Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
NPJ Regen Med ; 5: 3, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32133156

RESUMO

Therapeutic approaches requiring the intravenous injection of autologous or allogeneic mesenchymal stromal cells (MSCs) are currently being evaluated for treatment of a range of diseases, including orthopaedic injuries. An alternative approach would be to mobilise endogenous MSCs into the blood, thereby reducing costs and obviating regulatory and technical hurdles associated with development of cell therapies. However, pharmacological tools for MSC mobilisation are currently lacking. Here we show that ß3 adrenergic agonists (ß3AR) in combination with a CXCR4 antagonist, AMD3100/Plerixafor, can mobilise MSCs into the blood in mice and rats. Mechanistically we show that reversal of the CXCL12 gradient across the bone marrow endothelium and local generation of endocannabinoids may both play a role in this process. Using a spine fusion model we provide evidence that this pharmacological strategy for MSC mobilisation enhances bone formation.

2.
Blood Adv ; 1(22): 1934-1943, 2017 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-29296840

RESUMO

Pharmacological mobilization of hematopoietic progenitor cells (HPCs) is used clinically to harvest HPCs for bone marrow transplants. It is now widely accepted that the CXCR4:CXCL12 chemokine axis plays a critical role in the retention of HPCs in the bone marrow, and CXCR4 antagonists have been developed for their mobilization. The first of this class of drugs to be US Food and Drug Administration-approved was the bicyclam AMD3100. In addition to mobilizing HPCs and leukocytes in naïve mice, AMD3100 has been shown to mobilize mesenchymal progenitor cells (MPCs) in vascular endothelial growth factor (VEGF-A) pretreated mice. AMD3100 binds to the transmembrane region of CXCR4 and is thought to mobilize HPCs by reversing the gradient of CXCL12 across the bone marrow endothelium. Consistent with this hypothesis, our data show that selective neutralization of CXCL12, with chalcone 4-phosphate (C4P), inhibited AMD3100-stimulated mobilization of HPCs and leukocytes in naïve mice and MPCs in VEGF-A pretreated mice. In contrast it is shown here that the CXCR4 antagonist KRH3955 that binds to the extracellular loop of CXCR4 does not reverse the CXCL12 chemokine gradient. However, this drug efficiently mobilizes HPCs, a response that is not inhibited by C4P. In contrast, KRH3955 does not mobilize MPCs in VEGF-A pretreated mice. These data suggest that CXCR4 antagonists that bind to distinct regions of the receptor mobilize progenitor cells by distinct molecular mechanisms.

3.
Pharmacol Ther ; 151: 107-20, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25827580

RESUMO

Regenerative medicine using mesenchymal stem cells for the purposes of tissue repair has garnered considerable public attention due to the potential of returning tissues and organs to a normal, healthy state after injury or damage has occurred. To achieve this, progenitor cells such as pericytes and bone marrow-derived mesenchymal stem cells can be delivered exogenously, mobilised and recruited from within the body or transplanted in the form organs and tissues grown in the laboratory from stem cells. In this review, we summarise the recent evidence supporting the use of endogenously mobilised stem cell populations to enhance tissue repair along with the use of mesenchymal stem cells and pericytes in the development of engineered tissues. Finally, we conclude with an overview of currently available therapeutic options to manipulate endogenous stem cells to promote tissue repair.


Assuntos
Células-Tronco Mesenquimais/fisiologia , Pericitos/fisiologia , Regeneração , Engenharia Tecidual , Animais , Movimento Celular , Fibrose/patologia , Fibrose/terapia , Humanos , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/efeitos dos fármacos , Neovascularização Fisiológica , Pericitos/efeitos dos fármacos , Pericitos/transplante
4.
Adv Genet ; 89: 113-152, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25620010

RESUMO

DNA vectors have the potential to become powerful medical tools for treatment of human disease. The human body has, however, developed a range of defensive strategies to detect and silence foreign or misplaced DNA, which is more typically encountered during infection or chromosomal damage. A clinically relevant human gene therapy vector must overcome or avoid these protections whilst delivering sustained levels of therapeutic gene product without compromising the vitality of the recipient host. Many non-viral DNA vectors trigger these defense mechanisms and are subsequently destroyed or rendered silent. Thus, without modification or considered design, the clinical utility of a typical DNA vector is fundamentally limited due to the transient nature of its transgene expression. The development of safe and persistently expressing DNA vectors is a crucial prerequisite for its successful clinical application and subsequently remains, therefore, one of the main strategic tasks of non-viral gene therapy research. In this chapter we will describe our current understanding of the mechanisms that can destroy or silence DNA vectors and discuss strategies, which have been utilized to improve their sustenance and the level and duration of their transgene expression.


Assuntos
DNA/administração & dosagem , Expressão Gênica , Vetores Genéticos , Transgenes , Animais , Efeitos da Posição Cromossômica , DNA/imunologia , Epigênese Genética , Técnicas de Transferência de Genes , Vetores Genéticos/imunologia , Humanos , Nanopartículas/administração & dosagem
5.
Hepatology ; 60(3): 1035-43, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24585515

RESUMO

UNLABELLED: Host immune response to viral vectors, persistence of nonintegrating vectors, and sustained transgene expression are among the major challenges in gene therapy. To overcome these hurdles, we successfully used minicircle (MC) naked-DNA vectors devoid of any viral or bacterial sequences for the long-term treatment of murine phenylketonuria, a model for a genetic liver defect. MC-DNA vectors expressed the murine phenylalanine hydroxylase (Pah) complementary DNA (cDNA) from a liver-specific promoter coupled to a de novo designed hepatocyte-specific regulatory element, designated P3, which is a cluster of evolutionary conserved transcription factor binding sites. MC-DNA vectors were subsequently delivered to the liver by a single hydrodynamic tail vein (HTV) injection. The MC-DNA vector normalized blood phenylalanine concomitant with reversion of hypopigmentation in a dose-dependent manner for more than 1 year, whereas the corresponding parental plasmid did not result in any phenylalanine clearance. MC vectors persisted in an episomal state in the liver consistent with sustained transgene expression and hepatic PAH enzyme activity without any apparent adverse effects. Moreover, 14-20% of all hepatocytes expressed transgenic PAH, and the expression was observed exclusively in the liver and predominately around pericentral areas of the hepatic lobule, while there was no transgene expression in periportal areas. CONCLUSION: This study demonstrates that MC technology offers an improved safety profile and has the potential for the genetic treatment of liver diseases.


Assuntos
DNA Super-Helicoidal , Terapia Genética/métodos , Vetores Genéticos , Fígado/enzimologia , Fenilcetonúrias/terapia , Animais , Modelos Animais de Doenças , Feminino , Masculino , Camundongos Endogâmicos C57BL , Fenilalanina/sangue , Fenilalanina Hidroxilase/metabolismo , Regiões Promotoras Genéticas
6.
Mol Ther Nucleic Acids ; 2: e123, 2013 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-24045712

RESUMO

We previously identified short synthetic shRNAs (sshRNAs) that target a conserved hepatitis C virus (HCV) sequence within the internal ribosome entry site (IRES) of HCV and potently inhibit HCV IRES-linked gene expression. To assess in vivo liver delivery and activity, the HCV-directed sshRNA SG220 was formulated into lipid nanoparticles (LNP) and injected i.v. into mice whose livers supported stable HCV IRES-luciferase expression from a liver-specific promoter. After a single injection, RNase protection assays for the sshRNA and (3)H labeling of a lipid component of the nanoparticles showed efficient liver uptake of both components and long-lasting survival of a significant fraction of the sshRNA in the liver. In vivo imaging showed a dose-dependent inhibition of luciferase expression (>90% 1 day after injection of 2.5 mg/kg sshRNA) with t1/2 for recovery of about 3 weeks. These results demonstrate the ability of moderate levels of i.v.-injected, LNP-formulated sshRNAs to be taken up by liver hepatocytes at a level sufficient to substantially suppress gene expression. Suppression is rapid and durable, suggesting that sshRNAs may have promise as therapeutic agents for liver indications.Molecular Therapy-Nucleic Acids (2013) 2, e123; doi:10.1038/mtna.2013.50; published online 17 September 2013.

7.
Mol Ther Nucleic Acids ; 2: e115, 2013 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-23941867

RESUMO

The development of episomally maintained DNA vectors to genetically modify dividing cells efficiently and stably, without the risk of integration-mediated genotoxicity, should prove to be a valuable tool in genetic research. In this study, we demonstrate the utility of Scaffold/Matrix Attachment Region (S/MAR) DNA vectors to model the restoration of a functional wild-type copy of the gene folliculin (FLCN) implicated in the renal cancer Birt-Hogg-Dubé (BHD). Inactivation of FLCN has been shown to be involved in the development of sporadic renal neoplasia in BHD. S/MAR-modified BHD tumor cells (named UOK257-FS) show restored stable FLCN expression and have normalized downstream TGFß signals. We demonstrate that UOK257-FS cells show a reduced growth rate in vitro and suppression of xenograft tumor development in vivo, compared with the original FLCN-null UOK257 cell line. In addition, we demonstrate that mTOR signaling in serum-starved FLCN-restored cells is differentially regulated compared with the FLCN-deficient cell. The novel UOK257-FS cell line will be useful for studying the signaling pathways affected in BHD pathogenesis. Significantly, this study demonstrates the suitability of S/MAR vectors to successfully model the functional expression of a therapeutic gene in a cancer cell line and will aid the identification of novel cancer markers for diagnosis and therapy.Molecular Therapy-Nucleic Acids (2013) 2, e115; doi:10.1038/mtna.2013.40; published online 13 August 2013.

8.
Bioconjug Chem ; 24(3): 314-32, 2013 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-23305315

RESUMO

Recently, we reported for the first time the development of pH-triggered nanoparticles for the functional delivery of small interfering RNA (siRNA) to liver for treatment of hepatitis B virus infections in vivo. Here, we report on systematic formulation and biophysical studies of three different pH-triggered nanoparticle formulations looking for ways to improve on the capabilities of our previous nanoparticle system. We demonstrate how pH-triggered, PEGylated siRNA nanoparticles stable with respect to aggregation in 80% serum can still release siRNA payload at pH 5.5 within 30 min. This capability allows functional delivery to cultured murine hepatocyte cells in vitro, despite a high degree of PEGylation (5 mol %). We also demonstrate that pH-triggered, PEGylated siRNA nanoparticles typically enter cells by clathrin-coated pit endocytosis, but functional delivery requires membrane fusion events (fusogenicity). Biodistribution studies indicate that >70% of our administered nanoparticles are found in liver hepatocytes, post intravenous administration. Pharmacodynamic experiments show siRNA delivery to murine liver effecting maximum knockdown 48 h post administration from a single dose, while control (nontriggered) nanoparticles require 96 h and two doses to demonstrate the same effect. We also describe an anti-hepatitis C virus (HCV) proof-of-concept experiment indicating the possibility of RNAi therapy for HCV infections using pH-triggered, PEGylated siRNA nanoparticles.


Assuntos
Técnicas de Transferência de Genes , Hepatócitos/fisiologia , Nanopartículas/administração & dosagem , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/genética , Animais , Bovinos , Células Cultivadas , Feminino , Células HeLa , Células Hep G2 , Humanos , Concentração de Íons de Hidrogênio , Masculino , Camundongos , Nanopartículas/química , RNA Interferente Pequeno/química
9.
PLoS One ; 7(10): e47920, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23110132

RESUMO

The development of genetically marked animal tumour xenografts is an area of ongoing research to enable easier and more reliable testing of cancer therapies. Genetically marked tumour models have a number of advantages over conventional tumour models, including the easy longitudinal monitoring of therapies and the reduced number of animals needed for trials. Several different methods have been used in previous studies to mark tumours genetically, however all have limitations, such as genotoxicity and other artifacts related to the usage of integrating viral vectors. Recently, we have generated an episomally maintained plasmid DNA (pDNA) expression system based on Scaffold/Matrix Attachment Region (S/MAR), which permits long-term luciferase transgene expression in the mouse liver. Here we describe a further usage of this pDNA vector with the human Ubiquitin C promoter to create stably transfected human hepatoma (Huh7) and human Pancreatic Carcinoma (MIA-PaCa2) cell lines, which were delivered into "immune deficient" mice and monitored longitudinally over time using a bioluminometer. Both cell lines revealed sustained episomal long-term luciferase expression and formation of a tumour showing the pathological characteristics of hepatocellular carcinoma (HCC) and pancreatic carcinoma (PaCa), respectively. This is the first demonstration that a pDNA vector can confer sustained episomal luciferase transgene expression in various mouse tumour models and can thus be readily utilised to follow tumour formation without interfering with the cellular genome.


Assuntos
Linhagem Celular Tumoral , Regiões de Interação com a Matriz/genética , Plasmídeos/metabolismo , Transplante Heterólogo/métodos , Células Tumorais Cultivadas/metabolismo , Animais , Southern Blotting , Primers do DNA/genética , Vetores Genéticos/genética , Humanos , Imuno-Histoquímica , Estudos Longitudinais , Luciferases , Camundongos , Camundongos SCID , Plasmídeos/genética , Reação em Cadeia da Polimerase , Regiões Promotoras Genéticas/genética , Reação em Cadeia da Polimerase em Tempo Real , Ubiquitina C/genética
10.
Methods Mol Biol ; 891: 133-67, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22648771

RESUMO

Gene therapy vectors based on viruses are the most effective gene delivery systems in use today and although efficient at gene transfer their potential toxicity (Hacein-Bey-Abina et al., Science 302:415-419, 2003) provides impetus for the development of safer non-viral alternatives. An ideal vector for human gene therapy should deliver sustainable therapeutic levels of gene expression without affecting the viability of the host at either the cellular or somatic level. Vectors, which comprise entirely human elements, may provide the most suitable method of achieving this. Non-viral vectors are attractive alternatives to viral gene delivery systems because of their low toxicity, relatively easy production, and great versatility. The development of more efficient, economically prepared, and safer gene delivery vectors is a crucial prerequisite for their successful clinical application and remains a primary strategic task of gene therapy research.


Assuntos
Terapia Genética/métodos , Vetores Genéticos/biossíntese , Vetores Genéticos/genética , Cuidado Pré-Natal/métodos , Animais , Quitosana/administração & dosagem , DNA Nucleotidiltransferases/metabolismo , DNA Circular/administração & dosagem , DNA Circular/biossíntese , DNA Circular/genética , Endotoxinas/isolamento & purificação , Feto/metabolismo , Técnicas de Transferência de Genes , Humanos , Injeções , Camundongos , Especificidade de Órgãos , Plasmídeos/genética , Polietilenoimina/administração & dosagem , Vírus/metabolismo , Saco Vitelino/irrigação sanguínea
11.
Hum Gene Ther ; 22(8): 915-23, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21644838

RESUMO

Because of their high efficiency, virus-based vectors are currently used in most gene therapy trials. Because such vectors bear some potential safety risks, nonviral expression systems could be an attractive alternative. Ideally, these vectors should be completely based on chromosomal elements and replicate as an autonomous unit in the recipient cell, thus avoiding the risk of insertional mutagenesis or immunological reactions of the recipient organism. Our limited knowledge of the epigenetic regulation of replication in mammalian cells does not yet allow the rational design of such constructs. But in the late 1990s it was shown that scaffold/matrix attached region (S/MAR)-based vectors can promote episomal replication and maintenance in mammalian cells. These vectors have found broad application in basic research but are now improved for their use in the safe and reproducible genetic modification of cells and organisms and in gene therapy.


Assuntos
Técnicas de Transferência de Genes , Vetores Genéticos/química , Regiões de Interação com a Matriz , Animais , Replicação do DNA , Terapia Genética , Vetores Genéticos/metabolismo , Humanos , Mutagênese Insercional , Plasmídeos/genética , Transgenes
12.
Expert Opin Biol Ther ; 11(9): 1177-91, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21548848

RESUMO

INTRODUCTION: The early potential of gene therapy is slowly becoming realized following the recent treatment of patients with severe combined immunodeficiency and ocular diseases. However at present the field of gene therapy is tempered by the toxicity issues, mainly that of the integrated retroviral vector used in most trials which led to oncogenesis in several of the treated patients. The development of safer, alternative vectors is therefore vital for further progress in this field, in particular vectors which remain episomal and are therefore less genotoxic. One such unique class of vectors are those based on scaffold matrix attachment regions (S/MARs) elements, which are maintained extra-chromosomally and replicate in vitro and in vivo. AREAS COVERED: The overview here describes the most relevant studies utilizing the S/MAR element to episomally modify mammalian cells and tissues with a particular focus on liver tissue, as well as the brain, the muscle, the eye, cancer cells, embryonic cells and neonatal mice. For this purpose, recently published data in these areas (mainly articles published between 2000 and 2010) are reviewed. EXPERT OPINION: The utilisation of vectors harbouring an S/MAR element is an efficient, safe and cost-effective way to episomally modify mammalian cells.


Assuntos
Células/metabolismo , Terapia Genética , Vetores Genéticos/genética , Regiões de Interação com a Matriz/genética , Plasmídeos/metabolismo , Animais , Replicação do DNA , Expressão Gênica , Vetores Genéticos/uso terapêutico , Humanos , Camundongos
13.
J Mol Med (Berl) ; 89(5): 515-29, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21301798

RESUMO

We have previously described the development of a scaffold/matrix attachment region (S/MAR) episomal vector system for in vivo application and demonstrated its utility to sustain transgene expression in the mouse liver for at least 6 months following a single administration. Subsequently, we observed that transgene expression is sustained for the lifetime of the animal. The level of expression, however, does drop appreciably over time. We hypothesised that by eliminating the bacterial components in our vectors, we could improve their performance since bacterial sequences have been shown to be responsible for the immunotoxicity of the vector and the silencing of its expression when applied in vivo. We describe here the development of a minimally sized S/MAR vector, which is devoid of extraneous bacterial sequences. This minicircle vector comprises an expression cassette and an S/MAR moiety, providing higher and more sustained transgene expression for several months in the absence of selection, both in vitro and in vivo. In contrast to the expression of our original S/MAR plasmid vector, the novel S/MAR minicircle vectors mediate increased transgene expression, which becomes sustained at about twice the levels observed immediately after administration. These promising results demonstrate the utility of minimally sized S/MAR vectors for persistent, atoxic gene expression.


Assuntos
Vetores Genéticos/genética , Fígado/metabolismo , Transgenes/genética , Animais , Southern Blotting , Linhagem Celular Tumoral , Ensaio de Imunoadsorção Enzimática , Humanos , Camundongos , Reação em Cadeia da Polimerase
14.
J Control Release ; 150(3): 298-306, 2011 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-21192993

RESUMO

Non-viral vectors have not been extensively investigated in neonatal mice due to the poor efficiency of the delivery methods available. Understanding the effects of non-viral vectors during early development is vital to develop safe gene therapy treatments where irreversible pathological processes may be avoided by early gene reconstitution. Here we describe a simple and effective method for the systemic administration of non-viral vectors via the superior temporal vein of mouse pups at 1.5 days of age. We show that injection of polyethylenimine (PEI)-complexed plasmid DNA (pDNA) intravenously results in effective transfection in the liver, lung, heart, spleen, brain and kidney. We also investigate the specific targeting of transgene expression to the proliferating neonate liver using a liver-specific plasmid containing a Scaffold Matrix Attachment Region (S/MAR) element, which has previously been shown to confer long-term expression in adult mouse liver. Using bioluminescent imaging, a gradual increase in transgene expression was observed which peaked at days 11-12, before the reduction of expression to background levels by day 25, suggestive of vector copy number loss. We conclude that non-viral vectors can successfully be used for systemic delivery to neonatal mice and that this technique is likely to open a host of early therapeutic possibilities for gene transfer by a range of non-viral vector formulations.


Assuntos
DNA/administração & dosagem , Plasmídeos/administração & dosagem , Polietilenoimina/química , Transfecção , Animais , Encéfalo/metabolismo , DNA/química , Feminino , Expressão Gênica , Rim/metabolismo , Fígado/enzimologia , Fígado/metabolismo , Pulmão/metabolismo , Camundongos , Miocárdio/metabolismo , Plasmídeos/química , Baço/metabolismo , Transgenes
15.
BMC Biotechnol ; 10: 20, 2010 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-20230618

RESUMO

BACKGROUND: The episomal replication of the prototype vector pEPI-1 depends on a transcription unit starting from the constitutively expressed Cytomegalovirus immediate early promoter (CMV-IEP) and directed into a 2000 bp long matrix attachment region sequence (MARS) derived from the human beta-interferon gene. The original pEPI-1 vector contains two mammalian transcription units and a total of 305 CpG islands, which are located predominantly within the vector elements necessary for bacterial propagation and known to be counterproductive for persistent long-term transgene expression. RESULTS: Here, we report the development of a novel vector pEPito, which is derived from the pEPI-1 plasmid replicon but has considerably improved efficacy both in vitro and in vivo. The pEPito vector is significantly reduced in size, contains only one transcription unit and 60% less CpG motives in comparison to pEPI-1. It exhibits major advantages compared to the original pEPI-1 plasmid, including higher transgene expression levels and increased colony-forming efficiencies in vitro, as well as more persistent transgene expression profiles in vivo. The performance of pEPito-based vectors was further improved by replacing the CMV-IEP with the human CMV enhancer/human elongation factor 1 alpha promoter (hCMV/EF1P) element that is known to be less affected by epigenetic silencing events. CONCLUSIONS: The novel vector pEPito can be considered suitable as an improved vector for biotechnological applications in vitro and for non-viral gene delivery in vivo.


Assuntos
Vetores Genéticos/biossíntese , Plasmídeos/genética , Transfecção , Transgenes , Animais , Ilhas de CpG , Citomegalovirus/genética , Expressão Gênica , Humanos , Fígado/metabolismo , Masculino , Regiões de Interação com a Matriz , Camundongos , Células NIH 3T3 , Regiões Promotoras Genéticas , Replicon
16.
Curr Opin Mol Ther ; 11(4): 433-41, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19649988

RESUMO

The clinical application of gene therapy has become a reality with the treatment of patients with X-linked SCID (SCID-X1) using a modified retrovirus. This success has been tempered by the toxicity of the vector used in this trial, which led to oncogenesis in several of the treated patients. The development of safer, alternative vectors, which remain episomal and are therefore less genotoxic, is currently an area of active research. Notable recent developments include the application of modified lentiviral vectors, which stably express transgenes without the risk of integration; plasmid vectors, which exist episomally and are persistently expressed in the livers of mice; and the generation of replicating artificial chromosomes containing genomic loci. In addition, knowledge of the molecular mechanisms of nuclear retention and replication of the transgene is improving and will facilitate further developments in the use of episomal DNA for the genetic modification of cells. This review describes the development and application of gene therapy vectors, with a focus on those that are specifically designed to avoid integration and exist episomally.


Assuntos
Células/metabolismo , Plasmídeos/genética , Animais , Replicação do DNA , Vetores Genéticos/genética , Humanos , Regiões de Interação com a Matriz/genética
17.
Hum Gene Ther ; 19(10): 1050-6, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18847316

RESUMO

In vivo bioimaging of transgenic luciferase in the lung and nose is an expedient method by which to continually measure expression of this marker gene after gene transduction. Its substrate, luciferin, is typically injected into the peritoneal cavity before bioimaging. Here we demonstrate that, compared with intraperitoneal injection, intranasal instillation of luciferin confers approximately an order of magnitude increase in luciferase bioluminescence detection in both lung and nose. This effect was observed after administration of viral vectors based on adenovirus type 5, adeno-associated virus type 8, and gp64-pseudotyped HIV lentivirus and, to a lesser extent, after nonviral polyethylenimine (PEI)-DNA delivery. Detection increased relative to the concentration of luciferin; however, a standard concentration of 15 mg/ml was well beyond the saturation point. Compared with intraperitoneal injection, intranasal instillation yields about a 10-fold increase in sensitivity with an approximate 30-fold reduction in luciferin usage when bioimaging in the nasal and pulmonary airways of mice.


Assuntos
Adenoviridae , Dependovirus , Luciferina de Vaga-Lumes/farmacologia , Vetores Genéticos/farmacologia , Lentivirus , Luciferases , Substâncias Luminescentes/farmacologia , Transgenes , Administração Intranasal , Animais , Vetores Genéticos/genética , Vetores Genéticos/metabolismo , Imuno-Histoquímica , Injeções Intraperitoneais , Luminescência , Medições Luminescentes , Pulmão , Camundongos , Nariz , Imagem Corporal Total/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...