Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Pharmacol Res ; 161: 105094, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32795509

RESUMO

Oncolytic virus (OV) immunotherapy has demonstrated to be a promising approach in cancer treatment due to tumor-specific oncolysis. However, their clinical use so far has been largely limited due to the lack of suitable delivery strategies with high efficacy. Direct 'intratumoral' injection is the way to cross the hurdles of systemic toxicity, while providing local effects. Progress in this field has enabled the development of alternative way using 'systemic' oncolytic virotherapy for producing better results. One major potential roadblock to systemic OV delivery is the low virus persistence in the face of hostile immune system. The delivery challenge is even greater when attempting to target the oncolytic viruses into the entire tumor mass, where not all tumor cells are equally exposed to exactly the same microenvironment. The microenvironment of many tumors is known to be massively infiltrated with various types of leucocytes in both primary and metastatic sites. Interestingly, this intratumoral immune cell heterogeneity exhibits a degree of organized distribution inside the tumor bed as evidenced, for example, by the hypoxic tumor microenviroment where predominantly recruits tumor-associated macrophages. Although in vivo OV delivery seems complicated and challenging, recent results are encouraging for decreasing the limitations of systemically administered oncolytic viruses and an improved efficiency of oncolytic viral therapy in targeting cancerous tissues in vitro. Here, we review the latest developments of carrier cell-based oncolytic virus delivery using tumor-infiltrating immune cells with a focus on the main features of each cellular vehicle.


Assuntos
Fibroblastos Associados a Câncer/virologia , Células Matadoras Induzidas por Citocinas/virologia , Imunoterapia Adotiva , Linfócitos do Interstício Tumoral/virologia , Monócitos/virologia , Neoplasias/terapia , Terapia Viral Oncolítica , Vírus Oncolíticos/patogenicidade , Linfócitos T/virologia , Animais , Fibroblastos Associados a Câncer/imunologia , Fibroblastos Associados a Câncer/transplante , Células Matadoras Induzidas por Citocinas/imunologia , Células Matadoras Induzidas por Citocinas/transplante , Humanos , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/transplante , Monócitos/imunologia , Monócitos/transplante , Neoplasias/imunologia , Neoplasias/patologia , Neoplasias/virologia , Células-Tronco Neoplásicas/imunologia , Células-Tronco Neoplásicas/patologia , Células-Tronco Neoplásicas/virologia , Vírus Oncolíticos/genética , Vírus Oncolíticos/imunologia , Fenótipo , Linfócitos T/imunologia , Linfócitos T/transplante , Hipóxia Tumoral , Microambiente Tumoral
2.
Vaccine ; 36(25): 3613-3621, 2018 06 14.
Artigo em Inglês | MEDLINE | ID: mdl-29764681

RESUMO

Although some commercial vaccines against grass carp reovirus (GCRV) are available, given the many varieties of GCRV and limited types of vaccines, the disease caused by GCRV remains a major problem, which leads to economic losses in grass carp aquaculture. A reovirus strain (GCRV-HN14) was recently isolated from local diseased fish in our laboratory. The S11 segment of GCRV-HN14 was speculated to encode the virus capsid protein VP35. In our study, the S11 segment was cloned into the eukaryotic expression vector pcDNA3.1(+) to construct the recombinant plasmid pcDNA3.1-s11, which was then transfected into CIK cells, and the VP35 protein was successfully expressed. Grass carp was immunized with pcDNA3.1-s11, and the in vivo distribution and expression of the pcDNA3.1-s11 plasmids were analyzed by PCR and Western blot. Recombinant plasmids were detected in the blood, liver, spleen, kidney, and muscle. However, protein expression could only be detected in the muscle. The immune protection of the pcDNA3.1-s11 plasmid in grass carp was evaluated using a series of experiments. Results showed that the population of white blood cells significantly increased at 1, 7, and 14 days post-immunization (dpi) and reached a peak with (9.58 ±â€¯0.72) × 107/ml at 7 dpi (P < 0.01 or P < 0.05). The percentage of neutrophils reached a peak with (24.13 ±â€¯2.38)% at 7 dpi (P < 0.01), whereas the lymphocytes peaked with (93.30 ±â€¯4.71)% at 14 dpi (P < 0.05). Serum antibody levels were significantly enhanced in immunized fish at 14, 21, and 28 dpi (P < 0.01). The mRNA expression levels of type I interferon, immunoglobulin M, Toll-like receptor 22, and major histocompatibility complex class I were significantly up-regulated in the head kidney and spleen of immunized fish (P < 0.05). Grass carp immunized with pcDNA3.1-s11 exhibited a higher survival percentage (70.4%-73.3%) than the controls (5%-13%). Overall, as a DNA vaccine, the pcDNA3.1-s11 plasmid could induce immune protection against GCRV.


Assuntos
Anticorpos Antivirais/biossíntese , Células Matadoras Induzidas por Citocinas/imunologia , Doenças dos Peixes/prevenção & controle , Plasmídeos/imunologia , Infecções por Reoviridae/prevenção & controle , Vacinação , Vacinas Virais/imunologia , Animais , Aquicultura , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/imunologia , Carpas , Proliferação de Células , Clonagem Molecular , Células Matadoras Induzidas por Citocinas/virologia , Doenças dos Peixes/imunologia , Doenças dos Peixes/mortalidade , Doenças dos Peixes/virologia , Expressão Gênica , Imunoglobulina M/biossíntese , Interferon Tipo I/genética , Interferon Tipo I/imunologia , Músculos/imunologia , Músculos/virologia , Plasmídeos/administração & dosagem , Plasmídeos/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Reoviridae/imunologia , Infecções por Reoviridae/imunologia , Infecções por Reoviridae/mortalidade , Infecções por Reoviridae/veterinária , Análise de Sobrevida , Vacinas de DNA , Vacinas Virais/administração & dosagem , Vacinas Virais/genética
3.
Int J Mol Sci ; 18(11)2017 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-29135940

RESUMO

Grass carp (Ctenopharyngodon idellus) is an important worldwide commercial freshwater culture species. However, grass carp reovirus (GCRV) causes serious hemorrhagic disease in fingerlings and yearlings of fishes. To understand the molecular pathogenesis of host cells during GCRV infection, intensive proteomic quantification analysis of lysine acetylation in Ctenopharyngodon idella kidney (CIK) cells was performed. Using dimethylation labeling-based quantitative proteomics, 832 acetylated proteins with 1391 lysine acetylation sites were identified in response to GCRV infection, among which 792 proteins with 1323 sites were quantifiable. Bioinformatics analysis showed that differentially expressed lysine acetylated proteins are involved in diverse cellular processes and associated with multifarious functions, suggesting that extensive intracellular activities were changed upon viral infection. In addition, extensive alterations on host-protein interactions at the lysine acetylation level were also detected. Further biological experiments showed that the histone deacetylases (HDAC) inhibitor suberoylanilide hydroxamic acid (SAHA) could significantly suppress the GCRV replication. To our knowledge, this is the first to reveal the proteome-wide changes in host cell acetylome with aquatic virus infection. The results provided in this study laid a basis for further understanding the host response to aquareovirus infection in the post-translational modification aspect by regulating cell lysine acetylation conducive to viral replication.


Assuntos
Carpas/fisiologia , Células Matadoras Induzidas por Citocinas/metabolismo , Células Matadoras Induzidas por Citocinas/virologia , Doenças dos Peixes/virologia , Lisina/metabolismo , Proteômica , Reoviridae/fisiologia , Acetilação , Motivos de Aminoácidos , Animais , Benzoatos/farmacologia , Benzoatos/uso terapêutico , Linhagem Celular , Análise por Conglomerados , Células Matadoras Induzidas por Citocinas/efeitos dos fármacos , Doenças dos Peixes/metabolismo , Proteínas de Peixes/química , Proteínas de Peixes/metabolismo , Ontologia Genética , Ácidos Hidroxâmicos/farmacologia , Ácidos Hidroxâmicos/uso terapêutico , Nitrobenzenos , Domínios Proteicos , Mapas de Interação de Proteínas/efeitos dos fármacos , Proteoma/metabolismo , Pirazóis/farmacologia , Pirazóis/uso terapêutico , Pirazolonas , Frações Subcelulares/metabolismo , Replicação Viral/efeitos dos fármacos , Vorinostat
4.
Fish Shellfish Immunol ; 35(6): 1874-81, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24084043

RESUMO

The interferon-induced, dsRNA-activated protein kinase (PKR) is considered as an important component of innate immune system and as a representative effector protein of interferon system. In the present study, PKR gene (CiPKR, JX511974) from grass carp (Ctenopharyngodon idellus) was isolated and identified using homology-based PCR. CiPKR shares high sequence identity with the counterparts of goldfish (Crucian carp) and zebrafish (Danio rerio). The full-length cDNA of CiPKR was found to be 2436 bp, with an ORF of 2067 bp that encodes a polypeptide of 688 amino acids. The deduced polypeptide CiPKR contains three tandem dsRNA-binding motifs (dsRBMs) at the N-terminus and a conserved Ser/Thr kinase domain at the C-terminus. CiPKR was expressed ubiquitously at a low-level under normal conditions, but it could be up-regulated after intraperitoneal (ip) injection with grass carp haemorrhagic virus (GCHV). CiPKR was dramatically up-regulated at 6 h post-injection and then recovered rapidly to normal levels within 24 h; however, it was obviously up-regulated once again at 48 h or 72 h post-injection. It seemed that CiPKR could respond to GCHV infection in an IFN-independent as well as an IFN-dependent pathway. To further investigate its mechanism of biological actions, we constructed a series of recombinant plasmids including pcDNA3.1/PKR-wt, pcDNA3.1/PKR-K430R, pcDNA3.1/PKR-C (deletion of dsRBD sequence) and pcDNA3.1/PKR-C-K430R, and then each recombinant plasmid was transfected into CIK cells. In comparison with those of controls, a 79% and a 64% decrease of luciferase activities were detected in the tested cells transfected with CiPKR and CiPKR-C, respectively; however, luciferase activities were increased in those cells transfected with PKR-K430R and PKR-C-K430R, with a 160% and 115% up-regulation, respectively. Similarly, MTT colorimetric assay indicated that cell viabilities of CIK cells transfected with pcDNA3.1/PKR-wt, pcDNA3.1/PKR-K430R, pcDNA3.1/PKR-C and pcDNA3.1/PKR-C-K430R were 49%, 90%, 54% and 100%, respectively. Our observations suggested that the expression of CiPKR could be up-regulated following viral infection, and then resulted in the inhibition of protein synthesis and the induction of potential apoptosis.


Assuntos
Carpas/genética , Carpas/imunologia , Proteínas de Peixes/genética , eIF-2 Quinase/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Clonagem Molecular , Células Matadoras Induzidas por Citocinas/citologia , Células Matadoras Induzidas por Citocinas/virologia , DNA Complementar/genética , DNA Complementar/metabolismo , Proteínas de Peixes/química , Proteínas de Peixes/metabolismo , Regulação da Expressão Gênica , Dados de Sequência Molecular , Especificidade de Órgãos , Filogenia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase em Tempo Real/veterinária , Reoviridae , eIF-2 Quinase/química , eIF-2 Quinase/metabolismo
5.
Exp Hematol ; 41(6): 508-17, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23403007

RESUMO

Cytokine-induced killer (CIK) cells are in clinical testing against various tumor types, including multiple myeloma. In this study, we show that CIK cells have activity against subcutaneous and disseminated models of human myeloma (KAS-6/1), which can be enhanced by infecting the CIK cells with an oncolytic measles virus (MV) or by pretreating the myeloma cells with ionizing radiation (XRT). KAS-6/1 cells were killed by coculture with CIK or MV-infected CIK (CIK/MV) cells, and the addition of an anti-NKG2D antibody inhibited cytolysis by 50%. However, human bone marrow stromal cells can reduce CIK and CIK/MV mediated killing of myeloma cells (RPMI 8226, JJN-3 and MM1). In vivo, CIK and CIK/MV prolonged the survival of mice with systemic myeloma, although CIK/MV showed enhanced antitumor activity compared with CIK. Irradiation of the KAS-6/1 cells induced mRNA and protein expression of NKG2D ligands, MICA, and MICB in a dose-dependent manner and enhanced delivery of CIK/MV to the irradiated tumors. In both subcutaneous and disseminated myeloma models, XRT at 2 Gy resulted in superior prolongation of the survival of mice given CIK/MV therapy compared with CIK/MV with no XRT. This study demonstrates the potential of CIK against myeloma and that the combination of virotherapy with radiation could be used to further enhance therapeutic outcome using CIK cells.


Assuntos
Células Matadoras Induzidas por Citocinas/transplante , Imunoterapia Adotiva , Vírus do Sarampo/fisiologia , Mieloma Múltiplo/terapia , Terapia Viral Oncolítica , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacologia , Antígenos de Neoplasias/biossíntese , Antígenos de Neoplasias/genética , Células Cultivadas/imunologia , Células Cultivadas/transplante , Células Cultivadas/virologia , Técnicas de Cocultura , Terapia Combinada , Células Matadoras Induzidas por Citocinas/imunologia , Células Matadoras Induzidas por Citocinas/virologia , Citotoxicidade Imunológica , Feminino , Antígenos de Histocompatibilidade Classe I/biossíntese , Antígenos de Histocompatibilidade Classe I/genética , Humanos , Camundongos , Camundongos Endogâmicos ICR , Camundongos SCID , Mieloma Múltiplo/imunologia , Mieloma Múltiplo/patologia , Mieloma Múltiplo/radioterapia , Subfamília K de Receptores Semelhantes a Lectina de Células NK/imunologia , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Distribuição Aleatória , Células Estromais/transplante , Replicação Viral , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...