Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Development ; 151(13)2024 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-38828852

RESUMO

The cellular and genetic networks that contribute to the development of the zeugopod (radius and ulna of the forearm, tibia and fibula of the leg) are not well understood, although these bones are susceptible to loss in congenital human syndromes and to the action of teratogens such as thalidomide. Using a new fate-mapping approach with the Chameleon transgenic chicken line, we show that there is a small contribution of SHH-expressing cells to the posterior ulna, posterior carpals and digit 3. We establish that although the majority of the ulna develops in response to paracrine SHH signalling in both the chicken and mouse, there are differences in the contribution of SHH-expressing cells between mouse and chicken as well as between the chicken ulna and fibula. This is evidence that, although zeugopod bones are clearly homologous according to the fossil record, the gene regulatory networks that contribute to their development and evolution are not fixed.


Assuntos
Animais Geneticamente Modificados , Galinhas , Proteínas Hedgehog , Animais , Proteínas Hedgehog/metabolismo , Proteínas Hedgehog/genética , Galinhas/genética , Camundongos , Evolução Biológica , Embrião de Galinha , Ulna , Regulação da Expressão Gênica no Desenvolvimento , Fíbula/metabolismo , Rádio (Anatomia)/metabolismo , Humanos , Extremidades/embriologia
2.
PLoS Biol ; 17(2): e3000132, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30789897

RESUMO

Feathers are arranged in a precise pattern in avian skin. They first arise during development in a row along the dorsal midline, with rows of new feather buds added sequentially in a spreading wave. We show that the patterning of feathers relies on coupled fibroblast growth factor (FGF) and bone morphogenetic protein (BMP) signalling together with mesenchymal cell movement, acting in a coordinated reaction-diffusion-taxis system. This periodic patterning system is partly mechanochemical, with mechanical-chemical integration occurring through a positive feedback loop centred on FGF20, which induces cell aggregation, mechanically compressing the epidermis to rapidly intensify FGF20 expression. The travelling wave of feather formation is imposed by expanding expression of Ectodysplasin A (EDA), which initiates the expression of FGF20. The EDA wave spreads across a mesenchymal cell density gradient, triggering pattern formation by lowering the threshold of mesenchymal cells required to begin to form a feather bud. These waves, and the precise arrangement of feather primordia, are lost in the flightless emu and ostrich, though via different developmental routes. The ostrich retains the tract arrangement characteristic of birds in general but lays down feather primordia without a wave, akin to the process of hair follicle formation in mammalian embryos. The embryonic emu skin lacks sufficient cells to enact feather formation, causing failure of tract formation, and instead the entire skin gains feather primordia through a later process. This work shows that a reaction-diffusion-taxis system, integrated with mechanical processes, generates the feather array. In flighted birds, the key role of the EDA/Ectodysplasin A receptor (EDAR) pathway in vertebrate skin patterning has been recast to activate this process in a quasi-1-dimensional manner, imposing highly ordered pattern formation.


Assuntos
Padronização Corporal , Plumas/citologia , Plumas/embriologia , Transdução de Sinais , Animais , Fenômenos Biomecânicos , Aves/embriologia , Agregação Celular , Contagem de Células , Movimento Celular , Forma Celular , Ectodisplasinas/metabolismo , Receptor Edar/metabolismo , Fatores de Crescimento de Fibroblastos/metabolismo , Voo Animal/fisiologia , Mesoderma/citologia , Mesoderma/embriologia , Pele/citologia , Pele/embriologia , beta Catenina/metabolismo
3.
BMC Biotechnol ; 18(1): 82, 2018 12 29.
Artigo em Inglês | MEDLINE | ID: mdl-30594166

RESUMO

BACKGROUND: The global market for protein drugs has the highest compound annual growth rate of any pharmaceutical class but their availability, especially outside of the US market, is compromised by the high cost of manufacture and validation compared to traditional chemical drugs. Improvements in transgenic technologies allow valuable proteins to be produced by genetically-modified animals; several therapeutic proteins from such animal bioreactors are already on the market after successful clinical trials and regulatory approval. Chickens have lagged behind mammals in bioreactor development, despite a number of potential advantages, due to the historic difficulty in producing transgenic birds, but the production of therapeutic proteins in egg white of transgenic chickens would substantially lower costs across the entire production cycle compared to traditional cell culture-based production systems. This could lead to more affordable treatments and wider markets, including in developing countries and for animal health applications. RESULTS: Here we report the efficient generation of new transgenic chicken lines to optimize protein production in eggs. As proof-of-concept, we describe the expression, purification and functional characterization of three pharmaceutical proteins, the human cytokine interferon α2a and two species-specific Fc fusions of the cytokine CSF1. CONCLUSION: Our work optimizes and validates a transgenic chicken system for the cost-effective production of pure, high quality, biologically active protein for therapeutics and other applications.


Assuntos
Animais Geneticamente Modificados/genética , Biotecnologia/métodos , Galinhas/genética , Citocinas/genética , Animais , Animais Geneticamente Modificados/metabolismo , Reatores Biológicos/economia , Biotecnologia/economia , Galinhas/metabolismo , Citocinas/economia , Citocinas/metabolismo , Humanos , Interferon-alfa/economia , Interferon-alfa/genética , Interferon-alfa/metabolismo , Fator Estimulador de Colônias de Macrófagos/economia , Fator Estimulador de Colônias de Macrófagos/genética , Fator Estimulador de Colônias de Macrófagos/metabolismo , Proteínas Recombinantes/economia , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
4.
Development ; 141(16): 3255-65, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25063453

RESUMO

We have generated the first transgenic chickens in which reporter genes are expressed in a specific immune cell lineage, based upon control elements of the colony stimulating factor 1 receptor (CSF1R) locus. The Fms intronic regulatory element (FIRE) within CSF1R is shown to be highly conserved in amniotes and absolutely required for myeloid-restricted expression of fluorescent reporter genes. As in mammals, CSF1R-reporter genes were specifically expressed at high levels in cells of the macrophage lineage and at a much lower level in granulocytes. The cell lineage specificity of reporter gene expression was confirmed by demonstration of coincident expression with the endogenous CSF1R protein. In transgenic birds, expression of the reporter gene provided a defined marker for macrophage-lineage cells, identifying the earliest stages in the yolk sac, throughout embryonic development and in all adult tissues. The reporter genes permit detailed and dynamic visualisation of embryonic chicken macrophages. Chicken embryonic macrophages are not recruited to incisional wounds, but are able to recognise and phagocytose microbial antigens.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Macrófagos/citologia , Animais , Animais Geneticamente Modificados , Sequência de Bases , Aves , Linhagem da Célula , Galinhas , Células Dendríticas/citologia , Genes Reporter , Técnicas Genéticas , Sistema Imunitário , Íntrons , Dados de Sequência Molecular , Fagocitose , Receptor de Fator Estimulador de Colônias de Macrófagos/metabolismo , Homologia de Sequência do Ácido Nucleico , Especificidade da Espécie , Transgenes , Saco Vitelino/fisiologia
5.
J Immunol ; 194(5): 2338-44, 2015 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-25637020

RESUMO

We have identified differences in gene expression in macrophages grown from the bone marrow of male and female chickens in recombinant chicken M-CSF (CSF1). Cells were profiled with or without treatment with bacterial LPS for 24 h. Approximately 600 transcripts were induced by prolonged LPS stimulation to an equal extent in the male and female macrophages. Many transcripts encoded on the Z chromosome were expressed ∼1.6-fold higher in males, reflecting a lack of dosage compensation in the homogametic sex. A smaller set of W chromosome-specific genes was expressed only in females. LPS signaling in mammals is associated with induction of type 1 IFN-responsive genes. Unexpectedly, because IFNs are encoded on the Z chromosome of chickens, unstimulated macrophages from the female birds expressed a set of known IFN-inducible genes at much higher levels than male cells under the same conditions. To confirm that these differences were not the consequence of the actions of gonadal hormones, we induced gonadal sex reversal to alter the hormonal environment of the developing chick and analyzed macrophages cultured from male, female, and female sex-reversed embryos. Gonadal sex reversal did not alter the sexually dimorphic expression of either sex-linked or IFN-responsive genes. We suggest that female birds compensate for the reduced dose of inducible IFN with a higher basal set point of IFN-responsive genes.


Assuntos
Proteínas Aviárias/imunologia , Galinhas/imunologia , Gônadas/imunologia , Macrófagos/imunologia , RNA Mensageiro/imunologia , Cromossomos Sexuais/imunologia , Animais , Inibidores da Aromatase/farmacologia , Proteínas Aviárias/genética , Células Cultivadas , Embrião de Galinha , Galinhas/genética , Mecanismo Genético de Compensação de Dose , Fadrozol/farmacologia , Feminino , Expressão Gênica , Perfilação da Expressão Gênica , Gônadas/efeitos dos fármacos , Interferon-alfa/genética , Interferon-alfa/imunologia , Interferon beta/genética , Interferon beta/imunologia , Lipopolissacarídeos/farmacologia , Fator Estimulador de Colônias de Macrófagos/farmacologia , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Masculino , RNA Mensageiro/genética , Caracteres Sexuais
6.
BMC Biol ; 13: 12, 2015 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-25857347

RESUMO

BACKGROUND: Macrophages have many functions in development and homeostasis as well as innate immunity. Recent studies in mammals suggest that cells arising in the yolk sac give rise to self-renewing macrophage populations that persist in adult tissues. Macrophage proliferation and differentiation is controlled by macrophage colony-stimulating factor (CSF1) and interleukin 34 (IL34), both agonists of the CSF1 receptor (CSF1R). In the current manuscript we describe the origin, function and regulation of macrophages, and the role of CSF1R signaling during embryonic development, using the chick as a model. RESULTS: Based upon RNA-sequencing comparison to bone marrow-derived macrophages grown in CSF1, we show that embryonic macrophages contribute around 2% of the total embryo RNA in day 7 chick embryos, and have similar gene expression profiles to bone marrow-derived macrophages. To explore the origins of embryonic and adult macrophages, we injected Hamburger-Hamilton stage 16 to 17 chick embryos with either yolk sac-derived blood cells, or bone marrow cells from EGFP+ donors. In both cases, the transferred cells gave rise to large numbers of EGFP+ tissue macrophages in the embryo. In the case of the yolk sac, these cells were not retained in hatched birds. Conversely, bone marrow EGFP+ cells gave rise to tissue macrophages in all organs of adult birds, and regenerated CSF1-responsive marrow macrophage progenitors. Surprisingly, they did not contribute to any other hematopoietic lineage. To explore the role of CSF1 further, we injected embryonic or hatchling CSF1R-reporter transgenic birds with a novel chicken CSF1-Fc conjugate. In both cases, the treatment produced a large increase in macrophage numbers in all tissues examined. There were no apparent adverse effects of chicken CSF1-Fc on embryonic or post-hatch development, but there was an unexpected increase in bone density in the treated hatchlings. CONCLUSIONS: The data indicate that the yolk sac is not the major source of macrophages in adult birds, and that there is a macrophage-restricted, self-renewing progenitor cell in bone marrow. CSF1R is demonstrated to be limiting for macrophage development during development in ovo and post-hatch. The chicken provides a novel and tractable model to study the development of the mononuclear phagocyte system and CSF1R signaling.


Assuntos
Galinhas/imunologia , Sistema Fagocitário Mononuclear/embriologia , Sistema Fagocitário Mononuclear/metabolismo , Receptor de Fator Estimulador de Colônias de Macrófagos/metabolismo , Transdução de Sinais , Animais , Células Sanguíneas/efeitos dos fármacos , Células Sanguíneas/metabolismo , Densidade Óssea/efeitos dos fármacos , Células da Medula Óssea , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Embrião de Galinha , Galinhas/genética , Citometria de Fluxo , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Fator Estimulador de Colônias de Macrófagos/farmacologia , Sistema Fagocitário Mononuclear/efeitos dos fármacos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Análise de Sequência de RNA , Transdução de Sinais/efeitos dos fármacos , Saco Vitelino/citologia
7.
Proc Natl Acad Sci U S A ; 109(23): E1466-72, 2012 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-22586100

RESUMO

The derivation of germ-line competent avian primordial germ cells establishes a cell-based model system for the investigation of germ cell differentiation and the production of genetically modified animals. Current methods to modify primordial germ cells using DNA or retroviral vectors are inefficient and prone to epigenetic silencing. Here, we validate the use of transposable elements for the genetic manipulation of primordial germ cells. We demonstrate that chicken primordial germ cells can be modified in vitro using transposable elements. Both piggyBac and Tol2 transposons efficiently transpose primordial germ cells. Tol2 transposon integration sites were spread throughout both the macro- and microchromosomes of the chicken genome and were more prevalent in gene transcriptional units and intronic regions, consistent with transposon integrations observed in other species. We determined that the presence of insulator elements was not required for reporter gene expression from the integrated transposon. We further demonstrate that a gene-trap cassette carried in the Tol2 transposon can trap and mutate endogenous transcripts in primordial germ cells. Finally, we observed that modified primordial germ cells form functional gametes as demonstrated by the generation of transgenic offspring that correctly expressed a reporter gene carried in the transposon. Transposable elements are therefore efficient vectors for the genetic manipulation of primordial germ cells and the chicken genome.


Assuntos
Animais Geneticamente Modificados/genética , Elementos de DNA Transponíveis/genética , Técnicas de Transferência de Genes , Vetores Genéticos/genética , Células Germinativas/metabolismo , Animais , Southern Blotting , Técnicas de Cultura de Células , Embrião de Galinha , Primers do DNA/genética , Genes Reporter/genética , Reação em Cadeia da Polimerase/métodos
8.
Front Immunol ; 14: 1273661, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37954617

RESUMO

Conventional dendritic cells (cDCs) are antigen-presenting cells (APCs) that play a central role in linking innate and adaptive immunity. cDCs have been well described in a number of different mammalian species, but remain poorly characterised in the chicken. In this study, we use previously described chicken cDC specific reagents, a novel gene-edited chicken line and single-cell RNA sequencing (scRNAseq) to characterise chicken splenic cDCs. In contrast to mammals, scRNAseq analysis indicates that the chicken spleen contains a single, chemokine receptor XCR1 expressing, cDC subset. By sexual maturity the XCR1+ cDC population is the most abundant mononuclear phagocyte cell subset in the chicken spleen. scRNAseq analysis revealed substantial heterogeneity within the chicken splenic XCR1+ cDC population. Immature MHC class II (MHCII)LOW XCR1+ cDCs expressed a range of viral resistance genes. Maturation to MHCIIHIGH XCR1+ cDCs was associated with reduced expression of anti-viral gene expression and increased expression of genes related to antigen presentation via the MHCII and cross-presentation pathways. To visualise and transiently ablate chicken XCR1+ cDCs in situ, we generated XCR1-iCaspase9-RFP chickens using a CRISPR-Cas9 knockin transgenesis approach to precisely edit the XCR1 locus, replacing the XCR1 coding region with genes for a fluorescent protein (TagRFP), and inducible Caspase 9. After inducible ablation, the chicken spleen is initially repopulated by immature CD1.1+ XCR1+ cDCs. XCR1+ cDCs are abundant in the splenic red pulp, in close association with CD8+ T-cells. Knockout of XCR1 prevented this clustering of cDCs with CD8+ T-cells. Taken together these data indicate a conserved role for chicken and mammalian XCR1+ cDCs in driving CD8+ T-cells responses.


Assuntos
Linfócitos T CD8-Positivos , Galinhas , Animais , Apresentação de Antígeno , Células Dendríticas , Apresentação Cruzada , Mamíferos
9.
Nat Commun ; 14(1): 6136, 2023 10 10.
Artigo em Inglês | MEDLINE | ID: mdl-37816720

RESUMO

Chickens genetically resistant to avian influenza could prevent future outbreaks. In chickens, influenza A virus (IAV) relies on host protein ANP32A. Here we use CRISPR/Cas9 to generate homozygous gene edited (GE) chickens containing two ANP32A amino acid substitutions that prevent viral polymerase interaction. After IAV challenge, 9/10 edited chickens remain uninfected. Challenge with a higher dose, however, led to breakthrough infections. Breakthrough IAV virus contained IAV polymerase gene mutations that conferred adaptation to the edited chicken ANP32A. Unexpectedly, this virus also replicated in chicken embryos edited to remove the entire ANP32A gene and instead co-opted alternative ANP32 protein family members, chicken ANP32B and ANP32E. Additional genome editing for removal of ANP32B and ANP32E eliminated all viral growth in chicken cells. Our data illustrate a first proof of concept step to generate IAV-resistant chickens and show that multiple genetic modifications will be required to curtail viral escape.


Assuntos
Vírus da Influenza A , Influenza Aviária , Embrião de Galinha , Animais , Influenza Aviária/genética , Edição de Genes , RNA Polimerase Dependente de RNA/genética , RNA Polimerase Dependente de RNA/metabolismo , Galinhas/genética , Vírus da Influenza A/genética , Vírus da Influenza A/metabolismo
10.
Exp Cell Res ; 314(14): 2634-42, 2008 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-18571647

RESUMO

The expression of 4 pluripotency genes (Oct4, Sox2, c-Myc and Klf4) in mouse embryonic fibroblasts can reprogramme them to a pluripotent state. We have investigated the expression of these pluripotency genes when human somatic 293T cells are permeabilized and incubated in extracts of mouse embryonic stem (ES) cells. Expression of all 4 genes was induced over 1-8 h. Gene expression was associated with loss of repressive histone H3 modifications and increased recruitment of RNA polymerase II at the promoters. Lamin A/C, which is typically found only in differentiated cells, was also removed from the nuclei. When 293T cells were returned to culture after exposure to ES cell extract, the expression of the pluripotency genes continued to rise over the following 48 h of culture, suggesting that long-term reprogramming of gene expression had been induced. This provides a methodology for studying the de-differentiation of somatic cells that can potentially lead to an efficient way of reprogramming somatic cells to a pluripotent state without genetically altering them.


Assuntos
Extratos Celulares/farmacologia , Células-Tronco Embrionárias/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Células-Tronco Pluripotentes/metabolismo , Animais , Biomarcadores/metabolismo , Linhagem Celular , Permeabilidade da Membrana Celular/efeitos dos fármacos , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/efeitos dos fármacos , Histonas/metabolismo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Fator 4 Semelhante a Kruppel , Lamina Tipo A/isolamento & purificação , Camundongos , Proteína Homeobox Nanog , Fator 3 de Transcrição de Octâmero/genética , Fator 3 de Transcrição de Octâmero/metabolismo , Células-Tronco Pluripotentes/efeitos dos fármacos , Regiões Promotoras Genéticas/genética , Ligação Proteica/efeitos dos fármacos , Biossíntese de Proteínas/efeitos dos fármacos , RNA Polimerase II/metabolismo , Transcrição Gênica/efeitos dos fármacos , Xenopus
11.
Front Immunol ; 10: 2495, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31695701

RESUMO

The follicle-associated epithelium (FAE) is a specialized structure that samples luminal antigens and transports them into mucosa-associated lymphoid tissues (MALT). In mammals, transcytosis of antigens across the gut epithelium is performed by a subset of FAE cells known as M cells. Here we show that colony-stimulating factor 1 receptor (CSF1R) is expressed by a subset of cells in the avian bursa of Fabricius FAE. Expression was initially detected using a CSF1R-reporter transgene that also label subsets of bursal macrophages. Immunohistochemical detection using a specific monoclonal antibody confirmed abundant expression of CSF1R on the basolateral membrane of FAE cells. CSF1R-transgene expressing bursal FAE cells were enriched for expression of markers previously reported as putative M cell markers, including annexin A10 and CD44. They were further distinguished from a population of CSF1R-transgene negative epithelial cells within FAE by high apical F-actin expression and differential staining with the lectins jacalin, PHA-L and SNA. Bursal FAE cells that express the CSF1R-reporter transgene were responsible for the bulk of FAE transcytosis of labeled microparticles in the size range 0.02-0.1 µm. Unlike mammalian M cells, they did not readily take up larger bacterial sized microparticles (0.5 µm). Their role in uptake of bacteria was tested using Salmonella, which can enter via M cells in mammals. Labeled Salmonella enterica serovar Typhimurium entered bursal tissue via the FAE. Entry was partially dependent upon Type III secretion system-1. However, the majority of invading bacteria were localized to CSF1R-negative FAE cells and in resident phagocytes that express the phosphatidylserine receptor TIM4. CSF1R-expressing FAE cells in infected follicles showed evidence of cell death and shedding into the bursal lumen. In mammals, CSF1R expression in the gut is restricted to macrophages which only indirectly control M cell differentiation. The novel expression of CSF1R in birds suggests that these functional equivalents to mammalian M cells may have different ontological origins and their development and function are likely to be regulated by different growth factors.


Assuntos
Apresentação de Antígeno/imunologia , Proteínas Aviárias/imunologia , Bolsa de Fabricius/imunologia , Células Epiteliais/imunologia , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/imunologia , Infecções por Salmonella/imunologia , Salmonella typhimurium/imunologia , Animais , Antígenos de Bactérias , Antígenos de Diferenciação/imunologia , Bolsa de Fabricius/patologia , Galinhas , Humanos , Infecções por Salmonella/patologia
12.
Sci Rep ; 8(1): 15126, 2018 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-30310080

RESUMO

Primordial germ cells (PGCs), the embryonic precursors of the sperm and egg, are used for the introduction of genetic modifications into avian genome. Introduction of small defined sequences using genome editing has not been demonstrated in bird species. Here, we compared oligonucleotide-mediated HDR using wild type SpCas9 (SpCas9-WT) and high fidelity SpCas9-HF1 in PGCs and show that many loci in chicken PGCs can be precise edited using donors containing CRISPR/Cas9-blocking mutations positioned in the protospacer adjacent motif (PAM). However, targeting was more efficient using SpCas9-HF1 when mutations were introduced only into the gRNA target sequence. We subsequently employed an eGFP-to-BFP conversion assay, to directly compare HDR mediated by SpCas9-WT and SpCas9-HF1 and discovered that SpCas9-HF1 increases HDR while reducing INDEL formation. Furthermore, SpCas9-HF1 increases the frequency of single allele editing in comparison to SpCas9-WT. We used SpCas9-HF1 to demonstrate the introduction of monoallelic and biallelic point mutations into the FGF20 gene and generate clonal populations of edited PGCs with defined homozygous and heterozygous genotypes. Our results demonstrate the use of oligonucleotide donors and high fidelity CRISPR/Cas9 variants to perform precise genome editing with high efficiency in PGCs.


Assuntos
Alelos , Sistemas CRISPR-Cas , Edição de Genes , Células Germinativas/metabolismo , Animais , Sequência de Bases , Sítios de Ligação , Galinhas , Ordem dos Genes , Vetores Genéticos/genética , Células Germinativas/citologia , Heterozigoto , Mutação INDEL , Mutação , Ligação Proteica , Análise de Sequência de DNA
13.
Int J Dev Biol ; 62(1-2-3): 257-264, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29616734

RESUMO

After decades of research investment, techniques for the robust and efficient modification of the chicken genome are now with us. The biology of the chicken has provided many challenges, as have the methods by which transgenes can be readily, stably and functionally integrated into the genome. Now that these obstacles have been surmounted and the chicken has been 'updated' to a cutting-edge modern model organism, a future as a central and versatile model in developmental biology beckons. In this review, we describe recent advances in genetic modification of the chicken and some of the many transgenic models developed for the elucidation of the mechanisms of embryogenesis.


Assuntos
Embrião de Galinha , Galinhas/fisiologia , Engenharia Genética , Animais , Animais Geneticamente Modificados , Técnicas de Cultura Embrionária , Desenvolvimento Embrionário , Edição de Genes , Genoma , Células Germinativas/citologia , Proteínas de Fluorescência Verde/metabolismo , Lentivirus , Modelos Genéticos , Transgenes
14.
Genome Biol ; 19(1): 204, 2018 11 26.
Artigo em Inglês | MEDLINE | ID: mdl-30477539

RESUMO

The human population is growing, and as a result we need to produce more food whilst reducing the impact of farming on the environment. Selective breeding and genomic selection have had a transformational impact on livestock productivity, and now transgenic and genome-editing technologies offer exciting opportunities for the production of fitter, healthier and more-productive livestock. Here, we review recent progress in the application of genome editing to farmed animal species and discuss the potential impact on our ability to produce food.


Assuntos
Edição de Genes , Gado/genética , Criação de Animais Domésticos , Animais , Genômica
15.
Trends Mol Med ; 11(6): 293-8, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15949771

RESUMO

Recent advances in the efficiency of transgenic technology have important implications for medicine. The production of therapeutic proteins from animal bioreactors is well established and the first products are close to market. The genetic modification of pigs to improve their suitability as organ donors for xenotransplantation has been initiated, but many challenges remain. The use of transgenesis, in combination with the method of RNA interference to knock down gene expression, has been proposed as a method for making animals resistant to viral diseases, which could reduce the likelihood of transmission to humans. Here, the latest developments in transgenic technology and their applications relevant to medicine and human health will be discussed.


Assuntos
Transgenes , Animais , Reatores Biológicos , Biotecnologia , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Modelos Animais de Doenças , Engenharia Genética , Humanos , Interferência de RNA , RNA Mensageiro/metabolismo , Transplante , Transplante Heterólogo
16.
Drug Discov Today ; 10(3): 191-6, 2005 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-15708533

RESUMO

The potential of using transgenic animals for the synthesis of therapeutic proteins was suggested over twenty years ago. Considerable progress has been made in developing methods for the production of transgenic animals and specifically in the expression of therapeutic proteins in the mammary glands of cows, sheep and goats. Development of transgenic hens for protein production in eggs has lagged behind these systems. The positive features associated with the use of the chicken in terms of cost, speed of development of a production flock and potentially appropriate glycosylation of target proteins have led to significant advances in transgenic chicken models in the past few years.


Assuntos
Animais Geneticamente Modificados , Reatores Biológicos , Galinhas , Proteínas/metabolismo , Animais , Galinhas/genética , Óvulo/metabolismo , Proteínas/genética , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética
17.
Nat Cell Biol ; 17(4): 397-408, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25812521

RESUMO

Primitive streak formation in the chick embryo involves large-scale highly coordinated flows of more than 100,000 cells in the epiblast. These large-scale tissue flows and deformations can be correlated with specific anisotropic cell behaviours in the forming mesendoderm through a combination of light-sheet microscopy and computational analysis. Relevant behaviours include apical contraction, elongation along the apical-basal axis followed by ingression, and asynchronous directional cell intercalation of small groups of mesendoderm cells. Cell intercalation is associated with sequential, directional contraction of apical junctions, the onset, localization and direction of which correlate strongly with the appearance of active myosin II cables in aligned apical junctions in neighbouring cells. Use of class specific myosin inhibitors and gene-specific knockdown shows that apical contraction and intercalation are myosin II dependent and also reveal critical roles for myosin I and myosin V family members in the assembly of junctional myosin II cables.


Assuntos
Forma Celular/fisiologia , Miosina Tipo II/metabolismo , Miosina Tipo I/metabolismo , Miosina Tipo V/metabolismo , Linha Primitiva/embriologia , Animais , Animais Geneticamente Modificados , Linhagem Celular , Movimento Celular , Proliferação de Células , Embrião de Galinha , Galinhas , Gastrulação/fisiologia , Células HEK293 , Compostos Heterocíclicos de 4 ou mais Anéis/farmacologia , Humanos , Hidrocarbonetos Clorados/farmacologia , Miosina Tipo I/antagonistas & inibidores , Miosina Tipo I/genética , Miosina Tipo II/antagonistas & inibidores , Miosina Tipo II/genética , Miosina Tipo V/antagonistas & inibidores , Miosina Tipo V/genética , Fosforilação , Linha Primitiva/citologia , Pirróis/farmacologia , Interferência de RNA , RNA Interferente Pequeno
18.
Dev Comp Immunol ; 42(2): 278-85, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24084378

RESUMO

Macrophages contribute to innate and acquired immunity as well as many aspects of homeostasis and development. Studies of macrophage biology and function in birds have been hampered by a lack of definitive cell surface markers. As in mammals, avian macrophages proliferate and differentiate in response to CSF1 and IL34, acting through the shared receptor, CSF1R. CSF1R mRNA expression in the chicken is restricted to macrophages and their progenitors. To expedite studies of avian macrophage biology, we produced an avian CSF1R-Fc chimeric protein and generated a monoclonal antibody (designated ROS-AV170) against the chicken CSF1R using the chimeric protein as immunogen. Specific binding of ROS-AV170 to CSF1R was confirmed by FACS, ELISA and immunohistochemistry on tissue sections. CSF1 down-regulated cell surface expression of the CSF1R detected with ROS-AV170, but the antibody did not block CSF1 signalling. Expression of CSF1R was detected on the surface of bone marrow progenitors only after culture in the absence of CSF1, and was induced during macrophage differentiation. Constitutive surface expression of CSF1R distinguished monocytes from other myeloid cells, including heterophils and thrombocytes. This antibody will therefore be of considerable utility for the study of chicken macrophage biology.


Assuntos
Anticorpos Monoclonais/imunologia , Galinhas/imunologia , Macrófagos/imunologia , Receptor de Fator Estimulador de Colônias de Macrófagos/imunologia , Animais , Células da Medula Óssea/imunologia , Células CHO , Diferenciação Celular/imunologia , Linhagem Celular , Linhagem da Célula/imunologia , Cricetulus , Feminino , Fator Estimulador de Colônias de Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Monócitos/imunologia , RNA Mensageiro/biossíntese , Receptor de Fator Estimulador de Colônias de Macrófagos/biossíntese , Receptor de Fator Estimulador de Colônias de Macrófagos/genética
19.
PLoS One ; 8(11): e77222, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24223709

RESUMO

In this work, we describe a single piggyBac transposon system containing both a tet-activator and a doxycycline-inducible expression cassette. We demonstrate that a gene product can be conditionally expressed from the integrated transposon and a second gene can be simultaneously targeted by a short hairpin RNA contained within the transposon, both in vivo and in mammalian and avian cell lines. We applied this system to stably modify chicken primordial germ cell (PGC) lines in vitro and induce a reporter gene at specific developmental stages after injection of the transposon-modified germ cells into chicken embryos. We used this vector to express a constitutively-active AKT molecule during PGC migration to the forming gonad. We found that PGC migration was retarded and cells could not colonise the forming gonad. Correct levels of AKT activation are thus essential for germ cell migration during early embryonic development.


Assuntos
Movimento Celular , Elementos de DNA Transponíveis , Células Germinativas/fisiologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ativação Transcricional , Células-Tronco Adultas/metabolismo , Animais , Linhagem Celular , Embrião de Galinha , Galinhas , Clonagem Molecular , Células-Tronco Embrionárias/metabolismo , Fibroblastos/metabolismo , Expressão Gênica , Engenharia Genética , Células Germinativas/transplante , Proteínas de Fluorescência Verde/biossíntese , Proteínas de Fluorescência Verde/genética , Masculino , Camundongos , Camundongos Transgênicos , Sêmen/citologia , Transdução de Sinais
20.
PLoS One ; 5(11): e15518, 2010 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-21124737

RESUMO

BACKGROUND: Avian primordial germ cells (PGCs) have significant potential to be used as a cell-based system for the study and preservation of avian germplasm, and the genetic modification of the avian genome. It was previously reported that PGCs from chicken embryos can be propagated in culture and contribute to the germ cell lineage of host birds. PRINCIPAL FINDINGS: We confirm these results by demonstrating that PGCs from a different layer breed of chickens can be propagated for extended periods in vitro. We demonstrate that intracellular signalling through PI3K and MEK is necessary for PGC growth. We carried out an initial characterisation of these cells. We find that cultured PGCs contain large lipid vacuoles, are glycogen rich, and express the stem cell marker, SSEA-1. These cells also express the germ cell-specific proteins CVH and CDH. Unexpectedly, using RT-PCR we show that cultured PGCs express the pluripotency genes c-Myc, cKlf4, cPouV, cSox2, and cNanog. Finally, we demonstrate that the cultured PGCs will migrate to and colonise the forming gonad of host embryos. Male PGCs will colonise the female gonad and enter meiosis, but are lost from the gonad during sexual development. In male hosts, cultured PGCs form functional gametes as demonstrated by the generation of viable offspring. CONCLUSIONS: The establishment of in vitro cultures of germline competent avian PGCs offers a unique system for the study of early germ cell differentiation and also a comparative system for mammalian germ cell development. Primary PGC lines will form the basis of an alternative technique for the preservation of avian germplasm and will be a valuable tool for transgenic technology, with both research and industrial applications.


Assuntos
Linhagem da Célula , Proliferação de Células , Células-Tronco Embrionárias/citologia , Células Germinativas/citologia , Animais , Animais Geneticamente Modificados , Benzamidas/farmacologia , Células Cultivadas , Embrião de Galinha , Difenilamina/análogos & derivados , Difenilamina/farmacologia , Células-Tronco Embrionárias/efeitos dos fármacos , Células-Tronco Embrionárias/metabolismo , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Células Germinativas/metabolismo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Imuno-Histoquímica , Hibridização In Situ , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Meiose , Fosfatidilinositol 3-Quinases/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA