Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
1.
Reproduction ; 2024 Jan 01.
Article in English | MEDLINE | ID: mdl-38215284

ABSTRACT

The uterine epithelium is composed of a single layer of hormone responsive polarized epithelial cells that line the lumen and form tubular glands. Endometrial epithelial organoids (EEO) can be generated from uterine epithelia and recapitulate cell composition and hormone responses in vitro. As such, the development of EEO represents a major advance for facilitating mechanistic studies in vitro. However, a major limitation for the use of EEO cultured in basement membrane extract and other hydrogels is the inner location of apical membrane, thereby hindering direct access to the apical surface of the epithelium to study interactions with the embryo or infectious agents such as viruses and bacteria. Here, a straightforward strategy was developed that successfully reverses the polarity of EEO. The result is an apical-out organoid that preserves a distinct apical-basolateral orientation and remains responsive to ovarian steroid hormones. Our investigations highlight the utility of polarity-reversed EEO to study interactions with E. coli and blastocysts. This method of generating apical-out EEO lays the foundation for developing new in vitro functional assays, particularly regarding epithelial interactions with embryos during pregnancy or other luminal constituents in a pathological or diseased state.

2.
Proc Natl Acad Sci U S A ; 117(39): 24195-24204, 2020 09 29.
Article in English | MEDLINE | ID: mdl-32929012

ABSTRACT

Spermatogonial stem cell transplantation (SSCT) is an experimental technique for transfer of germline between donor and recipient males that could be used as a tool for biomedical research, preservation of endangered species, and dissemination of desirable genetics in food animal populations. To fully realize these potentials, recipient males must be devoid of endogenous germline but possess normal testicular architecture and somatic cell function capable of supporting allogeneic donor stem cell engraftment and regeneration of spermatogenesis. Here we show that male mice, pigs, goats, and cattle harboring knockout alleles of the NANOS2 gene generated by CRISPR-Cas9 editing have testes that are germline ablated but otherwise structurally normal. In adult pigs and goats, SSCT with allogeneic donor stem cells led to sustained donor-derived spermatogenesis. With prepubertal mice, allogeneic SSCT resulted in attainment of natural fertility. Collectively, these advancements represent a major step toward realizing the enormous potential of surrogate sires as a tool for dissemination and regeneration of germplasm in all mammalian species.


Subject(s)
Adult Germline Stem Cells/transplantation , RNA-Binding Proteins/physiology , Spermatogenesis , Animals , Cattle , Female , Goats , Male , Mice , Mice, Knockout , Swine , Testis/anatomy & histology , Testis/physiology , Transplantation, Homologous
3.
BMC Genomics ; 23(1): 181, 2022 Mar 05.
Article in English | MEDLINE | ID: mdl-35247961

ABSTRACT

BACKGROUND: Meiotic recombination is one of the important phenomena contributing to gamete genome diversity. However, except for human and a few model organisms, it is not well studied in livestock, including cattle. RESULTS: To investigate their distributions in the cattle sperm genome, we sequenced 143 single sperms from two Holstein bulls. We mapped meiotic recombination events at high resolution based on phased heterozygous single nucleotide polymorphism (SNP). In the absence of evolutionary selection pressure in fertilization and survival, recombination events in sperm are enriched near distal chromosomal ends, revealing that such a pattern is intrinsic to the molecular mechanism of meiosis. Furthermore, we further validated these findings in single sperms with results derived from sequencing its family trio of diploid genomes and our previous studies of recombination in cattle. CONCLUSIONS: To our knowledge, this is the first large-scale single sperm whole-genome sequencing effort in livestock, which provided useful information for future studies of recombination, genome instability, and male infertility.


Subject(s)
Meiosis , Recombination, Genetic , Animals , Cattle/genetics , Chromosome Mapping , Male , Meiosis/genetics , Polymorphism, Single Nucleotide , Sequence Analysis, DNA/methods , Spermatozoa
4.
Biol Reprod ; 106(4): 629-638, 2022 04 26.
Article in English | MEDLINE | ID: mdl-35094055

ABSTRACT

Increased knowledge of reproduction and health of domesticated animals is integral to sustain and improve global competitiveness of U.S. animal agriculture, understand and resolve complex animal and human diseases, and advance fundamental research in sciences that are critical to understanding mechanisms of action and identifying future targets for interventions. Historically, federal and state budgets have dwindled and funding for the United States Department of Agriculture (USDA) National Institute of Food and Agriculture (NIFA) competitive grants programs remained relatively stagnant from 1985 through 2010. This shortage in critical financial support for basic and applied research, coupled with the underappreciated knowledge of the utility of non-rodent species for biomedical research, hindered funding opportunities for research involving livestock and limited improvements in both animal agriculture and animal and human health. In 2010, the National Institutes of Health and USDA NIFA established an interagency partnership to promote the use of agriculturally important animal species in basic and translational research relevant to both biomedicine and agriculture. This interagency program supported 61 grants totaling over $107 million with 23 awards to new or early-stage investigators. This article will review the success of the 9-year Dual Purpose effort and highlight opportunities for utilizing domesticated agricultural animals in research.


Subject(s)
Agriculture , Animals, Domestic , Animals , Livestock , National Institutes of Health (U.S.) , United States , United States Department of Agriculture
5.
Adv Anat Embryol Cell Biol ; 234: 21-40, 2021.
Article in English | MEDLINE | ID: mdl-34694476

ABSTRACT

The preimplantation mammalian embryo is a simplistic, self-contained, and a superior model for investigating the inherent complexities of cell fate decision mechanisms. All mammals begin their humble journey from a single-cell fertilized zygote contained within a proteinaceous coat called the zona pellucida. The zygote embarks on a series of well-orchestrated events, beginning with the activation of embryonic genome, transition from meiotic to mitotic divisions, spatial organization of the cells, timely differentiation into committed trophectoderm (TE) and primitive endoderm (PrE), and ultimately escape from zona pellucida for implantation into the uterus. The entire development of preimplantation embryo can be studied in vitro using a minimalistic and defined culture system. The ease of culture along with the ability to manipulate gene expression and image the embryos makes them an ideal model system for investigation into the first two of several cell fate decisions made by the embryo that result in a pluripotent epiblast (EPI) and differentiated TE and PrE lineages. This chapter reviews our latest knowledge of preimplantation embryo development, setting the stage for understanding placental development in subsequent chapters in this Book.


Subject(s)
Blastocyst , Placenta , Animals , Blastocyst/metabolism , Cell Differentiation/physiology , Embryo Implantation/physiology , Embryo, Mammalian/metabolism , Embryonic Development , Endoderm , Female , Mammals , Pregnancy
6.
Biol Reprod ; 100(1): 208-216, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30085007

ABSTRACT

Placental hypoxia can stimulate oxidative stress and mitochondrial dysfunction reducing placental efficiency and inducing fetal growth restriction (FGR). We hypothesized that chronic hypoxia inhibits mitochondrial function in the placenta as an underlying cause of cellular mechanisms contributing to FGR. Pregnant guinea pigs were exposed to either normoxia (NMX) or hypoxia (HPX; 10.5% O2) at 25 day gestation until term (65 day). Guinea pigs were anesthetized, and fetuses and placentas were excised at either mid (40 day) or late gestation (64 day), weighed, and placental tissue stored at -80°C until assayed. Mitochondrial DNA content, protein expression of respiratory Complexes I-V, and nitration and activity rates of Complexes I and IV were measured in NMX and HPX male (N = 6 in each treatment) and female (N = 6 in each treatment) placentas. Mitochondrial density was not altered by HPX in either mid- or late-term placentas. In mid gestation, HPX slightly increased expression of Complexes I-III and V in male placentas only, but had no effect on either Complex I or IV activity rates or nitrotyrosine expression. In late gestation, HPX significantly decreased CI/CIV activity rates and increased CI/CIV nitration in male but not female placentas exhibiting a sexual dimorphism. Complex I-V expression was reduced from mid to late gestation in both male and female placentas regardless of treatment. We conclude that chronic HPX decreases mitochondrial function by inhibiting Complex I/IV activity via increased peroxynitrite in a sex-related manner. Further, there may be a progressive decrease in energy metabolism of placental cell types with gestation that increases the vulnerability of placental function to intrauterine stress.


Subject(s)
Hypoxia/physiopathology , Mitochondria/physiology , Placenta/physiopathology , Prenatal Exposure Delayed Effects , Sex Characteristics , Animals , DNA, Mitochondrial/metabolism , Female , Fetal Growth Retardation/genetics , Fetal Growth Retardation/metabolism , Fetal Growth Retardation/pathology , Fetal Growth Retardation/physiopathology , Fetal Hypoxia/genetics , Fetal Hypoxia/metabolism , Fetal Hypoxia/pathology , Fetal Hypoxia/physiopathology , Fetal Weight/physiology , Hypoxia/metabolism , Male , Organ Size , Oxidative Stress/physiology , Placenta/metabolism , Placenta/pathology , Pregnancy , Prenatal Exposure Delayed Effects/metabolism , Prenatal Exposure Delayed Effects/pathology , Prenatal Exposure Delayed Effects/physiopathology
7.
Mamm Genome ; 28(7-8): 338-347, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28712062

ABSTRACT

Genetic modification of livestock has a longstanding and successful history, starting with domestication several thousand years ago. Modern animal breeding strategies predominantly based on marker-assisted and genomic selection, artificial insemination, and embryo transfer have led to significant improvement in the performance of domestic animals, and are the basis for regular supply of high quality animal derived food. However, the current strategy of breeding animals over multiple generations to introduce novel traits is not realistic in responding to the unprecedented challenges such as changing climate, pandemic diseases, and feeding an anticipated 3 billion increase in global population in the next three decades. Consequently, sophisticated genetic modifications that allow for seamless introgression of novel alleles or traits and introduction of precise modifications without affecting the overall genetic merit of the animal are required for addressing these pressing challenges. The requirement for precise modifications is especially important in the context of modeling human diseases for the development of therapeutic interventions. The animal science community envisions the genome editors as essential tools in addressing these critical priorities in agriculture and biomedicine, and for advancing livestock genetic engineering for agriculture, biomedical as well as "dual purpose" applications.


Subject(s)
Agriculture , Biomedical Research , Gene Editing , Genetic Engineering , Genome , Livestock/genetics , Agriculture/methods , Animals , Animals, Domestic , Breeding , Gene Editing/methods , Genetic Engineering/methods , Humans
8.
J Pathol ; 238(2): 247-56, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26414877

ABSTRACT

The recent development of gene editing tools and methodology for use in livestock enables the production of new animal disease models. These tools facilitate site-specific mutation of the genome, allowing animals carrying known human disease mutations to be produced. In this review, we describe the various gene editing tools and how they can be used for a range of large animal models of diseases. This genomic technology is in its infancy but the expectation is that through the use of gene editing tools we will see a dramatic increase in animal model resources available for both the study of human disease and the translation of this knowledge into the clinic. Comparative pathology will be central to the productive use of these animal models and the successful translation of new therapeutic strategies.


Subject(s)
Disease Models, Animal , Genetic Engineering/methods , Mutation/genetics , Alleles , Animals , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Embryo Research , Forecasting , Frameshift Mutation/genetics , Genetic Engineering/trends , Humans , Nuclear Transfer Techniques , Primates , Swine , Transcriptional Activation/genetics , Zinc Fingers/genetics
9.
Biol Reprod ; 95(6): 128, 2016 12.
Article in English | MEDLINE | ID: mdl-27806942

ABSTRACT

Chronic placental hypoxia is one of the root causes of placental insufficiencies that result in pre-eclampsia and maternal hypertension. Chronic hypoxia causes disruption of trophoblast (TB) development, invasion into maternal decidua, and remodeling of maternal spiral arteries. The pregnant guinea pig shares several characteristics with humans such as hemomonochorial placenta, villous subplacenta, deep TB invasion, and remodeling of maternal arteries, and is an ideal animal model to study placental development. We hypothesized that chronic placental hypoxia of the pregnant guinea pig inhibits TB invasion and alters spiral artery remodeling. Time-mated pregnant guinea pigs were exposed to either normoxia (NMX) or three levels of hypoxia (HPX: 16%, 12%, or 10.5% O2) from 20 day gestation until midterm (39-40 days) or term (60-65 days). At term, HPX (10.5% O2) increased maternal arterial blood pressure (HPX 57.9 ± 2.3 vs. NMX 40.4 ± 2.3, P < 0.001), decreased fetal weight by 16.1% (P < 0.05), and increased both absolute and relative placenta weights by 10.1% and 31.8%, respectively (P < 0.05). At midterm, there was a significant increase in TB proliferation in HPX placentas as confirmed by increased PCNA and KRT7 staining and elevated ESX1 (TB marker) gene expression (P < 0.05). Additionally, quantitative image analysis revealed decreased invasion of maternal blood vessels by TB cells. In summary, this animal model of placental HPX identifies several aspects of abnormal placental development, including increased TB proliferation and decreased migration and invasion of TBs into the spiral arteries, the consequences of which are associated with maternal hypertension and fetal growth restriction.


Subject(s)
Hypertension, Pregnancy-Induced/etiology , Hypoxia/complications , Placenta/metabolism , Placental Insufficiency/etiology , Animals , Blood Pressure/physiology , Cell Proliferation/physiology , Disease Models, Animal , Female , Guinea Pigs , Hypertension, Pregnancy-Induced/metabolism , Hypertension, Pregnancy-Induced/physiopathology , Hypoxia/metabolism , Hypoxia/physiopathology , Keratin-7/metabolism , Organ Size/physiology , Placenta/physiopathology , Placental Insufficiency/metabolism , Placental Insufficiency/physiopathology , Placentation , Pregnancy , Proliferating Cell Nuclear Antigen/metabolism , Vascular Remodeling/physiology
10.
Int J Mol Sci ; 17(6)2016 May 26.
Article in English | MEDLINE | ID: mdl-27240344

ABSTRACT

The pig is an ideal large animal model for genetic engineering applications. A relatively short gestation interval and large litter size makes the pig a conducive model for generating and propagating genetic modifications. The domestic pig also shares close similarity in anatomy, physiology, size, and life expectancy, making it an ideal animal for modeling human diseases. Often, however, the technical difficulties in generating desired genetic modifications such as targeted knockin of short stretches of sequences or transgenes have impeded progress in this field. In this study, we have investigated and compared the relative efficiency of CRISPR/Cas ribonucleoproteins in engineering targeted knockin of pseudo attP sites downstream of a ubiquitously expressed COL1A gene in porcine somatic cells and generated live fetuses by somatic cell nuclear transfer (SCNT). By leveraging these knockin pseudo attP sites, we have demonstrated subsequent phiC31 integrase mediated integration of green fluorescent protein (GFP) transgene into the site. This work for the first time created an optimized protocol for CRISPR/Cas mediated knockin in porcine somatic cells, while simultaneously creating a stable platform for future transgene integration and generating transgenic animals.


Subject(s)
Collagen Type I/genetics , Gene Knock-In Techniques/methods , Ribonucleoproteins/metabolism , Swine/genetics , Animals , Animals, Genetically Modified , Attachment Sites, Microbiological , CRISPR-Cas Systems , Cells, Cultured , Fibroblasts/cytology , Gene Targeting , Genetic Engineering/methods , Humans , Integrases/metabolism , Nuclear Transfer Techniques
11.
Int J Mol Sci ; 17(12)2016 Dec 03.
Article in English | MEDLINE | ID: mdl-27918485

ABSTRACT

The domestic pig is an ideal "dual purpose" animal model for agricultural and biomedical research. With the availability of genome editing tools such as clustered regularly interspaced short palindromic repeat (CRISPR) and associated nuclease Cas9 (CRISPR/Cas9), it is now possible to perform site-specific alterations with relative ease, and will likely help realize the potential of this valuable model. In this article, we investigated for the first time a combination of somatic cell nuclear transfer (SCNT) and direct injection of CRISPR/Cas ribonucleoprotein complex targeting GRB10 into the reconstituted oocytes to generate GRB10 ablated Ossabaw fetuses. This strategy resulted in highly efficient (100%) generation of biallelic modifications in cloned fetuses. By combining SCNT with CRISPR/Cas9 microinjection, genome edited animals can now be produced without the need to manage a founder herd, while simultaneously eliminating the need for laborious in vitro culture and screening. Our approach utilizes standard cloning techniques while simultaneously performing genome editing in the cloned zygotes of a large animal model for agriculture and biomedical applications.


Subject(s)
CRISPR-Cas Systems/genetics , Gene Editing/methods , Genome , Microinjections/methods , Nuclear Transfer Techniques , Sus scrofa/genetics , Animals , Cloning, Organism , Genotyping Techniques , RNA, Guide, Kinetoplastida/metabolism , Zygote/metabolism
12.
Mol Reprod Dev ; 82(9): 709-21, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26118622

ABSTRACT

The creation of genetically modified goats provides a powerful approach for improving animal health, enhancing production traits, animal pharming, and for ensuring food safety all of which are high-priority goals for animal agriculture. The availability of goat embryonic stem cells (ESCs) that are characteristically immortal in culture would be of enormous benefit for developing genetically modified animals. As an alternative to long-sought goat ESCs, we generated induced pluripotent stem cells (iPSC) by forced expression of bovine POU5F1, SOX2, MYC, KLF4, LIN-28, and NANOG reprogramming factors in combination with a MIR302/367 cluster, delivered by lentiviral vectors. In order to minimize integrations, the reprogramming factor coding sequences were assembled with porcine teschovirus-1 2A (P2A) self-cleaving peptides that allowed for tri-cistronic expression from each vector. The lentiviral-transduced cells were cultured on irradiated mouse feeder cells in a semi-defined, serum-free medium containing fibroblast growth factor (FGF) and/or leukemia inhibitory factor (LIF). The resulting goat iPSC exhibit cell and colony morphology typical of human and mouse ESCs-that is, well-defined borders, a high nuclear-to-cytoplasmic ratio, a short cell-cycle interval, alkaline phosphatase expression, and the ability to generate teratomas in vivo. Additionally, these goat iPSC demonstrated the ability to differentiate into directed lineages in vitro. These results constitute the first steps in establishing integration and footprint-free iPSC from ruminants. Mol. Reprod. Dev. 82: 709-721, 2015. © 2015 Wiley Periodicals, Inc.


Subject(s)
Cell Differentiation/genetics , Cellular Reprogramming/genetics , Goats/genetics , Induced Pluripotent Stem Cells , Animals , Cattle , Cell Culture Techniques/methods , Cells, Cultured , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/physiology , Kruppel-Like Factor 4 , Mice , Stem Cell Research
13.
Reproduction ; 147(5): D1-12, 2014 May.
Article in English | MEDLINE | ID: mdl-24518070

ABSTRACT

It is imperative to unveil the full range of differentiated cell types into which human pluripotent stem cells (hPSCs) can develop. The need is twofold: it will delimit the therapeutic utility of these stem cells and is necessary to place their position accurately in the developmental hierarchy of lineage potential. Accumulated evidence suggested that hPSC could develop in vitro into an extraembryonic lineage (trophoblast (TB)) that is typically inaccessible to pluripotent embryonic cells during embryogenesis. However, whether these differentiated cells are truly authentic TB has been challenged. In this debate, we present a case for and a case against TB differentiation from hPSCs. By analogy to other differentiation systems, our debate is broadly applicable, as it articulates higher and more challenging standards for judging whether a given cell type has been genuinely produced from hPSC differentiation.


Subject(s)
Cell Differentiation/physiology , Embryonic Stem Cells/cytology , Trophoblasts/cytology , Cell Lineage , Cells, Cultured , Embryonic Stem Cells/physiology , Female , Humans , In Vitro Techniques , Morphogenesis/physiology , Placenta/cytology , Placenta/physiology , Pregnancy , Trophoblasts/physiology
14.
Sci Rep ; 14(1): 14822, 2024 06 27.
Article in English | MEDLINE | ID: mdl-38937564

ABSTRACT

Milk is a good source of nutrition but is also a source of allergenic proteins such as α-lactalbumin, ß-lactoglobulin (BLG), casein, and immunoglobulins. The Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)/Cas technology has the potential to edit any gene, including milk allergens. Previously, CRISPR/Cas has been successfully employed in dairy cows and goats, but buffaloes remain unexplored for any milk trait. In this study, we utilized the CRISPR/Cas9 system to edit the major milk allergen BLG gene in buffaloes. First, the editing efficiency of designed sgRNAs was tested in fibroblast cells using the T7E assay and Sanger sequencing. The most effective sgRNA was selected to generate clonal lines of BLG-edited cells. Analysis of 15 single-cell clones, through TA cloning and Sanger sequencing, revealed that 7 clones exhibited bi-allelic (-/-) heterozygous, bi-allelic (-/-) homozygous, and mono-allelic (-/+) disruptions in BLG. Bioinformatics prediction analysis confirmed that non-multiple-of-3 edited nucleotide cell clones have frame shifts and early truncation of BLG protein, while multiple-of-3 edited nucleotides resulted in slightly disoriented protein structures. Somatic cell nuclear transfer (SCNT) method was used to produce blastocyst-stage embryos that have similar developmental rates and quality with wild-type embryos. This study demonstrated the successful bi-allelic editing (-/-) of BLG in buffalo cells through CRISPR/Cas, followed by the production of BLG-edited blastocyst stage embryos using SCNT. With CRISPR and SCNT methods described herein, our long-term goal is to generate gene-edited buffaloes with BLG-free milk.


Subject(s)
Buffaloes , CRISPR-Cas Systems , Gene Editing , Lactoglobulins , Animals , Lactoglobulins/genetics , Buffaloes/genetics , Gene Editing/methods , RNA, Guide, CRISPR-Cas Systems/genetics , Milk/metabolism , Fibroblasts/metabolism
15.
Trends Cancer ; 10(3): 182-184, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38290969

ABSTRACT

Cancer remains a leading cause of morbidity and mortality, and a paradigm shift is needed to fundamentally revisit drug development efforts. Pigs share close similarities to humans and may serve as an alternative model. Recently, a transgenic 'Oncopig' line has been generated to induce solid tumors with organ specificity, opening the potential of Oncopigs as a platform for developing novel therapeutic regimens.


Subject(s)
Neoplasms , Animals , Swine , Humans , Disease Models, Animal , Animals, Genetically Modified , Neoplasms/drug therapy , Neoplasms/genetics
16.
World J Oncol ; 15(2): 149-168, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38545477

ABSTRACT

Pigs are playing an increasingly vital role as translational biomedical models for studying human pathophysiology. The annotation of the pig genome was a huge step forward in translatability of pigs as a biomedical model for various human diseases. Similarities between humans and pigs in terms of anatomy, physiology, genetics, and immunology have allowed pigs to become a comprehensive preclinical model for human diseases. With a diverse range, from craniofacial and ophthalmology to reproduction, wound healing, musculoskeletal, and cancer, pigs have provided a seminal understanding of human pathophysiology. This review focuses on the current research using pigs as preclinical models for cancer research and highlights the strengths and opportunities for studying various human cancers.

17.
CRISPR J ; 7(1): 12-28, 2024 02.
Article in English | MEDLINE | ID: mdl-38353617

ABSTRACT

Disease resistance genes in livestock provide health benefits to animals and opportunities for farmers to meet the growing demand for affordable, high-quality protein. Previously, researchers used gene editing to modify the porcine CD163 gene and demonstrated resistance to a harmful virus that causes porcine reproductive and respiratory syndrome (PRRS). To maximize potential benefits, this disease resistance trait needs to be present in commercially relevant breeding populations for multiplication and distribution of pigs. Toward this goal, a first-of-its-kind, scaled gene editing program was established to introduce a single modified CD163 allele into four genetically diverse, elite porcine lines. This effort produced healthy pigs that resisted PRRS virus infection as determined by macrophage and animal challenges. This founder population will be used for additional disease and trait testing, multiplication, and commercial distribution upon regulatory approval. Applying CRISPR-Cas to eliminate a viral disease represents a major step toward improving animal health.


Subject(s)
Porcine Reproductive and Respiratory Syndrome , Porcine respiratory and reproductive syndrome virus , Animals , Swine , Porcine respiratory and reproductive syndrome virus/genetics , Porcine Reproductive and Respiratory Syndrome/genetics , CRISPR-Cas Systems/genetics , Disease Resistance/genetics , Gene Editing , Livestock
18.
Reprod Fertil Dev ; 26(1): 65-73, 2013.
Article in English | MEDLINE | ID: mdl-24305178

ABSTRACT

The establishment of embryonic stem cells (ESCs) and gene targeting technologies in mice has revolutionised the field of genetics. The relative ease with which genes can be knocked out, and exogenous sequences introduced, has allowed the mouse to become the prime model for deciphering the genetic code. Not surprisingly, the lack of authentic ESCs has hampered the livestock genetics field and has forced animal scientists into adapting alternative technologies for genetic engineering. The recent discovery of the creation of induced pluripotent stem cells (iPSCs) by upregulation of a handful of reprogramming genes has offered renewed enthusiasm to animal geneticists. However, much like ESCs, establishing authentic iPSCs from the domestic animals is still beset with problems, including (but not limited to) the persistent expression of reprogramming genes and the lack of proven potential for differentiation into target cell types both in vitro and in vivo. Site-specific nucleases comprised of zinc finger nucleases (ZFNs), transcription activator-like effector nucleases (TALENs) and clustered regulated interspaced short palindromic repeats (CRISPRs) emerged as powerful genetic tools for precisely editing the genome, usurping the need for ESC-based genetic modifications even in the mouse. In this article, in the aftermath of these powerful genome editing technologies, the role of pluripotent stem cells in livestock genetics is discussed.


Subject(s)
Animals, Genetically Modified , Cellular Reprogramming , Clustered Regularly Interspaced Short Palindromic Repeats , Deoxyribonucleases/metabolism , Genetic Engineering/veterinary , Induced Pluripotent Stem Cells/metabolism , Livestock/genetics , Ribonucleases/metabolism , Transcription Factors/metabolism , Animals , Cell Line , Deoxyribonucleases/genetics , Gene Expression Regulation, Developmental , Gene Targeting/veterinary , Gene Transfer Techniques/veterinary , Genotype , Phenotype , Ribonucleases/genetics , Transcription Factors/genetics
19.
bioRxiv ; 2023 Aug 19.
Article in English | MEDLINE | ID: mdl-37645779

ABSTRACT

The uterine epithelium is composed of a single layer of hormone responsive polarized epithelial cells that line the lumen and form tubular glands. Endometrial epithelial organoids (EEO) can be generated from uterine epithelia and recapitulate cell composition and hormone responses in vitro . As such, the development of EEO represents a major advance for facilitating mechanistic studies in vitro . However, a major limitation for the use of EEO cultured in basement membrane extract and other hydrogels is the inner location of apical membrane, thereby hindering direct access to the apical surface of the epithelium to study interactions with the embryo or infectious agents such as viruses and bacteria. Here, a straightforward strategy was developed that successfully reverses the polarity of EEO. The result is an apical-out organoid that preserves a distinct apical-basolateral orientation and remains responsive to ovarian steroid hormones. Our investigations highlight the utility of polarity-reversed EEO to study interactions with E. coli and blastocysts. This method of generating apical-out EEO lays the foundation for developing new in vitro functional assays, particularly regarding epithelial interactions with embryos during pregnancy or other luminal constituents in a pathological or diseased state.

20.
Front Genome Ed ; 5: 1256451, 2023.
Article in English | MEDLINE | ID: mdl-37694158

ABSTRACT

Recent advances in CRISPR-Cas genome editing technology have been instrumental in improving the efficiency to produce genetically modified animal models. In this study we have combined four very promising approaches to come up with a highly effective pipeline to produce knock-in mouse and rat models. The four combined methods include: AAV-mediated DNA delivery, single-stranded DNA donor templates, 2-cell embryo modification, and CRISPR-Cas ribonucleoprotein (RNP) electroporation. Using this new combined approach, we were able to produce successfully targeted knock-in rat models containing either Cre or Flp recombinase sequences with knock-in efficiencies over 90%. Furthermore, we were able to produce a knock-in mouse model containing a Cre recombinase targeted insertion with over 50% knock-in efficiency directly comparing efficiencies to other commonly used approaches. Our modified AAV-mediated DNA delivery with 2-cell embryo CRISPR-Cas9 RNP electroporation technique has proven to be highly effective for generating both knock-in mouse and knock-in rat models.

SELECTION OF CITATIONS
SEARCH DETAIL