Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 68
Filter
1.
Hum Gene Ther ; 32(3-4): 158-165, 2021 02.
Article in English | MEDLINE | ID: mdl-33504253

ABSTRACT

Alphavirus M1 is a promising oncolytic virus for cancer therapy. Here, we constructed a fluorescent reporter virus for real-time visualization and quantification of M1 virus both in vitro and in vivo. The reporter-encoding M1 virus maintained the characteristics of parental virus in the aspects of structure, replication capacity, the feature to induce cytopathic cell death, and the property of tumor targeting. The fluorescence is positively correlated with virus replication both in vitro and in vivo. More importantly, the reporter can be stably expressed for at least 10 generations in a serial passage assay. In summary, we successfully constructed stable and authentic reporter viruses for studying M1 virus and provided a feasible technical route for gene modification of oncolytic virus M1.


Subject(s)
Alphavirus , Neoplasms , Oncolytic Virotherapy , Oncolytic Viruses , Alphavirus/genetics , Cell Line, Tumor , Humans , Neoplasms/genetics , Neoplasms/therapy , Oncolytic Viruses/genetics , Virus Replication
2.
Mol Oncol ; 13(7): 1589-1604, 2019 07.
Article in English | MEDLINE | ID: mdl-31162799

ABSTRACT

Activation of the cyclic adenosine monophosphate/protein kinase A (cAMP/PKA) pathway induces glial differentiation of glioblastoma (GBM) cells, but the mechanism by which microRNA (miRNA) regulate this process remains poorly understood. In this study, by performing miRNA genomics and loss- and gain-of-function assays in dibutyryl-cAMP-treated GBM cells, we identified a critical negative regulator, hsa-miR-1275, that modulates a set of genes involved in cancer progression, stem cell maintenance, and cell maturation and differentiation. Additionally, we confirmed that miR-1275 directly and negatively regulates the protein expression of glial fibrillary acidic protein (GFAP), a marker of mature astrocytes. Of note, tri-methyl-histone H3 (Lys27) (H3K27me3), downstream of the PKA/polycomb repressive complex 2 (PRC2) pathway, accounts for the downregulation of miR-1275. Furthermore, decreased miR-1275 expression and induction of GFAP expression were also observed in dibutyryl-cAMP-treated primary cultured GBM cells. In a patient-derived glioma stem cell tumor model, a cAMP elevator and an inhibitor of H3K27me3 methyltransferase inhibited tumor growth, induced differentiation, and reduced expression of miR-1275. In summary, our study shows that epigenetic inhibition of miR-1275 by the cAMP/PKA/PRC2/H3K27me3 pathway mediates glial induction of GBM cells, providing a new mechanism and novel targets for differentiation-inducing therapy.


Subject(s)
Brain Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Histones/metabolism , MicroRNAs/genetics , Animals , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Down-Regulation , Female , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Methylation , Mice, Inbred BALB C , Neuroglia/metabolism , Neuroglia/pathology , Transcriptome
3.
Mol Pharm ; 16(2): 779-785, 2019 02 04.
Article in English | MEDLINE | ID: mdl-30604617

ABSTRACT

Oncolytic viral therapy is an attractive novel strategy for cancer therapy. As a natural alphavirus, oncolytic virus M1 is able to infect and kill various zinc finger antiviral protein (ZAP)-deficient tumor cells selectively, while leaving normal cells undamaged. However, M1 can trigger the production of neutralizing antibodies that dramatically weaken its antitumor effect. In order to attenuate immunogenicity of the therapeutic M1 virus, we encapsulated it into liposomes (referred to as M-LPO) using the thin-film hydration method. The effect of anti-M1 neutralizing antibody on M-LPO was examined in LoVo and Hep 3B cell lines. In the absence of neutralizing antibodies, treating cells with naked M1, blank liposomes (LPO), M-LPO, or a simple mixture of M1 and liposomes (LPO+M1) inhibited cell growth. In the presence of neutralizing antibodies, only M-LPO inhibited cell growth. After intravenous administration, M-LPO reduced the production of the M1-neutralizing antibody and the corresponding immune response. Analysis of the M-LPO uptake by cells was examined by confocal microscopy using M1 labeled with FITC and liposomal shells labeled with RhB. The results suggest that M1 may be released from liposomes before or after M-LPO internalization. Taken together, our results suggest that encapsulating oncolytic virus M1 in liposomes may reduce intrinsic viral immunogenicity for improved anticancer therapy.


Subject(s)
Liposomes/chemistry , Oncolytic Viruses/physiology , Animals , Antibodies, Neutralizing/metabolism , Cell Line, Tumor , Cell Survival/physiology , Female , Humans , Mice , Mice, Inbred BALB C , Microscopy, Electron, Transmission , Oncolytic Virotherapy/methods , Oncolytic Viruses/chemistry
4.
Nat Commun ; 9(1): 4342, 2018 10 18.
Article in English | MEDLINE | ID: mdl-30337542

ABSTRACT

Oncolytic virotherapy is a promising therapeutic strategy that uses replication-competent viruses to selectively destroy malignancies. However, the therapeutic effect of certain oncolytic viruses (OVs) varies among cancer patients. Thus, it is necessary to overcome resistance to OVs through rationally designed combination strategies. Here, through an anticancer drug screening, we show that DNA-dependent protein kinase (DNA-PK) inhibition sensitizes cancer cells to OV M1 and improves therapeutic effects in refractory cancer models in vivo and in patient tumour samples. Infection of M1 virus triggers the transcription of interferons (IFNs) and the activation of the antiviral response, which can be abolished by pretreatment of DNA-PK inhibitor (DNA-PKI), resulting in selectively enhanced replication of OV M1 within malignancies. Furthermore, DNA-PK inhibition promotes the DNA damage response induced by M1 virus, leading to increased tumour cell apoptosis. Together, our study identifies the combination of DNA-PKI and OV M1 as a potential treatment for cancers.


Subject(s)
Antiviral Agents/pharmacology , DNA Damage , DNA-Activated Protein Kinase/antagonists & inhibitors , Oncolytic Viruses/physiology , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Combined Modality Therapy , DNA-Activated Protein Kinase/metabolism , Drug Evaluation, Preclinical , Endoplasmic Reticulum Stress/drug effects , Humans , Mice, Inbred BALB C , Mice, Nude , Models, Biological , Oncolytic Virotherapy , Protein Kinase Inhibitors/pharmacology , Rats
6.
Nat Commun ; 9(1): 1524, 2018 04 18.
Article in English | MEDLINE | ID: mdl-29670091

ABSTRACT

Oncolytic virus is an attractive anticancer agent that selectively lyses cancer through targeting cancer cells rather than normal cells. Although M1 virus is effective against several cancer types, certain cancer cells present low sensitivity to it. Here we identified that most of the components in the cholesterol biosynthesis pathway are downregulated after M1 virus infection. Further functional studies illustrate that mevalonate/protein farnesylation/ras homolog family member Q (RHOQ) axis inhibits M1 virus replication. Further transcriptome analysis shows that RHOQ knockdown obviously suppresses Rab GTPase and ATP-mediated membrane transporter system, which may mediate the antiviral effect of RHOQ. Based on this, inhibition of the above pathway significantly enhances the anticancer potency of M1 virus in vitro, in vivo, and ex vivo. Our research provides an intriguing strategy for the rational combination of M1 virus with farnesyl transferase inhibitors to enhance therapeutic efficacy.


Subject(s)
Cholesterol/chemistry , Mevalonic Acid/antagonists & inhibitors , Mevalonic Acid/metabolism , Oncolytic Virotherapy , Oncolytic Viruses/physiology , Virus Replication , Animals , Cell Line, Tumor , Cell Survival , Down-Regulation , Farnesyltranstransferase/antagonists & inhibitors , Female , Gene Knockdown Techniques , HCT116 Cells , Humans , Mice , Mice, Inbred BALB C , Neoplasm Transplantation , Protein Prenylation , RNA Interference , RNA, Small Interfering/metabolism , rab GTP-Binding Proteins/metabolism
7.
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
8.
J Neurochem ; 144(2): 186-200, 2018 01.
Article in English | MEDLINE | ID: mdl-29205357

ABSTRACT

Hyperglycolysis, observed within the penumbra zone during brain ischemia, was shown to be detrimental for tissue survival because of lactate accumulation and reactive oxygen species overproduction in clinical and experimental settings. Recently, mounting evidence suggests that glycolytic reprogramming and induced metabolic enzymes can fuel the activation of peripheral immune cells. However, the possible roles and details regarding hyperglycolysis in neuroinflammation during ischemia are relatively poorly understood. Here, we investigated whether overactivated glycolysis could activate microglia and identified the crucial regulators of neuroinflammatory responses in vitro and in vivo. Using BV 2 and primary microglial cultures, we found hyperglycolysis and induction of the key glycolytic enzyme hexokinase 2 (HK2) were essential for microglia-mediated neuroinflammation under hypoxia. Mechanistically, HK2 up-regulation led to accumulated acetyl-coenzyme A, which accounted for the subsequent histone acetylation and transcriptional activation of interleukin (IL)-1ß. The inhibition and selective knockdown of HK2 in vivo significantly protected against ischemic brain injury by suppressing microglial activation and IL-1ß production in male Sprague-Dawley rats subjected to transient middle cerebral artery occlusion (MCAo) surgery. We provide novel insights for HK2 specifically serving as a neuroinflammatory determinant, thus explaining the neurotoxic effect of hyperglycolysis and indicating the possibility of selectively targeting HK2 as a therapeutic strategy in acute ischemic stroke.


Subject(s)
Brain Ischemia/enzymology , Brain Ischemia/genetics , Glycolysis/genetics , Hexokinase/genetics , Hexokinase/metabolism , Macrophage Activation/genetics , Microglia/enzymology , Stroke/enzymology , Stroke/genetics , Acetyl Coenzyme A/metabolism , Acetylation , Animals , Enzyme Induction/genetics , Hexokinase/biosynthesis , Histones/metabolism , Infarction, Middle Cerebral Artery/genetics , Infarction, Middle Cerebral Artery/metabolism , Inflammation/genetics , Interleukin-1beta/metabolism , Male , RNA Interference , Rats , Rats, Sprague-Dawley
9.
J Virol ; 92(6)2018 03 15.
Article in English | MEDLINE | ID: mdl-29263275

ABSTRACT

Oncolytic virotherapy is an emerging treatment modality that uses replication-competent viruses to destroy cancer cells. M1 is a naturally occurring alphavirus (Togaviridae) which shows potent oncolytic activities against many cancers. Accumulation of unfolded proteins during virus replication leads to a transcriptional/translational response known as the unfolded protein response (UPR), which might counteract the antitumor effect of the oncolytic virus. In this report, we show that either pharmacological or biological inhibition of IRE1α or PERK, but not ATF6, substantially increases the oncolytic effects of the M1 virus. Moreover, inhibition of IRE1α blocks M1 virus-induced autophagy, which restricts the antitumor effects of the M1 virus through degradation of viral protein, in glioma cells. In addition, IRE1α suppression significantly increases the oncolytic effect of M1 virus in an orthotopic glioma model. From a molecular pathology study, we found that IRE1α is expressed at lower levels in higher-grade gliomas, suggesting greater antitumor efficacy of the oncolytic virus M1. Taken together, these findings illustrate a defensive mechanism of glioma cells against the oncolytic virus M1 and identify possible approaches to enhance the oncolytic viral protein accumulation and the subsequent lysis of tumor cells.IMPORTANCE Although oncolytic virotherapy is showing great promise in clinical applications, not all patients are benefiting. Identifying inhibitory signals in refractory cancer cells for each oncolytic virus would provide a good chance to increase the therapeutic effect. Here we describe that infection with the oncolytic virus M1 triggers the unfolded protein response (UPR) and subsequent autophagy, while blocking the UPR-autophagy axis significantly potentiates the antitumor efficacy of M1 in vitro and in vivo A survey of cancer tissue banks revealed that IRE1α, a key element in the UPR pathway, is commonly downregulated in higher-grade human gliomas, suggesting favorable prospects for the application of M1. Our work provides a potential predictor and target for enhancement of the therapeutic effectiveness of the M1 virus. We predict that the mechanism-based combination therapy will promote cancer virotherapy in the future.


Subject(s)
Autophagy/immunology , Endoribonucleases/deficiency , Glioma/therapy , Neoplasm Proteins/deficiency , Oncolytic Virotherapy , Oncolytic Viruses , Protein Serine-Threonine Kinases/deficiency , Togaviridae , Animals , Autophagy/genetics , Cell Line, Tumor , Chlorocebus aethiops , Cricetinae , Endoribonucleases/immunology , Female , Glioma/genetics , Glioma/immunology , Humans , Mice, Inbred BALB C , Mice, Nude , Neoplasm Proteins/immunology , Protein Serine-Threonine Kinases/immunology , Unfolded Protein Response/genetics , Unfolded Protein Response/immunology , Vero Cells , Xenograft Model Antitumor Assays
10.
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
11.
Neuroscience ; 350: 110-123, 2017 05 14.
Article in English | MEDLINE | ID: mdl-28336411

ABSTRACT

Acute stress impairs the hippocampus-dependent spatial memory retrieval, and its synaptic mechanisms are associated with hippocampal CA1 long-term depression (LTD) enhancement in the adult rats. Endogenous hydrogen sulfide (H2S) is recognized as a novel gasotransmitter and has the neural protective roles. However, very little attention has been paid to understanding the effects of H2S on spatial memory retrieval impairment. We observed the protective effects of NaHS (a donor of H2S) against spatial memory retrieval impairment caused by acute stress and its synaptic mechanisms. Our results showed that NaHS abolished spatial memory retrieval impairment and hippocampal CA1 LTD enhancement caused by acute stress, but not by glutamate transporter inhibitor l-trans-pyrrolidine-2,4-dicarboxylic (tPDC), indicating that the activation of glutamate transporters is necessary for exogenous H2S to exert its roles. Moreover, NaHS restored the decreased glutamate uptake in the hippocampal CA1 synaptosomal fraction caused by acute stress. Dithiothreitol (DTT, a disulfide reducing agent) abolished a decrease in the glutamate uptake caused by acute stress, and NaHS eradicated the decreased glutamate uptake caused by 5,5'-dithio-bis(2-nitrobenzoic)acid (DTNB, a thiol oxidizing agent), collectively, revealing that exogenous H2S increases glutamate uptake by reducing disulfide bonds of the glutamate transporters. Additionally, NaHS inhibited the increased expression level of phosphorylated c-Jun-N-terminal kinase (JNK) in the hippocampal CA1 region caused by acute stress. The JNK inhibitor SP600125 eliminated spatial memory retrieval impairment, hippocampal CA1 LTD enhancement and the decreased glutamate uptake caused by acute stress, indicating that exogenous H2S exerts these roles by inhibiting the activation of JNK signaling pathway.


Subject(s)
Glutamic Acid/metabolism , Hippocampus/drug effects , Hydrogen Sulfide/pharmacology , Spatial Memory/drug effects , Stress, Psychological/metabolism , Animals , Hippocampus/metabolism , Long-Term Synaptic Depression/physiology , Male , Memory Disorders/metabolism , Neuronal Plasticity/physiology , Rats, Sprague-Dawley
12.
Can J Physiol Pharmacol ; 95(5): 463-473, 2017 May.
Article in English | MEDLINE | ID: mdl-28177695

ABSTRACT

Diallyl trisulfide (DATS), a major garlic derivative, inhibits cell proliferation and triggers apoptosis in a variety of cancer cell lines. However, the effects of DATS on hepatic stellate cells (HSCs) remain unknown. The aim of this study was to analyze the effects of DATS on cell proliferation and apoptosis, as well as the protein expression profile in rat HSCs. Rat HSCs were treated with or without 12 and 24 µg/mL DATS for various time intervals. Cell proliferation and apoptosis were determined using tetrazolium dye (MTT) colorimetric assay, bromodeoxyuridine (5-bromo-2'-deoxyuridine; BrdU) assay, Hoechst 33342 staining, electroscopy, and flow cytometry. Protein expression patterns in HSCs were systematically studied using 2-dimensional electrophoresis and mass spectrometry. DATS inhibited cell proliferation and induced apoptosis of HSCs in a time-dependent manner. We observed clear morphological changes in apoptotic HSCs and dramatically increased annexin V-positive - propidium iodide negative apoptosis compared with the untreated control group. Twenty-one significant differentially expressed proteins, including 9 downregulated proteins and 12 upregulated proteins, were identified after DATS administration, and most of them were involved in apoptosis. Our results suggest that DATS is an inducer of apoptosis in HSCs, and several key proteins may be involved in the molecular mechanism of apoptosis induced by DATS.


Subject(s)
Allyl Compounds/pharmacology , Apoptosis/drug effects , Hepatic Stellate Cells/cytology , Hepatic Stellate Cells/drug effects , Proteomics , Sulfides/pharmacology , Animals , Cell Cycle/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Garlic/chemistry , Gene Expression Regulation/drug effects , Hepatic Stellate Cells/metabolism , Rats
13.
Cell Rep ; 18(2): 468-481, 2017 01 10.
Article in English | MEDLINE | ID: mdl-28076790

ABSTRACT

Glioblastoma multiforme (GBM) is among the most aggressive of human cancers. Although differentiation therapy has been proposed as a potential approach to treat GBM, the mechanisms of induced differentiation remain poorly defined. Here, we established an induced differentiation model of GBM using cAMP activators that specifically directed GBM differentiation into astroglia. Transcriptomic and proteomic analyses revealed that oxidative phosphorylation and mitochondrial biogenesis are involved in induced differentiation of GBM. Dibutyryl cyclic AMP (dbcAMP) reverses the Warburg effect, as evidenced by increased oxygen consumption and reduced lactate production. Mitochondrial biogenesis induced by activation of the CREB-PGC1α pathway triggers metabolic shift and differentiation. Blocking mitochondrial biogenesis using mdivi1 or by silencing PGC1α abrogates differentiation; conversely, overexpression of PGC1α elicits differentiation. In GBM xenograft models and patient-derived GBM samples, cAMP activators also induce tumor growth inhibition and differentiation. Our data show that mitochondrial biogenesis and metabolic switch to oxidative phosphorylation drive the differentiation of tumor cells.


Subject(s)
Astrocytes/pathology , Brain Neoplasms/pathology , Cell Differentiation , Cyclic AMP/metabolism , Glioblastoma/metabolism , Glioblastoma/pathology , Glycolysis , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , 8-Bromo Cyclic Adenosine Monophosphate/analogs & derivatives , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Astrocytes/metabolism , Astrocytes/ultrastructure , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/ultrastructure , Cell Death/drug effects , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclic AMP Response Element-Binding Protein/metabolism , Gene Expression Profiling , Glial Fibrillary Acidic Protein/metabolism , Glioblastoma/genetics , Glioblastoma/ultrastructure , Glycolysis/drug effects , Humans , Organelle Biogenesis , Oxidative Phosphorylation/drug effects , Proteomics , Signal Transduction , Xenograft Model Antitumor Assays
14.
Nat Commun ; 7: 13107, 2016 10 06.
Article in English | MEDLINE | ID: mdl-27708285

ABSTRACT

Snakes have numerous features distinctive from other tetrapods and a rich history of genome evolution that is still obscure. Here, we report the high-quality genome of the five-pacer viper, Deinagkistrodon acutus, and comparative analyses with other representative snake and lizard genomes. We map the evolutionary trajectories of transposable elements (TEs), developmental genes and sex chromosomes onto the snake phylogeny. TEs exhibit dynamic lineage-specific expansion, and many viper TEs show brain-specific gene expression along with their nearby genes. We detect signatures of adaptive evolution in olfactory, venom and thermal-sensing genes and also functional degeneration of genes associated with vision and hearing. Lineage-specific relaxation of functional constraints on respective Hox and Tbx limb-patterning genes supports fossil evidence for a successive loss of forelimbs then hindlimbs during snake evolution. Finally, we infer that the ZW sex chromosome pair had undergone at least three recombination suppression events in the ancestor of advanced snakes. These results altogether forge a framework for our deep understanding into snakes' history of molecular evolution.


Subject(s)
Biological Evolution , DNA Transposable Elements , Snakes/anatomy & histology , Snakes/genetics , Animals , Cell Lineage , Evolution, Molecular , Female , Forelimb , Gene Expression Profiling , Gene Expression Regulation , Genome , Hindlimb , Lizards/genetics , Male , Phylogeny , Recombination, Genetic , Sex Chromosomes , Transcriptome
15.
Anal Sci ; 32(9): 981-8, 2016.
Article in English | MEDLINE | ID: mdl-27682404

ABSTRACT

Sensitive and specific biomarkers are required for the diagnosis and treatment of depression because the existing diagnostic criteria are subjective and could produce false positives or negatives. Some endogenous neuroactive steroids that have shown either antidepressant effects or concentration changes in individuals with depression could provide potential biomarkers. In this study, a simple and specific method was developed to simultaneously determine seven endogenous neuroactive steroids in biological samples: cortisone, cortisol, dehydroepiandrosterone, estradiol, progesterone, pregnenolone, and testosterone. After liquid-liquid extraction, chromatographic separation was achieved on a C18 column with gradient elution using water-methanol at a flow rate of 300 µL min(-1). Detection and quantitation were performed by tandem mass spectrometry with atmospheric pressure chemical ionization and selected reaction monitoring. Plasma and brain neuroactive steroid levels were then determined in control rats and rats exposed to forced swimming, a classical rodent model of depression. The results showed that the plasma concentrations of testosterone, pregnenolone, and progesterone significantly increased in rats exposed to the forced swimming test. In contrast, brain homogenate levels of cortisol, estradiol, and progesterone decreased, while pregnenolone levels were elevated in this model of depression. In conclusion, a new method to quantify neuroactive steroids was successfully developed and applied to their investigation in rat plasma and brain. The findings of this study indicated that plasma testosterone, pregnenolone, and progesterone levels could provide potential biomarkers for the diagnosis and treatment of depression.


Subject(s)
Blood Chemical Analysis/methods , Brain/metabolism , Depression/metabolism , Steroids/blood , Analytic Sample Preparation Methods , Animals , Chromatography, High Pressure Liquid , Depression/blood , Male , Methanol/chemistry , Rats , Rats, Sprague-Dawley , Steroids/chemistry , Steroids/isolation & purification , Steroids/metabolism , Tandem Mass Spectrometry , Time Factors , Water/chemistry
16.
Int J Pharm ; 513(1-2): 347-356, 2016 Nov 20.
Article in English | MEDLINE | ID: mdl-27628782

ABSTRACT

Hydroxypropyl-ß-cyclodextrin (HP-ß-CD) has been widely used as an effective solubilizing agent in pharmaceutical industry for many years. However, the effect of degree of substitution (D.S.) of HP-ß-CD on solubilizing capacity and toxicity has not been concerned. In this study, solubilizing capacity of HP-ß-CDs with three different D.S. (4.55, 6.16 and 7.76) for 16 drugs were measured and their toxicities were compared by a 7-day i.v. administration (q.d.) study in rats. Generally, HP-ß-CD with high D.S. (7.76) showed weaker solubilizing capacity for steroids and BCS class II drugs, but lower hemolytic activity, compared with that of HP-ß-CD with low (4.55) or medium (6.16) D.S. HP-ß-CD with low D.S. resulted in more changes in hematological and biochemical parameters, but the effects were reversible after a 7-day recovery. Moreover, HP-ß-CD with medium D.S. may have slightly greater nephrotoxicity than the other two HP-ß-CDs. HP-ß-CDs with different D.S. had similar urine excretion percentage after i.v. administration and none of them was found to affect glomerular filtration function of rats. The results suggest that HP-ß-CD with low D.S. would be a better choice considering both the solubilizing capacity and toxicity. However, comparison in toxicity of HP-ß-CDs with different D.S. should be carried out in human in view of its species-dependence property.


Subject(s)
Excipients/chemistry , Excipients/toxicity , beta-Cyclodextrins/chemistry , beta-Cyclodextrins/toxicity , 2-Hydroxypropyl-beta-cyclodextrin , Animals , Erythrocytes/drug effects , Excipients/pharmacokinetics , Female , Glomerular Filtration Rate/drug effects , Hemolysis/drug effects , Male , Pharmaceutical Preparations/chemistry , Rabbits , Rats, Sprague-Dawley , Solubility , beta-Cyclodextrins/pharmacokinetics , beta-Cyclodextrins/urine
17.
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
18.
Mol Med Rep ; 13(3): 2499-505, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26821268

ABSTRACT

Glutamate is the principal neurotransmitter in the central nervous system. Glutamate-mediated excitotoxicity is the predominant cause of cerebral damage. Recent studies have shown that lysosomal membrane permeabilization (LMP) is involved in ischemia­associated neuronal death in non­human primates. This study was designed to investigate the effect of glutamate on lysosomal stability in primary cultured cortical neurons. Glutamate treatment for 30 min induced the permeabilization of lysosomal membranes as assessed by acridine orange redistribution and immunofluorescence of cathepsin B in the cytoplasm. Inhibition of glutamate excitotoxicity by the NMDA receptor antagonist MK­801 and the calcium chelator ethylene glycol­bis (2­aminoethylether)­N, N, N', N'­tetraacetic acid, rescued lysosomes from permeabilization. The role of calpain and reactive oxygen species (ROS) in inducing LMP was also investigated. Ca2+ overload following glutamate treatment induced the activation of calpain and the production of ROS, which are two major contributors to neuronal death. It has been reported that lysosomal­associated membrane protein 2 (LAMP2) and heat shock protein (HSP)70 are two calpain substrates that promote LMP in cancer cells; however, it was found that calpains were activated by glutamate, but only LAMP2 was subsequently degraded. Furthermore, LMP was not alleviated by treatment with the calpain inhibitors calpeptin and SJA6017, which blocked the cleavage of the calpain substrate α­fodrin. It was demonstrated that LMP was significantly alleviated by treatment with the antioxidant N­Acetyl­L­cysteine, indicating that LMP involvement in early glutamate excitotoxicity may be mediated partly by ROS rather than calpain activation. Overall, these data shed light on the role of ROS-mediated LMP in early glutamate excitotoxicity.


Subject(s)
Glutamic Acid/pharmacology , Lysosomes/metabolism , Neurons/metabolism , Animals , Calpain/metabolism , Cell Membrane Permeability/drug effects , Cells, Cultured , Cerebral Cortex/cytology , Lysosomes/drug effects , Primary Cell Culture , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism
19.
Steroids ; 105: 96-105, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26631550

ABSTRACT

Steroids have been shown to have multiple effects on the nervous system including neuroprotective activities, and they have the potential to be used for the treatment of neurodegenerative diseases. In this current study, we tested the hypothesis that the marine steroid 24-methylenecholestane-3ß,5α,6ß,19-tetraol (Tetrol) has a neuroprotective effect. (1) We synthesized Tetrol through a multiple step reaction starting from hyodeoxycholic acid (HDCA). (2) We then evaluated the neuroprotective effect of Tetrol with a glutamate-induced neuronal injury model in vitro. Tetrol concentration dependently increased the survival rate of cerebellar granule neurons challenged with toxic concentration of glutamate. Consistently, Tetrol significantly decreased glutamate-induced lactate dehydrogenase (LDH) release with a threshold concentration of 2.5 µM. (3) We further evaluated the neuroprotective effect of Tetrol in a middle cerebral artery occlusion (MCAO)-induced cerebral ischemia model in rat. Tetrol, at a dose of 12 mg/kg, significantly decreased MCAO-induced infarction volume by ∼50%. (4) Finally, we probed the mechanism and found that Tetrol concentration dependently attenuated N-methyl-d-aspartate (NMDA)-induced intracellular calcium ([Ca(2+)]i) increase with an IC50 of 7.8±0.62 µM, and inhibited NMDA currents in cortical neurons with an IC50 of 10.28±0.71 µM. Taken together, we have synthesized and characterized Tetrol as a novel neuroprotectant through negative modulation of NMDA receptors.


Subject(s)
Aquatic Organisms/chemistry , Cholestanols/pharmacology , Neuroprotective Agents/pharmacology , Animals , Brain Ischemia/drug therapy , Brain Ischemia/etiology , Brain Ischemia/pathology , Cerebral Cortex/pathology , Cholestanols/chemical synthesis , Cholestanols/chemistry , Cholestanols/therapeutic use , Glutamic Acid/toxicity , Infarction, Middle Cerebral Artery/complications , Infarction, Middle Cerebral Artery/drug therapy , Infarction, Middle Cerebral Artery/pathology , Magnetic Resonance Spectroscopy , Male , Mice , N-Methylaspartate/pharmacology , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Neuroprotective Agents/chemical synthesis , Neuroprotective Agents/chemistry , Neuroprotective Agents/therapeutic use , Rats, Sprague-Dawley
20.
Mol Ther ; 24(1): 156-65, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26373347

ABSTRACT

Oncolytic virotherapy is a novel and emerging treatment modality that uses replication-competent viruses to destroy cancer cells. Although diverse cancer cell types are sensitive to oncolytic viruses, one of the major challenges of oncolytic virotherapy is that the sensitivity to oncolysis ranges among different cancer cell types. Furthermore, the underlying mechanism of action is not fully understood. Here, we report that activation of cyclic adenosine monophosphate (cAMP) signaling significantly sensitizes refractory cancer cells to alphavirus M1 in vitro, in vivo, and ex vivo. We find that activation of the cAMP signaling pathway inhibits M1-induced expression of antiviral factors in refractory cancer cells, leading to prolonged and severe endoplasmic reticulum (ER) stress, and cell apoptosis. We also demonstrate that M1-mediated oncolysis, which is enhanced by cAMP signaling, involves the factor, exchange protein directly activated by cAMP 1 (Epac1), but not the classical cAMP-dependent protein kinase A (PKA). Taken together, cAMP/Epac1 signaling pathway activation inhibits antiviral factors and improves responsiveness of refractory cancer cells to M1-mediated virotherapy.


Subject(s)
Alphavirus/genetics , Colforsin/administration & dosage , Cyclic AMP/metabolism , Neoplasms/therapy , Signal Transduction/drug effects , Animals , Apoptosis , Cell Line, Tumor , Colforsin/pharmacology , Endoplasmic Reticulum Stress/drug effects , Guanine Nucleotide Exchange Factors/genetics , HCT116 Cells , Humans , Mice , Neoplasms/genetics , Oncolytic Virotherapy , Oncolytic Viruses/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...