Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 216
Filtrar
1.
Molecules ; 27(13)2022 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-35807435

RESUMO

Enterovirus 71 (EV71) is a dominant pathogenic agent that may cause severe central nervous system (CNS) diseases among infants and young children in the Asia-pacific. The inflammasome is closely implicated in EV71-induced CNS injuries through a series of signaling pathways. However, the activation pathway of NLRP3 inflammasome involved in EV71-mediated CNS injuries remains poorly defined. In the studies, EV71 infection, ERK1/2 phosphorylation, and activation of NLRP3 are abolished in glioblastoma cells with low vimentin expression by CRISPR/Cas9-mediated knockdown. PD098059, an inhibitor of p-ERK, remarkably blocks the vimentin-mediated ERK1/2 phosphorylation in EV71-infected cells. Nuclear translocation of NF-κB p65 is dependent on p-ERK in a time-dependent manner. Moreover, NLRP3 activation and caspase-1 production are limited in EV71-infected cells upon the caffeic acid phenethyl ester (CAPE) administration, an inhibitor of NF-κB, which contributes to the inflammasome regulation. In conclusion, these results suggest that EV71-mediated NLRP3 inflammasome could be activated via the VIM-ERK-NF-κB pathway, and the treatment of the dephosphorylation of ERK and NF-κB inhibitors is beneficial to host defense in EV71-infected CNS.


Assuntos
Enterovirus , Glioblastoma , NF-kappa B , Vimentina , Criança , Pré-Escolar , Enterovirus/metabolismo , Glioblastoma/genética , Glioblastoma/metabolismo , Glioblastoma/virologia , Humanos , Inflamassomos/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , NF-kappa B/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Fosforilação , Vimentina/genética , Vimentina/metabolismo
2.
Viruses ; 14(1)2022 01 07.
Artigo em Inglês | MEDLINE | ID: mdl-35062307

RESUMO

Glioblastoma (GBM) is an aggressive primary central nervous system neoplasia with limited therapeutic options and poor prognosis. Following reports of cytomegalovirus (HCMV) in GBM tumors, the anti-viral drug Valganciclovir was administered and found to significantly increase the longevity of GBM patients. While these findings suggest a role for HCMV in GBM, the relationship between them is not clear and remains controversial. Treatment with anti-viral drugs may prove clinically useful; however, their results do not explain the underlying mechanism between HCMV infection and GBM progression. We hypothesized that HCMV infection would metabolically reprogram GBM cells and that these changes would allow for increased tumor progression. We infected LN-18 GBM cells and employed a Seahorse Bioanalyzer to characterize cellular metabolism. Increased mitochondrial respiration and glycolytic rates were observed following infection. These changes were accompanied by elevated production of reactive oxygen species and lactate. Due to lactate's numerous tumor-promoting effects, we examined the impact of paracrine signaling of HCMV-infected GBM cells on uninfected stromal cells. Our results indicated that, independent of viral transmission, the secretome of HCMV-infected GBM cells was able to alter the expression of key metabolic proteins and epigenetic markers. This suggests a mechanism of action where reprogramming of GBM cells alters the surrounding tumor microenvironment to be permissive to tumor progression in a manner akin to the Reverse-Warburg Effect. Overall, this suggests a potential oncomodulatory role for HCMV in the context of GBM.


Assuntos
Infecções por Citomegalovirus/fisiopatologia , Citomegalovirus/fisiologia , Glioblastoma/metabolismo , Glioblastoma/virologia , Comunicação Parácrina , Secretoma , Linhagem Celular Tumoral , Citomegalovirus/genética , Infecções por Citomegalovirus/virologia , Glicólise , Humanos , Ácido Láctico/metabolismo , Potencial da Membrana Mitocondrial , Mitocôndrias/metabolismo , Fosforilação Oxidativa , Espécies Reativas de Oxigênio/metabolismo , Microambiente Tumoral , Replicação Viral
4.
Sci Rep ; 11(1): 21227, 2021 10 27.
Artigo em Inglês | MEDLINE | ID: mdl-34707174

RESUMO

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.


Assuntos
Neoplasias Encefálicas/virologia , Glioblastoma/virologia , Herpesvirus Humano 1/fisiologia , Proteínas de Resistência a Myxovirus/metabolismo , Replicação Viral , Neoplasias Encefálicas/metabolismo , Linhagem Celular Tumoral , RNA Helicases DEAD-box/genética , RNA Helicases DEAD-box/metabolismo , Glioblastoma/metabolismo , Herpesvirus Humano 1/patogenicidade , Humanos , Imunidade Inata , Fator Regulador 3 de Interferon/genética , Fator Regulador 3 de Interferon/metabolismo , Interferon-alfa/genética , Interferon-alfa/metabolismo , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/metabolismo , Proteínas de Resistência a Myxovirus/genética
5.
Iran Biomed J ; 25(6): 426-33, 2021 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-34696577

RESUMO

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.


Assuntos
Neoplasias Encefálicas/complicações , Infecções por Citomegalovirus/epidemiologia , Citomegalovirus/isolamento & purificação , Infecções por Vírus Epstein-Barr/epidemiologia , Glioblastoma/complicações , Herpesvirus Humano 4/isolamento & purificação , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Neoplasias Encefálicas/virologia , Estudos de Casos e Controles , Criança , Pré-Escolar , Infecções por Citomegalovirus/virologia , Infecções por Vírus Epstein-Barr/virologia , Feminino , Glioblastoma/virologia , Humanos , Irã (Geográfico)/epidemiologia , Masculino , Pessoa de Meia-Idade , Adulto Jovem
6.
Cell Rep ; 36(10): 109673, 2021 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-34496248

RESUMO

Cancer therapy urges targeting of malignant subsets within self-renewing heterogeneous stem cell populations. We dissect the genetic and functional heterogeneity of human glioblastoma stem cells (GSCs) within patients by their innate responses to non-pathogenic mouse parvoviruses that are tightly restrained by cellular physiology. GSC neurospheres accumulate assembled capsids but restrict viral NS1 cytotoxic protein expression by an innate PKR/eIF2α-P response counteractable by electric pulses. NS1 triggers a comprehensive DNA damage response involving cell-cycle arrest, neurosphere disorganization, and bystander disruption of GSC-derived brain tumor architecture in rodent models. GSCs and cancer cell lines permissive to parvovirus genome replication require p53-Ser15 phosphorylation (Pp53S15). NS1 expression is enhanced by exogeneous Pp53S15 induction but repressed by wtp53. Consistently, patient-specific GSC subpopulations harboring p53 gain-of-function mutants and/or Pp53S15 are selective viral targets. This study provides a molecular foundation for personalized biosafe viral therapies against devastating glioblastoma and other cancers with deregulated p53 signaling.


Assuntos
Neoplasias Encefálicas/patologia , Glioblastoma/virologia , Células-Tronco Neoplásicas/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Animais , Neoplasias Encefálicas/genética , Linhagem Celular Tumoral , Glioblastoma/patologia , Humanos , Fosforilação , Ratos Nus , Transdução de Sinais/fisiologia , Replicação Viral/fisiologia
7.
Histochem Cell Biol ; 156(6): 527-538, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34514517

RESUMO

Structural changes of two patient-derived glioblastoma cell lines after Zika virus infection were investigated using scanning transmission electron tomography on high-pressure-frozen, freeze-substituted samples. In Zika-virus-infected cells, Golgi structures were barely visible under an electron microscope, and viral factories appeared. The cytosol outside of the viral factories resembled the cytosol of uninfected cells. The viral factories contained largely deranged endoplasmic reticulum (ER), filled with many so-called replication organelles consisting of a luminal vesicle surrounded by the ER membrane. Viral capsids were observed in the vicinity of the replication organelles (cell line #12537 GB) or in ER cisternae at large distance from the replication organelles (cell line #15747 GB). Near the replication organelles, we observed many about 100-nm-long filaments that may represent viral ribonucleoprotein complexes (RNPs), which consist of the RNA genome and N protein oligomers. In addition, we compared Zika-virus-infected cells with cells infected with a phlebovirus (sandfly fever Turkey virus). Zika virions are formed in the ER, whereas phlebovirus virions are assembled in the Golgi apparatus. Our findings will help to understand the replication cycle in the virus factories and the building of the replication organelles in glioblastoma cells.


Assuntos
Retículo Endoplasmático/metabolismo , Glioblastoma/metabolismo , Glioblastoma/virologia , Organelas/metabolismo , Ribonucleoproteínas/metabolismo , Zika virus/metabolismo , Tomografia com Microscopia Eletrônica , Humanos , Células Tumorais Cultivadas , Replicação Viral
8.
Nat Commun ; 12(1): 3834, 2021 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-34158478

RESUMO

H-1 parvovirus (H-1PV) is a promising anticancer therapy. However, in-depth understanding of its life cycle, including the host cell factors needed for infectivity and oncolysis, is lacking. This understanding may guide the rational design of combination strategies, aid development of more effective viruses, and help identify biomarkers of susceptibility to H-1PV treatment. To identify the host cell factors involved, we carry out siRNA library screening using a druggable genome library. We identify one crucial modulator of H-1PV infection: laminin γ1 (LAMC1). Using loss- and gain-of-function studies, competition experiments, and ELISA, we validate LAMC1 and laminin family members as being essential to H-1PV cell attachment and entry. H-1PV binding to laminins is dependent on their sialic acid moieties and is inhibited by heparin. We show that laminins are differentially expressed in various tumour entities, including glioblastoma. We confirm the expression pattern of laminin γ1 in glioblastoma biopsies by immunohistochemistry. We also provide evidence of a direct correlation between LAMC1 expression levels and H-1PV oncolytic activity in 59 cancer cell lines and in 3D organotypic spheroid cultures with different sensitivities to H-1PV infection. These results support the idea that tumours with elevated levels of γ1 containing laminins are more susceptible to H-1PV-based therapies.


Assuntos
Parvovirus H-1/metabolismo , Laminina/metabolismo , Ácido N-Acetilneuramínico/metabolismo , Vírus Oncolíticos/metabolismo , Ligação Viral , Internalização do Vírus , Animais , Linhagem Celular Tumoral , Glioblastoma/patologia , Glioblastoma/terapia , Glioblastoma/virologia , Células HCT116 , Células HEK293 , Células HeLa , Humanos , Laminina/genética , Camundongos Endogâmicos NOD , Camundongos SCID , Terapia Viral Oncolítica/métodos , Ligação Proteica , Interferência de RNA , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
9.
Neuroreport ; 32(9): 771-775, 2021 06 09.
Artigo em Inglês | MEDLINE | ID: mdl-33994523

RESUMO

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.


Assuntos
Neoplasias Encefálicas/virologia , COVID-19/metabolismo , Proteínas do Nucleocapsídeo de Coronavírus/metabolismo , Glioblastoma/virologia , SARS-CoV-2/metabolismo , Adulto , Enzima de Conversão de Angiotensina 2/metabolismo , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/cirurgia , Neoplasias Encefálicas/ultraestrutura , COVID-19/virologia , Teste de Ácido Nucleico para COVID-19 , Convalescença , Glioblastoma/metabolismo , Glioblastoma/cirurgia , Glioblastoma/ultraestrutura , Humanos , Hibridização In Situ , Masculino , Microscopia Eletrônica de Transmissão , Fosfoproteínas/metabolismo , RNA Viral/metabolismo , Receptores de Coronavírus/metabolismo , SARS-CoV-2/ultraestrutura , Vírion/ultraestrutura
10.
Cancer Lett ; 513: 26-35, 2021 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-33989707

RESUMO

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.


Assuntos
Neoplasias Encefálicas/virologia , Infecções por Citomegalovirus/genética , Glioblastoma/virologia , NF-kappa B/metabolismo , Proteínas Proto-Oncogênicas c-met/metabolismo , Animais , Neoplasias Encefálicas/patologia , Infecções por Citomegalovirus/patologia , Glioblastoma/patologia , Humanos , Camundongos , Regulação para Cima
11.
Brain Res ; 1758: 147344, 2021 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-33556379

RESUMO

Modelling cell infection in-a-dish can represent a useful tool to understand the susceptibility of different cell types towards severe acute respiratory coronavirus-2 (SARS-CoV-2) and to decipher its neurotropism. In this perspective, retinoic acid (RA)-differentiated neuroblastoma cell lines, SH-SY5Y and SK-N-BE(2) and glioblastoma cell lines, U-87 MG and U-373 MG, were infected with a SARS-CoV-2 strain, at various multiplicity-of-infection (MOI). We first demonstrated that the common entry genes - needed for invading epithelial cells - were expressed. RA-differentiation induced an upregulation of ace2 and tmprss2 gene expression while inducing downregulation of ctsb and ctsl. Using in situ hybridization and confocal analysis, SARS-CoV-2 gene S RNA was detected intracellularly at MOI 5.0, and localized in both soma and neuritic-like or glial-like processes. The infection was confirmed by quantification of viral gene E RNA and showed a dose-dependency, with few infected cells at MOI 0.1. After 24 h of infection, no cytopathic effect was observed in SH-SY5Y abilities to maintain neuritic processes or in U-373 MG for the uptake of glutamate. Unlike the permissive Vero E6 cells, no significant apoptosis death was detected following SARS-CoV-2 infection of neuroblastoma or glioblastoma cells. This study demonstrates the susceptibility of neuronal- and glial-like cell lines towards SARS-CoV-2 infection at high MOIs. Once inside the cells, the virus does not seem to rapidly replicate nor exert major cytopathic effect. Overall, our results strengthen the idea that SARS-CoV-2 has a tropism for nervous cells that express commonly described entry genes.


Assuntos
COVID-19/virologia , Glioblastoma/virologia , Neuroblastoma/virologia , SARS-CoV-2/patogenicidade , Enzima de Conversão de Angiotensina 2/metabolismo , COVID-19/patologia , Linhagem Celular Tumoral , Citoplasma/metabolismo , Glioblastoma/patologia , Humanos , Modelos Biológicos , Neuroblastoma/patologia , SARS-CoV-2/metabolismo , Serina Endopeptidases/metabolismo
12.
J Neurovirol ; 27(1): 94-100, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33405205

RESUMO

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.


Assuntos
Neoplasias Encefálicas/virologia , Infecções por Citomegalovirus/imunologia , Glioblastoma/virologia , Hospedeiro Imunocomprometido , Ativação Viral/imunologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Quimiorradioterapia/efeitos adversos , Citomegalovirus , Infecções por Citomegalovirus/complicações , Feminino , Glioblastoma/terapia , Humanos , Infecção Latente/imunologia , Masculino , Pessoa de Meia-Idade , Fatores de Risco
13.
Mol Med Rep ; 22(4): 3066-3072, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32945485

RESUMO

Human cytomegalovirus (HCMV) is a prevalent viral pathogen, which can cause severe clinical consequences in neonates, immunocompromised individuals, patients with AIDS, and organ and stem cell transplant recipients. HCMV inhibits the host cell cycle progress while the immediate­early protein 1 (IE1) tethers to condensed chromatin in mitotic cells. The present study investigated the effect of HCMV on the cell cycle in human glioblastoma cells, as well as the role of RhoA GTPase during mitosis in the same context. Live cell microscopy showed that despite the apparent cell cycle arrest at late stages of mitosis in normal fibroblasts, HCMV­infected U373MG cells successfully went through all stages of cell division. HCMV IE1 protein exhibited a remarkably tight association with mitotic chromosomes from early mitosis to late cytokinesis. Depletion of RhoA significantly impaired the proliferation rate of HCMV­infected U373MG cells; consistent with this observation, the number of cells entering mitosis was also decreased. These results demonstrated the differential behavior of HCMV during mitosis in a normal and a cancer background. Furthermore, RhoA may be a critical component for the efficient cell division of HCMV­infected glioblastoma cells, which subsequently ensures the maintenance of viral genomes.


Assuntos
Neoplasias Encefálicas/virologia , Infecções por Citomegalovirus/metabolismo , Citomegalovirus/fisiologia , Glioblastoma/virologia , Proteínas Imediatamente Precoces/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Pontos de Checagem do Ciclo Celular , Linhagem Celular Tumoral , Proliferação de Células , Citocinese , Citomegalovirus/metabolismo , Infecções por Citomegalovirus/genética , Fibroblastos/citologia , Fibroblastos/metabolismo , Fibroblastos/virologia , Glioblastoma/genética , Glioblastoma/metabolismo , Humanos , Mitose , Replicação Viral , Proteína rhoA de Ligação ao GTP/genética
14.
J Neurovirol ; 26(6): 984-987, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32880872

RESUMO

In the current study, a 58-year-old male patient presented with recurrent glioblastoma multiforme (GBM). The patient underwent surgical resection, 4 months earlier, followed by radiotherapy and chemotherapy. During the second surgical intervention, tumour tissue and whole blood were sampled and analysed for human cytomegalovirus (HCMV) DNA, immediate early (IE) mRNA and pp65 mRNA. HCMV DNA was detected only in the recurrent tumour tissue but not in the whole blood. Neither IE mRNA nor pp65 mRNA was expressed. Our result suggests HCMV latency in the brain tumour with detectable level of viral DNA. More data are needed to understand the HCMV infection chronology in the brain tumours but our data could be important for further studies of HCMV antigens on the tumour surface and anti-GBM therapy.


Assuntos
Neoplasias Encefálicas/patologia , Infecções por Citomegalovirus/patologia , Citomegalovirus/genética , DNA Viral/genética , Glioblastoma/patologia , Recidiva Local de Neoplasia/patologia , Neoplasias Encefálicas/diagnóstico por imagem , Neoplasias Encefálicas/cirurgia , Neoplasias Encefálicas/virologia , Citomegalovirus/isolamento & purificação , Infecções por Citomegalovirus/diagnóstico por imagem , Infecções por Citomegalovirus/cirurgia , Infecções por Citomegalovirus/virologia , Evolução Fatal , Glioblastoma/diagnóstico por imagem , Glioblastoma/cirurgia , Glioblastoma/virologia , Humanos , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Recidiva Local de Neoplasia/diagnóstico por imagem , Recidiva Local de Neoplasia/cirurgia , Recidiva Local de Neoplasia/virologia , Reação em Cadeia da Polimerase , RNA Mensageiro/genética , Proteínas da Matriz Viral/genética , Latência Viral/genética
15.
Cancer Med ; 9(18): 6776-6790, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32738030

RESUMO

The glioblastoma multiforme (GBM) is one of the deadliest tumors. It has been speculated that virus plays a role in GBM but the evidences are controversy. Published researches are mainly limited to studies on the presence of human cytomegalovirus (HCMV) in GBM. No comprehensive assessment of the brain virome, the collection of viral material in the brain, based on recently sequenced data has been performed. Here, we characterized the virome from 111 GBM samples and 57 normal brain samples from eight projects in the SRA database by a tested and comprehensive assembly approach. The annotation of the assembled contigs showed that most viral sequences in the brain belong to the viral family Retroviridae. In some GBM samples, we also detected full genome sequence of a novel picornavirus recently discovered in invertebrates. Unlike previous reports, our study did not detect herpes virus such as HCMV in GBM from the data we used. However, some contigs that cannot be annotated with any known genes exhibited antibody epitopes in their sequences. These findings provide several avenues for potential cancer therapy: the newly discovered picornavirus could be a starting point to engineer novel oncolytic virus; and the exhibited antibody epitopes could be a source to explore potential drug targets for immune cancer therapy. By characterizing the virosphere in GBM and normal brain at a global level, the results from this study strengthen the link between GBM and viral infection which warrants the further investigation.


Assuntos
Neoplasias Encefálicas/virologia , Encéfalo/virologia , Glioblastoma/virologia , Sequenciamento de Nucleotídeos em Larga Escala , Metagenômica , Picornaviridae/genética , Retroviridae/genética , Viroma/genética , Encéfalo/patologia , Neoplasias Encefálicas/patologia , Estudos de Casos e Controles , Mapeamento de Sequências Contíguas , Bases de Dados de Ácidos Nucleicos , Glioblastoma/patologia , Humanos , RNA Viral/genética , RNA-Seq
16.
Clin Cancer Res ; 26(20): 5297-5303, 2020 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-32719000

RESUMO

Despite standard of care for glioblastoma, including gross total resection, high-dose radiation, and dose-limited chemotherapy, this tumor remains one of the most aggressive and therapeutically challenging. The relatively small number of patients with this diagnosis compared with more common solid tumors in clinical trials commits new glioblastoma therapies to testing in small, underpowered, nonrandomized settings. Among approximately 200 registered glioblastoma trials identified between 2005 and 2015, nearly half were single-arm studies with sample sizes not exceeding 50 patients. These constraints have made demonstrating efficacy for novel therapies difficult in glioblastoma and other rare and aggressive cancers. Novel immunotherapies for glioblastoma such as vaccination with dendritic cells (DC) have yielded mixed results in clinical trials. To address limited numbers, we sequentially conducted three separate clinical trials utilizing cytomegalovirus (CMV)-specific DC vaccines in patients with newly diagnosed glioblastoma whereby each follow-up study had nearly doubled in sample size. Follow-up data from the first blinded, randomized phase II clinical trial (NCT00639639) revealed that nearly one third of this cohort is without tumor recurrence at 5 years from diagnosis. A second clinical trial (NCT00639639) resulted in a 36% survival rate at 5 years from diagnosis. Results of the first two-arm trial (NCT00639639) showed increased migration of the DC vaccine to draining lymph nodes, and this increased migration has been recapitulated in our larger confirmatory clinical study (NCT02366728). We have now observed that nearly one third of the glioblastoma study patient population receiving CMV-specific DC vaccines results in exceptional long-term survivors.


Assuntos
Neoplasias Encefálicas/tratamento farmacológico , Vacinas Anticâncer/administração & dosagem , Glioblastoma/tratamento farmacológico , Recidiva Local de Neoplasia/tratamento farmacológico , Adulto , Idoso , Neoplasias Encefálicas/imunologia , Neoplasias Encefálicas/patologia , Vacinas Anticâncer/efeitos adversos , Vacinas Anticâncer/imunologia , Citomegalovirus/efeitos dos fármacos , Citomegalovirus/patogenicidade , Células Dendríticas/imunologia , Células Dendríticas/virologia , Feminino , Glioblastoma/imunologia , Glioblastoma/virologia , Humanos , Imunoterapia , Masculino , Pessoa de Meia-Idade , Recidiva Local de Neoplasia/imunologia , Recidiva Local de Neoplasia/patologia
17.
Viruses ; 12(5)2020 05 25.
Artigo em Inglês | MEDLINE | ID: mdl-32466170

RESUMO

We studied whether cytosine phosphate-guanine (CpG) recoding in a viral genome may provide oncolytic candidates with reduced infection kinetics in nonmalignant brain cells, but with high virulence in glioblastoma stem cells (GSCs). As a model, we used well-characterized CpG-recoded Zika virus vaccine candidates that previously showed genetic stability and safety in animal models. In vitro, one of the CpG-recoded Zika virus variants had reduced infection kinetics in nonmalignant brain cells but high infectivity and oncolytic activity in GSCs as represented by reduced cell proliferation. The recoded virus also efficiently replicated in GSC-derived tumors in ovo with a significant reduction of tumor growth. We also showed that some GSCs may be resistant to Zika virus oncolytic activity, emphasizing the need for personalized oncolytic therapy or a strategy to overcome resistance in GSCs. Collectively, we demonstrated the potential of the CpG recoding approach for oncolytic virus development that encourages further research towards a better understanding of host-tumor-CpG-recoded virus interactions.


Assuntos
Ilhas de CpG/fisiologia , Glioblastoma/metabolismo , Células-Tronco/metabolismo , Zika virus/metabolismo , Animais , Linhagem Celular Tumoral , Proliferação de Células , Ilhas de CpG/genética , Glioblastoma/virologia , Humanos , Vírus Oncolíticos/genética , Fosfatos , Replicação Viral , Infecção por Zika virus
18.
Clin Cancer Res ; 26(14): 3565-3577, 2020 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-32299815

RESUMO

PURPOSE: Cytomegalovirus (CMV) antigens occur in glioblastoma but not in normal brains, making them desirable immunologic targets. PATIENTS AND METHODS: Highly functional autologous polyclonal CMV pp65-specific T cells from patients with glioblastoma were numerically expanded under good manufacturing practice compliant conditions and administered after 3 weeks of lymphodepleting dose-dense temozolomide (100 mg/m2) treatment. The phase I component used a 3+3 design, ascending through four dose levels (5 × 106-1 × 108 cells). Treatment occurred every 6 weeks for four cycles. In vivo persistence and effector function of CMV-specific T cells was determined by dextramer staining and multiparameter flow cytometry in serially sampled peripheral blood and in the tumor microenvironment. RESULTS: We screened 65 patients; 41 were seropositive for CMV; 25 underwent leukapheresis; and 20 completed ≥1 cycle. No dose-limiting toxicities were observed. Radiographic response was complete in 1 patient, partial in 2. Median progression-free survival (PFS) time was 1.3 months [95% confidence interval (CI), 0-8.3 months]; 6-month PFS was 19% (95% CI, 7%-52%); and median overall survival time was 12 months (95% CI, 6 months to not reached). Repeated infusions of CMV-T cells paralleled significant increases in circulating CMV+ CD8+ T cells, but cytokine production showing effector activity was suppressed, especially from T cells obtained directly from glioblastomas. CONCLUSIONS: Adoptive infusion of CMV-specific T cells after lymphodepletion with dose-dense temozolomide was well tolerated. But apparently CMV seropositivity does not guarantee tumor susceptibility to CMV-specific T cells, suggesting heterogeneity in CMV antigen expression. Moreover, effector function of these T cells was attenuated, indicating a requirement for further T-cell modulation to prevent their dysfunction before conducting large-scale clinical studies.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Infecções por Citomegalovirus/terapia , Glioblastoma/terapia , Imunoterapia Adotiva/métodos , Proteínas da Matriz Viral/imunologia , Adulto , Citomegalovirus/imunologia , Citomegalovirus/isolamento & purificação , Infecções por Citomegalovirus/imunologia , Infecções por Citomegalovirus/mortalidade , Infecções por Citomegalovirus/virologia , Feminino , Glioblastoma/imunologia , Glioblastoma/mortalidade , Glioblastoma/virologia , Humanos , Leucaférese , Depleção Linfocítica/métodos , Masculino , Pessoa de Meia-Idade , Intervalo Livre de Progressão , Temozolomida/administração & dosagem , Transplante Autólogo/métodos , Microambiente Tumoral/imunologia
19.
J Virol ; 94(8)2020 03 31.
Artigo em Inglês | MEDLINE | ID: mdl-32051271

RESUMO

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.


Assuntos
Neoplasias Encefálicas/metabolismo , Ebolavirus/imunologia , Glicoproteínas/imunologia , Doença pelo Vírus Ebola/virologia , Mucinas/imunologia , Animais , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/virologia , Linhagem Celular Tumoral , Modelos Animais de Doenças , Ebolavirus/genética , Glioblastoma/virologia , Glioma/patologia , Glioma/virologia , Proteínas de Fluorescência Verde , Xenoenxertos , Humanos , Camundongos , Camundongos SCID , Mucinas/genética , Vírus da Estomatite Vesicular Indiana/imunologia
20.
Cancer Lett ; 468: 41-47, 2020 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-31605777

RESUMO

Gliomas are tumors that originate from the glial tissue, thus involving the central nervous system with varying degrees of malignancy. The most aggressive and frequent form is glioblastoma multiforme, a disease characterized by resistance to therapies, frequent recurrences, and extremely poor median survival time. Data on overall glioma case studies demonstrate clear sex disparities regarding incidence, prognosis, drug toxicity, clinical outcome, and, recently, prediction of therapeutic response. In this study, we analyze data in the literature regarding malignant glioma, mainly glioblastoma multiforme, focusing on epidemiological and clinical evaluations. Less discussed issues, such as the role of viral infections, energy metabolism, and predictive aspects concerning the possible use of dedicated therapeutic approaches for male or female patients, will be reported together with different estimated pathogenetic mechanisms underlying astrocyte transformation and glioma chemosensitivity. In this era, where personalized/precision medicine is the most important driver for targeted cancer therapies, the lines of evidence discussed herein strongly suggest that clinical approaches to malignant glioma should consider the patient's sex. Furthermore, retrospectively revising previous clinical studies considering patient sex as a crucial variable is recommended.


Assuntos
Neoplasias Encefálicas/terapia , Glioblastoma/terapia , Disparidades nos Níveis de Saúde , Recidiva Local de Neoplasia/terapia , Medicina de Precisão/métodos , Neoplasias Encefálicas/epidemiologia , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/virologia , Citomegalovirus/isolamento & purificação , Citomegalovirus/patogenicidade , Receptores ErbB/genética , Receptores ErbB/metabolismo , Feminino , Glioblastoma/epidemiologia , Glioblastoma/genética , Glioblastoma/virologia , Humanos , Incidência , Masculino , Recidiva Local de Neoplasia/epidemiologia , Recidiva Local de Neoplasia/genética , Recidiva Local de Neoplasia/virologia , Neuroglia/patologia , Neuroglia/virologia , Papillomaviridae/isolamento & purificação , Papillomaviridae/patogenicidade , Polyomavirus/isolamento & purificação , Polyomavirus/patogenicidade , Prognóstico , Fatores de Risco , Fatores Sexuais , Transdução de Sinais/genética , Telomerase/genética , Telomerase/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...