Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Marie-Cecile van de Lavoir is active.

Publication


Featured researches published by Marie-Cecile van de Lavoir.


Nature | 2006

Germline transmission of genetically modified primordial germ cells

Marie-Cecile van de Lavoir; Jennifer H. Diamond; Philip A. Leighton; Christine Mather-Love; Babette Heyer; Renee Bradshaw; Allyn Kerchner; Lisa T. Hooi; Terri M. Gessaro; Susan E. Swanberg; Mary E. Delany; Robert J. Etches

Primordial germ cells (PGCs) are the precursors of sperm and eggs. In most animals, segregation of the germ line from the somatic lineages is one of the earliest events in development; in avian embryos, PGCs are first identified in an extra-embryonic region, the germinal crescent, after approximately 18 h of incubation. After 50–55 h of development, PGCs migrate to the gonad and subsequently produce functional sperm and oocytes. So far, cultures of PGCs that remain restricted to the germ line have not been reported in any species. Here we show that chicken PGCs can be isolated, cultured and genetically modified while maintaining their commitment to the germ line. Furthermore, we show that chicken PGCs can be induced in vitro to differentiate into embryonic germ cells that contribute to somatic tissues. Retention of the commitment of PGCs to the germ line after extended periods in culture and after genetic modification combined with their capacity to acquire somatic competence in vitro provides a new model for developmental biology. The utility of the model is enhanced by the accessibility of the avian embryo, which facilitates access to the earliest stages of development and supplies a facile route for the reintroduction of PGCs into the embryonic vasculature. In addition, these attributes create new opportunities to manipulate the genome of chickens for agricultural and pharmaceutical applications.


Nature Biotechnology | 2005

Production of human monoclonal antibody in eggs of chimeric chickens

Lei Zhu; Marie-Cecile van de Lavoir; Jenny Albanese; David O. Beenhouwer; Pina M. Cardarelli; Severino Cuison; David F Deng; Shrikant Deshpande; Jennifer H. Diamond; Lynae Green; Edward L. Halk; Babette Heyer; Robert M. Kay; Allyn Kerchner; Philip Albert Leighton; Christine Mather; Sherie L. Morrison; Zivko L. Nikolov; David Passmore; Alicia Pradas-Monne; Benjamin T. Preston; Vangipuram S Rangan; Mingxia Shi; Mohan Srinivasan; Steven G White; Peggy Winters-Digiacinto; Susan Wong; Wen Zhou; Robert J. Etches

The tubular gland of the chicken oviduct is an attractive system for protein expression as large quantities of proteins are deposited in the egg, the production of eggs is easily scalable and good manufacturing practices for therapeutics from eggs have been established. Here we examined the ability of upstream and downstream DNA sequences of ovalbumin, a protein produced exclusively in very high quantities in chicken egg white, to drive tissue-specific expression of human mAb in chicken eggs. To accommodate these large regulatory regions, we established and transfected lines of chicken embryonic stem (cES) cells and formed chimeras that express mAb from cES cell–derived tubular gland cells. Eggs from high-grade chimeras contained up to 3 mg of mAb that possesses enhanced antibody-dependent cellular cytotoxicity (ADCC), nonantigenic glycosylation, acceptable half-life, excellent antigen recognition and good rates of internalization.


Mechanisms of Development | 2006

High-grade transgenic somatic chimeras from chicken embryonic stem cells.

Marie-Cecile van de Lavoir; Christine Mather-Love; Philip A. Leighton; Jennifer H. Diamond; Babette Heyer; Rhys Roberts; Lei Zhu; Peggy Winters-Digiacinto; Allyn Kerchner; Terri M. Gessaro; Susan E. Swanberg; Mary E. Delany; Robert J. Etches

Male and female embryonic stem (ES) cell lines were derived from the area pellucidae of Stage X (EG&K) chicken embryos. These ES cell lines were grown in culture for extended periods of time and the majority of the cells retained a diploid karyotype. When reintroduced into Stage VI-X (EG&K) recipient embryos, the cES cells were able to contribute to all somatic tissues. By combining irradiation of the recipient embryo with exposure of the cES cells to the embryonic environment in diapause, a high frequency and extent of chimerism was obtained. High-grade chimeras, indistinguishable from the donor phenotype by feather pigmentation, were produced. A transgene encoding GFP was incorporated into the genome of cES cells under control of the ubiquitous promoter CX and GFP was widely expressed in somatic tissues. Although cES cells made extensive contributions to the somatic tissues, contribution to the germline was not observed.


Proceedings of the National Academy of Sciences of the United States of America | 2013

Immunoglobulin knockout chickens via efficient homologous recombination in primordial germ cells

Benjamin Schusser; Ellen J. Collarini; Henry Yi; Shelley Izquierdo; Jeffrey Fesler; Darlene Pedersen; Kirk C. Klasing; Bernd Kaspers; William Harriman; Marie-Cecile van de Lavoir; Robert J. Etches; Philip A. Leighton

Significance Chickens have been an important animal model in the fields of developmental biology and immunology over the last century and have contributed a number of basic findings in these areas. However, their use has been limited more recently because there has been no way to genetically engineer specific mutations in the chicken. In addition to basic research applications, genetic modification in birds could also be used to mitigate the threat of avian influenza by production of flu-resistant bird stocks in agriculture. Here we describe an efficient way to target genes of interest in the chicken genome and create knockout chickens. We targeted the immunoglobulin heavy chain gene, leading to loss of antibody production and a block in B-cell development. 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.


Molecular Reproduction and Development | 2008

Genetic modification of primordial germ cells by gene trapping, gene targeting, and ϕC31 integrase

Philip A. Leighton; Marie-Cecile van de Lavoir; Jennifer H. Diamond; Chunyao Xia; Robert J. Etches

The genome of germline committed cells is thought to be protected by mechanisms of transcriptional silencing, posing a barrier to transgenesis using cultured germline cells. We found that selection for transgene integration into the primordial germ cell genome required that the transgenes be flanked by the chicken β‐globin insulator. However, integration frequency was low, and sequencing of the insertion sites revealed that the transgenes preferentially inserted into active promoter regions, implying that silencing prohibited recovery of insertions in other regions. Much higher frequencies of integration were achieved when the ϕC31 integrase was used to insert transgenes into endogenous pseudo attP sites. Despite the evidence for transcriptional silencing in PGCs, gene targeting of a nonexpressed gene was also achieved. The ability to make genetic modifications in PGCs provides unprecedented opportunities to study the biology of PGCs, as well as produce transgenic chickens for applications in biotechnology and developmental biology. Mol. Reprod. Dev. 75: 1163–1175, 2008.


Biology of Reproduction | 2014

Characteristics of Long-Term Cultures of Avian Primordial Germ Cells and Gonocytes

Yonghong Song; Sivakumar Duraisamy; Jahabar Ali; Jaleel Kizhakkayil; Victor Doss Jacob; Mohammed Ahmed Mohammed; Mohammed A. Eltigani; Suresh Amisetty; Manoj Kumar Shukla; Robert J. Etches; Marie-Cecile van de Lavoir

ABSTRACT Avian cell lines derived from germinal crescent primordial germ cells and gonadal gonocytes with long-term proliferative capacity in vitro and their subsequent rates of colonization and germline transmission are described. In general, male cultures proliferate more rapidly than female cultures although both can be developed into cell lines of >2 × 106 cells, at which time, they can be grown indefinitely and a cell bank can be established. All the cell lines injected into embryos transmitted through the germline with the percentage of germline transmission of both male and female cell lines varying from single digits to the high 90s. The derivation of these primordial germ cell and gonadal cell lines and the subsequent robustness of germline transmission validates these cells as suitable for establishment of lines of chickens bearing novel genetic modifications.


PLOS ONE | 2016

Germline Gene Editing in Chickens by Efficient CRISPR-Mediated Homologous Recombination in Primordial Germ Cells

Lazar Dimitrov; Darlene Pedersen; Kathryn H. Ching; Henry Yi; Ellen J. Collarini; Shelley Izquierdo; Marie-Cecile van de Lavoir; Philip A. Leighton

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.


PLOS ONE | 2012

Interspecific germline transmission of cultured primordial germ cells.

Marie-Cecile van de Lavoir; Ellen J. Collarini; Philip A. Leighton; Jeffrey Fesler; Daniel R. Lu; William D. Harriman; T. S. Thiyagasundaram; Robert J. Etches

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.


European Journal of Immunology | 2016

Expression of heavy chain-only antibodies can support B-cell development in light chain knockout chickens.

Benjamin Schusser; Ellen J. Collarini; Darlene Pedersen; Henry Yi; Kathryn H. Ching; Shelley Izquierdo; Theresa Thoma; Sarah Lettmann; Bernd Kaspers; Robert J. Etches; Marie-Cecile van de Lavoir; William D. Harriman; Philip A. Leighton

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.


mAbs | 2018

Chickens with humanized immunoglobulin genes generate antibodies with high affinity and broad epitope coverage to conserved targets

Kathryn H. Ching; Ellen J. Collarini; Yasmina Noubia Abdiche; Daniel Bedinger; Darlene Pedersen; Shelley Izquierdo; Rian Harriman; Lei Zhu; Robert J. Etches; Marie-Cecile van de Lavoir; William D. Harriman; Philip A. Leighton

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.

Collaboration


Dive into the Marie-Cecile van de Lavoir's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Philip A. Leighton

Howard Hughes Medical Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kathryn H. Ching

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Lei Zhu

Princeton University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mary E. Delany

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge