma, in Japanese native chickens used for both egg-laying and meat production. These birds were also affected by non-suppurative encephalitis and glioma in their brains. Four ALV strains (GifN_001, GifN_002, GifN_004, GifN_005) were isolated, and a phylogenic analysis of envSU showed that these isolates were classified into different clusters from FGVp and the variants previously reported. Whereas the envSU shared a high identity (94.7%) with that of Rous sarcoma virus (strain Schmidt-Ruppin B) (RSV-SRB), the identity between envTM of GifN_001 and that of FGVp was high (94.5%), indicating that GifN_strains may emerge by recombination between FGVp and other exogenous ALVs. Specific-pathogen-free chickens inoculated in ovo with GifN_001 revealed fowl glioma and cerebellar hypoplasia. These results suggest that the newly isolated strains have acquired neuropathogenicity to chickens.


Subject(s)
Avian Leukosis Virus/pathogenicity , Avian Leukosis/virology , Chickens/virology , Glioma/veterinary , Osteopetrosis/veterinary , Poultry Diseases/virology , Animals , Avian Leukosis Virus/classification , Avian Leukosis Virus/genetics , Cerebellum/abnormalities , Cerebellum/virology , Chick Embryo , Developmental Disabilities/virology , Encephalitis/veterinary , Encephalitis/virology , Female , Glioma/virology , Myxosarcoma/veterinary , Myxosarcoma/virology , Nervous System Malformations/veterinary , Nervous System Malformations/virology , Osteopetrosis/virology , Phylogeny , Recombination, Genetic , Rhabdomyosarcoma/veterinary , Rhabdomyosarcoma/virology , Specific Pathogen-Free Organisms
16.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 51(2): 159-164, 2020 Mar.
Article in Chinese | MEDLINE | ID: mdl-32220182

ABSTRACT

OBJECTIVE: To investigate the effects of enterovirus 71 (EV71) on mitochondrial dynamics in human Glioma U251 cells. METHODS: The EV71 was replicated in Vero cells and the 50% tissue culture infective dose (TCID 50) was calculated based on the Reed-Muench formula. After the U251 cells were infected with EV71, the cellular morphology was assessed through the light microscope. The mitochondrial morphology was detected by MitoTracker Deep Red staining under laser confocal microscopy and the mitochondrial ultrastructure was visualized by transmission electron microscopy. The expressions of mitochondrial fission proteins Drp1, p-Drp1 and fusion protein Opa1 were examined by Western blot. The level of ATP was measured by a commercial ATP assay kit. The generation of mitochondrial superoxide was detected by MitoSOX staining. RESULTS: The TCID 50 of EV71 was 10 -5.4/0.1 mL. Twenty-four or 48 h after EV71 infection, the U251 cells appeared shrunken, round and dead. The laser confocal microscopy and transmission electron microscopy images showed that the EV71 infection induced mitochondrial elongation and cristae damage. Moreover, Western blot analysis demonstrated that the protein expressions of Drp1 and Opa1 were downregulated at both 24 and 48 h after EV71 infection in U251 cells, companied with a significant increase in Drp1 phosphorylation at 48 h after infection ( P<0.05). In addition, a decreased ATP level and elevated mitochondrial superoxide generation were observed in the EV71 infected group, as compared to the control group. CONCLUSION: Our study demonstrated that infection with EV71 led to changes of mitochondrial morphology and dynamics in U251 cells, which may impair mitochondrial function and contribute to nervous system dysfunction.


Subject(s)
Brain Neoplasms/virology , Enterovirus A, Human , Enterovirus Infections , Enterovirus , Glioma/virology , Mitochondrial Dynamics , Animals , Chlorocebus aethiops , Enterovirus A, Human/pathogenicity , Enterovirus Infections/complications , Humans , Nervous System/physiopathology , Nervous System/virology , Tumor Cells, Cultured , Vero Cells
17.
J Neurovirol ; 26(3): 347-357, 2020 06.
Article in English | MEDLINE | ID: mdl-32124265

ABSTRACT

The gliomagenesis remains not fully established and their etiological factors still remain obscure. Polyomaviruses were detected and involved in several human tumors. Their potential implication in gliomas has been not yet surveyed in Africa and Arab World. Herein, we investigated the prevalence of six polyomaviruses (SV40, JCPyV, BKPyV, MCPyV, KIPyV, and WUPyV) in 112 gliomas from Tunisian patients. The DNA sequences of polyomaviruses were examined by PCR assays. Viral infection was confirmed by DNA in situ hybridization (ISH) and/or immunohistochemistry (IHC). The relationships between polyomavirus infection and tumor features were evaluated. Specific SV40 Tag, viral regulatory, and VP1 regions were identified in 12 GBM (10.7%). DNA ISH targeting the whole SV40 genome and SV40 Tag IHC confirmed the PCR findings. Five gliomas yielded JCPyV positivity by PCR and DNA ISH (2.7%). However, no BKPyV, KIPyV, and WUPyV DNA sequences were identified in all samples. MCPyV DNA was identified in 30 gliomas (26.8%). For GBM samples, MCPyV was significantly related to patient age (p = 0.037), tumor recurrence (p = 0.024), and SV40 (p = 0.045) infection. No further significant association was identified with the remaining tumor features (p > 0.05) and patient survival (Log Rank, p > 0.05). Our study indicates the presence of SV40, JCPyV, and MCPyV DNA in Tunisian gliomas. Further investigations are required to more elucidate the potential involvement of polyomaviruses in these destructive malignancies.


Subject(s)
Brain Neoplasms/virology , Glioma/virology , JC Virus/genetics , Merkel cell polyomavirus/genetics , Neoplasm Recurrence, Local/virology , Polyomavirus Infections/virology , Simian virus 40/genetics , Adult , Age Factors , Brain Neoplasms/genetics , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Capsid Proteins/genetics , Capsid Proteins/metabolism , DNA, Viral/genetics , DNA, Viral/metabolism , Female , Follow-Up Studies , Glioma/genetics , Glioma/mortality , Glioma/pathology , Humans , Immunohistochemistry , In Situ Hybridization , JC Virus/growth & development , JC Virus/pathogenicity , Male , Merkel cell polyomavirus/growth & development , Merkel cell polyomavirus/pathogenicity , Middle Aged , Neoplasm Grading , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/mortality , Neoplasm Recurrence, Local/pathology , Polyomavirus Infections/genetics , Polyomavirus Infections/mortality , Polyomavirus Infections/pathology , Simian virus 40/growth & development , Simian virus 40/pathogenicity , Survival Analysis , Viral Load
18.
J Virol ; 94(8)2020 03 31.
Article in English | MEDLINE | ID: mdl-32051271

ABSTRACT

Given that the Ebola virus (EBOV) infects a wide array of organs and cells yet displays a relative lack of neurotropism, we asked whether a chimeric vesicular stomatitis virus (VSV) expressing the EBOV glycoprotein (GP) might selectively target brain tumors. The mucin-like domain (MLD) of the EBOV GP may enhance virus immune system evasion. Here, we compared chimeric VSVs in which EBOV GP replaces the VSV glycoprotein, thereby reducing the neurotoxicity associated with wild-type VSV. A chimeric VSV expressing the full-length EBOV GP (VSV-EBOV) containing the MLD was substantially more effective and safer than a parallel construct with an EBOV GP lacking the MLD (VSV-EBOVΔMLD). One-step growth, reverse transcription-quantitative PCR, and Western blotting assessments showed that VSV-EBOVΔMLD produced substantially more progeny faster than VSV-EBOV. Using immunodeficient SCID mice, we focused on targeting human brain tumors with these VSV-EBOVs. Similar to the findings of our previous study in which we used an attenuated VSV-EBOV with no MLD that expressed green fluorescent protein (GFP) (VSV-EBOVΔMLD-GFP), VSV-EBOVΔMLD without GFP targeted glioma but yielded only a modest extension of survival. In contrast, VSV-EBOV containing the MLD showed substantially better targeting and elimination of brain tumors after intravenous delivery and increased the survival of brain tumor-bearing mice. Despite the apparent destruction of most tumor cells by VSV-EBOVΔMLD, the virus remained active within the SCID mouse brain and showed widespread infection of normal brain cells. In contrast, VSV-EBOV eliminated the tumors and showed relatively little infection of normal brain cells. Parallel experiments with direct intracranial virus infection generated similar results. Neither VSV-EBOV nor VSV-EBOVΔMLD showed substantive infection of the brains of normal immunocompetent mice.IMPORTANCE The Ebola virus glycoprotein contains a mucin-like domain which may play a role in immune evasion. Chimeric vesicular stomatitis viruses with the EBOV glycoprotein substituted for the VSV glycoprotein show greater safety and efficacy in targeting brain tumors in immunodeficient mice when the MLD was expressed within the EBOV glycoprotein than when EBOV lacked the mucin-like domain.


Subject(s)
Brain Neoplasms/metabolism , Ebolavirus/immunology , Glycoproteins/immunology , Hemorrhagic Fever, Ebola/virology , Mucins/immunology , Animals , Brain Neoplasms/pathology , Brain Neoplasms/virology , Cell Line, Tumor , Disease Models, Animal , Ebolavirus/genetics , Glioblastoma/virology , Glioma/pathology , Glioma/virology , Green Fluorescent Proteins , Heterografts , Humans , Mice , Mice, SCID , Mucins/genetics , Vesicular stomatitis Indiana virus/immunology
19.
J Med Virol ; 92(8): 1266-1276, 2020 08.
Article in English | MEDLINE | ID: mdl-31944314

ABSTRACT

Human cytomegalovirus (HCMV), as a ubiquitous and opportunistic virus, is a matter for consideration in broad-spectrum diseases, specifically in immunocompromised individuals. In recent decades, many studies that have evaluated the role of HCMV in inflammation and malignancies, especially in high-grade gliomas, have reported inconsistent results. Thus, this study was conducted to analyze 97 primary gliomas for human CMV UL83 gene and protein through TaqMan real-time polymerase chain reaction and immunohistochemistry, respectively. The results were positive for the UL83 gene and pp65 protein in 71% and 24% of samples, respectively. The frequency of HCMV was significantly higher in glioblastomas than other glioma grades (P < .01 and P < .05 for the UL83 gene and protein, respectively). In addition, the association between the prevalence of HCMV and aging strengthened the virus reactivation hypothesis in gliomas. In conclusion, a high frequency of HCMV infection was found in gliomas that correlated with tumor aggressiveness and age. This study recommends a thorough investigation to determine HCMV infection in gliomas to improve the existing knowledge of its role in glial tumors, its prognostic value, and possible efficient antiviral target therapy.


Subject(s)
Aging , Cytomegalovirus Infections/epidemiology , Glioma/virology , Adolescent , Adult , Aged , Child , Child, Preschool , Cytomegalovirus , DNA, Viral/analysis , Female , Glioma/epidemiology , Humans , Immunocompromised Host , Immunohistochemistry , Iran/epidemiology , Male , Middle Aged , Young Adult
20.
J Neurosurg Sci ; 64(2): 173-180, 2020 Apr.
Article in English | MEDLINE | ID: mdl-28407709

ABSTRACT

BACKGROUND: Gene therapy is regarded as a new and promising therapeutic modality for cancers, and adenovirus is one of the most frequently used vectors. However, because of low or absent coxsackievirus and adenovirus receptor levels on the surface of many kinds of tumor cells, the efficiency of adenovirus infection of target tumor cells may be low. Meanwhile, gene therapy by a single vector carrying two or more antioncogenes can improve treatment effects and reduce side effects from vectors. In this research, we aimed to detect the antitumor effect of ING4/PTEN double tumor suppressors mediated by adenovirus modified with arginine(R)-glycine(G)-aspartate(D) (RGD) on glioma cells. METHODS: We treated U87 glioma cells with PBS, blank adenovirus or adenovirus carrying RGD, ING4, PTEN, or both ING4 and PTEN, then we detected and compared the U87 cells' growth, apoptosis, and invasion. Moreover, we established a U87 glioma transplantation tumor model to study the antitumor effect in vivo by measuring the volume and weight of tumor masses in each condition. In addition, we analyzed the transcription of related genes by fluorescent quantitative PCR and detected their expression by immunohistochemistry staining to reveal the underlying mechanisms. RESULTS: The double tumor suppressors ING4/PTEN could inhibit the growth of U87 glioma cells with a synergistic antitumor effect, and the RGD modification also acted as an antioncogene to inhibit U87 cell invasion and tumor angiogenesis. CONCLUSIONS: The ING4/PTEN double tumor suppressors mediated by adenovirus modified with RGD had a significantly synergistic antitumor effect on glioma.


Subject(s)
Adenoviridae Infections/therapy , Adenoviridae/genetics , Cell Cycle Proteins/antagonists & inhibitors , Glioma/therapy , Homeodomain Proteins/antagonists & inhibitors , Membrane Proteins/antagonists & inhibitors , PTEN Phosphohydrolase/antagonists & inhibitors , Tumor Suppressor Proteins/antagonists & inhibitors , Amino Acids/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Proliferation/drug effects , Genetic Therapy/methods , Glioma/virology , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Membrane Proteins/genetics , Neovascularization, Pathologic , PTEN Phosphohydrolase/genetics , Tumor Suppressor Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL