Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Rep Med ; 4(10): 101229, 2023 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-37820722

RESUMEN

Although promising, dendritic cell (DC) vaccines still provide limited clinical benefits, mainly due to the immunosuppressive tumor microenvironment (TME) and the lack of tumor-associated antigens (TAAs). Oncolytic virus therapy is an ideal strategy to overcome immunosuppression and expose TAAs; therefore, they may work synergistically with DC vaccines. In this study, we demonstrate that oncolytic virus M1 (OVM) can enhance the antitumor effects of DC vaccines across diverse syngeneic mouse tumor models by increasing the infiltration of CD8+ effector T cells in the TME. Mechanically, we show that tumor cells counteract DC vaccines through the SIRPα-CD47 immune checkpoint, while OVM can downregulate SIRPα in DCs and CD47 in tumor cells. Since OVM upregulates PD-L1 in DCs, combining PD-L1 blockade with DC vaccines and OVM further enhances antitumor activity. Overall, OVM strengthens the antitumor efficacy of DC vaccines by targeting the SIRPα-CD47 axis, which exerts dominant immunosuppressive effects on DC vaccines.


Asunto(s)
Virus Oncolíticos , Vacunas , Ratones , Animales , Virus Oncolíticos/genética , Antígeno CD47/genética , Antígeno B7-H1 , Línea Celular Tumoral , Antígenos de Neoplasias
2.
Hum Gene Ther ; 32(3-4): 158-165, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33504253

RESUMEN

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.


Asunto(s)
Alphavirus , Neoplasias , Viroterapia Oncolítica , Virus Oncolíticos , Alphavirus/genética , Línea Celular Tumoral , Humanos , Neoplasias/genética , Neoplasias/terapia , Virus Oncolíticos/genética , Replicación Viral
3.
Cell Death Dis ; 11(12): 1062, 2020 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-33311488

RESUMEN

Reversing the highly immunosuppressive tumor microenvironment (TME) is essential to achieve long-term efficacy with cancer immunotherapy. Despite the impressive clinical response to checkpoint blockade in multiple types of cancer, only a minority of patients benefit from this approach. Here, we report that the oncolytic virus M1 induces immunogenic tumor cell death and subsequently restores the ability of dendritic cells to prime antitumor T cells. Intravenous injection of M1 disrupts immune tolerance in the privileged TME, reprogramming immune-silent (cold) tumors into immune-inflamed (hot) tumors. M1 elicits potent CD8+ T cell-dependent therapeutic effects and establishes long-term antitumor immune memory in poorly immunogenic tumor models. Pretreatment with M1 sensitizes refractory tumors to subsequent checkpoint blockade by boosting T-cell recruitment and upregulating the expression of PD-L1. These findings reveal the antitumor immunological mechanism of the M1 virus and indicated that oncolytic viruses are ideal cotreatments for checkpoint blockade immunotherapy.


Asunto(s)
Resistencia a Antineoplásicos/efectos de los fármacos , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología , Virus Oncolíticos/metabolismo , Linfocitos T/inmunología , Animales , Antígeno B7-H1/metabolismo , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , Células Dendríticas/efectos de los fármacos , Células Dendríticas/metabolismo , Resistencia a Antineoplásicos/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Inhibidores de Puntos de Control Inmunológico/farmacología , Muerte Celular Inmunogénica/efectos de los fármacos , Memoria Inmunológica/efectos de los fármacos , Inflamación/genética , Inyecciones Intravenosas , Ratones Endogámicos C57BL , Virus Oncolíticos/efectos de los fármacos , Linfocitos T/efectos de los fármacos , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología
4.
Mol Oncol ; 13(7): 1589-1604, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31162799

RESUMEN

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.


Asunto(s)
Neoplasias Encefálicas/genética , Regulación Neoplásica de la Expresión Génica , Glioblastoma/genética , Histonas/metabolismo , MicroARNs/genética , Animales , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Regulación hacia Abajo , Femenino , Glioblastoma/metabolismo , Glioblastoma/patología , Humanos , Metilación , Ratones Endogámicos BALB C , Neuroglía/metabolismo , Neuroglía/patología , Transcriptoma
5.
Nat Commun ; 9(1): 4342, 2018 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-30337542

RESUMEN

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.


Asunto(s)
Antivirales/farmacología , Daño del ADN , Proteína Quinasa Activada por ADN/antagonistas & inhibidores , Virus Oncolíticos/fisiología , Animales , Antineoplásicos/farmacología , Línea Celular Tumoral , Terapia Combinada , Proteína Quinasa Activada por ADN/metabolismo , Evaluación Preclínica de Medicamentos , Estrés del Retículo Endoplásmico/efectos de los fármacos , Humanos , Ratones Endogámicos BALB C , Ratones Desnudos , Modelos Biológicos , Viroterapia Oncolítica , Inhibidores de Proteínas Quinasas/farmacología , Ratas
7.
Nat Commun ; 9(1): 1524, 2018 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-29670091

RESUMEN

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.


Asunto(s)
Colesterol/química , Ácido Mevalónico/antagonistas & inhibidores , Ácido Mevalónico/metabolismo , Viroterapia Oncolítica , Virus Oncolíticos/fisiología , Replicación Viral , Animales , Línea Celular Tumoral , Supervivencia Celular , Regulación hacia Abajo , Farnesiltransferasa/antagonistas & inhibidores , Femenino , Técnicas de Silenciamiento del Gen , Células HCT116 , Humanos , Ratones , Ratones Endogámicos BALB C , Trasplante de Neoplasias , Prenilación de Proteína , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Proteínas de Unión al GTP rab/metabolismo
8.
Hum Gene Ther ; 29(8): 950-961, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-28750564

RESUMEN

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.


Asunto(s)
Neoplasias/genética , Viroterapia Oncolítica/métodos , Virus Oncolíticos/efectos de los fármacos , Proteínas Proto-Oncogénicas c-bcl-2/genética , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Terapia Combinada , Humanos , Ratones , Mitocondrias/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Neoplasias/virología , Virus Oncolíticos/genética , Proteínas Proto-Oncogénicas c-bcl-2/antagonistas & inhibidores , Ensayos Antitumor por Modelo de Xenoinjerto
9.
J Neurochem ; 144(2): 186-200, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29205357

RESUMEN

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.


Asunto(s)
Isquemia Encefálica/enzimología , Isquemia Encefálica/genética , Glucólisis/genética , Hexoquinasa/genética , Hexoquinasa/metabolismo , Activación de Macrófagos/genética , Microglía/enzimología , Accidente Cerebrovascular/enzimología , Accidente Cerebrovascular/genética , Acetilcoenzima A/metabolismo , Acetilación , Animales , Inducción Enzimática/genética , Hexoquinasa/biosíntesis , Histonas/metabolismo , Infarto de la Arteria Cerebral Media/genética , Infarto de la Arteria Cerebral Media/metabolismo , Inflamación/genética , Interleucina-1beta/metabolismo , Masculino , Interferencia de ARN , Ratas , Ratas Sprague-Dawley
10.
J Virol ; 92(6)2018 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-29263275

RESUMEN

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.


Asunto(s)
Autofagia/inmunología , Endorribonucleasas/deficiencia , Glioma/terapia , Proteínas de Neoplasias/deficiencia , Viroterapia Oncolítica , Virus Oncolíticos , Proteínas Serina-Treonina Quinasas/deficiencia , Togaviridae , Animales , Autofagia/genética , Línea Celular Tumoral , Chlorocebus aethiops , Cricetinae , Endorribonucleasas/inmunología , Femenino , Glioma/genética , Glioma/inmunología , Humanos , Ratones Endogámicos BALB C , Ratones Desnudos , Proteínas de Neoplasias/inmunología , Proteínas Serina-Treonina Quinasas/inmunología , Respuesta de Proteína Desplegada/genética , Respuesta de Proteína Desplegada/inmunología , Células Vero , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Sci Transl Med ; 9(404)2017 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-28835517

RESUMEN

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.


Asunto(s)
Antineoplásicos/metabolismo , Carcinoma Hepatocelular/terapia , Carcinoma Hepatocelular/virología , Neoplasias Hepáticas/terapia , Neoplasias Hepáticas/virología , Virus Oncolíticos/metabolismo , Proteína que Contiene Valosina/antagonistas & inhibidores , Animales , Apoptosis , Efecto Espectador , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Terapia Combinada , Estrés del Retículo Endoplásmico , Endorribonucleasas/metabolismo , Ensayos Analíticos de Alto Rendimiento , Humanos , Neoplasias Hepáticas/patología , Virus Oncolíticos/patogenicidad , Primates , Proteínas Serina-Treonina Quinasas/metabolismo , Proteína que Contiene Valosina/metabolismo , Proteína 1 de Unión a la X-Box/metabolismo
12.
Neuroscience ; 350: 110-123, 2017 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-28336411

RESUMEN

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.


Asunto(s)
Ácido Glutámico/metabolismo , Hipocampo/efectos de los fármacos , Sulfuro de Hidrógeno/farmacología , Memoria Espacial/efectos de los fármacos , Estrés Psicológico/metabolismo , Animales , Hipocampo/metabolismo , Depresión Sináptica a Largo Plazo/fisiología , Masculino , Trastornos de la Memoria/metabolismo , Plasticidad Neuronal/fisiología , Ratas Sprague-Dawley
13.
Cell Rep ; 18(2): 468-481, 2017 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-28076790

RESUMEN

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.


Asunto(s)
Astrocitos/patología , Neoplasias Encefálicas/patología , Diferenciación Celular , AMP Cíclico/metabolismo , Glioblastoma/metabolismo , Glioblastoma/patología , Glucólisis , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , 8-Bromo Monofosfato de Adenosina Cíclica/análogos & derivados , 8-Bromo Monofosfato de Adenosina Cíclica/farmacología , Astrocitos/metabolismo , Astrocitos/ultraestructura , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/ultraestructura , Muerte Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Perfilación de la Expresión Génica , Proteína Ácida Fibrilar de la Glía/metabolismo , Glioblastoma/genética , Glioblastoma/ultraestructura , Glucólisis/efectos de los fármacos , Humanos , Biogénesis de Organelos , Fosforilación Oxidativa/efectos de los fármacos , Proteómica , Transducción de Señal , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Nat Commun ; 7: 13107, 2016 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-27708285

RESUMEN

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.


Asunto(s)
Evolución Biológica , Elementos Transponibles de ADN , Serpientes/anatomía & histología , Serpientes/genética , Animales , Linaje de la Célula , Evolución Molecular , Femenino , Miembro Anterior , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Genoma , Miembro Posterior , Lagartos/genética , Masculino , Filogenia , Recombinación Genética , Cromosomas Sexuales , Transcriptoma
15.
Hum Gene Ther ; 27(9): 700-11, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27296553

RESUMEN

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.


Asunto(s)
Alphavirus/inmunología , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Vectores Genéticos/administración & dosificación , Virus Oncolíticos/inmunología , Alphavirus/genética , Animales , Modelos Animales de Enfermedad , Femenino , Humanos , Inyecciones Intravenosas , Macaca fascicularis , Masculino , Virus Oncolíticos/genética
16.
Steroids ; 105: 96-105, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26631550

RESUMEN

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.


Asunto(s)
Organismos Acuáticos/química , Colestanoles/farmacología , Fármacos Neuroprotectores/farmacología , Animales , Isquemia Encefálica/tratamiento farmacológico , Isquemia Encefálica/etiología , Isquemia Encefálica/patología , Corteza Cerebral/patología , Colestanoles/síntesis química , Colestanoles/química , Colestanoles/uso terapéutico , Ácido Glutámico/toxicidad , Infarto de la Arteria Cerebral Media/complicaciones , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Infarto de la Arteria Cerebral Media/patología , Espectroscopía de Resonancia Magnética , Masculino , Ratones , N-Metilaspartato/farmacología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Fármacos Neuroprotectores/síntesis química , Fármacos Neuroprotectores/química , Fármacos Neuroprotectores/uso terapéutico , Ratas Sprague-Dawley
17.
Mol Ther ; 24(1): 156-65, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26373347

RESUMEN

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.


Asunto(s)
Alphavirus/genética , Colforsina/administración & dosificación , AMP Cíclico/metabolismo , Neoplasias/terapia , Transducción de Señal/efectos de los fármacos , Animales , Apoptosis , Línea Celular Tumoral , Colforsina/farmacología , Estrés del Retículo Endoplásmico/efectos de los fármacos , Factores de Intercambio de Guanina Nucleótido/genética , Células HCT116 , Humanos , Ratones , Neoplasias/genética , Viroterapia Oncolítica , Virus Oncolíticos/genética
18.
CNS Neurosci Ther ; 21(6): 486-95, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25678034

RESUMEN

BACKGROUND: Neuroactive steroids represent promising candidates for the treatment of neurological disorders. Our previous studies identified an endogenous steroid cholestane-3ß, 5α, 6ß-triol (Triol) as a novel neuroprotectant. AIM: We aimed to identify a potent candidate for stroke treatment through a screening of Triol analogs. METHODS: Hypoxia- and glutamate-induced neuronal injury models in vitro, middle cerebral artery occlusion (MCAO)-induced cerebral ischemia model in vivo, fluorescein diacetate (FDA) for alive and propidium iodide (PI) for dead staining, LDH assay, and calcium imaging techniques were used. RESULTS: 24-keto-cholest-5-en-3ß, 19-diol (Diol) showed the most potent neuroprotective effect among the screened structurally related compounds. FDA and PI staining showed that Diol concentration dependently increased the survival rate of cerebellar granule neurons (CGNs) challenged with glutamate or hypoxia, with an effective threshold concentration of 2.5 µM. Consistently, the quantitative LDH release assay showed the same concentration-dependent protection in both models. Diol, at 10 µM, potently decreased glutamate- and hypoxia-induced LDH release from 51.6 to 18.2% and 62.1 to 21.7%, respectively, which values are close to the normal LDH release (~16-18%). Moreover, we found Diol effectively decreased MCAO-induced infarction volume in mice from ~23% to 7%, at a dose of 6 mg/kg. We further explored the underlying mechanism and found that Diol attenuated NMDA-induced intracellular calcium ([Ca(2+) ]i ) increase in cortical neurons, suggesting a negative modulatory effect on NMDA receptor. CONCLUSION: Taken together, we identified Diol as a potent neuroprotectant. It may represent a novel and promising neuroprotectant for stroke intervention.


Asunto(s)
Colestanoles/uso terapéutico , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Animales , Animales Recién Nacidos , Calcio/metabolismo , Hipoxia de la Célula/efectos de los fármacos , Células Cultivadas , Cerebelo/citología , Colestanoles/química , Modelos Animales de Enfermedad , Agonistas de Aminoácidos Excitadores/toxicidad , Ácido Glutámico/toxicidad , L-Lactato Deshidrogenasa/metabolismo , N-Metilaspartato/toxicidad , Oxígeno/administración & dosificación , Ratas , Ratas Sprague-Dawley
19.
Mol Med Rep ; 9(5): 1773-9, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24626525

RESUMEN

The biotoxin cholera toxin has been demonstrated to have anti-tumor activity in numerous types of cancer, including glioma. However, the role of cholera toxin in the tumorigenesis of transitional cell carcinoma (TCC), the most common malignant tumor of the bladder, remains to be elucidated. To address this, in the present study, two TCC cell lines, T24 and UM-UC-3, were treated with cholera toxin [protein kinase A (PKA) activator] and KT5720 (PKA inhibitor). Cell survival and proliferation, cell cycle alterations and apoptosis were analyzed using Hoechst staining, the MTT assay, fluorescence microscopy and flow cytometry. Western blot analysis was used to detect the expression of proteins involved in cell cycle regulation. The results revealed that cholera toxin significantly induced G1 arrest and downregulated the expression of cyclin D1 and cyclin-dependent kinase 4/6 in the TCC cell lines, and this was rescued by KT5720. Furthermore, it was demonstrated that cholera toxin downregulated the activation of the c-Raf/Mek/Erk cascade, an important mediator of tumor cell proliferation, via the PKA-dependent c-Raf phosphorylation at Ser-43. Furthermore, inhibition of Mek activity with UO126 mimicked the effects of cholera toxin. In conclusion, these results confirmed that cholera toxin specifically inhibited proliferation and induced G1 phase arrest in human bladder TCC cells. This effect was due to PKA-dependent inactivation of the c-Raf/Mek/Erk pathway. This suggested that cholera toxin may be a viable therapeutic treatment against tumorigenesis and proliferation in bladder cancer.


Asunto(s)
Carcinoma de Células Transicionales/metabolismo , Toxina del Cólera/farmacología , Puntos de Control de la Fase G1 del Ciclo Celular/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proteínas Proto-Oncogénicas c-raf/metabolismo , Neoplasias de la Vejiga Urinaria/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Humanos
20.
Respiration ; 84(6): 509-17, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23006535

RESUMEN

BACKGROUND: Idiopathic pulmonary fibrosis (IPF) is a progressive and fatal lung disease without beneficial therapy, except for lung transplantation. A high oral dose of N-acetylcysteine (NAC) added to prednisone and azathioprine has been found to improve lung function in IPF patients, though the mechanism of action remains poorly understood. OBJECTIVE: Based on our previous findings showing elevation of glutathione (GSH) content associated with downregulation of lysyl oxidase (LOX) activity, which is essential for collagen deposition, the aim of the present study was to test the hypothesis that NAC alleviates IPF by regulating LOX function. METHODS: We firstly analyzed the time course of collagen deposition in lung tissue, hydroxyproline content, LOX activity, GSH levels, and transforming growth factor-ß(1) (TGF-ß(1)) and α-smooth muscle actin (α-SMA) expression in bleomycin (BLM)-induced pulmonary fibrosis in a rat model. Then, we focused our studies on NAC modulation of LOX activity. RESULTS: LOX activity was increased on day 9 and peaked 14 days after BLM administration, while TGF-ß(1) protein peaked on day 9. Interestingly, NAC treatment for 14 days from day 0 reversed LOX activity to normal levels and increased GSH levels in the lung of BLM-dosed rats. Consistently, NAC partially attenuated pulmonary fibrosis and inhibited TGF-ß(1) and α-SMA expression in this model. CONCLUSIONS: Our study supports a novel mechanism of NAC alleviating IPF by inhibition of LOX activity via elevation of lung GSH in BLM-induced pulmonary fibrosis. The TGF-ß(1)/α-SMA pathway may also play an important role in modulation of LOX activity.


Asunto(s)
Acetilcisteína/farmacología , Depuradores de Radicales Libres/farmacología , Proteína-Lisina 6-Oxidasa/metabolismo , Fibrosis Pulmonar/metabolismo , Actinas/efectos de los fármacos , Actinas/metabolismo , Animales , Antibióticos Antineoplásicos/efectos adversos , Bleomicina/efectos adversos , Colágeno/efectos de los fármacos , Colágeno/metabolismo , Modelos Animales de Enfermedad , Regulación hacia Abajo , Glutatión/efectos de los fármacos , Glutatión/metabolismo , Fibrosis Pulmonar Idiopática/tratamiento farmacológico , Fibrosis Pulmonar Idiopática/metabolismo , Pulmón/efectos de los fármacos , Pulmón/patología , Masculino , Proteína-Lisina 6-Oxidasa/efectos de los fármacos , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/tratamiento farmacológico , Ratas , Ratas Sprague-Dawley , Factor de Crecimiento Transformador beta1/efectos de los fármacos , Factor de Crecimiento Transformador beta1/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA