Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Anim Biotechnol ; 34(4): 775-784, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32707002

RESUMO

Development of simple and readily adoptable methods to mediate germline engineering of the chicken genome will have many applications in research, agriculture and industrial biotechnology. We report germline targeting of the endogenous chicken Interferon Alpha and Beta Receptor Subunit 1 (IFNAR1) gene by in vivo transgenic expression of the high-fidelity Cas9 (Cas9-HF1) and guide RNAs (gRNAs) in chickens. First, we developed a Tol2 transposon vector carrying Cas9-HF1, IFNAR1-gRNAs (IF-gRNAs) and green fluorescent protein (GFP) transgenes (pTgRCG) and validated in chicken fibroblast DF1 cells. Next, the pTgRCG plasmid was directly injected into the dorsal aorta of embryonic day (ED) 2.5 chicken embryos targeting the circulating primordial germ cells (PGCs). The resulting chimera roosters generated a fully transgenic generation 1 (G1) hen with constitutive expression of Cas9-HF1 and IF-gRNAs (G1_Tol2-Cas9/IF-gRNA). We detected a spectrum of indels at gRNA-targeted loci in the G1_Tol2-Cas9/IF-gRNA hen and the indels were stably inherited by the G2 progeny. Breeding of the G1_Tol2-Cas9/IF-gRNA hen resulted in up to 10% transgene-free heterozygote IFNAR1 mutants, following null-segregation of the Tol2 insert. The method described here will provide new opportunities for genome editing in chicken and other avian species that lack PGC culture.


Assuntos
Sistemas CRISPR-Cas , Galinhas , Animais , Embrião de Galinha , Feminino , Masculino , Galinhas/genética , Sistemas CRISPR-Cas/genética , Transfecção , Animais Geneticamente Modificados/genética , Edição de Genes/métodos , Células Germinativas/metabolismo
2.
Anim Biotechnol ; 33(6): 1235-1245, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-33650465

RESUMO

Efficient isolation of genetically modified cells that are phenotypically indistinguishable from the unmodified cells remains a major technical barrier for the broader utilization of CRISPR/Cas9. Here, we report a novel enrichment approach to select the genome engineered cells by co-targeting a genomically integrated GFP gene along with the endogenous gene of interest (GOI). Using this co-targeting approach, multiple genomic loci were successfully targeted in chicken (DF1) and quail (CEC-32) fibroblast cell lines by transient transfection of Cas9 and guide RNAs (gRNAs). Clonal isolation of co-targeted DF1 cells showed 75% of cell clones had deletion of GFP and biallelic deletion of the GOI. To assess the utility of this approach to generate genome modified animals, we tested it on chicken primordial germ cells (PGCs) expressing GFP by co-targeting with gRNAs against GFP and endogenous ovomucoid (OVM) gene. PGCs enriched for loss of GFP and confirmed for OVM deletion, derived by co-targeting, were injected into Hamburger and Hamilton stage 14-15 chicken embryos, and their ability to migrate to the genital ridge was confirmed. This simple, efficient enrichment approach could easily be applied to the creation of knock-out or edited cell lines or animals.


Assuntos
Sistemas CRISPR-Cas , Edição de Genes , Embrião de Galinha , Animais , Sistemas CRISPR-Cas/genética , RNA Guia de Cinetoplastídeos/genética , Células Germinativas/metabolismo , Galinhas/genética , Linhagem Celular
3.
Microorganisms ; 9(1)2021 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-33450980

RESUMO

Marek's disease (MD), caused by MD herpesvirus (MDV), is an economically important disease in chickens. The efficacy of the existing vaccines against evolving virulent stains may become limited and necessitates the development of novel antiviral strategies to protect poultry from MDV strains with increased virulence. The CRISPR/Cas9 system has emerged as a powerful genome editing tool providing an opportunity to develop antiviral strategies for the control of MDV infection. Here, we characterized Tol2 transposon constructs encoding Cas9 and guide RNAs (gRNAs) specific to the immediate early infected-cell polypeptide-4 (ICP4) of MDV. We generated transgenic chickens that constitutively express Cas9 and ICP4-gRNAs (gICP4) and challenged them via intraabdominal injection of MDV-1 Woodlands strain passage-19 (p19). Transgenic chickens expressing both gRNA/Cas9 had a significantly reduced replication of MDV in comparison to either transgenic Cas9-only or the wild-type (WT) chickens. We further confirmed that the designed gRNAs exhibited sequence-specific virus interference in transgenic chicken embryo fibroblast (CEF) expressing Cas9/gICP4 when infected with MDV but not with herpesvirus of turkeys (HVT). These results suggest that CRISPR/Cas9 can be used as an antiviral approach to control MDV infection in chickens, allowing HVT to be used as a vector for recombinant vaccines.

4.
Transgenic Res ; 28(1): 51-76, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30374651

RESUMO

Zoonotic and foodborne diseases pose a significant burden, decreasing both human and animal health. Modifying chickens to overexpress antimicrobials has the potential to decrease bacterial growth on poultry products and boost chicken innate immunity. Chickens overexpressing either ovotransferrin or avian ß-defensin-3 (AvßD3) were generated using Tol-2 transposons. Transgene expression at the RNA and protein level was seen in egg white, breast muscle, and serum. There were significant differences in the immune cell populations in the blood, bursa, and spleen associated with transgene expression including an increased proportion of CD8+ cells in the blood of ovotransferrin and AvßD3 transgenic birds. Expression of the antimicrobials inhibited the in vitro growth of human and chicken bacterial pathogens and spoilage bacteria. For example, transgene expression significantly reduced growth of aerobic and coliform bacteria in breast muscle and decreased the growth of Salmonella enterica in egg white. Overall these results indicate that overexpression of antimicrobials in the chicken can impact the immune system and increase the antimicrobial capacity of poultry products.


Assuntos
Animais Geneticamente Modificados/genética , Conalbumina/genética , Imunidade Inata/genética , beta-Defensinas/genética , Animais , Animais Geneticamente Modificados/microbiologia , Anti-Infecciosos/sangue , Galinhas/sangue , Galinhas/genética , Conalbumina/sangue , Conalbumina/imunologia , Elementos de DNA Transponíveis/genética , Clara de Ovo/química , Regulação da Expressão Gênica/genética , Humanos , Músculos/metabolismo , Produtos Avícolas/microbiologia , beta-Defensinas/sangue , beta-Defensinas/imunologia
5.
Transgenic Res ; 26(3): 331-347, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-27896535

RESUMO

Generating transgenic and gene edited mammals involves in vitro manipulation of oocytes or single cell embryos. Due to the comparative inaccessibility of avian oocytes and single cell embryos, novel protocols have been developed to produce transgenic and gene edited birds. While these protocols are relatively efficient, they involve two generation intervals before reaching complete somatic and germline expressing transgenic or gene edited birds. Most of this work has been done with chickens, and many protocols require in vitro culturing of primordial germ cells (PGCs). However, for many other bird species no methodology for long term culture of PGCs exists. Developing methodologies to produce germline transgenic or gene edited birds in the first generation would save significant amounts of time and resource. Furthermore, developing protocols that can be readily adapted to a wide variety of avian species would open up new research opportunities. Here we report a method using sperm as a delivery mechanism for gene editing vectors which we call sperm transfection assisted gene editing (STAGE). We have successfully used this method to generate GFP knockout embryos and chickens, as well as generate embryos with mutations in the doublesex and mab-3 related transcription factor 1 (DMRT1) gene using the CRISPR/Cas9 system. The efficiency of the method varies from as low as 0% to as high as 26% with multiple factors such as CRISPR guide efficiency and mRNA stability likely impacting the outcome. This straightforward methodology could simplify gene editing in many bird species including those for which no methodology currently exists.


Assuntos
Animais Geneticamente Modificados , Edição de Genes/métodos , Espermatozoides/fisiologia , Transfecção/métodos , Animais , Embrião de Galinha , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Feminino , Proteínas de Fluorescência Verde/genética , Inseminação Artificial , Masculino , RNA Guia de Cinetoplastídeos , Fatores de Transcrição/genética
6.
Endocrinology ; 157(1): 83-90, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26556534

RESUMO

Estrogens play a key role in sexual differentiation of both the gonads and external traits in birds. The production of estrogen occurs via a well-characterized steroidogenic pathway, which is a multistep process involving several enzymes, including cytochrome P450 aromatase. In chicken embryos, the aromatase gene (CYP19A1) is expressed female-specifically from the time of gonadal sex differentiation. Ectopic overexpression of aromatase in male chicken embryos induces gonadal sex reversal, and male embryos treated with estradiol become feminized; however, this is not permanent. To test whether a continuous supply of estrogen in adult chickens could induce stable male to female sex reversal, 2 transgenic male chickens overexpressing aromatase were generated using the Tol2/transposase system. These birds had robust ectopic aromatase expression, which resulted in the production of high serum levels of estradiol. Transgenic males had female-like wattle and comb growth and feathering, but they retained male weights, displayed leg spurs, and developed testes. Despite the small sample size, this data strongly suggests that high levels of circulating estrogen are insufficient to maintain a female gonadal phenotype in adult birds. Previous observations of gynandromorph birds and embryos with mixed sex chimeric gonads have highlighted the role of cell autonomous sex identity in chickens. This might imply that in the study described here, direct genetic effects of the male chromosomes largely prevailed over the hormonal profile of the aromatase transgenic birds. This data therefore support the emerging view of at least partial cell autonomous sex development in birds. However, a larger study will confirm this intriguing observation.


Assuntos
Animais Geneticamente Modificados/metabolismo , Aromatase/metabolismo , Proteínas Aviárias/metabolismo , Galinhas/metabolismo , Estrogênios/sangue , Feminização/veterinária , Regulação para Cima , Animais , Animais Geneticamente Modificados/sangue , Animais Geneticamente Modificados/genética , Aromatase/genética , Proteínas Aviárias/genética , Doenças das Aves/sangue , Doenças das Aves/metabolismo , Doenças das Aves/patologia , Doenças das Aves/fisiopatologia , Galinhas/sangue , Galinhas/genética , Galinhas/crescimento & desenvolvimento , Estrogênios/metabolismo , Feminino , Feminização/metabolismo , Feminização/patologia , Feminização/fisiopatologia , Masculino , Microscopia de Fluorescência/veterinária , Tamanho do Órgão , Ovário/crescimento & desenvolvimento , Ovário/metabolismo , Ovário/patologia , Índice de Gravidade de Doença , Maturidade Sexual , Testículo/crescimento & desenvolvimento , Testículo/metabolismo , Testículo/patologia , Aumento de Peso
7.
Transgenic Res ; 22(6): 1257-64, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23807321

RESUMO

Traditional methods of avian transgenesis involve complex manipulations involving either retroviral infection of blastoderms or the ex vivo manipulation of primordial germ cells (PGCs) followed by injection of the cells back into a recipient embryo. Unlike in mammalian systems, avian embryonic PGCs undergo a migration through the vasculature on their path to the gonad where they become the sperm or ova producing cells. In a development which simplifies the procedure of creating transgenic chickens we have shown that PGCs are directly transfectable in vivo using commonly available transfection reagents. We used Lipofectamine 2000 complexed with Tol2 transposon and transposase plasmids to stably transform PGCs in vivo generating transgenic offspring that express a reporter gene carried in the transposon. The process has been shown to be highly effective and as robust as the other methods used to create germ-line transgenic chickens while substantially reducing time, infrastructure and reagents required. The method described here defines a simple direct approach for transgenic chicken production, allowing researchers without extensive PGC culturing facilities or skills with retroviruses to produce transgenic chickens for wide-ranging applications in research, biotechnology and agriculture.


Assuntos
Galinhas/genética , Elementos de DNA Transponíveis/genética , Técnicas de Transferência de Genes , Células Germinativas , Animais , Animais Geneticamente Modificados , Lipídeos/genética , Plasmídeos , Transfecção/métodos
8.
J Interferon Cytokine Res ; 28(7): 435-44, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18597621

RESUMO

Increasing resistance to anthelmintic drugs indicates a vital need to develop alternative strategies to control helminth infections. Interleukin-3 (IL-3) is a multilineage hematopoietic growth regulator produced by activated T lymphocytes in response to infection. In helminth infections, eosinophils play an important role in the elimination of parasites through their recruitment of inflammatory cells and the release of granules. The ability of IL-3 to stimulate the development of eosinophils makes it a particularly important candidate for therapeutic use to protect against parasites. To enable the role of IL-3 in the development, growth, and differentiation of porcine eosinophils to be elucidated, recombinant IL-3 (rPoIL-3) was expressed and purified. As the amino acid sequence identities between porcine IL-3 and other reported species were quite low ( approximately 39% between human and pig), an assessment of the in vitro activity of rPoIL-3 was made. The culture of porcine bone marrow (BM) cells with rPoIL-3 stimulated the proliferation of SWC3a(hi) myeloid cells, conA rming that rPoIL-3 acted as a hematopoietic cell growth factor. Since rPoIL-3 stimulated the development of myeloid cells in culture, the in vivo potential to produce elevated eosinophil proportions was assessed. In vivo administration of rPoIL-3 induced a signiA cant increase in the number of eosinophils in blood. These results suggest that rPoIL-3 is a potent inducer of eosinophils in swine and supports the inclusion of rPoIL-3 in therapeutic strategies.


Assuntos
Células da Medula Óssea/efeitos dos fármacos , Eosinófilos/citologia , Interleucina-3/administração & dosagem , Proteínas Recombinantes/administração & dosagem , Animais , Células da Medula Óssea/citologia , Células da Medula Óssea/imunologia , Bovinos , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Eosinófilos/efeitos dos fármacos , Eosinófilos/imunologia , Humanos , Interleucina-3/imunologia , Contagem de Leucócitos , Camundongos , Modelos Biológicos , Conformação Molecular , Doenças Parasitárias/sangue , Doenças Parasitárias/tratamento farmacológico , Proteínas Recombinantes/imunologia , Análise de Sequência de Proteína , Ovinos , Suínos
9.
J Interferon Cytokine Res ; 28(6): 341-50, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18593329

RESUMO

Interferons (IFN) provide a critical first line of defense against viral infection in vertebrates. Moreover, IFN-lambda, a recently identified group of mammalian IFN, has demonstrated antiviral potential in the treatment of mammalian viruses. With the growing concern over such diseases as avian influenza (AI), there is a pressing need for new antiviral strategies to manage problem viruses in poultry. Furthermore, the use of immune molecules, such as IFN-lambda, provides an attractive option for treating poultry by augmenting the host response to virus. With this in mind, we report here the first cloning, expression, and analysis of biologic activity of chicken IFN-lambda (ChIFN-lambda). We compared the similarity of ChIFN-lambda to those identified in other species and demonstrate that ChIFN-lambda has antiviral properties similar to those of human IFN-lambda (HuIFN-lambda). Our results demonstrate that in the chicken, as in human, the antiviral activity demonstrated by ChIFN-lambda supports its inclusion in therapeutic strategies directed against viral infections.


Assuntos
Interleucinas/genética , Proteínas Recombinantes/genética , Animais , Antivirais/imunologia , Antivirais/farmacologia , Linhagem Celular , Embrião de Galinha , Galinhas , Clonagem Molecular , Escherichia coli , Humanos , Imunoterapia Ativa , Vírus da Influenza A/efeitos dos fármacos , Vírus da Influenza A/imunologia , Influenza Aviária/imunologia , Influenza Aviária/terapia , Interleucinas/imunologia , Macrófagos/imunologia , Macrófagos/metabolismo , Proteínas Recombinantes/imunologia , Alinhamento de Sequência , Receptor 3 Toll-Like/biossíntese , Receptor 3 Toll-Like/imunologia
10.
J Interferon Cytokine Res ; 27(11): 937-46, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18052728

RESUMO

Escherichia coli infection of the respiratory system in chickens occurs as a sequel to a variety of environmental stressors or microbial infections, culminating as chronic respiratory disease (CRD) syndrome or colibacillosis. These diseases cause significant production losses in poultry. With the growing concerns about the use of antibiotics in animal production, for diseases such as CRD, alternative natural agents, like cytokines, may be considered for enhancing health by stimulating the immune system. The current study was aimed at understanding the in vivo effects of recombinant chicken interferon-gamma (ChIFN-gamma) treatment on a variety of immunologic parameters during E. coli infection in chickens. Administration of ChIFN-gamma to chickens increased the percentage of phagocytes in lung and blood of E. coli-infected birds. At the phenotypic level, there was an increase in the percentage of cells expressing MHC II in the air sac, with a concomitant reduction in the proportion of these cells in blood. Furthermore, the blood plasma from ChIFN-gamma-treated infected birds showed an increased level of interleukin-6 (IL-6) activity. Cumulatively, these findings are indicative of in vivo enhancement of immune responses due to ChIFN-gamma. However, administration of ChIFN-gamma protein did not mitigate the development of air sac lesions following E. coli infection.


Assuntos
Infecções por Escherichia coli/veterinária , Antígenos de Histocompatibilidade Classe II/sangue , Interferon gama/uso terapêutico , Interleucina-6/sangue , Doenças das Aves Domésticas/tratamento farmacológico , Sacos Aéreos/imunologia , Animais , Galinhas , Infecções por Escherichia coli/tratamento farmacológico , Infecções por Escherichia coli/imunologia , Leucócitos/imunologia , Pulmão/imunologia , Fagocitose , Doenças das Aves Domésticas/imunologia , Proteínas Recombinantes
11.
Exp Hematol ; 35(1): 171-8, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17198886

RESUMO

OBJECTIVES: The isolation of porcine hematopoietic stem cells (HSC) would be an important step toward development of porcine-to-human chimerism for induction of tolerance in clinical xenotransplantation. CD34 is a common marker of HSC and has not been developed as a marker in pigs. In this study we have generated and characterized a monoclonal antibody (mAb) that identifies porcine CD34 on a subset of porcine bone marrow (BM) stem/progenitor cells. METHODS: The porcine CD34 gene was cloned and a recombinant protein produced. An anti-porcine CD34 mAb was produced that could detect both the recombinant protein and a subset of porcine BM cells. The CD34(+) cells were phenotyped by lineage and HSC associated markers. Furthermore, the CD34(+) cells were analyzed by colony-forming unit (CFU) assay. RESULTS: Two splice variants of the porcine CD34 gene were cloned and a recombinant protein produced for mAb production. The mAb developed can detect both the recombinant protein and the native CD34 protein on a range of pig tissues, including BM. This subset of BM cells was negative for hematopoietic lineage makers, including CD3, CD14, and CD21 and positive for other known porcine HSC markers, including CD90, CD172a, histocompatibility complex (MHC) class I, and MHC class II. Moreover, the CD34(+) BM cells were enriched for multilineage progenitor cells as determined by CFU assay. CONCLUSIONS: Similar to human and mouse CD34, pig CD34 detects a subset of BM progenitor cells. This mAb will now provide a means for isolating porcine CD34(+) cells to be further analyzed for HSC activity and to assess their potential to develop pig-to-human chimeras to induce xenograft tolerance.


Assuntos
Anticorpos Monoclonais , Antígenos CD34/imunologia , Separação Celular/métodos , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/imunologia , Animais , Antígenos CD34/genética , Células da Medula Óssea , Técnicas de Cultura de Células , Clonagem Molecular , Ensaio de Unidades Formadoras de Colônias , Imunofenotipagem/métodos , Suínos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...