Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 80
Filter
1.
J Pediatr Hematol Oncol ; 43(8): 314-315, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34673712

ABSTRACT

The interaction of coronavirus disease-2019 (COVID-19) and chemotherapy may result in worse outcomes. However, there may be more indirect effects of COVID. We report 3 cases in which treatment was delayed because of COVID-related inability or reluctance to travel. Oncology programs should consider such indirect effects when devising treatments.


Subject(s)
COVID-19/transmission , Osteosarcoma/drug therapy , Retinoblastoma/drug therapy , SARS-CoV-2/isolation & purification , Time-to-Treatment/statistics & numerical data , Transportation/statistics & numerical data , Antineoplastic Combined Chemotherapy Protocols , Bone Neoplasms/drug therapy , Bone Neoplasms/virology , COVID-19/virology , Child , Child, Preschool , Female , Humans , Infant , Male , Osteosarcoma/virology , Prognosis , Retinal Neoplasms/drug therapy , Retinal Neoplasms/virology , Retinoblastoma/virology
2.
Commun Biol ; 4(1): 510, 2021 04 30.
Article in English | MEDLINE | ID: mdl-33931711

ABSTRACT

G-quadruplexes (G4s) are four-stranded nucleic acid structures abundant at gene promoters. They can adopt several distinctive conformations. G4s have been shown to form in the herpes simplex virus-1 (HSV-1) genome during its viral cycle. Here by cross-linking/pull-down assay we identified ICP4, the major HSV-1 transcription factor, as the protein that most efficiently interacts with viral G4s during infection. ICP4 specific and direct binding and unfolding of parallel G4s, including those present in HSV-1 immediate early gene promoters, induced transcription in vitro and in infected cells. This mechanism was also exploited by ICP4 to promote its own transcription. Proximity ligation assay allowed visualization of G4-protein interaction at the single selected G4 in cells. G4 ligands inhibited ICP4 binding to G4s. Our results indicate the existence of a well-defined G4-viral protein network that regulates the productive HSV-1 cycle. They also point to G4s as elements that recruit transcription factors to activate transcription in cells.


Subject(s)
G-Quadruplexes , Herpes Simplex/complications , Herpesvirus 1, Human/genetics , Immediate-Early Proteins/metabolism , Osteosarcoma/virology , Promoter Regions, Genetic , Viral Transcription , Bone Neoplasms/genetics , Bone Neoplasms/pathology , Bone Neoplasms/virology , DNA Replication , Herpes Simplex/genetics , Herpes Simplex/virology , Humans , Immediate-Early Proteins/genetics , Osteosarcoma/genetics , Osteosarcoma/pathology , Tumor Cells, Cultured
3.
J Immunother Cancer ; 9(3)2021 03.
Article in English | MEDLINE | ID: mdl-33737338

ABSTRACT

BACKGROUND: Osteosarcoma is the most common malignant solid tumor that affects bones, however, survival rates of patients with relapsed osteosarcoma have not improved in the last 30 years. Oncolytic virotherapy, which uses viruses designed to selectively replicate in cancer cells, has emerged as a promising treatment for solid tumors. Our group uses mesenchymal stem cells (MSCs) to transport oncolytic adenoviruses (OAds) to the tumor site, a therapeutic strategy called Celyvir. This treatment has been already applied in human patients, canine patients and different mouse models. In parallel, previous results have probed that administration of granulocyte-colony stimulating factor (G-CSF) increased immune infiltration in tumors. We then hypothesized that the mobilization of immune cells by G-CSF may increase the antitumor efficacy of Celyvir treatment by increasing the immune infiltration into the tumors. METHODS: In this study, we use a murine version of Celyvir consisting in murine MSCs carrying the murine OAd dlE102-here called OAd-MSCs-in an immunocompetent model of osteosarcoma. We tested the antitumoral efficacy of the combination of OAd-MSCs plus G-CSF. RESULTS: Our results show that treatment with OAd-MSCs or the union of OAd-MSCs with G-CSF (Combination) significantly reduced tumor growth of osteosarcoma in vivo. Moreover, treated tumors presented higher tumor infiltration of immune cells-especially tumor-infiltrating lymphocytes-and reduced T cell exhaustion, which seems to be enhanced in tumors treated with the Combination. The comparison of our results to those obtained from a cohort of pediatric osteosarcoma patients showed that the virotherapy induces immunological changes similar to those observed in patients with good prognosis. CONCLUSIONS: The results open the possibility of using cellular virotherapy for the treatment of bone cancers. Indeed, its combination with G-CSF may be considered for the improvement of the therapy.


Subject(s)
Adenoviridae/pathogenicity , Bone Neoplasms/therapy , Granulocyte Colony-Stimulating Factor/pharmacology , Immunomodulating Agents/pharmacology , Mesenchymal Stem Cells/virology , Oncolytic Virotherapy , Oncolytic Viruses/pathogenicity , Osteosarcoma/therapy , Adenoviridae/immunology , Animals , Bone Neoplasms/immunology , Bone Neoplasms/pathology , Bone Neoplasms/virology , Cell Line, Tumor , Combined Modality Therapy , Cytopathogenic Effect, Viral , Databases, Genetic , Humans , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Mesenchymal Stem Cell Transplantation , Mice, Inbred BALB C , Oncolytic Viruses/immunology , Osteosarcoma/immunology , Osteosarcoma/pathology , Osteosarcoma/virology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Tumor Microenvironment
4.
Proc Natl Acad Sci U S A ; 118(10)2021 03 09.
Article in English | MEDLINE | ID: mdl-33674386

ABSTRACT

Osteosarcoma is the most common malignant tumor of bone predominately affecting adolescents and young adults. Based on animal studies, a viral etiology of osteosarcoma was proposed more than a half-century ago, but no viral association with human osteosarcoma has been found. The Uyghur ethnic population in Xinjiang, China, has an unusually high prevalence of Kaposi's sarcoma-associated herpesvirus (KSHV) infection and elevated incidence of osteosarcoma. In the current study, we explored the possible association of KSHV infection and osteosarcoma occurrence. Our seroepidemiological study revealed that KSHV prevalence was significantly elevated in Uyghur osteosarcoma patients versus the general Uyghur population (OR, 10.23; 95%CI, 4.25, 18.89). The KSHV DNA genome and viral latent nuclear antigen LANA were detected in most osteosarcoma tumor cells. Gene expression profiling analysis showed that KSHV-positive osteosarcoma represents a distinct subtype of osteosarcomas with viral gene-activated signaling pathways important for osteosarcoma development. We conclude that KSHV infection is a risk factor for osteosarcoma, and KSHV is associated with some osteosarcomas, representing a newly identified viral-associated endemic cancer.


Subject(s)
Herpesviridae Infections , Herpesvirus 8, Human/metabolism , Osteosarcoma , Adolescent , Adult , Antigens, Viral/metabolism , Child , Child, Preschool , China/epidemiology , China/ethnology , DNA, Viral/metabolism , Female , Genome, Viral , Herpesviridae Infections/epidemiology , Herpesviridae Infections/ethnology , Herpesviridae Infections/metabolism , Herpesviridae Infections/virology , Humans , Male , Middle Aged , Neoplasm Proteins/metabolism , Nuclear Proteins/metabolism , Osteosarcoma/epidemiology , Osteosarcoma/ethnology , Osteosarcoma/metabolism , Osteosarcoma/virology , Prevalence , Viral Proteins/metabolism
5.
BMC Vet Res ; 17(1): 84, 2021 Feb 18.
Article in English | MEDLINE | ID: mdl-33602243

ABSTRACT

BACKGROUND: Osteosarcoma is a malignant mesenchymal bone tumor. Although it is a common tumor in the appendicular skeleton of dogs and cats, it is rarely reported in birds. Retroviruses are usually associated with solid tumor development in different avian species. CASE PRESENTATION: This report aims to describe a case of osteosarcoma associated with the avian leukosis virus in a captive bare-faced curassow (Crax fasciolata). A captive adult female bare-faced curassow presented with lameness, hyporexia, and a non-ulcerative and firm tumor in the right femur. The bird was euthanized due to the poor prognosis. Histopathology revealed an infiltrative mesenchymal neoplasm consisting of spindle cells with moderate cell pleomorphism, organized in bundles and interspersed by marked deposition of the osteoid matrix, which was compatible with osteosarcoma affecting both femur and tibiotarsus, with renal metastasis. Immunohistochemistry of the primary and metastatic tumor demonstrated vimentin expression by neoplastic cells. Samples of the neoplasm, bone marrow, and spleen were processed for PCR, which enabled the demonstration of proviral avian leukosis virus (ALV) DNA. CONCLUSIONS: To the best of our knowledge, this is the first report of an osteosarcoma in a bare-faced curassow with an unusual polyostotic manifestation and associated with ALV infection.


Subject(s)
Avian Leukosis , Bird Diseases/pathology , Bone Neoplasms/veterinary , Osteosarcoma/veterinary , Animals , Avian Leukosis Virus/isolation & purification , Bird Diseases/virology , Bone Marrow/virology , Bone Neoplasms/virology , Female , Galliformes/virology , Kidney Neoplasms/secondary , Kidney Neoplasms/veterinary , Osteosarcoma/virology , Spleen/virology , Vimentin/metabolism
6.
J Pediatr Hematol Oncol ; 43(6): e859-e860, 2021 08 01.
Article in English | MEDLINE | ID: mdl-32852398

ABSTRACT

The novel coronavirus, SARS-CoV-2, causes much more severe disease in adults than in children. Although it is anticipated that immune compromised children and children with cancer may be at higher risk of developing severe or fatal COVID-19, there are no currently published reports of fatal disease in a child with cancer. Because of the discrepancy in disease severity between adult and pediatric patients, we report the case of an adolescent with pulmonary metastatic osteosarcoma who died of COVID-19 early in the course of the pandemic in New York City in the hope that heightening awareness that pulmonary metastatic disease may predispose to a more severe outcome will increase surveillance in this vulnerable population.


Subject(s)
Bone Neoplasms/pathology , COVID-19/complications , Lung Neoplasms/secondary , Osteosarcoma/pathology , Respiratory Insufficiency/pathology , SARS-CoV-2/isolation & purification , Bone Neoplasms/complications , Bone Neoplasms/virology , COVID-19/virology , Child , Humans , Lung Neoplasms/complications , Lung Neoplasms/virology , Male , Osteosarcoma/complications , Osteosarcoma/virology , Respiratory Insufficiency/etiology , Severity of Illness Index
7.
Tumour Biol ; 42(11): 1010428320974247, 2020 Nov.
Article in English | MEDLINE | ID: mdl-33234011

ABSTRACT

Epstein-Barr virus is an etiologic agent of several malignancies. In this study, we explored the association of Epstein-Barr virus-encoded RNA1 and Epstein-Barr virus latent membrane protein 1 co-expression with osteosarcoma. Epstein-Barr virus-encoded RNA1 expression in tumor cells was quantified using reverse transcriptase polymerase chain reaction and in situ hybridization and Epstein-Barr virus latent membrane protein 1 expression was measured using immunohistochemistry staining. There was a statistically significant association between Epstein-Barr virus latent membrane protein 1 and Epstein-Barr virus-encoded RNA1 co-expression and characteristics of osteosarcoma such as nodal stage (p < 0.04), metastasis (p < 0.04), Ki67 index (p < 0.03), and tumor stage (p < 0.05). Co-expression of Epstein-Barr virus-encoded RNA1 and Epstein-Barr virus latent membrane protein 1 in tumors correlated with advanced osteosarcoma and indicated the aggressiveness of bone sarcoma.


Subject(s)
Osteosarcoma/virology , RNA, Viral/metabolism , Viral Matrix Proteins/metabolism , Adolescent , Adult , Biomarkers, Tumor/metabolism , Child , Epstein-Barr Virus Infections/mortality , Epstein-Barr Virus Infections/pathology , Epstein-Barr Virus Infections/virology , Female , Herpesvirus 4, Human/physiology , Humans , Iran/epidemiology , Male , Osteosarcoma/mortality , Osteosarcoma/pathology , RNA, Viral/genetics , Retrospective Studies , Survival Analysis , Young Adult
8.
Gen Physiol Biophys ; 38(4): 335-342, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31219428

ABSTRACT

Ultraviolet-inactivated Sendai virus strain Tianjin (UV-Tianjin) has been proved to have antitumor effects in many kinds of tumor cells. Here, we investigated the anticancer properties of UV-Tianjin on human osteosarcoma HOS cells and the underlying molecular mechanism. Apoptosis, intracellular reactive oxygen species (ROS) levels and mitochondrial membrane potential were determined by flow cytometry analysis. The expression levels of apoptosis-related proteins were tested by western blotting. The results showed that UV-Tianjin concentration-dependently induced apoptosis in HOS cells. UV-Tianjin-induced apoptosis was mediated by the mitochondrial pathway, which was confirmed by mitochondrial dysfunction, downregulation of B-cell lymphoma 2 (Bcl-2), B-cell lymphoma-xL (Bcl-xL) and myeloid cell leukemia-1 (Mcl-1), upregulation of B-cell lymphoma 2 associated X protein (Bax) and Bcl-2 Homologous Antagonist/Killer (Bak), as well as the cleavage of caspase-9 and -3. Further analysis showed that UV-Tianjin augmented the phosphorylation of c-Jun N-terminal kinase, the extracellular-regulated kinase and p38, the major components of mitogen-activated protein kinase (MAPK) pathways, as well as the generation of ROS. Moreover, UV-Tianjin-induced apoptosis was remarkably attenuated by MAPK inhibitors and ROS inhibitor. Taken together, our results indicated that UV-Tianjin exerts antitumor effects by inducing mitochondria-dependent apoptosis involving ROS generation and MAPK pathway in human osteosarcoma HOS cells.


Subject(s)
Apoptosis , Mitochondria/metabolism , Oncolytic Virotherapy , Osteosarcoma/therapy , Osteosarcoma/virology , Sendai virus/classification , Sendai virus/radiation effects , Cell Line, Tumor , Humans , MAP Kinase Signaling System , Membrane Potential, Mitochondrial , Mitochondria/enzymology , Osteosarcoma/pathology , Reactive Oxygen Species/metabolism
9.
PLoS Pathog ; 15(5): e1007798, 2019 05.
Article in English | MEDLINE | ID: mdl-31116799

ABSTRACT

Cellular antiviral programs encode molecules capable of targeting multiple steps in the virus lifecycle. Zinc-finger antiviral protein (ZAP) is a central and general regulator of antiviral activity that targets pathogen mRNA stability and translation. ZAP is diffusely cytoplasmic, but upon infection ZAP is targeted to particular cytoplasmic structures, termed stress granules (SGs). However, it remains unclear if ZAP's antiviral activity correlates with SG localization, and what molecular cues are required to induce this localization event. Here, we use Sindbis virus (SINV) as a model infection and find that ZAP's localization to SGs can be transient. Sometimes no apparent viral infection follows ZAP SG localization but ZAP SG localization always precedes accumulation of SINV non-structural protein, suggesting virus replication processes trigger SG formation and ZAP recruitment. Data from single-molecule RNA FISH corroborates this finding as the majority of cells with ZAP localization in SGs contain low levels of viral RNA. Furthermore, ZAP recruitment to SGs occurred in ZAP-expressing cells when co-cultured with cells replicating full-length SINV, but not when co-cultured with cells replicating a SINV replicon. ZAP recruitment to SGs is functionally important as a panel of alanine ZAP mutants indicate that the anti-SINV activity is correlated with ZAP's ability to localize to SGs. As ZAP is a central component of the cellular antiviral programs, these data provide further evidence that SGs are an important cytoplasmic antiviral hub. These findings provide insight into how antiviral components are regulated upon virus infection to inhibit virus spread.


Subject(s)
Alphavirus Infections/prevention & control , Antiviral Agents/pharmacology , Cytoplasmic Granules/metabolism , RNA-Binding Proteins/pharmacology , Sindbis Virus/pathogenicity , Stress, Physiological , Virus Replication/drug effects , Alphavirus Infections/metabolism , Alphavirus Infections/virology , Antiviral Agents/metabolism , Bone Neoplasms/drug therapy , Bone Neoplasms/metabolism , Bone Neoplasms/virology , Humans , Osteosarcoma/drug therapy , Osteosarcoma/metabolism , Osteosarcoma/virology , Protein Transport , RNA Interference , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Tumor Cells, Cultured
10.
PLoS Pathog ; 15(5): e1007788, 2019 05.
Article in English | MEDLINE | ID: mdl-31091289

ABSTRACT

Inhibition of human papillomavirus (HPV) replication is a promising therapeutic approach for intervening with HPV-related pathologies. Primary targets for interference are two viral proteins, E1 and E2, which are required for HPV replication. Both E1 and E2 are phosphoproteins; thus, the protein kinases that phosphorylate them might represent secondary targets to achieve inhibition of HPV replication. In the present study, we show that CX4945, an ATP-competitive small molecule inhibitor of casein kinase 2 (CK2) catalytic activity, suppresses replication of different HPV types, including novel HPV5NLuc, HPV11NLuc and HPV18NLuc marker genomes, but enhances the replication of HPV16 and HPV31. We further corroborate our findings using short interfering RNA (siRNA)-mediated knockdown of CK2 α and α' subunits in U2OS and CIN612 cells; we show that while both subunits are expressed in these cell lines, CK2α is required for HPV replication, but CK2α' is not. Furthermore, we demonstrate that CK2α acts in a kinase activity-dependent manner and regulates the stability and nuclear retention of endogenous E1 proteins of HPV11 and HPV18. This unique feature of CK2α makes it an attractive target for developing antiviral agents.


Subject(s)
Papillomaviridae/physiology , Papillomavirus Infections/virology , Phosphoproteins/metabolism , Viral Proteins/metabolism , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Bone Neoplasms/virology , Casein Kinase II/genetics , Casein Kinase II/metabolism , Humans , Osteosarcoma/metabolism , Osteosarcoma/pathology , Osteosarcoma/virology , Papillomavirus Infections/genetics , Papillomavirus Infections/metabolism , Phosphoproteins/genetics , Phosphorylation , Tumor Cells, Cultured , Viral Proteins/genetics
11.
Sci Rep ; 9(1): 5938, 2019 04 11.
Article in English | MEDLINE | ID: mdl-30976051

ABSTRACT

High-risk Human Papillomavirus infections are responsible for anogenital and oropharyngeal cancers. Alternative splicing is an important mechanism controlling HPV16 gene expression. Modulation in the splice pattern leads to polycistronic HPV16 early transcripts encoding a full length E6 oncoprotein or truncated E6 proteins, commonly named E6*. Spliced E6*I transcripts are the most abundant RNAs produced in HPV-related cancers. To date, the biological function of the E6*I isoform remains controversial. In this study, we identified, by RNA sequencing, cellular targets deregulated by E6*I, among which genes related to ROS metabolism. Concomitantly, E6*I-overexpressing cells display high levels of ROS. However, co-overexpression of both E6 and E6*I has no effect on ROS production. In HPV16-infected cells expressing different E6/E6*I levels, we show that the newly identified targets CCL2 and RAC2 are increased by E6*I but decreased by E6 expression, suggesting that E6 abrogates the effect of E6*I. Taken together, these data support the idea that E6*I acts independently of E6 to increase ROS production and that E6 has the ability to counteract the effects of E6*I. This asks the question of how E6*I can be considered separately of E6 in the natural history of HPV16 infection.


Subject(s)
Alternative Splicing , Host-Pathogen Interactions/genetics , Oncogene Proteins, Viral/genetics , Osteosarcoma/virology , Papillomavirus Infections/genetics , Reactive Oxygen Species/metabolism , Repressor Proteins/genetics , Sequence Analysis, RNA/methods , Uterine Cervical Neoplasms/virology , Bone Neoplasms/epidemiology , Bone Neoplasms/genetics , Bone Neoplasms/pathology , Bone Neoplasms/virology , Female , Human papillomavirus 16/isolation & purification , Humans , Osteosarcoma/epidemiology , Osteosarcoma/genetics , Osteosarcoma/pathology , Papillomavirus Infections/pathology , Papillomavirus Infections/virology , Tumor Cells, Cultured , Uterine Cervical Neoplasms/epidemiology , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/pathology
12.
J Cell Physiol ; 234(4): 4179-4190, 2019 04.
Article in English | MEDLINE | ID: mdl-30146726

ABSTRACT

Sendai virus strain Tianjin, a novel genotype of Sendai virus, has been proven to possess potent antitumor effect on certain cancer cell types although inactivated by ultraviolet (UV). This study was carried out to investigate the in vitro anticancer properties of UV-inactivated Sendai virus strain Tianjin (UV-Tianjin) on human osteosarcoma cells and the underlying molecular mechanism. Our studies demonstrated UV-Tianjin significantly inhibited the viability of human osteosarcoma cell lines and triggered apoptosis through activation of both extrinsic and intrinsic pathways in MG-63 cells. Meanwhile, autophagy occurred in UV-Tianjin-treated cells. Blockade of autophagy with 3-methyladenine remarkably attenuated the inhibition of cell proliferation by UV-Tianjin, suggesting that UV-Tianjin-induced autophagy may be contributing to cell death. Furthermore, UV-Tianjin induced reactive oxygen species (ROS) production, which was involved in the execution of MG-63 cell apoptosis and autophagy, as evidenced by the result that treatment of N-acetyl-L-cysteine, a ROS scavenger, attenuated both apoptosis and autophagy. In addition, inhibition of apoptosis promoted autophagy, whereas suppression of autophagy attenuated apoptosis. Our results suggest that UV-Tianjin triggers apoptosis and autophagic cell death via generation of the ROS in MG-63 cells, which might provide important insights into the effectiveness of novel strategies for osteosarcoma therapy.


Subject(s)
Apoptosis , Autophagy , Bone Neoplasms/therapy , Oncolytic Virotherapy , Oncolytic Viruses , Osteosarcoma/therapy , Oxidative Stress , Reactive Oxygen Species/metabolism , Sendai virus , Apoptosis Regulatory Proteins/metabolism , Autophagy-Related Proteins/metabolism , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Bone Neoplasms/virology , Cell Line, Tumor , Cell Proliferation , Host-Pathogen Interactions , Humans , Oncolytic Viruses/radiation effects , Osteosarcoma/metabolism , Osteosarcoma/pathology , Osteosarcoma/virology , Sendai virus/radiation effects , Ultraviolet Rays
13.
J Virol ; 93(3)2019 02 01.
Article in English | MEDLINE | ID: mdl-30429337

ABSTRACT

Human adenovirus (HAdV) encodes a multifunctional DNA-binding protein pVII, which is involved in virus DNA packaging and extracellular immune signaling regulation. Although the pVII is an essential viral protein, its exact role in the virus life cycle and interplay with cellular proteins have remained to a large extent unclear. We have recently identified the cellular zinc finger protein 622 (ZNF622) as a potential pVII-interacting protein. In this study, we describe the functional consequences of the ZNF622-pVII interplay and the role of ZNF622 in the HAdV life cycle. ZNF622 protein expression increased, and it accumulated similarly to the pVII protein in the nuclei of virus-infected cells. The lack of the ZNF622 protein specifically increased pVII binding to viral DNA in the infected cells and elevated the pVII protein levels in the purified virions. In addition, ZNF622 knockout cells showed an increased cell lysis and enhanced accumulation of the infectious virus particles. Protein interaction studies revealed that ZNF622 forms a trimeric complex with the pVII protein and the cellular histone chaperon protein nucleophosmin 1 (NPM1). The integrity of this complex is important since ZNF622 mutations and NPM1 deficiency changed pVII ability to bind viral DNA. Collectively, our results implicate that ZNF622 may act as a cellular antiviral protein hindering lytic HAdV growth and limiting pVII protein binding to viral DNA.IMPORTANCE Human adenoviruses (HAdVs) are common human pathogens causing a wide range of acute infections. To counteract viral pathogenicity, cells encode a variety of antiviral proteins and noncoding RNAs to block virus growth. In this study, we show that the cellular zinc finger protein 622 (ZNF622) interacts with an essential HAdV protein known as pVII. This mutual interaction limits pVII binding to viral DNA. Further, ZNF622 has a role in HAdV life cycle since the lack of ZNF622 correlates with increased lysis of the infected cells and accumulation of the infectious virions. Together, our study reveals a novel cellular antiviral protein ZNF622, which may impede lytic HAdV growth.


Subject(s)
Adenovirus Infections, Human/virology , Adenoviruses, Human/growth & development , DNA, Viral/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , Viral Core Proteins/metabolism , Virus Replication , Adenovirus Infections, Human/metabolism , Adenoviruses, Human/genetics , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Bone Neoplasms/virology , Cell Survival , Genome, Viral , Humans , Nucleophosmin , Osteosarcoma/genetics , Osteosarcoma/metabolism , Osteosarcoma/virology , Tumor Cells, Cultured
14.
Anticancer Drugs ; 29(3): 197-207, 2018 03.
Article in English | MEDLINE | ID: mdl-29438228

ABSTRACT

Newcastle disease virus (NDV) can specifically kill cancer cells and has less toxicity to normal cells. The hemagglutinin-neuraminidase (HN) protein is an important structural protein in NDV pathogenesis and has been postulated as a promising candidate for antitumor therapy. The aim of this study was to investigate the anticancer potential of recombinant adenovirus Ad-HN-PEG3p-E1a. An MTS assay was performed to determine viral proliferation after viral infection, the data showed that the proliferation ability of osteosarcoma cells decreased, whereas there was no significant change in normal hepatic cells. DAPI and Annexin V experiments showed that osteosarcoma cells were killed because of apoptosis, active oxygen content, and augmented mitochondrial membrane potential loss. Caspase Activity Assay Kits were used to detect the caspase-3 activities of the treated OS-732 for increased expression. Western blot analysis showed that cytochrome C increased significantly and apoptosis of the virus was confirmed in tumor cells. In-vivo experiments show that NDV has an inhibitory effect on tumor growth. The recombinant adenovirus, which is composed of a HN protein and progressive increment promoter PEG3p, could inhibit the growth of OS-732 and promote the apoptosis of tumor cells. However, there was no clear relationship with normal cell (L02) apoptosis.


Subject(s)
Bone Neoplasms/therapy , Hemagglutinins, Viral/genetics , Neuraminidase/genetics , Newcastle disease virus/genetics , Oncolytic Virotherapy/methods , Osteosarcoma/therapy , Animals , Apoptosis/genetics , Bone Neoplasms/genetics , Bone Neoplasms/pathology , Bone Neoplasms/virology , Cell Line, Tumor , Female , HEK293 Cells , Hemagglutinins, Viral/biosynthesis , Humans , Mice , Neuraminidase/biosynthesis , Newcastle disease virus/enzymology , Osteosarcoma/genetics , Osteosarcoma/pathology , Osteosarcoma/virology , Random Allocation
15.
Viruses ; 9(10)2017 10 17.
Article in English | MEDLINE | ID: mdl-29039746

ABSTRACT

Osteosarcoma is the most frequent malignant disease of the bone. On the basis of early clinical experience in the 1960s with H-1 protoparvovirus (H-1PV) in osteosarcoma patients, this effective oncolytic virus was selected for systematic preclinical testing on various osteosarcoma cell cultures. A panel of five human osteosarcoma cell lines (CAL 72, H-OS, MG-63, SaOS-2, U-2OS) was tested. Virus oncoselectivity was confirmed by infecting non-malignant human neonatal fibroblasts and osteoblasts used as culture models of non-transformed mesenchymal cells. H-1PV was found to enter osteosarcoma cells and to induce viral DNA replication, transcription of viral genes, and translation to viral proteins. After H-1PV infection, release of infectious viral particles from osteosarcoma cells into the supernatant indicated successful viral assembly and egress. Crystal violet staining revealed progressive cytomorphological changes in all osteosarcoma cell lines. Infection of osteosarcoma cell lines with the standard H-1PV caused an arrest of the cell cycle in the G2 phase, and these lines had a limited capacity for standard H-1PV virus replication. The cytotoxicity of wild-type H-1PV virus towards osteosarcoma cells was compared in vitro with that of two variants, Del H-1PV and DM H-1PV, previously described as fitness variants displaying higher infectivity and spreading in human transformed cell lines of different origins. Surprisingly, wild-type H-1PV displayed the strongest cytostatic and cytotoxic effects in this analysis and thus seems the most promising for the next preclinical validation steps in vivo.


Subject(s)
Cell Death , H-1 parvovirus/physiology , Oncolytic Viruses/physiology , Osteosarcoma/pathology , Osteosarcoma/virology , Cell Cycle , Cell Line , Cell Line, Tumor , Humans , Oncolytic Virotherapy , Virus Replication
16.
Int J Cancer ; 141(6): 1257-1264, 2017 09 15.
Article in English | MEDLINE | ID: mdl-28568891

ABSTRACT

The poor prognosis of patients with advanced bone and soft-tissue sarcoma has not changed in the past several decades, highlighting the necessity for new therapeutic approaches. Immunotherapies, including oncolytic viral (OV) therapy, have shown great promise in a number of clinical trials for a variety of tumor types. However, the effective application of OV in treating sarcoma still remains to be demonstrated. Although few pre-clinical studies using distinct OVs have been performed and demonstrated therapeutic benefit in sarcoma models, a side-by-side comparison of clinically relevant OV platforms has not been performed. Four clinically relevant OV platforms (Reovirus, Vaccinia virus, Herpes-simplex virus and Rhabdovirus) were screened for their ability to infect and kill human and canine sarcoma cell lines in vitro, and human sarcoma specimens ex vivo. In vivo treatment efficacy was tested in a murine model. The rhabdovirus MG1 demonstrated the highest potency in vitro. Ex vivo, MG1 productively infected more than 80% of human sarcoma tissues tested, and treatment in vivo led to a significant increase in long-lasting cures in sarcoma-bearing mice. Importantly, MG1 treatment induced the generation of memory immune response that provided protection against a subsequent tumor challenge. This study opens the door for the use of MG1-based oncolytic immunotherapy strategies as treatment for sarcoma or as a component of a combined therapy.


Subject(s)
Oncolytic Virotherapy/methods , Rhabdoviridae/physiology , Sarcoma/therapy , Sarcoma/virology , Animals , Bone Neoplasms/therapy , Bone Neoplasms/virology , Cell Line, Tumor , Dogs , Female , Humans , Mice , Mice, Inbred BALB C , Osteosarcoma/therapy , Osteosarcoma/virology , Sarcoma, Ewing/therapy , Sarcoma, Ewing/virology , Sarcoma, Synovial/therapy , Sarcoma, Synovial/virology
17.
J Virol ; 91(9)2017 05 01.
Article in English | MEDLINE | ID: mdl-28179534

ABSTRACT

Human herpes simplex virus 1 (HSV-1) is a widespread pathogen, with 80% of the population being latently infected. To successfully evade the host, the virus has evolved strategies to counteract antiviral responses, including the gene-silencing and innate immunity machineries. The immediately early protein of the virus, infected cell protein 0 (ICP0), plays a central role in these processes. ICP0 blocks innate immunity, and one mechanism is by degrading hostile factors with its intrinsic E3 ligase activity. ICP0 also functions as a promiscuous transactivator, and it blocks repressor complexes to enable viral gene transcription. For these reasons, the growth of a ΔICP0 virus is impaired in most cells, except cells of the human osteosarcoma cell line U2OS, and it is only partially impaired in cells of the human osteosarcoma cell line Saos-2. We found that the two human osteosarcoma cell lines that supported the growth of the ΔICP0 virus failed to activate innate immune responses upon treatment with 2'3'-cyclic GAMP (2'3'-cGAMP), the natural agonist of STING (i.e., stimulator of interferon genes) or after infection with the ΔICP0 mutant virus. Innate immune responses were restored in these cells by transient expression of the STING protein but not after overexpression of interferon-inducible protein 16 (IFI16). Restoration of STING expression resulted in suppression of ΔICP0 virus gene expression and a decrease in viral yields. Overexpression of IFI16 also suppressed ΔICP0 virus gene expression, albeit to a lesser extent than STING. These data suggest that the susceptibility of U2OS and Saos-2 cells to the ΔICP0 HSV-1 is in part due to an impaired STING pathway.IMPORTANCE The DNA sensor STING plays pivotal role in controlling HSV-1 infection both in cell culture and in mice. The HSV-1 genome encodes numerous proteins that are dedicated to combat host antiviral responses. The immediate early protein of the virus ICP0 plays major role in this process as it targets hostile host proteins for degradation with its E3 ligase activity, and it disrupts repressor complexes via protein-protein interaction to enable viral gene transcription. Therefore, the ΔICP0 HSV-1 virus is defective for growth in most cells, except the human osteosarcoma cell lines U2OS and Saos-2. We found that both cell lines that support ΔICP0 virus infection have defects in the STING DNA-sensing pathway, which partially accounts for the rescue of the ΔICP0 virus growth. Restoration of STING expression in these cells rescued innate immunity and suppressed ΔICP0 virus infection. This study underscores the importance of STING in the control of HSV-1.


Subject(s)
Bone Neoplasms/pathology , Immediate-Early Proteins/genetics , Membrane Proteins/genetics , Osteosarcoma/pathology , Simplexvirus , Ubiquitin-Protein Ligases/genetics , Bone Neoplasms/genetics , Bone Neoplasms/virology , Cell Line, Tumor , Cyclic GMP/pharmacology , Humans , Immunity, Innate/genetics , Immunity, Innate/immunology , Membrane Proteins/agonists , Membrane Proteins/metabolism , Nuclear Proteins/metabolism , Osteosarcoma/genetics , Osteosarcoma/virology , Phosphoproteins/metabolism , Simplexvirus/genetics , Simplexvirus/growth & development , Simplexvirus/immunology , Trans-Activators/genetics , Virus Replication/genetics
18.
Vet Radiol Ultrasound ; 57(5): 551-6, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27374864

ABSTRACT

Coarsely fractionated radiation is commonly used as a method for pain control in dogs with appendicular osteosarcoma, however there is little published information on optimal protocols. The aim of this retrospective, descriptive study was to report safety and toxicity findings in a sample of dogs with appendicular osteosarcoma that had been treated with a radiation scheme of 10 Gy delivered over two consecutive days for a total of 20 Gy. Dogs were included in the study if they had osteosarcoma that was treated with the aforementioned protocol. Dogs were excluded if treated with the same protocol for any other bone tumor besides osteosarcoma or inadequate follow-up. Thirteen of the 14 patients received adjuvant therapy with pamidronate and a nonsteroidal anti-inflammatory. Nine dogs received adjuvant chemotherapy with carboplatin after radiation was complete. Within a median of 14 days, 92.8% of dogs subjectively had improved pain control. Median duration of response (DOR) was 80 days (range 20-365). The majority of patients developed VRTOG grade one toxicity, primarily alopecia. Five dogs (35.7%) developed pathologic fracture postradiation treatment. Timing of fracture was variable ranging from 24 to 250 days. This radiation protocol was well tolerated, with minimal toxicity, subjectively improved survival time, and had the benefit of being completed in two consecutive days.


Subject(s)
Bone Neoplasms/veterinary , Dog Diseases/radiotherapy , Osteosarcoma/virology , Radiography, Thoracic/veterinary , Animals , Bone Density Conservation Agents/administration & dosage , Bone Neoplasms/etiology , Bone Neoplasms/radiotherapy , Combined Modality Therapy/adverse effects , Combined Modality Therapy/veterinary , Diphosphonates/administration & dosage , Dog Diseases/etiology , Dogs , Dose-Response Relationship, Radiation , Female , Forelimb , Hindlimb , Male , Osteosarcoma/etiology , Osteosarcoma/radiotherapy , Pamidronate , Retrospective Studies
19.
Clin Cancer Res ; 22(9): 2217-25, 2016 05 01.
Article in English | MEDLINE | ID: mdl-26603261

ABSTRACT

PURPOSE: Osteosarcoma is the most common malignant bone tumor in children and adolescents. Despite aggressive chemotherapy, more than 30% of patients do not respond and develop bone or lung metastasis. Oncolytic adenoviruses engineered to specifically destroy cancer cells are a feasible option for osteosarcoma treatment. VCN-01 is a replication-competent adenovirus specifically engineered to replicate in tumors with a defective RB pathway, presents an enhanced infectivity through a modified fiber and an improved distribution through the expression of a soluble hyaluronidase. The aim of this study is to elucidate whether the use of VCN-01 would be an effective therapeutic strategy for pediatric osteosarcoma. EXPERIMENTAL DESIGN: We used osteosarcoma cell lines established from patients with metastatic disease (531MII, 678R, 588M, and 595M) and a commercial cell line (143B). MTT assays were carried out to evaluate the cytotoxicity of VCN-01. Hexon assays were used to evaluate the replication of the virus. Western blot analysis was performed to assess the expression levels of viral proteins and autophagic markers. The antitumor effect of VCN-01 was evaluated in orthotopic and metastatic osteosarcoma murine animal models. RESULTS: This study found that VCN-01, a new generation genetically modified oncolytic adenovirus, administered locally or systemically, had a potent antisarcoma effect in vitro and in vivo in mouse models of intratibial and lung metastatic osteosarcoma. Moreover, VCN-01 administration showed a safe toxicity profile. CONCLUSIONS: These results uncover VCN-01 as a promising strategy for osteosarcoma, setting the bases to propel a phase I/II trial for kids with this disease. Clin Cancer Res; 22(9); 2217-25. ©2015 AACR.


Subject(s)
Adenoviridae/genetics , Bone Neoplasms/virology , Oncolytic Viruses/genetics , Osteosarcoma/virology , Animals , Bone Neoplasms/pathology , Cell Line , Cell Line, Tumor , Female , Genetic Therapy/methods , HEK293 Cells , Humans , Lung Neoplasms/pathology , Lung Neoplasms/virology , Mice , Mice, Nude , Oncolytic Virotherapy/methods , Pediatrics , Virus Replication/genetics , Xenograft Model Antitumor Assays/methods
20.
Proteomics ; 15(12): 1957-67, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25809282

ABSTRACT

The herpes simplex virus 1 virion protein 16 (VP16) tegument protein forms a transactivation complex with the cellular proteins host cell factor 1 (HCF-1) and octamer-binding transcription factor 1 (Oct-1) upon entry into the host cell. VP16 has also been shown to interact with a number of virion tegument proteins and viral glycoprotein H to promote viral assembly, but no comprehensive study of the VP16 proteome has been performed at early times postinfection. We therefore performed a proteomic analysis of VP16-interacting proteins at 3 h postinfection. We confirmed the interaction of VP16 with HCF-1 and a large number of cellular Mediator complex proteins, but most surprisingly, we found that the major viral protein associating with VP16 is the infected cell protein 4 (ICP4) immediate-early (IE) transactivator protein. These results raise the potential for a new function for VP16 in associating with the IE ICP4 and playing a role in transactivation of early and late gene expression, in addition to its well-documented function in transactivation of IE gene expression.


Subject(s)
Bone Neoplasms/metabolism , Fibroblasts/metabolism , Herpes Simplex Virus Protein Vmw65/metabolism , Herpes Simplex/metabolism , Immediate-Early Proteins/metabolism , Osteosarcoma/metabolism , Proteome/analysis , Animals , Bone Neoplasms/pathology , Bone Neoplasms/virology , Cells, Cultured , Chlorocebus aethiops , Fibroblasts/cytology , Fibroblasts/virology , Herpes Simplex/pathology , Herpes Simplex/virology , Herpesvirus 1, Human/physiology , Humans , Immunoblotting , Immunoprecipitation , Osteosarcoma/pathology , Osteosarcoma/virology , Proteomics/methods , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Trans-Activators/metabolism , Vero Cells
SELECTION OF CITATIONS
SEARCH DETAIL
...