Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 177
Filtrar
1.
Retrovirology ; 19(1): 29, 2022 12 16.
Artigo em Inglês | MEDLINE | ID: mdl-36527061

RESUMO

BACKGROUND: Friend virus (FV) is a complex of the Friend murine leukemia virus (F-MuLV) and the replication-defective, pathogenic spleen focus forming virus (SFFV). In the past, we used a fluorescently labeled F-MuLV to analyze FV target cells. To build on these findings, we have now created a double-labeled FV that contains a Katushka-labeled F-MuLV and an mTagBFP-labeled SFFV, which we have used to study the infection by the two individual viruses in the FV infection of highly susceptible BALB/c mice. RESULTS: Our data show that the target cells of SFFV largely mirror those of F-MuLV, with the highest virus loads in erythroblasts, B cells and myeloid cells. The early phase of infection was dominated by cells infected by either SFFV or F-MuLV, whereas double-infected cells became dominant later in the course of infection with increasing viral loads. In the late phase of infection, the frequency of double-infected cells was similarly high as the frequencies of SFFV or F-MuLV single-infected cells, and single- and double-infected cells outnumbered the uninfected cells in the most highly infected cell populations such as erythroblasts. FV and retroviruses in general have been shown to induce interleukin 10 (IL-10) as a means of suppressing immune responses. Interestingly, we found in infected IL-10-eGFP reporter mice that SFFV-infected cells contributed to the IL-10-producing cell pool much more significantly than F-MuLV-infected cells, suggesting that the truncated SFFV envelope protein gp55 might play a role in IL-10 induction. Even though BALB/c mice mount notoriously weak immune responses against FV, infection of mice with an ablation of IL-10 expression in T cells showed transiently lower viral loads and stronger T cell activation, suggesting that IL-10 induction by FV and by SFFV in particular may contribute to a suppressed immune response in BALB/c mice. CONCLUSION: Our data provide detailed information about both F-MuLV- and SFFV-infected cells during the course of FV infection in highly susceptible mice and imply that the pathogenic SFFV contributes to immune suppression.


Assuntos
Vírus da Leucemia Murina de Friend , Leucemia Experimental , Camundongos , Animais , Vírus Formadores de Foco no Baço , Interleucina-10 , Baço , Camundongos Endogâmicos BALB C , Imunidade
2.
Sci Rep ; 10(1): 2121, 2020 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-32034258

RESUMO

We have previously designed a library of lentiviral vectors to generate somatic-transgenic rodents to monitor signalling pathways in diseased organs using whole-body bioluminescence imaging, in conscious, freely moving rodents. We have now expanded this technology to adeno-associated viral vectors. We first explored bio-distribution by assessing GFP expression after neonatal intravenous delivery of AAV8. We observed widespread gene expression in, central and peripheral nervous system, liver, kidney and skeletal muscle. Next, we selected a constitutive SFFV promoter and NFκB binding sequence for bioluminescence and biosensor evaluation. An intravenous injection of AAV8 containing firefly luciferase and eGFP under transcriptional control of either element resulted in strong and persistent widespread luciferase expression. A single dose of LPS-induced a 10-fold increase in luciferase expression in AAV8-NFκB mice and immunohistochemistry revealed GFP expression in cells of astrocytic and neuronal morphology. Importantly, whole-body bioluminescence persisted up to 240 days. We have validated a novel biosensor technology in an AAV system by using an NFκB response element and revealed its potential to monitor signalling pathway in a non-invasive manner in a model of LPS-induced inflammation. This technology complements existing germline-transgenic models and may be applicable to other rodent disease models.


Assuntos
Dependovirus/genética , Vetores Genéticos/genética , Camundongos Transgênicos/genética , Animais , Técnicas Biossensoriais/métodos , Expressão Gênica/genética , Proteínas de Fluorescência Verde/genética , Inflamação/genética , Luciferases de Vaga-Lume/genética , Camundongos , NF-kappa B/genética , Regiões Promotoras Genéticas/genética , Transdução de Sinais/genética , Vírus Formadores de Foco no Baço/genética , Transcrição Gênica/genética
3.
J Evol Biol ; 31(2): 314-322, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29266576

RESUMO

The virulence levels attained by serial passage of pathogens through similar host genotypes are much higher than observed in natural systems; however, it is unknown what keeps natural virulence levels below these empirically demonstrated maximum levels. One hypothesis suggests that host diversity impedes pathogen virulence, because adaptation to one host genotype carries trade-offs in the ability to replicate and cause disease in other host genotypes. To test this hypothesis, with the simplest level of population diversity within the loci of the major histocompatibility complex (MHC), we serially passaged Friend virus complex (FVC) through two rounds, in hosts with either the same MHC genotypes (pure passage) or hosts with different MHC genotypes (alternated passage). Alternated passages showed a significant overall reduction in viral titre (31%) and virulence (54%) when compared to pure passages. Furthermore, a resistant host genotype initially dominated any effects due to MHC diversity; however, when FVC was allowed to adapt to the resistant host genotype, predicted MHC effects emerged; that is, alternated lines show reduced virulence. These data indicate serial exposure to diverse MHC genotypes is an impediment to pathogen adaptation, suggesting genetic variation at MHC loci is important for limiting virulence in a rapidly evolving pathogen and supports negative frequency-dependent selection as a force maintaining MHC diversity in host populations.


Assuntos
Evolução Biológica , Vírus da Leucemia Murina de Friend/patogenicidade , Complexo Principal de Histocompatibilidade , Vírus Formadores de Foco no Baço/patogenicidade , Animais , Variação Genética , Camundongos , Camundongos Endogâmicos BALB C
4.
Mol Med Rep ; 16(3): 3041-3048, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28713922

RESUMO

Gene transfer into normal quiescent human B cells is a challenging procedure. The present study aimed to investigate whether it is possible to increase the levels of transgene expression by using various types of promoters to drive the expression of selected genes­of­interest. To produce lentiviral particles, the present study used the 2nd generation psPAX2 packaging vector and the vesicular stomatitis virus ­expressing envelope vector pMD2.G. Subsequently, lentiviral vectors were generated containing various promoters, including cytomegalovirus (CMV), elongation factor­1 alpha (EF1α) and spleen focus­forming virus (SFFV). The present study was unable to induce satisfactory transduction efficiency in quiescent normal B cells; however, infection of normal B cells with Epstein­Barr virus resulted in increased susceptibility to lentiviral transduction. In addition, the SFFV promoter resulted in a higher level of transgene expression compared with CMV or EF1α promoters. As a proof­of concept that this approach allows for stable gene expression in normal B cells, the present study used bicistronic lentiviral vectors with genes encoding fluorescent reporter proteins, as well as X­box binding protein­1 and binding immunoglobulin protein.


Assuntos
Linfócitos B/metabolismo , Técnicas de Transferência de Genes , Regiões Promotoras Genéticas , Adulto , Linfócitos B/virologia , Biomarcadores/metabolismo , Ligante de CD40/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Técnicas de Cocultura , Células Alimentadoras/metabolismo , Feminino , Fluorescência , Expressão Gênica , Células HEK293 , Herpesvirus Humano 4/fisiologia , Humanos , Sítios Internos de Entrada Ribossomal/genética , Lentivirus/genética , Masculino , Pessoa de Meia-Idade , Vírus Formadores de Foco no Baço/fisiologia , Transdução Genética , Transgenes
5.
Reprod Fertil Dev ; 29(3): 521-543, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28442062

RESUMO

Restricted availability of retinoic acid (RA) in the testicular milieu regulates transcriptional activity of c-kit (KIT, CD117), which aids in the determination of spermatogonial stem-cell differentiation. The effect of RA on c-kit has been reported previously, but its mode of genomic action remains unresolved. We studied the molecular machinery guiding RA responsiveness to the c-kit gene using spermatogonial stem-cell line C18-4 and primary spermatogonial cells. A novel retinoic acid response element (RARE) positioned at -989 nucleotides upstream of the transcription start site (TSS) was identified, providing a binding site for a dimeric RA receptor (i.e. retinoic acid receptor gamma (RARγ) and retinoic X receptor). RA treatment influenced c-kit promoter activity, along with endogenous c-kit expression in C18-4 cells. A comprehensive promoter deletion assay using the pGL3B reporter system characterised the region spanning -271bp and -1011bp upstream of the TSS, which function as minimal promoter and maximal promoter, respectively. In silico analysis predicted that the region -1011 to +58bp comprised the distal enhancer RARE and activators such as spleen focus forming virus proviral integration oncogene (SPFI1) (PU.1), specificity protein 1 (SP1) and four E26 transformation-specific (ETS) tandem binding sites at the proximal region. Gel retardation and chromatin immunoprecipitation (ChIP) assays showed binding for RARγ, PU.1 and SP1 to the predicted consensus binding sequences, whereas GABPα occupied only two out of four ETS binding sites within the c-kit promoter region. We propose that for RA response, an enhanceosome is orchestrated through scaffolding of a CREB-binding protein (CBP)/p300 molecule between RARE and elements in the proximal promoter region, controlling germ-line expression of the c-kit gene. This study outlines the fundamental role played by RARγ, along with other non-RAR transcription factors (PU.1, SP1 and GABPα), in the regulation of c-kit expression in spermatogonial stem cells in response to RA.


Assuntos
Células-Tronco Germinativas Adultas/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-kit/genética , Tretinoína/farmacologia , Células-Tronco Germinativas Adultas/metabolismo , Animais , Sítios de Ligação , Linhagem Celular , Expressão Gênica/efeitos dos fármacos , Humanos , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas c-ets/genética , Proteínas Proto-Oncogênicas c-ets/metabolismo , Proteínas Proto-Oncogênicas c-kit/metabolismo , Receptores do Ácido Retinoico/genética , Receptores do Ácido Retinoico/metabolismo , Vírus Formadores de Foco no Baço/genética , Vírus Formadores de Foco no Baço/metabolismo
6.
Stem Cell Reports ; 6(6): 873-884, 2016 06 14.
Artigo em Inglês | MEDLINE | ID: mdl-27161365

RESUMO

We previously reported the generation of integration-free induced pluripotent stem cells from adult peripheral blood (PB) with an improved episomal vector (EV) system, which uses the spleen focus-forming virus U3 promoter and an extra factor BCL-XL (B). Here we show an ∼100-fold increase in efficiency by optimizing the vector combination. The two most critical factors are: (1) equimolar expression of OCT4 (O) and SOX2 (S), by using a 2A linker; (2) a higher and gradual increase in the MYC (M) to KLF4 (K) ratio during the course of reprogramming, by using two individual vectors to express M and K instead of one. The combination of EV plasmids (OS + M + K + B) is comparable with Sendai virus in reprogramming efficiency but at a fraction of the cost. The generated iPSCs are indistinguishable from those from our previous approach in pluripotency and phenotype. This improvement lays the foundation for broad applications of episomal vectors in PB reprogramming.


Assuntos
Reprogramação Celular , Engenharia Genética/métodos , Vetores Genéticos/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Leucócitos Mononucleares/metabolismo , Plasmídeos/metabolismo , Adulto , Biomarcadores/metabolismo , Diferenciação Celular , Expressão Gênica , Engenharia Genética/economia , Vetores Genéticos/química , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Leucócitos Mononucleares/citologia , Fator 3 de Transcrição de Octâmero/genética , Fator 3 de Transcrição de Octâmero/metabolismo , Plasmídeos/química , Cultura Primária de Células , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo , Fatores de Transcrição SOXB1/genética , Fatores de Transcrição SOXB1/metabolismo , Vírus Formadores de Foco no Baço/genética , Vírus Formadores de Foco no Baço/metabolismo , Proteína bcl-X/genética , Proteína bcl-X/metabolismo
7.
Methods Mol Biol ; 1357: 57-69, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-25403468

RESUMO

Peripheral blood is the easy-to-access, minimally invasive, and the most abundant cell source to use for cell reprogramming. The episomal vector is among the best approaches for generating integration-free induced pluripotent stem (iPS) cells due to its simplicity and affordability. Here we describe the detailed protocol for the efficient generation of integration-free iPS cells from peripheral blood mononuclear cells. With this optimized protocol, one can readily generate hundreds of iPS cell colonies from 1 ml of peripheral blood.


Assuntos
Vetores Genéticos/genética , Células-Tronco Pluripotentes Induzidas/citologia , Leucócitos Mononucleares/citologia , Plasmídeos/genética , Técnicas de Cultura de Células/métodos , Diferenciação Celular , Células Cultivadas , Reprogramação Celular , Expressão Gênica , Vírus da Hepatite B da Marmota/genética , Humanos , Regiões Promotoras Genéticas/genética , Proteínas Recombinantes , Elementos de Resposta/genética , Vírus Formadores de Foco no Baço/genética , Transfecção , Proteína bcl-X/genética , Proteína bcl-X/fisiologia
8.
Proc Natl Acad Sci U S A ; 112(29): E3893-900, 2015 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-26150503

RESUMO

Substantial advances have been made in the past two decades in the management of osteoporosis. However, none of the current medications can eliminate the risk of fracture and rejuvenate the skeleton. To this end, we recently reported that transplantation of hematopoietic stem/progenitor cells (HSCs) or Sca1(+) cells engineered to overexpress FGF2 results in a significant increase in lamellar bone matrix formation at the endosteum; but this increase was attended by the development of secondary hyperparathyroidism and severe osteomalacia. Here we switch the therapeutic gene to PDGFB, another potent mitogen for mesenchymal stem cells (MSCs) but potentially safer than FGF2. We found that modest overexpression of PDGFB using a relatively weak phosphoglycerate kinase (PGK) promoter completely avoided osteomalacia and secondary hyperparathyroidism, and simultaneously increased trabecular bone formation and trabecular connectivity, and decreased cortical porosity. These effects led to a 45% increase in the bone strength. Transplantation of PGK-PDGFB-transduced Sca1(+) cells increased MSC proliferation, raising the possibility that PDGF-BB enhances expansion of MSC in the vicinity of the hematopoietic niche where the osteogenic milieu propels the differentiation of MSCs toward an osteogenic destination. Our therapy should have potential clinical applications for patients undergoing HSC transplantation, who are at high risk for osteoporosis and bone fractures after total body irradiation preconditioning. It could eventually have wider application once the therapy can be applied without the preconditioning.


Assuntos
Osso e Ossos/fisiopatologia , Terapia Genética , Transplante de Células-Tronco Hematopoéticas , Proteínas Proto-Oncogênicas c-sis/genética , Proteínas Proto-Oncogênicas c-sis/uso terapêutico , Fosfatase Alcalina/sangue , Animais , Antígenos Ly/metabolismo , Peso Corporal , Remodelação Óssea , Diferenciação Celular , Proliferação de Células , Hiperparatireoidismo/complicações , Hiperparatireoidismo/metabolismo , Hiperparatireoidismo/fisiopatologia , Hiperparatireoidismo/terapia , Antígeno Ki-67/metabolismo , Lentivirus/metabolismo , Proteínas de Membrana/metabolismo , Células-Tronco Mesenquimais/citologia , Camundongos , Modelos Biológicos , Neovascularização Fisiológica , Osteoblastos/metabolismo , Osteoblastos/patologia , Osteocalcina/sangue , Osteogênese , Osteomalacia/complicações , Osteomalacia/fisiopatologia , Fosfoglicerato Quinase/genética , Fosfoglicerato Quinase/metabolismo , Regiões Promotoras Genéticas/genética , Vírus Formadores de Foco no Baço/metabolismo , Transdução Genética , Transgenes , Suporte de Carga
9.
Curr Gene Ther ; 14(4): 276-88, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25039614

RESUMO

Lentiviral vectors (LVs) represent suitable candidates to mediate gene therapy for muscular dystrophies as they infect dividing and non-dividing cells and integrate their genetic material into the host genome, thereby theoretically mediating longterm expression. We evaluated the ability of LVs where a GFP reporter gene was under the control of five different promoters, to transduce and mediate expression in myogenic and non-myogenic cells in vitro and in skeletal muscle fibres and stem (satellite) cells in vivo. We further analysed lentivirally-transduced satellite cell-derived myoblasts following their transplantation into dystrophic, immunodeficient mouse muscles. The spleen focus-forming virus promoter mediated the highest gene expression in all cell types; the CBX3-HNRPA2B1 ubiquitously-acting chromatin opening element (UCOE) promoter was also active in all cells, whereas the human desmin promoter in isolation or fused with UCOE had lower activity in non-muscle cells. Surprisingly, the human skeletal muscle actin promoter was also active in immune cells. The human desmin promoter mediated robust, persistent reporter gene expression in myogenic cells in vitro, and satellite cells and muscle fibres in vivo. The human desmin promoter combined with UCOE did not significantly increase transgene expression. Therefore, our data indicate that the desmin promoter is suitable for the development of therapeutic purposes.


Assuntos
Desmina/genética , Terapia Genética/métodos , Vetores Genéticos , Células Satélites de Músculo Esquelético/fisiologia , Animais , Diferenciação Celular , Células Cultivadas , Proteínas Cromossômicas não Histona/genética , Regulação da Expressão Gênica , Humanos , Camundongos Endogâmicos mdx , Músculo Esquelético/citologia , Mioblastos Esqueléticos/citologia , Especificidade de Órgãos , Regiões Promotoras Genéticas , Vírus Formadores de Foco no Baço/genética , Transgenes
10.
Exp Hematol ; 42(9): 793-803.e1, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24746876

RESUMO

Mesenchymal stromal cells (MSCs) are a promising treatment modality for a variety of diseases. Strategies to investigate the fate of MSCs in vivo are important to unravel their therapeutic mechanisms. However, currently available techniques are hampered by their low sensitivity. We therefore aimed to optimize in vivo bioluminescence imaging of MSCs. We compared MSCs transduced with firefly luciferase (Fluc) and transmembrane-bound Gaussia luciferase driven by the human cytomegalovirus, spleen focus-forming virus (SFFV), and elongation factor 1-α (EF1α) promoters. Although cytomegalovirus-transmembrane-bound Gaussia luciferase-transduced MSCs showed the highest light intensity in vitro, the signal was almost undetectable in vivo. Spleen focus-forming virus-Fluc-transduced MSCs revealed a bright signal in vivo, but transgene expression was silenced upon in vitro stimulation with interferon (IFN)-γ. Therefore, the SFFV promoter was replaced by the EF1α promoter. Light emission of Fluc under the control of EF1α was similar to SFFV-Fluc. Although EF1α-Fluc light emission was decreased tenfold in the presence of IFN-γ when compared with unstimulated MSCs, the bioluminescent signal could still be detected and was clearly distinguishable from untransduced MSCs. Furthermore, stimulation of MSCs with tumor necrosis factor-α hardly affected transgene expression in EF1α-Fluc-transduced MSCs. Thus, the use of the EF1α promoter partially overcomes silencing and allows in vivo bioluminescence imaging of IFN-γ-stimulated MSCs.


Assuntos
Antivirais/farmacologia , Genes Reporter , Interferon gama/farmacologia , Luciferases de Vaga-Lume/biossíntese , Medições Luminescentes/métodos , Células-Tronco Mesenquimais/metabolismo , Fator 1 de Elongação de Peptídeos/genética , Regiões Promotoras Genéticas , Animais , Citomegalovirus/genética , Citomegalovirus/metabolismo , Humanos , Luciferases de Vaga-Lume/genética , Masculino , Células-Tronco Mesenquimais/citologia , Camundongos , Camundongos Endogâmicos BALB C , Vírus Formadores de Foco no Baço/genética , Vírus Formadores de Foco no Baço/metabolismo , Transdução Genética
11.
Rev. esp. geriatr. gerontol. (Ed. impr.) ; 49(1): 15-19, ene.-feb. 2014.
Artigo em Espanhol | IBECS | ID: ibc-118622

RESUMO

Introducción. En la enfermedad del Alzheimer (EA) se ha comprobado que existe un deterioro en la comunicación neuroinmunoendocrina. Sin embargo, apenas hay estudios a un nivel periférico en esta enfermedad neurodegenerativa, en concreto en lo que respecta a la función inmunitaria. Dado que recientemente se ha establecido que ciertos parámetros de función inmunitaria son marcadores de la velocidad de envejecimiento y pueden predecir la longevidad, el propósito del presente trabajo fue valorar algunas de esas funciones en leucocitos esplénicos de ratones transgénicos para la EA de diversas edades. Material y métodos. Se emplearon ratones triple-transgénicos para la EA (3 xTgAD) hembras, así como sus controles no transgénicos (NTg) jóvenes (4 ± 1 meses), adultos (9 ± 1 meses) y maduros (12 ± 1 meses). Se valoraron la quimiotaxis, la actividad citotóxica de las «natural killer» (NK) y la respuesta linfoproliferativa en presencia de los mitógenos concanavalina A y lipopolisacárido en leucocitos esplénicos, funciones que disminuyen al envejecer. Además, se realizó una curva de supervivencia en otro grupo de animales 3 xTgAD y NTg. Resultados. En los 3 xTgAD, con respecto a los NTg, la quimiotaxis se encuentra disminuida en todas las edades, mientras que dicha disminución se observa a los 4 y 9 meses en la linfoproliferación y solo en los jóvenes en el caso de la actividad NK. Los 3 xTgAD mostraron una menor supervivencia que los NTg. Conclusiones. Los ratones 3 xTgAD presentan una inmunosenescencia prematura, lo que podría explicar su temprana mortalidad. La valoración a nivel periférico de las funciones inmunológicas estudiadas podría ser un indicador del desarrollo de la enfermedad de Alzheimer (AU)


Introduction. A deterioration of the neuroimmunoendocrine network has been observed in Alzheimer's disease (AD). However, the peripheral immune response has hardly been investigated in this pathology. Since some immune function parameters have been established as good markers of the rate of ageing, and can predict longevity, the aim of the present work was to study some of these functions in splenic leucocytes in transgenic mice for AD of different ages. Material and methods. Young female (4 ± 1 months), adult (9 ± 1 months), and mature (12 ± 1 months) triple-transgenic mice for AD (3 xTgAD) and non-transgenic (NTg) control mice of the same ages were used. The chemotaxis, the anti-tumour activity of «natural killer» (NK) cells and the lymphoproliferative response in the presence of the mitogens concanavalin A and lipopolysaccharide, functions that decrease with age, were determined in splenic leucocytes. In addition, the differences in lifespan between 3 xTgAD and NTg were studied in parallel using other animals, until their death through natural causes. Results. In 3 xTgAD, with respect to NTg, chemotaxis decreased at all ages studied, whereas in lymphoproliferative response this reduction was shown at 4 months and 9 months. NK activity was diminished only in young 3 xTgAD with respect to NTg. The 3 xTgAD showed a shorter lifespan than the NTg control group. Conclusions. The 3 xTgAD mice show a premature immunosenescence, which could explain their early mortality. The determination of these immune functions at peripheral level could serve as a marker of the progression of the Alzheimer's disease (AU)


Assuntos
Humanos , Masculino , Feminino , Camundongos , Animais , Envelhecimento/imunologia , Doença de Alzheimer/epidemiologia , Doença de Alzheimer/imunologia , Doença de Alzheimer/prevenção & controle , Vírus Formadores de Foco no Baço/imunologia , Monitorização Imunológica , Imunoterapia Ativa/métodos , Imunoterapia Ativa , Quimiotaxia/imunologia , Quimiotaxia/fisiologia
12.
J Virol ; 87(24): 13760-74, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24109240

RESUMO

To assess the possible contribution of host immune responses to the exertion of Fv2-associated resistance to Friend virus (FV)-induced disease development, we inoculated C57BL/6 (B6) mice that lacked various subsets of lymphocytes with FV containing no lactate dehydrogenase-elevating virus. Fv2(r) B6 mice lacking CD4(+) T cells developed early polycythemia and fatal erythroleukemia, while B6 mice lacking CD8(+) T cells remained resistant. Erythroid progenitor cells infected with spleen focus-forming virus (SFFV) were eliminated, and no polycythemia was observed in B cell-deficient B6 mice, but they later developed myeloid leukemia associated with oligoclonal integration of ecotropic Friend murine leukemia virus. Additional depletion of natural killer and/or CD8(+) T cells from B cell-deficient B6 mice resulted in the expansion of SFFV proviruses and the development of polycythemia, indicating that SFFV-infected erythroid cells are not only restricted in their growth but are actively eliminated in Fv2(r) mice through cellular immune responses.


Assuntos
Vírus da Leucemia Murina de Friend/imunologia , Imunidade Celular , Imunidade Humoral , Leucemia Eritroblástica Aguda/veterinária , Doenças dos Roedores/imunologia , Animais , Linfócitos B/imunologia , Progressão da Doença , Resistência à Doença , Feminino , Vírus da Leucemia Murina de Friend/genética , Humanos , Leucemia Eritroblástica Aguda/imunologia , Leucemia Eritroblástica Aguda/virologia , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Doenças dos Roedores/virologia , Vírus Formadores de Foco no Baço/genética , Vírus Formadores de Foco no Baço/imunologia , Linfócitos T/imunologia
13.
J Virol ; 87(17): 9886-94, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23824803

RESUMO

Human cytomegalovirus (HCMV) lytic phase gene expression is repressed upon entry into myeloid lineage cells where the virus establishes latency. Lytic infection is not initiated because the tegument-delivered transactivator protein pp71 fails to enter the nucleus and inactivate the Daxx-mediated cellular intrinsic defense that silences the viral genome. When pp71 is expressed de novo in THP-1 monocytes, it localizes to the nucleus, inactivates the Daxx defense, and initiates lytic infection. We speculated that replacing the native viral promoter that drives pp71 expression with one that is highly and constitutively active in myeloid cells would permit pp71 de novo expression upon infection and that this newly expressed pp71 would accumulate in the nucleus, inactivate the intrinsic defense, and initiate the cascade of lytic gene expression. Surprisingly, we found that this promoter was still subject to normal silencing mechanisms in THP-1 monocytes and primary CD34(+) cells, two independent myeloid lineage cells. A second constitutively active heterologous viral promoter located in a different region of the HCMV genome was also silenced in THP-1 and CD34(+) cells. Furthermore, these two independent heterologous viral promoters inserted into three different regions of the HCMV genome in three different viral strains all required prior expression of the viral immediate early proteins for activation in fibroblasts. From this, we conclude that incorporation within the HCMV genome impacts the proclivity of heterologous viral promoters to initiate transcription. These observations have mechanistic implications for the expression of viral genes and transgenes during both lytic infection and latency.


Assuntos
Citomegalovirus/genética , Latência Viral/genética , Animais , Antígenos CD34/metabolismo , Linhagem Celular , Células Cultivadas , Citomegalovirus/fisiologia , Inativação Gênica , Genoma Viral , Interações Hospedeiro-Patógeno , Humanos , Proteínas Imediatamente Precoces/genética , Proteínas Imediatamente Precoces/metabolismo , Camundongos , Monócitos/virologia , Células Mieloides/imunologia , Células Mieloides/virologia , Regiões Promotoras Genéticas , Recombinação Genética , Vírus Formadores de Foco no Baço/genética , Vírus Formadores de Foco no Baço/fisiologia , Sequências Repetidas Terminais , Proteínas Virais/genética , Proteínas Virais/metabolismo , Latência Viral/fisiologia
14.
PLoS One ; 8(4): e62333, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23626802

RESUMO

Hematopoietic stem cell gene therapy requires the use of integrating retroviral vectors in order to stably transmit a therapeutic gene to mature blood cells. Human clinical trials have shown that some vector integration events lead to disrupted regulation of proto-oncogenes resulting in disordered hematopoiesis including T-cell leukemia. Newer vectors have been designed to decrease the incidence of these adverse events but require appropriate pre-clinical assays to demonstrate safety. We have used two distinct mouse serial transplant assays to evaluate the safety of a self-inactivating lentiviral vector intended for use in X-linked severe combined immunodeficiency (XSCID) gene therapy trials. These experiments entailed 28 months of total follow-up and included 386 mice. There were no cases in which the XSCID lentiviral vector clearly caused hematopoietic malignancies, although a single case of B cell malignancy was observed that contained the lentiviral vector as a likely passenger event. In contrast, a SFFV-DsRed γ-retroviral vector resulted in clonal transformation events in multiple secondary recipients. Non-specific pathology not related to vector insertions was noted including T cell leukemias arising from irradiated recipient cells. Overall, this comprehensive study of mouse transplant safety assays demonstrate the relative safety of the XSCID lentiviral vector but also highlight the limitations of these assays.


Assuntos
Transformação Celular Viral , Vetores Genéticos/efeitos adversos , Vetores Genéticos/genética , Transplante de Células-Tronco Hematopoéticas , Lentivirus/genética , Animais , Células da Medula Óssea/metabolismo , Células da Medula Óssea/patologia , Transformação Celular Neoplásica/genética , Feminino , Ordem dos Genes , Humanos , Linfoma de Células B/genética , Linfoma de Células B/virologia , Camundongos , Camundongos Knockout , Células Mieloides/metabolismo , Células Mieloides/patologia , Fator 1 de Elongação de Peptídeos/genética , Vírus Formadores de Foco no Baço/genética , Linfócitos T/metabolismo , Linfócitos T/patologia , Linfócitos T/virologia , Doenças por Imunodeficiência Combinada Ligada ao Cromossomo X/genética
15.
Hum Gene Ther Methods ; 23(6): 408-15, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23240650

RESUMO

Primary human B cells are an attractive target for gene-therapeutic applications, but have been found to be relatively resistant toward transduction with lentiviral vectors (LVVs), even though a number of different envelope pseudotypes were tested. Moreover, low transgene expression in primary human B cells has impeded the use of LVVs for this target cell. We investigated the transduction potential of gibbon-ape leukemia virus (GALV) Env-pseudotyped LVVs for primary human B cells. By establishing optimized transduction kinetics and multiplicities of infection, we were able to regularly obtain transduction efficiencies of more than 50% in CD40L-activated B cells. Noteworthy, with the use of GALV-pseudotyped LVVs we could achieve a more than 10-fold higher yield of transduced activated B cells in direct comparison with LVVs pseudotyped with measles virus glycoproteins. Phenotyping of transduced primary B cells revealed a majority of memory B cells, a long-lived phenotype, presumed to be well suited for enduring therapeutic interventions. Finally, by combining the enhancer (Eµ) and the matrix/scaffold-attachment regions (MARs) of the human immunoglobulin heavy chain with the promoter of spleen focus-forming virus (SFFV) we aimed at generating a novel LVV particularly suitable for B cell transgenesis. We show that the optimized vector facilitated significantly higher transgene expression in various B cell lines and, more importantly, primary human B cells (mean factor of three). In summary, we have established a novel protocol for the efficient lentiviral transduction of primary human B cells and have improved transgene expression in B cells by a specific vector modification.


Assuntos
Linfócitos B/metabolismo , Vetores Genéticos/genética , Vírus da Leucemia do Macaco Gibão/genética , Linfócitos B/citologia , Linfócitos B/imunologia , Antígenos CD40/metabolismo , Células Cultivadas , Vetores Genéticos/metabolismo , Células HEK293 , Humanos , Cadeias Pesadas de Imunoglobulinas/genética , Regiões de Interação com a Matriz/genética , Regiões Promotoras Genéticas , Vírus Formadores de Foco no Baço/genética , Transdução Genética
16.
J Immunol ; 189(5): 2521-9, 2012 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-22821964

RESUMO

The immune system is tasked with defending against a myriad of microbial infections, and its response to a given infectious microbe may be strongly influenced by coinfection with another microbe. It was shown that infection of mice with lactate dehydrogenase-elevating virus (LDV) impairs early adaptive immune responses to Friend virus (FV) coinfection. To investigate the mechanism of this impairment, we examined LDV-induced innate immune responses and found LDV-specific induction of IFN-α and IFN-γ. LDV-induced IFN-α had little effect on FV infection or immune responses, but unexpectedly, LDV-induced IFN-γ production dampened Th1 adaptive immune responses and enhanced FV infection. Two distinct effects were identified. First, LDV-induced IFN-γ signaling indirectly modulated FV-specific CD8+ T cell responses. Second, intrinsic IFN-γ signaling in B cells promoted polyclonal B cell activation and enhanced early FV infection, despite promotion of germinal center formation and neutralizing Ab production. Results from this model reveal that IFN-γ production can have detrimental effects on early adaptive immune responses and virus control.


Assuntos
Imunidade Adaptativa , Regulação para Baixo/imunologia , Interferon gama/fisiologia , Vírus da Leucemia Murina/imunologia , Infecções por Retroviridae/imunologia , Imunidade Adaptativa/genética , Animais , Modelos Animais de Doenças , Regulação para Baixo/genética , Feminino , Vírus da Leucemia Murina de Friend/imunologia , Vírus da Leucemia Murina de Friend/patogenicidade , Interferon gama/deficiência , Interferon gama/genética , Vírus Elevador do Lactato Desidrogenase/imunologia , Vírus Elevador do Lactato Desidrogenase/patogenicidade , Vírus da Leucemia Murina/patogenicidade , Leucemia Experimental/genética , Leucemia Experimental/imunologia , Leucemia Experimental/virologia , Camundongos , Camundongos Congênicos , Camundongos Endogâmicos A , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Mutantes , Infecções por Retroviridae/genética , Infecções por Retroviridae/patologia , Vírus Formadores de Foco no Baço/imunologia , Vírus Formadores de Foco no Baço/patogenicidade , Infecções Tumorais por Vírus/genética , Infecções Tumorais por Vírus/imunologia , Infecções Tumorais por Vírus/virologia
17.
Mol Ther ; 20(10): 1968-80, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22692499

RESUMO

Recombination activating gene 2 (RAG2) deficiency results in severe combined immunodeficiency (SCID) with complete lack of T and B lymphocytes. Initial gammaretroviral gene therapy trials for other types of SCID proved effective, but also revealed the necessity of safe vector design. We report the development of lentiviral vectors with the spleen focus forming virus (SF) promoter driving codon-optimized human RAG2 (RAG2co), which improved phenotype amelioration compared to native RAG2 in Rag2(-/-) mice. With the RAG2co therapeutic transgene, T-cell receptor (TCR) and immunoglobulin repertoire, T-cell mitogen responses, plasma immunoglobulin levels and T-cell dependent and independent specific antibody responses were restored. However, the thymus double positive T-cell population remained subnormal, possibly due to the SF virus derived element being sensitive to methylation/silencing in the thymus, which was prevented by replacing the SF promoter by the previously reported silencing resistant element (ubiquitous chromatin opening element (UCOE)), and also improved B-cell reconstitution to eventually near normal levels. Weak cellular promoters were effective in T-cell reconstitution, but deficient in B-cell reconstitution. We conclude that immune functions are corrected in Rag2(-/-) mice by genetic modification of stem cells using the UCOE driven codon-optimized RAG2, providing a valid optional vector for clinical implementation.


Assuntos
Proteínas de Ligação a DNA/genética , Terapia Genética/métodos , Vetores Genéticos/genética , Imunodeficiência Combinada Severa/genética , Imunodeficiência Combinada Severa/terapia , Animais , Linfócitos B/metabolismo , Proliferação de Células , Quimerismo , Cromatina , Códon/genética , Feminino , Dosagem de Genes , Rearranjo Gênico , Genes Codificadores da Cadeia beta de Receptores de Linfócitos T , Células-Tronco Hematopoéticas/metabolismo , Lentivirus/genética , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Camundongos SCID , Fenótipo , Plasmídeos , Regiões Promotoras Genéticas , Análise de Sequência de DNA , Baço/citologia , Baço/metabolismo , Vírus Formadores de Foco no Baço/genética , Linfócitos T/metabolismo , Transdução Genética , Transgenes
18.
Mol Ther ; 20(5): 1014-21, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22434137

RESUMO

Lentiviral vectors (LV) are widely used to stably transfer genes into target cells investigating or treating gene functions. In addition, gene transfer into early murine embryos may be improved to efficiently generate transgenic mice. We applied lentiviral gene transfer to generate a mouse model transgenic for SET binding protein-1 (Setbp1) and enhanced green fluorescent protein (eGFP). Neither transgenic founders nor their vector-positive offspring transcribed or expressed the transgenes. Bisulfite sequencing of the internal spleen focus-forming virus (SFFV) promoter demonstrated extensive methylation of all analyzed CpGs in the transgenic mice. To analyze the impact of Setbp1 on epigenetic silencing, embryonic stem cells (ESC) were differentiated into cardiomyocytes (CM) in vitro. In contrast to human promoters in LV, virally derived promoter sequences were strongly methylated during differentiation, independent of the transgene. Moreover, the commonly used SFFV promoter (SFFVp) was highly methylated with remarkable strength and frequency during hematopoietic differentiation in vivo in LV but less in γ-retroviral (γ-RV) backbones. In summary, we conclude that LV using an internal SFFVp are not suitable to generate transgenic mice or perform constitutive expression studies in differentiating cells. Choosing the appropriate promoter is also crucial to allow stable transgene expression in clinical gene therapy.


Assuntos
Proteínas de Transporte/genética , Vetores Genéticos , Lentivirus/genética , Camundongos Transgênicos/genética , Vírus Formadores de Foco no Baço/genética , Células-Tronco/metabolismo , Animais , Diferenciação Celular , Ilhas de CpG/genética , Metilação de DNA , Epigênese Genética , Efeito Fundador , Inativação Gênica , Genes Essenciais , Proteínas de Fluorescência Verde/genética , Humanos , Camundongos , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Regiões Promotoras Genéticas , Análise de Sequência de DNA , Células-Tronco/citologia , Transgenes
19.
Gene Ther ; 19(4): 396-403, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21900964

RESUMO

Gene transfer to the early-stage embryonic brain using the ultrasound image-guided gene delivery (UIGD) technique has proven to be valuable for investigating brain development. Thus far, this technology has been restricted to the study of embryonic neurogenesis. When this technique is designed to be employed for the study in adult animals, a long-term stable gene expression will be required. We attempted to develop a retroviral vector suitable for expressing exogenous genes in the brains of postnatal and adult mice in the context of the UIGD technique. Retroviral vectors containing four different long terminal repeats (LTRs) (each from Moloney murine leukemia virus (MoMLV), murine stem cell virus (MSCV), myeloproliferative sarcoma virus (MPSV) and spleen focus-forming virus (SFFV)) were compared using the well-known CE vector having the EF1α internal promoter as a control. The MS vector containing MSCV LTR produced a higher viral titer and a higher level of gene expression than other vectors including CE. The MS vector drove the gene expression in cultured neural stem cells for 3 weeks. Furthermore, the MS vector could efficiently deliver the gene to the mouse central nervous system, as transgene expression was found in various regions of the brains and spinal cords as well as in all major neural cell types. The data from an in vivo luciferase imaging analysis showed that the gene expression from the MS vector was sustainable for almost 3 months. Our data suggested that the MS vector would be suitable to construct mice containing the transgene expressed in the brain or spinal cord in a quick and cost-effective manner.


Assuntos
Encéfalo , Técnicas de Transferência de Genes , Vetores Genéticos , Retroviridae/genética , Ultrassonografia , Animais , Encéfalo/crescimento & desenvolvimento , Linhagem Celular , Expressão Gênica , Genes Reporter , Luciferases/genética , Masculino , Camundongos , Vírus da Leucemia Murina de Moloney/genética , Células-Tronco Neurais , Vírus Formadores de Foco no Baço/genética , Sequências Repetidas Terminais
20.
Int J Cancer ; 131(5): 1083-94, 2012 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-22034044

RESUMO

Infection of erythroid cells by Friend spleen focus-forming virus (SFFV) leads to acute erythroid hyperplasia in mice, due to expression of its unique envelope glycoprotein, gp55. Erythroid cells expressing SFFV gp55 proliferate in the absence of their normal regulator, erythropoietin, because of the interaction among the viral envelope protein, the erythropoietin receptor, and a short form of the receptor tyrosine kinase Stk (sf-Stk). This leads to constitutive activation of several signal transduction pathways. Our previous studies showed that sf-Stk interacts with SFFV gp55, forming disulfide-linked complexes. This covalent interaction, along with other noncovalent interactions with SFFV-gp55, results in constitutive tyrosine phosphorylation of sf-Stk and rodent fibroblast transformation. Here, we determined the precise amino acid region within sf-Stk that contributes to fibroblast transformation by the polycythemia-inducing (SFFV-P) and the anemia-inducing (SFFV-A) strains of SFFV. Sf-Stk deletion mutants showed different transforming abilities in fibroblasts infected with SFFV-P and SFFV-A, although the N-terminal extracellular domain of sf-Stk was essential for fibroblast transformation by both viruses. Point mutations of sf-Stk indicated that cysteine 19 was critical for fibroblast transformation by SFFV-P, although all four cysteines (8, 19, 37 and 42) appeared to be important for fibroblast transformation by both SFFV-P and SFFV-A. Mutation of sf-Stk cysteine 19 abolished its ability to form dimers with SFFV-P and SFFV-A gp55. These results suggest that the interaction between sf-Stk and the envelope proteins of the polycythemia- and anemia-inducing variants of SFFV is architecturally different.


Assuntos
Anemia/etiologia , Transformação Celular Neoplásica/patologia , Fibroblastos/patologia , Leucemia Experimental/genética , Policitemia/etiologia , Receptores Proteína Tirosina Quinases/metabolismo , Infecções por Retroviridae/genética , Vírus Formadores de Foco no Baço/genética , Infecções Tumorais por Vírus/genética , Sequência de Aminoácidos , Anemia/metabolismo , Anemia/patologia , Animais , Western Blotting , Transformação Celular Neoplásica/metabolismo , Células Cultivadas , Fibroblastos/metabolismo , Fibroblastos/virologia , Humanos , Leucemia Experimental/metabolismo , Leucemia Experimental/virologia , Camundongos , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Mutação/genética , Fosforilação , Plasmídeos/genética , Policitemia/metabolismo , Policitemia/patologia , Estrutura Terciária de Proteína , Receptores Proteína Tirosina Quinases/genética , Infecções por Retroviridae/metabolismo , Infecções por Retroviridae/virologia , Homologia de Sequência de Aminoácidos , Transdução de Sinais , Infecções Tumorais por Vírus/metabolismo , Infecções Tumorais por Vírus/virologia , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...