1
|
Summers KM. Genetic models of fibrillinopathies. Genetics 2024; 226:iyad189. [PMID: 37972149 PMCID: PMC11021029 DOI: 10.1093/genetics/iyad189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 10/16/2023] [Indexed: 11/19/2023] Open
Abstract
The fibrillinopathies represent a group of diseases in which the 10-12 nm extracellular microfibrils are disrupted by genetic variants in one of the genes encoding fibrillin molecules, large glycoproteins of the extracellular matrix. The best-known fibrillinopathy is Marfan syndrome, an autosomal dominant condition affecting the cardiovascular, ocular, skeletal, and other systems, with a prevalence of around 1 in 3,000 across all ethnic groups. It is caused by variants of the FBN1 gene, encoding fibrillin-1, which interacts with elastin to provide strength and elasticity to connective tissues. A number of mouse models have been created in an attempt to replicate the human phenotype, although all have limitations. There are also natural bovine models and engineered models in pig and rabbit. Variants in FBN2 encoding fibrillin-2 cause congenital contractural arachnodactyly and mouse models for this condition have also been produced. In most animals, including birds, reptiles, and amphibians, there is a third fibrillin, fibrillin-3 (FBN3 gene) for which the creation of models has been difficult as the gene is degenerate and nonfunctional in mice and rats. Other eukaryotes such as the nematode C. elegans and zebrafish D. rerio have a gene with some homology to fibrillins and models have been used to discover more about the function of this family of proteins. This review looks at the phenotype, inheritance, and relevance of the various animal models for the different fibrillinopathies.
Collapse
Affiliation(s)
- Kim M Summers
- Mater Research Institute-University of Queensland, Translational Research Institute, Woolloongabba QLD 4102, Australia
| |
Collapse
|
2
|
Nix JL, Schettini GP, Speckhart SL, Ealy AD, Biase FH. Ablation of OCT4 function in cattle embryos by double electroporation of CRISPR-Cas for DNA and RNA targeting (CRISPR-DART). PNAS NEXUS 2023; 2:pgad343. [PMID: 37954164 PMCID: PMC10637268 DOI: 10.1093/pnasnexus/pgad343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 10/11/2023] [Indexed: 11/14/2023]
Abstract
CRISPR-Cas ribonucleoproteins (RNPs) are important tools for gene editing in preimplantation embryos. However, the inefficient production of biallelic deletions in cattle zygotes has hindered mechanistic studies of gene function. In addition, the presence of maternal RNAs that support embryo development until embryonic genome activation may cause confounding phenotypes. Here, we aimed to improve the efficiency of biallelic deletions and deplete specific maternal RNAs in cattle zygotes using CRISPR-Cas editing technology. Two electroporation sessions with Cas9D10A RNPs targeting exon 1 and the promoter of OCT4 produced biallelic deletions in 91% of the embryos tested. In most cases, the deletions were longer than 1,000 nucleotides long. Electroporation of Cas13a RNPs prevents the production of the corresponding proteins. We electroporated Cas9D10A RNPs targeting exon 1, including the promoter region, of OCT4 in two sessions with inclusion of Cas13a RNPs targeting OCT4 mRNAs in the second session to ablate OCT4 function in cattle embryos. A lack of OCT4 resulted in embryos arresting development prior to blastocyst formation at a greater proportion (13%) than controls (31.6%, P < 0.001). The few embryos that developed past the morula stage did not form a normal inner cell mass. Transcriptome analysis of single blastocysts, confirmed to lack exon 1 and promoter region of OCT4, revealed a significant (False Discovery Rate, FDR < 0.1) reduction in transcript abundance of many genes functionally connected to stemness, including markers of pluripotency (CADHD1, DPPA4, GNL3, RRM2). The results confirm that OCT4 is a key regulator of genes that modulate pluripotency and is required to form a functional blastocyst in cattle.
Collapse
Affiliation(s)
- Jada L Nix
- School of Animal Sciences, Virginia Polytechnic Institute and State University, 175 W Campus dr, Blacksburg, VA 24061, USA
| | - Gustavo P Schettini
- School of Animal Sciences, Virginia Polytechnic Institute and State University, 175 W Campus dr, Blacksburg, VA 24061, USA
| | - Savannah L Speckhart
- School of Animal Sciences, Virginia Polytechnic Institute and State University, 175 W Campus dr, Blacksburg, VA 24061, USA
| | - Alan D Ealy
- School of Animal Sciences, Virginia Polytechnic Institute and State University, 175 W Campus dr, Blacksburg, VA 24061, USA
| | - Fernando H Biase
- School of Animal Sciences, Virginia Polytechnic Institute and State University, 175 W Campus dr, Blacksburg, VA 24061, USA
| |
Collapse
|
3
|
Park CH, Jeoung YH, Zhang L, Yeddula SGR, Park KE, Waters J, Telugu BP. Establishment, characterization, and validation of novel porcine embryonic fibroblasts as a potential source for genetic modification. Front Cell Dev Biol 2022; 10:1059710. [PMID: 36438568 PMCID: PMC9685398 DOI: 10.3389/fcell.2022.1059710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Accepted: 10/27/2022] [Indexed: 11/11/2022] Open
Abstract
Fibroblasts are the common cell type in the connective tissue-the most abundant tissue type in the body. Fibroblasts are widely used for cell culture, for the generation of induced pluripotent stem cells (iPSCs), and as nuclear donors for somatic cell nuclear transfer (SCNT). We report for the first time, the derivation of embryonic fibroblasts (EFs) from porcine embryonic outgrowths, which share similarities in morphology, culture characteristics, molecular markers, and transcriptional profile to fetal fibroblasts (FFs). We demonstrated the efficient use of EFs as nuclear donors in SCNT, for enhanced post-blastocyst development, implantation, and pregnancy outcomes. We further validated EFs as a source for CRISPR/Cas genome editing with overall editing frequencies comparable to that of FFs. Taken together, we established an alternative and efficient pipeline for genome editing and for the generation of genetically engineered animals.
Collapse
Affiliation(s)
- Chi-Hun Park
- Division of Animal Sciences, University of Missouri, Columbia, MO, United States
- RenOVAte Biosciences Inc., Reisterstown, MD, United States
| | - Young-Hee Jeoung
- Division of Animal Sciences, University of Missouri, Columbia, MO, United States
- RenOVAte Biosciences Inc., Reisterstown, MD, United States
| | - Luhui Zhang
- Division of Animal Sciences, University of Missouri, Columbia, MO, United States
| | | | - Ki-Eun Park
- RenOVAte Biosciences Inc., Reisterstown, MD, United States
| | - Jerel Waters
- RenOVAte Biosciences Inc., Reisterstown, MD, United States
| | - Bhanu P. Telugu
- Division of Animal Sciences, University of Missouri, Columbia, MO, United States
- RenOVAte Biosciences Inc., Reisterstown, MD, United States
- *Correspondence: Bhanu P. Telugu,
| |
Collapse
|
4
|
Mathur T, Kumar A, Flanagan JM, Jain A. Vascular Transcriptomics: Investigating Endothelial Activation and Vascular Dysfunction Using Blood Outgrowth Endothelial Cells, Organ-Chips, and RNA Sequencing. Curr Protoc 2022; 2:e582. [PMID: 36300922 PMCID: PMC9627633 DOI: 10.1002/cpz1.582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Vascular organ-chip or vessel-chip technology has significantly impacted our ability to model microphysiological vasculature. These biomimetic platforms have garnered significant interest from scientists and pharmaceutical companies as drug screening models. However, these models still lack the inclusion of patient-specific vasculature in the form of patient-derived endothelial cells. Blood outgrowth endothelial cells are patient blood-derived endothelial progenitors that have gained interest from the vascular biology community as an autologous endothelial cell alternative and have also been incorporated with the vessel-chip model. Next-generation sequencing techniques like RNA sequencing can further unlock the potential of personalized vessel-chips in discerning patient-specific hallmarks of endothelial dysfunction. Here we present a detailed protocol for (1) isolating blood outgrowth endothelial cells from patient blood samples, (2) culturing them in microfluidic vessel-chips, (3) isolating and preparing RNA from individual vessel-chips for sequencing, and (4) performing differential gene expression and bioinformatics analyses of vascular dysfunction and endothelial activation pathways. This method focuses specifically on identification of pathways and genes involved in vascular homeostasis and pathology, but can easily be adapted for the requirements of other systems. © 2022 Wiley Periodicals LLC. Basic Protocol 1: Isolation of blood outgrowth endothelial cells from patient blood Basic Protocol 2: Culture of blood outgrowth endothelial cells in microfluidic vessel-chips Basic Protocol 3: Isolation of RNA from autologous vessel-chips Basic Protocol 4: Differential gene expression and bioinformatics analyses of endothelial activation pathways.
Collapse
Affiliation(s)
- Tanmay Mathur
- Department of Biomedical Engineering, Texas A&M University, 101 Bizzell St, College Station, USA
| | - Ankit Kumar
- Department of Biomedical Engineering, Texas A&M University, 101 Bizzell St, College Station, USA
| | - Jonathan M. Flanagan
- Department of Pediatrics, Section of Hematology-Oncology, Baylor College of Medicine, Houston, USA
| | - Abhishek Jain
- Department of Biomedical Engineering, Texas A&M University, 101 Bizzell St, College Station, USA
- Department of Medical Physiology, College of Medicine, Texas A&M Health Science Center, Bryan, USA
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, USA
| |
Collapse
|
5
|
Eaton SL, Murdoch F, Rzechorzek NM, Thompson G, Hartley C, Blacklock BT, Proudfoot C, Lillico SG, Tennant P, Ritchie A, Nixon J, Brennan PM, Guido S, Mitchell NL, Palmer DN, Whitelaw CBA, Cooper JD, Wishart TM. Modelling Neurological Diseases in Large Animals: Criteria for Model Selection and Clinical Assessment. Cells 2022; 11:cells11172641. [PMID: 36078049 PMCID: PMC9454934 DOI: 10.3390/cells11172641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 08/19/2022] [Accepted: 08/22/2022] [Indexed: 11/16/2022] Open
Abstract
Issue: The impact of neurological disorders is recognised globally, with one in six people affected in their lifetime and few treatments to slow or halt disease progression. This is due in part to the increasing ageing population, and is confounded by the high failure rate of translation from rodent-derived therapeutics to clinically effective human neurological interventions. Improved translation is demonstrated using higher order mammals with more complex/comparable neuroanatomy. These animals effectually span this translational disparity and increase confidence in factors including routes of administration/dosing and ability to scale, such that potential therapeutics will have successful outcomes when moving to patients. Coupled with advancements in genetic engineering to produce genetically tailored models, livestock are increasingly being used to bridge this translational gap. Approach: In order to aid in standardising characterisation of such models, we provide comprehensive neurological assessment protocols designed to inform on neuroanatomical dysfunction and/or lesion(s) for large animal species. We also describe the applicability of these exams in different large animals to help provide a better understanding of the practicalities of cross species neurological disease modelling. Recommendation: We would encourage the use of these assessments as a reference framework to help standardise neurological clinical scoring of large animal models.
Collapse
Affiliation(s)
- Samantha L. Eaton
- Royal (Dick) School of Veterinary Studies and Roslin Institute, University of Edinburgh, Easter Bush Campus, Roslin, Midlothian EH25 9RG, UK
- Correspondence: (S.L.E.); (T.M.W.); Tel.: +44-(0)-131-651-9125 (S.L.E.); +44-(0)-131-651-9233 (T.M.W.)
| | - Fraser Murdoch
- Royal (Dick) School of Veterinary Studies and Roslin Institute, University of Edinburgh, Easter Bush Campus, Roslin, Midlothian EH25 9RG, UK
| | - Nina M. Rzechorzek
- Medical Research Council Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - Gerard Thompson
- Centre for Clinical Brain Sciences, University of Edinburgh, Chancellor’s Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
- Department of Clinical Neurosciences, NHS Lothian, 50 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Claudia Hartley
- Royal (Dick) School of Veterinary Studies and Roslin Institute, University of Edinburgh, Easter Bush Campus, Roslin, Midlothian EH25 9RG, UK
| | - Benjamin Thomas Blacklock
- Royal (Dick) School of Veterinary Studies and Roslin Institute, University of Edinburgh, Easter Bush Campus, Roslin, Midlothian EH25 9RG, UK
| | - Chris Proudfoot
- The Large Animal Research & Imaging Facility, Royal (Dick) School of Veterinary Studies and Roslin Institute, University of Edinburgh, Easter Bush Campus, Roslin, Midlothian EH25 9RG, UK
| | - Simon G. Lillico
- Royal (Dick) School of Veterinary Studies and Roslin Institute, University of Edinburgh, Easter Bush Campus, Roslin, Midlothian EH25 9RG, UK
| | - Peter Tennant
- The Large Animal Research & Imaging Facility, Royal (Dick) School of Veterinary Studies and Roslin Institute, University of Edinburgh, Easter Bush Campus, Roslin, Midlothian EH25 9RG, UK
| | - Adrian Ritchie
- The Large Animal Research & Imaging Facility, Royal (Dick) School of Veterinary Studies and Roslin Institute, University of Edinburgh, Easter Bush Campus, Roslin, Midlothian EH25 9RG, UK
| | - James Nixon
- The Large Animal Research & Imaging Facility, Royal (Dick) School of Veterinary Studies and Roslin Institute, University of Edinburgh, Easter Bush Campus, Roslin, Midlothian EH25 9RG, UK
| | - Paul M. Brennan
- Translational Neurosurgery, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
| | - Stefano Guido
- Royal (Dick) School of Veterinary Studies and Roslin Institute, University of Edinburgh, Easter Bush Campus, Roslin, Midlothian EH25 9RG, UK
- Bioresearch & Veterinary Services, University of Edinburgh, Chancellor’s Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Nadia L. Mitchell
- Faculty of Agriculture and Life Sciences, Lincoln University, P.O. Box 85084, Lincoln 7647, New Zealand
| | - David N. Palmer
- Faculty of Agriculture and Life Sciences, Lincoln University, P.O. Box 85084, Lincoln 7647, New Zealand
| | - C. Bruce A. Whitelaw
- Royal (Dick) School of Veterinary Studies and Roslin Institute, University of Edinburgh, Easter Bush Campus, Roslin, Midlothian EH25 9RG, UK
| | - Jonathan D. Cooper
- Departments of Pediatrics, Genetics, and Neurology, Washington University School of Medicine in St. Louis, 660 S Euclid Ave, St. Louis, MO 63110, USA
| | - Thomas M. Wishart
- Royal (Dick) School of Veterinary Studies and Roslin Institute, University of Edinburgh, Easter Bush Campus, Roslin, Midlothian EH25 9RG, UK
- Correspondence: (S.L.E.); (T.M.W.); Tel.: +44-(0)-131-651-9125 (S.L.E.); +44-(0)-131-651-9233 (T.M.W.)
| |
Collapse
|
6
|
Mice in translational neuroscience: What R we doing? Prog Neurobiol 2022; 217:102330. [PMID: 35872220 DOI: 10.1016/j.pneurobio.2022.102330] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 07/13/2022] [Accepted: 07/19/2022] [Indexed: 12/28/2022]
Abstract
Animal models play a pivotal role in translational neuroscience but recurrent problems in data collection, analyses, and interpretation, lack of biomarkers, and a tendency to over-reliance on mice have marred neuroscience progress, leading to one of the highest attrition rates in drug translation. Global initiatives to improve reproducibility and model selection are being implemented. Notwithstanding, mice are still the preferred animal species to model human brain disorders even when the translation has been shown to be limited. Non-human primates are better positioned to provide relevant translational information because of their higher brain complexity and homology to humans. Among others, lack of resources and formal training, strict legislation, and ethical issues may impede broad access to large animals. We propose that instead of increasingly restrictive legislation, more resources for training, education, husbandry, and data sharing are urgently needed. The creation of multidisciplinary teams, in which veterinarians need to play a key role, would be critical to improve translational efficiency. Furthermore, it is not usually acknowledged by researchers and regulators the value of comparative studies in lower species, that are instrumental in toxicology, target identification, and mechanistic studies. Overall, we highlight here the need for a conceptual shift in neuroscience research and policies to reach the patients.
Collapse
|
7
|
Abstract
Cardiovascular disease remains the leading cause of morbidity and mortality in the developed world. In recent decades, extraordinary effort has been devoted to defining the molecular and pathophysiological characteristics of the diseased heart and vasculature. Mouse models have been especially powerful in illuminating the complex signaling pathways, genetic and epigenetic regulatory circuits, and multicellular interactions that underlie cardiovascular disease. The advent of CRISPR genome editing has ushered in a new era of cardiovascular research and possibilities for genetic correction of disease. Next-generation sequencing technologies have greatly accelerated the identification of disease-causing mutations, and advances in gene editing have enabled the rapid modeling of these mutations in mice and patient-derived induced pluripotent stem cells. The ability to correct the genetic drivers of cardiovascular disease through delivery of gene editing components in vivo, while still facing challenges, represents an exciting therapeutic frontier. In this review, we provide an overview of cardiovascular disease mechanisms and the potential applications of CRISPR genome editing for disease modeling and correction. We also discuss the extent to which mice can faithfully model cardiovascular disease and the opportunities and challenges that lie ahead.
Collapse
Affiliation(s)
- Ning Liu
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas
| | - Eric N Olson
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas
| |
Collapse
|
8
|
Rim CS, Kim YS, Rim CH, Ri YJ, Choe JS, Kim DS, Kim GS, Il Ri J, Kim RC, Chen H, Xiao L, Fu Z, Pak YJ, Jong UM. Effect of roscovitine pretreatment for increased utilization of small follicle-derived oocytes on developmental competence of somatic cell nuclear transfer embryos in pigs. Anim Reprod Sci 2022; 241:106987. [DOI: 10.1016/j.anireprosci.2022.106987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 04/23/2022] [Accepted: 05/01/2022] [Indexed: 11/25/2022]
|
9
|
Garcia-Prieto CA, Álvarez-Errico D, Musulen E, Bueno-Costa A, N Vazquez B, Vaquero A, Esteller M. Validation of a DNA methylation microarray for 285,000 CpG sites in the mouse genome. Epigenetics 2022; 17:1677-1685. [PMID: 35297293 DOI: 10.1080/15592294.2022.2053816] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The mouse has been extensively used as the model organism in many studies to characterize biological pathways, drug effects and to mimic human diseases. The similar DNA sequences between both species facilitates these type of experiments. However, much less is known about the mouse epigenome, particularly for DNA methylation. Progress in delivering mouse DNA methylomes has been slow due to the currently available time-consuming and expensive methodologies. Following the great acceptance of the human DNA methylation microarrays, we have herein validated a newly developed DNA methylation microarray (Infinium Mouse Methylation BeadChip) that interrogates 280,754 unique CpG sites within the mouse genome. The CpGs included in the platform cover CpG islands, shores, shelfs and open sea sequences; and loci surrounding transcription start sites and gene bodies. From the functional standpoint, mouse ENCODE representative DNase hypersensitivity sites (rDHSs) and candidate cis-Regulatory Elements (cCREs) are also included. Herein, we show that the profiled mouse DNA methylation microarray provides reliable values among technical replicates; matched results from fresh frozen versus formalin-fixed samples; detects hemimethylated X-chromosome and imprinted CpG sites; and is able to determine CpG methylation changes in mouse cell lines treated with a DNA demethylating agent or upon genetic disruption of a DNA methyltransferase. Most important, using unsupervised hierarchical clustering and t-SNE approaches, the platform is able to classify all types of normal mouse tissues and organs. These data underscore the great features of the assessed microarray to obtain comprehensive DNA methylation profiles of the mouse genome.
Collapse
Affiliation(s)
- Carlos A Garcia-Prieto
- Cancer Epigenetics Group, Josep Carreras Leukaemia Research Institute (IJC), Badalona, Barcelona, Catalonia, Spain.,Life Sciences Department, Barcelona Supercomputing Center (BSC), Barcelona, Catalonia Spain
| | - Damiana Álvarez-Errico
- Cancer Epigenetics Group, Josep Carreras Leukaemia Research Institute (IJC), Badalona, Barcelona, Catalonia, Spain
| | - Eva Musulen
- Cancer Epigenetics Group, Josep Carreras Leukaemia Research Institute (IJC), Badalona, Barcelona, Catalonia, Spain.,Department of Pathology, Hospital Universitari General de Catalunya Grupo-QuirónSalud, Sant Cugat del Vallès, Barcelona, Catalonia, Spain
| | - Alberto Bueno-Costa
- Cancer Epigenetics Group, Josep Carreras Leukaemia Research Institute (IJC), Badalona, Barcelona, Catalonia, Spain
| | - Berta N Vazquez
- Chromatin Biology Group, Josep Carreras Leukaemia Research Institute (IJC), Badalona, Barcelona, Catalonia, Spain
| | - Alejandro Vaquero
- Chromatin Biology Group, Josep Carreras Leukaemia Research Institute (IJC), Badalona, Barcelona, Catalonia, Spain
| | - Manel Esteller
- Cancer Epigenetics Group, Josep Carreras Leukaemia Research Institute (IJC), Badalona, Barcelona, Catalonia, Spain.,Centro de Investigacion Biomedica en Red Cancer (CIBERONC), Madrid, Spain.,Institucio Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Catalonia, Spain.,Physiological Sciences Department, School of Medicine and Health Sciences, University of Barcelona (UB), Barcelona, Catalonia, Spain
| |
Collapse
|
10
|
Navarro-Serna S, Piñeiro-Silva C, Luongo C, Parrington J, Romar R, Gadea J. Effect of Aphidicolin, a Reversible Inhibitor of Eukaryotic Nuclear DNA Replication, on the Production of Genetically Modified Porcine Embryos by CRISPR/Cas9. Int J Mol Sci 2022; 23:ijms23042135. [PMID: 35216252 PMCID: PMC8880323 DOI: 10.3390/ijms23042135] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 02/09/2022] [Accepted: 02/12/2022] [Indexed: 01/27/2023] Open
Abstract
Mosaicism is the most important limitation for one-step gene editing in embryos by CRISPR/Cas9 because cuts and repairs sometimes take place after the first DNA replication of the zygote. To try to minimize the risk of mosaicism, in this study a reversible DNA replication inhibitor was used after the release of CRISPR/Cas9 in the cell. There is no previous information on the use of aphidicolin in porcine embryos, so the reversible inhibition of DNA replication and the effect on embryo development of different concentrations of this drug was first evaluated. The effect of incubation with aphidicolin was tested with CRISPR/Cas9 at different concentrations and different delivery methodologies. As a result, the reversible inhibition of DNA replication was observed, and it was concentration dependent. An optimal concentration of 0.5 μM was established and used for subsequent experiments. Following the use of this drug with CRISPR/Cas9, a halving of mosaicism was observed together with a detrimental effect on embryo development. In conclusion, the use of reversible inhibition of DNA replication offers a way to reduce mosaicism. Nevertheless, due to the reduction in embryo development, it would be necessary to reach a balance for its use to be feasible.
Collapse
Affiliation(s)
- Sergio Navarro-Serna
- Department of Physiology, International Excellence Campus for Higher Education and Research “Campus Mare Nostrum”, University of Murcia, 30100 Murcia, Spain; (S.N.-S.); (C.P.-S.); (C.L.); (R.R.)
- Institute for Biomedical Research of Murcia (IMIB-Arrixaca), 30120 Murcia, Spain
| | - Celia Piñeiro-Silva
- Department of Physiology, International Excellence Campus for Higher Education and Research “Campus Mare Nostrum”, University of Murcia, 30100 Murcia, Spain; (S.N.-S.); (C.P.-S.); (C.L.); (R.R.)
- Institute for Biomedical Research of Murcia (IMIB-Arrixaca), 30120 Murcia, Spain
| | - Chiara Luongo
- Department of Physiology, International Excellence Campus for Higher Education and Research “Campus Mare Nostrum”, University of Murcia, 30100 Murcia, Spain; (S.N.-S.); (C.P.-S.); (C.L.); (R.R.)
- Institute for Biomedical Research of Murcia (IMIB-Arrixaca), 30120 Murcia, Spain
| | - John Parrington
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK;
| | - Raquel Romar
- Department of Physiology, International Excellence Campus for Higher Education and Research “Campus Mare Nostrum”, University of Murcia, 30100 Murcia, Spain; (S.N.-S.); (C.P.-S.); (C.L.); (R.R.)
- Institute for Biomedical Research of Murcia (IMIB-Arrixaca), 30120 Murcia, Spain
| | - Joaquín Gadea
- Department of Physiology, International Excellence Campus for Higher Education and Research “Campus Mare Nostrum”, University of Murcia, 30100 Murcia, Spain; (S.N.-S.); (C.P.-S.); (C.L.); (R.R.)
- Institute for Biomedical Research of Murcia (IMIB-Arrixaca), 30120 Murcia, Spain
- Correspondence:
| |
Collapse
|
11
|
Xiang D, Jia B, Guo J, Shao Q, Hong Q, Wei H, Quan G, Wu G. Transcriptome Analysis of mRNAs and Long Non-Coding RNAs During Subsequent Embryo Development of Porcine Cloned Zygotes After Vitrification. Front Genet 2022; 12:753327. [PMID: 34976007 PMCID: PMC8718616 DOI: 10.3389/fgene.2021.753327] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 11/17/2021] [Indexed: 11/15/2022] Open
Abstract
Cryopreservation of porcine cloned zygotes has important implications for biotechnology and biomedicine research; however, lower embryo developmental potential remains an urgent problem to be resolved. For exploring the sublethal cryodamages during embryo development, this study was designed to acquire the mRNA and long non-coding RNA (lncRNA) profiles of 2-cells, 4-cells and blastocysts derived from vitrified porcine cloned zygotes using transcriptome sequencing. We identified 167 differentially expressed (DE) mRNAs and 516 DE lncRNAs in 2-cell stage, 469 DE mRNAs and 565 lncRNAs in 4-cell stage, and 389 DE mRNAs and 816 DE lncRNAs in blastocyst stage. Functional enrichment analysis revealed that the DE mRNAs during embryo development were involved in many regulatory mechanisms related to cell cycle, cell proliferation, apoptosis, metabolism and others. Moreover, the target genes of DE lncRNAs in the three embryonic stages were also enriched in many key GO terms or pathways such as “defense response”, “linoleic acid metabolic process”, “embryonic axis specification”, “negative regulation of protein neddylation”, etc., In conclusion, the present study provided comprehensive transcriptomic data about mRNAs and lncRNAs for the vitrified porcine cloned zygotes during different developmental stages, which contributed to further understand the potential cryodamage mechanisms responsible for impaired embryo development.
Collapse
Affiliation(s)
- Decai Xiang
- Yunnan Provincial Genebank of Livestock and Poultry Genetic Resources, Yunnan Provincial Engineering Laboratory of Animal Genetic Resource Conservation and Germplasm Enhancement, Yunnan Animal Science and Veterinary Institute, Kunming, China
| | - Baoyu Jia
- Key Laboratory of Animal Gene Editing and Animal Cloning in Yunnan Province, College of Veterinary Medicine, Yunnan Agricultural University, Kunming, China
| | - Jianxiong Guo
- Key Laboratory of Animal Gene Editing and Animal Cloning in Yunnan Province, College of Veterinary Medicine, Yunnan Agricultural University, Kunming, China
| | - Qingyong Shao
- Yunnan Provincial Genebank of Livestock and Poultry Genetic Resources, Yunnan Provincial Engineering Laboratory of Animal Genetic Resource Conservation and Germplasm Enhancement, Yunnan Animal Science and Veterinary Institute, Kunming, China
| | - Qionghua Hong
- Yunnan Provincial Genebank of Livestock and Poultry Genetic Resources, Yunnan Provincial Engineering Laboratory of Animal Genetic Resource Conservation and Germplasm Enhancement, Yunnan Animal Science and Veterinary Institute, Kunming, China
| | - Hongjiang Wei
- Key Laboratory of Animal Gene Editing and Animal Cloning in Yunnan Province, College of Veterinary Medicine, Yunnan Agricultural University, Kunming, China
| | - Guobo Quan
- Yunnan Provincial Genebank of Livestock and Poultry Genetic Resources, Yunnan Provincial Engineering Laboratory of Animal Genetic Resource Conservation and Germplasm Enhancement, Yunnan Animal Science and Veterinary Institute, Kunming, China
| | - Guoquan Wu
- Yunnan Provincial Genebank of Livestock and Poultry Genetic Resources, Yunnan Provincial Engineering Laboratory of Animal Genetic Resource Conservation and Germplasm Enhancement, Yunnan Animal Science and Veterinary Institute, Kunming, China
| |
Collapse
|
12
|
Zárybnický T, Heikkinen A, Kangas SM, Karikoski M, Martínez-Nieto GA, Salo MH, Uusimaa J, Vuolteenaho R, Hinttala R, Sipilä P, Kuure S. Modeling Rare Human Disorders in Mice: The Finnish Disease Heritage. Cells 2021; 10:cells10113158. [PMID: 34831381 PMCID: PMC8621025 DOI: 10.3390/cells10113158] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 11/04/2021] [Accepted: 11/06/2021] [Indexed: 12/31/2022] Open
Abstract
The modification of genes in animal models has evidently and comprehensively improved our knowledge on proteins and signaling pathways in human physiology and pathology. In this review, we discuss almost 40 monogenic rare diseases that are enriched in the Finnish population and defined as the Finnish disease heritage (FDH). We will highlight how gene-modified mouse models have greatly facilitated the understanding of the pathological manifestations of these diseases and how some of the diseases still lack proper models. We urge the establishment of subsequent international consortiums to cooperatively plan and carry out future human disease modeling strategies. Detailed information on disease mechanisms brings along broader understanding of the molecular pathways they act along both parallel and transverse to the proteins affected in rare diseases, therefore also aiding understanding of common disease pathologies.
Collapse
Affiliation(s)
- Tomáš Zárybnický
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, P.O. Box 63, 00014 Helsinki, Finland;
| | - Anne Heikkinen
- Biocenter Oulu, University of Oulu, P.O. Box 5000, 90014 Oulu, Finland; (A.H.); (S.M.K.); (M.H.S.); (R.V.)
- Oulu Center for Cell-Matrix Research, Faculty of Biochemistry and Molecular Medicine, University of Oulu, P.O. Box 8000, 90014 Oulu, Finland
| | - Salla M. Kangas
- Biocenter Oulu, University of Oulu, P.O. Box 5000, 90014 Oulu, Finland; (A.H.); (S.M.K.); (M.H.S.); (R.V.)
- PEDEGO Research Unit, University of Oulu, P.O. Box 8000, 90014 Oulu, Finland;
- Medical Research Center, Oulu University Hospital, University of Oulu, P.O. Box 5000, 90014 Oulu, Finland
| | - Marika Karikoski
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, 20520 Turku, Finland; (M.K.); (G.A.M.-N.)
| | - Guillermo Antonio Martínez-Nieto
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, 20520 Turku, Finland; (M.K.); (G.A.M.-N.)
- Turku Center for Disease Modelling (TCDM), Institute of Biomedicine, University of Turku, 20520 Turku, Finland
| | - Miia H. Salo
- Biocenter Oulu, University of Oulu, P.O. Box 5000, 90014 Oulu, Finland; (A.H.); (S.M.K.); (M.H.S.); (R.V.)
- PEDEGO Research Unit, University of Oulu, P.O. Box 8000, 90014 Oulu, Finland;
- Medical Research Center, Oulu University Hospital, University of Oulu, P.O. Box 5000, 90014 Oulu, Finland
| | - Johanna Uusimaa
- PEDEGO Research Unit, University of Oulu, P.O. Box 8000, 90014 Oulu, Finland;
- Medical Research Center, Oulu University Hospital, University of Oulu, P.O. Box 5000, 90014 Oulu, Finland
- Clinic for Children and Adolescents, Division of Pediatric Neurology, Oulu University Hospital, P.O. Box 20, 90029 Oulu, Finland
| | - Reetta Vuolteenaho
- Biocenter Oulu, University of Oulu, P.O. Box 5000, 90014 Oulu, Finland; (A.H.); (S.M.K.); (M.H.S.); (R.V.)
| | - Reetta Hinttala
- Biocenter Oulu, University of Oulu, P.O. Box 5000, 90014 Oulu, Finland; (A.H.); (S.M.K.); (M.H.S.); (R.V.)
- PEDEGO Research Unit, University of Oulu, P.O. Box 8000, 90014 Oulu, Finland;
- Medical Research Center, Oulu University Hospital, University of Oulu, P.O. Box 5000, 90014 Oulu, Finland
- Correspondence: (R.H.); (P.S.); (S.K.)
| | - Petra Sipilä
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, 20520 Turku, Finland; (M.K.); (G.A.M.-N.)
- Turku Center for Disease Modelling (TCDM), Institute of Biomedicine, University of Turku, 20520 Turku, Finland
- Correspondence: (R.H.); (P.S.); (S.K.)
| | - Satu Kuure
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, P.O. Box 63, 00014 Helsinki, Finland;
- GM-Unit, Laboratory Animal Center, Helsinki Institute of Life Science, University of Helsinki, 00790 Helsinki, Finland
- Correspondence: (R.H.); (P.S.); (S.K.)
| |
Collapse
|
13
|
Vats P, Kaushik R, Rawat N, Sharma A, Sharma T, Dua D, Singh MK, Palta P, Singla SK, Manik RS, Chauhan MS. Production of Transgenic Handmade Cloned Goat ( Capra hircus) Embryos by Targeted Integration into Rosa 26 Locus Using Transcription Activator-like Effector Nucleases. Cell Reprogram 2021; 23:250-262. [PMID: 34348041 DOI: 10.1089/cell.2021.0011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Transgenic goats are ideal bioreactors for the production of therapeutic proteins in their mammary glands. However, random integration of the transgene within-host genome often culminates in unstable expression and unpredictable phenotypes. Targeting desired genes to a safe locus in the goat genome using advanced targeted genome-editing tools, such as transcription activator-like effector nucleases (TALENs) might assist in overcoming these hurdles. We identified Rosa 26 locus, a safe harbor for transgene integration, on chromosome 22 in the goat genome for the first time. We further demonstrate that TALEN-mediated targeting of GFP gene cassette at Rosa 26 locus exhibited stable and ubiquitous expression of GFP gene in goat fetal fibroblasts (GFFs) and after that, transgenic cloned embryos generated by handmade cloning (HMC). The transfection of GFFs by the TALEN pair resulted in 13.30% indel frequency at the target site. Upon cotransfection with TALEN and donor vectors, four correctly targeted cell colonies were obtained and all of them showed monoallelic gene insertions. The blastocyst rate for transgenic cloned embryos (3.92% ± 1.12%) was significantly (p < 0.05) lower than cloned embryos (7.84% ± 0.68%) used as control. Concomitantly, 2 out of 15 embryos of morulae and blastocyst stage (13.30%) exhibited site-specific integration. In conclusion, the present study demonstrates TALEN-mediated transgene integration at Rosa 26 locus in caprine fetal fibroblasts and the generation of transgenic cloned embryos using HMC.
Collapse
Affiliation(s)
- Preeti Vats
- Embryo Biotechnology Laboratory, Animal Biotechnology Centre, ICAR-National Dairy Research Institute, Karnal, India
| | - Ramakant Kaushik
- Embryo Biotechnology Laboratory, Animal Biotechnology Centre, ICAR-National Dairy Research Institute, Karnal, India
| | - Nidhi Rawat
- Embryo Biotechnology Laboratory, Animal Biotechnology Centre, ICAR-National Dairy Research Institute, Karnal, India
| | - Ankur Sharma
- Embryo Biotechnology Laboratory, Animal Biotechnology Centre, ICAR-National Dairy Research Institute, Karnal, India
| | - Tushar Sharma
- Embryo Biotechnology Laboratory, Animal Biotechnology Centre, ICAR-National Dairy Research Institute, Karnal, India
| | - Diksha Dua
- Embryo Biotechnology Laboratory, Animal Biotechnology Centre, ICAR-National Dairy Research Institute, Karnal, India
| | - Manoj Kumar Singh
- Embryo Biotechnology Laboratory, Animal Biotechnology Centre, ICAR-National Dairy Research Institute, Karnal, India
| | - Prabhat Palta
- Embryo Biotechnology Laboratory, Animal Biotechnology Centre, ICAR-National Dairy Research Institute, Karnal, India
| | - Suresh Kumar Singla
- Embryo Biotechnology Laboratory, Animal Biotechnology Centre, ICAR-National Dairy Research Institute, Karnal, India
| | - Radhey Sham Manik
- Embryo Biotechnology Laboratory, Animal Biotechnology Centre, ICAR-National Dairy Research Institute, Karnal, India
| | - Manmohan Singh Chauhan
- Embryo Biotechnology Laboratory, Animal Biotechnology Centre, ICAR-National Dairy Research Institute, Karnal, India
| |
Collapse
|
14
|
Li JT, Zeng N, Yan ZP, Liao T, Ni GX. A review of applications of metabolomics in osteoarthritis. Clin Rheumatol 2021; 40:2569-2579. [PMID: 33219452 DOI: 10.1007/s10067-020-05511-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 11/10/2020] [Accepted: 11/15/2020] [Indexed: 02/08/2023]
Abstract
Osteoarthritis (OA) represents the most prevalent and disabling arthritis worldwide due to its heterogeneous and progressive articular degradation. However, effective and timely diagnosis and fundamental treatment for this disorder are lacking. Metabolomics, a growing field in life science research in recent years, has the potential to detect many metabolites and thus explains the underlying pathophysiological processes. Hence, new specific metabolic markers and related metabolic pathways can be identified for OA. In this review, we aimed to provide an overview of studies related to the metabolomics of OA in animal models and humans to describe the metabolic changes and related pathways for OA. The present metabolomics studies reveal that the pathogenesis of OA may be significantly related to perturbations of amino acid metabolism. These altered amino acids (e.g., branched-chain amino acids, arginine, and alanine), as well as phospholipids, were identified as potential biomarkers to distinguish patients with OA from healthy individuals.
Collapse
Affiliation(s)
- Jie-Ting Li
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, People's Republic of China
| | - Ni Zeng
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, People's Republic of China
| | - Zhi-Peng Yan
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, People's Republic of China
| | - Tao Liao
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, People's Republic of China
| | - Guo-Xin Ni
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing, 100084, People's Republic of China.
| |
Collapse
|
15
|
Jin L, Tang Q, Hu S, Chen Z, Zhou X, Zeng B, Wang Y, He M, Li Y, Gui L, Shen L, Long K, Ma J, Wang X, Chen Z, Jiang Y, Tang G, Zhu L, Liu F, Zhang B, Huang Z, Li G, Li D, Gladyshev VN, Yin J, Gu Y, Li X, Li M. A pig BodyMap transcriptome reveals diverse tissue physiologies and evolutionary dynamics of transcription. Nat Commun 2021; 12:3715. [PMID: 34140474 PMCID: PMC8211698 DOI: 10.1038/s41467-021-23560-8] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 05/04/2021] [Indexed: 12/13/2022] Open
Abstract
A comprehensive transcriptomic survey of pigs can provide a mechanistic understanding of tissue specialization processes underlying economically valuable traits and accelerate their use as a biomedical model. Here we characterize four transcript types (lncRNAs, TUCPs, miRNAs, and circRNAs) and protein-coding genes in 31 adult pig tissues and two cell lines. We uncover the transcriptomic variability among 47 skeletal muscles, and six adipose depots linked to their different origins, metabolism, cell composition, physical activity, and mitochondrial pathways. We perform comparative analysis of the transcriptomes of seven tissues from pigs and nine other vertebrates to reveal that evolutionary divergence in transcription potentially contributes to lineage-specific biology. Long-range promoter–enhancer interaction analysis in subcutaneous adipose tissues across species suggests evolutionarily stable transcription patterns likely attributable to redundant enhancers buffering gene expression patterns against perturbations, thereby conferring robustness during speciation. This study can facilitate adoption of the pig as a biomedical model for human biology and disease and uncovers the molecular bases of valuable traits. A comprehensive transcriptomic survey of the pig could enable mechanistic understanding of tissue specialization and accelerate its use as a biomedical model. Here the authors characterize four distinct transcript types in 31 adult pig tissues to dissect their distinct structural and transcriptional features and uncover transcriptomic variability related to tissue physiology.
Collapse
Affiliation(s)
- Long Jin
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Qianzi Tang
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, China.
| | - Silu Hu
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Zhongxu Chen
- Department of Life Science, Tcuni Inc., Chengdu, Sichuan, China
| | - Xuming Zhou
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Bo Zeng
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Yuhao Wang
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Mengnan He
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Yan Li
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Lixuan Gui
- Department of Life Science, Tcuni Inc., Chengdu, Sichuan, China
| | - Linyuan Shen
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Keren Long
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Jideng Ma
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Xun Wang
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Zhengli Chen
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Yanzhi Jiang
- College of Life Science, Sichuan Agricultural University, Ya'an, Sichuan, China
| | - Guoqing Tang
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Li Zhu
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Fei Liu
- Information and Educational Technology Center, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Bo Zhang
- Ya'an Digital Economy Operation Company, Ya'an, Sichuan, China
| | - Zhiqing Huang
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Guisen Li
- Renal Department and Nephrology Institute, Sichuan Provincial People's Hospital, Chengdu, Sichuan, China
| | - Diyan Li
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Vadim N Gladyshev
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Jingdong Yin
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Yiren Gu
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu, Sichuan, China
| | - Xuewei Li
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Mingzhou Li
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, China.
| |
Collapse
|
16
|
Knockout of the HMG domain of the porcine SRY gene causes sex reversal in gene-edited pigs. Proc Natl Acad Sci U S A 2021; 118:2008743118. [PMID: 33443157 PMCID: PMC7812820 DOI: 10.1073/pnas.2008743118] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The present work characterizes the porcine sex-determining region on the Y chromosome (SRY) gene and demonstrates its pivotal role in sex determination. We provide evidence that genetically male pigs with a knockout of the SRY gene undergo sex reversal of the external and internal genitalia. This discovery of SRY as the main switch for sex determination in pigs may provide an alternative for surgical castration in pig production, preventing boar taint. As the pig shares many genetic, physiological, and anatomical similarities with humans, it also provides a suitable large animal model for human sex reversal syndromes, allowing for the development of new interventions for human sex disorders. The sex-determining region on the Y chromosome (SRY) is thought to be the central genetic element of male sex development in mammals. Pathogenic modifications within the SRY gene are associated with a male-to-female sex reversal syndrome in humans and other mammalian species, including rabbits and mice. However, the underlying mechanisms are largely unknown. To understand the biological function of the SRY gene, a site-directed mutational analysis is required to investigate associated phenotypic changes at the molecular, cellular, and morphological level. Here, we successfully generated a knockout of the porcine SRY gene by microinjection of two CRISPR-Cas ribonucleoproteins, targeting the centrally located “high mobility group” (HMG), followed by a frameshift mutation of the downstream SRY sequence. This resulted in the development of genetically male (XY) pigs with complete external and internal female genitalia, which, however, were significantly smaller than in 9-mo-old age-matched control females. Quantitative digital PCR analysis revealed a duplication of the SRY locus in Landrace pigs similar to the known palindromic duplication in Duroc breeds. Our study demonstrates the central role of the HMG domain in the SRY gene in male porcine sex determination. This proof-of-principle study could assist in solving the problem of sex preference in agriculture to improve animal welfare. Moreover, it establishes a large animal model that is more comparable to humans with regard to genetics, physiology, and anatomy, which is pivotal for longitudinal studies to unravel mammalian sex determination and relevant for the development of new interventions for human sex development disorders.
Collapse
|
17
|
Barazesh M, Mohammadi S, Bahrami Y, Mokarram P, Morowvat MH, Saidijam M, Karimipoor M, Kavousipour S, Vosoughi AR, Khanaki K. CRISPR/Cas9 Technology as a Modern Genetic Manipulation Tool for Recapitulating of Neurodegenerative Disorders in Large Animal Models. Curr Gene Ther 2021; 21:130-148. [PMID: 33319680 DOI: 10.2174/1566523220666201214115024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 11/12/2020] [Accepted: 11/23/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Neurodegenerative diseases are often the consequence of alterations in structures and functions of the Central Nervous System (CNS) in patients. Despite obtaining massive genomic information concerning the molecular basis of these diseases and since the neurological disorders are multifactorial, causal connections between pathological pathways at the molecular level and CNS disorders development have remained obscure and need to be elucidated to a great extent. OBJECTIVE Animal models serve as accessible and valuable tools for understanding and discovering the roles of causative factors in the development of neurodegenerative disorders and finding appropriate treatments. Contrary to rodents and other small animals, large animals, especially non-human primates (NHPs), are remarkably similar to humans; hence, they establish suitable models for recapitulating the main human's neuropathological manifestations that may not be seen in rodent models. In addition, they serve as useful models to discover effective therapeutic targets for neurodegenerative disorders due to their similarity to humans in terms of physiology, evolutionary distance, anatomy, and behavior. METHODS In this review, we recommend different strategies based on the CRISPR-Cas9 system for generating animal models of human neurodegenerative disorders and explaining in vivo CRISPR-Cas9 delivery procedures that are applied to disease models for therapeutic purposes. RESULTS With the emergence of CRISPR/Cas9 as a modern specific gene-editing technology in the field of genetic engineering, genetic modification procedures such as gene knock-in and knock-out have become increasingly easier compared to traditional gene targeting techniques. Unlike the old techniques, this versatile technology can efficiently generate transgenic large animal models without the need to complicate lab instruments. Hence, these animals can accurately replicate the signs of neurodegenerative disorders. CONCLUSION Preclinical applications of CRISPR/Cas9 gene-editing technology supply a unique opportunity to establish animal models of neurodegenerative disorders with high accuracy and facilitate perspectives for breakthroughs in the research on the nervous system disease therapy and drug discovery. Furthermore, the useful outcomes of CRISPR applications in various clinical phases are hopeful for their translation to the clinic in a short time.
Collapse
Affiliation(s)
- Mahdi Barazesh
- School of Paramedical, Gerash University of Medical Sciences, Gerash, Iran
| | - Shiva Mohammadi
- Department of Medical Biotechnology, School of Medicine, Lorestan University of Medical Sciences, Khoram Abad, Iran
| | - Yadollah Bahrami
- Molecular Biology Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Pooneh Mokarram
- Autophagy Research center, Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Massoud Saidijam
- Department of Molecular Medicine and Genetics, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Morteza Karimipoor
- Molecular Medicine Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Soudabeh Kavousipour
- Molecular Medicine Research Center, Hormozgan Health Institute, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Amir Reza Vosoughi
- Department of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, Canada
| | - Korosh Khanaki
- Medical Biotechnology Research Center, Paramedicine Faculty, Guilan University of Medical Sciences, Rasht, Iran
| |
Collapse
|
18
|
Hornyik T, Rieder M, Castiglione A, Major P, Baczko I, Brunner M, Koren G, Odening KE. Transgenic rabbit models for cardiac disease research. Br J Pharmacol 2021; 179:938-957. [PMID: 33822374 DOI: 10.1111/bph.15484] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 02/23/2021] [Accepted: 03/11/2021] [Indexed: 12/20/2022] Open
Abstract
To study the pathophysiology of human cardiac diseases and to develop novel treatment strategies, complex interactions of cardiac cells on cellular, tissue and on level of the whole heart need to be considered. As in vitro cell-based models do not depict the complexity of the human heart, animal models are used to obtain insights that can be translated to human diseases. Mice are the most commonly used animals in cardiac research. However, differences in electrophysiological and mechanical cardiac function and a different composition of electrical and contractile proteins limit the transferability of the knowledge gained. Moreover, the small heart size and fast heart rate are major disadvantages. In contrast to rodents, electrophysiological, mechanical and structural cardiac characteristics of rabbits resemble the human heart more closely, making them particularly suitable as an animal model for cardiac disease research. In this review, various methodological approaches for the generation of transgenic rabbits for cardiac disease research, such as pronuclear microinjection, the sleeping beauty transposon system and novel genome-editing methods (ZFN and CRISPR/Cas9)will be discussed. In the second section, we will introduce the different currently available transgenic rabbit models for monogenic cardiac diseases (such as long QT syndrome, short-QT syndrome and hypertrophic cardiomyopathy) in detail, especially in regard to their utility to increase the understanding of pathophysiological disease mechanisms and novel treatment options.
Collapse
Affiliation(s)
- Tibor Hornyik
- Translational Cardiology, Department of Cardiology, Inselspital, Bern University Hospital, and Institute of Physiology, University of Bern, Bern, Switzerland.,Department of Cardiology and Angiology I, University Heart Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Marina Rieder
- Translational Cardiology, Department of Cardiology, Inselspital, Bern University Hospital, and Institute of Physiology, University of Bern, Bern, Switzerland
| | - Alessandro Castiglione
- Translational Cardiology, Department of Cardiology, Inselspital, Bern University Hospital, and Institute of Physiology, University of Bern, Bern, Switzerland
| | - Peter Major
- Institute for Genetics and Biotechnology, Hungarian University of Agriculture and Life Sciences, Gödöllő, Hungary
| | - Istvan Baczko
- Department of Pharmacology and Pharmacotherapy, University of Szeged, Szeged, Hungary
| | - Michael Brunner
- Department of Cardiology and Angiology I, University Heart Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Department of Cardiology and Medical Intensive Care, St. Josefskrankenhaus, Freiburg, Germany
| | - Gideon Koren
- Cardiovascular Research Center, Division of Cardiology, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, Rhode Island, USA
| | - Katja E Odening
- Translational Cardiology, Department of Cardiology, Inselspital, Bern University Hospital, and Institute of Physiology, University of Bern, Bern, Switzerland.,Department of Cardiology and Angiology I, University Heart Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
19
|
Teplitz GM, Shi M, Sirard MA, Lombardo DM. Coculture of porcine luteal cells during in vitro porcine oocyte maturation affects blastocyst gene expression and developmental potential. Theriogenology 2021; 166:124-134. [PMID: 33735666 DOI: 10.1016/j.theriogenology.2021.02.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 01/26/2021] [Accepted: 02/22/2021] [Indexed: 12/22/2022]
Abstract
Oocyte maturation in culture is still the weakest part of in vitro fertilization (IVF) and coculture with somatic cells may be an alternative to improve suboptimal culture conditions, especially in the pig in which maturation takes more than 44 h. In the present study, we investigated the effect of a coculture system of porcine luteal cells (PLC) during in vitro maturation (IVM) on embryo development and gene expression. Cumulus-oocyte complexes were matured in vitro in TCM-199 with human menopausal gonadotrophin (control) and in coculture with PLC. IVF was performed with frozen-thawed boar semen in Tris-buffered medium. Presumptive zygotes were cultured in PZM for 7 days. The coculture with PLC significantly increased blastocysts rates. Gene expression changes were measured with a porcine embryo-specific microarray and confirmed by RT-qPCR. The global transcription pattern of embryos developing after PLC coculture exhibited overall downregulation of gene expression. Following global gene expression pattern analysis, genes associated with lipid metabolism, mitochondrial function, endoplasmic reticulum stress, and apoptosis were found downregulated, and genes associated with cell cycle and proliferation were found upregulated in the PLC coculture. Canonical pathway analysis by Ingenuity Pathway revealed that differential expression transcripts were associated with the sirtuin signaling pathway, oxidative phosphorylation pathway, cytokines and ephrin receptor signaling. To conclude, the coculture system of PLC during IVM has a lasting effect on the embryo until the blastocyst stage, modifying gene expression, with a positive effect on embryo development. Our model could be an alternative to replace the conventional maturation medium with gonadotrophins with higher rates of embryo development, a key issue in porcine in vitro embryo production.
Collapse
Affiliation(s)
- G M Teplitz
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Godoy Cruz 2290 C1425TQB, Buenos Aires, Argentina; Universidad de Buenos Aires, Facultad de Ciencias Veterinarias, Instituto de Investigación y Tecnología en Reproducción Animal, Cátedra de Histología y Embriología, Chorroarín 280 C1427CWO, Buenos Aires, Argentina
| | - M Shi
- Departement des Sciences Animales, Centre de Recherche en Reproduction, Développement et Santé Inter-générationnelle (CRDSI), Université Laval, Quebec, Canada. Pavillon Des Services, Local 2732, Université Laval, Quebec G1V 0A6, Canada
| | - M A Sirard
- Departement des Sciences Animales, Centre de Recherche en Reproduction, Développement et Santé Inter-générationnelle (CRDSI), Université Laval, Quebec, Canada. Pavillon Des Services, Local 2732, Université Laval, Quebec G1V 0A6, Canada
| | - D M Lombardo
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Godoy Cruz 2290 C1425TQB, Buenos Aires, Argentina; Universidad de Buenos Aires, Facultad de Ciencias Veterinarias, Instituto de Investigación y Tecnología en Reproducción Animal, Cátedra de Histología y Embriología, Chorroarín 280 C1427CWO, Buenos Aires, Argentina.
| |
Collapse
|
20
|
Osum SH, Watson AL, Largaespada DA. Spontaneous and Engineered Large Animal Models of Neurofibromatosis Type 1. Int J Mol Sci 2021; 22:1954. [PMID: 33669386 PMCID: PMC7920315 DOI: 10.3390/ijms22041954] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 02/02/2021] [Accepted: 02/04/2021] [Indexed: 12/13/2022] Open
Abstract
Animal models are crucial to understanding human disease biology and developing new therapies. By far the most common animal used to investigate prevailing questions about human disease is the mouse. Mouse models are powerful tools for research as their small size, limited lifespan, and defined genetic background allow researchers to easily manipulate their genome and maintain large numbers of animals in general laboratory spaces. However, it is precisely these attributes that make them so different from humans and explains, in part, why these models do not accurately predict drug responses in human patients. This is particularly true of the neurofibromatoses (NFs), a group of genetic diseases that predispose individuals to tumors of the nervous system, the most common of which is Neurofibromatosis type 1 (NF1). Despite years of research, there are still many unanswered questions and few effective treatments for NF1. Genetically engineered mice have drastically improved our understanding of many aspects of NF1, but they do not exemplify the overall complexity of the disease and some findings do not translate well to humans due to differences in body size and physiology. Moreover, NF1 mouse models are heavily reliant on the Cre-Lox system, which does not accurately reflect the molecular mechanism of spontaneous loss of heterozygosity that accompanies human tumor development. Spontaneous and genetically engineered large animal models may provide a valuable supplement to rodent studies for NF1. Naturally occurring comparative models of disease are an attractive prospect because they occur on heterogeneous genetic backgrounds and are due to spontaneous rather than engineered mutations. The use of animals with naturally occurring disease has been effective for studying osteosarcoma, lymphoma, and diabetes. Spontaneous NF-like symptoms including neurofibromas and malignant peripheral nerve sheath tumors (MPNST) have been documented in several large animal species and share biological and clinical similarities with human NF1. These animals could provide additional insight into the complex biology of NF1 and potentially provide a platform for pre-clinical trials. Additionally, genetically engineered porcine models of NF1 have recently been developed and display a variety of clinical features similar to those seen in NF1 patients. Their large size and relatively long lifespan allow for longitudinal imaging studies and evaluation of innovative surgical techniques using human equipment. Greater genetic, anatomic, and physiologic similarities to humans enable the engineering of precise disease alleles found in human patients and make them ideal for preclinical pharmacokinetic and pharmacodynamic studies of small molecule, cellular, and gene therapies prior to clinical trials in patients. Comparative genomic studies between humans and animals with naturally occurring disease, as well as preclinical studies in large animal disease models, may help identify new targets for therapeutic intervention and expedite the translation of new therapies. In this review, we discuss new genetically engineered large animal models of NF1 and cases of spontaneous NF-like manifestations in large animals, with a special emphasis on how these comparative models could act as a crucial translational intermediary between specialized murine models and NF1 patients.
Collapse
Affiliation(s)
- Sara H. Osum
- Masonic Cancer Center, Department of Pediatrics, Division of Hematology and Oncology, University of Minnesota, Minneapolis, MN 55455, USA;
| | | | - David A. Largaespada
- Masonic Cancer Center, Department of Pediatrics, Division of Hematology and Oncology, University of Minnesota, Minneapolis, MN 55455, USA;
| |
Collapse
|
21
|
Perisse IV, Fan Z, Singina GN, White KL, Polejaeva IA. Improvements in Gene Editing Technology Boost Its Applications in Livestock. Front Genet 2021; 11:614688. [PMID: 33603767 PMCID: PMC7885404 DOI: 10.3389/fgene.2020.614688] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 12/07/2020] [Indexed: 12/18/2022] Open
Abstract
Accelerated development of novel CRISPR/Cas9-based genome editing techniques provides a feasible approach to introduce a variety of precise modifications in the mammalian genome, including introduction of multiple edits simultaneously, efficient insertion of long DNA sequences into specific targeted loci as well as performing nucleotide transitions and transversions. Thus, the CRISPR/Cas9 tool has become the method of choice for introducing genome alterations in livestock species. The list of new CRISPR/Cas9-based genome editing tools is constantly expanding. Here, we discuss the methods developed to improve efficiency and specificity of gene editing tools as well as approaches that can be employed for gene regulation, base editing, and epigenetic modifications. Additionally, advantages and disadvantages of two primary methods used for the production of gene-edited farm animals: somatic cell nuclear transfer (SCNT or cloning) and zygote manipulations will be discussed. Furthermore, we will review agricultural and biomedical applications of gene editing technology.
Collapse
Affiliation(s)
- Iuri Viotti Perisse
- Department of Animal, Dairy and Veterinary Sciences, Utah State University, Logan, UT, United States
| | - Zhiqiang Fan
- Department of Animal, Dairy and Veterinary Sciences, Utah State University, Logan, UT, United States
| | - Galina N. Singina
- L.K. Ernst Federal Research Center for Animal Husbandry, Podolsk, Russia
| | - Kenneth L. White
- Department of Animal, Dairy and Veterinary Sciences, Utah State University, Logan, UT, United States
| | - Irina A. Polejaeva
- Department of Animal, Dairy and Veterinary Sciences, Utah State University, Logan, UT, United States
| |
Collapse
|
22
|
Santini L, Palandri C, Nediani C, Cerbai E, Coppini R. Modelling genetic diseases for drug development: Hypertrophic cardiomyopathy. Pharmacol Res 2020; 160:105176. [DOI: 10.1016/j.phrs.2020.105176] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 08/16/2020] [Accepted: 08/22/2020] [Indexed: 12/13/2022]
|
23
|
Polkoff KM, Chung J, Simpson SG, Gleason K, Piedrahita JA. In Vitro Validation of Transgene Expression in Gene-Edited Pigs Using CRISPR Transcriptional Activators. CRISPR J 2020; 3:409-418. [PMID: 33095051 PMCID: PMC7580606 DOI: 10.1089/crispr.2020.0037] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The use of CRISPR-Cas and RNA-guided endonucleases has drastically changed research strategies for understanding and exploiting gene function, particularly for the generation of gene-edited animal models. This has resulted in an explosion in the number of gene-edited species, including highly biomedically relevant pig models. However, even with error-free DNA insertion or deletion, edited genes are occasionally not expressed and/or translated as expected. Therefore, there is a need to validate the expression outcomes gene modifications in vitro before investing in the costly generation of a gene-edited animal. Unfortunately, many gene targets are tissue specific and/or not expressed in cultured primary cells, making validation difficult without generating an animal. In this study, using pigs as a proof of concept, we show that CRISPR-dCas9 transcriptional activators can be used to validate functional transgene insertion in nonexpressing easily cultured cells such as fibroblasts. This is a tool that can be used across disciplines and animal species to save time and resources by verifying expected outcomes of gene edits before generating live animals.
Collapse
Affiliation(s)
- Kathryn M. Polkoff
- Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina, USA
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA
| | - Jaewook Chung
- Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina, USA
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA
| | - Sean G. Simpson
- Deparment of Animal and Avian Sciences, University of Maryland, College Park, Maryland, USA
- RenOVAte Biosciences, Inc., Reisterstown, Maryland, USA
| | - Katherine Gleason
- Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina, USA
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA
| | - Jorge A. Piedrahita
- Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina, USA
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA
| |
Collapse
|
24
|
Khalil AS, Jaenisch R, Mooney DJ. Engineered tissues and strategies to overcome challenges in drug development. Adv Drug Deliv Rev 2020; 158:116-139. [PMID: 32987094 PMCID: PMC7518978 DOI: 10.1016/j.addr.2020.09.012] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 08/29/2020] [Accepted: 09/23/2020] [Indexed: 12/16/2022]
Abstract
Current preclinical studies in drug development utilize high-throughput in vitro screens to identify drug leads, followed by both in vitro and in vivo models to predict lead candidates' pharmacokinetic and pharmacodynamic properties. The goal of these studies is to reduce the number of lead drug candidates down to the most likely to succeed in later human clinical trials. However, only 1 in 10 drug candidates that emerge from preclinical studies will succeed and become an approved therapeutic. Lack of efficacy or undetected toxicity represents roughly 75% of the causes for these failures, despite these parameters being the primary exclusion criteria in preclinical studies. Recently, advances in both biology and engineering have created new tools for constructing new preclinical models. These models can complement those used in current preclinical studies by helping to create more realistic representations of human tissues in vitro and in vivo. In this review, we describe current preclinical models to identify their value and limitations and then discuss select areas of research where improvements in preclinical models are particularly needed to advance drug development. Following this, we discuss design considerations for constructing preclinical models and then highlight recent advances in these efforts. Taken together, we aim to review the advances as of 2020 surrounding the prospect of biological and engineering tools for adding enhanced biological relevance to preclinical studies to aid in the challenges of failed drug candidates and the burden this poses on the drug development enterprise and thus healthcare.
Collapse
Affiliation(s)
- Andrew S Khalil
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA 02115, USA; Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Rudolf Jaenisch
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA.
| | - David J Mooney
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA 02115, USA.
| |
Collapse
|
25
|
Measurable Cytokine Concentrations in Pig Seminal Plasma Are Modified by Semen Handling and Storage. BIOLOGY 2020; 9:biology9090276. [PMID: 32906591 PMCID: PMC7565580 DOI: 10.3390/biology9090276] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 08/30/2020] [Accepted: 09/04/2020] [Indexed: 12/12/2022]
Abstract
Sample handling and storing are critical steps for the reliable measurement of circulating biomolecules in biological fluids. This study evaluates how cytokine measurements in pig seminal plasma (SP) vary depending on semen handling and SP storage. Thirteen cytokines (GM-CSF, IFNγ, IL-1α, IL-1β, IL-1ra, IL-2, IL-4, IL-6, IL-8, IL-10, IL-12, IL-18 and TNFα) were measured using Luminex xMAP® technology in individual seminal plasma (SP) samples (n = 62) from healthy breeding boars. Three separate experiments explored the delay (2 h and 24 h) in SP collection after ejaculation (Experiment 1) and SP storage, either short-term (5 °C, −20 °C and −80 °C for 72 h, Experiment 2) or long-term (at −20 °C and −80 °C for two months, Experiment 3), before analysis. Levels in fresh SP-samples were used as baseline control values. Delays in SP harvesting of up to 24 h did not substantially impact SP cytokine measurements. Some cytokines showed instability in stored SP samples, mainly in long-term storage. Ideally, cytokines in pig SP should be measured in fresh samples harvested within 24 h after ejaculation. If storage of SP is imperative, storage conditions should be adjusted for each cytokine.
Collapse
|
26
|
Zettler S, Renner S, Kemter E, Hinrichs A, Klymiuk N, Backman M, Riedel EO, Mueller C, Streckel E, Braun-Reichhart C, Martins AS, Kurome M, Keßler B, Zakhartchenko V, Flenkenthaler F, Arnold GJ, Fröhlich T, Blum H, Blutke A, Wanke R, Wolf E. A decade of experience with genetically tailored pig models for diabetes and metabolic research. Anim Reprod 2020; 17:e20200064. [PMID: 33029223 PMCID: PMC7534555 DOI: 10.1590/1984-3143-ar2020-0064] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023] Open
Abstract
The global prevalence of diabetes mellitus and other metabolic diseases is rapidly increasing. Animal models play pivotal roles in unravelling disease mechanisms and developing and testing therapeutic strategies. Rodents are the most widely used animal models but may have limitations in their resemblance to human disease mechanisms and phenotypes. Findings in rodent models are consequently often difficult to extrapolate to human clinical trials. To overcome this ‘translational gap’, we and other groups are developing porcine disease models. Pigs share many anatomical and physiological traits with humans and thus hold great promise as translational animal models. Importantly, the toolbox for genetic engineering of pigs is rapidly expanding. Human disease mechanisms and targets can therefore be reproduced in pigs on a molecular level, resulting in precise and predictive porcine (PPP) models. In this short review, we summarize our work on the development of genetically (pre)diabetic pig models and how they have been used to study disease mechanisms and test therapeutic strategies. This includes the generation of reporter pigs for studying beta-cell maturation and physiology. Furthermore, genetically engineered pigs are promising donors of pancreatic islets for xenotransplantation. In summary, genetically tailored pig models have become an important link in the chain of translational diabetes and metabolic research.
Collapse
Affiliation(s)
- Silja Zettler
- Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany.,Center for Innovative Medical Models, Department of Veterinary Sciences, LMU Munich, Oberschleißheim, Germany.,German Center for Diabetes Research, Neuherberg, Germany
| | - Simone Renner
- Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany.,Center for Innovative Medical Models, Department of Veterinary Sciences, LMU Munich, Oberschleißheim, Germany.,German Center for Diabetes Research, Neuherberg, Germany
| | - Elisabeth Kemter
- Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany.,Center for Innovative Medical Models, Department of Veterinary Sciences, LMU Munich, Oberschleißheim, Germany.,German Center for Diabetes Research, Neuherberg, Germany
| | - Arne Hinrichs
- Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany.,Center for Innovative Medical Models, Department of Veterinary Sciences, LMU Munich, Oberschleißheim, Germany
| | - Nikolai Klymiuk
- Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany.,Center for Innovative Medical Models, Department of Veterinary Sciences, LMU Munich, Oberschleißheim, Germany
| | - Mattias Backman
- Laboratory for Functional Genome Analysis, Gene Center, LMU Munich, Munich
| | | | - Christiane Mueller
- Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany.,Center for Innovative Medical Models, Department of Veterinary Sciences, LMU Munich, Oberschleißheim, Germany
| | - Elisabeth Streckel
- Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany.,Center for Innovative Medical Models, Department of Veterinary Sciences, LMU Munich, Oberschleißheim, Germany
| | - Christina Braun-Reichhart
- Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany.,Center for Innovative Medical Models, Department of Veterinary Sciences, LMU Munich, Oberschleißheim, Germany
| | - Ana Sofia Martins
- Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany.,Center for Innovative Medical Models, Department of Veterinary Sciences, LMU Munich, Oberschleißheim, Germany
| | - Mayuko Kurome
- Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany.,Center for Innovative Medical Models, Department of Veterinary Sciences, LMU Munich, Oberschleißheim, Germany
| | - Barbara Keßler
- Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany.,Center for Innovative Medical Models, Department of Veterinary Sciences, LMU Munich, Oberschleißheim, Germany
| | - Valeri Zakhartchenko
- Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany.,Center for Innovative Medical Models, Department of Veterinary Sciences, LMU Munich, Oberschleißheim, Germany
| | | | - Georg Josef Arnold
- Laboratory for Functional Genome Analysis, Gene Center, LMU Munich, Munich
| | - Thomas Fröhlich
- Laboratory for Functional Genome Analysis, Gene Center, LMU Munich, Munich
| | - Helmut Blum
- Laboratory for Functional Genome Analysis, Gene Center, LMU Munich, Munich
| | - Andreas Blutke
- Research Unit Analytical Pathology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Rüdiger Wanke
- Institute of Veterinary Pathology, Center for Clinical Veterinary Medicine, LMU Munich, Munich, Germany
| | - Eckhard Wolf
- Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany.,Center for Innovative Medical Models, Department of Veterinary Sciences, LMU Munich, Oberschleißheim, Germany.,German Center for Diabetes Research, Neuherberg, Germany.,Laboratory for Functional Genome Analysis, Gene Center, LMU Munich, Munich
| |
Collapse
|
27
|
Jeong PS, Sim BW, Park SH, Kim MJ, Kang HG, Nanjidsuren T, Lee S, Song BS, Koo DB, Kim SU. Chaetocin Improves Pig Cloning Efficiency by Enhancing Epigenetic Reprogramming and Autophagic Activity. Int J Mol Sci 2020; 21:ijms21144836. [PMID: 32650566 PMCID: PMC7402317 DOI: 10.3390/ijms21144836] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 07/04/2020] [Accepted: 07/07/2020] [Indexed: 12/21/2022] Open
Abstract
Efficient epigenetic reprogramming is crucial for the in vitro development of mammalian somatic cell nuclear transfer (SCNT) embryos. The aberrant levels of histone H3 lysine 9 trimethylation (H3K9me3) is an epigenetic barrier. In this study, we evaluated the effects of chaetocin, an H3K9me3-specific methyltransferase inhibitor, on the epigenetic reprogramming and developmental competence of porcine SCNT embryos. The SCNT embryos showed abnormal levels of H3K9me3 at the pronuclear, two-cell, and four-cell stages compared to in vitro fertilized embryos. Moreover, the expression levels of H3K9me3-specific methyltransferases (suv39h1 and suv39h2) and DNA methyltransferases (DNMT1, DNMT3a, and DNMT3b) were higher in SCNT embryos. Treatment with 0.5 nM chaetocin for 24 h after activation significantly increased the developmental competence of SCNT embryos in terms of the cleavage rate, blastocyst formation rate, hatching rate, cell number, expression of pluripotency-related genes, and cell survival rate. In particular, chaetocin enhanced epigenetic reprogramming by reducing the H3K9me3 and 5-methylcytosine levels and restoring the abnormal expression of H3K9me3-specific methyltransferases and DNA methyltransferases. Chaetocin induced autophagic activity, leading to a significant reduction in maternal mRNA levels in embryos at the pronuclear and two-cell stages. These findings revealed that chaetocin enhanced the developmental competence of porcine SCNT embryos by regulating epigenetic reprogramming and autophagic activity and so could be used to enhance the production of transgenic pigs for biomedical research.
Collapse
Affiliation(s)
- Pil-Soo Jeong
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do 28116, Korea; (P.-S.J.); (B.-W.S.); (S.-H.P.); (M.J.K.); (H.-G.K.); (T.N.); (S.L.); (B.-S.S.)
- Department of Biotechnology, Daegu University, Gyeongsangbuk-do 38453, Korea
| | - Bo-Woong Sim
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do 28116, Korea; (P.-S.J.); (B.-W.S.); (S.-H.P.); (M.J.K.); (H.-G.K.); (T.N.); (S.L.); (B.-S.S.)
| | - Soo-Hyun Park
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do 28116, Korea; (P.-S.J.); (B.-W.S.); (S.-H.P.); (M.J.K.); (H.-G.K.); (T.N.); (S.L.); (B.-S.S.)
| | - Min Ju Kim
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do 28116, Korea; (P.-S.J.); (B.-W.S.); (S.-H.P.); (M.J.K.); (H.-G.K.); (T.N.); (S.L.); (B.-S.S.)
| | - Hyo-Gu Kang
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do 28116, Korea; (P.-S.J.); (B.-W.S.); (S.-H.P.); (M.J.K.); (H.-G.K.); (T.N.); (S.L.); (B.-S.S.)
| | - Tsevelmaa Nanjidsuren
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do 28116, Korea; (P.-S.J.); (B.-W.S.); (S.-H.P.); (M.J.K.); (H.-G.K.); (T.N.); (S.L.); (B.-S.S.)
| | - Sanghoon Lee
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do 28116, Korea; (P.-S.J.); (B.-W.S.); (S.-H.P.); (M.J.K.); (H.-G.K.); (T.N.); (S.L.); (B.-S.S.)
| | - Bong-Seok Song
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do 28116, Korea; (P.-S.J.); (B.-W.S.); (S.-H.P.); (M.J.K.); (H.-G.K.); (T.N.); (S.L.); (B.-S.S.)
| | - Deog-Bon Koo
- Department of Biotechnology, Daegu University, Gyeongsangbuk-do 38453, Korea
- Correspondence: (D.-B.K.); (S.-U.K.); Tel.: +82-43-240-6321 (S.-U.K.); Fax: +82-43-240-6309 (S.-U.K.)
| | - Sun-Uk Kim
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do 28116, Korea; (P.-S.J.); (B.-W.S.); (S.-H.P.); (M.J.K.); (H.-G.K.); (T.N.); (S.L.); (B.-S.S.)
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon 34113, Korea
- Correspondence: (D.-B.K.); (S.-U.K.); Tel.: +82-43-240-6321 (S.-U.K.); Fax: +82-43-240-6309 (S.-U.K.)
| |
Collapse
|
28
|
Eun K, Hong N, Jeong YW, Park MG, Hwang SU, Jeong YIK, Choi EJ, Olsson PO, Hwang WS, Hyun SH, Kim H. Transcriptional activities of human elongation factor-1α and cytomegalovirus promoter in transgenic dogs generated by somatic cell nuclear transfer. PLoS One 2020; 15:e0233784. [PMID: 32492024 PMCID: PMC7269240 DOI: 10.1371/journal.pone.0233784] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 05/12/2020] [Indexed: 11/30/2022] Open
Abstract
Recent advances in somatic cell nuclear transfer (SCNT) in canines facilitate the production of canine transgenic models. Owing to the importance of stable and strong promoter activity in transgenic animals, we tested human elongation factor 1α (hEF1α) and cytomegalovirus (CMV) promoter sequences in SCNT transgenic dogs. After transfection, transgenic donor fibroblasts with the hEF1α-enhanced green fluorescence protein (EGFP) transgene were successfully isolated using fluorescence-activated cell sorting (FACS). We obtained four puppies, after SCNT, and identified three puppies as being transgenic using PCR analysis. Unexpectedly, EGFP regulated by hEF1α promoter was not observed at the organismal and cellular levels in these transgenic dogs. EGFP expression was rescued by the inhibition of DNA methyltransferases, implying that the hEF1α promoter is silenced by DNA methylation. Next, donor cells with CMV-EGFP transgene were successfully established and SCNT was performed. Three puppies of six born puppies were confirmed to be transgenic. Unlike hEF1α-regulated EGFP, CMV-regulated EGFP was strongly detectable at both the organismal and cellular levels in all transgenic dogs, even after 19 months. In conclusion, our study suggests that the CMV promoter is more suitable, than the hEF1α promoter, for stable transgene expression in SCNT-derived transgenic canine model.
Collapse
Affiliation(s)
- Kiyoung Eun
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seongbuk-gu, Seoul, Republic of Korea
- Institute of Animal Molecular Biotechnology, Korea University, Seongbuk-gu, Seoul, Republic of Korea
| | - Nayoung Hong
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seongbuk-gu, Seoul, Republic of Korea
- Institute of Animal Molecular Biotechnology, Korea University, Seongbuk-gu, Seoul, Republic of Korea
| | - Yeon Woo Jeong
- Sooam Biotech Research Foundation, Guro-gu, Seoul, Republic of Korea
| | - Min Gi Park
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seongbuk-gu, Seoul, Republic of Korea
- Institute of Animal Molecular Biotechnology, Korea University, Seongbuk-gu, Seoul, Republic of Korea
| | - Seon-Ung Hwang
- Laboratory of Veterinary Embryology and Biotechnology, College of Veterinary Medicine, Chungbuk National University, Seowon-gu, Cheongju, Republic of Korea
- Institute of Stem Cell & Regenerative Medicine, Chungbuk National University, Seowon-gu, Cheongju, Republic of Korea
| | - Yeon I. K. Jeong
- Sooam Biotech Research Foundation, Guro-gu, Seoul, Republic of Korea
| | - Eun Ji Choi
- Sooam Biotech Research Foundation, Guro-gu, Seoul, Republic of Korea
| | - P. Olof Olsson
- Sooam Biotech Research Foundation, Guro-gu, Seoul, Republic of Korea
| | - Woo Suk Hwang
- Sooam Biotech Research Foundation, Guro-gu, Seoul, Republic of Korea
| | - Sang-Hwan Hyun
- Laboratory of Veterinary Embryology and Biotechnology, College of Veterinary Medicine, Chungbuk National University, Seowon-gu, Cheongju, Republic of Korea
- Institute of Stem Cell & Regenerative Medicine, Chungbuk National University, Seowon-gu, Cheongju, Republic of Korea
- * E-mail: (SHH); (HK)
| | - Hyunggee Kim
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seongbuk-gu, Seoul, Republic of Korea
- Institute of Animal Molecular Biotechnology, Korea University, Seongbuk-gu, Seoul, Republic of Korea
- * E-mail: (SHH); (HK)
| |
Collapse
|
29
|
Zhao X, Nie J, Tang Y, He W, Xiao K, Pang C, Liang X, Lu Y, Zhang M. Generation of Transgenic Cloned Buffalo Embryos Harboring the EGFP Gene in the Y Chromosome Using CRISPR/Cas9-Mediated Targeted Integration. Front Vet Sci 2020; 7:199. [PMID: 32426378 PMCID: PMC7212351 DOI: 10.3389/fvets.2020.00199] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 03/25/2020] [Indexed: 11/16/2022] Open
Abstract
Sex control technology is of great significance in the production of domestic animals, especially for rapidly breeding water buffalo (bubalus bubalis), which served as a research model in the present study. We have confirmed that a fluorescence protein integrated into the Y chromosome is fit for sexing pre-implantation embryos in the mouse. Firstly, we optimized the efficiency of targeted integration of exogenous gene encoding enhanced green fluorescent protein (eGFP) and mCherry in Neuro-2a cells, mouse embryonic stem cells, mouse embryonic cells (NIH3T3), buffalo fetal fibroblast (BFF) cells. The results showed that a homology arm length of 800 bp on both sides of the target is more efficient that 300 bp or 300 bp/800 bp. Homology-directed repair (HDR)-mediated knock-in in BFF cells was also significantly improved when cells were supplemented with pifithrin-μ, which is a small molecule that inhibits the binding of p53 to mitochondria. Three pulses at 250 V resulted in the most efficient electroporation in BFF cells and 1.5 μg/mL puromycin was found to be the optimal concentration for screening. Moreover, Y-Chr-eGFP transgenic BFF cells and cloned buffalo embryos were successfully generated using CRISPR/Cas9-mediated gene editing combined with the somatic cell nuclear transfer (SCNT) technique. At passage numbers 6–8, the growth rate and cell proliferation rate were significantly lower in Y-Chr-eGFP transgenic than in non-transgenic BFF cells; the expression levels of the methylation-related genes DNMT1 and DNMT3a were similar; however, the expression levels of the acetylation-related genes HDAC1, HDAC2, and HDAC3 were significantly higher (p < 0.05) in Y-Chr-eGFP transgenic BFF cells compared with non-transgenic cells. Y-Chr-eGFP transgenic BFFs were used as donors for SCNT, the results showed that eGFP reporter is suitable for the visualization of the sex of embryos. The blastocyst rates of cloned buffalo embryos were similar; however, the cleavage rates of transgenic cloned embryos were significantly lower compared with control. In summary, we optimized the protocol for generating transgenic BFF cells and successfully generated Y-Chr-eGFP transgenic embryos using these cells as donors.
Collapse
Affiliation(s)
- Xiuling Zhao
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Animal Reproduction Institute, Guangxi University, Nanning, China
| | - Junyu Nie
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Animal Reproduction Institute, Guangxi University, Nanning, China
| | - Yuyan Tang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Animal Reproduction Institute, Guangxi University, Nanning, China
| | - Wengtan He
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Animal Reproduction Institute, Guangxi University, Nanning, China
| | - Kai Xiao
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Animal Reproduction Institute, Guangxi University, Nanning, China
| | - Chunying Pang
- Key Laboratory of Buffalo Genetics, Breeding and Reproduction Technology, Ministry of Agriculture and Buffalo Research Institute, Chinese Academy of Agricultural Science, Nanning, China
| | - Xianwei Liang
- Key Laboratory of Buffalo Genetics, Breeding and Reproduction Technology, Ministry of Agriculture and Buffalo Research Institute, Chinese Academy of Agricultural Science, Nanning, China
| | - Yangqing Lu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Animal Reproduction Institute, Guangxi University, Nanning, China
| | - Ming Zhang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Animal Reproduction Institute, Guangxi University, Nanning, China
| |
Collapse
|
30
|
Livestock Gene Editing by One-step Embryo Manipulation. J Equine Vet Sci 2020; 89:103025. [PMID: 32563448 DOI: 10.1016/j.jevs.2020.103025] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 04/06/2020] [Accepted: 04/07/2020] [Indexed: 12/11/2022]
Abstract
The breakthrough and rapid advance of clustered regularly interspaced short palindromic repeat (CRISPR)/CRISPR-associated protein 9 (Cas9) technology has enabled the efficient generation of gene-edited animals by one-step embryo manipulation. Clustered regularly interspaced short palindromic repeat/CRISPR-associated protein 9 delivery to the livestock embryos has been typically achieved by intracytoplasmic microinjection; however, recent studies show that electroporation may be a reliable, efficient, and practical method for CRISPR/Cas9 delivery. The source of embryos used to generate gene-edited animals varies from in vivo to in vitro produced, depending mostly on the species of interest. In addition, different Cas9 and gRNA reagents can be used for embryo editing, ranging from Cas9-coding plasmid or messenger RNA to Cas9 recombinant protein, which can be combined with in vitro transcribed or synthetic guide RNAs. Mosaicism is reported as one of the main problems with generation of animals by embryo editing. On the other hand, off-target mutations are rarely found in livestock derived from one-step editing. In this review, we discussed these and other aspects of generating gene-edited animals by single-step embryo manipulation.
Collapse
|
31
|
Summers KM, Bush SJ, Wu C, Su AI, Muriuki C, Clark EL, Finlayson HA, Eory L, Waddell LA, Talbot R, Archibald AL, Hume DA. Functional Annotation of the Transcriptome of the Pig, Sus scrofa, Based Upon Network Analysis of an RNAseq Transcriptional Atlas. Front Genet 2020; 10:1355. [PMID: 32117413 PMCID: PMC7034361 DOI: 10.3389/fgene.2019.01355] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 12/11/2019] [Indexed: 12/15/2022] Open
Abstract
The domestic pig (Sus scrofa) is both an economically important livestock species and a model for biomedical research. Two highly contiguous pig reference genomes have recently been released. To support functional annotation of the pig genomes and comparative analysis with large human transcriptomic data sets, we aimed to create a pig gene expression atlas. To achieve this objective, we extended a previous approach developed for the chicken. We downloaded RNAseq data sets from public repositories, down-sampled to a common depth, and quantified expression against a reference transcriptome using the mRNA quantitation tool, Kallisto. We then used the network analysis tool Graphia to identify clusters of transcripts that were coexpressed across the merged data set. Consistent with the principle of guilt-by-association, we identified coexpression clusters that were highly tissue or cell-type restricted and contained transcription factors that have previously been implicated in lineage determination. Other clusters were enriched for transcripts associated with biological processes, such as the cell cycle and oxidative phosphorylation. The same approach was used to identify coexpression clusters within RNAseq data from multiple individual liver and brain samples, highlighting cell type, process, and region-specific gene expression. Evidence of conserved expression can add confidence to assignment of orthology between pig and human genes. Many transcripts currently identified as novel genes with ENSSSCG or LOC IDs were found to be coexpressed with annotated neighbouring transcripts in the same orientation, indicating they may be products of the same transcriptional unit. The meta-analytic approach to utilising public RNAseq data is extendable to include new data sets and new species and provides a framework to support the Functional Annotation of Animals Genomes (FAANG) initiative.
Collapse
Affiliation(s)
- Kim M. Summers
- Mater Research Institute-University of Queensland, Translational Research Institute, Woolloongabba, QLD, Australia
| | - Stephen J. Bush
- Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
| | - Chunlei Wu
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, United States
| | - Andrew I. Su
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, United States
| | - Charity Muriuki
- The Roslin Institute, University of Edinburgh, Midlothian, United Kingdom
| | - Emily L. Clark
- The Roslin Institute, University of Edinburgh, Midlothian, United Kingdom
| | | | - Lel Eory
- The Roslin Institute, University of Edinburgh, Midlothian, United Kingdom
| | - Lindsey A. Waddell
- The Roslin Institute, University of Edinburgh, Midlothian, United Kingdom
| | - Richard Talbot
- The Roslin Institute, University of Edinburgh, Midlothian, United Kingdom
| | - Alan L. Archibald
- The Roslin Institute, University of Edinburgh, Midlothian, United Kingdom
| | - David A. Hume
- Mater Research Institute-University of Queensland, Translational Research Institute, Woolloongabba, QLD, Australia
| |
Collapse
|
32
|
Lee K, Uh K, Farrell K. Current progress of genome editing in livestock. Theriogenology 2020; 150:229-235. [PMID: 32000993 DOI: 10.1016/j.theriogenology.2020.01.036] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 01/18/2020] [Indexed: 12/12/2022]
Abstract
Historically, genetic engineering in livestock proved to be challenging. Without stable embryonic stem cell lines to utilize, somatic cell nuclear transfer (SCNT) had to be employed to produce many of the genetically engineered (GE) livestock models. Through the genetic engineering of somatic cells followed by SCNT, GE livestock models could be generated carrying site-specific modifications. Although successful, only a few GE livestock models were generated because of low efficiency and associated birth defects. Recently, there have been major strides in the development of genome editing tools: Zinc-Finger Nucleases (ZFNs), Transcription activator-like effector nucleases (TALENS), and Clustered regularly interspaced short palindromic repeats (CRISPR) and CRISPR-associated 9 (Cas9) system. These tools rely on the generation of a double strand DNA break, followed by one of two repair pathways: non-homologous end joining (NHEJ) or homology directed repair (HDR). Compared to the traditional approaches, these tools dramatically reduce time and effort needed to establish a GE animal. Another benefit of utilizing genome editing tools is the application of direct injection into developing embryos to induce targeted mutations, therefore, eliminating side effects associated with SCNT. Emerging technological advancements of genome editing systems have dramatically improved efficiency to generate GE livestock models for both biomedical and agricultural purposes. Although the efficiency of genome editing tools has revolutionized GE livestock production, improvements for safe and consistent application are desired. This review will provide an overview of genome editing techniques, as well as examples of GE livestock models for agricultural and biomedical purposes.
Collapse
Affiliation(s)
- Kiho Lee
- Department of Animal and Poultry Sciences, Virginia Tech, Blacksburg, VA, USA.
| | - Kyungjun Uh
- Department of Animal and Poultry Sciences, Virginia Tech, Blacksburg, VA, USA
| | - Kayla Farrell
- Department of Animal and Poultry Sciences, Virginia Tech, Blacksburg, VA, USA
| |
Collapse
|
33
|
From Mice to Monkeys? Beyond Orthodox Approaches to the Ethics of Animal Model Choice. Animals (Basel) 2020; 10:ani10010077. [PMID: 31906319 PMCID: PMC7022287 DOI: 10.3390/ani10010077] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 12/06/2019] [Accepted: 12/11/2019] [Indexed: 11/30/2022] Open
Abstract
Simple Summary New tools, allowing scientists to make precise changes to mammal genomes, have made possible future increased use of larger mammals in biomedical research, such as primates, pigs, and dogs. This paper addresses ethical issues that are raised by using larger mammals instead of smaller ones in this research. Because scientists who use animals in research follow strict guidelines, we first examine what those guidelines suggest for using larger mammals. We then consider what philosophers, who write about the ethics of animal use, consider as the important questions in evaluating which (if any) animals are acceptable to use in research. We find that philosophical perspectives have typically been interested in the question of when or if animal use is justified, while biomedical research guidance has assumed that animal use is justified but defined specific limits to that use. To address directly the ethical questions that arise in the practice of biomedical research in selecting which animals to use, we consider an approach to ethics that is focused on character and living well (or flourishing). This paper is valuable to society in drawing attention to the ethical questions, rather than merely the scientific issues, that are important in selecting which animals to use in biomedical research. Abstract Recent developments in genome editing tools, along with limits in the translational potential of rodent models of human disease, have spurred renewed biomedical research interest in large mammals like nonhuman primates, pigs, and dogs. Such scientific developments raise ethical issues about the use of these animals in comparison with smaller mammals, such as mice and rats. To examine these ethical questions, we first consider standard (or “orthodox”) approaches, including ethics oversight within biomedical research communities, and critical theoretical reflections on animal research, including rights-based and utilitarian approaches. We argue that oversight of biomedical research offers guidance on the profession’s permitted uses of animals within a research setting and orthodox approaches to animal ethics questions when and whether animals should be used in biomedicine; however, neither approach sufficiently investigates the nuances of ethical practices within the research setting. To fill this lacuna, we consider a virtue ethical approach to the use of specific animal models in biomedicine. From this perspective, we argued that limitations on flourishing for large mammals in a research setting, as well as potential human-animal bonds, are two sources of likely ethical tensions in animal care and use in the context of larger mammals.
Collapse
|
34
|
Abstract
Safe and reliable large animal diabetes models are a key prerequisite for advanced preclinical studies on diabetes. Chemical induction is the standard model of diabetes in rodents but is often critiqued in higher animals due to reduced efficacy, relevant side effects, and inadequate mortality rate. In this chapter, we aim to describe both pharmacological and surgical approaches for reproducible and safe diabetes models in minipigs and primates. In addition, genetically modified pig models for diabetes research are described.
Collapse
Affiliation(s)
- Barbara Ludwig
- Department of Medicine III, University Hospital Carl Gustav Carus, Dresden, Germany. .,Paul Langerhans Institute Dresden (PLID) of the Helmholtz Center Munich at the University Hospital Carl Gustav Carus and Faculty of Medicine of the TU Dresden, Dresden, Germany. .,DFG-Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany. .,Department of Endocrinology and Diabetology, University Hospital Zurich, Zurich, Switzerland.
| | - Eckhard Wolf
- Department of Molecular Animal Breeding and Biotechnology, Ludwig Maximilian University, Munich, Germany
| | - Uwe Schönmann
- German Primate Center, Leibniz-Institute for Primate Research, Göttingen, Germany
| | - Stefan Ludwig
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital Carl Gustav Carus, Dresden, Germany
| |
Collapse
|
35
|
Pires CF, Rosa FF, Kurochkin I, Pereira CF. Understanding and Modulating Immunity With Cell Reprogramming. Front Immunol 2019; 10:2809. [PMID: 31921109 PMCID: PMC6917620 DOI: 10.3389/fimmu.2019.02809] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 11/15/2019] [Indexed: 12/30/2022] Open
Abstract
Cell reprogramming concepts have been classically developed in the fields of developmental and stem cell biology and are currently being explored for regenerative medicine, given its potential to generate desired cell types for replacement therapy. Cell fate can be experimentally reversed or modified by enforced expression of lineage specific transcription factors leading to pluripotency or attainment of another somatic cell type identity. The possibility to reprogram fibroblasts into induced dendritic cells (DC) competent for antigen presentation creates a paradigm shift for understanding and modulating the immune system with direct cell reprogramming. PU.1, IRF8, and BATF3 were identified as sufficient and necessary to impose DC fate in unrelated cell types, taking advantage of Clec9a, a C-type lectin receptor with restricted expression in conventional DC type 1. The identification of such minimal gene regulatory networks helps to elucidate the molecular mechanisms governing development and lineage heterogeneity along the hematopoietic hierarchy. Furthermore, the generation of patient-tailored reprogrammed immune cells provides new and exciting tools for the expanding field of cancer immunotherapy. Here, we summarize cell reprogramming concepts and experimental approaches, review current knowledge at the intersection of cell reprogramming with hematopoiesis, and propose how cell fate engineering can be merged to immunology, opening new opportunities to understand the immune system in health and disease.
Collapse
Affiliation(s)
- Cristiana F. Pires
- Cell Reprogramming in Hematopoiesis and Immunity Laboratory, Lund Stem Cell Center, Molecular Medicine and Gene Therapy, Lund University, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Fábio F. Rosa
- Cell Reprogramming in Hematopoiesis and Immunity Laboratory, Lund Stem Cell Center, Molecular Medicine and Gene Therapy, Lund University, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Ilia Kurochkin
- Center for Neurobiology and Brain Restoration, Skolkovo Institute of Science and Technology, Moscow, Russia
| | - Carlos-Filipe Pereira
- Cell Reprogramming in Hematopoiesis and Immunity Laboratory, Lund Stem Cell Center, Molecular Medicine and Gene Therapy, Lund University, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
36
|
Draper ACE, Cahalan SD, Goodwin D, Perkins J, Piercy RJ. Assessing pathological changes within the nucleus ambiguus of horses with recurrent laryngeal neuropathy: An extreme, length-dependent axonopathy. Muscle Nerve 2019; 60:762-768. [PMID: 31498901 DOI: 10.1002/mus.26699] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 09/03/2019] [Accepted: 09/03/2019] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Equine recurrent laryngeal neuropathy (RLN) is a naturally occurring model of length-dependent axonopathy characterized by asymmetrical degeneration of recurrent laryngeal nerve axons (RLn). Distal RLn degeneration is marked, but it is unclear whether degeneration extends to include cell bodies (consistent with a neuronopathy). METHODS With examiners blinded to RLN severity, brainstem location, and side, we examined correlations between RLN severity (assessed using left distal RLn myelinated axon count) and histopathological features (including chromatolysis and glial responses) in the nucleus ambiguus cell bodies, and myelinated axon count of the right distal RLn of 16 horses. RESULTS RLN severity was not associated with RLn cell body number (P > .05), or degeneration. A positive correlation between the left and right distal RLn myelinated axon counts was identified (R2 = 0.57, P < .05). DISCUSSION We confirm that RLN, a length-dependent distal axonopathy, occurs in the absence of detectable neuronopathy.
Collapse
Affiliation(s)
- Alexandra C E Draper
- Comparative Neuromuscular Disease Laboratory, Department of Clinical Sciences and Services, Royal Veterinary College, University of London, London, UK
| | - Stephen D Cahalan
- Comparative Neuromuscular Disease Laboratory, Department of Clinical Sciences and Services, Royal Veterinary College, University of London, London, UK
| | - David Goodwin
- Comparative Neuromuscular Disease Laboratory, Department of Clinical Sciences and Services, Royal Veterinary College, University of London, London, UK
| | - Justin Perkins
- Comparative Neuromuscular Disease Laboratory, Department of Clinical Sciences and Services, Royal Veterinary College, University of London, London, UK
| | - Richard J Piercy
- Comparative Neuromuscular Disease Laboratory, Department of Clinical Sciences and Services, Royal Veterinary College, University of London, London, UK
| |
Collapse
|
37
|
Li G, Zhou S, Li C, Cai B, Yu H, Ma B, Huang Y, Ding Y, Liu Y, Ding Q, He C, Zhou J, Wang Y, Zhou G, Li Y, Yan Y, Hua J, Petersen B, Jiang Y, Sonstegard T, Huang X, Chen Y, Wang X. Base pair editing in goat: nonsense codon introgression into FGF5 results in longer hair. FEBS J 2019; 286:4675-4692. [PMID: 31276295 DOI: 10.1111/febs.14983] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 04/21/2019] [Accepted: 07/03/2019] [Indexed: 12/26/2022]
Abstract
The ability to alter single bases without homology directed repair (HDR) of double-strand breaks provides a potential solution for editing livestock genomes for economic traits, which are often multigenic. Progress toward multiplex editing in large animals has been hampered by the costly inefficiencies of HDR via microinjection of in vitro manipulated embryos. Here, we designed sgRNAs to induce nonsense codons (C-to-T transitions) at four target sites in caprine FGF5, which is a crucial regulator of hair length in mammals. Initial transfections of the third generation Base Editor (BE3) plasmid and four different sgRNAs into caprine fibroblasts were ineffective in altering FGF5. In contrast, all five progenies produced from microinjected single-cell embryos had alleles with a targeted nonsense mutation. The effectiveness of BE3 to make single base changes varied considerably based on sgRNA design. In addition, the rate of mosaicism differed between animals, target sites, and tissue type. The phenotypic effects on hair fiber were characterized by hematoxylin and eosin, immunofluorescence staining, and western blotting. Differences in morphology were detectable, even though mosaicism was probably affecting the levels of FGF5 expression. PCR amplicon and whole-genome resequencing analyses for off-target changes caused by BE3 were low at a genome-wide scale. This study provided the first evidence of base editing in large mammals produced from microinjected single-cell embryos. Our results support further optimization of BEs for introgressing complex human disease alleles into large animal models, to evaluate potential genetic improvement of complex health and production traits in a single generation.
Collapse
Affiliation(s)
- Guanwei Li
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Shiwei Zhou
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Chao Li
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Bei Cai
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Honghao Yu
- College of Biotechnology, Guilin Medical University, China
| | - Baohua Ma
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Yu Huang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Yige Ding
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Yao Liu
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Qiang Ding
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Chong He
- College of Information and Engineering, Northwest A&F University, Yangling, China
| | - Jiankui Zhou
- School of Life Science and Technology, ShanghaiTech University, China
| | - Ying Wang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Guangxian Zhou
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Yan Li
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Yuan Yan
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Jinlian Hua
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Bjoern Petersen
- Institute of Farm Animal Genetics, Friedrich-Loeffler-Institut, Neustadt, Germany
| | - Yu Jiang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | | | - Xingxu Huang
- School of Life Science and Technology, ShanghaiTech University, China
| | - Yulin Chen
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Xiaolong Wang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| |
Collapse
|
38
|
CRISPR/Cas9 mediated generation of an ovine model for infantile neuronal ceroid lipofuscinosis (CLN1 disease). Sci Rep 2019; 9:9891. [PMID: 31289301 PMCID: PMC6616324 DOI: 10.1038/s41598-019-45859-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 06/12/2019] [Indexed: 01/09/2023] Open
Abstract
The neuronal ceroid lipofuscinoses (NCLs) are a group of devastating monogenetic lysosomal disorders that affect children and young adults with no cure or effective treatment currently available. One of the more severe infantile forms of the disease (INCL or CLN1 disease) is due to mutations in the palmitoyl-protein thioesterase 1 (PPT1) gene and severely reduces the child's lifespan to approximately 9 years of age. In order to better translate the human condition than is possible in mice, we sought to produce a large animal model employing CRISPR/Cas9 gene editing technology. Three PPT1 homozygote sheep were generated by insertion of a disease-causing PPT1 (R151X) human mutation into the orthologous sheep locus. This resulted in a morphological, anatomical and biochemical disease phenotype that closely resembles the human condition. The homozygous sheep were found to have significantly reduced PPT1 enzyme activity and accumulate autofluorescent storage material, as is observed in CLN1 patients. Clinical signs included pronounced behavioral deficits as well as motor deficits and complete loss of vision, with a reduced lifespan of 17 ± 1 months at a humanely defined terminal endpoint. Magnetic resonance imaging (MRI) confirmed a significant decrease in motor cortical volume as well as increased ventricular volume corresponding with observed brain atrophy and a profound reduction in brain mass of 30% at necropsy, similar to alterations observed in human patients. In summary, we have generated the first CRISPR/Cas9 gene edited NCL model. This novel sheep model of CLN1 disease develops biochemical, gross morphological and in vivo brain alterations confirming the efficacy of the targeted modification and potential relevance to the human condition.
Collapse
|
39
|
Romar R, Cánovas S, Matás C, Gadea J, Coy P. Pig in vitro fertilization: Where are we and where do we go? Theriogenology 2019; 137:113-121. [PMID: 31182223 DOI: 10.1016/j.theriogenology.2019.05.045] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The pig is an important livestock animal. Biotechnological interest in this species has increased due to its use, among others, in the generation of transgenic animals for use in biomedicine based on its greater physiological proximity to the human species than other large domestic animals. This development has paralleled an improvement in Assisted Reproduction Techniques (ART) used for this species. However, the ability to generate animals from embryos produced entirely in vitro is still limited and a wide margin for improvement remains. Here we review the procedures, additives, and devices used during pig in vitro fertilization (IVF), focusing on the main points of each step that have offered the best results in terms of increased efficiency of the system. The lack of standardized protocols and consensus on the parameters to be assessed makes it difficult to compare results across different studies, but some conclusions are drawn from the literature. We anticipate that new physiological protocols will advance the field of swine IVF, including induction of prefertilization ZP hardening with oviductal fluid, sperm preparation by swim-up method, increased viscosity through the addition of inert molecules or reproductive biofluids, and the incorporation of 3D devices. Here we also reflect on the need to expand the variables on which the efficiency of pig IVF is based, providing new parameters that should be considered to supply more objective and quantitative assessment of IVF additives and protocols.
Collapse
Affiliation(s)
- Raquel Romar
- Department of Physiology, Faculty of Veterinary, University of Murcia, Campus Mare Nostrum, IMIB-Arrixaca, Murcia, Spain.
| | - Sebastián Cánovas
- Department of Physiology, Faculty of Veterinary, University of Murcia, Campus Mare Nostrum, IMIB-Arrixaca, Murcia, Spain
| | - Carmen Matás
- Department of Physiology, Faculty of Veterinary, University of Murcia, Campus Mare Nostrum, IMIB-Arrixaca, Murcia, Spain
| | - Joaquín Gadea
- Department of Physiology, Faculty of Veterinary, University of Murcia, Campus Mare Nostrum, IMIB-Arrixaca, Murcia, Spain
| | - Pilar Coy
- Department of Physiology, Faculty of Veterinary, University of Murcia, Campus Mare Nostrum, IMIB-Arrixaca, Murcia, Spain
| |
Collapse
|
40
|
de Graeff N, Jongsma KR, Johnston J, Hartley S, Bredenoord AL. The ethics of genome editing in non-human animals: a systematic review of reasons reported in the academic literature. Philos Trans R Soc Lond B Biol Sci 2019; 374:20180106. [PMID: 30905297 PMCID: PMC6452271 DOI: 10.1098/rstb.2018.0106] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/10/2018] [Indexed: 12/16/2022] Open
Abstract
In recent years, new genome editing technologies have emerged that can edit the genome of non-human animals with progressively increasing efficiency. Despite ongoing academic debate about the ethical implications of these technologies, no comprehensive overview of this debate exists. To address this gap in the literature, we conducted a systematic review of the reasons reported in the academic literature for and against the development and use of genome editing technologies in animals. Most included articles were written by academics from the biomedical or animal sciences. The reported reasons related to seven themes: human health, efficiency, risks and uncertainty, animal welfare, animal dignity, environmental considerations and public acceptability. Our findings illuminate several key considerations about the academic debate, including a low disciplinary diversity in the contributing academics, a scarcity of systematic comparisons of potential consequences of using these technologies, an underrepresentation of animal interests, and a disjunction between the public and academic debate on this topic. As such, this article can be considered a call for a broad range of academics to get increasingly involved in the discussion about genome editing, to incorporate animal interests and systematic comparisons, and to further discuss the aims and methods of public involvement. This article is part of a discussion meeting issue 'The ecology and evolution of prokaryotic CRISPR-Cas adaptive immune systems'.
Collapse
Affiliation(s)
- Nienke de Graeff
- Department of Medical Humanities, Julius Center, University Medical Center Utrecht/Utrecht University, PO Box 85500, Utrecht, GA 3508, The Netherlands
| | - Karin R. Jongsma
- Department of Medical Humanities, Julius Center, University Medical Center Utrecht/Utrecht University, PO Box 85500, Utrecht, GA 3508, The Netherlands
| | - Josephine Johnston
- Research Department, The Hastings Center, 21 Malcolm Gordon Road, Garrison, NY 10524, USA
| | - Sarah Hartley
- The University of Exeter Business School, University of Exeter, Rennes Drive, Exeter EX4 4PU, UK
| | - Annelien L. Bredenoord
- Department of Medical Humanities, Julius Center, University Medical Center Utrecht/Utrecht University, PO Box 85500, Utrecht, GA 3508, The Netherlands
| |
Collapse
|
41
|
García-Martínez S, Sánchez Hurtado MA, Gutiérrez H, Sánchez Margallo FM, Romar R, Latorre R, Coy P, López Albors O. Mimicking physiological O2 tension in the female reproductive tract improves assisted reproduction outcomes in pig. Mol Hum Reprod 2019; 24:260-270. [PMID: 29490063 DOI: 10.1093/molehr/gay008] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 02/26/2018] [Indexed: 01/20/2023] Open
Abstract
STUDY QUESTION Is O2 tension in the pig oviduct and uterus affected by the estrous cycle stage and the animal's age, and can the outcome of in vitro embryo development be improved by mimicking these physiological values? SUMMARY ANSWER O2 tension within the pig reproductive organs is affected by the animal's age, and values close to those measured in vivo have a positive impact on embryo development and quality when used during IVF and embryo culture (EC). WHAT IS KNOWN ALREADY To obtain a healthy embryo in vitro, it is necessary to adopt a culture microenvironment that approximates physiological conditions. Despite advances in surgical procedures and sensitive probes that allow accurate assessment of in vivo O2 tension, few such studies have been conducted recently in mammals. In addition, no reference values of physiological O2 tension in the reproductive tract exist for large animal models such as pig, and the effect of O2 tension on ART outcomes is unknown. STUDY DESIGN, SIZE, DURATION This study was conducted in pigs. We measured oviductal and uterine O2 tension (n = 29 and 13, respectively) and then examined how the use of the physiological values in pig IVF and EC affected pig ART output (n = 1447 oocytes). PARTICIPANTS/MATERIALS, SETTING, METHODS The oviductal and uterine O2 tension at the different stages of the estrous cycle was monitored using a laparo-endoscopic single-site surgery (LESS) assisted approach along with a flexible and thin miniaturized luminescent probe. Two groups of pigs, Large-white × Landrace breed, were used: for the first group, 16 pre-pubertal gilts (5 months old and 95 kg) were induced to ovulate with equine chorionic gonadotropin (eCG) and human chorionic gonadotropin (hCG); in the second group 13 mature sows (24-48 months and 185 kg) were used. IVF and EC were performed at two different O2 tensions: Atmospheric O2 (20%) and the mean in vivo value measured (7%). At 18-20 h post-insemination (hpi), a small sample of presumptive zygotes were fixed, stained and examined under epifluorescence microscopy to assess the fertilization rates. At 48 hpi, cleavage was evaluated under the stereomicroscope. Finally, at 180 hpi, development to the blastocyst stage was quantified, blastocyst morphology was assessed, and embryos were fixed and stained to count the mean cell number per blastocyst. MAIN RESULTS AND THE ROLE OF CHANCE The mean O2 content within the pig oviduct and uterus was always lower than in ambient air. The average O2 percentage was higher in gilts (10.0%) than in sows (7.6%) (P < 0.0001). The cleavage rate of porcine in vitro fertilized embryos maintained under 7% O2 during IVF and EC was significantly higher (60.0 ± 2.3) compared with those cultured under 20% O2 (32.0 ± 2.2) (P < 0.05). An increase in the number of cells in embryos cultured under the low O2 concentration (88.9 ± 5.9) was observed compared to those cultured under 20% O2 (59.0 ± 5.0) (P < 0.05). LARGE SCALE DATA None. LIMITATIONS, REASONS FOR CAUTION Although minimally invasive surgery was used the effect of anesthesia and manipulations on O2 tension within the organs are unknown. WIDER IMPLICATIONS OF THE FINDINGS Using physiological oxygen concentrations in IVF/EC could improve ART outcomes. STUDY FUNDING AND COMPETING INTEREST(S) This study was funded by Spanish Ministry of Economy and Competitiveness (MINECO) and European Regional Development Fund (FEDER). Grants AGL2012-40180-C03-01 and AGL2015-66341-R. The authors declare no conflict of interest.
Collapse
Affiliation(s)
- S García-Martínez
- Dept. Physiology, Facultad de Veterinaria, Campus Internacional de Excelencia de Educación Superior (Campus Mare Nostrum), Universidad de Murcia, Murcia 30100, España.,Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca), Murcia, España
| | - M A Sánchez Hurtado
- Laparoscopy Unit, Minimally Invasive Surgery Center Jesús Usón, Cáceres 10071, España
| | - H Gutiérrez
- Dept. Animal Health, Faculty of Veterinary Medicine and Zootechnics, University Nacional of Colombia, Carrera 30 45-03, Bogotá, Colombia.,Dept. Anatomy & Comparative Physiology, Facultad de Veterinaria, Campus Internacional de Excelencia de Educación Superior (Campus Mare Nostrum), Universidad de Murcia, Murcia 30100, España
| | - F M Sánchez Margallo
- Laparoscopy Unit, Minimally Invasive Surgery Center Jesús Usón, Cáceres 10071, España
| | - R Romar
- Dept. Physiology, Facultad de Veterinaria, Campus Internacional de Excelencia de Educación Superior (Campus Mare Nostrum), Universidad de Murcia, Murcia 30100, España.,Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca), Murcia, España
| | - R Latorre
- Dept. Anatomy & Comparative Physiology, Facultad de Veterinaria, Campus Internacional de Excelencia de Educación Superior (Campus Mare Nostrum), Universidad de Murcia, Murcia 30100, España
| | - P Coy
- Dept. Physiology, Facultad de Veterinaria, Campus Internacional de Excelencia de Educación Superior (Campus Mare Nostrum), Universidad de Murcia, Murcia 30100, España.,Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca), Murcia, España
| | - O López Albors
- Dept. Anatomy & Comparative Physiology, Facultad de Veterinaria, Campus Internacional de Excelencia de Educación Superior (Campus Mare Nostrum), Universidad de Murcia, Murcia 30100, España
| |
Collapse
|
42
|
Volobueva AS, Orekhov AN, Deykin AV. An update on the tools for creating transgenic animal models of human diseases - focus on atherosclerosis. ACTA ACUST UNITED AC 2019; 52:e8108. [PMID: 31038578 PMCID: PMC6487744 DOI: 10.1590/1414-431x20198108] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 02/25/2019] [Indexed: 12/19/2022]
Abstract
Animal models of diseases are invaluable tools of modern medicine. More than forty years have passed since the first successful experiments and the spectrum of available models, as well as the list of methods for creating them, have expanded dramatically. The major step forward in creating specific disease models was the development of gene editing techniques, which allowed for targeted modification of the animal's genome. In this review, we discuss the available tools for creating transgenic animal models, such as transgenesis methods, recombinases, and nucleases, including zinc finger nuclease (ZFN), transcription activator-like effector nuclease (TALEN), and CRISPR/Cas9 systems. We then focus specifically on the models of atherosclerosis, especially mouse models that greatly contributed to improving our understanding of the disease pathogenesis and we outline their characteristics and limitations.
Collapse
Affiliation(s)
- A S Volobueva
- Laboratory of Gene Therapy, Biocad Biotechnology Company, Strelnya, Russia
| | - A N Orekhov
- Institute of Gene Biology, Russian Academy of Sciences, Moscow, Russia.,Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Moscow, Russia.,Institute for Atherosclerosis Research, Skolkovo Innovative Center, Moscow, Russia
| | - A V Deykin
- Institute of Gene Biology, Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
43
|
Dorado B, Pløen GG, Barettino A, Macías A, Gonzalo P, Andrés-Manzano MJ, González-Gómez C, Galán-Arriola C, Alfonso JM, Lobo M, López-Martín GJ, Molina A, Sánchez-Sánchez R, Gadea J, Sánchez-González J, Liu Y, Callesen H, Filgueiras-Rama D, Ibáñez B, Sørensen CB, Andrés V. Generation and characterization of a novel knockin minipig model of Hutchinson-Gilford progeria syndrome. Cell Discov 2019; 5:16. [PMID: 30911407 PMCID: PMC6423020 DOI: 10.1038/s41421-019-0084-z] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 01/15/2019] [Accepted: 01/22/2019] [Indexed: 01/22/2023] Open
Abstract
Hutchinson-Gilford progeria syndrome (HGPS) is an extremely rare genetic disorder for which no cure exists. The disease is characterized by premature aging and inevitable death in adolescence due to cardiovascular complications. Most HGPS patients carry a heterozygous de novo LMNA c.1824C > T mutation, which provokes the expression of a dominant-negative mutant protein called progerin. Therapies proven effective in HGPS-like mouse models have yielded only modest benefit in HGPS clinical trials. To overcome the gap between HGPS mouse models and patients, we have generated by CRISPR-Cas9 gene editing the first large animal model for HGPS, a knockin heterozygous LMNA c.1824C > T Yucatan minipig. Like HGPS patients, HGPS minipigs endogenously co-express progerin and normal lamin A/C, and exhibit severe growth retardation, lipodystrophy, skin and bone alterations, cardiovascular disease, and die around puberty. Remarkably, the HGPS minipigs recapitulate critical cardiovascular alterations seen in patients, such as left ventricular diastolic dysfunction, altered cardiac electrical activity, and loss of vascular smooth muscle cells. Our analysis also revealed reduced myocardial perfusion due to microvascular damage and myocardial interstitial fibrosis, previously undescribed readouts potentially useful for monitoring disease progression in patients. The HGPS minipigs provide an appropriate preclinical model in which to test human-size interventional devices and optimize candidate therapies before advancing to clinical trials, thus accelerating the development of effective applications for HGPS patients.
Collapse
Affiliation(s)
- Beatriz Dorado
- 1Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain.,CIBER en Enfermedades Cardiovasculares (CIBER-CV), Madrid, Spain
| | - Gro Grunnet Pløen
- 3Department of Clinical Medicine, Aarhus University, 8200 Aarhus, Denmark.,4Department of Cardiology, Aarhus University Hospital, 8200 Aarhus, Denmark
| | - Ana Barettino
- 1Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain.,CIBER en Enfermedades Cardiovasculares (CIBER-CV), Madrid, Spain
| | - Alvaro Macías
- 1Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain
| | - Pilar Gonzalo
- 1Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain.,CIBER en Enfermedades Cardiovasculares (CIBER-CV), Madrid, Spain
| | - María Jesús Andrés-Manzano
- 1Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain.,CIBER en Enfermedades Cardiovasculares (CIBER-CV), Madrid, Spain
| | - Cristina González-Gómez
- 1Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain.,CIBER en Enfermedades Cardiovasculares (CIBER-CV), Madrid, Spain
| | - Carlos Galán-Arriola
- 1Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain.,CIBER en Enfermedades Cardiovasculares (CIBER-CV), Madrid, Spain
| | - José Manuel Alfonso
- 1Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain
| | - Manuel Lobo
- 1Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain.,CIBER en Enfermedades Cardiovasculares (CIBER-CV), Madrid, Spain
| | | | - Antonio Molina
- 1Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain
| | - Raúl Sánchez-Sánchez
- 5Laboratory of Physiology and Biotechnology of Reproduction in Swine, INIA (Spanish National Institute for Agricultural and Food Research and Technology), Madrid, Spain
| | - Joaquín Gadea
- 6Department of Physiology, University of Murcia and IMIB-Arrixaca, 30100 Murcia, Spain
| | | | - Ying Liu
- 8Department of Animal Science, Aarhus University, 8830 Tjele, Denmark
| | - Henrik Callesen
- 8Department of Animal Science, Aarhus University, 8830 Tjele, Denmark
| | - David Filgueiras-Rama
- 1Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain.,CIBER en Enfermedades Cardiovasculares (CIBER-CV), Madrid, Spain.,9Department of Cardiology, Cardiac Electrophysiology Unit, Hospital Clínico San Carlos, Madrid, Spain
| | - Borja Ibáñez
- 1Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain.,CIBER en Enfermedades Cardiovasculares (CIBER-CV), Madrid, Spain.,10Department of Cardiology, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz Hospital, Madrid, Spain
| | - Charlotte Brandt Sørensen
- 3Department of Clinical Medicine, Aarhus University, 8200 Aarhus, Denmark.,4Department of Cardiology, Aarhus University Hospital, 8200 Aarhus, Denmark
| | - Vicente Andrés
- 1Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain.,CIBER en Enfermedades Cardiovasculares (CIBER-CV), Madrid, Spain
| |
Collapse
|
44
|
Chansel‐Debordeaux L, Bezard E. Local transgene expression and whole-body transgenesis to model brain diseases in nonhuman primate. Animal Model Exp Med 2019; 2:9-17. [PMID: 31016282 PMCID: PMC6431118 DOI: 10.1002/ame2.12055] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 12/10/2018] [Indexed: 12/26/2022] Open
Abstract
Animal model is an essential tool in the life sciences research, notably in understanding the pathogenesis of the diseases and for further therapeutic intervention success. Rodents have been the most frequently used animals to model human disease since the establishment of gene manipulation technique. However, they remain inadequate to fully mimic the pathophysiology of human brain disease, partially due to huge differences between rodents and humans in terms of anatomy, brain function, and social behaviors. Nonhuman primates are more suitable in translational perspective. Thus, genetically modified animals have been generated to investigate neurologic and psychiatric disorders. The classical transgenesis technique is not efficient in that model; so, viral vector-mediated transgene delivery and the new genome-editing technologies have been promoted. In this review, we summarize some of the technical progress in the generation of an ad hoc animal model of brain diseases by gene delivery and real transgenic nonhuman primate.
Collapse
Affiliation(s)
- Lucie Chansel‐Debordeaux
- Institut des Maladies NeurodégénérativesUniversity of BordeauxUMR 5293BordeauxFrance
- CNRSInstitut des Maladies NeurodégénérativesUMR 5293BordeauxFrance
- CHU BordeauxService de Biologie de la reproduction‐CECOSBordeauxFrance
| | - Erwan Bezard
- Institut des Maladies NeurodégénérativesUniversity of BordeauxUMR 5293BordeauxFrance
- CNRSInstitut des Maladies NeurodégénérativesUMR 5293BordeauxFrance
| |
Collapse
|
45
|
Production of Cloned Pigs Derived from Double Gene Knockout Cells Using CRISPR/Cas9 System and MACS-based Enrichment System. JOURNAL OF ANIMAL REPRODUCTION AND BIOTECHNOLOGY 2018. [DOI: 10.12750/jet.2018.33.4.245] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
|
46
|
Many Cells Make Life Work-Multicellularity in Stem Cell-Based Cardiac Disease Modelling. Int J Mol Sci 2018; 19:ijms19113361. [PMID: 30373227 PMCID: PMC6274721 DOI: 10.3390/ijms19113361] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 10/23/2018] [Accepted: 10/24/2018] [Indexed: 12/22/2022] Open
Abstract
Cardiac disease causes 33% of deaths worldwide but our knowledge of disease progression is still very limited. In vitro models utilising and combining multiple, differentiated cell types have been used to recapitulate the range of myocardial microenvironments in an effort to delineate the mechanical, humoral, and electrical interactions that modulate the cardiac contractile function in health and the pathogenesis of human disease. However, due to limitations in isolating these cell types and changes in their structure and function in vitro, the field is now focused on the development and use of stem cell-derived cell types, most notably, human-induced pluripotent stem cell-derived CMs (hiPSC-CMs), in modelling the CM function in health and patient-specific diseases, allowing us to build on the findings from studies using animal and adult human CMs. It is becoming increasingly appreciated that communications between cardiomyocytes (CMs), the contractile cell of the heart, and the non-myocyte components of the heart not only regulate cardiac development and maintenance of health and adult CM functions, including the contractile state, but they also regulate remodelling in diseases, which may cause the chronic impairment of the contractile function of the myocardium, ultimately leading to heart failure. Within the myocardium, each CM is surrounded by an intricate network of cell types including endothelial cells, fibroblasts, vascular smooth muscle cells, sympathetic neurons, and resident macrophages, and the extracellular matrix (ECM), forming complex interactions, and models utilizing hiPSC-derived cell types offer a great opportunity to investigate these interactions further. In this review, we outline the historical and current state of disease modelling, focusing on the major milestones in the development of stem cell-derived cell types, and how this technology has contributed to our knowledge about the interactions between CMs and key non-myocyte components of the heart in health and disease, in particular, heart failure. Understanding where we stand in the field will be critical for stem cell-based applications, including the modelling of diseases that have complex multicellular dysfunctions.
Collapse
|
47
|
Xia T, Yu F, Zhang K, Wu Z, Shi D, Teng H, Shen J, Yang X, Jiang Q. The effectiveness of allogeneic mesenchymal stem cells therapy for knee osteoarthritis in pigs. ANNALS OF TRANSLATIONAL MEDICINE 2018; 6:404. [PMID: 30498731 DOI: 10.21037/atm.2018.09.55] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Background Intraarticular injection of the mesenchymal stem cells (MSCs) has shown to be successful for treating osteoarthritis (OA). Nevertheless, many studies have been focusing on autologous MSCs. The following study investigates the safety and effectiveness of intraarticular injection of allogenic MSCs in a pig OA model. Methods Superparamagnetic iron oxide (SPIO) nanoparticles were labelled with bone marrow-derived mesenchymal stem cells (BM-MSCs) to allow cells tracking using magnetic resonance imaging (MRI). A pig OA model was established by bilateral medial meniscectomy. Next, SPIO-BM-MSCs were injected into the right knee, while the left knee was left untreated. MRI and radiography were used to assess the degree of OA and to evaluate the effectiveness of allogenic MSCs. Hematoxylin and eosin (H&E), safranin-o fast green staining, toluidine blue, and immunohistochemical staining were used to evaluate the therapeutic effect of the injections. Results At concentration of ≤20 µg/mL, SPIO caused no toxicity to BM-MSCs. Four weeks after surgery, OA changes were observed on MRI scan. The SPIO labeled BM-MSCs were found moving towards the impaired part of the cartilage 8 to 24 h after injections. In addition, no significant differences between the right side (therapeutic side) and the left side (untreated side) were observed following histological and immunohistochemistry analysis. Conclusions The suitable concentration of SPIO for labelling BMSCs was 20 µg/mL, while the allogenic MSCs could move towards and accumulate around the impaired cartilage. No significant difference was found between treatment and control group.
Collapse
Affiliation(s)
- Tianwei Xia
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210046, China
| | - Fei Yu
- Drum Tower of Clinical Medicine, Nanjing Medical University, Nanjing 210046, China.,Department of Sports Medicine and Adult Reconstructive Surgery, Drum Tower Hospital, School of Medicine, Nanjing University, Nanjing 210008, China
| | - Kaijia Zhang
- Department of Sports Medicine and Adult Reconstructive Surgery, Drum Tower Hospital, School of Medicine, Nanjing University, Nanjing 210008, China
| | - Zongfang Wu
- Drum Tower of Clinical Medicine, Nanjing Medical University, Nanjing 210046, China.,Department of Sports Medicine and Adult Reconstructive Surgery, Drum Tower Hospital, School of Medicine, Nanjing University, Nanjing 210008, China
| | - Dongquan Shi
- Department of Sports Medicine and Adult Reconstructive Surgery, Drum Tower Hospital, School of Medicine, Nanjing University, Nanjing 210008, China
| | - Huajian Teng
- Joint Research Center for Bone and Joint Disease, Model Animal Research Center (MARC), Nanjing University, Nanjing 210008, China
| | - Jirong Shen
- Department of Orthopedics, Jiangsu Provincial Hospital of Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210046, China
| | - Xianfeng Yang
- Department of Radiology, Drum Tower Hospital, School of Medicine, Nanjing University, Nanjing 210008, China
| | - Qing Jiang
- Drum Tower of Clinical Medicine, Nanjing Medical University, Nanjing 210046, China.,Department of Sports Medicine and Adult Reconstructive Surgery, Drum Tower Hospital, School of Medicine, Nanjing University, Nanjing 210008, China
| |
Collapse
|
48
|
A comparative study of cartilage engineered constructs in immunocompromised, humanized and immunocompetent mice. ACTA ACUST UNITED AC 2018. [DOI: 10.1016/j.regen.2018.09.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
49
|
Silveira MM, Salgado Bayão HX, Dos Santos Mendonça A, Borges NA, Vargas LN, Caetano AR, Rumpf R, Franco MM. DNA methylation profile at a satellite region is associated with aberrant placentation in cloned calves. Placenta 2018; 70:25-33. [PMID: 30316323 DOI: 10.1016/j.placenta.2018.08.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 08/18/2018] [Accepted: 08/28/2018] [Indexed: 01/21/2023]
Abstract
INTRODUCTION Cloning via somatic cell nuclear transfer (SCNT) has been associated with a variety of pathologies, primarily in the placenta, and these alterations may be associated with aberrant epigenetic reprogramming of the donor cell genome. We tested the hypothesis that DNA methylation patterns are not appropriately established after nuclear transfer and that those altered patterns are associated with specific aberrant phenotypes. METHODS We compared global and specific placental DNA methylation patterns between aberrant and healthy SCNT-produced calves. Foetal cotyledon samples of ten SCNT pregnancies were collected. Global DNA methylation and hydroxymethylation levels were measured using an ELISA-based assay and specific DNA methylation of satellite I, and α-satellite repeat elements were measured using bisulfite PCR. RESULTS Our analysis revealed that the SCNT-produced calves, which showed aberrant phenotypes, exhibited a reduced methylation pattern of the satellite I region compared to that of healthy calves. In contrast, global methylation and hydroxymethylation analyses showed higher levels for both cytosine modifications in SCNT-produced female calves with aberrant phenotypes. The satellite I region showed most of the sequences to be hypermethylated in live cloned calves compared with those in deceased calves. DISCUSSION Our results suggest that this satellite I region could be used as an epigenetic biomarker for predicting offspring viability. Studies evaluating DNA methylation patterns of this satellite region in the donor cell genome or embryo biopsies could shed light on how to improve the efficiency of SCNT cloning.
Collapse
Affiliation(s)
- Márcia Marques Silveira
- Laboratory of Animal Reproduction, Embrapa Genetic Resources and Biotechnology, Brasília, Distrito Federal, Brazil; Institute of Genetics and Biochemistry, Federal University of Uberlândia, Uberlândia, Minas Gerais, Brazil.
| | | | - Anelise Dos Santos Mendonça
- Laboratory of Animal Reproduction, Embrapa Genetic Resources and Biotechnology, Brasília, Distrito Federal, Brazil; Institute of Genetics and Biochemistry, Federal University of Uberlândia, Uberlândia, Minas Gerais, Brazil.
| | - Naiara Araújo Borges
- Laboratory of Animal Reproduction, Embrapa Genetic Resources and Biotechnology, Brasília, Distrito Federal, Brazil; Institute of Genetics and Biochemistry, Federal University of Uberlândia, Uberlândia, Minas Gerais, Brazil.
| | - Luna Nascimento Vargas
- Laboratory of Animal Reproduction, Embrapa Genetic Resources and Biotechnology, Brasília, Distrito Federal, Brazil; Institute of Genetics and Biochemistry, Federal University of Uberlândia, Uberlândia, Minas Gerais, Brazil.
| | | | - Rodolfo Rumpf
- GENEAL Genetics and Animal Biotechnology, Uberaba, Minas Gerais, Brazil.
| | - Maurício Machaim Franco
- Laboratory of Animal Reproduction, Embrapa Genetic Resources and Biotechnology, Brasília, Distrito Federal, Brazil; Institute of Genetics and Biochemistry, Federal University of Uberlândia, Uberlândia, Minas Gerais, Brazil.
| |
Collapse
|
50
|
Lamas-Toranzo I, Ramos-Ibeas P, Pericuesta E, Bermejo-Álvarez P. Directions and applications of CRISPR technology in livestock research. Anim Reprod 2018; 15:292-300. [PMID: 34178152 PMCID: PMC8202460 DOI: 10.21451/1984-3143-ar2018-0075] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The ablation (KO) or targeted insertion (KI) of specific genes or sequences has been essential
to test their roles on a particular biological process. Unfortunately, such genome modifications
have been largely limited to the mouse model, as the only way to achieve targeted mutagenesis
in other mammals required from somatic cell nuclear transfer, a time- and resource-consuming
technique. This difficulty has left research in livestock species largely devoided of KO
and targeted KI models, crucial tools to uncover the molecular roots of any physiological
or pathological process. Luckily, the eruption of site-specific endonucleases, and particularly
CRISPR technology, has empowered farm animal scientists to consider projects that could
not develop before. In this sense, the availability of genome modification in livestock species
is meant to change the way research is performed on many fields, switching from descriptive
and correlational approaches to experimental research. In this review we will provide some
guidance about how the genome can be edited by CRISPR and the possible strategies to achieve
KO or KI, paying special attention to an initially overlooked phenomenon: mosaicism. Mosaicism
is produced when the zygote´s genome edition occurs after its DNA has replicated,
and is characterized by the presence of more than two alleles in the same individual, an undesirable
outcome when attempting direct KO generation. Finally, the possible applications on different
fields of livestock research, such as reproduction or infectious diseases are discussed.
Collapse
Affiliation(s)
| | | | - Eva Pericuesta
- Department Reproducción Animal, INIA, 28040 Madrid, Spain
| | | |
Collapse
|