Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
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.
Immunology ; 165(2): 171-194, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34767637

RESUMO

Conventional dendritic cells (cDC) are bone marrow-derived immune cells that play a central role in linking innate and adaptive immunity. cDCs efficiently uptake, process and present antigen to naïve T cells, driving clonal expansion of antigen-specific T-cell responses. In chicken, vital reagents are lacking for the efficient and precise identification of cDCs. In this study, we have developed several novel reagents for the identification and characterization of chicken cDCs. Chicken FLT3 cDNA was cloned and a monoclonal antibody to cell surface FLT3 was generated. This antibody identified a distinct FLT3HI splenic subset which lack expression of signature markers for B cells, T cells or monocyte/macrophages. By combining anti-FLT3 and CSF1R-eGFP transgenic expression, three major populations within the mononuclear phagocyte system were identified in the spleen. The cDC1 subset of mammalian cDCs express the chemokine receptor XCR1. To characterize chicken cDCs, a synthetic chicken chemokine (C motif) ligand (XCL1) peptide conjugated to Alexa Fluor 647 was developed (XCL1AF647 ). Flow cytometry staining of XCL1AF647 on splenocytes showed that all chicken FLT3HI cells exclusively express XCR1, supporting the hypothesis that this population comprises bona fide chicken cDCs. Further analysis revealed that chicken cDCs expressed CSF1R but lacked the expression of CSF2R. Collectively, the cell surface phenotypes of chicken cDCs were partially conserved with mammalian XCR1+ cDC1, with distinct differences in CSF1R and CSF2R expression compared with mammalian orthologues. These original reagents allow the efficient identification of chicken cDCs to investigate their important roles in the chicken immunity and diseases.


Assuntos
Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/metabolismo , Tirosina Quinase 3 Semelhante a fms/metabolismo , Animais , Anticorpos Monoclonais , Biomarcadores , Técnicas de Cultura de Células , Galinhas , Imunofluorescência , Expressão Gênica , Humanos , Imunofenotipagem , Receptores Acoplados a Proteínas G/genética , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Tirosina Quinase 3 Semelhante a fms/genética
3.
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
4.
Dysphagia ; 37(5): 1305-1313, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-34981254

RESUMO

Infants with congenital diaphragmatic hernia (CDH) who require non-invasive positive pressure ventilation or high flow nasal cannula are at risk for aspiration and delayed initiation of oral feeding. We developed a dysphagia provider-led protocol that involved early consultation with an occupational therapist or speech/language pathologist and modified barium swallow study (MBSS) to assess for readiness for oral feeding initiation/advancement on non-invasive positive pressure ventilation. The objective of this study was to retrospectively compare this intervention cohort to a historical control cohort to evaluate the protocol's impact on the time to initiate oral feeding. We describe the development and implementation of the protocol, the MBSS findings of the intervention cohort, and compared the control (n = 64) and intervention (n = 37) cohorts using Fischer's exact test and Mann-Whitney test. We found that both cohorts had similar prenatal and neonatal characteristics including age at extubation. Significantly more infants in the intervention cohort were on non-invasive positive pressure ventilation or high flow nasal cannula at the time of oral feeding initiation (84% vs. 28%, p < 0.0001). None of the control cohort infants underwent MBSS while on respiratory support. Of the intervention cohort, 15 infants underwent a MBSS while on non-invasive positive pressure ventilation; 6 had no evidence of laryngeal penetration and/or aspiration during swallowing. Infants in the control cohort initiated oral feeds significantly sooner after extubation (6 versus 11 days, p = 0.001) and attained full oral feeds earlier (20 days versus 28 days, p = 0.02) than the intervention group. There was no difference in the rate of gastrostomy tube placement (38%). Appropriate monitoring by a dysphagia provider and evaluation with clinical and radiological means are crucial to determine the safety of initiating oral feeding in term infants with CDH. Continued surveillance is needed to determine the long-term impact on oral feeding progression in this population.


Assuntos
Transtornos de Deglutição , Hérnias Diafragmáticas Congênitas , Deglutição , Transtornos de Deglutição/epidemiologia , Transtornos de Deglutição/etiologia , Transtornos de Deglutição/terapia , Hérnias Diafragmáticas Congênitas/complicações , Humanos , Lactente , Recém-Nascido , Respiração com Pressão Positiva/métodos , Estudos Retrospectivos
5.
PLoS Biol ; 16(4): e2004162, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29708962

RESUMO

The vertebrate neuroepithelium is composed of elongated progenitors whose reciprocal attachments ensure the continuity of the ventricular wall. As progenitors commit to differentiation, they translocate their nucleus basally and eventually withdraw their apical endfoot from the ventricular surface. However, the mechanisms allowing this delamination process to take place while preserving the integrity of the neuroepithelial tissue are still unclear. Here, we show that Notch signaling, which is classically associated with an undifferentiated state, remains active in prospective neurons until they delaminate. During this transition period, prospective neurons rapidly reduce their apical surface and only later down-regulate N-Cadherin levels. Upon Notch blockade, nascent neurons disassemble their junctions but fail to reduce their apical surface. This disrupted sequence weakens the junctional network and eventually leads to breaches in the ventricular wall. We also provide evidence that the Notch ligand Delta-like 1 (Dll1) promotes differentiation by reducing Notch signaling through a Cis-inhibition mechanism. However, during the delamination process, the ubiquitin ligase Mindbomb1 (Mib1) transiently blocks this Cis-inhibition and sustains Notch activity to defer differentiation. We propose that the fine-tuned balance between Notch Trans-activation and Cis-inhibition allows neuroepithelial cells to seamlessly delaminate from the ventricular wall as they commit to differentiation.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteínas de Membrana/genética , Células Neuroepiteliais/metabolismo , Neurogênese/genética , Receptores Notch/genética , Ubiquitina-Proteína Ligases/genética , Animais , Animais Geneticamente Modificados , Caderinas/genética , Caderinas/metabolismo , Diferenciação Celular , Embrião de Galinha , Galinhas , Feminino , Vetores Genéticos/química , Vetores Genéticos/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Lentivirus/genética , Lentivirus/metabolismo , Masculino , Proteínas de Membrana/metabolismo , Células Neuroepiteliais/citologia , Neurônios/citologia , Neurônios/metabolismo , Plasmídeos/química , Plasmídeos/metabolismo , Receptores Notch/metabolismo , Transdução de Sinais , Transfecção , Ubiquitina-Proteína Ligases/metabolismo
6.
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
7.
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
8.
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
9.
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
10.
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
11.
Development ; 138(12): 2613-24, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21610037

RESUMO

The infundibulum links the nervous and endocrine systems, serving as a crucial integrating centre for body homeostasis. Here we describe that the chick infundibulum derives from two subsets of anterior ventral midline cells. One set remains at the ventral midline and forms the posterior-ventral infundibulum. A second set migrates laterally, forming a collar around the midline. We show that collar cells are composed of Fgf3(+) SOX3(+) proliferating progenitors, the induction of which is SHH dependent, but the maintenance of which requires FGF signalling. Collar cells proliferate late into embryogenesis, can generate neurospheres that passage extensively, and differentiate to distinct fates, including hypothalamic neuronal fates and Fgf10(+) anterior-dorsal infundibular cells. Together, our study shows that a subset of anterior floor plate-like cells gives rise to Fgf3(+) SOX3(+) progenitor cells, demonstrates a dual origin of infundibular cells and reveals a crucial role for FGF signalling in governing extended infundibular growth.


Assuntos
Fator 3 de Crescimento de Fibroblastos/fisiologia , Fatores de Crescimento de Fibroblastos/fisiologia , Neuro-Hipófise/embriologia , Células-Tronco/citologia , Animais , Padronização Corporal , Embrião de Galinha , Fator 3 de Crescimento de Fibroblastos/análise , Neuro-Hipófise/citologia , Neuro-Hipófise/crescimento & desenvolvimento , Fatores de Transcrição SOXB1/análise , Fatores de Transcrição SOXB1/fisiologia , Células-Tronco/fisiologia
12.
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
13.
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
14.
Dev Biol ; 357(1): 108-16, 2011 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-21741963

RESUMO

The forelimbs of higher vertebrates are composed of two portions: the appendicular region (stylopod, zeugopod and autopod) and the less prominent proximal girdle elements (scapula and clavicle) that brace the limb to the main trunk axis. We show that the formation of the muscles of the proximal limb occurs through two distinct mechanisms. The more superficial girdle muscles (pectoral and latissimus dorsi) develop by the "In-Out" mechanism whereby migration of myogenic cells from the somites into the limb bud is followed by their extension from the proximal limb bud out onto the thorax. In contrast, the deeper girdle muscles (e.g. rhomboideus profundus and serratus anterior) are induced by the forelimb field which promotes myotomal extension directly from the somites. Tbx5 inactivation demonstrated its requirement for the development of all forelimb elements which include the skeletal elements, proximal and distal muscles as well as the sternum in mammals and the cleithrum of fish. Intriguingly, the formation of the diaphragm musculature is also dependent on the Tbx5 programme. These observations challenge our classical views of the boundary between limb and trunk tissues. We suggest that significant structures located in the body should be considered as components of the forelimb.


Assuntos
Padronização Corporal , Membro Anterior/embriologia , Músculo Esquelético/embriologia , Animais , Embrião de Galinha , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Membro Anterior/citologia , Camundongos , Músculo Esquelético/anatomia & histologia , Músculo Esquelético/citologia , Somitos/citologia , Proteínas com Domínio T/genética , Proteínas com Domínio T/metabolismo , Peixe-Zebra/embriologia , Peixe-Zebra/genética
16.
Dev Dyn ; 240(5): 1163-72, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21465618

RESUMO

Point mutations in the intronic ZRS region of Lmbr1, a limb specific cis-regulatory element of Sonic hedgehog (Shh), are associated with polydactyly in humans, cats, and mice. We and others have recently mapped the dominant preaxial polydactyly (Po) locus in Silkie chickens to a single nucleotide polymorphism (SNP) in the ZRS region. Using polymorphisms in the chicken Shh sequence, we confirm that the ZRS region directly regulates Shh expression in the developing limb causing ectopic Shh expression in the anterior leg, prolonged Shh expression in the posterior limb, and allelic imbalance between wt and Slk Shh alleles in heterozygote limbs. Using Silkie legs, we have explored the consequences of increased Shh expression in the posterior leg on the patterning of the toes, and the induction of preaxial polydactyly.


Assuntos
Extremidades/embriologia , Proteínas Hedgehog/metabolismo , Animais , Gatos , Embrião de Galinha , Galinhas , Genótipo , Proteínas Hedgehog/genética , Hibridização In Situ , Camundongos , Polidactilia , Polimorfismo de Fragmento de Restrição/genética , Polimorfismo de Nucleotídeo Único/genética , Locos de Características Quantitativas/genética
17.
BMC Dev Biol ; 10: 26, 2010 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-20184756

RESUMO

BACKGROUND: Regulatory elements that control expression of specific genes during development have been shown in many cases to contain functionally-conserved modules that can be transferred between species and direct gene expression in a comparable developmental pattern. An example of such a module has been identified at the rat myosin light chain (MLC) 1/3 locus, which has been well characterised in transgenic mouse studies. This locus contains two promoters encoding two alternatively spliced isoforms of alkali myosin light chain. These promoters are differentially regulated during development through the activity of two enhancer elements. The MLC3 promoter alone has been shown to confer expression of a reporter gene in skeletal and cardiac muscle in transgenic mice and the addition of the downstream MLC enhancer increased expression levels in skeletal muscle. We asked whether this regulatory module, sufficient for striated muscle gene expression in the mouse, would drive expression in similar domains in the chicken. RESULTS: We have observed that a conserved downstream MLC enhancer is present in the chicken MLC locus. We found that the rat MLC1/3 regulatory elements were transcriptionally active in chick skeletal muscle primary cultures. We observed that a single copy lentiviral insert containing this regulatory cassette was able to drive expression of a lacZ reporter gene in the fast-fibres of skeletal muscle in chicken in three independent transgenic chicken lines in a pattern similar to the endogenous MLC locus. Reporter gene expression in cardiac muscle tissues was not observed for any of these lines. CONCLUSIONS: From these results we conclude that skeletal expression from this regulatory module is conserved in a genomic context between rodents and chickens. This transgenic module will be useful in future investigations of muscle development in avian species.


Assuntos
Regulação da Expressão Gênica , Músculo Esquelético/metabolismo , Animais , Animais Geneticamente Modificados , Sequência de Bases , Células Cultivadas , Galinhas , Elementos Facilitadores Genéticos , Humanos , Camundongos , Camundongos Transgênicos , Dados de Sequência Molecular , Cadeias Leves de Miosina/genética , Regiões Promotoras Genéticas , Ratos , Alinhamento de Sequência
18.
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
19.
Nat Cell Biol ; 21(11): 1334-1345, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31685991

RESUMO

It is well established that haematopoietic stem and progenitor cells (HSPCs) are generated from a transient subset of specialized endothelial cells termed haemogenic, present in the yolk sac, placenta and aorta, through an endothelial-to-haematopoietic transition (EHT). HSPC generation via EHT is thought to be restricted to the early stages of development. By using experimental embryology and genetic approaches in birds and mice, respectively, we document here the discovery of a bone marrow haemogenic endothelium in the late fetus/young adult. These cells are capable of de novo producing a cohort of HSPCs in situ that harbour a very specific molecular signature close to that of aortic endothelial cells undergoing EHT or their immediate progenies, i.e., recently emerged HSPCs. Taken together, our results reveal that HSPCs can be generated de novo past embryonic stages. Understanding the molecular events controlling this production will be critical for devising innovative therapies.


Assuntos
Células da Medula Óssea/metabolismo , Linhagem da Célula/genética , Regulação da Expressão Gênica no Desenvolvimento , Hemangioblastos/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Animais , Animais Geneticamente Modificados , Aorta/citologia , Aorta/metabolismo , Células da Medula Óssea/citologia , Diferenciação Celular , Galinhas , Embrião de Mamíferos , Embrião não Mamífero , Feminino , Feto , Perfilação da Expressão Gênica , Redes Reguladoras de Genes , Hemangioblastos/citologia , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/citologia , Heterozigoto , Homozigoto , Masculino , Camundongos , Gravidez , Saco Vitelino/citologia , Saco Vitelino/crescimento & desenvolvimento , Saco Vitelino/metabolismo
20.
Elife ; 82019 06 04.
Artigo em Inglês | MEDLINE | ID: mdl-31159925

RESUMO

Influenza A viruses (IAV) are subject to species barriers that prevent frequent zoonotic transmission and pandemics. One of these barriers is the poor activity of avian IAV polymerases in human cells. Differences between avian and mammalian ANP32 proteins underlie this host range barrier. Human ANP32A and ANP32B homologues both support function of human-adapted influenza polymerase but do not support efficient activity of avian IAV polymerase which requires avian ANP32A. We show here that the gene currently designated as avian ANP32B is evolutionarily distinct from mammalian ANP32B, and that chicken ANP32B does not support IAV polymerase activity even of human-adapted viruses. Consequently, IAV relies solely on chicken ANP32A to support its replication in chicken cells. Amino acids 129I and 130N, accounted for the inactivity of chicken ANP32B. Transfer of these residues to chicken ANP32A abolished support of IAV polymerase. Understanding ANP32 function will help develop antiviral strategies and aid the design of influenza virus resilient genome edited chickens.


Assuntos
Especificidade de Hospedeiro , Interações Hospedeiro-Patógeno , Vírus da Influenza A/crescimento & desenvolvimento , Proteínas Nucleares/metabolismo , Proteínas de Ligação a RNA/metabolismo , Animais , Linhagem Celular , Galinhas , Humanos , Vírus da Influenza A/enzimologia , RNA Polimerase Dependente de RNA/metabolismo , Replicação Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA