Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Immunol ; 212(11): 1744-1753, 2024 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-38629917

RESUMO

H chain-only Igs are naturally produced in camelids and sharks. Because these Abs lack the L chain, the Ag-binding domain is half the size of a traditional Ab, allowing this type of Ig to bind to targets in novel ways. Consequently, the H chain-only single-domain Ab (sdAb) structure has the potential to increase the repertoire and functional range of an active humoral immune system. The majority of vertebrates use the standard heterodimeric (both H and L chains) structure and do not produce sdAb format Igs. To investigate if other animals are able to support sdAb development and function, transgenic chickens (Gallus gallus) were designed to produce H chain-only Abs by omitting the L chain V region and maintaining only the LC region to serve as a chaperone for Ab secretion from the cell. These birds produced 30-50% normal B cell populations within PBMCs and readily expressed chicken sequence sdAbs. Interestingly, the H chains contained a spontaneous CH1 deletion. Although no isotype switching to IgY or IgA occurred, the IgM repertoire was diverse, and immunization with a variety of protein immunogens rapidly produced high and specific serum titers. mAbs of high affinity were efficiently recovered by single B cell screening. In in vitro functional assays, the sdAbs produced by birds immunized against SARS-CoV-2 were also able to strongly neutralize and prevent viral replication. These data suggest that the truncated L chain design successfully supported sdAb development and expression in chickens.


Assuntos
Animais Geneticamente Modificados , Galinhas , Cadeias Pesadas de Imunoglobulinas , Anticorpos de Domínio Único , Animais , Galinhas/imunologia , Anticorpos de Domínio Único/imunologia , Anticorpos de Domínio Único/genética , Cadeias Pesadas de Imunoglobulinas/genética , Cadeias Pesadas de Imunoglobulinas/imunologia , SARS-CoV-2/imunologia , SARS-CoV-2/genética , COVID-19/imunologia , Transgenes/genética , Linfócitos B/imunologia , Anticorpos Antivirais/imunologia , Cadeias Leves de Imunoglobulina/genética , Cadeias Leves de Imunoglobulina/imunologia , Humanos
2.
MAbs ; 13(1): 1862451, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33491549

RESUMO

Bispecific antibodies are an important and growing segment in antibody therapeutics, particularly in the immuno-oncology space. Manufacturing of a bispecific antibody with two different heavy chains is greatly simplified if the light chains can be the same for both arms of the antibody. Here, we introduce a strain of common light chain chickens, called OmniClic®, that produces antibody repertoires largely devoid of light chain diversity. The antibody repertoire in these chickens is composed of diverse human heavy chain variable regions capable of high-affinity antigen-specific binding and broad epitope diversity when paired with the germline human kappa light chain. OmniClic birds can be used in immunization campaigns for discovery of human heavy chains to different targets. Subsequent pairing of the heavy chain with a germline human kappa light chain serves to facilitate bispecific antibody production by increasing the efficiency of correct pairing. Abbreviations: AID: activation-induced cytidine deaminase; bsAb: bispecific antibody; CDR: complementarity-determining region; CL: light chain constant region; CmLC: common light chain; D: diversity region; ELISA: enzyme-linked immunosorbent assay; FACS: fluorescence-activated cell sorting; Fc: fragment crystallizable; FcRn: neonatal Fc receptor; FR: framework region; GEM: gel-encapsulated microenvironment; Ig: immunoglobulin; IMGT: the international ImMunoGeneTics information system®; J: joining region; KO: knockout; mAb: monoclonal antibody; NGS: next-generation sequencing; PBS: phosphate-buffered saline; PCR: polymerase chain reaction; PGC: primordial germ cell; PGRN: progranulin; TCR: T cell receptor; V: variable region; VK: kappa light chain variable region; VL: light chain variable region; VH: heavy chain variable region.


Assuntos
Anticorpos Biespecíficos/imunologia , Anticorpos Monoclonais/imunologia , Afinidade de Anticorpos/imunologia , Galinhas/imunologia , Epitopos/imunologia , Cadeias Leves de Imunoglobulina/imunologia , Animais , Antígenos/imunologia , Linfócitos B/imunologia , Linfócitos B/metabolismo , Citometria de Fluxo/métodos , Humanos , Imunização/métodos , Cadeias Pesadas de Imunoglobulinas/imunologia , Cadeias kappa de Imunoglobulina/imunologia , Engenharia de Proteínas/métodos
3.
PLoS One ; 15(1): e0228164, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31995598

RESUMO

Most of the approved monoclonal antibodies used in the clinic were initially discovered in mice. However, many targets of therapeutic interest are highly conserved proteins that do not elicit a robust immune response in mice. There is a need for non-mammalian antibody discovery platforms which would allow researchers to access epitopes that are not recognized in mammalian hosts. Recently, we introduced the OmniChicken®, a transgenic animal carrying human VH3-23 and VK3-15 at its immunoglobulin loci. Here, we describe a new version of the OmniChicken which carries VH3-23 and either VL1-44 or VL3-19 at its heavy and light chain loci, respectively. The Vλ-expressing birds showed normal B and T populations in the periphery. A panel of monoclonal antibodies demonstrated comparable epitope coverage of a model antigen compared to both wild-type and Vκ-expressing OmniChickens. Kinetic analysis identified binders in the picomolar range. The Vλ-expressing bird increases the antibody diversity available in the OmniChicken platform, further enabling discovery of therapeutic leads.


Assuntos
Animais Geneticamente Modificados/genética , Galinhas/genética , Cadeias lambda de Imunoglobulina/genética , Animais , Animais Geneticamente Modificados/imunologia , Anticorpos Monoclonais/genética , Anticorpos Monoclonais/imunologia , Linfócitos B/imunologia , Galinhas/imunologia , Humanos , Imunidade Humoral , Cadeias Pesadas de Imunoglobulinas/genética , Região Variável de Imunoglobulina/genética , Cadeias lambda de Imunoglobulina/imunologia , Progranulinas/imunologia , Linfócitos T/imunologia , Transgenes/genética
4.
MAbs ; 11(6): 1036-1052, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31257988

RESUMO

Targeting the CD47-signal-regulatory protein α (SIRPα) pathway represents a novel therapeutic approach to enhance anti-cancer immunity by promoting both innate and adaptive immune responses. Unlike CD47, which is expressed ubiquitously, SIRPα expression is mainly restricted to myeloid cells and neurons. Therefore, compared to CD47-targeted therapies, targeting SIRPα may result in differential safety and efficacy profiles, potentially enabling lower effective doses and improved pharmacokinetics and pharmacodynamics. The development of effective SIRPα antagonists is restricted by polymorphisms within the CD47-binding domain of SIRPα, necessitating pan-allele reactive anti-SIRPα antibodies for therapeutic intervention in diverse patient populations. We immunized wild-type and human antibody transgenic chickens with a multi-allele and multi-species SIRPα regimen in order to discover pan-allelic and pan-mammalian reactive anti-SIRPα antibodies suitable for clinical translation. A total of 200 antibodies were isolated and screened for SIRPα reactivity from which approximately 70 antibodies with diverse SIRPα binding profiles, sequence families, and epitopes were selected for further characterization. A subset of anti-SIRPα antibodies bound to both human SIRPα v1 and v2 alleles with high affinity ranging from low nanomolar to picomolar, potently antagonized the CD47/SIRPα interaction, and potentiated macrophage-mediated antibody-dependent cellular phagocytosis in vitro. X-ray crystal structures of five anti-SIRPα antigen-binding fragments, each with unique epitopes, in complex with SIRPα (PDB codes 6NMV, 6NMU, 6NMT, 6NMS, and 6NMR) are reported. Furthermore, some of the anti-SIRPα antibodies cross-react with cynomolgus SIRPα and various mouse SIRPα alleles (BALB/c, NOD, BL/6), which can facilitate preclinical to clinical development. These properties provide an attractive rationale to advance the development of these anti-SIRPα antibodies as a novel therapy for advanced malignancies. Abbreviations: ADCC: antibody-dependent cellular cytotoxicity; ADCP: antibody-dependent cellular phagocytosis; CFSE: carboxyfluorescein succinimidyl ester; Fab: fragment antigen binding; Fc: fragment crystallizable; FcγR: Fcγ receptor; Ig: immunoglobulin; IND: investigational new drug; MDM⊘: monocyte-derived macrophage; NOD: non-obese diabetic; scFv: single chain fragment variable; SCID: severe combined immunodeficiency; SIRP: signal-regulatory protein.


Assuntos
Anticorpos Monoclonais , Especificidade de Anticorpos , Antígenos de Diferenciação , Receptores Imunológicos , Animais , Anticorpos Monoclonais/química , Anticorpos Monoclonais/genética , Anticorpos Monoclonais/imunologia , Antígenos de Diferenciação/química , Antígenos de Diferenciação/imunologia , Antígeno CD47/imunologia , Galinhas , Cristalografia por Raios X , Epitopos/química , Epitopos/genética , Epitopos/imunologia , Feminino , Humanos , Imunoterapia , Masculino , Neoplasias/imunologia , Neoplasias/terapia , Domínios Proteicos , Receptores Imunológicos/antagonistas & inibidores , Receptores Imunológicos/química , Receptores Imunológicos/imunologia
5.
MAbs ; 10(4): 636-650, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29494279

RESUMO

Identifying monoclonal antibodies that block human voltage-gated ion channels (VGICs) is a challenging endeavor exacerbated by difficulties in producing recombinant ion channel proteins in amounts that support drug discovery programs. We have developed a general strategy to address this challenge by combining high-level expression of recombinant VGICs in Tetrahymena thermophila with immunization of phylogenetically diverse species and unique screening tools that allow deep-mining for antibodies that could potentially bind functionally important regions of the protein. Using this approach, we targeted human Kv1.3, a voltage-gated potassium channel widely recognized as a therapeutic target for the treatment of a variety of T-cell mediated autoimmune diseases. Recombinant Kv1.3 was used to generate and recover 69 full-length anti-Kv1.3 mAbs from immunized chickens and llamas, of which 10 were able to inhibit Kv1.3 current. Select antibodies were shown to be potent (IC50<10 nM) and specific for Kv1.3 over related Kv1 family members, hERG and hNav1.5.


Assuntos
Anticorpos Monoclonais , Descoberta de Drogas/métodos , Canal de Potássio Kv1.3/antagonistas & inibidores , Animais , Camelídeos Americanos , Galinhas , Humanos , Proteínas Recombinantes , Tetrahymena thermophila
6.
MAbs ; 10(1): 71-80, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29035625

RESUMO

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.


Assuntos
Anticorpos Monoclonais Humanizados/biossíntese , Anticorpos Monoclonais Humanizados/imunologia , Afinidade de Anticorpos , Especificidade de Anticorpos , Antígenos/imunologia , Galinhas/imunologia , Epitopos/imunologia , Imunoglobulinas/imunologia , Animais , Animais Geneticamente Modificados , Antígenos/administração & dosagem , Linfócitos B/imunologia , Galinhas/sangue , Galinhas/genética , Mapeamento de Epitopos , Humanos , Imunização , Imunoglobulinas/sangue , Imunoglobulinas/genética , Especificidade da Espécie , Linfócitos T/imunologia
7.
Eur J Immunol ; 46(9): 2137-48, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27392810

RESUMO

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.


Assuntos
Anticorpos/genética , Linfócitos B/imunologia , Linfócitos B/metabolismo , Expressão Gênica , Cadeias Pesadas de Imunoglobulinas/genética , Cadeias Leves de Imunoglobulina/genética , Animais , Animais Geneticamente Modificados , Anticorpos/imunologia , Formação de Anticorpos/genética , Formação de Anticorpos/imunologia , Diferenciação Celular/genética , Diferenciação Celular/imunologia , Galinhas , Deleção de Genes , Técnicas de Inativação de Genes , Ordem dos Genes , Marcação de Genes , Vetores Genéticos/genética , Cadeias Leves de Imunoglobulina/química , Plasmócitos/imunologia , Plasmócitos/metabolismo , Domínios Proteicos/genética
8.
PLoS One ; 11(4): e0154303, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27099923

RESUMO

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.


Assuntos
Sistemas CRISPR-Cas , Galinhas/genética , Edição de Genes/métodos , Genoma , Recombinação Homóloga , Cadeias Pesadas de Imunoglobulinas/genética , Animais , Animais Geneticamente Modificados , Sequência de Bases , Galinhas/crescimento & desenvolvimento , Clonagem de Organismos , Embrião não Mamífero , Feminino , Técnicas de Inativação de Genes , Vetores Genéticos/química , Vetores Genéticos/metabolismo , Células Germinativas , Proteínas de Fluorescência Verde/deficiência , Proteínas de Fluorescência Verde/genética , Masculino , RNA Guia de Cinetoplastídeos/genética , RNA Guia de Cinetoplastídeos/metabolismo
9.
Transgenic Res ; 25(5): 609-16, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27034267

RESUMO

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.


Assuntos
Galinhas/genética , Integrases/genética , Recombinação Genética , Animais , Animais Geneticamente Modificados , Regulação da Expressão Gênica , Proteínas de Fluorescência Verde , Especificidade de Órgãos , Regiões Promotoras Genéticas , Transgenes
10.
Poult Sci ; 94(4): 799-803, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25828572

RESUMO

During the past decade, modifications to the chicken genome have evolved from random insertions of small transgenes using viral vectors to site-specific deletions using homologous recombination vectors and nontargeted insertions of large transgenes using phi-31 integrase. Primordial germ cells (PGC) and gonocytes are the germline-competent cell lines in which targeted modifications and large transgenes are inserted into the genome. After extended periods of in vitro culture, PGC retain their capacity to form functional gametes when reintroduced in vivo. Rates of stable germline modification vary from 1×10(-5) for nontargeted insertions to 1×10(-8) for targeted insertions. Following transfection, clonally derived cell lines are expanded, injected into Stage 13-15 Hamburger and Hamilton embryos, and putative chimeras are incubated to term in surrogate shells. Green fluorescent protein (GFP) is incorporated into transgenes to reveal the presence of genetically modified PGC in culture and the extent of colonization of the gonad during the first week posthatch. If the extent of colonization is adequate, cohorts of putative chimeras are reared to sexual maturity. Semen is collected and the contribution from donor PGC is estimated by evaluating GFP expression using flow cytometry and PCR. The most promising candidates are selected for breeding to obtain G1 heterozygote offspring. To date, this protocol has been used to (1) knockout the immunoglobulin heavy and light chain genes and produce chickens lacking humoral immunity, (2) insert human V genes and arrays of pseudo V genes into the heavy and light immunoglobulin loci to produce chickens making antibodies with human V regions, (3) insert GFP into nontargeted locations within the genome to produce chickens expressing GFP, and (4) insert Cre recombinase into the genome to produce chickens that excise sequences of DNA flanked by loxP sites.


Assuntos
Galinhas/genética , Genoma , Mutagênese Insercional , Transgenes , Animais , Humanos
11.
Proc Natl Acad Sci U S A ; 110(50): 20170-5, 2013 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-24282302

RESUMO

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.


Assuntos
Linfócitos B/citologia , Galinhas/genética , Técnicas de Inativação de Genes/métodos , Engenharia Genética/métodos , Células Germinativas/química , Cadeias Pesadas de Imunoglobulinas/genética , Animais , Linfócitos B/metabolismo , Southern Blotting , Galinhas/imunologia , Metilação de DNA , Primers do DNA/genética , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Genótipo , Células Germinativas/metabolismo , Imuno-Histoquímica
12.
Stem Cells ; 27(6): 1255-64, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19489081

RESUMO

We recently demonstrated that somatic cells from adult primates could be reprogrammed into a pluripotent state by somatic cell nuclear transfer. However, the low efficiency with donor cells from one monkey necessitated the need for large oocyte numbers. Here, we demonstrate nearly threefold higher blastocyst development and embryonic stem (ES) cell derivation rates with different nuclear donor cells. Two ES cell lines were isolated using adult female rhesus macaque skin fibroblasts as nuclear donors and oocytes retrieved from one female, following a single controlled ovarian stimulation. In addition to routine pluripotency tests involving in vitro and in vivo differentiation into various somatic cell types, primate ES cells derived from reprogrammed somatic cells were also capable of contributing to cells expressing markers of germ cells. Moreover, imprinted gene expression, methylation, telomere length, and X-inactivation analyses were consistent with accurate and extensive epigenetic reprogramming of somatic cells by oocyte-specific factors.


Assuntos
Epigênese Genética , Fibroblastos/citologia , Técnicas de Transferência Nuclear , Oócitos/citologia , Células-Tronco Pluripotentes/citologia , Animais , Diferenciação Celular , Reprogramação Celular/genética , Células-Tronco Embrionárias/citologia , Feminino , Expressão Gênica , Macaca mulatta , Reação em Cadeia da Polimerase Via Transcriptase Reversa
13.
Gene Expr Patterns ; 9(2): 94-108, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18996226

RESUMO

The rhesus monkey embryonic stem cell line 366.4 differentiates into serotonin neurons. We examined the genetic cascade during differentiation and compared ESC-derived serotonin neurons to adult monkey serotonin neurons. RNA was extracted from ESC colonies, embryoid bodies (EBs), neurospheres in selection (N1) and proliferation stages (N2), differentiated serotonin neurons (N3) and from laser captured (LC) serotonin neurons of spayed female macaques treated with placebo, estrogen (E), progesterone (P) or E+P. The RNA was labeled and hybridized to Rhesus Monkey Affymetrix Gene Chips (n=1 per stage and 2 per animal treatment). Gene expression was examined with GeneSifter software. 545 genes that were related to developmental processes showed a threefold or greater change between stages. TGFb, Wnt, VEGF and Hedgehog signaling pathways showed the highest percent of probe set changes during differentiation. Genes in the categories (a) homeobox binding and transcription factors, (b) growth factors and receptors, (c) brain and neural specific factors and (d) serotonin specific factors are reported. Pivotal genes were confirmed with quantitative RT-PCR. In the serotonin developmental cascade, FGFR2 was robustly expressed at each stage. GATA3 was robustly expressed in EBs. Sonic hedgehog (Shh), PTCH (Shh-R) and Fev1 transcription factor expression coincided with the induction of serotonin specific marker genes during N1-selection. A majority of the examined genes were expressed in adult serotonin neurons. However, in the ESC-derived neurons, there was significant over-representation of probe sets related to cell cycle, axon guidance & dorso-ventral axis formation. This analysis suggests that the 366.4 cell line possesses cues for serotonin differentiation at early stages of differentiation, but that ESC-derived serotonin neurons are still immature.


Assuntos
Diferenciação Celular/genética , Células-Tronco Embrionárias/fisiologia , Perfilação da Expressão Gênica , Neurônios/fisiologia , Serotonina/metabolismo , Envelhecimento/genética , Envelhecimento/fisiologia , Animais , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Células-Tronco Embrionárias/efeitos dos fármacos , Estradiol/farmacologia , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Macaca mulatta , Camundongos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Progesterona/farmacologia , Transdução de Sinais/genética
14.
Stem Cells ; 26(3): 756-66, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18192229

RESUMO

Monoparental parthenotes represent a potential source of histocompatible stem cells that should be isogenic with the oocyte donor and therefore suitable for use in cell or tissue replacement therapy. We generated five rhesus monkey parthenogenetic embryonic stem cell (PESC) lines with stable, diploid female karyotypes that were morphologically indistinguishable from biparental controls, expressed key pluripotent markers, and generated cell derivatives representative of all three germ layers following in vivo and in vitro differentiation. Interestingly, high levels of heterozygosity were observed at the majority of loci that were polymorphic in the oocyte donors. Some PESC lines were also heterozygous in the major histocompatibility complex region, carrying haplotypes identical to those of the egg donor females. Expression analysis revealed transcripts from some imprinted genes that are normally expressed from only the paternal allele. These results indicate that limitations accompanying the potential use of PESC-derived phenotypes in regenerative medicine, including aberrant genomic imprinting and high levels of homozygosity, are cell line-dependent and not always present. PESC lines were derived in high enough yields to be practicable, and their derivatives are suitable for autologous transplantation into oocyte donors or could be used to establish a bank of histocompatible cell lines for a broad spectrum of patients.


Assuntos
Células-Tronco Embrionárias/citologia , Macaca mulatta/embriologia , Partenogênese , Animais , Diferenciação Celular , Linhagem Celular , Separação Celular , Metilação de DNA , Embrião de Mamíferos , Células-Tronco Embrionárias/metabolismo , Feminino , Fertilização in vitro , Regulação da Expressão Gênica , Impressão Genômica , Genótipo , Heterozigoto , Histocompatibilidade , Humanos , Repetições de Microssatélites/genética , Polimorfismo de Nucleotídeo Único/genética , Síndrome de Prader-Willi/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...