Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Vet Res ; 44: 17, 2013 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-23497101

RESUMO

Porcine reproductive and respiratory syndrome virus (PRRSV) is responsible for significant economic losses in the porcine industry. Currently available commercial vaccines do not allow optimal and safe protection. In this study, replicating but nondisseminating adenovectors (rAdV) were used for the first time in pigs for vaccinal purposes. They were expressing the PRRSV matrix M protein in fusion with either the envelope GP5 wild-type protein (M-GP5) which carries the major neutralizing antibody (NAb)-inducing epitope or a mutant form of GP5 (M-GP5m) developed to theoretically increase the NAb immune response. Three groups of fourteen piglets were immunized both intramuscularly and intranasally at 3-week intervals with rAdV expressing the green fluorescent protein (GFP, used as a negative control), M-GP5 or M-GP5m. Two additional groups of pigs were primed with M-GP5m-expressing rAdV followed by a boost with bacterially-expressed recombinant wild-type GP5 or were immunized twice with a PRRSV inactivated commercial vaccine. The results show that the rAdV expressing the fusion proteins of interest induced systemic and mucosal PRRSV GP5-specific antibody response as determined in an ELISA. Moreover the prime with M-GP5m-expressing rAdV and boost with recombinant GP5 showed the highest antibody response against GP5. Following PRRSV experimental challenge, pigs immunized twice with rAdV expressing either M-GP5 or M-GP5m developed partial protection as shown by a decrease in viremia overtime. The lowest viremia levels and/or percentages of macroscopic lung lesions were obtained in pigs immunized twice with either the rAdV expressing M-GP5m or the PRRSV inactivated commercial vaccine.


Assuntos
Anticorpos Antivirais/sangue , Síndrome Respiratória e Reprodutiva Suína/imunologia , Vírus da Síndrome Respiratória e Reprodutiva Suína/imunologia , Proteínas do Envelope Viral/genética , Proteínas da Matriz Viral/genética , Vacinas Virais/imunologia , Adenoviridae , Animais , Anticorpos Neutralizantes/sangue , Ensaio de Imunoadsorção Enzimática/veterinária , Síndrome Respiratória e Reprodutiva Suína/virologia , Suínos , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/imunologia , Proteínas do Envelope Viral/metabolismo , Proteínas da Matriz Viral/metabolismo , Vacinas Virais/administração & dosagem , Viremia/imunologia
2.
J Gene Med ; 14(12): 746-60, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23071006

RESUMO

BACKGROUND: Gutless adenovirus (helper-dependent adenoviral vector; HDAd) and lentiviral vectors (LV) are attractive vectors for the gene therapy of muscle diseases. Because the organization of their DNA (episomal versus integrated) differs, we investigated whether the strength and specificity of ΔUSEx3, a novel muscle-specific promoter previously tested with plasmid, were maintained in the context of these vectors. METHODS: Two HDAds expressing ß-galactosidase regulated by ΔUSEx3 or CAG [cytomegalovirus (CMV) enhancer/ß-actin promoter], and three LV expressing green fluorescent protein regulated by ΔUSEx3, CMV or a modified skeletal α-actin promoter (SPcΔ5-12), were constructed. Gene expression was compared in cell culture and after intravenous (HDAd only) and intramuscular injection of mice. RESULTS: Irrespective of the vector used, ΔUSEx3 remained poorly active in nonmuscle cells and tissues. In myotubes, ΔUSEx3 was as strong as CMV and SPcΔ5-12, although it was ten-fold weaker than CAG, a proven powerful promoter in muscle. In cell culture, ΔUSEx3 activity in the context of LV was more stable than CMV, indicating it is less prone to silencing. In the context of HDAd, the behavior of ΔUSEx3 in skeletal muscle mirrored that of cell culture (10% of the CAG activity and half the number of transduced fibers). Surprisingly, in muscles treated with LV, ΔUSEx3 activity was five-fold lower than SPcΔ5-12. CONCLUSIONS: The data obtained in the present study confirm that ΔUSEx3 is a strong and robust muscle-specific promoter in the context of HDAd (cell culture and in vivo) and LV (cell culture). However, it was less efficient in vivo in the context of LV.


Assuntos
Adenoviridae/genética , Vetores Genéticos/genética , Lentivirus/genética , Músculo Esquelético/metabolismo , Regiões Promotoras Genéticas , Troponina I/genética , Animais , Linhagem Celular , Citomegalovirus/genética , Elementos Facilitadores Genéticos , Expressão Gênica , Ordem dos Genes , Humanos , Camundongos , Especificidade de Órgãos/genética , Troponina I/metabolismo
3.
Apoptosis ; 16(3): 256-71, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21107701

RESUMO

We previously reported that HSV-2 R1, the R1 subunit (ICP10; UL39) of herpes simplex virus type-2 ribonucleotide reductase, protects cells against apoptosis induced by the death receptor (DR) ligands tumor necrosis factor-alpha- (TNFα) and Fas ligand (FasL) by interrupting DR-mediated signaling at, or upstream of, caspase-8 activation. Further investigation of the molecular mechanism underlying HSV-2 R1 protection showed that extracellular-regulated kinase 1/2 (ERK1/2), phosphatidylinositol 3-kinase (PI3-K)/Akt, NF-κB and JNK survival pathways do not play a major role in this antiapoptotic function. Interaction studies revealed that HSV-2 R1 interacted constitutively with caspase-8. The HSV-2 R1 deletion mutant R1(1-834)-GFP and Epstein-Barr virus (EBV) R1, which did not protect against apoptosis induced by DR ligands, did not interact with caspase-8, indicating that interaction is required for protection. HSV-2 R1 impaired caspase-8 activation induced by caspase-8 over-expression, suggesting that interaction between the two proteins prevents caspase-8 dimerization/activation. HSV-2 R1 bound to caspase-8 directly through its prodomain but did not interact with either its caspase domain or Fas-associated death domain protein (FADD). Interaction between HSV-2 R1 and caspase-8 disrupted FADD-caspase-8 binding. We further demonstrated that individually expressed HSV-1 R1 (ICP6) shares, with HSV-2 R1, the ability to bind caspase-8 and to protect cells against DR-induced apoptosis. Finally, as the long-lived Fas protein remained stable during the early period of infection, experiments with the HSV-1 UL39 deletion mutant ICP6∆ showed that HSV-1 R1 could be essential for the protection of HSV-1-infected cells against FasL.


Assuntos
Apoptose/efeitos dos fármacos , Caspase 8/metabolismo , Proteína Ligante Fas/farmacologia , Herpesvirus Humano 1/enzimologia , Herpesvirus Humano 2/enzimologia , Ribonucleotídeo Redutases/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Caspase 8/química , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Citoproteção/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Proteína de Domínio de Morte Associada a Fas/metabolismo , Deleção de Genes , Herpes Simples/metabolismo , Herpes Simples/virologia , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , NF-kappa B/metabolismo , Ligação Proteica/efeitos dos fármacos , Estrutura Terciária de Proteína , Subunidades Proteicas/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos
4.
Liver Int ; 31(9): 1306-14, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22093453

RESUMO

BACKGROUND: Epidemiological and laboratory evidences led to the hypothesis that molecular mimicry between viruses and self-proteins could be linked to the onset of autoimmune hepatitis (AIH). Hepatotropic viruses could be good candidates, as a pro-inflammatory environment may facilitate the development of AIH. AIMS: The aims of this study were to test a virus ability to induce an AIH through molecular mimicry and the influence of hepatic inflammation in this process. METHODS: C57BL/6 mice were injected i.v. or i.m. with recombinant adenoviral vectors (RecAdV) encoding for human type 2 AIH antigens to target xenoantigens expression in the liver and to create a transient hepatitis (i.v.) or for 'peripheral' xenoantigens expression (i.m.). Liver injury and B-cell response were evaluated. RESULTS: Late-onset hepatitis was observed 8 months after i.v. or i.m. RecAdV injections, despite presence or absence of an initial transient hepatitis. Intensity of B-cell response was similar for both type of injections, but the Ig isotypes produced were different. B-cell autoimmune response spread to several liver proteins. CONCLUSIONS: Liver autoimmune response can be initiated using molecular mimicry over a long period of time, validating the hit-and-run hypothesis. Initial liver inflammatory injury is neither necessary, nor detrimental to the development of AIH. These results highlight the significance of initial events on the pathogenesis of autoimmune liver injury.


Assuntos
Antígenos Heterófilos/imunologia , Hepatite Autoimune/imunologia , Fígado/imunologia , Mimetismo Molecular , Adenoviridae/genética , Amônia-Liases/genética , Amônia-Liases/imunologia , Animais , Antígenos Heterófilos/biossíntese , Antígenos Heterófilos/genética , Linfócitos B/imunologia , Linfócitos B/virologia , Antígeno CTLA-4/genética , Antígeno CTLA-4/imunologia , Citocromo P-450 CYP2D6/genética , Citocromo P-450 CYP2D6/imunologia , Feminino , Técnicas de Transferência de Genes , Vetores Genéticos , Glutamato Formimidoiltransferase/genética , Glutamato Formimidoiltransferase/imunologia , Hepatite Autoimune/patologia , Hepatite Autoimune/virologia , Humanos , Injeções Intramusculares , Injeções Intravenosas , Fígado/patologia , Fígado/virologia , Camundongos , Camundongos Endogâmicos C57BL , Enzimas Multifuncionais , Células Th1/imunologia , Células Th1/virologia , Células Th2/imunologia , Células Th2/virologia , Fatores de Tempo
5.
Appl Environ Microbiol ; 76(15): 5058-66, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20562288

RESUMO

A novel tightly regulated gene expression system was developed for Escherichia coli by applying the regulatory elements of the Pseudomonas putida F1 cym and cmt operons to control target gene expression at the transcriptional level by using p-isopropylbenzoate (cumate) as an inducer. This novel expression system, referred to as the cumate gene switch, includes a specific expression vector, pNEW, that contains a partial T5 phage promoter combined with the Pseudomonas-based synthetic operator and the cymR repressor protein-encoding gene designed to express constitutively in the host strain. The induction of transcription relies on the addition of the exogenous inducer (cumate), which is nontoxic to the culture, water soluble, and inexpensive. The characteristics and potential of the expression system were determined. Using flow cytometry and fed-batch fermentations, we have shown that, with the newly developed cumate-regulated system, (i) higher recombinant product yields can be obtained than with the pET (isopropyl-beta-D-thiogalactopyranoside [IPTG])-induced expression system, (ii) expression is tightly regulated, (iii) addition of cumate quickly results in a fully induced and homogenous protein-expressing population in contrast to the bimodal expression profile of an IPTG-induced population, (iv) expression can be modulated by varying the cumate concentration, and (v) the cumate-induced population remains induced and fully expressing even at 8 h following induction, resulting in high yields of the target protein Furthermore, the cumate gene switch described in this article is applicable to a wide range of E. coli strains.


Assuntos
Clonagem Molecular/métodos , Escherichia coli/genética , Regulação Bacteriana da Expressão Gênica/efeitos dos fármacos , Expressão Gênica , Engenharia Genética/métodos , Proteínas Recombinantes/biossíntese , Benzoatos/metabolismo , Vetores Genéticos , Pseudomonas , Pseudomonas putida/genética , Proteínas Recombinantes/genética , Ativação Transcricional
6.
Biotechnol Bioeng ; 106(2): 203-15, 2010 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-20178120

RESUMO

Fast and efficient production of recombinant proteins for structural and functional studies is a crucial issue for research and for industry. To this end, we have developed an efficient system to generate in less than 2 months, starting from the cDNA, pools of CHO cells stably expressing high-level of recombinant proteins. It is based on lentiviral vectors (LVs) for stable transduction coupled with the cumate gene-switch for inducible and efficient gene expression. Transcription is initiated upon binding of the cumate transactivator (cTA) or the reverse cTA (rcTA) to the CR5 promoter. Binding of cTA or rcTA is prevented or induced by addition of cumate respectively. We first validated the CHO/LV production system with an LV carrying the secreted alkaline phosphatase (SEAP), whose expression was linked to the green fluorescent protein (GFP) through an internal ribosome entry site (IRES). CHO cells stably expressing the cTA (CHO-cTA) were transduced at various multiplicity of infection (MOI). Pools of cells were incubated at 37 and 30 degrees C during 10 days. Optimal SEAP production (65 microg/mL) was achieved at 30 degrees C with a MOI of 200. The pool stability was demonstrated for 48 days of culture by GFP expression analysis. The system was also evaluated using LV expressing three typical therapeutic proteins (a protein made up of the extracellular domain of CD200 fused to IgG Fc region [CD200Fc], a chimeric antibody [chB43], and erythropoietin [EPO]). CHO cells expressing rcTA (CHO-Cum2) were transduced with these LVs at a MOI of 200 and production was tested at 30 degrees C. After 13 days of culture, 235, 160, and 206 microg/mL of CD200Fc, chB43, and EPO were produced, respectively. The ON/OFF ratio of these pools was equal to 6 for CD200Fc, 16 for chB43, and 74 for EPO. In conclusion, this system should be very useful to produce mg quantities of recombinant proteins in a timely manner in serum free suspension culture of CHO cells for preclinical studies.


Assuntos
Benzoatos/metabolismo , Células CHO/fisiologia , Vetores Genéticos/genética , Lentivirus/genética , Engenharia de Proteínas/métodos , Proteínas Recombinantes/metabolismo , Animais , Cricetinae , Cricetulus , Melhoramento Genético/métodos
7.
Protein Expr Purif ; 69(1): 1-8, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19635567

RESUMO

Islet Neogenesis Associated Protein (INGAP) is implicated in pancreatic islet neogenesis. INGAP peptide, a pentadecapeptide comprising amino acids 104-118, reverses diabetes in rodents and improves glucose homeostasis in patients with diabetes. The mechanism of INGAP action is unknown, but such studies would benefit from the availability of the full-length recombinant protein (rINGAP). Here we report the production of rINGAP from 293-SF cells following lentiviral transduction, and its characterization by MALDI-TOF and Q-TOF Mass Spectrometry, and HPLC. Importantly, we show that rINGAP exhibits 100x the bioactivity of INGAP peptide on a molar basis in an in vitro assay of human islet regeneration.


Assuntos
Antígenos de Neoplasias/biossíntese , Biomarcadores Tumorais/biossíntese , Lectinas Tipo C/biossíntese , Proteínas Recombinantes/biossíntese , Sequência de Aminoácidos , Animais , Antígenos de Neoplasias/química , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/isolamento & purificação , Biomarcadores Tumorais/química , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/isolamento & purificação , Células Cultivadas , Cromatografia Líquida de Alta Pressão , Cricetinae , Regulação da Expressão Gênica , Humanos , Ilhotas Pancreáticas/fisiologia , Lectinas Tipo C/química , Lectinas Tipo C/genética , Lectinas Tipo C/isolamento & purificação , Lentivirus/genética , Espectrometria de Massas , Mesocricetus , Dados de Sequência Molecular , Peso Molecular , Proteínas Associadas a Pancreatite , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/isolamento & purificação , Regeneração/fisiologia , Frações Subcelulares/metabolismo , Transdução Genética
8.
BMC Biotechnol ; 9: 42, 2009 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-19432976

RESUMO

BACKGROUND: Despite the powerful impact in recent years of gene expression markers like the green fluorescent protein (GFP) to link the expression of recombinant protein for selection of high producers, there is a strong incentive to develop rapid and efficient methods for isolating mammalian cell clones secreting high levels of marker-free recombinant proteins. Recently, a method combining cell colony growth in methylcellulose-based medium with detection by a fluorescently labeled secondary antibody or antigen has shown promise for the selection of Chinese Hamster Ovary (CHO) cell lines secreting recombinant antibodies. Here we report an extension of this method referred to as fluorescent labeling in semi-solid medium (FLSSM) to detect recombinant proteins significantly smaller than antibodies, such as IGF-E5, a 25 kDa insulin-like growth factor derivative. RESULTS: CHO cell clones, expressing 300 microg/ml IGF-E5 in batch culture, were isolated more easily and quickly compared to the classic limiting dilution method. The intensity of the detected fluorescent signal was found to be proportional to the amount of IGF-E5 secreted, thus allowing the highest producers in the population to be identified and picked. CHO clones producing up to 9.5 microg/ml of Tissue-Plasminogen Activator (tPA, 67 kDa) were also generated using FLSSM. In addition, IGF-E5 high-producers were isolated from 293SF transfectants, showing that cell selection in semi-solid medium is not limited to CHO and lymphoid cells. The best positive clones were collected with a micromanipulator as well as with an automated colony picker, thus demonstrating the method's high throughput potential. CONCLUSION: FLSSM allows rapid visualization of the high secretors from transfected pools prior to picking, thus eliminating the tedious task of screening a high number of cell isolates. Because of its rapidity and its simplicity, FLSSM is a versatile method for the screening of high producers for research and industry.


Assuntos
Técnicas de Cultura de Células/métodos , Meios de Cultura , Corantes Fluorescentes , Proteínas Recombinantes/biossíntese , Animais , Células CHO , Separação Celular , Cricetinae , Cricetulus , Fator de Crescimento Insulin-Like I/biossíntese , Ativador de Plasminogênio Tecidual/biossíntese
9.
Protein Expr Purif ; 64(2): 108-17, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19061959

RESUMO

Heterodimerizing peptides, such as the de novo designed E5/K5 peptide pair, have several applications including as tags for protein purification or immobilization. Recently, we demonstrated that E5-tagged epidermal growth factor (EGF), when bound to a K4 expressing adenovirus, promotes retargeting of the adenovirus to EGFR expressing target cells. In this study, we present the Escherichia coli expression, refolding and purification of human EGF fused with the E5-coil (E5-coil-EGF) or with the K5-coil (K5-coil-EGF). EGF receptor phosphorylation and cell proliferation assays demonstrated that the biological activity of the coil-tagged EGF versions was comparable to that of non-tagged EGF. Additionally, analysis of the binding of E5/K5-coil-EGF to cell surface EGFR or to soluble EGFR ectodomain, as measured by cell-based binding competition assays and by SPR-based biosensor experiments, indicated that the coil-tagged EGF versions bound to EGFR with affinities similar to that of non-tagged EGF. Finally, we show that E-coil-tagged EGF, but not non-tagged EGF, can retarget a K-coil containing adenovirus to EGF receptor expressing glioblastoma tumor cells. Overall these results indicate that E. coli expression offers a practical platform for the reproducible production of fully biologically active E5/K5-coil-tagged EGF, and support applications of heterodimerizing coil-tagged ligands, e.g. the targeting of viruses or other entities such as nanoparticles to tumor cells, or growth factor immobilization on cell culture scaffolds for tissue engineering.


Assuntos
Fator de Crescimento Epidérmico/genética , Fator de Crescimento Epidérmico/metabolismo , Escherichia coli/genética , Peptídeos/química , Linhagem Celular Tumoral , Células Cultivadas , Dimerização , Receptores ErbB/genética , Receptores ErbB/metabolismo , Escherichia coli/metabolismo , Expressão Gênica , Humanos , Corpos de Inclusão/metabolismo , Peptídeos/genética , Peptídeos/metabolismo , Fosforilação , Dobramento de Proteína , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Ressonância de Plasmônio de Superfície , Engenharia Tecidual , Tirosina/química , Tirosina/metabolismo
10.
Mol Ther ; 16(3): 500-7, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18180776

RESUMO

We have developed new packaging cell lines (293SF-PacLV) that can produce lentiviral vectors (LVs) in serum-free suspension cultures. A cell line derived from 293SF cells, expressing the repressor (CymR) of the cumate switch and the reverse transactivator (rtTA2(S)-M2) of the tetracycline (Tet) switch, was established first. We next generated clones stably expressing the Gag/Pol and Rev genes of human immunodeficiency virus-1, and the glycoprotein of vesicular stomatitis virus (VSV-G). Expression of Rev and VSV-G was tightly regulated by the cumate and Tet switches. Our best packaging cells produced up to 2.6 x 10(7) transducing units (TU)/ml after transfection with the transfer vector. Up to 3.4 x 10(7) TU/ml were obtained using stable producers generated by transducing the packaging cells with conditional-SIN-LV. The 293SF-PacLV was stable, as shown by the fact that some producers maintained high-level LV production for 18 weeks without selective pressure. The utility of the 293SF-PacLV for scaling up production in serum-free medium was demonstrated in suspension cultures and in a 3.5-L bioreactor. In shake flasks, the best packaging cells produced between 3.0 and 8.0 x 10(6) TU/ml/day for 3 days, and the best producer cells, between 1.0 and 3.4 x 10(7) TU/ml/day for 5 days. In the bioreactor, 2.8 liters containing 2.0 x 10(6) TU/ml was obtained after 3 days of batch culture following the transfection of packaging cells. In summary, the 293SF-PacLV possesses all the attributes necessary to become a valuable tool for scaling up LV production for preclinical and clinical applications.


Assuntos
Vetores Genéticos/biossíntese , Lentivirus/crescimento & desenvolvimento , Reatores Biológicos/virologia , Técnicas de Cultura de Células , Linhagem Celular , Meios de Cultura Livres de Soro/farmacologia , Proteínas de Fusão gag-pol/genética , Genes rev/genética , Vetores Genéticos/genética , Humanos , Lentivirus/efeitos dos fármacos , Lentivirus/genética , Modelos Genéticos , Plasmídeos/genética , Regiões Promotoras Genéticas/genética
12.
Cancer Res ; 67(7): 3387-95, 2007 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-17409449

RESUMO

Gene therapy is a promising approach for cancer treatment; however, efficacy of current vectors remains insufficient. To improve the success of suicide gene therapy, we constructed a replication-competent adenoviral vector that has its protease gene deleted and expresses bacterial cytosine deaminase fused with bacterial uracil phosphoribosyltransferase (CU). The prodrug, 5-fluorocytosine, is transformed into the highly toxic and tissue-diffusible 5-fluorouracil by CU in infected cells. This vector is incapable of producing infectious particles but is able to undergo a single round of replication, thereby increasing transgene copy number and expression. In the presence of 5-FC, compared with the first-generation vector (AdCU), the replication-competent vector, Ad(dPS)CU-IRES-E1A, was significantly more efficacious for in vitro tumor cell killing and in bystander assays, whereas 25-fold fewer viral particles were required in a three-dimensional spheroid model. For in vivo experiments, in which virus was injected into preestablished intracranial glioma xenografts, followed by 5-FC treatment, mice receiving Ad(dPS)CU-IRES-E1A had significantly smaller tumors at 35 days postinjection as well as significantly longer median survival than mice treated with the replication-deficient, protease-deleted vector [Ad(dPS)CU]. In an immunocompetent syngeneic model, Ad(dPS)CU + 5-FC-treated mice had a median survival of only 23 days, whereas Ad(dPS)CU-IRES-E1A + 5-FC-treated animals had a survival of 57.1% at 365 days. In conclusion, Ad(dPS)CU-IRES-E1A in the presence of 5-FC produces more potent tumoricidal effects than its replication-deficient counterparts.


Assuntos
Adenoviridae/genética , Terapia Genética/métodos , Glioblastoma/terapia , Peptídeo Hidrolases/deficiência , Adenoviridae/enzimologia , Adenoviridae/fisiologia , Animais , Linhagem Celular Tumoral , Citosina Desaminase/genética , Citosina Desaminase/metabolismo , Flucitosina/farmacocinética , Fluoruracila/farmacocinética , Fluoruracila/farmacologia , Amplificação de Genes , Vetores Genéticos/genética , Genoma Viral , Glioblastoma/genética , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Pentosiltransferases/genética , Pentosiltransferases/metabolismo , Peptídeo Hidrolases/genética , Esferoides Celulares , Transgenes , Replicação Viral
13.
J Biotechnol ; 296: 32-41, 2019 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-30885656

RESUMO

Chinese hamster ovary (CHO) cells are the most widely used mammalian host for industrial-scale production of monoclonal antibodies (mAbs) and other protein biologics. Isolation of rare high-producing CHO cell lines from heterogeneous populations of stable transfectants is a daunting task and delays the process of manufacturing of novel biologics. A variety of factors that contribute to the low frequency of high-producing clones have been described; however, the impact of metabolic burden and other stresses (eg. ER stress) associated with sustained high-level expression of recombinant protein (r-protein) during selection of stable transfectants has not been fully appreciated. CHO cell line development has not traditionally received much optimization in this area because the vast majority of platforms use constitutive expression systems to produce biologics. Previously, we developed a cell line (CHOBRI/rcTA) containing a robust inducible expression system, based on the cumate gene switch, that allows r-protein expression to be down-regulated during selection. Using this switch, we generated inducible CHOBRI/rcTA pools expressing an Fc-fusion protein within two weeks of transfection with volumetric productivity of up to 1.1 g/L at 17 days post-induction in a fed-batch culture process. Herein, we show that the ability to regulate r-protein expression during pool generation confers a substantial advantage for selecting high-producing stable clones. Reducing expression levels ("off-state") during pool selection dramatically enhances high-producer frequency compared to a pool in which expression was maintained at a high level during selection ("on-state", mimicking a constitutive expression system). Overexpression of the r-protein during the pool selection process negatively affects pool recovery and is associated with subtle but significant increases in BiP expression and cell death compared to pool selection in the "off-state". Our data shows that the cumate gene switch is a valuable platform for stable clone generation and supports the wider application of inducible systems for scalable production of biologics in CHO cells.


Assuntos
Anticorpos Monoclonais/biossíntese , Técnicas de Cultura Celular por Lotes/métodos , Fragmentos Fc das Imunoglobulinas/biossíntese , Proteínas Recombinantes/biossíntese , Animais , Anticorpos Monoclonais/genética , Anticorpos Monoclonais/imunologia , Células CHO , Cricetinae , Cricetulus , Regulação da Expressão Gênica/imunologia , Fragmentos Fc das Imunoglobulinas/genética , Fragmentos Fc das Imunoglobulinas/imunologia , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Estresse Fisiológico/genética , Transfecção
14.
Neuron ; 36(5): 815-29, 2002 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-12467586

RESUMO

Quaking viable (qk(v)) mice fail to properly compact myelin in their central nervous systems. Although the defect in the qk(v) mice involves a mutation affecting the expression of the alternatively spliced qk gene products, their roles in myelination are unknown. We show that the QKI RNA binding proteins regulate the nuclear export of MBP mRNAs. Disruption of the QKI nucleocytoplasmic equilibrium in oligodendrocytes results in nuclear and perikaryal retention of the MBP mRNAs and lack of export to cytoplasmic processes, as it occurs in qk(v) mice. MBP mRNA export defect leads to a reduction in the MBP levels and their improper cellular targeting to the periphery. Our findings suggest that QKI participates in myelination by regulating the mRNA export of key protein components.


Assuntos
Proteína Básica da Mielina/metabolismo , Isoformas de Proteínas/metabolismo , Proteínas de Ligação a RNA/metabolismo , Regiões 3' não Traduzidas/metabolismo , Transporte Ativo do Núcleo Celular/fisiologia , Processamento Alternativo , Animais , Sítios de Ligação , Encéfalo/citologia , Encéfalo/metabolismo , Células Cultivadas , Doenças Desmielinizantes/fisiopatologia , Éxons/genética , Humanos , Camundongos , Camundongos Quaking , Proteína Básica da Mielina/genética , Oligodendroglia/citologia , Oligodendroglia/fisiologia , Mutação Puntual , Ligação Proteica , Isoformas de Proteínas/genética , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/genética , Ratos , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo
15.
Lab Invest ; 88(7): 761-72, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18490899

RESUMO

Cultured human islets can be dedifferentiated to duct-like structures composed mainly of cytokeratin+ and nestin+ cells. Given that these structures possess the potential to redifferentiate into islet-like structures, we sought to elucidate their specific cellular origins. Adenoviral vectors were engineered for beta-, alpha-, delta- or PP-cell-specific GFP expression. A double-stranded system was designed whereby cultures were infected with two vectors: one expressed GFP behind the cumate-inducible promoter sequence, and the other expressed the requisite transactivator behind the human insulin, glucagon, somatostatin or pancreatic polypeptide promoter. This system labels hormone+ cells in the islet in a cell-specific manner, allowing these cells to be tracked during the course of transformation from islet to duct-like structure. Post-infection, islets were cultured to induce dedifferentiation. Fluorescence microscopy demonstrated that alpha-, delta- and PP-cells contributed equally to the cytokeratin+ population, with minimal beta-cell contribution, whereas the converse was true for nestin+ cells. Complementary targeted cell ablation studies, using streptozotocin or similar adenoviral expression of the Bax (Bcl2-associated X protein) toxigene, validated these findings and suggested a redundancy between alpha-, delta- and PP-cells with respect to cytokeratin+ cell derivation. These results call into question the traditional understanding of islet cells as being terminally differentiated and provide support for the concept of adult islet morphogenetic plasticity.


Assuntos
Ilhotas Pancreáticas/citologia , Adenoviridae/genética , Adulto , Diferenciação Celular , Células Cultivadas , Vetores Genéticos , Células Secretoras de Glucagon/citologia , Células Secretoras de Glucagon/metabolismo , Proteínas de Fluorescência Verde/biossíntese , Proteínas de Fluorescência Verde/genética , Humanos , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/metabolismo , Proteínas de Filamentos Intermediários/metabolismo , Ilhotas Pancreáticas/metabolismo , Queratinas/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Nestina , Ductos Pancreáticos/citologia , Células Secretoras de Polipeptídeo Pancreático/citologia , Células Secretoras de Polipeptídeo Pancreático/metabolismo , Regiões Promotoras Genéticas , Somatostatina/fisiologia , Células Secretoras de Somatostatina/citologia , Células Secretoras de Somatostatina/metabolismo , Células-Tronco/citologia , Estreptozocina/farmacologia , Proteína X Associada a bcl-2/biossíntese , Proteína X Associada a bcl-2/genética
16.
J Gene Med ; 10(4): 355-67, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18189335

RESUMO

BACKGROUND: Delivery of transgenes into specific tissues by adenovirus vectors (AdVs) relies on ablations of their natural tropism and on introduction of a new tropism. If the interaction with its natural receptor is ablated, a new packaging cell line is required to produce the AdV. In the present study, we have used two de novo designed peptides (E-Coil and K-Coil) that interact with each other with high affinity to establish a new receptor-ligand system for the propagation of retargeted AdVs. METHODS: We produced a cell line (293E) expressing on its surface a pseudoreceptor containing the E-Coil. An AdV (AdFK4m/GFP) lacking the interaction with the primary receptor for adenovirus (CAR) and containing the K-Coil inserted at the fiber C-terminus was constructed and tested using two strategies: (1) an RGD motif (Arg-Gly-Asp) was inserted into the HI-loop of the fiber; (2) AdFK4m/GFP was conjugated to a bispecific adaptor for the epidermal growth factor receptor (EGFR). RESULTS: AdFK4m/GFP infected 293E cells more efficiently than cells lacking the pseudoreceptor. The transduction was due to the K-Coil/E-Coil specific interaction since it was competed by addition of soluble K-Coil, but not soluble fiber. We demonstrated that the modified AdV was retargeted toward alpha v integrin by inclusion of the RGD motif, or toward EGFR using the bispecific adaptor. CONCLUSIONS: We have established a new system to produce AdVs ablated of natural tropism. This system should permit the retargeting of AdVs by inserting new ligands within the fiber or through the interaction with bispecific adaptors.


Assuntos
Adenoviridae/isolamento & purificação , Vetores Genéticos/isolamento & purificação , Peptídeos/metabolismo , Internalização do Vírus , Replicação Viral , Adenoviridae/genética , Adenoviridae/fisiologia , Motivos de Aminoácidos , Linhagem Celular , Proteína de Membrana Semelhante a Receptor de Coxsackie e Adenovirus , Vetores Genéticos/genética , Vetores Genéticos/fisiologia , Proteínas de Fluorescência Verde/genética , Humanos , Ligantes , Peptídeos/genética , Receptores Virais/metabolismo , Transdução Genética , Montagem de Vírus
17.
Cancer Res ; 66(21): 10576-85, 2006 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-17079482

RESUMO

When properly activated, macrophages can be tumoricidal, thus making them attractive additions to standard cancer therapies. To this end, tolerance and activity of human autologous IFN-gamma-activated macrophages, produced in large scale for clinical use (MAK cells), have been assessed in pilot trials in cancer patients. In the present study, we tested the hypothesis that activation of IFN regulatory factor (IRF)-3 and IRF-7, with subsequent type I IFN production, may be involved in the acquisition of new antitumor functions by macrophages. Adenoviral vectors were generated for the delivery of constitutively active forms of IRF-3 (Ad-IRF-3) or IRF-7 (Ad-IRF-7) into primary human macrophages. Cell death was observed in Ad-IRF-3-transduced macrophages, whereas Ad-IRF-7-transduced macrophages produced type I IFNs and displayed increased expression of genes encoding tumor necrosis factor (TNF)-related apoptosis-inducing ligand, interleukin (IL)-12, IL-15, and CD80, persisting for at least 96 hours. Expression of iNOS, TNF-alpha, FasL, IL-1, and IL-6 genes was unaltered by Ad-IRF-7 transduction. Interestingly, Ad-IRF-3 or Ad-IRF-7 transduction negatively regulated the transcription of protumorigenic genes encoding vascular endothelial growth factor and matrix metalloproteinase-2. Furthermore, Ad-IRF-7-transduced macrophages exerted a cytostatic activity on different cancer cell lines, including SK-BR-3, MCF-7, and COLO-205; the latter cells were shown previously to be insensitive to MAK cells. In conclusion, transduction of active forms of IRF-3 or IRF-7 differentially modulate the apoptotic and antitumor properties of primary macrophages, with active IRF-7 leading to the acquisition of novel antitumor effector functions.


Assuntos
Citotoxicidade Imunológica , Fator Regulador 3 de Interferon/fisiologia , Fator Regulador 7 de Interferon/fisiologia , Macrófagos/imunologia , Apoptose , Antígeno B7-1/genética , Linhagem Celular , Humanos , Fator Regulador 7 de Interferon/genética , Interferon Tipo I/genética , Interleucina-15/genética , Ativação de Macrófagos , RNA Mensageiro/análise , Ligante Indutor de Apoptose Relacionado a TNF/genética , Transdução Genética
18.
Sci Rep ; 8(1): 17361, 2018 11 26.
Artigo em Inglês | MEDLINE | ID: mdl-30478273

RESUMO

The insulin-like growth factor (IGF) axis has been implicated in the progression of malignant disease and identified as a clinically important therapeutic target. Several IGF-1 receptor (IGF-1R) targeting drugs including humanized monoclonal antibodies have advanced to phase II/III clinical trials, but to date, have not progressed to clinical use, due, at least in part, to interference with insulin receptor signalling. We previously reported on the production of a soluble fusion protein consisting of the extracellular domain of human IGF-1R fused to the Fc portion of human IgG1 (first generation IGF-TRAP) that bound human IGF-1 and IGF-2 with a 3 log higher affinity than insulin. We showed that the IGF-TRAP had potent anti-cancer activity in several pre-clinical models of aggressive carcinomas. Here we report on the re-engineering of the IGF-TRAP with the aim of improving physicochemical properties and suitability for clinical applications. We show that cysteine-serine substitutions in the Fc hinge region of IGF-TRAP eliminated high-molecular-weight oligomerized species, while a further addition of a flexible linker, not only improved the pharmacokinetic profile, but also enhanced the therapeutic profile of the IGF-TRAP, as evaluated in an experimental colon carcinoma metastasis model. Dose-response profiles of the modified IGF-TRAPs correlated with their bio-availability profiles, as measured by the IGF kinase-receptor-activation (KIRA) assay, providing a novel, surrogate biomarker for drug efficacy. This study provides a compelling example of structure-based re-engineering of Fc-fusion-based biologics for better manufacturability that also significantly improved pharmacological parameters. It identifies the re-engineered IGF-TRAP as a potent anti-cancer therapeutic.


Assuntos
Antineoplásicos/farmacologia , Carcinoma/tratamento farmacológico , Neoplasias do Colo/tratamento farmacológico , Fator de Crescimento Insulin-Like I/farmacologia , Proteínas Recombinantes de Fusão/farmacologia , Animais , Produtos Biológicos/farmacologia , Biomarcadores Tumorais/metabolismo , Carcinoma/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Neoplasias do Colo/metabolismo , Feminino , Células HEK293 , Humanos , Imunoglobulina G/metabolismo , Fator de Crescimento Insulin-Like II/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Receptor IGF Tipo 1/metabolismo
19.
J Neurosci ; 26(26): 7116-20, 2006 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-16807340

RESUMO

Haploinsufficency of Sim1, which codes for a basic helix-loop-helix-PAS (PER-ARNT-SIM) transcription factor, causes hyperphagia in mice and humans, without decrease in energy expenditure. Sim1 is expressed in several areas of the brain, including the developing and postnatal paraventricular nucleus (PVN), a region of the hypothalamus that controls food intake. We have previously found that the number of PVN cells is decreased in Sim1+/- mice, suggesting that their hyperphagia is caused by a developmental mechanism. However, the possibility that Sim1 functions in the postnatal PVN to control food intake cannot be ruled out. To explore this hypothesis, we used adenoviral vectors to modulate Sim1 expression in the postnatal PVN of wild-type mice. Unilateral stereotaxic injection into the PVN of an adenoviral vector producing a short hairpin RNA directed against Sim1 resulted in a significant increase in food intake, which peaked to 22% 6 d after the procedure, compared with the injection of a control virus. In contrast, injection of an adenovirus that expresses Sim1 induced a decrease in food intake that was maximal on the seventh day after the procedure, reaching 20%. The impact of bilateral injections of these vectors into the PVN was not greater than that of unilateral injections. Together, these results strongly suggest that Sim1 functions along a physiological pathway to control food intake.


Assuntos
Adenoviridae/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/fisiologia , Ingestão de Alimentos/fisiologia , Técnicas de Transferência de Genes , Vetores Genéticos , Núcleo Hipotalâmico Paraventricular/fisiologia , Proteínas Repressoras/fisiologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos , Núcleo Hipotalâmico Paraventricular/metabolismo , Interferência de RNA , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Fatores de Tempo
20.
J Biotechnol ; 255: 16-27, 2017 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-28625678

RESUMO

To rapidly produce large amounts of recombinant proteins, the generation of stable Chinese Hamster Ovary (CHO) cell pools represents a useful alternative to large-scale transient gene expression (TGE). We have developed a cell line (CHOBRI/rcTA) allowing the inducible expression of recombinant proteins, based on the cumate gene switch. After the identification of optimal plasmid DNA topology (supercoiled vs linearized plasmid) for PEIpro™ mediated transfection and of optimal conditions for methionine sulfoximine (MSX) selection, we were able to generate CHOBRI/rcTA pools producing high levels of recombinant proteins. Volumetric productivities of up to 900mg/L were reproducibly achieved for a Fc fusion protein and up to 350mg/L for an antibody after 14days post-induction in non-optimized fed-batch cultures. In addition, we show that CHO pool volumetric productivities are not affected by a freeze-thaw cycle or following maintenance in culture for over one month in the presence of MSX. Finally, we demonstrate that volumetric protein production with the CR5 cumate-inducible promoter is three- to four-fold higher than with the human CMV or hybrid EF1α-HTLV constitutive promoters. These results suggest that the cumate-inducible CHOBRI/rcTA stable pool platform is a powerful and robust system for the rapid production of gram amounts of recombinant proteins.


Assuntos
Plasmídeos/genética , Engenharia de Proteínas/métodos , Proteínas Recombinantes/metabolismo , Animais , Técnicas de Cultura Celular por Lotes , Células CHO , Cricetinae , Cricetulus , Expressão Gênica , Vetores Genéticos , Metionina Sulfoximina/farmacologia , Plasmídeos/metabolismo , Regiões Promotoras Genéticas , Proteínas Recombinantes/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA