Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Methods Mol Biol ; 1874: 403-430, 2019.
Article in English | MEDLINE | ID: mdl-30353528

ABSTRACT

The unique characteristics of the avian embryo, with its large opaque yolk, have necessitated the development of different approaches to transgenesis from those that have been successful in mammalian species. Genetic modification of birds was greatly advanced by the ability to grow long-term cultures of primordial germ cells (PGCs). These cells are obtained from embryos, established in culture, and can be propagated without losing the ability to contribute to the germline when reintroduced into a host animal. PGCs can be genetically modified in culture using traditional transfection and selection techniques, including gene targeting and site-specific nuclease approaches. Here, we describe our methods for deriving cell lines, long-term culture, genetic modification, production of germline chimeras and obtaining fully transgenic birds with the desired genetic modifications.


Subject(s)
Animals, Genetically Modified/growth & development , Chickens/genetics , Chimera/growth & development , Germ Cells/cytology , Animals , Cell Line , Cells, Cultured , Chickens/growth & development , Coculture Techniques , Female , Gene Transfer Techniques , Germ Cells/metabolism , Male , Rats
2.
MAbs ; 10(1): 71-80, 2018 01.
Article in English | MEDLINE | ID: mdl-29035625

ABSTRACT

Transgenic animal platforms for the discovery of human monoclonal antibodies have been developed in mice, rats, rabbits and cows. The immune response to human proteins is limited in these animals by their tolerance to mammalian-conserved epitopes. To expand the range of epitopes that are accessible, we have chosen an animal host that is less phylogenetically related to humans. Specifically, we generated transgenic chickens expressing antibodies from immunoglobulin heavy and light chain loci containing human variable regions and chicken constant regions. From these birds, paired human light and heavy chain variable regions are recovered and cloned as fully human recombinant antibodies. The human antibody-expressing chickens exhibit normal B cell development and raise immune responses to conserved human proteins that are not immunogenic in mice. Fully human monoclonal antibodies can be recovered with sub-nanomolar affinities. Binning data of antibodies to a human protein show epitope coverage similar to wild type chickens, which we previously showed is broader than that produced from rodent immunizations.


Subject(s)
Antibodies, Monoclonal, Humanized/biosynthesis , Antibodies, Monoclonal, Humanized/immunology , Antibody Affinity , Antibody Specificity , Antigens/immunology , Chickens/immunology , Epitopes/immunology , Immunoglobulins/immunology , Animals , Animals, Genetically Modified , Antigens/administration & dosage , B-Lymphocytes/immunology , Chickens/blood , Chickens/genetics , Epitope Mapping , Humans , Immunization , Immunoglobulins/blood , Immunoglobulins/genetics , Species Specificity , T-Lymphocytes/immunology
3.
Eur J Immunol ; 46(9): 2137-48, 2016 09.
Article in English | MEDLINE | ID: mdl-27392810

ABSTRACT

Since the discovery of antibody-producing B cells in chickens six decades ago, chickens have been a model for B-cell development in gut-associated lymphoid tissue species. Here we describe targeting of the immunoglobulin light chain locus by homologous recombination in chicken primordial germ cells (PGCs) and generation of VJCL knockout chickens. In contrast to immunoglobulin heavy chain knockout chickens, which completely lack mature B cells, homozygous light chain knockout (IgL(-/-) ) chickens have a small population of B lineage cells that develop in the bursa and migrate to the periphery. This population of B cells expresses the immunoglobulin heavy chain molecule on the cell surface. Soluble heavy-chain-only IgM and IgY proteins of reduced molecular weight were detectable in plasma in 4-week-old IgL(-/-) chickens, and antigen-specific IgM and IgY heavy chain proteins were produced in response to immunization. Circulating heavy-chain-only IgM showed a deletion of the CH1 domain of the constant region enabling the immunoglobulin heavy chain to be secreted in the absence of the light chain. Our data suggest that the heavy chain by itself is enough to support all the important steps in B-cell development in a gut-associated lymphoid tissue species.


Subject(s)
Antibodies/genetics , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Gene Expression , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Light Chains/genetics , Animals , Animals, Genetically Modified , Antibodies/immunology , Antibody Formation/genetics , Antibody Formation/immunology , Cell Differentiation/genetics , Cell Differentiation/immunology , Chickens , Gene Deletion , Gene Knockout Techniques , Gene Order , Gene Targeting , Genetic Vectors/genetics , Immunoglobulin Light Chains/chemistry , Plasma Cells/immunology , Plasma Cells/metabolism , Protein Domains/genetics
4.
PLoS One ; 11(4): e0154303, 2016.
Article in English | MEDLINE | ID: mdl-27099923

ABSTRACT

The CRISPR/Cas9 system has been applied in a large number of animal and plant species for genome editing. In chickens, CRISPR has been used to knockout genes in somatic tissues, but no CRISPR-mediated germline modification has yet been reported. Here we use CRISPR to target the chicken immunoglobulin heavy chain locus in primordial germ cells (PGCs) to produce transgenic progeny. Guide RNAs were co-transfected with a donor vector for homology-directed repair of the double-strand break, and clonal populations were selected. All of the resulting drug-resistant clones contained the correct targeting event. The targeted cells gave rise to healthy progeny containing the CRISPR-targeted locus. The results show that gene-edited chickens can be obtained by modifying PGCs in vitro with the CRISPR/Cas9 system, opening up many potential applications for efficient genetic modification in birds.


Subject(s)
CRISPR-Cas Systems , Chickens/genetics , Gene Editing/methods , Genome , Homologous Recombination , Immunoglobulin Heavy Chains/genetics , Animals , Animals, Genetically Modified , Base Sequence , Chickens/growth & development , Cloning, Organism , Embryo, Nonmammalian , Female , Gene Knockout Techniques , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Germ Cells , Green Fluorescent Proteins/deficiency , Green Fluorescent Proteins/genetics , Male , RNA, Guide, Kinetoplastida/genetics , RNA, Guide, Kinetoplastida/metabolism
5.
Microscopy (Oxf) ; 65(4): 341-52, 2016 08.
Article in English | MEDLINE | ID: mdl-27107009

ABSTRACT

The analysis of secreted antibody from large and diverse populations of B cells in parallel at the clonal level can reveal desirable antibodies for diagnostic or therapeutic applications. By immobilizing B cells in microdroplets with particulate reporters, decoding and isolating them in a microscopy environment, we have recovered panels of antibodies with rare attributes to therapeutically relevant targets. The ability to screen up to 100 million cells in a single experiment can be fully leveraged by accessing primary B-cell populations from evolutionarily divergent species such as chickens.


Subject(s)
Antibodies, Monoclonal/immunology , B-Lymphocytes/metabolism , Hybridomas/immunology , Receptors, CCR5/immunology , Receptors, Purinergic P2X3/immunology , Receptors, TNF-Related Apoptosis-Inducing Ligand/immunology , Animals , Antibodies, Monoclonal/therapeutic use , B-Lymphocytes/immunology , CHO Cells , Cell Line, Tumor , Chickens , Cricetulus , Drug Discovery/methods , Humans , Hybridomas/metabolism , Jurkat Cells , Spleen/cytology
6.
Transgenic Res ; 25(5): 609-16, 2016 10.
Article in English | MEDLINE | ID: mdl-27034267

ABSTRACT

Cre recombinase has been extensively used for genome engineering in transgenic mice yet its use in other species has been more limited. Here we describe the generation of transgenic chickens expressing Cre recombinase. Green fluorescent protein (GFP)-positive chicken primordial germ cells were stably transfected with ß-actin-Cre-recombinase using phiC31 integrase and transgenic chickens were generated. Cre recombinase activity was verified by mating Cre birds to birds carrying a floxed transgene. Floxed sequences were only excised in offspring from roosters that inherited the Cre recombinase but were excised in all offspring from hens carrying the Cre recombinase irrespective of the presence of the Cre transgene. The Cre recombinase transgenic birds were healthy and reproductively normal. The Cre and GFP genes in two of the lines were closely linked whereas the genes segregated independently in a third line. These founders allowed development of GFP-expressing and non-GFP-expressing Cre recombinase lines. These lines of birds create a myriad of opportunities to study developmentally-regulated and tissue-specific expression of transgenes in chickens.


Subject(s)
Chickens/genetics , Integrases/genetics , Recombination, Genetic , Animals , Animals, Genetically Modified , Gene Expression Regulation , Green Fluorescent Proteins , Organ Specificity , Promoter Regions, Genetic , Transgenes
7.
PLoS One ; 8(11): e80108, 2013.
Article in English | MEDLINE | ID: mdl-24278246

ABSTRACT

Transgenic chickens expressing human sequence antibodies would be a powerful tool to access human targets and epitopes that have been intractable in mammalian hosts because of tolerance to conserved proteins. To foster the development of the chicken platform, it is beneficial to validate transgene constructs using a rapid, cell culture-based method prior to generating fully transgenic birds. We describe a method for the expression of human immunoglobulin variable regions in the chicken DT40 B cell line and the further diversification of these genes by gene conversion. Chicken VL and VH loci were knocked out in DT40 cells and replaced with human VK and VH genes. To achieve gene conversion of human genes in chicken B cells, synthetic human pseudogene arrays were inserted upstream of the functional human VK and VH regions. Proper expression of chimeric IgM comprised of human variable regions and chicken constant regions is shown. Most importantly, sequencing of DT40 genetic variants confirmed that the human pseudogene arrays contributed to the generation of diversity through gene conversion at both the Igl and Igh loci. These data show that engineered pseudogene arrays produce a diverse pool of human antibody sequences in chicken B cells, and suggest that these constructs will express a functional repertoire of chimeric antibodies in transgenic chickens.


Subject(s)
Antibodies/genetics , B-Lymphocytes/metabolism , Chickens/genetics , Gene Conversion , Amino Acid Sequence , Animals , Animals, Genetically Modified , Antibodies/chemistry , Base Sequence , Blotting, Western , Cell Line , DNA Primers , Flow Cytometry , Humans , Molecular Sequence Data , Sequence Homology, Amino Acid
8.
Proc Natl Acad Sci U S A ; 110(50): 20170-5, 2013 Dec 10.
Article in English | MEDLINE | ID: mdl-24282302

ABSTRACT

Gene targeting by homologous recombination or by sequence-specific nucleases allows the precise modification of genomes and genes to elucidate their functions. Although gene targeting has been used extensively to modify the genomes of mammals, fish, and amphibians, a targeting technology has not been available for the avian genome. Many of the principles of humoral immunity were discovered in chickens, yet the lack of gene targeting technologies in birds has limited biomedical research using this species. Here we describe targeting the joining (J) gene segment of the chicken Ig heavy chain gene by homologous recombination in primordial germ cells to establish fully transgenic chickens carrying the knockout. In homozygous knockouts, Ig heavy chain production is eliminated, and no antibody response is elicited on immunization. Migration of B-lineage precursors into the bursa of Fabricius is unaffected, whereas development into mature B cells and migration from the bursa are blocked in the mutants. Other cell types in the immune system appear normal. Chickens lacking the peripheral B-cell population will provide a unique experimental model to study avian immune responses to infectious disease. More generally, gene targeting in avian primordial germ cells will foster advances in diverse fields of biomedical research such as virology, stem cells, and developmental biology, and provide unique approaches in biotechnology, particularly in the field of antibody discovery.


Subject(s)
B-Lymphocytes/cytology , Chickens/genetics , Gene Knockout Techniques/methods , Genetic Engineering/methods , Germ Cells/chemistry , Immunoglobulin Heavy Chains/genetics , Animals , B-Lymphocytes/metabolism , Blotting, Southern , Chickens/immunology , DNA Methylation , DNA Primers/genetics , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Genotype , Germ Cells/metabolism , Immunohistochemistry
9.
PLoS One ; 7(5): e35664, 2012.
Article in English | MEDLINE | ID: mdl-22629301

ABSTRACT

In birds, the primordial germ cell (PGC) lineage separates from the soma within 24 h following fertilization. Here we show that the endogenous population of about 200 PGCs from a single chicken embryo can be expanded one million fold in culture. When cultured PGCs are injected into a xenogeneic embryo at an equivalent stage of development, they colonize the testis. At sexual maturity, these donor PGCs undergo spermatogenesis in the xenogeneic host and become functional sperm. Insemination of semen from the xenogeneic host into females from the donor species produces normal offspring from the donor species. In our model system, the donor species is chicken (Gallus domesticus) and the recipient species is guinea fowl (Numida meleagris), a member of a different avian family, suggesting that the mechanisms controlling proliferation of the germline are highly conserved within birds. From a pragmatic perspective, these data are the basis of a novel strategy to produce endangered species of birds using domesticated hosts that are both tractable and fecund.


Subject(s)
Chimera/genetics , Galliformes/genetics , Germ Cells/cytology , Animals , Cell Line , Cells, Cultured , Female , Male
10.
J Immunol ; 183(10): 6338-45, 2009 Nov 15.
Article in English | MEDLINE | ID: mdl-19841167

ABSTRACT

Native human Abs represent attractive drug candidates; however, the low frequency of B cells expressing high-quality Abs has posed a barrier to discovery. Using a novel single-cell phenotyping technology, we have overcome this barrier to discover human Abs targeting the conserved but poorly immunogenic central motif of respiratory syncytial virus (RSV) G protein. For the entire cohort of 24 subjects with recent RSV infection, B cells producing Abs meeting these stringent specificity criteria were rare, <10 per million. Several of the newly cloned Abs bind to the RSV G protein central conserved motif with very high affinity (K(d) 1-24 pM). Two of the Abs were characterized in detail and compared with palivizumab, a humanized mAb against the RSV F protein. Relative to palivizumab, the anti-G Abs showed improved viral neutralization potency in vitro and enhanced reduction of infectious virus in a prophylaxis mouse model. Furthermore, in a mouse model for postinfection treatment, both anti-G Abs were significantly more effective than palivizumab at reducing viral load. The combination of activity in mouse models for both prophylaxis and treatment makes these high-affinity human-derived Abs promising candidates for human clinical testing.


Subject(s)
Antibodies, Viral/therapeutic use , B-Lymphocytes/immunology , Respiratory Syncytial Virus Infections/therapy , Respiratory Syncytial Virus, Human/immunology , Viral Fusion Proteins/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Antibodies, Viral/immunology , Antibody Affinity/immunology , Antigens, Viral/immunology , Antigens, Viral/metabolism , B-Lymphocytes/virology , Cell Line , Humans , Mice , Neutralization Tests , Palivizumab , Recombinant Proteins/immunology , Respiratory Syncytial Virus Infections/prevention & control , Transfection , Viral Load/drug effects , Viral Load/immunology
11.
J Immunol Methods ; 341(1-2): 127-34, 2009 Feb 28.
Article in English | MEDLINE | ID: mdl-19084532

ABSTRACT

Micron scale latex beads are well established as highly biocompatible reagents. Imbibing two fluorescent dyes into the interior of the beads enables the creation of a family of combinatorially colored labels. Previous use of such beads, in flow cytometry for example, has focused on beads of approximately 5 microm diameter. We show here that 280 nm combinatorially labeled particles can be used to create ELISA-style assays in 200 microm scale virtual wells, using digital microscopy as the readout. The utility of this technique is illustrated by profiling the secreted cytokine footprints of peripheral blood mononuclear cells in a multiparametric version of the popular Elispot assay. Doing so reveals noncanonical classes of T lymphocytes. We further show that the secreting cell type can be concurrently identified by surface staining with a cell type specific antibody conjugated to the same multiplexed beads.


Subject(s)
Antibodies/chemistry , Flow Cytometry/methods , Immunoassay/methods , Microspheres , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Antibodies/immunology , Cytokines/analysis , Cytokines/immunology , Humans
12.
J Immunol Methods ; 341(1-2): 135-45, 2009 Feb 28.
Article in English | MEDLINE | ID: mdl-19087879

ABSTRACT

The secreted immunoglobulin footprint of single hybridoma cells, containing ~10 fg of antibody purified in situ, has been probed for 9 properties concurrently by use of detection labels comprising 280 nm combinatorially colored fluorescent latex beads functionalized with proteins. Specificity of each individual hybridoma cell's product has thereby been assessed in a primary screen. Varying the density of antigen on beads to modulate the avidity of the interaction between bead and secreted antibody footprint allowed rank ordering by affinity in the same primary screen. As more criteria were added to the selection process, the frequency of positive cells went down; in some cases, the favorable cell was present at <1/50,000. Recovery of the cell of interest was accomplished by plating the cells in a viscous medium on top of a membrane. After collecting the antibody footprint on a capture surface beneath the membrane, the immobilized cells were transferred to an incubator while the footprints were analyzed to locate the hybridoma cells of interest. The desired cells were then cloned by picking them from the corresponding locations on the membrane.


Subject(s)
Antibody Affinity/immunology , Antibody Specificity/immunology , Hybridomas/cytology , Hybridomas/immunology , Immunoassay/methods , Immunoglobulins/immunology , Animals , Hybridomas/metabolism , Immunoglobulins/metabolism , Mice , Mice, Inbred BALB C , Microspheres
SELECTION OF CITATIONS
SEARCH DETAIL
...