Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Mol Ther ; 28(2): 422-430, 2020 02 05.
Article in English | MEDLINE | ID: mdl-31843447

ABSTRACT

Short hairpin RNAs that are delivered by recombinant adeno-associated virus (rAAV) have the potential to elicit long-term RNAi therapy for human disease. However, the discovery that short hairpin sequences can cause truncation of the rAAV genome calls into question the efficiency and gene-silencing specificity of this strategy in humans. Here, we report that embedding the guide strand of a small silencing RNA into an artificial microRNA (miRNA) scaffold derived from mouse miRNA-33 ensures rAAV genomic integrity and reduces off-targeting by 10-fold, while maintaining effective in vivo target gene repression in mice.


Subject(s)
Dependovirus/genetics , Gene Silencing , Genetic Vectors/genetics , MicroRNAs/genetics , Animals , Genome, Viral , Humans , Mice , Nucleic Acid Conformation , RNA Interference , RNA Stability , RNA, Small Interfering/genetics , RNA, Viral
2.
Cancer Cell Int ; 20(1): 532, 2020 Nov 02.
Article in English | MEDLINE | ID: mdl-33292203

ABSTRACT

BACKGROUND: Viruses are obligate parasites that depend on host cells to provide the energy and molecular precursors necessary for successful infection. The main component of virus-induced metabolic reprogramming is the activation of glycolysis, which provides biomolecular resources for viral replication. However, little is known about the crosstalk between oncolytic viruses and host glycolytic processes. METHODS: A MTT assay was used to detect M1 virus-induced cell killing. Flow cytometry was used to monitor infection of M1 virus expressing the GFP reporter gene. qPCR and western blotting were used to detect gene expression. RNA sequencing was performed to evaluate gene expression under different drug treatments. Scanning electron microscopy was performed to visualize the endoplasmic reticulum (ER). Caspase activity was detected. Last, a mouse xenograft model was established to evaluate the antitumor effect in vivo. Most data were analyzed with a two-tailed Student's t test or one-way ANOVA with Dunnett's test for pairwise comparisons. Tumor volumes were analyzed by repeated measures of ANOVA. The Wilcoxon signed-rank test was used to compare nonnormally distributed data. RESULTS: Here, we showed that the glucose analog 2-deoxy-D-glucose (2-DG) inhibited infection by M1 virus, which we identified as a novel type of oncolytic virus, and decreased its oncolytic effect, indicating the dependence of M1 replication on glycolysis. In contrast, lonidamine, a reported hexokinase 2 (HK2) inhibitor, enhanced the infection and oncolytic effect of M1 virus independent of HK2. Further transcriptomic analysis revealed that downregulation of the antiviral immune response contributes to the lonidamine-mediated potentiation of the infection and oncolytic effect of M1 virus, and that MYC is the key factor in the pool of antiviral immune response factors inhibited by lonidamine. Moreover, lonidamine potentiated the irreversible ER stress-mediated apoptosis induced by M1 virus. Enhancement of M1's oncolytic effect by lonidamine was also identified in vivo. CONCLUSIONS: This research demonstrated the dependence of M1 virus on glycolysis and identified a candidate synergist for M1 virotherapy.

3.
Mol Ther ; 25(6): 1363-1374, 2017 06 07.
Article in English | MEDLINE | ID: mdl-28462820

ABSTRACT

Short hairpin (sh)RNAs delivered by recombinant adeno-associated viruses (rAAVs) are valuable tools to study gene function in vivo and a promising gene therapy platform. Our data show that incorporation of shRNA transgenes into rAAV constructs reduces vector yield and produces a population of truncated and defective genomes. We demonstrate that sequences with hairpins or hairpin-like structures drive the generation of truncated AAV genomes through a polymerase redirection mechanism during viral genome replication. Our findings reveal the importance of genomic secondary structure when optimizing viral vector designs. We also discovered that shDNAs could be adapted to act as surrogate mutant inverted terminal repeats (mTRs), sequences that were previously thought to be required for functional self-complementary AAV vectors. The use of shDNAs as artificial mTRs opens the door to engineering a new generation of AAV vectors with improved potency, genetic stability, and safety for both preclinical studies and human gene therapy.


Subject(s)
DNA, Viral , Dependovirus/genetics , Genetic Vectors/genetics , Genome, Viral , Inverted Repeat Sequences , Animals , Cell Line , DNA Replication , Gene Expression , Gene Order , Gene Transfer Techniques , Genes, Reporter , Humans , Male , Mice , Models, Biological , Nucleic Acid Conformation , Plasmids/genetics , RNA, Small Interfering , Sequence Analysis, DNA , Sequence Deletion , Transduction, Genetic
4.
Signal Transduct Target Ther ; 7(1): 100, 2022 04 08.
Article in English | MEDLINE | ID: mdl-35393389

ABSTRACT

Over the last decade, oncolytic virus (OV) therapy has shown its promising potential in tumor treatment. The fact that not every patient can benefit from it highlights the importance for defining biomarkers that help predict patients' responses. As particular self-amplifying biotherapeutics, the anti-tumor effects of OVs are highly dependent on the host factors for viral infection and replication. By using weighted gene co-expression network analysis (WGCNA), we found matrix remodeling associated 8 (MXRA8) is positively correlated with the oncolysis induced by oncolytic virus M1 (OVM). Consistently, MXRA8 promotes the oncolytic efficacy of OVM in vitro and in vivo. Moreover, the interaction of MXRA8 and OVM studied by single-particle cryo-electron microscopy (cryo-EM) showed that MXRA8 directly binds to this virus. Therefore, MXRA8 acts as the entry receptor of OVM. Pan-cancer analysis showed that MXRA8 is abundant in most solid tumors and is highly expressed in tumor tissues compared with adjacent normal ones. Further study in cancer cell lines and patient-derived tumor tissues revealed that the tumor selectivity of OVM is predominantly determined by a combinational effect of the cell membrane receptor MXRA8 and the intracellular factor, zinc-finger antiviral protein (ZAP). Taken together, our study may provide a novel dual-biomarker for precision medicine in OVM therapy.


Subject(s)
Neoplasms , Oncolytic Virotherapy , Oncolytic Viruses , Cryoelectron Microscopy , Humans , Immunoglobulins , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/therapy , Oncolytic Viruses/genetics
5.
Hum Gene Ther ; 31(21-22): 1203-1213, 2020 11.
Article in English | MEDLINE | ID: mdl-32829653

ABSTRACT

Oncolytic viruses are emerging as important tools for immunotherapy for cancer treatment; however, most of the clinically tested oncolytic candidates are still administered by intratumoral injection, and new viruses capable of intravenous injection are urgently needed. The M1 virus is a positive-sense single-stranded RNA virus that belongs to the alphavirus family, and it was identified as an oncolytic virus that can selectively replicate in and kill tumor cells after intravenous injection. To further develop M1 for clinical research through intravenous injection, we systematically investigated the biodistribution characteristics of the M1 virus in normal rats, cynomolgus monkeys, and tumor-bearing immunocompromised mice. The data showed that the M1 virus was eliminated gradually from normal tissue but replicated and increased rapidly in tumor tissue. More importantly, the virus also infiltrated the blood-brain barrier and specifically replicated in and killed malignant glioma in immunocompetent mice. Our data proved the tumor selectivity and safety of the M1 virus, supporting its further clinical development.


Subject(s)
Brain/metabolism , Glioma/therapy , Oncolytic Virotherapy/methods , Oncolytic Viruses/metabolism , Animals , Female , Glioma/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Mice, SCID , Rats , Rats, Sprague-Dawley , Tissue Distribution
6.
Hum Gene Ther ; 29(8): 950-961, 2018 08.
Article in English | MEDLINE | ID: mdl-28750564

ABSTRACT

Oncolytic virotherapy is a novel and intriguing treatment strategy for cancer therapy. However, the clinical potential of oncolytic virus as single agent is limited. M1 virus is a promising oncolytic virus that has been tested in preclinical studies. In this study, we investigated the effect of the combination use of M1 virus and Bcl-2 family inhibitors. A chemical compounds screening including ten Bcl-2 family inhibitors demonstrated that pan-Bcl-2 inhibitors selectively augmented M1 virus oncolysis in cancer cells at very low doses. The mechanism of the enhanced antitumor effect of pan-Bcl-2 inhibitors with M1 virus is mainly due to the inhibition of Bcl-xL, which synergizes with M1-induced upregulation of Bak to trigger apoptosis. In xenograft mouse models and patient-derived tumor tissues, the combination of M1 and pan-Bcl-2 inhibitors significantly inhibited tumor growth and prolonged survival, suggesting the potential therapeutic value of this strategy. These findings offer insights into the synergy between Bcl-xL inhibition and oncolytic virus M1 as a combination anticancer treatment modality.


Subject(s)
Neoplasms/genetics , Oncolytic Virotherapy/methods , Oncolytic Viruses/drug effects , Proto-Oncogene Proteins c-bcl-2/genetics , Animals , Apoptosis/drug effects , Cell Line, Tumor , Combined Modality Therapy , Humans , Mice , Mitochondria/drug effects , Neoplasms/drug therapy , Neoplasms/virology , Oncolytic Viruses/genetics , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Xenograft Model Antitumor Assays
7.
Sci Transl Med ; 9(404)2017 Aug 23.
Article in English | MEDLINE | ID: mdl-28835517

ABSTRACT

Oncolytic virotherapy is rapidly progressing through clinical evaluation. However, the therapeutic efficacy of oncolytic viruses in humans has been less than expected from preclinical studies. We describe an anticancer drug screen for compounds that enhance M1 oncolytic virus activity in hepatocellular carcinoma (HCC). An inhibitor of the valosin-containing protein (VCP) was identified as the top sensitizer, selectively increasing potency of the oncolytic virus up to 3600-fold. Further investigation revealed that VCP inhibitors cooperated with M1 virus-suppressed inositol-requiring enzyme 1α (IRE1α)-X-box binding protein 1 (XBP1) pathway and triggered irresolvable endoplasmic reticulum (ER) stress, subsequently promoting robust apoptosis in HCC. We show that VCP inhibitor improved the oncolytic efficacy of M1 virus in several mouse models of HCC and primary HCC tissues. Finally, this combinatorial therapeutic strategy was well tolerated in nonhuman primates. Our study identifies combined VCP inhibition and oncolytic virus as a potential treatment for HCC and demonstrates promising therapeutic potential.


Subject(s)
Antineoplastic Agents/metabolism , Carcinoma, Hepatocellular/therapy , Carcinoma, Hepatocellular/virology , Liver Neoplasms/therapy , Liver Neoplasms/virology , Oncolytic Viruses/metabolism , Valosin Containing Protein/antagonists & inhibitors , Animals , Apoptosis , Bystander Effect , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Combined Modality Therapy , Endoplasmic Reticulum Stress , Endoribonucleases/metabolism , High-Throughput Screening Assays , Humans , Liver Neoplasms/pathology , Oncolytic Viruses/pathogenicity , Primates , Protein Serine-Threonine Kinases/metabolism , Valosin Containing Protein/metabolism , X-Box Binding Protein 1/metabolism
8.
Neurosci Lett ; 407(3): 214-8, 2006 Oct 30.
Article in English | MEDLINE | ID: mdl-16973276

ABSTRACT

Cerebellar granule neurons (CGNs) depend on potassium depolarization for survival and undergo apoptosis when deprived of depolarizing concentration of potassium. Extracellular signal-regulated kinases (ERK1/2) are thought to be activated in response to potassium depolarization and responsible for the activity-dependent survival in CGNs, but one recent study has revealed that ERK1/2 is activated by potassium deprivation and is required for apoptosis of CGNs. In this study we showed that ERK1/2 was inactivated, rather than activated, by potassium deprivation, indicating a lack of ERK1/2 involvement in potassium deprivation-induced apoptosis. Furthermore, suppression of potassium depolarization-induced activation of ERK1/2 with chemical inhibitor U0126 or PD98059 had no influence on the pro-survival effect of potassium depolarisation. Thus, ERK1/2 was not required for potassium depolarization-dependent survival of CGNs. Taken together, our findings suggest that ERK1/2 is not involved in activity-dependent survival or apoptosis of CGNs.


Subject(s)
Apoptosis , Cerebellum/cytology , Extracellular Signal-Regulated MAP Kinases/physiology , Neurons/physiology , Animals , Animals, Newborn , Cell Survival , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/metabolism , MAP Kinase Kinase Kinases/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Neurons/enzymology , Rats , Rats, Sprague-Dawley
9.
Hum Gene Ther ; 27(9): 700-11, 2016 09.
Article in English | MEDLINE | ID: mdl-27296553

ABSTRACT

Cancers figure among the leading causes of morbidity and mortality worldwide. The number of new cases is expected to rise by about 70% over the next 2 decades. Development of novel therapeutic agents is urgently needed for clinical cancer therapy. Alphavirus M1 is a Getah-like virus isolated from China with a genome of positive single-strand RNA. We have previously identified that alphavirus M1 is a naturally existing oncolytic virus with significant anticancer activity against different kinds of cancer (e.g., liver cancer, bladder cancer, and colon cancer). To support the incoming clinical trial of intravenous administration of alphavirus M1 to cancer patients, we assessed the safety of M1 in adult nonhuman primates. We previously presented the genome sequencing data of the cynomolgus macaques (Macaca fascicularis), which was demonstrated as an ideal animal species for virus infection study. Therefore, we chose cynomolgus macaques of either sex for the present safety study of oncolytic virus M1. In the first round of administration, five experimental macaques were intravenously injected with six times of oncolytic virus M1 (1 × 10(9) pfu/dose) in 1 week, compared with five vehicle-injected control animals. The last two rounds of injections were further completed in the following months in the same way as the first round. Body weight, temperature, complete blood count, clinical biochemistries, cytokine profiles, lymphocytes subsets, neutralizing antibody, and clinical symptoms were closely monitored at different time points. Magnetic resonance imaging was also performed to assess the possibility of encephalitis or arthritis. As a result, no clinical, biochemical, immunological, or medical imaging or other pathological evidence of toxicity was found during the whole process of the study. Our results in cynomolgus macaques suggested the safety of intravenous administration of oncolytic virus M1 in cancer patients in the future.


Subject(s)
Alphavirus/immunology , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Genetic Vectors/administration & dosage , Oncolytic Viruses/immunology , Alphavirus/genetics , Animals , Disease Models, Animal , Female , Humans , Injections, Intravenous , Macaca fascicularis , Male , Oncolytic Viruses/genetics
10.
Neurosci Lett ; 375(1): 7-12, 2005 Feb 25.
Article in English | MEDLINE | ID: mdl-15664113

ABSTRACT

Bcl-2-interacting mediator of cell death (Bim), a proapoptotic BH3-only protein, plays a critical role in neuronal apoptosis. Cerebellar granule neurons (CGNs) depend on activity for their survival and undergo apoptosis when deprived of depolarizing concentration of KCl. While it has been proposed that the activation of c-Jun NH2-terminal protein kinase (JNK)/c-Jun pathway contributes to the upregulation of bim gene in neurons subjected to survival signaling withdrawal, here we show that neither inhibition of JNK activity nor expression of dominant-negative c-Jun suppresses the expression of bim gene induced by activity deprivation in CGNs. We conclude that induction of bim gene is independent of the activation of JNK/c-Jun signaling pathway by activity deprivation during apoptosis of CGNs.


Subject(s)
Apoptosis/physiology , Cerebellum/cytology , Gene Expression Regulation/physiology , Neurons/metabolism , Neuropeptides/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Animals , Animals, Newborn , Apoptosis Regulatory Proteins , Blotting, Western/methods , Cells, Cultured , Dose-Response Relationship, Drug , Drug Interactions/physiology , Enzyme Activation/physiology , Enzyme Inhibitors/pharmacology , Gene Expression Regulation/drug effects , Green Fluorescent Proteins/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Neural Inhibition/drug effects , Neuropeptides/genetics , Phosphorylation , Potassium Chloride/pharmacology , RNA, Messenger/biosynthesis , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction/methods , Serine/metabolism , Signal Transduction/physiology , Time Factors , Transfection/methods
11.
Mol Cell Biol ; 29(9): 2431-42, 2009 May.
Article in English | MEDLINE | ID: mdl-19255142

ABSTRACT

The activator protein 1 (AP-1) transcription factor c-Jun is crucial for neuronal apoptosis. However, c-Jun dimerization partners and the regulation of these proteins in neuronal apoptosis remain unknown. Here we report that c-Jun-mediated neuronal apoptosis requires the concomitant activation of activating transcription factor-2 (ATF2) and downregulation of c-Fos. Furthermore, we have observed that c-Jun predominantly heterodimerizes with ATF2 and that the c-Jun/ATF2 complex promotes apoptosis by triggering ATF activity. Inhibition of c-Jun/ATF2 heterodimerization using dominant negative mutants, small hairpin RNAs, or decoy oligonucleotides was able to rescue neurons from apoptosis, whereas constitutively active ATF2 and c-Jun mutants were found to synergistically stimulate apoptosis. Bimolecular fluorescence complementation analysis confirmed that, in living neurons, c-Fos downregulation facilitates c-Jun/ATF2 heterodimerization. A chromatin immunoprecipitation assay also revealed that c-Fos expression prevents the binding of c-Jun/ATF2 heterodimers to conserved ATF sites. Moreover, the presence of c-Fos is able to suppress the expression of c-Jun/ATF2-mediated target genes and, therefore, apoptosis. Taken together, our findings provide evidence that potassium deprivation-induced neuronal apoptosis is mediated by concurrent upregulation of c-Jun/ATF2 heterodimerization and downregulation of c-Fos expression. This paradigm demonstrates opposing roles for ATF2 and c-Fos in c-Jun-mediated neuronal apoptosis.


Subject(s)
Activating Transcription Factor 2/metabolism , Apoptosis/physiology , Neurons/physiology , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Activating Transcription Factor 2/chemistry , Activating Transcription Factor 2/genetics , Animals , Cells, Cultured , Dimerization , Neurons/cytology , Potassium/metabolism , Protein Structure, Quaternary , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-jun/chemistry , Proto-Oncogene Proteins c-jun/genetics , RNA Interference , Rats , Rats, Sprague-Dawley , Signal Transduction/physiology
12.
Sheng Wu Gong Cheng Xue Bao ; 23(6): 971-5, 2007 Nov.
Article in Zh | MEDLINE | ID: mdl-18257221

ABSTRACT

Heat shock factor 1 (HSF1) is the key protein in regulating stress response. It can be activated under heat, oxidative or another stress conditions. Dominant-positive and dominant-negative HSF1 are two types of HSF1 mutants. Both of them gain the DNA binding activity in the absence of stress. In addition, dominant-positive HSF1 acquires transcriptional activity, which dominant-negative HSF1 does not acquire. In this paper, the progress of using these HSF1 mutants in the research of cancer, neurodegenerative disorders and cardiovascular diseases will be discussed.


Subject(s)
Genetic Therapy , Heat-Shock Proteins/genetics , Mutant Proteins/genetics , Neoplasms/therapy , Neurodegenerative Diseases/therapy , Heat-Shock Proteins/therapeutic use , Humans
SELECTION OF CITATIONS
SEARCH DETAIL