Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Reprod Immunol ; 162: 104213, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38364342

RESUMO

Endometriosis (EMS) is known to be closely associated with inflammation. We evaluate the possible mechanism linking the PI3K/AKT signaling pathway with pyroptosis and inflammation in EMS. We collected 30 patients undergoing laparoscopic for endometriosis as the EMS group and those undergoing surgery for uterine fibroids as the control group, from whom we collected serum, normal endometrium, eutopic endometrium and ectopic endometrium. Transmission electron microscopy (TEM) was used to observe the internal structure of endometrial cells. Western Blot was used to detect the protein expression of PI3K, P-PI3K, AKT, P-AKT, NLRP3, Caspase-1, GSDMD, and GSDMD-N. Immunohistochemistry (IHC) staining was used to detect the expression of PI3K, AKT, NLRP3, Caspase-1, GSDMD, and GSDMD-N proteins. Immunofluorescence (IF) staining was used to observe the expression of GSDMD-N. Quantitative real-time polymerase chain reaction (qRT-PCR) was used to analyze the mRNA levels of PI3K, AKT, NLRP3, Caspase-1, GSDMD, and GSDMD-N. ELISA was used to detect serum levels of IL-1ß, IL-18, TLR4, and NF-κB. We found that activation of PI3K/AKT signaling pathway in endometriosis significantly increased the level of cellular pyroptosis and inflammatory factors. Our results suggest that there is a positive correlation between the PI3K/AKT signaling pathway and pyroptosisas well as inflammation in EMS patients.


Assuntos
Endometriose , Piroptose , Feminino , Humanos , Proteína 3 que Contém Domínio de Pirina da Família NLR , Fosfatidilinositol 3-Quinases , Proteínas Proto-Oncogênicas c-akt , Inflamação , Caspase 1 , Transdução de Sinais
2.
Mol Ther Methods Clin Dev ; 25: 136, 2022 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-35402631

RESUMO

[This corrects the article DOI: 10.1016/j.omtm.2021.08.007.].

3.
Mol Ther Methods Clin Dev ; 23: 78-86, 2021 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-34631928

RESUMO

The current methods for detecting circulating tumor cells (CTCs) suffer from several drawbacks. We report a novel method that is based on a chimeric virus probe and can detect CTCs with extremely high specificity and sensitivity. Moreover, it exclusively detects live CTCs, and its detection efficacy is not impacted by the variation of epithelial cell adhesion molecule (EpCAM) expression. The chimeric virus probe is composed of a capsid from human papillomavirus that provides the detection with high specificity and an SV40-based genome that can amplify extensively inside CTCs and, hence, endows the detection with high sensitivity. Furthermore, different marker genes can be incorporated into the probe to provide detection with versatility. These unique capabilities will likely improve the validity and utility of this CTC detection in several clinical applications, which is one of the drawbacks suffered by many of the current CTC detection methods.

4.
Int J Mol Sci ; 22(16)2021 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-34445669

RESUMO

Coronavirus Disease 2019 (COVID-19) remains a global health crisis, despite the development and success of vaccines in certain countries. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the virus that causes COVID-19, uses its spike protein to bind to the human cell surface receptor angiotensin-converting enzyme 2 (ACE2), which allows the virus to enter the human body. Using our unique cell screening technology, we identified two ACE2-binding peptoid compounds and developed dimeric derivatives (ACE2P1D1 and ACE2P2D1) that effectively blocked spike protein-ACE2 interaction, resulting in the inhibition of SARS-CoV-2 pseudovirus entry into human cells. ACE2P1D1 and ACE2P2D1 also blocked infection by a D614G mutant pseudovirus. More importantly, these compounds do not decrease ACE2 expression nor its enzyme activity (which is important in normal blood pressure regulation), suggesting safe applicability in humans.


Assuntos
Enzima de Conversão de Angiotensina 2/metabolismo , COVID-19/prevenção & controle , Peptidil Dipeptidase A/metabolismo , Peptoides/farmacologia , SARS-CoV-2/efeitos dos fármacos , Internalização do Vírus/efeitos dos fármacos , COVID-19/virologia , Humanos , Células MCF-7 , Peptoides/metabolismo , Ligação Proteica/efeitos dos fármacos , SARS-CoV-2/metabolismo , SARS-CoV-2/fisiologia , Glicoproteína da Espícula de Coronavírus/metabolismo , Tratamento Farmacológico da COVID-19
5.
Mol Ther Oncolytics ; 19: 33-46, 2020 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-33024817

RESUMO

One of the major hurdles for cancer immunotherapy is the host's innate antiviral defense mechanisms. They include innate immune cells, such as natural killer (NK) cells and macrophages, which can be recruited within hours to the site of injection to clear the introduced oncolytic viruses. Here, we report a strategy to redirect these infiltrating innate immune cells to attack tumor cells instead by arming herpes simplex virus (HSV)-derived oncolytic viruses with secreted chimeric molecules that can engage these innate immune cells with tumor cells to kill the latter. These chimeric molecules have, at their N terminus, a custom-binding moiety for a tumor-associated antigen (TAA) and at their C terminus, protein L (PL) that binds to immunoglobulins (Igs). The binding of PL to Igs exposes the Fc to the Fc receptors on the surface of the innate immune cells, trigging them to attack the engaged tumor cells. In vitro and in vivo evaluation in a murine tumor model with limited permissiveness to oncolytic HSVs showed that arming the viruses with these chimeric molecules significantly boosts the killing effect and therapeutic activity. Moreover, our data also showed that the combined killing effect from the engaged innate immune cells and the oncolytic virus resulted in a more efficient stimulation of neoantigen-specific antitumor immunity than the virotherapy alone. Our data suggest that arming an oncolytic virus with this strategy represents a unique and pragmatic way of potentiating the oncolytic and immunotherapeutic effect of virotherapy.

6.
BMC Geriatr ; 19(1): 214, 2019 08 07.
Artigo em Inglês | MEDLINE | ID: mdl-31390985

RESUMO

BACKGROUND: Hearing loss is one of the most common modifiable factors associated with cognitive and functional decline in geriatric populations. An accurate, easy-to-apply, and inexpensive hearing screening method is needed to detect hearing loss in community-dwelling elderly people, intervene early and reduce the negative consequences and burden of untreated hearing loss on individuals, families and society. However, available hearing screening tools do not adequately meet the need for large-scale geriatric hearing detection due to several barriers, including time, personnel training and equipment costs. This study aimed to propose an efficient method that could potentially satisfy this need. METHODS: In total, 1793 participants (≥60 years) were recruited to undertake a standard audiometric air conduction pure tone test at 4 frequencies (0.5-4 kHz). Audiometric data from one community were used to train the decision tree model and generate a pure tone screening rule to classify people with or without moderate or more serious hearing impairment. Audiometric data from another community were used to validate the tree model. RESULTS: In the decision tree analysis, 2 kHz and 0.5 kHz were found to be the most important frequencies for hearing severity classification. The tree model suggested a simple two-step screening procedure in which a 42 dB HL tone at 2 kHz is presented first, followed by a 47 dB HL tone at 0.5 kHz, depending on the individual's response to the first tone. This approach achieved an accuracy of 91.20% (91.92%), a sensitivity of 95.35% (93.50%) and a specificity of 86.85% (90.56%) in the training dataset (testing dataset). CONCLUSIONS: A simple two-step screening procedure using the two tones (2 kHz and 0.5 kHz) selected by the decision tree analysis can be applied to screen moderate-to-profound hearing loss in a community-based geriatric population in Shanghai. The decision tree analysis is useful in determining the optimal hearing screening criteria for local elderly populations. Implanting the pair of tones into a well-calibrated sound generator may create a simple, practical and time-efficient screening tool with high accuracy that is readily available at healthcare centers of all levels, thereby facilitating the initiation of extensive nationwide hearing screening in older adults.


Assuntos
Árvores de Decisões , Avaliação Geriátrica/métodos , Perda Auditiva/diagnóstico , Vida Independente , Programas de Rastreamento/métodos , Vigilância da População/métodos , Idoso , Idoso de 80 Anos ou mais , Audiometria de Tons Puros/métodos , Audiometria de Tons Puros/tendências , China/epidemiologia , Feminino , Perda Auditiva/epidemiologia , Humanos , Vida Independente/tendências , Masculino , Programas de Rastreamento/tendências , Pessoa de Meia-Idade
7.
Oncotarget ; 9(77): 34543-34553, 2018 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-30349648

RESUMO

Current oncolytic virotherapy is primarily administered by intratumoral injection. However, systemic delivery is desirable for treating patients, particularly for those who have developed metastatic diseases. Several components are impeding the systemic delivery efficiency of oncolytic viruses. Chief among them is the rapid clearance of viral particles by the host's mononuclear phagocyte system (MPS). We explored the possibility of genetically engrafting CD47, a "don't eat me" signal molecule, to the membrane envelop of an oncolytic herpes simplex virus (HSV) to enable it to escape from the MPS for systemic delivery. Our results show that this modification indeed allows the virus to be more efficiently delivered to local tumors by the systemic route. Moreover, this modification also prolongs the virus persistence in local tumors after it arrives there. Consequently, systemic delivery of the modified virus produced a measurable antitumor effect against a murine tumor model that is otherwise resistant to the parental virus delivered by the same route. Our data thus suggest that engrafting enveloped oncolytic viruses such as those derived from HSV with CD47 molecule represents a conceivable strategy to enhance the efficiency of systemic delivery.

8.
Oncotarget ; 9(30): 21348-21358, 2018 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-29765544

RESUMO

Herpes simplex virus (HSV) is one of the many viruses that have been modified or adapted for oncolytic purposes. There are two serotypes of HSV, HSV-1 and HSV-2. The majority of oncolytic HSVs, including T-VEC which has recently been approved by the US Food and Drug Administration (FDA) for clinical use in treating late stage melanoma patients, are derived from HSV-1. Recently, we and others have developed several HSV-2 based oncolytic viruses. During our in vitro characterization of oncolytic viruses developed from both serotypes (Baco-1 from HSV-1 and FusOn-H2 from HSV-2), we noticed there is a subpopulation of cancer cells in which both viruses could infect but only FusOn-H2 could spread from cell to cell on monolayers. This observation prompted us to investigate the virus receptor expression profiles in these and other tumor cells. Our data show the following: 1) This subpopulation of tumor cells only express nectin-2, not the other two major receptors (HVEM or nectin-1). 2) Baco-1 grows to a higher titer than FusOn-H2 in this subpopulation of tumor cells, but the latter kills these tumor cells more efficiently than the former. 3) FusOn-H2 is effective at treating tumors formed from these tumor cells while Baco-1 is completely ineffective. Our results suggest that this subpopulation of tumor cells may be intrinsically resistant to the therapeutic effect of a HSV-1 based oncolytic virus but they remain sensitive to a HSV-2 based virotherapy.

9.
BMC Cancer ; 15: 964, 2015 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-26674012

RESUMO

BACKGROUND: Many commonly used xenograft tumor models do not spontaneously metastasize to distant organs following subcutaneous or orthotopic implantation, limiting their usefulness in preclinical studies. It is generally believed that natural killer cells are the key component of the innate immune system in determining tumor metastatic potential in xenograft models. However, recent studies suggest that macrophages may play an important role, as resident macrophages can eliminate the invading tumor cells if they do not express adequate levels of the CD47 molecule. METHODS: We investigated the effect of overexpressing murine CD47 (mCD47) in PC-3 cells, a commonly used human prostate cancer line, on the metastatic potential in three mouse strains with different genetic background and varying degrees of immunodeficiency. We implanted the tumor cells either subcutaneously or orthotopically and then examined their local and distant metastases. RESULTS: Our results show that mCD47-expressing PC-3 cells subcutaneously implanted in NSG and CB17. Scid mice metastasized to the sentinel lymph node, lung and liver significantly more efficiently than the control cells. When implanted orthotopically to NOD. Scid mice, these cells spontaneously metastasized to lung and liver. CONCLUSIONS: Our data demonstrate that mCD47 can facilitate human tumor cell metastasis in murine models, and that these mCD47-expressing tumor cells may be useful for in vivo studies where spontaneous metastases are desirable.


Assuntos
Antígeno CD47/biossíntese , Modelos Animais de Doenças , Neoplasias da Próstata/patologia , Animais , Antígeno CD47/imunologia , Linhagem Celular Tumoral , Separação Celular , Citometria de Fluxo , Xenoenxertos , Humanos , Macrófagos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Invasividade Neoplásica/imunologia , Transplante de Neoplasias , Neoplasias da Próstata/imunologia , Transfecção
10.
Oncotarget ; 6(2): 902-14, 2015 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-25460506

RESUMO

Adoptive T-cell therapy has shown promises for cancer treatment. However, for treating solid tumors, there is a need for improving the ability of the adoptively transferred T cells to home to tumor sites. We explored the possibility of using an oncolytic virus derived from HSV-2, which can actively pull T effector cells to the site of infection, as a local attractant for migration of adoptively transferred T cells. Our data show that intratumoral administration of this virus can indeed attract active migration of the adoptively transferred T cells to the treated tumor. Moreover, once attracted to the tumor site by the virus, T cells persisted in there significantly longer than in mock-treated tumor. Chemokine profiling identified significant elevation of CXCL9 and CXCL10, as well as several other chemokines belonging to the inflammatory chemokine family in the virus-treated tumors. These chemokines initially guided the T-cell migration to and then maintained their persistence in the tumor site, leading to a significantly enhanced therapeutic effect. Our data suggests that this virotherapy may be combined with adoptive T-cell therapy to potentiate its therapeutic effect against solid tumors that are otherwise difficult to manage with the treatment alone.


Assuntos
Movimento Celular/imunologia , Herpesvirus Humano 2/imunologia , Neoplasias Experimentais/imunologia , Vírus Oncolíticos/imunologia , Linfócitos T/imunologia , Animais , Linhagem Celular , Linhagem Celular Tumoral , Quimiocinas/imunologia , Quimiocinas/metabolismo , Chlorocebus aethiops , Terapia Combinada , Herpesvirus Humano 2/fisiologia , Humanos , Imunoterapia Adotiva/métodos , Subunidade gama Comum de Receptores de Interleucina/deficiência , Subunidade gama Comum de Receptores de Interleucina/genética , Subunidade gama Comum de Receptores de Interleucina/imunologia , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Camundongos Transgênicos , Neoplasias Experimentais/terapia , Neoplasias Experimentais/virologia , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/fisiologia , Receptores de Quimiocinas/imunologia , Receptores de Quimiocinas/metabolismo , Linfócitos T/transplante , Carga Tumoral/imunologia , Células Vero
11.
Cell Biochem Biophys ; 69(3): 583-7, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24510538

RESUMO

The aim of this study is to observe the in vitro-targeted destruction of lung adenocarcinoma using recombinant Type I herpes simplex virus (HSV-I)-mediated gibbon ape leukemia virus envelope glycoprotein (GALV.fus), controlled by UL38 promoter and cytomegalovirus promoter (CMVP). A recombinant HSV-I plasmid encoding the GALV.fus was transfected into green monkey kidney cells, the lung adenocarcinoma line A549, and the human fetal fibroblast cell line HFL-I GNHu5 in various doses. The effects and expression of in vitro GALV.fus were observed using an inverted microscope. Enhanced green fluorescence protein expression served as the contro1 for GALV.fus. Recombinant HSV-I virus was produced. Fusogenic recombinant virus infection led to cell fusions in A549 in a dose-dependent manner. Nonfusogenic viruses only produced conventional cytotoxic effects. Recombinant HSV-I with the CMVP initiated cell fusions in HFL-1 GNHu5 cells with arrested cell cycles or as quiescence. HSV-I regulated by UL38p caused cell fusion only in growing cells. Protein expression of GALV.fus was confirmed by Western Blot in infected A549 and HFL-1 GNHu5. Delivery and tumor-specific expression of GALV.fus gene can selectively and safely target lung cancer in vitro, and may prove to be a novel gene therapy for lung cancer.


Assuntos
Adenocarcinoma/genética , Adenocarcinoma/terapia , Terapia Genética/métodos , Herpesvirus Humano 1/genética , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/terapia , Proteínas Virais de Fusão/genética , Adenocarcinoma/patologia , Adenocarcinoma de Pulmão , Animais , Linhagem Celular Tumoral , Chlorocebus aethiops , DNA Recombinante/genética , Expressão Gênica , Terapia Genética/efeitos adversos , Vetores Genéticos/genética , Herpesvirus Humano 1/fisiologia , Humanos , Vírus da Leucemia do Macaco Gibão/genética , Neoplasias Pulmonares/patologia , Terapia Viral Oncolítica , Plasmídeos/genética , Células Vero
12.
Cell Biochem Biophys ; 69(3): 577-82, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24519667

RESUMO

This study examined the efficacy of gene therapy of lung adenocarcinoma using specifically controlled type I herpes simplex virus recombinant vector expressing Gibbon ape leukemia virus membrane fusion glycoprotein gene (GALV.fus). Recombinant HSV-I plasmid carrying target transgene was constructed, and recombinant viral vector was generated in Vero cells using Lipofectamine transfection. Viral vector was introduced into lung adenocarcinoma A549 cells or human fetal fibroblast HFL-I GNHu 5 cells, or inoculated into human lung adenocarcinoma xenografts in nude mice. The anti-tumor and cytotoxic effects of GALV-FMG, the transgene, were examined in these cell and animal models. Expression of GALV-FMG in xenographs achieved 100 % tumorigenicity. Recombinant HSV-I viral vector also exhibited significant tumor cell killing effect in vitro. Relative survival rates of tumor cells treated with GALV-FMG or control vectors were, respectively, 20 and 70 %. GALV.fus has a potent anti-tumor effect against lung cancer both in vitro and in vivo. This anti-tumor potential provides foundation for further studies with this vector.


Assuntos
Adenocarcinoma/genética , Adenocarcinoma/terapia , Terapia Genética/métodos , Herpesvirus Humano 1/genética , Vírus da Leucemia do Macaco Gibão/genética , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/terapia , Proteínas Virais de Fusão/genética , Adenocarcinoma/patologia , Adenocarcinoma de Pulmão , Animais , Linhagem Celular Tumoral , Transformação Celular Neoplásica , Chlorocebus aethiops , DNA Recombinante/genética , Expressão Gênica , Terapia Genética/efeitos adversos , Vetores Genéticos/genética , Humanos , Neoplasias Pulmonares/patologia , Masculino , Camundongos , Células Vero
13.
Int J Cancer ; 133(10): 2483-92, 2013 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-23661285

RESUMO

Converting T cells into tumor cell killers by grafting them with a chimeric antigen receptor (CAR) has shown promise as a cancer immunotherapeutic. However, the inability of these cells to actively migrate and extravasate into tumor parenchyma has limited their effectiveness in vivo. Here we report the construction of a CAR containing an echistatin as its targeting moiety (eCAR). As echistatin has high binding affinity to αvß3 integrin that is highly expressed on the surface of endothelial cells of tumor neovasculature, T cells engrafted with eCAR (T-eCAR) can efficiently lyse human umbilical vein endothelial cells and tumor cells that express αvß3 integrin when tested in vitro. Systemic administration of T-eCAR led to extensive bleeding in tumor tissues with no evidence of damage to blood vessels in normal tissues. Destruction of tumor blood vessels by T-eCAR significantly inhibited the growth of established bulky tumors. Moreover, when T-eCAR was codelivered with nanoparticles in a strategically designed temporal order, it dramatically increased nanoparticle deposition in tumor tissues, pointing to the possibility that it may be used together with nanocarriers to increase their capability to selectively deliver antineoplastic drugs to tumor tissues.


Assuntos
Melanoma Experimental/irrigação sanguínea , Melanoma Experimental/terapia , Nanopartículas/administração & dosagem , Linfócitos T/fisiologia , Sequência de Aminoácidos , Animais , Antígenos de Neoplasias/biossíntese , Antígenos de Neoplasias/genética , Vasos Sanguíneos/patologia , Linhagem Celular , Linhagem Celular Tumoral , Células Endoteliais da Veia Umbilical Humana/patologia , Humanos , Integrina alfaVbeta3/metabolismo , Masculino , Melanoma Experimental/metabolismo , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Proteínas Mutantes Quiméricas/biossíntese , Proteínas Mutantes Quiméricas/genética , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/genética , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Receptores de Antígenos/metabolismo , Linfócitos T/imunologia
14.
Mol Ther ; 20(10): 1871-81, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22692498

RESUMO

Interferon (IFN) antiviral defense mechanism plays a critical role in controlling virus infection. It thus represents a formidable hurdle for virotherapy. Despite the reported ability of herpes simplex virus (HSV) to counteract this defense, the duration and extent of HSV infection in vivo is still largely dictated by host's IFN activity status. Because the HSV genes that have been reported to block IFN activity mainly act intracellularly, we hypothesized that their inhibitory effect could be enhanced by exploiting a gene whose product acts extracellularly. The B18R gene from vaccinia virus encodes a secreted decoy receptor with a broad antagonizing effect against type I IFNs. We therefore cloned B18R into an HSV-1-based oncolytic virus to generate Synco-B18R. In the presence of increased IFN levels in vitro, Synco-B18R largely retained its oncolytic effect, whereas the tumor-killing ability of the parental virus, Synco-2D, was severely compromised. When injected intratumorally in vivo, Synco-B18R showed significantly greater oncolytic activity than Synco-2D. Our results suggest that incorporation of the vaccinia virus B18R gene can safely potentiate the antitumor effect of an oncolytic HSV, and that similar strategies may be useful with other types of oncolytic viruses.


Assuntos
Herpesvirus Humano 1/genética , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/genética , Vaccinia virus/genética , Proteínas Virais/genética , Animais , Antivirais/farmacologia , Linhagem Celular Tumoral , Chlorocebus aethiops , Clonagem Molecular , Feminino , Herpesvirus Humano 1/fisiologia , Humanos , Interferons/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Vírus Oncolíticos/fisiologia , Plasmídeos , Recombinação Genética , Células Vero , Replicação Viral
15.
Mol Ther ; 20(2): 339-46, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22146341

RESUMO

Selective replication in tumor cells is a highly desirable feature for oncolytic viruses. Recent studies have shown that microRNAs (miRNAs) play important roles in controlling gene expression, and that certain tissue-specific miRNAs are frequently downregulated in malignant cells. miR-122 is a liver-specific microRNA. It is abundantly expressed in normal hepatocytes but is absent in many hepatocellular carcinoma (HCC) cells. We hypothesized that expression of an essential viral gene by a liver-specific promoter would initially restrict virus replication to cells of hepatic origin and that adding miR-122 complementary sequences to the viral gene would make the transcripts degradable by miR-122 in normal hepatocytes, thus further confining its replication to HCC. We have constructed such an oncolytic herpes simplex virus by linking the essential viral glycoprotein H gene with the liver-specific apolipoprotein E (apoE)-AAT promoter and by adding the miR-122a complimentary sequence to the 3' untranslated region (3'UTR). To further increase the safety of this virus, complementary sequences from miR-124a and let-7 were also engineered into the same 3'UTR. Designated liver-cancer specific oncolytic virus (LCSOV), it was highly selective in killing HCC cells and in shrinking HCC xenografts. We conclude that LCSOV is a highly specific oncolytic virus that can precisely target HCC.


Assuntos
Carcinoma Hepatocelular/terapia , Vetores Genéticos/genética , Neoplasias Hepáticas/terapia , Vírus Oncolíticos/genética , Simplexvirus/genética , Tropismo Viral , Animais , Apolipoproteínas E/genética , Carcinoma Hepatocelular/metabolismo , Linhagem Celular , Chlorocebus aethiops , Feminino , Expressão Gênica , Ordem dos Genes , Genes Reporter , Vetores Genéticos/administração & dosagem , Vetores Genéticos/efeitos adversos , Humanos , Fígado/metabolismo , Neoplasias Hepáticas/metabolismo , Camundongos , Camundongos Nus , MicroRNAs/genética , Vírus Oncolíticos/fisiologia , Especificidade de Órgãos/genética , Regiões Promotoras Genéticas , Simplexvirus/fisiologia , Replicação Viral/genética , Ensaios Antitumorais Modelo de Xenoenxerto
16.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 42(2): 152-6, 2011 Mar.
Artigo em Chinês | MEDLINE | ID: mdl-21500543

RESUMO

OBJECTIVE: To observe the killing effect of recombinant type I herpes simplex virus (HSV-I) with Gibbon ape leukemia virus membrane fusion glycoprotein (GALV.fus) gene on lung adenocarcinoma in vitro and in vivo. METHODS: Recombinant HSV-I plasmids (HSV-UL38P-GALV.fus, HSV-CMVP-GALV.fus, HSV-CMVP-EGFP) was introduced into green monkey kidney cells (Vero) by liposome to amplify the virus, which were propagated in Vero cells and purified by cesium chloride density purification, titrated by TCID50 method. The three recombinant viruses were named as Synco-2, Synco-1 and Baco-1 respectively, and were transfected into lung adenocarcinoma cell line A549 cell and human lung adenocarcinoma xenografts which were established in nude mice subcutaneously to observe the expression and transfection of recombinant plasmids; mice model was divided to A (Control) group, B (Baco-1) group, C (Synco-1) group, D (Synco-2) group and E (Synco-2) group. The antitumor and cytotoxic effects of the virus in vitro or in vivo were investigated simultaneously. RESULTS: Recombinated HSV-I virus were packed successfully, the titre of Baco-1, Synco-1 and Synco-2 were 3 x 10(10) pfu/mL, 1X 10(11) pfu/mL and 4 x 10(10) pfu/mL respectively. The virus produced clear antitumor effects in vitro, the oncolytic activity of Synco-2 and Synco-1 was superior to that of Baco-1 (P < 0.01). The striking antitumor effect was seen when the virus was given subcutaneously in established xenografts in the animals. Tumor volume in Group C and D decreased significantly compared those in Group A and B (P < 0.01). The same result was observed in tumour weight (P < 0.01), and we also find that there was statistical significance between Group C and D in tumour quality at last two weeks (P < 0.01). CONCLUSIONS: The three recombinant HSV-I were packaged, amplificated and purified successfully. Recombinant GALV. fus gene system controlled by special promoter and mediated by available carrier has potent activity against lung cancer both in vitro or in vivo, and maybe a new promising candidate for investigative gene therapy of this malignancy.


Assuntos
Terapia Genética/métodos , Herpesvirus Humano 1/metabolismo , Vírus da Leucemia do Macaco Gibão/genética , Neoplasias Pulmonares/terapia , Glicoproteínas de Membrana/genética , Proteínas Virais de Fusão/farmacologia , Adenocarcinoma/patologia , Adenocarcinoma/terapia , Animais , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Chlorocebus aethiops , Herpesvirus Humano 1/genética , Humanos , Neoplasias Pulmonares/patologia , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Nus , Transplante de Neoplasias , Transfecção , Células Vero , Proteínas Virais de Fusão/genética
17.
Int J Cancer ; 129(6): 1503-10, 2011 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-21128236

RESUMO

Oncolytic herpes simplex virus (HSV) is currently in phase III clinical trials for development as a novel therapeutic agent against a broad range of human tumors. Although results have been promising, clinical outcome is likely to be compromised by intrinsic and acquired resistance to HSV replication, leading us to test agents that may overcome this obstacle. We found that, despite showing no effect on HSV replication in tumor cells fully permissive to the virus growth, the mTOR inhibitor rapamycin markedly increased the yield and dissemination of oncolytic HSVs in semipermissive tumor cells. Similar results were obtained in tumor-bearing mice. Co-administration of rapamycin with an HSV-derived oncolytic virus either blocked or reversed the growth of tumor xenografts established from semipermissive human tumor cells, while use of either agent alone produced only transient inhibitory effect. Together, our results suggest that rapamycin could be used to potentiate the activity of oncolytic HSVs against difficult-to-treat human tumors or perhaps to prevent the emergence of resistant tumor cells during virotherapy.


Assuntos
Antibióticos Antineoplásicos/farmacologia , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos , Simplexvirus , Sirolimo/farmacologia , Replicação Viral/efeitos dos fármacos , Animais , Linhagem Celular , Linhagem Celular Tumoral , Terapia Combinada , Feminino , Humanos , Camundongos , Camundongos Nus , Ensaios Antitumorais Modelo de Xenoenxerto
18.
PLoS One ; 5(7): e11867, 2010 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-20686618

RESUMO

A simple and sensitive method to quantitatively measure the cytolytic effect of tumor-specific T killer cells is highly desirable for basic and clinical studies. Chromium (51Cr) release assay has been the "gold standard" for quantifying cytolytic activities of cytotoxic T lymphocytes (CTLs) against target cells and this method is still being used in many laboratories. However, a major drawback of this method is the use of radioactive materials, which is inconvenient to handle because of environmental safety concerns and expensive due to the short half-life of the isotope. Consequently, several nonradioactive methods have been reported recently. Here we report a new method that we recently developed for quantifying antigen-specific cytolytic activity of CTLs. This method fully exploits the high sensitivity and the relative simplicity of luciferase quantitative assay. We initially expected the released luciferase in the supernatant to be the adequate source for monitoring cell death. However, to our total surprise, incubation of these killer T cells with the tumor cell targets did not result in significant release of luciferase in the culture medium. Instead, we found that the remaining luciferase inside the cells could accurately reflect the overall cell viability.


Assuntos
Bioensaio/métodos , Linfócitos T Citotóxicos/imunologia , Animais , Sobrevivência Celular/genética , Sobrevivência Celular/fisiologia , Humanos , Luciferases/genética , Luciferases/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Linfócitos T Citotóxicos/metabolismo
19.
Hum Gene Ther ; 21(12): 1687-96, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20583863

RESUMO

It has been reported that certain polypeptides derived from aggregation-prone cellular proteins can turn soluble green fluorescent protein (GFP) into aggregates. Here we report our finding that a short peptide derived from a viral gene, ICP10 of herpes simplex virus (HSV)-2, also possesses such a property. A sequence as short as 13 amino acids from the middle region of the gene can convert GFP into an aggregation-prone toxic protein once it is fused to the C terminus. Moreover, this short peptide can direct a surrogate tumor antigen into the autophagosome/lysosome degradation pathway, drastically increasing both MHC class I and class II antigen presentation. The simultaneous induction of both arms of the T cell immune response to the tumor antigen effectively protects the immunized animals from tumor challenge. Designated VIPA (i.e., viral inducer of protein aggregation), this unique viral sequence may represent an attractive candidate as a molecular adjuvant for cancer immunotherapy and for other immunologically preventable diseases.


Assuntos
Apresentação de Antígeno , Autofagia , Vacinas Anticâncer/imunologia , Herpesvirus Humano 2 , Lisossomos/metabolismo , Peptídeos/imunologia , Fagossomos/metabolismo , Proteínas Serina-Treonina Quinases/genética , Ribonucleotídeo Redutases/genética , Animais , Antígenos de Neoplasias/imunologia , Vacinas Anticâncer/genética , Chlorocebus aethiops , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Transplante de Neoplasias , Ovalbumina/genética , Ovalbumina/imunologia , Peptídeos/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Transfecção , Células Tumorais Cultivadas , Células Vero
20.
Cancer Res ; 67(16): 7850-5, 2007 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-17699791

RESUMO

Despite their unique property of selective replication and propagation in tumor tissues, oncolytic viruses have had only limited antitumor effects in cancer patients. One of the major reasons is probably the host's immune defense mechanisms, which can restrict the ability of the virus to replicate and spread within tumors. The innate immune system, which can be rapidly activated during virus infection, likely plays a more pivotal antiviral role than does acquired immunity, as the antitumor effect of an oncolytic virus is mainly generated during the acute phase of virus replication. To exploit the potential of cyclophosphamide, a cancer chemotherapeutic drug that also inhibits innate immune responses, to enhance the activity of oncolytic viruses, we evaluated the effect of coadministration of this drug with a herpes simplex virus-2-based oncolytic virus (FusOn-H2) against Lewis lung carcinoma, which is only semipermissive to infection with FusOn-H2. This strategy synergistically enhanced the antitumor effect against lung carcinoma growing in mice. It also potentiated the ability of FusOn-H2 to induce tumor-specific immune responses. Together, our results suggest that coadministration of FusOn-H2 with cyclophosphamide would be a feasible way to enhance the antitumor effects of this oncolytic virus in future clinical trials.


Assuntos
Carcinoma Pulmonar de Lewis/imunologia , Carcinoma Pulmonar de Lewis/terapia , Ciclofosfamida/farmacologia , Herpesvirus Humano 2/fisiologia , Terapia Viral Oncolítica/métodos , Animais , Antineoplásicos Alquilantes/farmacologia , Carcinoma Pulmonar de Lewis/virologia , Chlorocebus aethiops , Terapia Combinada , Feminino , Herpesvirus Humano 2/genética , Imunossupressores/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Células Vero , Replicação Viral/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA