Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Cells ; 12(5)2023 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-36899831

RESUMO

Circulating microRNAs (miRNAs) have aroused a lot of interest as reliable blood diagnostic biomarkers of Alzheimer's disease (AD). Here, we investigated the panel of expressed blood miRNAs in response to aggregated Aß1-42 peptides infused in the hippocampus of adult rats to mimic events of the early onset of non-familial AD disorder. Aß1-42 peptides in the hippocampus led to cognitive impairments associated with an astrogliosis and downregulation of circulating miRNA-146a-5p, -29a-3p, -29c-3p, -125b-5p, and-191-5p. We established the kinetics of expression of selected miRNAs and found differences with those detected in the APPswe/PS1dE9 transgenic mouse model. Of note, miRNA-146a-5p was exclusively dysregulated in the Aß-induced AD model. The treatment of primary astrocytes with Aß1-42 peptides led to miRNA-146a-5p upregulation though the activation of the NF-κB signaling pathway, which in turn downregulated IRAK-1 but not TRAF-6 expression. As a consequence, no induction of IL-1ß, IL-6, or TNF-α was detected. Astrocytes treated with a miRNA-146-5p inhibitor rescued IRAK-1 and changed TRAF-6 steady-state levels that correlated with the induction of IL-6, IL-1ß, and CXCL1 production, indicating that miRNA-146a-5p operates anti-inflammatory functions through a NF-κB pathway negative feedback loop. Overall, we report a panel of circulating miRNAs that correlated with Aß1-42 peptides' presence in the hippocampus and provide mechanistic insights into miRNA-146a-5p biological function in the development of the early stage of sporadic AD.


Assuntos
Doença de Alzheimer , MicroRNAs , Animais , Camundongos , Ratos , Doença de Alzheimer/metabolismo , Anti-Inflamatórios/metabolismo , Astrócitos/metabolismo , Interleucina-6/metabolismo , MicroRNAs/metabolismo , NF-kappa B/metabolismo
2.
RNA Biol ; 18(sup1): 198-214, 2021 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-34570661

RESUMO

A major unresolved challenge in miRNA biology is the capacity to monitor the spatiotemporal activity of miRNAs expressed in animal disease models. We recently reported that the miRNA-ON monitoring system called RILES (RNAi-inducible expression Luciferase system) implanted in lentivirus expression system (LentiRILES) offers unique opportunity to decipher the kinetics of miRNA activity in vitro, in relation with their intracellular trafficking in glioblastoma cells. In this study, we describe in detail the method for the production of LentiRILES stable cell lines and employed it in several applications in the field of miRNA biology and therapy. We show that LentiRILES is a robust, highly specific and sensitive miRNA sensor system that can be used in vitro as a single-cell miRNA monitoring method, cell-based screening platform for miRNA therapeutics and as a tool to analyse the structure-function relationship of the miRNA duplex. Furthermore, we report the kinetics of miRNA activity upon the intracranial delivery of miRNA mimics in an orthotopic animal model of glioblastoma. This information is exploited to evaluate the tumour suppressive function of miRNA-200c as locoregional therapeutic modality to treat glioblastoma. Our data provide evidence that LentiRILES is a robust system, well suited to resolve the activity of endogenous and exogenously expressed miRNAs from basic research to gene and cell therapy.


Assuntos
Biomarcadores Tumorais/genética , Técnicas Biossensoriais/métodos , Regulação Neoplásica da Expressão Gênica , Genes Reporter , Glioblastoma/patologia , Lentivirus/genética , MicroRNAs/análise , Animais , Apoptose , Biomarcadores Tumorais/metabolismo , Ciclo Celular , Movimento Celular , Proliferação de Células , Feminino , Glioblastoma/genética , Glioblastoma/metabolismo , Humanos , Medições Luminescentes , Masculino , Camundongos , Camundongos SCID , MicroRNAs/genética , MicroRNAs/metabolismo , Pessoa de Meia-Idade , Células Tumorais Cultivadas
3.
J Control Release ; 327: 429-443, 2020 11 10.
Artigo em Inglês | MEDLINE | ID: mdl-32853728

RESUMO

MicroRNA (miRNA) oligonucleotides therapeutics are potent and attractive drugs for cancer treatment, but the kinetics of their intracellular trafficking, RISC processing and interaction with their mRNA targets in the cells are still not well understood. Moreover, the absence of efficient carriers impairs their translation into the clinic. Here, we compare the kinetics of miRNA-133a activity after transfection of U87MG glioblastoma cells with either a home-made lipopolyplexes (LPRi) or with the RNAiMax transfection reagent. For this purpose, we combined miRNA intracellular trafficking studies by confocal microscopy with our previously described RILES miRNA-ON reporter system subcloned here in a lentivirus expression vector (LentiRILES) for longitudinal analysis of miRNA activity in transfected cells. Using the LentiRILES system, we report significant differences in terms of miRNA delivery kinetics performed by these two transfection regents. We decipher the mechanisms of miRNA delivery by LPRi and investigate the main steps of miRNA internalization and cytosolic processing. We demonstrate that LPRi preferentially uses caveolae-mediated endocytosis as the main internalization pathway, releases miRNA into the cytosol after the first 3 h of incubation, and addresses the cytosolic miRNAs to P-bodies, while a fraction of miRNAs are exported to the extracellular space through exosomes which were found fully capable to re-transfect the cells. We implanted the LentiRILES cells in the brain of mice and infused the tumours with LPRi.miRNA using the convection-enhanced delivery method. Bioluminescence imaging of the live mice revealed efficient delivery of miRNAs in glioblastoma tumours, attesting successful miRNA uptake, internalization and RISC activation in vivo. Overall, our study provides a comprehensive overview of miRNA intracellular trafficking and processing in a glioblastoma context and highlights the potential use of LPRi for miRNA-based therapy.


Assuntos
Exossomos , Glioblastoma , MicroRNAs , Animais , Endocitose , Glioblastoma/genética , Glioblastoma/terapia , Camundongos , MicroRNAs/genética , Transfecção
4.
Int J Mol Sci ; 21(5)2020 Mar 03.
Artigo em Inglês | MEDLINE | ID: mdl-32138292

RESUMO

In order to harness local resources to improve well-being and human health, we aim in this study to investigate if the microalgae Dunaliella sp. isolated from the Tunisian coastal zone possesses any anticancer activity. Dunaliella sp. was cultured under normal (DSC) or stressed (DSS) conditions and extracted using different procedures. The biological activity assessment was performed on the Triple Negative Breast Cancer (TNBC) using 4T1 murine cells as a model. Results indicate that: (i) aqueous extract was the most cytotoxic compared to ethanolic and hydroalcoholic extracts; (ii) DSS activity was superior to that of DSC. DSS extracts induced apoptosis rather than necrosis, as evidenced by DNA fragmentation, PARP-1 cleavage and caspase-3 activation. Evaluation in an orthotopic TNBC model validated the anticancer activity in vivo. Intratumoral injection of DSS extract resulted in reduced tumor growth and an enhanced immune system activation. On the transcriptional side, the expression level of the immunosuppressive enzyme Arg-1 was decreased, as well as those of NOS-2 and COX-2 genes. These results suggest a potential anticancer activity of Tunisian Dunaliella sp. deserving further attention.


Assuntos
Antineoplásicos/química , Antineoplásicos/uso terapêutico , Microalgas/química , Extratos Vegetais/química , Extratos Vegetais/uso terapêutico , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Feminino , Marcação In Situ das Extremidades Cortadas , Camundongos , Camundongos Endogâmicos BALB C
5.
Anal Bioanal Chem ; 412(6): 1419-1430, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31940089

RESUMO

Insect venom is a highly complex mixture of bioactive compounds, containing proteins, peptides, and small molecules. Environmental factors can alter the venom composition and lead to intraspecific variation in its bioactivity properties. The investigation of discriminating compounds caused by variation impacts can be a key to manage sampling and explore the bioactive compounds. The present study reports the development of a peptidomic methodology based on UHPLC-ESI-QTOF-HRMS analysis followed by a nontargeted multivariate analysis to reveal the profile variance of Vespa velutina venom collected in different conditions. The reliability of the approach was enhanced by optimizing certain XCMS data processing parameters and determining the sample peak threshold to eliminate the interfering features. This approach demonstrated a good repeatability and a criterion coefficient of variation (CV) > 30% was set for deleting nonrepeatable features from the matrix. The methodology was then applied to investigate the impact of collection period variation. PCA and PLS-DA models were used and validated by cross-validation and permutation tests. A slight discrimination was found between winter and summer hornet venom in two successive years with 10 common discriminating compounds. Graphical abstract.


Assuntos
Cromatografia Líquida/métodos , Peptídeos/análise , Espectrometria de Massas por Ionização por Electrospray/métodos , Venenos de Vespas/química , Animais , Análise Multivariada , Reprodutibilidade dos Testes , Manejo de Espécimes
7.
ACS Nano ; 12(10): 9815-9829, 2018 10 23.
Artigo em Inglês | MEDLINE | ID: mdl-30256609

RESUMO

In vitro transcribed mRNA constitutes a versatile platform to encode antigens and to evoke CD8 T-cell responses. Systemic delivery of mRNA packaged into cationic liposomes (lipoplexes) has proven particularly powerful in achieving effective antitumor immunity in animal models. Yet, T-cell responses to mRNA lipoplexes critically depend on the induction of type I interferons (IFN), potent pro-inflammatory cytokines, which inflict dose-limiting toxicities. Here, we explored an advanced hybrid lipid polymer shell mRNA nanoparticle (lipopolyplex) endowed with a trimannose sugar tree as an alternative delivery vehicle for systemic mRNA vaccination. Like mRNA lipoplexes, mRNA lipopolyplexes were extremely effective in conferring antitumor T-cell immunity upon systemic administration. Conversely to mRNA lipoplexes, mRNA lipopolyplexes did not rely on type I IFN for effective T-cell immunity. This differential mode of action of mRNA lipopolyplexes enabled the incorporation of N1 methyl pseudouridine nucleoside modified mRNA to reduce inflammatory responses without hampering T-cell immunity. This feature was attributed to mRNA lipopolyplexes, as the incorporation of thus modified mRNA into lipoplexes resulted in strongly weakened T-cell immunity. Taken together, we have identified lipopolyplexes containing N1 methyl pseudouridine nucleoside modified mRNA as potent yet low-inflammatory alternatives to the mRNA lipoplexes currently explored in early phase clinical trials.


Assuntos
Inflamação/imunologia , Lipídeos/imunologia , RNA Mensageiro/imunologia , Linfócitos T/imunologia , Animais , Células Dendríticas/imunologia , Feminino , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Tamanho da Partícula , Polímeros/química , Propriedades de Superfície , Células Tumorais Cultivadas
8.
Trends Cancer ; 4(3): 222-238, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29506672

RESUMO

MicroRNAs (miRNAs) are key regulatory elements encoded by the genome. A single miRNA can downregulate the expression of multiple genes involved in diverse functions. Because cancer is a disease with multiple gene aberrations, developing novel approaches to identify and modulate miRNA pathways may result in a breakthrough for cancer treatment. With a special focus on glioblastoma (GBM), this review provides an up-to-date summary of miRNA biogenesis, the role of miRNA in cancer resistance, and essential tools for modulating miRNA expression, as well as of clinically promising RNAi delivery systems and how they can be adapted for therapy.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Glioblastoma/tratamento farmacológico , MicroRNAs/uso terapêutico , Animais , Barreira Hematoencefálica/metabolismo , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Terapia Genética/métodos , Glioblastoma/genética , Glioblastoma/metabolismo , Humanos
9.
Nat Biotechnol ; 34(7): 746-51, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27272383

RESUMO

Widespread application of gene therapy will depend on the development of simple methods to regulate the expression of therapeutic genes. Here we harness an endogenous signaling pathway to regulate therapeutic gene expression through diet. The GCN2-eIF2α signaling pathway is specifically activated by deficiencies in any essential amino acid (EAA); EAA deficiency leads to rapid expression of genes regulated by ATF4-binding cis elements. We found that therapeutic genes under the control of optimized amino acid response elements (AAREs) had low basal expression and high induced expression. We applied our system to regulate the expression of TNFSF10 (TRAIL) in the context of glioma therapy and found that intermittent activation of this gene by EEA-deficient meals retained its therapeutic efficacy while abrogating its toxic effects on normal tissue. The GCN2-eIF2α pathway is expressed in many tissues, including the brain, and is highly specific to EAA deficiency. Our system may be particularly well suited for intermittent regulation of therapeutic transgenes over short or long time periods.


Assuntos
Aminoácidos Essenciais/administração & dosagem , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/terapia , Terapia Genética/métodos , Ligante Indutor de Apoptose Relacionado a TNF/genética , Ligante Indutor de Apoptose Relacionado a TNF/uso terapêutico , Administração Oral , Aminoácidos Essenciais/farmacocinética , Animais , Suplementos Nutricionais , Relação Dose-Resposta a Droga , Ingestão de Alimentos/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Masculino , Camundongos , Transgenes/genética , Resultado do Tratamento
10.
Hum Gene Ther ; 27(2): 127-33, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26603492

RESUMO

Unlike for other digestive cancer entities, chemotherapy, radiotherapy, and targeted therapies have, so far, largely failed to improve patient survival in pancreatic adenocarcinoma (PDAC), which remains the fourth leading cause of cancer-related death in Europe and the United States. In this context, gene therapy may offer a new avenue for patients with PDAC. In this review, we explore the research currently ongoing in French laboratories aimed at defeating PDAC using nonviral therapeutic gene delivery, targeted transgene expression, or oncolytic virotherapy that recently or will soon bridge the gap between experimental models of cancer and clinical trials. These studies are likely to change clinical practice or thinking about PDAC management, as they represent a major advance not only for PDAC but may also significantly influence the field of gene-based molecular treatment of cancer.


Assuntos
Adenocarcinoma/terapia , Regulação Neoplásica da Expressão Gênica , Terapia Genética/métodos , Terapia Viral Oncolítica/métodos , Neoplasias Pancreáticas/terapia , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Adenovírus Humanos/genética , Adenovírus Humanos/metabolismo , Ensaios Clínicos como Assunto , Desoxicitidina Quinase/genética , Desoxicitidina Quinase/metabolismo , Vetores Genéticos/química , Vetores Genéticos/metabolismo , Humanos , MicroRNAs/genética , MicroRNAs/metabolismo , Núcleosídeo-Fosfato Quinase/genética , Núcleosídeo-Fosfato Quinase/metabolismo , Orthoreovirus de Mamíferos/genética , Orthoreovirus de Mamíferos/metabolismo , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Receptores de Somatostatina/genética , Receptores de Somatostatina/metabolismo , Transgenes
11.
J Control Release ; 213: 175-191, 2015 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-26184050

RESUMO

The co-delivery of minicircle DNA (mcDNA) and small anti-cancer drugs via stimuli-sensitive nanocarriers is a promising approach for combinatorial cancer therapy. However, the simultaneous loading of drugs and DNA in nanosized delivery systems is remarkably challenging. In this study we describe the synthesis of triblock copolymer micelles based on poly(2-ethyl-2-oxazoline)-poly(L-lactide) grafted with bioreducible polyethylenimine (PEOz-PLA-g-PEI-SS) for co-delivery of supercoiled (sc) mcDNA vectors and Doxorubicin (Dox). These amphiphilic carriers take advantage of non-fouling oxazolines to confer biological stability, of PLA to provide a hydrophobic core for drug encapsulation and of bioreducible PEI-SS to provide mcDNA complexation and an on-demand stimuli-responsive release. The obtained results show that mcDNA-loaded micelleplexes penetrate into in vitro tumor spheroid models with specific kinetics and exhibit a higher gene expression when compared to non-bioreducible nanocarriers. Moreover, in vivo bioluminescence imaging showed that gene expression is detected up to 8days following mcDNA-micelles intratumoral administration. Furthermore, drug-gene co-delivery in PEOz-PLA-g-PEI-SS carriers was verified by successful encapsulation of both Dox and mcDNA with high efficacy. Moreover, dual-loaded micelleplexes presented significant uptake and a cytotoxic effect in 2D cultures of cancer cells. The co-delivery of mcDNA-Dox to B16F10-Luciferase tumor bearing mice resulted in a reduction in tumor volume and cancer cells viability. Overall, such findings indicate that bioreducible triblock micelles are efficient for focal delivery in vivo and have potential for future application in combinatorial DNA-drug therapy.


Assuntos
Antibióticos Antineoplásicos/administração & dosagem , DNA Circular/administração & dosagem , Preparações de Ação Retardada/química , Doxorrubicina/administração & dosagem , Melanoma Experimental/tratamento farmacológico , Poliaminas/química , Polietilenoimina/química , Animais , Antibióticos Antineoplásicos/farmacocinética , Antibióticos Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , DNA Circular/farmacocinética , DNA Circular/uso terapêutico , Doxorrubicina/farmacocinética , Doxorrubicina/uso terapêutico , Feminino , Humanos , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos C57BL , Micelas , Poliésteres/química
12.
Nanomedicine ; 11(7): 1735-44, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26048315

RESUMO

Tendon injury is a major musculoskeletal disorder with a high public health impact. We propose a non-viral based strategy of gene therapy for the treatment of tendon injuries using histidylated vectors. Gene delivery of fibromodulin, a proteoglycan involved in collagen assembly was found to promote rat Achilles tendon repair in vivo and in vitro. In vivo liposome-based transfection of fibromodulin led to a better healing after surgical injury, biomechanical properties were better restored compared to untransfected control. These measures were confirmed by histological observations and scoring. To get better understandings of the mechanisms underlying fibromodulin transfection, an in vitro tendon healing model was developed. In vitro, polymer-based transfection of fibromodulin led to the best wound enclosure speed and a pronounced migration of tenocytes primary cultures was observed. These results suggest that fibromodulin non-viral gene therapy could be proposed as a new therapeutic strategy to accelerate tendon healing. FROM THE CLINICAL EDITOR: Tendon injury is relatively common and healing remains unsatisfactory. In this study, the effects of liposomal-based delivery of fibromodulin gene were investigated in a rat Achilles tendon injury model. The positive results observed would provide a new therapeutic strategy in clinical setting in the future.


Assuntos
Proteínas da Matriz Extracelular/genética , Técnicas de Transferência de Genes , Terapia Genética , Proteoglicanas/genética , Traumatismos dos Tendões/terapia , Tendão do Calcâneo/patologia , Adenoviridae/genética , Animais , Proteínas da Matriz Extracelular/biossíntese , Fibromodulina , Vetores Genéticos , Humanos , Lipossomos/química , Masculino , Proteoglicanas/biossíntese , Ratos , Traumatismos dos Tendões/genética , Traumatismos dos Tendões/patologia , Cicatrização/genética
13.
Int J Mol Sci ; 16(3): 4947-72, 2015 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-25749473

RESUMO

MicroRNAs (miRNAs) are a class of small non-coding RNAs that regulate gene expression by binding mRNA targets via sequence complementary inducing translational repression and/or mRNA degradation. A current challenge in the field of miRNA biology is to understand the functionality of miRNAs under physiopathological conditions. Recent evidence indicates that miRNA expression is more complex than simple regulation at the transcriptional level. MiRNAs undergo complex post-transcriptional regulations such miRNA processing, editing, accumulation and re-cycling within P-bodies. They are dynamically regulated and have a well-orchestrated spatiotemporal localization pattern. Real-time and spatio-temporal analyses of miRNA expression are difficult to evaluate and often underestimated. Therefore, important information connecting miRNA expression and function can be lost. Conventional miRNA profiling methods such as Northern blot, real-time PCR, microarray, in situ hybridization and deep sequencing continue to contribute to our knowledge of miRNA biology. However, these methods can seldom shed light on the spatiotemporal organization and function of miRNAs in real-time. Non-invasive molecular imaging methods have the potential to address these issues and are thus attracting increasing attention. This paper reviews the state-of-the-art of methods used to detect miRNAs and discusses their contribution in the emerging field of miRNA biology and therapy.


Assuntos
MicroRNAs/metabolismo , Imagem Molecular , Animais , Terapia Baseada em Transplante de Células e Tecidos , Corantes Fluorescentes/química , Corantes Fluorescentes/metabolismo , Perfilação da Expressão Gênica , Terapia Genética , Modelos Animais , Neoplasias/genética , Neoplasias/patologia , Interferência de RNA
14.
J Gene Med ; 13(12): 692-701, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22028274

RESUMO

BACKGROUND: Preclinical studies have demonstrated that, unlike oncolytic adenoviruses, oncolytic vaccinia viruses can reach implanted tumors upon systemic injection. However, the biodistribution of this oncolytic agent in in situ autochthonous tumor models remains poorly characterized. In the present study, we assessed this biodistribution in a model of mouse hepatocellular carcinoma (HCC) obtained after injection of the carcinogen diethylnitrosamine (DEN). METHODS: Twelve months after DEN administration, histology, quantitative reverse transcription-polymerase chain reaction, in situ hybridization and viral titration were used to characterize tumors, as well as to assess the viral load of the livers upon either intravenous or intraperitoineal injection. RESULTS: The results obtained showed that the architecture of the liver was lost, with a noticeable absence of sinusoids, as well as the presence of steatosis and α-fetoprotein-positive HCC tumor nodules. Bioluminescence imaging and measures of the infective virus load demonstrated that intravenous injection of 10(8) plaque-forming units of the recombinant vaccinia virus led to a predominant transduction of the liver, whereas intraperitoneal injection resulted in a lower level of liver transduction accompanied by an increased infection of the lungs, spleen, kidneys and bowels. Immunohistochemical analysis of liver sections of animals injected intravenously with the virus revealed a preferential localization of vaccinia-specific immunoreactivity in the tumors. CONCLUSIONS: The findings of the present study emphasize the importance of the route of administration of the vector and highlight the relevance of systemic injection of oncolytic vaccinia virus in the context of hepatocellular carcinoma.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Neoplasias Experimentais , Vírus Oncolíticos/genética , Poxviridae/genética , Animais , Vacinas Anticâncer/administração & dosagem , Vacinas Anticâncer/genética , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/terapia , Carcinoma Hepatocelular/virologia , Linhagem Celular Tumoral , Vetores Genéticos , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/terapia , Neoplasias Hepáticas/virologia , Camundongos , Neoplasias Experimentais/genética , Neoplasias Experimentais/terapia , Neoplasias Experimentais/virologia , Terapia Viral Oncolítica , Distribuição Tecidual
15.
Eur J Nucl Med Mol Imaging ; 37(7): 1377-85, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20140612

RESUMO

PURPOSE: In vivo imaging of the spread of oncolytic viruses using the Na/I symporter (NIS) has been proposed. Here, we assessed whether the presence of NIS in the viral genome affects the therapeutic efficacy of the oncolytic adenovirus dl922-947 following intraperitoneal administration, in a mouse model of peritoneal ovarian carcinoma. METHODS: We generated AdAM7, a dl922-947 oncolytic adenovirus encoding the NIS coding sequence. Iodide uptake, NIS expression, infectivity and cell-killing activity of AdAM7, as well as that of relevant controls, were determined in vitro. In vivo, the propagation of this virus in the peritoneal cavity of tumour-bearing mice was determined using SPECT/CT imaging and its therapeutic efficacy was evaluated. RESULTS: In vitro infection of ovarian carcinoma IGROV-1 cells with ADAM7 led to functional expression of NIS. However, the insertion of NIS into the viral genome resulted in a loss of efficacy of the virus in terms of replication and cytotoxicity. In vivo, on SPECT/CT imaging AdAM7 was only detectable in the peritoneal cavity of animals bearing peritoneal ovarian tumours for up to 5 days after intraperitoneal administration. Therapeutic experiments in vivo demonstrated that AdAM7 is as potent as its NIS-negative counterpart. CONCLUSION: This study demonstrated that despite the detrimental effect observed in vitro, insertion of the reporter gene NIS in an oncolytic adenovirus did not affect its therapeutic efficacy in vivo. We conclude that NIS is a highly relevant reporter gene to monitor the fate of oncolytic adenovectors in live subjects.


Assuntos
Adenoviridae/fisiologia , Genes Reporter/genética , Imagem Molecular/métodos , Vírus Oncolíticos/fisiologia , Neoplasias Peritoneais/virologia , Simportadores/genética , Replicação Viral , Adenoviridae/genética , Animais , Linhagem Celular Tumoral , Feminino , Regulação Neoplásica da Expressão Gênica , Genoma Viral/genética , Injeções Intraperitoneais , Camundongos , Vírus Oncolíticos/genética , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/terapia , Neoplasias Ovarianas/virologia , Neoplasias Peritoneais/genética , Neoplasias Peritoneais/terapia
16.
Br J Pharmacol ; 159(4): 761-71, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19814733

RESUMO

Biotherapies involve the utilization of antibodies, genetically modified viruses, bacteria or cells for therapeutic purposes. Molecular imaging has the potential to provide unique information that will guarantee their biosafety in humans and provide a rationale for the future development of new generations of reagents. In this context, non-invasive imaging of gene expression is an attractive prospect, allowing precise, spacio-temporal measurements of gene expression in longitudinal studies involving gene transfer vectors. With the emergence of cell therapies in regenerative medicine, it is also possible to track cells injected into subjects. In this context, the Na/I symporter (NIS) has been used in preclinical studies. Associated with a relevant radiotracer ((123)I(-), (124)I(-), (99m)TcO4(-)), NIS can be used to monitor gene transfer and the spread of selectively replicative viruses in tumours as well as in cells with a therapeutic potential. In addition to its imaging potential, NIS can be used as a therapeutic transgene through its ability to concentrate therapeutic doses of radionuclides in target cells. This dual property has applications in cancer treatment and could also be used to eradicate cells with therapeutic potential in the case of adverse events. Through experience acquired in preclinical studies, we can expect that non-invasive molecular imaging using NIS as a transgene will be pivotal for monitoring in vivo the exact distribution and pharmacodynamics of gene expression in a precise and quantitative way. This review highlights the applications of NIS in biotherapy, with a particular emphasis on image-guided radiotherapy, monitoring of gene and vector biodistribution and trafficking of stem cells.


Assuntos
Terapia Biológica/métodos , Regulação da Expressão Gênica , Genes Reporter , Imagem Molecular , Técnicas de Sonda Molecular , Simportadores/genética , Animais , Transplante de Células , Terapia Genética , Humanos , Terapia Viral Oncolítica , Dosagem Radioterapêutica , Simportadores/metabolismo
17.
Clin Cancer Res ; 15(21): 6595-601, 2009 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-19861465

RESUMO

PURPOSE: The Na/I symporter (hNIS) promotes concentration of iodine in cells. In cancer gene therapy, this transgene has potential as a reporter gene for molecular imaging of viral biodistribution and as a therapeutic protein promoting (131)I-mediated radiotherapy. Here, we combined the imaging and therapeutic potential of hNIS in an oncolytic adenoviruses targeting colorectal cancer cells. EXPERIMENTAL DESIGN: We generated an adenovirus (AdIP2) encoding hNIS and capable of selective replication in colorectal carcinoma cells. The selectivity of this virus was verified in vitro and in vivo. Its spread in tumors was monitored in vivo using single-photon emission computed tomography/CT imaging upon (99m)TcO(4)(-) injection and confirmed by immunohistochemistry. Metabolic radiotherapy was done through injection of therapeutic doses of (131)I(-). RESULTS: We showed in vitro and in vivo the selectivity of AdIP2 and that hNIS expression is restricted to the target cells. Imaging and immunohistochemical data showed that viral spread is limited and that the point of maximal hNIS expression is reached 48 hours after a single intratumoral injection. Administration of a single therapeutic dose of (131)I at this time point led to a dramatic reduction in tumor size not observed in hNIS-negative viruses. CONCLUSIONS: This report showed for the first time that the combination of the imaging and therapeutic potentials of hNIS can be applied to oncolytic adenoviruses in experimental models of cancer.


Assuntos
Adenoviridae/genética , Neoplasias Colorretais/diagnóstico por imagem , Neoplasias Colorretais/terapia , Genes Reporter , Terapia Genética/métodos , Radioisótopos do Iodo/uso terapêutico , Simportadores/genética , Proteínas Wnt/metabolismo , Animais , Linhagem Celular Tumoral , Diagnóstico por Imagem , Marcação de Genes , Vetores Genéticos , Camundongos , Camundongos Nus , Cintilografia , Simportadores/metabolismo , Transgenes
18.
J Immunother ; 32(8): 845-55, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19752749

RESUMO

Through their ability to induce cytotoxic T-lymphocytes and inhibit Foxp3 T-regulatory cells, Escherichia coli expressing listeriolysin-O (LLO) and a model tumor antigen have been shown to exert strong antitumor activity. The aim of this study is to extend these observations to a self-protein and clinically relevant tumor antigen associated with most types of adult leukemia: Wilms tumor gene 1 (WT1). We demonstrate that an E. coli coexpressing LLO and WT1 is capable of inducing a strong antitumor effect against WT1-expressing tumors in vivo through its ability to induce cytotoxic T-lymphocytes and inhibit the function of Foxp3 T-regulatory cells. Furthermore, we have characterized the immunodominant epitope involved in this effect (NAPYLPSCL) and demonstrated that coinjection of NAPYLPSCL with E. coli-LLO resulted in an antitumor effect largely equivalent to that obtained with E. coli-LLO/WT1. Our data demonstrate that the results obtained with a clinically irrelevant model tumor antigen remain valid with a "real" tumor antigen and that the adjuvant properties of the E. coli-LLO vaccine can be exploited in conjunction with peptides. The results obtained in this study will facilitate the translation of this work to human studies by combining antigenic motifs relevant to specific human leukocyte antigen haplotypes with the adjuvant effect of E. coli-LLO.


Assuntos
Adjuvantes Imunológicos/metabolismo , Toxinas Bacterianas/metabolismo , Vacinas Anticâncer , Escherichia coli/imunologia , Proteínas de Choque Térmico/metabolismo , Proteínas Hemolisinas/metabolismo , Leucemia/imunologia , Proteínas WT1/metabolismo , Adjuvantes Imunológicos/genética , Adulto , Animais , Toxinas Bacterianas/genética , Toxinas Bacterianas/imunologia , Linhagem Celular Tumoral , Escherichia coli/genética , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/imunologia , Proteínas Hemolisinas/genética , Proteínas Hemolisinas/imunologia , Humanos , Imunização , Epitopos Imunodominantes/genética , Epitopos Imunodominantes/imunologia , Epitopos Imunodominantes/metabolismo , Imunoglobulinas/sangue , Leucemia/patologia , Leucemia/terapia , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Transplante de Neoplasias , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/imunologia , Fragmentos de Peptídeos/metabolismo , Engenharia de Proteínas , Linfócitos T Citotóxicos/imunologia , Proteínas WT1/genética , Proteínas WT1/imunologia
19.
Cancer Res ; 69(6): 2655-62, 2009 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-19258509

RESUMO

The lack of safe and efficient systemic gene delivery vectors has largely reduced the potential of gene therapy in the clinic. Previously, we have reported that polypropylenimine dendrimer PPIG3/DNA nanoparticles are capable of tumor transfection upon systemic administration in tumor-bearing mice. To be safely applicable in the clinic, it is crucial to investigate the colloidal stability of nanoparticles and to monitor the exact biodistribution of gene transfer in the whole body of the live subject. Our biophysical characterization shows that dendrimers, when complexed with DNA, are capable of forming spontaneously in solution a supramolecular assembly that possesses all the features required to diffuse in experimental tumors through the enhanced permeability and retention effect. We show that these nanoparticles are of sizes ranging from 33 to 286 nm depending on the DNA concentration, with a colloidal stable and well-organized fingerprint-like structure in which DNA molecules are condensed with an even periodicity of 2.8 nm. Whole-body nuclear imaging using small-animal nano-single-photon emission computed tomography/computer tomography scanner and the human Na/I symporter (NIS) as reporter gene shows unique and highly specific tumor targeting with no detection of gene transfer in any of the other tissues of tumor-bearing mice. Tumor-selective transgene expression was confirmed by quantitative reverse transcription-PCR at autopsy of scanned animals, whereas genomic PCR showed that the tumor sites are the predominant sites of nanoparticle accumulation. Considering that NIS imaging of transgene expression has been recently validated in humans, our data highlight the potential of these nanoparticles as a new formulation for cancer gene therapy.


Assuntos
DNA/química , Técnicas de Transferência de Genes , Nanopartículas/química , Polipropilenos/química , Animais , Coloides/química , DNA/genética , Dendrímeros/química , Estabilidade de Medicamentos , Feminino , Análise de Fourier , Células HeLa , Humanos , Luz , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Camundongos Nus , Microscopia Eletrônica de Transmissão/métodos , Plasmídeos/química , Plasmídeos/genética , Espalhamento de Radiação , Transplante Heterólogo
20.
Clin Transl Oncol ; 10(3): 137-42, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18321815

RESUMO

Melanoma is a malignant tumour derived from melanocytes (dendritic cells originated from the neural crest and capable to produce melanin synthesis) that could be established on the skin or less frequently on the uvea. The cellular origin from both kind of melanoma seems to be the same but the melanocytes migrates to the epithelia for cutaneous melanoma, while for uveal melanoma, they migrate to mesodermic tissues. Despite the common origin, both melanomas show extreme differences in their metastatic potential, clinical response to treatments, immune response and genetic alterations. We will describe some of those differences in this review.


Assuntos
Melanoma/patologia , Neoplasias Cutâneas/patologia , Neoplasias Uveais/patologia , Humanos , Melanoma/genética , Melanoma/imunologia , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/imunologia , Neoplasias Uveais/genética , Neoplasias Uveais/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA