Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Proc Natl Acad Sci U S A ; 117(1): 292-299, 2020 01 07.
Article in English | MEDLINE | ID: mdl-31879340

ABSTRACT

We describe a Kappa-on-Heavy (KoH) mouse that produces a class of highly diverse, fully human, antibody-like agents. This mouse was made by replacing the germline variable sequences of both the Ig heavy-chain (IgH) and Ig kappa (IgK) loci with the human IgK germline variable sequences, producing antibody-like molecules with an antigen binding site made up of 2 kappa variable domains. These molecules, named KoH bodies, structurally mimic naturally existing Bence-Jones light-chain dimers in their variable domains and remain wild-type in their antibody constant domains. Unlike artificially diversified, nonimmunoglobulin alternative scaffolds (e.g., DARPins), KoH bodies consist of a configuration of normal Ig scaffolds that undergo natural diversification in B cells. Monoclonal KoH bodies have properties similar to those of conventional antibodies but exhibit an enhanced ability to bind small molecules such as the endogenous cardiotonic steroid marinobufagenin (MBG) and nicotine. A comparison of crystal structures of MBG bound to a KoH Fab versus a conventional Fab showed that the KoH body has a much deeper binding pocket, allowing MBG to be held 4 Å further down into the combining site between the 2 variable domains.


Subject(s)
Antibodies/chemistry , Antibodies/immunology , Antigens/immunology , Immunoglobulin Heavy Chains/chemistry , Immunoglobulin Variable Region/chemistry , Immunoglobulin Variable Region/immunology , Immunoglobulin kappa-Chains/chemistry , Animals , Antibodies/genetics , Antibodies/therapeutic use , Base Sequence , Binding Sites, Antibody/genetics , Bufanolides , Genetic Engineering , Humans , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Variable Region/genetics , Immunoglobulin kappa-Chains/genetics , Mice , Models, Molecular , Nicotine , Protein Conformation
2.
Biol Reprod ; 100(3): 686-696, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30289441

ABSTRACT

The Adisintegrin and metalloprotease domain-containing (ADAM) family of proteins is involved in cell adhesion, migration, proteolysis, and signaling. Many ADAMs are required for reproduction; however, the role of Adam6 has remained largely unknown. In the course of humanizing the mouse immunoglobulin heavy chain (IgH) locus, we generated Adam6-deficient mice that demonstrate severe subfertility. We decided to elucidate the role of ADAM6 in fertility and explore the underlying mechanisms. Despite normal sperm development and motility, Adam6-deficient mice display diminished male fertility, have abnormal sperm adhesion, and most importantly cannot transition from uterus to oviduct. To test whether ADAM6 is required for sperm's binding to extracellular matrix (ECM) components, we used a panel of ECM components and showed that unlike normal sperm, Adam6-deficient sperm cannot bind fibronectin, laminin, and tenascin. Reintroduction of Adam6 into these deficient mice repaired sperm interaction with ECM, restored male fertility, and corrected the sperm transport deficit. Together, our data suggest that ADAM6, either alone or in complex with other proteins, aids sperm transport through the female reproductive tract by providing a temporary site of attachment of sperm to ECM components prior to ascent into the oviduct.


Subject(s)
ADAM Proteins/metabolism , Infertility, Male/genetics , Sperm Motility/physiology , Spermatozoa/physiology , ADAM Proteins/genetics , Animals , Female , Male , Mice , Mice, Knockout , Oviducts , Sperm Motility/genetics
3.
Proc Natl Acad Sci U S A ; 111(14): 5147-52, 2014 Apr 08.
Article in English | MEDLINE | ID: mdl-24706858

ABSTRACT

Genetic humanization, which involves replacing mouse genes with their human counterparts, can create powerful animal models for the study of human genes and diseases. One important example of genetic humanization involves mice humanized for their Ig genes, allowing for human antibody responses within a mouse background (HumAb mice) and also providing a valuable platform for the generation of fully human antibodies as therapeutics. However, existing HumAb mice do not have fully functional immune systems, perhaps because of the manner in which they were genetically humanized. Heretofore, most genetic humanizations have involved disruption of the endogenous mouse gene with simultaneous introduction of a human transgene at a new and random location (so-called KO-plus-transgenic humanization). More recent efforts have attempted to replace mouse genes with their human counterparts at the same genetic location (in situ humanization), but such efforts involved laborious procedures and were limited in size and precision. We describe a general and efficient method for very large, in situ, and precise genetic humanization using large compound bacterial artificial chromosome-based targeting vectors introduced into mouse ES cells. We applied this method to genetically humanize 3-Mb segments of both the mouse heavy and κ light chain Ig loci, by far the largest genetic humanizations ever described. This paper provides a detailed description of our genetic humanization approach, and the companion paper reports that the humoral immune systems of mice bearing these genetically humanized loci function as efficiently as those of WT mice.


Subject(s)
Genes, Immunoglobulin , Animals , Chromosomes, Artificial, Bacterial , Embryonic Stem Cells/immunology , Homologous Recombination , Humans , Mice , Mice, Knockout , Polymerase Chain Reaction , Transgenes
4.
Proc Natl Acad Sci U S A ; 110(34): E3179-88, 2013 Aug 20.
Article in English | MEDLINE | ID: mdl-23918385

ABSTRACT

Conditional mutagenesis is becoming a method of choice for studying gene function, but constructing conditional alleles is often laborious, limited by target gene structure, and at times, prone to incomplete conditional ablation. To address these issues, we developed a technology termed conditionals by inversion (COIN). Before activation, COINs contain an inverted module (COIN module) that lies inertly within the antisense strand of a resident gene. When inverted into the sense strand by a site-specific recombinase, the COIN module causes termination of the target gene's transcription and simultaneously provides a reporter for tracking this event. COIN modules can be inserted into natural introns (intronic COINs) or directly into coding exons as part of an artificial intron (exonic COINs), greatly simplifying allele design and increasing flexibility over previous conditional KO approaches. Detailed analysis of over 20 COIN alleles establishes the reliability of the method and its broad applicability to any gene, regardless of exon-intron structure. Our extensive testing provides rules that help ensure success of this approach and also explains why other currently available conditional approaches often fail to function optimally. Finally, the ability to split exons using the COIN's artificial intron opens up engineering modalities for the generation of multifunctional alleles.


Subject(s)
Alleles , Gene Silencing , Genetic Engineering/methods , Mutagenesis, Insertional/methods , Sequence Inversion/genetics , DNA Nucleotidyltransferases/metabolism
5.
Transgenic Res ; 24(1): 19-29, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25087174

ABSTRACT

Known examples of male to female sex reversal in mice are caused by either strain incompatibilities or mutations in genes required for male sex determination. The resultant XY females are often sterile or exhibit very poor fertility. We describe here embryonic stem (ES) cell growth conditions that promote the production of healthy, anatomically normal fertile and fecund female F0 generation mice completely derived from gene-targeted XY male ES cells. The sex reversal is a transient trait that is not transmitted to the F1 progeny. Growth media with low osmolality and reduced sodium bicarbonate, maintained throughout the gene targeting process, enhance the yield of XY females. As a practical application of the induced sex reversal, we demonstrate the generation of homozygous mutant mice ready for phenotypic studies by the breeding of F0 XY females with their isogenic XY male clonal siblings, thereby eliminating one generation of breeding and the associated costs.


Subject(s)
Disorders of Sex Development/genetics , Fertility/genetics , Gonadal Dysgenesis, 46,XY/genetics , Sex Determination Processes , Animals , Embryonic Stem Cells/cytology , Female , Gene Targeting , Male , Mice , Microinjections , Mutation
6.
Proc Natl Acad Sci U S A ; 108(6): 2390-5, 2011 Feb 08.
Article in English | MEDLINE | ID: mdl-21262803

ABSTRACT

Mice with a functional human immune system have the potential to allow in vivo studies of human infectious diseases and to enable vaccine testing. To this end, mice need to fully support the development of human immune cells, allow infection with human pathogens, and be capable of mounting effective human immune responses. A major limitation of humanized mice is the poor development and function of human myeloid cells and the absence of human immune responses at mucosal surfaces, such as the lung. To overcome this, we generated human IL-3/GM-CSF knock-in (hIL-3/GM-CSF KI) mice. These mice faithfully expressed human GM-CSF and IL-3 and developed pulmonary alveolar proteinosis because of elimination of mouse GM-CSF. We demonstrate that hIL-3/GM-CSF KI mice engrafted with human CD34(+) hematopoietic cells had improved human myeloid cell reconstitution in the lung. In particular, hIL-3/GM-CSF KI mice supported the development of human alveolar macrophages that partially rescued the pulmonary alveolar proteinosis syndrome. Moreover, human alveolar macrophages mounted correlates of a human innate immune response against influenza virus. The hIL-3/GM-CSF KI mice represent a unique mouse model that permits the study of human mucosal immune responses to lung pathogens.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Immunity, Innate , Influenza A Virus, H1N1 Subtype/immunology , Interleukin-3/immunology , Lung/immunology , Macrophages, Alveolar/immunology , Orthomyxoviridae Infections/immunology , Animals , Cord Blood Stem Cell Transplantation , Gene Knock-In Techniques , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Humans , Immunity, Mucosal/genetics , Interleukin-3/genetics , Lung/virology , Macrophages, Alveolar/virology , Mice , Mice, Transgenic , Models, Immunological , Orthomyxoviridae Infections/genetics , Transplantation Chimera/genetics , Transplantation Chimera/immunology , Transplantation Chimera/virology , Transplantation, Heterologous
7.
Proc Natl Acad Sci U S A ; 108(6): 2378-83, 2011 Feb 08.
Article in English | MEDLINE | ID: mdl-21262827

ABSTRACT

Hematopoietic stem cells (HSCs) both self-renew and give rise to all blood cells for the lifetime of an individual. Xenogeneic mouse models are broadly used to study human hematopoietic stem and progenitor cell biology in vivo. However, maintenance, differentiation, and function of human hematopoietic cells are suboptimal in these hosts. Thrombopoietin (TPO) has been demonstrated as a crucial cytokine supporting maintenance and self-renewal of HSCs. We generated RAG2(-/-)γ(c)(-/-) mice in which we replaced the gene encoding mouse TPO by its human homolog. Homozygous humanization of TPO led to increased levels of human engraftment in the bone marrow of the hosts, and multilineage differentiation of hematopoietic cells was improved, with an increased ratio of myelomonocytic verus lymphoid lineages. Moreover, maintenance of human stem and progenitor cells was improved, as demonstrated by serial transplantation. Therefore, RAG2(-/-)γ(c)(-/-) TPO-humanized mice represent a useful model to study human hematopoiesis in vivo.


Subject(s)
Hematopoiesis , Thrombopoietin/metabolism , Animals , Gene Knock-In Techniques , Hematopoietic Stem Cell Transplantation , Humans , Mice , Mice, Transgenic , Thrombopoietin/genetics , Transplantation Chimera/genetics , Transplantation Chimera/metabolism , Transplantation, Heterologous
8.
Stem Cell Reports ; 18(1): 394-409, 2023 01 10.
Article in English | MEDLINE | ID: mdl-36525967

ABSTRACT

Rats were more frequently used than mice to model human disease before mouse embryonic stem cells (mESCs) revolutionized genetic engineering in mice. Rat ESCs (rESCs) were first reported over 10 years ago, yet they are not as frequently used as mESCs. CRISPR-based gene editing in zygotes is widely used in rats but is limited by the difficulty of inserting or replacing DNA sequences larger than about 10 kb. We report here the generation of germline-competent rESC lines from several rat strains. These rESC lines maintain their potential for germline transmission after serial targeting with bacterial artificial chromosome (BAC)-based targeting vectors, and CRISPR-Cas9 cutting can increase targeting efficiency. Using these methods, we have successfully replaced entire rat genes spanning up to 101 kb with the human ortholog.


Subject(s)
Embryonic Stem Cells , Retinal Degeneration , Humans , Rats , Animals , Mice , Gene Editing , Genetic Engineering , CRISPR-Cas Systems/genetics
9.
Proc Natl Acad Sci U S A ; 106(52): 22399-404, 2009 Dec 29.
Article in English | MEDLINE | ID: mdl-20018779

ABSTRACT

Inhibiting angiogenesis has become an effective approach for treating cancer and other diseases. However, our understanding of signaling pathways in tumor angiogenesis has been limited by the embryonic lethality of many gene knockouts. To overcome this limitation, we used the plasticity of embryonic stem (ES) cells to develop a unique approach to study tumor angiogenesis. Murine ES cells can be readily manipulated genetically; in addition, ES cells implanted subcutaneously in mice develop into tumors that contain a variety of cell types (teratomas). We show that ES cells differentiate into bona fide endothelial cells within the teratoma, and that these ES-derived endothelial cells form part of the functional tumor vasculature. Using this powerful and flexible system, the Angiopoietin/Tie2 system is shown to have a key role in the regulation of tumor vessel size. Endothelial differentiation in the ES teratoma model allows gene-targeting methods to be used in the study of tumor angiogenesis.


Subject(s)
Embryonic Stem Cells/enzymology , Embryonic Stem Cells/pathology , Neoplasms, Experimental/blood supply , Neoplasms, Experimental/enzymology , Neovascularization, Pathologic , Receptor Protein-Tyrosine Kinases/physiology , Receptor-Like Protein Tyrosine Phosphatases, Class 3/physiology , Angiopoietins/antagonists & inhibitors , Animals , Cell Differentiation , Cell Line , Disease Models, Animal , Endothelial Cells/enzymology , Endothelial Cells/pathology , Mice , Mice, SCID , Neoplasms, Experimental/etiology , Receptor, TIE-2 , Receptor-Like Protein Tyrosine Phosphatases, Class 3/deficiency , Receptor-Like Protein Tyrosine Phosphatases, Class 3/genetics , Teratoma/blood supply , Teratoma/enzymology , Teratoma/etiology , Vascular Endothelial Growth Factor Receptor-2/physiology
10.
Nat Biotechnol ; 25(1): 91-9, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17187059

ABSTRACT

A useful approach for exploring gene function involves generating mutant mice from genetically modified embryonic stem (ES) cells. Recent advances in genetic engineering of ES cells have shifted the bottleneck in this process to the generation of mice. Conventional injections of ES cells into blastocyst hosts produce F0 generation chimeras that are only partially derived from ES cells, requiring additional breeding to obtain mutant mice that can be phenotyped. The tetraploid complementation approach directly yields mice that are almost entirely derived from ES cells, but it is inefficient, works only with certain hybrid ES cell lines and suffers from nonspecific lethality and abnormalities, complicating phenotypic analyses. Here we show that laser-assisted injection of either inbred or hybrid ES cells into eight cell-stage embryos efficiently yields F0 generation mice that are fully ES cell-derived and healthy, exhibit 100% germline transmission and allow immediate phenotypic analysis, greatly accelerating gene function assignment.


Subject(s)
Embryonic Stem Cells/cytology , Embryonic Stem Cells/transplantation , Gene Targeting/methods , Laser Therapy/methods , Mice, Transgenic/genetics , Microinjections/methods , Stem Cell Transplantation/methods , Animals , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Mutant Strains , Mice, Transgenic/anatomy & histology , Mice, Transgenic/surgery , Microsurgery/methods , Phenotype
11.
Methods Mol Biol ; 530: 311-24, 2009.
Article in English | MEDLINE | ID: mdl-19266341

ABSTRACT

With the completion of the human and mouse genome sequences and the development of high-throughput knockout mouse technologies, there is now a need for equally high-throughput methods for the production of mice for phenotypic studies. In response to this challenge, we recently developed a new method termed VelociMouse for the production of F0-generation mice that are fully derived from gene-targeted ES cells. In the version of the VelociMouse method described here, laser ablation of a portion of the zona pellucid (zp) of a normal eight-cell-stage embryo facilitates ES cell injection. Upon gestation in a surrogate mother, the injected embryos produce F0 mice that carry no detectable host embryo contribution (<0.1%). The fully ES cell-derived mice are normal, healthy, and fertile and exhibit 100% germline transmission for optimal breeding efficiency. The VelociMouse method accommodates both inbred or hybrid ES cells and either inbred or outbred eight-cell host embryos. Because the F0 mice produced are suitable for direct phenotyping studies, the VelociMouse method, coupled with high-throughput ES cell targeting technologies, such as VelociGene, offers an accelerated path to new drug target discovery and validation and a revolutionary approach to realize the full value of large-scale functional genomic efforts, such as the NIH Knockout Mouse Project ( 1 ) and the European Conditional Mouse Mutagenesis Project( 9 ).


Subject(s)
Embryo Culture Techniques/methods , Embryo, Mammalian/cytology , Embryonic Stem Cells/cytology , Embryonic Stem Cells/transplantation , Gene Targeting/methods , Animals , Cell Differentiation , Female , Male , Mice , Mice, Inbred C57BL , Phenotype , Pregnancy
12.
Mol Cell Biol ; 25(6): 2431-40, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15743835

ABSTRACT

Prosaposin is a multifunctional protein with diverse functions. Intracellularly, prosaposin is a precursor of four sphingolipid activator proteins, saposins A to D, which are required for hydrolysis of sphingolipids by several lysosomal exohydrolases. Secreted prosaposin has been implicated as a neurotrophic, myelinotrophic, and myotrophic factor as well as a spermatogenic factor. It has also been implicated in fertilization. The human and the mouse prosaposin gene has a 9-bp exon (exon 8) that is alternatively spliced, resulting in an isoform with three extra amino acids, Gln-Asp-Gln, within the saposin B domain. Alternative splicing in the prosaposin gene is conserved from fish to humans, tissue specific, and regulated in the brain during development and nerve regeneration-degeneration processes. To elucidate the physiological role of alternative splicing, we have generated a mouse lacking exon 8 by homologous recombination. The exon 8 prosaposin mutant mice are healthy and fertile with no obvious phenotype. No changes were detected in prosaposin secretion or in accumulation and metabolism of gangliosides, sulfatides, neutral glycosphingolipids, neutral phospholipids, other neutral lipids, and ceramide. These data strongly indicate that the prosaposin variant containing the exon 8-encoded three amino acids is dispensable for normal mouse development and fertility as well as for prosaposin secretion and its lysosomal function, at least in the presence of the prosaposin variant missing the exon 8-encoded three amino acids.


Subject(s)
Alternative Splicing/physiology , Exons/genetics , Mice/growth & development , Saposins/genetics , Saposins/physiology , Alternative Splicing/genetics , Animals , Asparagine/genetics , Embryo, Mammalian/cytology , Fertility/genetics , Fertility/physiology , Glutamine/genetics , Lipid Metabolism , Lipids/analysis , Lysosomes/physiology , Male , Mice/genetics , Mice, Mutant Strains , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Isoforms/physiology , RNA, Messenger/analysis , RNA, Messenger/metabolism , Saposins/metabolism , Sequence Deletion , Stem Cells/metabolism , Testis/metabolism , Tissue Distribution
13.
Mol Cell Biol ; 24(16): 7235-48, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15282322

ABSTRACT

The fission yeast Schizosaccharomyces pombe rad9 gene promotes cell survival through activation of cell cycle checkpoints induced by DNA damage. Mouse embryonic stem cells with a targeted deletion of Mrad9, the mouse ortholog of this gene, were created to evaluate its function in mammals. Mrad9(-/-) cells demonstrated a marked increase in spontaneous chromosome aberrations and HPRT mutations, indicating a role in the maintenance of genomic integrity. These cells were also extremely sensitive to UV light, gamma rays, and hydroxyurea, and heterozygotes were somewhat sensitive to the last two agents relative to Mrad9(+/+) controls. Mrad9(-/-) cells could initiate but not maintain gamma-ray-induced G(2) delay and retained the ability to delay DNA synthesis rapidly after UV irradiation, suggesting that checkpoint abnormalities contribute little to the radiosensitivity observed. Ectopic expression of Mrad9 or human HRAD9 complemented Mrad9(-/-) cell defects, indicating that the gene has radioresponse and genomic maintenance functions that are evolutionarily conserved. Mrad9(+/-) mice were generated, but heterozygous intercrosses failed to yield Mrad9(-/-) pups, since embryos died at midgestation. Furthermore, Mrad9(-/-) mouse embryo fibroblasts were not viable. These investigations establish Mrad9 as a key mammalian genetic element of pathways that regulate the cellular response to DNA damage, maintenance of genomic integrity, and proper embryonic development.


Subject(s)
Cell Cycle Proteins/metabolism , DNA Damage , Embryo, Mammalian/physiology , Gene Deletion , Animals , Apoptosis/physiology , Cell Cycle/physiology , Cell Cycle Proteins/genetics , Cells, Cultured , Chromosome Aberrations , Embryo, Mammalian/anatomy & histology , Gene Expression Regulation , Genes, cdc , Genomic Instability , Humans , Hydroxyurea/pharmacology , In Situ Nick-End Labeling , Mice , Mice, Knockout , Nucleic Acid Synthesis Inhibitors/pharmacology , Radiation, Ionizing , Stem Cells/cytology , Stem Cells/drug effects , Stem Cells/physiology , Stem Cells/radiation effects , Survival Rate
14.
Nat Biotechnol ; 21(6): 652-9, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12730667

ABSTRACT

One of the most effective approaches for determining gene function involves engineering mice with mutations or deletions in endogenous genes of interest. Historically, this approach has been limited by the difficulty and time required to generate such mice. We describe the development of a high-throughput and largely automated process, termed VelociGene, that uses targeting vectors based on bacterial artificial chromosomes (BACs). VelociGene permits genetic alteration with nucleotide precision, is not limited by the size of desired deletions, does not depend on isogenicity or on positive-negative selection, and can precisely replace the gene of interest with a reporter that allows for high-resolution localization of target-gene expression. We describe custom genetic alterations for hundreds of genes, corresponding to about 0.5-1.0% of the entire genome. We also provide dozens of informative expression patterns involving cells in the nervous system, immune system, vasculature, skeleton, fat and other tissues.


Subject(s)
Chromosomes, Artificial, Bacterial/genetics , Chromosomes, Artificial, Bacterial/metabolism , Gene Expression Profiling/methods , Genetic Engineering/methods , Genome , Animals , Animals, Genetically Modified/genetics , Animals, Genetically Modified/metabolism , Electroporation/methods , Gene Targeting/methods , Mice/genetics , Mutagenesis, Insertional/methods , Mutagenesis, Site-Directed , Quality Control , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Stem Cells/metabolism
15.
Cold Spring Harb Protoc ; 2017(9): pdb.prot094367, 2017 Sep 01.
Article in English | MEDLINE | ID: mdl-28864571

ABSTRACT

In this protocol, eight-cell-stage precompaction embryos from outbred mouse strains are used for the injection of hybrid or inbred embryonic stem (ES) cells. This process often leads to generation of fully ES cell-derived so-called F0 mice (VelociMice). Postinjection culture of embryos is necessary to achieve the highest ratio of fully ES cell-derived mice and high-degree chimeras. Typically, 50 embryos are injected per ES cell clone.


Subject(s)
Embryo, Mammalian/cytology , Injections/methods , Mouse Embryonic Stem Cells/cytology , Animals , Female , Mice
16.
Sci Rep ; 6: 23204, 2016 Mar 16.
Article in English | MEDLINE | ID: mdl-26979938

ABSTRACT

The expansion of a hexanucleotide (GGGGCC) repeat in C9ORF72 is the most common cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Both the function of C9ORF72 and the mechanism by which the repeat expansion drives neuropathology are unknown. To examine whether C9ORF72 haploinsufficiency induces neurological disease, we created a C9orf72-deficient mouse line. Null mice developed a robust immune phenotype characterized by myeloid expansion, T cell activation, and increased plasma cells. Mice also presented with elevated autoantibodies and evidence of immune-mediated glomerulonephropathy. Collectively, our data suggest that C9orf72 regulates immune homeostasis and an autoimmune response reminiscent of systemic lupus erythematosus (SLE) occurs in its absence. We further imply that haploinsufficiency is unlikely to be the causative factor in C9ALS/FTD pathology.


Subject(s)
Autoantibodies/biosynthesis , Autoimmunity , Glomerulonephritis, Membranoproliferative/genetics , Guanine Nucleotide Exchange Factors/genetics , Animals , Autoantibodies/blood , C9orf72 Protein , Cytokines/blood , Female , Glomerulonephritis, Membranoproliferative/blood , Glomerulonephritis, Membranoproliferative/immunology , Guanine Nucleotide Exchange Factors/metabolism , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/immunology , Lymphocyte Activation , Lymphoid Tissue/pathology , Macrophages/immunology , Male , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Plasma Cells/immunology , Sequence Analysis, RNA , Transcriptome
17.
PLoS One ; 10(4): e0125522, 2015.
Article in English | MEDLINE | ID: mdl-25909911

ABSTRACT

In a survey of 20 knockout mouse lines designed to examine the biological functions of large intergenic non-coding RNAs (lincRNAs), we have found a variety of phenotypes, ranging from perinatal lethality to defects associated with premature aging and morphological and functional abnormalities in the lungs, skeleton, and muscle. Each mutant allele carried a lacZ reporter whose expression profile highlighted a wide spectrum of spatiotemporal and tissue-specific transcription patterns in embryos and adults that informed our phenotypic analyses and will serve as a guide for future investigations of these genes. Our study shows that lincRNAs are a new class of encoded molecules that, like proteins, serve essential and important functional roles in embryonic development, physiology, and homeostasis of a broad array of tissues and organs in mammals.


Subject(s)
RNA, Long Noncoding/genetics , Transcription, Genetic/genetics , Transcriptome/genetics , Alleles , Animals , Embryonic Development/genetics , Female , Genes, Reporter/genetics , Male , Mammals/genetics , Membrane Transport Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype
18.
Methods Enzymol ; 476: 285-94, 2010.
Article in English | MEDLINE | ID: mdl-20691872

ABSTRACT

In conventional methods for the generation of genetically modified mice, gene-targeted embryonic stem (ES) cells are injected into blastocyst-stage embryos or are aggregated with morula-stage embryos, which are then transferred to the uterus of a surrogate mother. F0 generation mice born from the embryos are chimeras composed of genetic contributions from both the modified ES cells and the recipient embryos. Obtaining a mouse strain that carries the gene-targeted mutation requires breeding the chimeras to transmit the ES cell genetic component through the germ line to the next (F1) generation (germ line transmission, GLT). To skip the chimera stage, we developed the VelociMouse method, in which injection of genetically modified ES cells into eight-cell embryos followed by maturation to the blastocyst stage and transfer to a surrogate mother produces F0 generation mice that are fully derived from the injected ES cells and exhibit a 100% GLT efficiency. The method is simple and flexible. Both male and female ES cells can be introduced into the eight-cell embryo by any method of injection or aggregation and using all ES cell and host embryo combinations from inbred, hybrid, and outbred genetic backgrounds. The VelociMouse method provides several unique opportunities for shortening project timelines and reducing mouse husbandry costs. First, as VelociMice exhibit 100% GLT, there is no need to test cross chimeras to establish GLT. Second, because the VelociMouse method permits efficient production of ES cell-derived mice from female ES cells, XO ES cell subclones, identified by screening for spontaneous loss of the Y chromosome, can be used to generate F0 females that can be bred with isogenic F0 males derived from the original targeted ES cell clone to obtain homozygous mutant mice in the F1 generation. Third, as VelociMice are genetically identical to the ES cells from which they were derived, the VelociMouse method opens up myriad possibilities for creating mice with complex genotypes in a defined genetic background directly from engineered ES cells without the need for inefficient and lengthy breeding schemes. Examples include creation of F0 knockout mice from ES cells carrying a homozygous null mutation, and creation of a mouse with a tissue-specific gene inactivation by combining null and floxed conditional alleles for the target gene with a transgenic Cre recombinase allele controlled by a tissue-specific promoter. VelociMice with the combinatorial alleles are ready for immediate phenotypic studies, which greatly accelerates gene function assignment and the creation of valuable models of human disease.


Subject(s)
Embryo Culture Techniques/methods , Embryo Transfer/methods , Embryo, Mammalian , Embryonic Stem Cells , Animals , Cell Line , Chimera , Embryo Culture Techniques/instrumentation , Embryo Transfer/instrumentation , Embryonic Stem Cells/cytology , Embryonic Stem Cells/physiology , Female , Humans , Male , Mice , Mice, Inbred C57BL , Microinjections/methods , Pregnancy
19.
Traffic ; 7(1): 52-60, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16445686

ABSTRACT

EHD1 is a member of the EHD family that contains four mammalian homologs. Among the invertebrate orthologs are a single Drosophila and Caenorhabditis elegans proteins and two plant members. They all contain three modules, a N-terminal domain that contains nucleotide-binding motifs, a central coiled-coil domain involved in oligomerization and a C-terminal region that harbors the EH domain. Studies in C. elegans and EHD1 depletion by RNA interference in human cells have demonstrated that it regulates recycling of membrane proteins. We addressed the physiological role of EHD1 through its inactivation in the mouse. Ehd1 knockout mice were indistinguishable from normal mice, had a normal life span and showed no histological abnormalities. Analysis of transferrin uptake in Ehd1(-/-) embryonic fibroblasts demonstrated delayed recycling to the plasma membrane with accumulation of transferrin in the endocytic recycling compartment. Our results corroborate the established role of EHD1 in the exit of membrane proteins from recycling endosomes in vivo in a mouse model.


Subject(s)
Cell Membrane/metabolism , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/physiology , Alleles , Animals , Cell Proliferation , Fibroblasts/cytology , Fibroblasts/metabolism , Mice , Mice, Knockout , Models, Animal , Mutation , Protein Transport/genetics , Protein Transport/physiology , Time Factors , Transferrin/metabolism
20.
Cell ; 124(2): 315-29, 2006 Jan 27.
Article in English | MEDLINE | ID: mdl-16439206

ABSTRACT

The Sir2 histone deacetylase functions as a chromatin silencer to regulate recombination, genomic stability, and aging in budding yeast. Seven mammalian Sir2 homologs have been identified (SIRT1-SIRT7), and it has been speculated that some may have similar functions to Sir2. Here, we demonstrate that SIRT6 is a nuclear, chromatin-associated protein that promotes resistance to DNA damage and suppresses genomic instability in mouse cells, in association with a role in base excision repair (BER). SIRT6-deficient mice are small and at 2-3 weeks of age develop abnormalities that include profound lymphopenia, loss of subcutaneous fat, lordokyphosis, and severe metabolic defects, eventually dying at about 4 weeks. We conclude that one function of SIRT6 is to promote normal DNA repair, and that SIRT6 loss leads to abnormalities in mice that overlap with aging-associated degenerative processes.


Subject(s)
Aging/metabolism , Genetic Diseases, Inborn/genetics , Genomic Instability , Sirtuins/genetics , Sirtuins/physiology , Animals , Cell Proliferation , Chromatin/metabolism , DNA Damage , DNA Repair , Genetic Diseases, Inborn/pathology , Humans , Ki-1 Antigen/metabolism , Lymphocytes/immunology , Mice , Mice, Knockout , Phenotype , Radiation Tolerance , Signal Transduction , Sirtuins/deficiency
SELECTION OF CITATIONS
SEARCH DETAIL