51
|
Küchler EC, Mazzi-Chaves JF, Antunes LS, Kirschneck C, Baratto-Filho F, Sousa-Neto MD. Current trends of genetics in apical periodontitis research. Braz Oral Res 2018; 32:e72. [DOI: 10.1590/1807-3107bor-2018.vol32.0072] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 06/06/2018] [Indexed: 12/12/2022] Open
|
52
|
Delgado Caceres M, Pfeifer CG, Docheva D. Understanding Tendons: Lessons from Transgenic Mouse Models. Stem Cells Dev 2018; 27:1161-1174. [PMID: 29978741 PMCID: PMC6121181 DOI: 10.1089/scd.2018.0121] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 07/05/2018] [Indexed: 12/26/2022] Open
Abstract
Tendons and ligaments are connective tissues that have been comparatively less studied than muscle and cartilage/bone, even though they are crucial for proper function of the musculoskeletal system. In tendon biology, considerable progress has been made in identifying tendon-specific genes (Scleraxis, Mohawk, and Tenomodulin) in the past decade. However, besides tendon function and the knowledge of a small number of important players in tendon biology, neither the ontogeny of the tenogenic lineage nor signaling cascades have been fully understood. This results in major drawbacks in treatment and repair options following tendon degeneration. In this review, we have systematically evaluated publications describing tendon-related genes, which were studied in depth and characterized by using knockout technologies and the subsequently generated transgenic mouse models (Tg) (knockout mice, KO). We report in a tabular manner, that from a total of 24 tendon-related genes, in 22 of the respective knockout mouse models, phenotypic changes were detected. Additionally, in some of the models it was described at which developmental stages these changes appeared and progressed. To summarize, only loss of Scleraxis and TGFβ signaling led to severe tendon developmental phenotypes, while mice deficient for various proteoglycans, Mohawk, EGR1 and 2, and Tenomodulin presented mild phenotypes. These data suggest that the tendon developmental system is well organized, orchestrated, and backed up; this is even more evident among the members of the proteoglycan family, where the compensatory effects are much clearer. In future, it will be of great importance to discover additional master tendon transcription factors and the genes that play crucial roles in tendon development. This would improve our understanding of the genetic makeup of tendons, and will increase the chances of generating tendon-specific drugs to advance overall treatment strategies.
Collapse
Affiliation(s)
- Manuel Delgado Caceres
- Experimental Trauma Surgery, Department of Trauma Surgery, University Regensburg Medical Centre, Regensburg, Germany
| | - Christian G. Pfeifer
- Experimental Trauma Surgery, Department of Trauma Surgery, University Regensburg Medical Centre, Regensburg, Germany
- Department of Trauma Surgery, University Regensburg Medical Centre, Regensburg, Germany
| | - Denitsa Docheva
- Experimental Trauma Surgery, Department of Trauma Surgery, University Regensburg Medical Centre, Regensburg, Germany
- Department of Medical Biology, Medical University-Plovdiv, Plovdiv, Bulgaria
| |
Collapse
|
53
|
Li T, Su Y, Yu X, Mouniir DSA, Masau JF, Wei X, Yang J. Trop2 Guarantees Cardioprotective Effects of Cortical Bone-Derived Stem Cells on Myocardial Ischemia/Reperfusion Injury. Cell Transplant 2018; 27:1256-1268. [PMID: 30008230 PMCID: PMC6434467 DOI: 10.1177/0963689718786882] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Stem cell transplantation represents a promising therapeutic approach for myocardial ischemia/reperfusion (I/R) injury, where cortical bone-derived stem cells (CBSCs) stand out and hold superior cardioprotective effects on myocardial infarction than other types of stem cells. However, the molecular mechanism underlying CBSCs function on myocardial I/R injury is poorly understood. In a previous study, we reported that Trop2 (trophoblast cell-surface antigen 2) is expressed exclusively on the CBSCs membrane, and is involved in regulation of proliferation and differentiation of CBSCs. In this study, we found that the Trop2 is essential for the ameliorative effects of CBSCs on myocardial I/R-induced heart damage via promoting angiogenesis and inhibiting cardiomyocytes apoptosis in a paracrine manner. Trop2 is required for the colonization of CBSCs in recipient hearts. When Trop2 was knocked out, CBSCs largely lost their functions in lowering myocardial infarction size, improving heart function, enhancing capillary density, and suppressing myocardial cell death. Mechanistically, activating the AKT/GSK3β/β-Catenin signaling axis contributes to the essential role of Trop2 in CBSCs-rendered cardioprotective effects on myocardial I/R injury. In conclusion, maintaining the expression and/or activation of Trop2 in CBSCs might be a promising strategy for treating myocardial infarction, I/R injury, and other related heart diseases.
Collapse
Affiliation(s)
- Tianyu Li
- 1 Division of Cardiothoracic and Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.,2 Division of Trauma Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yunshu Su
- 1 Division of Cardiothoracic and Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiongli Yu
- 3 Division of Biliary-Pancreatic Surgery and Endoscopy Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Durgahee S A Mouniir
- 1 Division of Cardiothoracic and Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jackson Ferdinand Masau
- 1 Division of Cardiothoracic and Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiang Wei
- 1 Division of Cardiothoracic and Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jianye Yang
- 1 Division of Cardiothoracic and Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
54
|
Chang CY, Ting HC, Su HL, Jeng JR. Combining Induced Pluripotent Stem Cells and Genome Editing Technologies for Clinical Applications. Cell Transplant 2018; 27:379-392. [PMID: 29806481 PMCID: PMC6038034 DOI: 10.1177/0963689718754560] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
In this review, we introduce current developments in induced pluripotent stem cells (iPSCs), site-specific nuclease (SSN)-mediated genome editing tools, and the combined application of these two novel technologies in biomedical research and therapeutic trials. The sustainable pluripotent property of iPSCs in vitro not only provides unlimited cell sources for basic research but also benefits precision medicines for human diseases. In addition, rapidly evolving SSN tools efficiently tailor genetic manipulations for exploring gene functions and can be utilized to correct genetic defects of congenital diseases in the near future. Combining iPSC and SSN technologies will create new reliable human disease models with isogenic backgrounds in vitro and provide new solutions for cell replacement and precise therapies.
Collapse
Affiliation(s)
- Chia-Yu Chang
- 1 Bio-innovation Center, Tzu Chi Foundation, Hualien, Taiwan.,2 Department of Medical Research, Buddhist Tzu Chi General Hospital, Hualien, Taiwan
| | | | - Hong-Lin Su
- 3 Department of Life Sciences, Agricultural Biotechnology Center, National Chung Hsing University, Taichung, Taiwan
| | - Jing-Ren Jeng
- 4 Division of Cardiology, Department of Internal Medicine, Buddhist Tzu Chi General Hospital, Tzu Chi University, Hualien, Taiwan
| |
Collapse
|
55
|
Lee CC, Hsieh TS. Wuho/WDR4 deficiency inhibits cell proliferation and induces apoptosis via DNA damage in mouse embryonic fibroblasts. Cell Signal 2018; 47:16-26. [PMID: 29574139 DOI: 10.1016/j.cellsig.2018.03.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 03/16/2018] [Accepted: 03/19/2018] [Indexed: 12/27/2022]
Abstract
Wuho known as WDR4 encodes a highly conserved WD40-repeat protein, which has known homologues of WDR4 in human and mouse. Wuho-FEN1 interaction may have a critical role in the growth and development, and in the maintenance of genome stability. However, how Wuho gene deletion contributes to cell growth inhibition and apoptosis is still unknown. We utilized CAGGCre-ER transgenic mice have a tamoxifen-inducible cre-mediated recombination cassette to prepare primary mouse embryonic fibroblasts (MEFs) with Wuho deficiency. We have demonstrated that Wuho deficiency would induces γH2AX protein level elevation, heterochromatin relaxation and DNA damage down-stream sequences, including p53 activation, caspase-mediated apoptotic pathway, and p21-mediated G2/M cell cycle arrest.
Collapse
Affiliation(s)
- Chi-Chiu Lee
- Institute of Cellular and Organismic Biology, Academia Sinica, No. 128, Academia Road, Sec. 2, Nangang, Taipei 11529, Taiwan.
| | - Tao-Shih Hsieh
- Institute of Cellular and Organismic Biology, Academia Sinica, No. 128, Academia Road, Sec. 2, Nangang, Taipei 11529, Taiwan; Department of Biochemistry, Duke University, Durham, NC, United States
| |
Collapse
|
56
|
Upadhaya S, Reizis B, Sawai CM. New genetic tools for the in vivo study of hematopoietic stem cell function. Exp Hematol 2018; 61:26-35. [PMID: 29501466 DOI: 10.1016/j.exphem.2018.02.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 02/13/2018] [Accepted: 02/14/2018] [Indexed: 11/18/2022]
Abstract
The production of blood cells is dependent on the activity of a rare stem cell population that normally resides in the bone marrow (BM) of the organism. These hematopoietic stem cells (HSCs) have the ability to both self-renew and differentiate, ensuring this lifelong hematopoiesis. Determining the regulation of HSC functions should thus provide critical insight to advancing regenerative medicine. Until quite recently, HSCs were primarily studied using in vitro studies and transplantations into immunodeficient hosts. Indeed, the definition of a bona fide HSC is its ability to reconstitute lymphopenic hosts. In this review, we discuss the development of novel, HSC-specific genetic reporter systems that enable the prospective identification of HSCs and the study of their functions in the absence of transplantation. Coupled with additional technological advances, these studies are now defining the fundamental properties of HSCs in vivo. Furthermore, complex cellular and molecular mechanisms that regulate HSC dormancy, self-renewal, and differentiation are being identified and further dissected. These novel reporter systems represent a major technological advance for the stem cell field and allow new questions to be addressed.
Collapse
Affiliation(s)
- Samik Upadhaya
- Graduate Program in Pathobiology and Molecular Medicine, Columbia University Medical Center, New York, NY, USA; Department of Pathology, New York University Langone Medical Center, New York, NY, USA
| | - Boris Reizis
- Department of Pathology, New York University Langone Medical Center, New York, NY, USA; Department of Medicine, New York University Langone Medical Center, New York, NY, USA
| | - Catherine M Sawai
- ACTION Laboratory, INSERM Unit 1218, University of Bordeaux, Bordeaux, France.
| |
Collapse
|
57
|
Meier RPH, Muller YD, Balaphas A, Morel P, Pascual M, Seebach JD, Buhler LH. Xenotransplantation: back to the future? Transpl Int 2018; 31:465-477. [PMID: 29210109 DOI: 10.1111/tri.13104] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 10/05/2017] [Accepted: 11/26/2017] [Indexed: 12/26/2022]
Abstract
The field of xenotransplantation has fluctuated between great optimism and doubts over the last 50 years. The initial clinical attempts were extremely ambitious but faced technical and ethical issues that prompted the research community to go back to preclinical studies. Important players left the field due to perceived xenozoonotic risks and the lack of progress in pig-to-nonhuman-primate transplant models. Initial apparently unsurmountable issues appear now to be possible to overcome due to progress of genetic engineering, allowing the generation of multiple-xenoantigen knockout pigs that express human transgenes and the genomewide inactivation of porcine endogenous retroviruses. These important steps forward were made possible by new genome editing technologies, such as CRISPR/Cas9, allowing researchers to precisely remove or insert genes anywhere in the genome. An additional emerging perspective is the possibility of growing humanized organs in pigs using blastocyst complementation. This article summarizes the current advances in xenotransplantation research in nonhuman primates, and it describes the newly developed genome editing technology tools and interspecific organ generation.
Collapse
Affiliation(s)
- Raphael P H Meier
- Visceral and Transplant Surgery, University Hospitals of Geneva, Geneva, Switzerland
| | - Yannick D Muller
- Division of Clinical Immunology and Allergy, Department of Medical Specialties, University Hospitals and Medical Faculty, Geneva, Switzerland.,Transplantation Center, Lausanne University Hospital, Lausanne, Switzerland
| | - Alexandre Balaphas
- Visceral and Transplant Surgery, University Hospitals of Geneva, Geneva, Switzerland
| | - Philippe Morel
- Visceral and Transplant Surgery, University Hospitals of Geneva, Geneva, Switzerland
| | - Manuel Pascual
- Transplantation Center, Lausanne University Hospital, Lausanne, Switzerland
| | - Jörg D Seebach
- Division of Clinical Immunology and Allergy, Department of Medical Specialties, University Hospitals and Medical Faculty, Geneva, Switzerland
| | - Leo H Buhler
- Visceral and Transplant Surgery, University Hospitals of Geneva, Geneva, Switzerland
| |
Collapse
|
58
|
Abbasi F, Miyata H, Ikawa M. Revolutionizing male fertility factor research in mice by using the genome editing tool CRISPR/Cas9. Reprod Med Biol 2018; 17:3-10. [PMID: 29371815 PMCID: PMC5768971 DOI: 10.1002/rmb2.12067] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 09/01/2014] [Indexed: 12/21/2022] Open
Abstract
Background Reproductive research is quintessential in understanding not only the cause of infertility, but also for creating family planning tools. The knockout (KO) system approach is conducive to discovering genes that are essential for fertility in mice. However, in vivo research has been limited due to its high cost and length of time needed to establish KO mice. Methods The mechanisms behind the CRISPR/Cas9 system and its application in investigating male fertility in mice are described by using original and review articles. Results The CRISPR/CAS9 SYSTEM has enabled researchers to rapidly, efficiently, and inexpensively produce genetically modified mice to study male fertility. Several genes have been highlighted that were found to be indispensable for male fertility by using the CRISPR/Cas9 system, as well as more complicated gene manipulation techniques, such as point mutations, tag insertions, and double knockouts, which have become easier with this new technology. Conclusion In order to increase efficiency and usage, new methods of CRISPR/Cas9 integration are being developed, such as electroporation and applying the system to embryonic stem cells. The hidden mysteries of male fertility will be unraveled with the help of this new technology.
Collapse
Affiliation(s)
- Ferheen Abbasi
- Research Institute for Microbial DiseasesOsaka UniversitySuitaJapan
- Graduate School of MedicineOsaka UniversitySuitaJapan
| | - Haruhiko Miyata
- Research Institute for Microbial DiseasesOsaka UniversitySuitaJapan
| | - Masahito Ikawa
- Research Institute for Microbial DiseasesOsaka UniversitySuitaJapan
- Graduate School of MedicineOsaka UniversitySuitaJapan
| |
Collapse
|
59
|
Wu Q, Han Y, Tong Q. Current Genetic Techniques in Neural Circuit Control of Feeding and Energy Metabolism. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1090:211-233. [PMID: 30390293 DOI: 10.1007/978-981-13-1286-1_12] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The current epidemic of obesity and its associated metabolic syndromes imposes unprecedented challenges to our society. Despite intensive research focus on obesity pathogenesis, an effective therapeutic strategy to treat and cure obesity is still lacking. The obesity development is due to a disturbed homeostatic control of feeding and energy expenditure, both of which are controlled by an intricate neural network in the brain. Given the inherent complexity of brain networks in controlling feeding and energy expenditure, the understanding of brain-based pathophysiology for obesity development is limited. One key limiting factor in dissecting neural pathways for feeding and energy expenditure is unavailability of techniques that can be used to effectively reduce the complexity of the brain network to a tractable paradigm, based on which a strong hypothesis can be tested. Excitingly, emerging techniques have been involved to be able to link specific groups of neurons and neural pathways to behaviors (i.e., feeding and energy expenditure). In this chapter, novel techniques especially those based on animal models and viral vector approaches will be discussed. We hope that this chapter will provide readers with a basis that can help to understand the literatures using these techniques and with a guide to apply these exciting techniques to investigate brain mechanisms underlying feeding and energy expenditure.
Collapse
Affiliation(s)
- Qi Wu
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA. .,Children's Nutrition Research Center, Research Service of Department of Agriculture of USA, Houston, TX, USA.
| | - Yong Han
- Department of Pediatrics, Baylor College of Medicine, USDA-ARS, Houston, TX, USA
| | - Qingchun Tong
- Center for Metabolic and Degenerative Diseases, Brown Foundation Institute of Molecular Medicine, University of Texas McGovern Medical School, Houston, TX, USA.
| |
Collapse
|
60
|
Cheng ZF, Pai RK, Cartwright CA. Rack1 function in intestinal epithelia: regulating crypt cell proliferation and regeneration and promoting differentiation and apoptosis. Am J Physiol Gastrointest Liver Physiol 2018; 314:G1-G13. [PMID: 28935684 PMCID: PMC5866376 DOI: 10.1152/ajpgi.00240.2017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 09/07/2017] [Accepted: 09/11/2017] [Indexed: 01/31/2023]
Abstract
Previously, we showed that receptor for activated C kinase 1 (Rack1) regulates growth of colon cells in vitro, partly by suppressing Src kinase activity at key cell cycle checkpoints, in apoptotic and cell survival pathways and at cell-cell adhesions. Here, we generated mouse models of Rack1 deficiency to assess Rack1's function in intestinal epithelia in vivo. Intestinal Rack1 deficiency resulted in proliferation of crypt cells, diminished differentiation of crypt cells into enterocyte, goblet, and enteroendocrine cell lineages, and expansion of Paneth cell populations. Following radiation injury, the morphology of Rack1-deleted small bowel was strikingly abnormal with development of large polypoid structures that contained many partly formed villi, numerous back-to-back elongated and regenerating crypts, and high-grade dysplasia in surface epithelia. These abnormalities were not observed in Rack1-expressing areas of intestine or in control mice. Following irradiation, apoptosis of enterocytes was strikingly reduced in Rack1-deleted epithelia. These novel findings reveal key functions for Rack1 in regulating growth of intestinal epithelia: suppressing crypt cell proliferation and regeneration, promoting differentiation and apoptosis, and repressing development of neoplasia. NEW & NOTEWORTHY Our findings reveal novel functions for receptor for activated C kinase 1 (Rack1) in regulating growth of intestinal epithelia: suppressing crypt cell proliferation and regeneration, promoting differentiation and apoptosis, and repressing development of neoplasia.
Collapse
Affiliation(s)
- Zhuan-Fen Cheng
- Department of Medicine, Stanford University , Stanford, California
| | - Reetesh K Pai
- Department of Pathology, University of Pittsburgh , Pittsburgh, Pennsylvania
| | | |
Collapse
|
61
|
The Impact of CRISPR/Cas9 Technology on Cardiac Research: From Disease Modelling to Therapeutic Approaches. Stem Cells Int 2017; 2017:8960236. [PMID: 29434642 PMCID: PMC5757142 DOI: 10.1155/2017/8960236] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 11/16/2017] [Indexed: 12/19/2022] Open
Abstract
Genome-editing technology has emerged as a powerful method that enables the generation of genetically modified cells and organisms necessary to elucidate gene function and mechanisms of human diseases. The clustered regularly interspaced short palindromic repeats- (CRISPR-) associated 9 (Cas9) system has rapidly become one of the most popular approaches for genome editing in basic biomedical research over recent years because of its simplicity and adaptability. CRISPR/Cas9 genome editing has been used to correct DNA mutations ranging from a single base pair to large deletions in both in vitro and in vivo model systems. CRISPR/Cas9 has been used to increase the understanding of many aspects of cardiovascular disorders, including lipid metabolism, electrophysiology and genetic inheritance. The CRISPR/Cas9 technology has been proven to be effective in creating gene knockout (KO) or knockin in human cells and is particularly useful for editing induced pluripotent stem cells (iPSCs). Despite these progresses, some biological, technical, and ethical issues are limiting the therapeutic potential of genome editing in cardiovascular diseases. This review will focus on various applications of CRISPR/Cas9 genome editing in the cardiovascular field, for both disease research and the prospect of in vivo genome-editing therapies in the future.
Collapse
|
62
|
Márkus B, Pató Z, Sarang Z, Albert R, Tőzsér J, Petrovski G, Csősz É. The proteomic profile of a mouse model of proliferative vitreoretinopathy. FEBS Open Bio 2017; 7:1166-1177. [PMID: 28781956 PMCID: PMC5537063 DOI: 10.1002/2211-5463.12252] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 05/02/2017] [Accepted: 05/27/2017] [Indexed: 11/24/2022] Open
Abstract
Proliferative vitreoretinopathy (PVR) develops as a complication of retinal detachment surgery and represents a devastating condition leading to serious vision loss. A good animal model that permits extensive functional studies and drug testing is crucial in finding better therapeutic modalities for PVR. A previously established mouse model, using dispase injection, was analyzed from the proteomic point of view, examining global protein profile changes by 2D electrophoresis, image analysis and HPLC–tandem mass spectrometry‐based protein identification. The easy applicability of the mouse model was used to study the role of transglutaminase 2 (TG2) in PVR formation by proteomic examination of dispase‐induced TG2 knockout vitreous samples. Our data demonstrate that, despite the altered appearance of crystallin proteins, the lack of TG2 did not prevent the development of PVR.
Collapse
Affiliation(s)
- Bernadett Márkus
- Department of Biochemistry and Molecular Biology Faculty of Medicine University of Debrecen Hungary
| | - Zsuzsanna Pató
- Department of Biochemistry and Molecular Biology Faculty of Medicine University of Debrecen Hungary
| | - Zsolt Sarang
- Department of Biochemistry and Molecular Biology Faculty of Medicine University of Debrecen Hungary
| | - Réka Albert
- Department of Ophthalmology Faculty of Medicine University of Szeged Hungary
| | - József Tőzsér
- Department of Biochemistry and Molecular Biology Faculty of Medicine University of Debrecen Hungary
| | - Goran Petrovski
- Department of Ophthalmology Faculty of Medicine University of Szeged Hungary.,Department of Ophthalmology Oslo University Hospital and University of Oslo Norway
| | - Éva Csősz
- Department of Biochemistry and Molecular Biology Faculty of Medicine University of Debrecen Hungary
| |
Collapse
|
63
|
Wefers B, Bashir S, Rossius J, Wurst W, Kühn R. Gene editing in mouse zygotes using the CRISPR/Cas9 system. Methods 2017; 121-122:55-67. [PMID: 28263886 DOI: 10.1016/j.ymeth.2017.02.008] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2016] [Revised: 02/09/2017] [Accepted: 02/27/2017] [Indexed: 12/26/2022] Open
Abstract
The generation of targeted mouse mutants is a key technology for biomedical research. Using the CRISPR/Cas9 system for induction of targeted double-strand breaks, gene editing can be performed in a single step directly in mouse zygotes. This article covers the design of knockout and knockin alleles, preparation of reagents, microinjection or electroporation of zygotes and the genotyping of pups derived from gene editing projects. In addition we include a section for the control of experimental settings by targeting the Rosa26 locus and PCR based genotyping of blastocysts.
Collapse
Affiliation(s)
- Benedikt Wefers
- German Center for Neurodegenerative Diseases (DZNE), Feodor-Lynen Str. 17, 81377 Munich, Germany; Helmholtz Zentrum München, German Research Center for Environmental Health, Institute of Developmental Genetics, Ingolstädter Landstr. 1, 85764 Neuherberg, Germany.
| | - Sanum Bashir
- Max-Delbrück-Centrum für Molekulare Medizin, Robert-Rössle Str. 10, 13125 Berlin, Germany; Berlin Institute of Health, Kapelle-Ufer 2, 10117 Berlin, Germany.
| | - Jana Rossius
- Max-Delbrück-Centrum für Molekulare Medizin, Robert-Rössle Str. 10, 13125 Berlin, Germany.
| | - Wolfgang Wurst
- German Center for Neurodegenerative Diseases (DZNE), Feodor-Lynen Str. 17, 81377 Munich, Germany; Helmholtz Zentrum München, German Research Center for Environmental Health, Institute of Developmental Genetics, Ingolstädter Landstr. 1, 85764 Neuherberg, Germany; Technische Universität München-Weihenstephan, Chair of Developmental Genetics, c/o Helmholtz Zentrum München, Ingolstädter Landstr. 1, 85764 Neuherberg, Germany; Munich Cluster for Systems Neurology (SyNergy), Feodor-Lynen-Str. 17, 81377 Munich, Germany.
| | - Ralf Kühn
- Max-Delbrück-Centrum für Molekulare Medizin, Robert-Rössle Str. 10, 13125 Berlin, Germany; Berlin Institute of Health, Kapelle-Ufer 2, 10117 Berlin, Germany.
| |
Collapse
|
64
|
Miller MR, Miller EW, Blystone SD. Non-canonical activity of the podosomal formin FMNL1γ supports immune cell migration. J Cell Sci 2017; 130:1730-1739. [PMID: 28348104 DOI: 10.1242/jcs.195099] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 03/16/2017] [Indexed: 12/14/2022] Open
Abstract
Having previously located the formin FMNL1 in macrophage podosomes, we developed an in vivo model to assess the role of FMNL1 in the migration activities of primary macrophages. Deletion of FMNL1 in mice was genetically lethal; however, targeted deletion in macrophages was achieved by employing macrophage-specific Cre. Unchallenged FMNL1-deficient mice exhibited an unexpected reduction in tissue-resident macrophages despite normal blood monocyte numbers. Upon immune stimulus, the absence of FMNL1 resulted in reduced macrophage recruitment in vivo, decreased migration in two-dimensional in vitro culture and a decrease in the number of macrophages exhibiting podosomes. Of the three described isoforms of FMNL1 - α, β and γ - only FMNL1γ rescued macrophage migration when expressed exogenously in depleted macrophages. Surprisingly, mutation of residues in the FH2 domain of FMNL1γ that disrupt barbed-end actin binding did not limit rescue of macrophage migration and podosome numbers. These observations suggest that FMNL1 contributes to macrophage migration activity by stabilizing the lifespan of podosomes without interaction of fast-growing actin termini.
Collapse
Affiliation(s)
- Matthew R Miller
- Department of Cell and Developmental Biology, SUNY Upstate Medical University, 750 East Adams St., Syracuse, NY 13210, USA
| | - Eric W Miller
- Department of Cell and Developmental Biology, SUNY Upstate Medical University, 750 East Adams St., Syracuse, NY 13210, USA
| | - Scott D Blystone
- Department of Cell and Developmental Biology, SUNY Upstate Medical University, 750 East Adams St., Syracuse, NY 13210, USA
| |
Collapse
|
65
|
McLellan MA, Rosenthal NA, Pinto AR. Cre-loxP-Mediated Recombination: General Principles and Experimental Considerations. ACTA ACUST UNITED AC 2017; 7:1-12. [DOI: 10.1002/cpmo.22] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Micheal A. McLellan
- The Jackson Laboratory; Bar Harbor Maine
- Graduate Program in Genetics, Sackler School of Graduate Biomedical Sciences, Tufts University; Boston Massachusetts
| | - Nadia A. Rosenthal
- The Jackson Laboratory; Bar Harbor Maine
- Australian Regenerative Medicine Institute, Monash University; Melbourne Victoria Australia
- National Heart and Lung Institute, Imperial College London; London United Kingdom
| | - Alexander R. Pinto
- The Jackson Laboratory; Bar Harbor Maine
- Australian Regenerative Medicine Institute, Monash University; Melbourne Victoria Australia
| |
Collapse
|
66
|
May I Cut in? Gene Editing Approaches in Human Induced Pluripotent Stem Cells. Cells 2017; 6:cells6010005. [PMID: 28178187 PMCID: PMC5371870 DOI: 10.3390/cells6010005] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2016] [Revised: 01/20/2017] [Accepted: 01/30/2017] [Indexed: 12/16/2022] Open
Abstract
In the decade since Yamanaka and colleagues described methods to reprogram somatic cells into a pluripotent state, human induced pluripotent stem cells (hiPSCs) have demonstrated tremendous promise in numerous disease modeling, drug discovery, and regenerative medicine applications. More recently, the development and refinement of advanced gene transduction and editing technologies have further accelerated the potential of hiPSCs. In this review, we discuss the various gene editing technologies that are being implemented with hiPSCs. Specifically, we describe the emergence of technologies including zinc-finger nuclease (ZFN), transcription activator-like effector nuclease (TALEN), and clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 that can be used to edit the genome at precise locations, and discuss the strengths and weaknesses of each of these technologies. In addition, we present the current applications of these technologies in elucidating the mechanisms of human development and disease, developing novel and effective therapeutic molecules, and engineering cell-based therapies. Finally, we discuss the emerging technological advances in targeted gene editing methods.
Collapse
|
67
|
Albers E, Sbroggiò M, Martin-Gonzalez J, Avram A, Munk S, Lopez-Contreras AJ. A simple DNA recombination screening method by RT-PCR as an alternative to Southern blot. Transgenic Res 2017; 26:429-434. [PMID: 28105543 DOI: 10.1007/s11248-017-0008-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 01/04/2017] [Indexed: 11/30/2022]
Abstract
The generation of genetically engineered mouse models (GEMMs), including knock-out (KO) and knock-in (KI) models, often requires genomic screening of many mouse ES cell (mESC) clones by Southern blot. The use of large targeting constructs facilitates the recombination of exogenous DNA in a specific genomic locus, but limits the detection of its correct genomic integration by standard PCR methods. Genomic Long Range PCR (LR-PCR), using primers adjacent to the homology arms, has been used as an alternative to radioactive-based Southern blot screenings. However, LR-PCRs are often difficult and render many false positive and false negative results. Here, we propose an alternative screening method based on the detection of a genetic modification at the mRNA level, which we successfully optimized in two mouse models. This screening method consists of a reverse-transcription PCR (RT-PCR) using primers that match exons flanking the targeting construct. The detection of the expected modification in this PCR product confirms the integration at the correct genomic location and shows that the mutant mRNA is expressed. This is a simple and sensitive strategy to screen locus-specific recombination of targeting constructs which can also be useful to screen KO and KI mutant mice or cell lines including those generated by CRISPR/Cas9.
Collapse
Affiliation(s)
- Eliene Albers
- Department of Cellular and Molecular Medicine, Center for Chromosome Stability and Center for Healthy Aging, Panum Institute, University of Copenhagen, Copenhagen, Denmark
| | - Mauro Sbroggiò
- Department of Cellular and Molecular Medicine, Center for Chromosome Stability and Center for Healthy Aging, Panum Institute, University of Copenhagen, Copenhagen, Denmark
| | - Javier Martin-Gonzalez
- Transgenic Core Facility, Department of Experimental Medicine, Panum Institute, University of Copenhagen, Copenhagen, Denmark
| | - Alexandra Avram
- Department of Cellular and Molecular Medicine, Center for Chromosome Stability and Center for Healthy Aging, Panum Institute, University of Copenhagen, Copenhagen, Denmark
| | - Stephanie Munk
- Department of Cellular and Molecular Medicine, Center for Chromosome Stability and Center for Healthy Aging, Panum Institute, University of Copenhagen, Copenhagen, Denmark
| | - Andres J Lopez-Contreras
- Department of Cellular and Molecular Medicine, Center for Chromosome Stability and Center for Healthy Aging, Panum Institute, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
68
|
Abstract
The generation of a new genetically modified mouse strain is a big hurdle to take for many researchers. It is often unclear which steps and decisions have to be made prior to obtaining the desired mouse model. This review aims to help researchers by providing a decision guide that answers the essential questions that need to be asked before generating the most suitable genetically modified mouse line in the most optimal timeframe. The review includes the latest technologies in both the stem cell culture and gene editing tools, particularly CRISPR/Cas9, and provides compatibility guidelines for selecting among the different types of genetic modifications that can be introduced in the mouse genome and the various routes for introducing these modifications into the mouse germline.
Collapse
Affiliation(s)
- Ivo J Huijbers
- Mouse Clinic for Cancer and Aging, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands.
| |
Collapse
|
69
|
Adam S, Melguizo Sanchis D, El-Kamah G, Samarasinghe S, Alharthi S, Armstrong L, Lako M. Concise Review: Getting to the Core of Inherited Bone Marrow Failures. Stem Cells 2016; 35:284-298. [PMID: 27870251 PMCID: PMC5299470 DOI: 10.1002/stem.2543] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Revised: 10/15/2016] [Accepted: 10/28/2016] [Indexed: 12/20/2022]
Abstract
Bone marrow failure syndromes (BMFS) are a group of disorders with complex pathophysiology characterized by a common phenotype of peripheral cytopenia and/or hypoplastic bone marrow. Understanding genetic factors contributing to the pathophysiology of BMFS has enabled the identification of causative genes and development of diagnostic tests. To date more than 40 mutations in genes involved in maintenance of genomic stability, DNA repair, ribosome and telomere biology have been identified. In addition, pathophysiological studies have provided insights into several biological pathways leading to the characterization of genotype/phenotype correlations as well as the development of diagnostic approaches and management strategies. Recent developments in bone marrow transplant techniques and the choice of conditioning regimens have helped improve transplant outcomes. However, current morbidity and mortality remain unacceptable underlining the need for further research in this area. Studies in mice have largely been unable to mimic disease phenotype in humans due to difficulties in fully replicating the human mutations and the differences between mouse and human cells with regard to telomere length regulation, processing of reactive oxygen species and lifespan. Recent advances in induced pluripotency have provided novel insights into disease pathogenesis and have generated excellent platforms for identifying signaling pathways and functional mapping of haplo‐insufficient genes involved in large‐scale chromosomal deletions–associated disorders. In this review, we have summarized the current state of knowledge in the field of BMFS with specific focus on modeling the inherited forms and how to best utilize these models for the development of targeted therapies. Stem Cells2017;35:284–298
Collapse
Affiliation(s)
- Soheir Adam
- Department of Medicine, Duke University Medical Center, Durham, North Carolina, USA.,Hematology Department, Medical School, King Abdulaziz University, Jeddah, KSA
| | | | - Ghada El-Kamah
- Division of Human Genetics & Genome Research, National Research Center, Cairo, Egypt
| | - Sujith Samarasinghe
- Department of Hematology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom
| | - Sameer Alharthi
- Princess Al Jawhara Al-Brahim Center of Excellence in Research of Hereditary Disorders, King Abdulaziz University, KSA
| | - Lyle Armstrong
- Institute of Genetic Medicine, Newcastle University, United Kingdom
| | - Majlinda Lako
- Institute of Genetic Medicine, Newcastle University, United Kingdom
| |
Collapse
|
70
|
Zuberi A, Lutz C. Mouse Models for Drug Discovery. Can New Tools and Technology Improve Translational Power? ILAR J 2016; 57:178-185. [PMID: 28053071 PMCID: PMC5886322 DOI: 10.1093/ilar/ilw021] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 08/05/2016] [Accepted: 08/12/2016] [Indexed: 12/24/2022] Open
Abstract
The use of mouse models in biomedical research and preclinical drug evaluation is on the rise. The advent of new molecular genome-altering technologies such as CRISPR/Cas9 allows for genetic mutations to be introduced into the germ line of a mouse faster and less expensively than previous methods. In addition, the rapid progress in the development and use of somatic transgenesis using viral vectors, as well as manipulations of gene expression with siRNAs and antisense oligonucleotides, allow for even greater exploration into genomics and systems biology. These technological advances come at a time when cost reductions in genome sequencing have led to the identification of pathogenic mutations in patient populations, providing unprecedented opportunities in the use of mice to model human disease. The ease of genetic engineering in mice also offers a potential paradigm shift in resource sharing and the speed by which models are made available in the public domain. Predictively, the knowledge alone that a model can be quickly remade will provide relief to resources encumbered by licensing and Material Transfer Agreements. For decades, mouse strains have provided an exquisite experimental tool to study the pathophysiology of the disease and assess therapeutic options in a genetically defined system. However, a major limitation of the mouse has been the limited genetic diversity associated with common laboratory mice. This has been overcome with the recent development of the Collaborative Cross and Diversity Outbred mice. These strains provide new tools capable of replicating genetic diversity to that approaching the diversity found in human populations. The Collaborative Cross and Diversity Outbred strains thus provide a means to observe and characterize toxicity or efficacy of new therapeutic drugs for a given population. The combination of traditional and contemporary mouse genome editing tools, along with the addition of genetic diversity in new modeling systems, are synergistic and serve to make the mouse a better model for biomedical research, enhancing the potential for preclinical drug discovery and personalized medicine.
Collapse
Affiliation(s)
- Aamir Zuberi
- Cathleen Lutz holds a PhD in biochemistry and an MBA and is the director of the Mouse Repository at The Jackson Laboratory as well as the director of the Rare and Orphan Disease Center and the lead for the in vivo pharmacology program at The Jackson Laboratory in Bar Harbor, Maine. Aamir Zuberi holds a PhD in molecular genetics and is a research associate in the laboratory of Dr. Lutz at the Jackson Laboratory, Bar Harbor, Maine
| | - Cathleen Lutz
- Cathleen Lutz holds a PhD in biochemistry and an MBA and is the director of the Mouse Repository at The Jackson Laboratory as well as the director of the Rare and Orphan Disease Center and the lead for the in vivo pharmacology program at The Jackson Laboratory in Bar Harbor, Maine. Aamir Zuberi holds a PhD in molecular genetics and is a research associate in the laboratory of Dr. Lutz at the Jackson Laboratory, Bar Harbor, Maine
| |
Collapse
|
71
|
Ferrara G, Errede M, Girolamo F, Morando S, Ivaldi F, Panini N, Bendotti C, Perris R, Furlan R, Virgintino D, Kerlero de Rosbo N, Uccelli A. NG2, a common denominator for neuroinflammation, blood-brain barrier alteration, and oligodendrocyte precursor response in EAE, plays a role in dendritic cell activation. Acta Neuropathol 2016; 132:23-42. [PMID: 27026411 PMCID: PMC4911384 DOI: 10.1007/s00401-016-1563-z] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Revised: 03/15/2016] [Accepted: 03/15/2016] [Indexed: 02/07/2023]
Abstract
In adult CNS, nerve/glial-antigen 2 (NG2) is expressed by oligodendrocyte progenitor cells (OPCs) and is an early marker of pericyte activation in pathological conditions. NG2 could, therefore, play a role in experimental autoimmune encephalomyelitis (EAE), a disease associated with increased blood–brain barrier (BBB) permeability, inflammatory infiltrates, and CNS damage. We induced EAE in NG2 knock-out (NG2KO) mice and used laser confocal microscopy immunofluorescence and morphometry to dissect the effect of NG2 KO on CNS pathology. NG2KO mice developed milder EAE than their wild-type (WT) counterparts, with less intense neuropathology associated with a significant improvement in BBB stability. In contrast to WT mice, OPC numbers did not change in NG2KO mice during EAE. Through FACS and confocal microscopy, we found that NG2 was also expressed by immune cells, including T cells, macrophages, and dendritic cells (DCs). Assessment of recall T cell responses to the encephalitogen by proliferation assays and ELISA showed that, while WT and NG2KO T cells proliferated equally to the encephalitogenic peptide MOG35-55, NG2KO T cells were skewed towards a Th2-type response. Because DCs could be responsible for this effect, we assessed their expression of IL-12 by PCR and intracellular FACS. IL-12-expressing CD11c+ cells were significantly decreased in MOG35-55-primed NG2KO lymph node cells. Importantly, in WT mice, the proportion of IL-12-expressing cells was significantly lower in CD11c+ NG2- cells than in CD11c+ NG2+ cells. To assess the relevance of NG2 at immune system and CNS levels, we induced EAE in bone-marrow chimeric mice, generated with WT recipients of NG2KO bone-marrow cells and vice versa. Regardless of their original phenotype, mice receiving NG2KO bone marrow developed milder EAE than those receiving WT bone marrow. Our data suggest that NG2 plays a role in EAE not only at CNS/BBB level, but also at immune response level, impacting on DC activation and thereby their stimulation of reactive T cells, through controlling IL-12 expression.
Collapse
|
72
|
Lokken AA, Ralston A. The Genetic Regulation of Cell Fate During Preimplantation Mouse Development. Curr Top Dev Biol 2016; 120:173-202. [PMID: 27475852 DOI: 10.1016/bs.ctdb.2016.04.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The adult body is estimated to contain several hundred distinct cell types, each with a specialized physiological function. Failure to maintain cell fate can lead to devastating diseases and cancer, but understanding how cell fates are assigned and maintained during animal development provides new opportunities for human health intervention. The mouse is a premier model for evaluating the genetic regulation of cell fate during development because of the wide variety of tools for measuring and manipulating gene expression levels, the ability to access embryos at desired developmental stages, and the similarities between mouse and human development, particularly during the early stages of development. During the first 3 days of mouse development, the preimplantation embryo sets aside cells that will contribute to the extraembryonic tissues. The extraembryonic tissues are essential for establishing pregnancy and ensuring normal fetal development in both mice and humans. Genetic analyses of mouse preimplantation development have permitted identification of genes that are essential for specification of the extraembryonic lineages. In this chapter, we review the tools and concepts of mouse preimplantation development. We describe genes that are essential for cell fate specification during preimplantation stages, and we describe diverse models proposed to account for the mechanisms of cell fate specification during early development.
Collapse
Affiliation(s)
- A A Lokken
- Michigan State University, East Lansing, MI, United States
| | - A Ralston
- Michigan State University, East Lansing, MI, United States.
| |
Collapse
|
73
|
|
74
|
Lines KE, Stevenson M, Thakker RV. Animal models of pituitary neoplasia. Mol Cell Endocrinol 2016; 421:68-81. [PMID: 26320859 PMCID: PMC4721536 DOI: 10.1016/j.mce.2015.08.024] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 08/25/2015] [Accepted: 08/25/2015] [Indexed: 01/21/2023]
Abstract
Pituitary neoplasias can occur as part of a complex inherited disorder, or more commonly as sporadic (non-familial) disease. Studies of the molecular and genetic mechanisms causing such pituitary tumours have identified dysregulation of >35 genes, with many revealed by studies in mice, rats and zebrafish. Strategies used to generate these animal models have included gene knockout, gene knockin and transgenic over-expression, as well as chemical mutagenesis and drug induction. These animal models provide an important resource for investigation of tissue-specific tumourigenic mechanisms, and evaluations of novel therapies, illustrated by studies into multiple endocrine neoplasia type 1 (MEN1), a hereditary syndrome in which ∼ 30% of patients develop pituitary adenomas. This review describes animal models of pituitary neoplasia that have been generated, together with some recent advances in gene editing technologies, and an illustration of the use of the Men1 mouse as a pre clinical model for evaluating novel therapies.
Collapse
Affiliation(s)
- K E Lines
- Academic Endocrine Unit, Radcliffe Department of Medicine, University of Oxford, Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), Churchill Hospital, Headington, Oxford OX3 7LJ, UK
| | - M Stevenson
- Academic Endocrine Unit, Radcliffe Department of Medicine, University of Oxford, Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), Churchill Hospital, Headington, Oxford OX3 7LJ, UK
| | - R V Thakker
- Academic Endocrine Unit, Radcliffe Department of Medicine, University of Oxford, Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), Churchill Hospital, Headington, Oxford OX3 7LJ, UK.
| |
Collapse
|
75
|
Specks J, Nieto-Soler M, Lopez-Contreras AJ, Fernandez-Capetillo O. Modeling the study of DNA damage responses in mice. Methods Mol Biol 2015; 1267:413-37. [PMID: 25636482 DOI: 10.1007/978-1-4939-2297-0_21] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Damaged DNA has a profound impact on mammalian health and overall survival. In addition to being the source of mutations that initiate cancer, the accumulation of toxic amounts of DNA damage can cause severe developmental diseases and accelerate aging. Therefore, understanding how cells respond to DNA damage has become one of the most intense areas of biomedical research in the recent years. However, whereas most mechanistic studies derive from in vitro or in cellulo work, the impact of a given mutation on a living organism is largely unpredictable. For instance, why BRCA1 mutations preferentially lead to breast cancer whereas mutations compromising mismatch repair drive colon cancer is still not understood. In this context, evaluating the specific physiological impact of mutations that compromise genome integrity has become crucial for a better dimensioning of our knowledge. We here describe the various technologies that can be used for modeling mutations in mice and provide a review of the genes and pathways that have been modeled so far in the context of DNA damage responses.
Collapse
Affiliation(s)
- Julia Specks
- Genomic Instability Group, Spanish National Cancer Research Center (CNIO), C/Melchor Fernandez Almagro, 3, E-28029, Madrid, Spain
| | | | | | | |
Collapse
|
76
|
Guna A, Butcher NJ, Bassett AS. Comparative mapping of the 22q11.2 deletion region and the potential of simple model organisms. J Neurodev Disord 2015; 7:18. [PMID: 26137170 PMCID: PMC4487986 DOI: 10.1186/s11689-015-9113-x] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Accepted: 05/26/2015] [Indexed: 01/18/2023] Open
Abstract
Background 22q11.2 deletion syndrome (22q11.2DS) is the most common micro-deletion syndrome. The associated 22q11.2 deletion conveys the strongest known molecular risk for schizophrenia. Neurodevelopmental phenotypes, including intellectual disability, are also prominent though variable in severity. Other developmental features include congenital cardiac and craniofacial anomalies. Whereas existing mouse models have been helpful in determining the role of some genes overlapped by the hemizygous 22q11.2 deletion in phenotypic expression, much remains unknown. Simple model organisms remain largely unexploited in exploring these genotype-phenotype relationships. Methods We first developed a comprehensive map of the human 22q11.2 deletion region, delineating gene content, and brain expression. To identify putative orthologs, standard methods were used to interrogate the proteomes of the zebrafish (D. rerio), fruit fly (D. melanogaster), and worm (C. elegans), in addition to the mouse. Spatial locations of conserved homologues were mapped to examine syntenic relationships. We systematically cataloged available knockout and knockdown models of all conserved genes across these organisms, including a comprehensive review of associated phenotypes. Results There are 90 genes overlapped by the typical 2.5 Mb deletion 22q11.2 region. Of the 46 protein-coding genes, 41 (89.1 %) have documented expression in the human brain. Identified homologues in the zebrafish (n = 37, 80.4 %) were comparable to those in the mouse (n = 40, 86.9 %) and included some conserved gene cluster structures. There were 22 (47.8 %) putative homologues in the fruit fly and 17 (37.0 %) in the worm involving multiple chromosomes. Individual gene knockdown mutants were available for the simple model organisms, but not for mouse. Although phenotypic data were relatively limited for knockout and knockdown models of the 17 genes conserved across all species, there was some evidence for roles in neurodevelopmental phenotypes, including four of the six mitochondrial genes in the 22q11.2 deletion region. Conclusions Simple model organisms represent a powerful but underutilized means of investigating the molecular mechanisms underlying the elevated risk for neurodevelopmental disorders in 22q11.2DS. This comparative multi-species study provides novel resources and support for the potential utility of non-mouse models in expression studies and high-throughput drug screening. The approach has implications for other recurrent copy number variations associated with neurodevelopmental phenotypes. Electronic supplementary material The online version of this article (doi:10.1186/s11689-015-9113-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Alina Guna
- Clinical Genetics Research Program and Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON Canada
| | - Nancy J Butcher
- Clinical Genetics Research Program and Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON Canada ; Institute of Medical Science, University of Toronto, Toronto, ON Canada
| | - Anne S Bassett
- Clinical Genetics Research Program and Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON Canada ; Institute of Medical Science, University of Toronto, Toronto, ON Canada ; Dalglish Family Hearts and Minds Clinic for Adults with 22q11.2 Deletion Syndrome, Division of Cardiology, Department of Medicine, Department of Psychiatry, and Toronto General Research Institute, University Health Network, Toronto, ON Canada ; Department of Psychiatry, University of Toronto, Toronto, ON Canada ; Centre for Addiction and Mental Health, 33 Russell Street, Room 1100, M5S 2S1 Toronto, ON Canada
| |
Collapse
|
77
|
Parikh BA, Beckman DL, Patel SJ, White JM, Yokoyama WM. Detailed phenotypic and molecular analyses of genetically modified mice generated by CRISPR-Cas9-mediated editing. PLoS One 2015; 10:e0116484. [PMID: 25587897 PMCID: PMC4294663 DOI: 10.1371/journal.pone.0116484] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Accepted: 12/08/2014] [Indexed: 11/20/2022] Open
Abstract
The bacterial CRISPR-Cas9 system has been adapted for use as a genome editing tool. While several recent reports have indicated that successful genome editing of mice can be achieved, detailed phenotypic and molecular analyses of the mutant animals are limited. Following pronuclear micro-injection of fertilized eggs with either wild-type Cas9 or the nickase mutant (D10A) and single or paired guide RNA (sgRNA) for targeting of the tyrosinase (Tyr) gene, we assessed genome editing in mice using rapid phenotypic readouts (eye and coat color). Mutant mice with insertions or deletions (indels) in Tyr were efficiently generated without detectable off-target cleavage events. Gene correction of a single nucleotide by homologous recombination (HR) could only occur when the sgRNA recognition sites in the donor DNA were modified. Gene repair did not occur if the donor DNA was not modified because Cas9 catalytic activity was completely inhibited. Our results indicate that allelic mosaicism can occur following -Cas9-mediated editing in mice and appears to correlate with sgRNA cleavage efficiency at the single-cell stage. We also show that larger than expected deletions may be overlooked based on the screening strategy employed. An unbiased analysis of all the deleted nucleotides in our experiments revealed that the highest frequencies of nucleotide deletions were clustered around the predicted Cas9 cleavage sites, with slightly broader distributions than expected. Finally, additional analysis of founder mice and their offspring indicate that their general health, fertility, and the transmission of genetic changes were not compromised. These results provide the foundation to interpret and predict the diverse outcomes following CRISPR-Cas9-mediated genome editing experiments in mice.
Collapse
Affiliation(s)
- Bijal A. Parikh
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, 63110, United States of America
| | - Diana L. Beckman
- Division of Rheumatology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, United States of America
| | - Swapneel J. Patel
- Division of Rheumatology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, United States of America
| | - J. Michael White
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, 63110, United States of America
| | - Wayne M. Yokoyama
- Division of Rheumatology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, United States of America
- Howard Hughes Medical Institute, Washington University School of Medicine, St. Louis, MO, 63110, United States of America
| |
Collapse
|
78
|
Harms DW, Quadros RM, Seruggia D, Ohtsuka M, Takahashi G, Montoliu L, Gurumurthy CB. Mouse Genome Editing Using the CRISPR/Cas System. CURRENT PROTOCOLS IN HUMAN GENETICS 2014; 83:15.7.1-27. [PMID: 25271839 PMCID: PMC4519007 DOI: 10.1002/0471142905.hg1507s83] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The availability of techniques to create desired genetic mutations has enabled the laboratory mouse as an extensively used model organism in biomedical research including human genetics. A new addition to this existing technical repertoire is the CRISPR/Cas system. Specifically, this system allows editing of the mouse genome much more quickly than the previously used techniques, and, more importantly, multiple mutations can be created in a single experiment. Here we provide protocols for preparation of CRISPR/Cas reagents and microinjection into one-cell mouse embryos to create knockout or knock-in mouse models.
Collapse
Affiliation(s)
- Donald W Harms
- Mouse Genome Engineering Core Facility, Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, Nebraska; These authors contributed equally to this work
| | | | | | | | | | | | | |
Collapse
|
79
|
Scott JNF, Kupinski AP, Boyes J. Targeted genome regulation and modification using transcription activator-like effectors. FEBS J 2014; 281:4583-97. [DOI: 10.1111/febs.12973] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Revised: 08/07/2014] [Accepted: 08/13/2014] [Indexed: 11/30/2022]
Affiliation(s)
- James N. F. Scott
- School of Molecular and Cellular Biology; Faculty of Biological Sciences; University of Leeds; UK
| | - Adam P. Kupinski
- School of Molecular and Cellular Biology; Faculty of Biological Sciences; University of Leeds; UK
| | - Joan Boyes
- School of Molecular and Cellular Biology; Faculty of Biological Sciences; University of Leeds; UK
| |
Collapse
|
80
|
Investigation of inflammation and tissue patterning in the gut using a Spatially Explicit General-purpose Model of Enteric Tissue (SEGMEnT). PLoS Comput Biol 2014; 10:e1003507. [PMID: 24675765 PMCID: PMC3967920 DOI: 10.1371/journal.pcbi.1003507] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Accepted: 01/10/2014] [Indexed: 01/22/2023] Open
Abstract
The mucosa of the intestinal tract represents a finely tuned system where tissue structure strongly influences, and is turn influenced by, its function as both an absorptive surface and a defensive barrier. Mucosal architecture and histology plays a key role in the diagnosis, characterization and pathophysiology of a host of gastrointestinal diseases. Inflammation is a significant factor in the pathogenesis in many gastrointestinal diseases, and is perhaps the most clinically significant control factor governing the maintenance of the mucosal architecture by morphogenic pathways. We propose that appropriate characterization of the role of inflammation as a controller of enteric mucosal tissue patterning requires understanding the underlying cellular and molecular dynamics that determine the epithelial crypt-villus architecture across a range of conditions from health to disease. Towards this end we have developed the Spatially Explicit General-purpose Model of Enteric Tissue (SEGMEnT) to dynamically represent existing knowledge of the behavior of enteric epithelial tissue as influenced by inflammation with the ability to generate a variety of pathophysiological processes within a common platform and from a common knowledge base. In addition to reproducing healthy ileal mucosal dynamics as well as a series of morphogen knock-out/inhibition experiments, SEGMEnT provides insight into a range of clinically relevant cellular-molecular mechanisms, such as a putative role for Phosphotase and tensin homolog/phosphoinositide 3-kinase (PTEN/PI3K) as a key point of crosstalk between inflammation and morphogenesis, the protective role of enterocyte sloughing in enteric ischemia-reperfusion and chronic low level inflammation as a driver for colonic metaplasia. These results suggest that SEGMEnT can serve as an integrating platform for the study of inflammation in gastrointestinal disease.
Collapse
|
81
|
Fujita H, Aoki H, Ajioka I, Yamazaki M, Abe M, Oh-Nishi A, Sakimura K, Sugihara I. Detailed expression pattern of aldolase C (Aldoc) in the cerebellum, retina and other areas of the CNS studied in Aldoc-Venus knock-in mice. PLoS One 2014; 9:e86679. [PMID: 24475166 PMCID: PMC3903578 DOI: 10.1371/journal.pone.0086679] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Accepted: 12/13/2013] [Indexed: 11/22/2022] Open
Abstract
Aldolase C (Aldoc, also known as "zebrin II"), a brain type isozyme of a glycolysis enzyme, is expressed heterogeneously in subpopulations of cerebellar Purkinje cells (PCs) that are arranged longitudinally in a complex striped pattern in the cerebellar cortex, a pattern which is closely related to the topography of input and output axonal projections. Here, we generated knock-in Aldoc-Venus mice in which Aldoc expression is visualized by expression of a fluorescent protein, Venus. Since there was no obvious phenotypes in general brain morphology and in the striped pattern of the cerebellum in mutants, we made detailed observation of Aldoc expression pattern in the nervous system by using Venus expression in Aldoc-Venus heterozygotes. High levels of Venus expression were observed in cerebellar PCs, cartwheel cells in the dorsal cochlear nucleus, sensory epithelium of the inner ear and in all major types of retinal cells, while moderate levels of Venus expression were observed in astrocytes and satellite cells in the dorsal root ganglion. The striped arrangement of PCs that express Venus to different degrees was carefully traced with serial section alignment analysis and mapped on the unfolded scheme of the entire cerebellar cortex to re-identify all individual Aldoc stripes. A longitudinally striped boundary of Aldoc expression was first identified in the mouse flocculus, and was correlated with the climbing fiber projection pattern and expression of another compartmental marker molecule, heat shock protein 25 (HSP25). As in the rat, the cerebellar nuclei were divided into the rostrodorsal negative and the caudoventral positive portions by distinct projections of Aldoc-positive and negative PC axons in the mouse. Identification of the cerebellar Aldoc stripes in this study, as indicated in sample coronal and horizontal sections as well as in sample surface photos of whole-mount preparations, can be referred to in future experiments.
Collapse
Affiliation(s)
- Hirofumi Fujita
- Department of Systems Neurophysiology, Tokyo Medical and Dental University Graduate School, Bunkyo-ku, Tokyo, Japan
- Systems Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, California, United States of America
- Department of Otolaryngology-Head and Neck Surgery, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Hanako Aoki
- Department of Systems Neurophysiology, Tokyo Medical and Dental University Graduate School, Bunkyo-ku, Tokyo, Japan
| | - Itsuki Ajioka
- Center for Brain Integration Research, Tokyo Medical and Dental University Graduate School, Bunkyo-ku, Tokyo, Japan
| | - Maya Yamazaki
- Department of Cellular Neurobiology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Manabu Abe
- Department of Cellular Neurobiology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Arata Oh-Nishi
- Molecular Neuroimaging Program, Molecular Imaging Center, National Institute of Radiological Sciences, Inage-ku, Chiba, Japan
| | - Kenji Sakimura
- Department of Cellular Neurobiology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Izumi Sugihara
- Department of Systems Neurophysiology, Tokyo Medical and Dental University Graduate School, Bunkyo-ku, Tokyo, Japan
- Center for Brain Integration Research, Tokyo Medical and Dental University Graduate School, Bunkyo-ku, Tokyo, Japan
| |
Collapse
|
82
|
Davila D, Thibault K, Fiacco TA, Agulhon C. Recent molecular approaches to understanding astrocyte function in vivo. Front Cell Neurosci 2013; 7:272. [PMID: 24399932 PMCID: PMC3871966 DOI: 10.3389/fncel.2013.00272] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Accepted: 12/06/2013] [Indexed: 12/11/2022] Open
Abstract
Astrocytes are a predominant glial cell type in the nervous systems, and are becoming recognized as important mediators of normal brain function as well as neurodevelopmental, neurological, and neurodegenerative brain diseases. Although numerous potential mechanisms have been proposed to explain the role of astrocytes in the normal and diseased brain, research into the physiological relevance of these mechanisms in vivo is just beginning. In this review, we will summarize recent developments in innovative and powerful molecular approaches, including knockout mouse models, transgenic mouse models, and astrocyte-targeted gene transfer/expression, which have led to advances in understanding astrocyte biology in vivo that were heretofore inaccessible to experimentation. We will examine the recently improved understanding of the roles of astrocytes – with an emphasis on astrocyte signaling – in the context of both the healthy and diseased brain, discuss areas where the role of astrocytes remains debated, and suggest new research directions.
Collapse
Affiliation(s)
- David Davila
- Glia-Glia and Glia-Neuron Interactions Group, National Center for Scientific Research, UFR Biomedicale, Paris Descartes University Paris, France
| | - Karine Thibault
- Glia-Glia and Glia-Neuron Interactions Group, National Center for Scientific Research, UFR Biomedicale, Paris Descartes University Paris, France
| | - Todd A Fiacco
- Department of Cell Biology and Neuroscience, and Center for Glial-Neuronal Interactions and Program in Cellular, Molecular and Developmental Biology, University of California at Riverside Riverside, CA, USA
| | - Cendra Agulhon
- Glia-Glia and Glia-Neuron Interactions Group, National Center for Scientific Research, UFR Biomedicale, Paris Descartes University Paris, France
| |
Collapse
|
83
|
Yu W, Hill WG. Lack of specificity shown by P2Y6 receptor antibodies. Naunyn Schmiedebergs Arch Pharmacol 2013; 386:885-91. [PMID: 23793102 DOI: 10.1007/s00210-013-0894-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Accepted: 06/04/2013] [Indexed: 01/11/2023]
Abstract
P2Y6 receptor in bladder smooth muscle responds to UDP by increasing muscle tone and augmenting bladder contractions. The exact cellular location of the receptor is however unknown. Three commercially available antibodies to P2Y6 receptor gave clean bands on Western blot which were eliminated by specific peptide competition. Two of the three also immunostained bladder smooth muscle cells while leaving adjacent interstitial cells of Cajal unstained. However, attempts to validate the specificity of these antibodies by performing the same assays on bladders from P2Y6 knockout mice were unsuccessful. In Western blots, all three antibodies bound similar proteins in both wild type and P2Y6 knockout tissue. Immunostaining of knockout tissue sections also showed no difference in staining patterns or intensity. We conclude that rigorous controls are required when using commercial reagents to this G-protein coupled receptor and perhaps to other members of the P2Y receptor family.
Collapse
Affiliation(s)
- Weiqun Yu
- Harvard Medical School, Harvard University, Boston, MA, USA
| | | |
Collapse
|
84
|
YANG JIANYE, ZHU ZHAOHUI, WANG HONGFEI, LI FEIFEI, DU XINLING, MA RUNLINZ. Trop2 regulates the proliferation and differentiation of murine compact-bone derived MSCs. Int J Oncol 2013; 43:859-67. [DOI: 10.3892/ijo.2013.1987] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Accepted: 06/03/2013] [Indexed: 11/05/2022] Open
|
85
|
Li P, Xiao Y, Liu Z, Liu P. Using mouse models to study function of transcriptional factors in T cell development. CELL REGENERATION (LONDON, ENGLAND) 2012; 1:8. [PMID: 25408871 PMCID: PMC4230505 DOI: 10.1186/2045-9769-1-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/08/2012] [Accepted: 10/08/2012] [Indexed: 02/03/2023]
Abstract
Laboratory mice have widely been used as tools for basic biological research and models for studying human diseases. With the advances of genetic engineering and conditional knockout (CKO) mice, we now understand hematopoiesis is a dynamic stepwise process starting from hematopoietic stem cells (HSCs) which are responsible for replenishing all blood cells. Transcriptional factors play important role in hematopoiesis. In this review we compile several studies on using genetic modified mice and humanized mice to study function of transcriptional factors in lymphopoiesis, including T lymphocyte and Natural killer (NK) cell development. Finally, we focused on the key transcriptional factor Bcl11b and its function in regulating T cell specification and commitment.
Collapse
Affiliation(s)
- Peng Li
- Key Laboratory of Regenerative Biology, Guangzchou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China ; Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou, China
| | - Yiren Xiao
- Key Laboratory of Regenerative Biology, Guangzchou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China ; Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou, China
| | - Zhixin Liu
- Key Laboratory of Regenerative Biology, Guangzchou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China ; Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou, China
| | - Pentao Liu
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, CB10 1HH UK
| |
Collapse
|
86
|
Tzfira T, Weinthal D, Marton I, Zeevi V, Zuker A, Vainstein A. Genome modifications in plant cells by custom-made restriction enzymes. PLANT BIOTECHNOLOGY JOURNAL 2012; 10:373-89. [PMID: 22469004 DOI: 10.1111/j.1467-7652.2011.00672.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Genome editing, i.e. the ability to mutagenize, insert, delete and replace sequences, in living cells is a powerful and highly desirable method that could potentially revolutionize plant basic research and applied biotechnology. Indeed, various research groups from academia and industry are in a race to devise methods and develop tools that will enable not only site-specific mutagenesis but also controlled foreign DNA integration and replacement of native and transgene sequences by foreign DNA, in living plant cells. In recent years, much of the progress seen in gene targeting in plant cells has been attributed to the development of zinc finger nucleases and other novel restriction enzymes for use as molecular DNA scissors. The induction of double-strand breaks at specific genomic locations by zinc finger nucleases and other novel restriction enzymes results in a wide variety of genetic changes, which range from gene addition to the replacement, deletion and site-specific mutagenesis of endogenous and heterologous genes in living plant cells. In this review, we discuss the principles and tools for restriction enzyme-mediated gene targeting in plant cells, as well as their current and prospective use for gene targeting in model and crop plants.
Collapse
Affiliation(s)
- Tzvi Tzfira
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel.
| | | | | | | | | | | |
Collapse
|
87
|
Leavitt AD, Hamlett I. Homologous recombination in human embryonic stem cells: a tool for advancing cell therapy and understanding and treating human disease. Clin Transl Sci 2011; 4:298-305. [PMID: 21884519 DOI: 10.1111/j.1752-8062.2011.00281.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Human embryonic stem cells (hESCs) hold great promise for ushering in an era of novel cell therapies to treat a wide range of rare and common diseases, yet they also provide an unprecedented opportunity for basic research to yield clinical benefit. HESCs can be used to better understand human development, to model human diseases, to understand the contribution of specific mutations to the pathogenesis of disease, and to develop human cell-based screening systems to identify novel therapeutic agents and evaluate potential toxicity of therapeutic agents under development. Such basic research will benefit greatly from efficient methods to perform targeted gene modification, an area of hESC investigation that is currently in its infancy. Moreover, the reality of hESC-based cellular therapies will require improved methods for generating the specific cells of interest, and reporter cell lines generated through targeted gene modifications are expected to play an important role in developing optimal cell-specific differentiation protocols. Herein, we review the current status of homologous recombination in hESCs, a gene targeting technique that is sure to continue to improve, and to play an important role in realizing the maximal human benefit from hESCs.
Collapse
Affiliation(s)
- Andrew D Leavitt
- Laboratory Medicine, University of California, San Francisco, California, USA.
| | | |
Collapse
|
88
|
Marton I, Zuker A, Shklarman E, Zeevi V, Tovkach A, Roffe S, Ovadis M, Tzfira T, Vainstein A. Nontransgenic genome modification in plant cells. PLANT PHYSIOLOGY 2010; 154:1079-87. [PMID: 20876340 PMCID: PMC2971589 DOI: 10.1104/pp.110.164806] [Citation(s) in RCA: 100] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2010] [Accepted: 09/23/2010] [Indexed: 05/21/2023]
Abstract
Zinc finger nucleases (ZFNs) are a powerful tool for genome editing in eukaryotic cells. ZFNs have been used for targeted mutagenesis in model and crop species. In animal and human cells, transient ZFN expression is often achieved by direct gene transfer into the target cells. Stable transformation, however, is the preferred method for gene expression in plant species, and ZFN-expressing transgenic plants have been used for recovery of mutants that are likely to be classified as transgenic due to the use of direct gene-transfer methods into the target cells. Here we present an alternative, nontransgenic approach for ZFN delivery and production of mutant plants using a novel Tobacco rattle virus (TRV)-based expression system for indirect transient delivery of ZFNs into a variety of tissues and cells of intact plants. TRV systemically infected its hosts and virus ZFN-mediated targeted mutagenesis could be clearly observed in newly developed infected tissues as measured by activation of a mutated reporter transgene in tobacco (Nicotiana tabacum) and petunia (Petunia hybrida) plants. The ability of TRV to move to developing buds and regenerating tissues enabled recovery of mutated tobacco and petunia plants. Sequence analysis and transmission of the mutations to the next generation confirmed the stability of the ZFN-induced genetic changes. Because TRV is an RNA virus that can infect a wide range of plant species, it provides a viable alternative to the production of ZFN-mediated mutants while avoiding the use of direct plant-transformation methods.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Tzvi Tzfira
- Danziger Innovations Ltd., Mishmar Hashiva Village, Beit Dagan 50297, Israel (I.M., A.Z., S.R.); Institute of Plant Sciences and Genetics in Agriculture, Robert H. Smith Faculty of Agriculture, Food, and Environment, Hebrew University of Jerusalem, Rehovot 76100, Israel (I.M., E.S., S.R., M.O., A.V.); Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan 48109 (V.Z., A.T., T.T.)
| | | |
Collapse
|