51
|
Donovan CH, Wong SA, Randolph SH, Stark RA, Gibb RL, Gruber AJ. Sex differences in rat decision-making: The confounding role of extraneous feeder sampling between trials. Behav Brain Res 2018; 342:62-69. [PMID: 29355674 DOI: 10.1016/j.bbr.2018.01.018] [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: 11/16/2017] [Revised: 01/08/2018] [Accepted: 01/16/2018] [Indexed: 01/19/2023]
Abstract
Although male and female rats appear to perform differently in some tasks, a clear picture of sex differences in decision-making has yet to develop. This is in part due to significant variability arising from differences in strains and tasks. The aim of this study was to characterize the effects of sex on specific response elements in a reinforcement learning task so as to help identify potential explanations for this variability. We found that the primary difference between sexes was the propensity to approach feeders out of the task context. This extraneous feeder sampling affects choice on subsequent trials in both sexes by promoting a lose-shift response away from the last feeder sampled. Female rats, however, were more likely to engage in this extraneous feeder sampling, and therefore exhibited a greater rate of this effect. Once trials following extraneous sampling were removed, there were no significant sex differences in any of the tested measures. These data suggest that feeder approach outside of the task context, which is often not recorded, could produce a confound in sex-based differences of reinforcement sensitivity in some tasks.
Collapse
Affiliation(s)
- Clifford H Donovan
- Canadian Centre for Behavioural Neuroscience, Department of Neuroscience, University of Lethbridge, 4401 University Drive West, Lethbridge, AB, T1K 6T5, Canada
| | - Scott A Wong
- Canadian Centre for Behavioural Neuroscience, Department of Neuroscience, University of Lethbridge, 4401 University Drive West, Lethbridge, AB, T1K 6T5, Canada
| | - Sienna H Randolph
- Canadian Centre for Behavioural Neuroscience, Department of Neuroscience, University of Lethbridge, 4401 University Drive West, Lethbridge, AB, T1K 6T5, Canada
| | - Rachel A Stark
- Canadian Centre for Behavioural Neuroscience, Department of Neuroscience, University of Lethbridge, 4401 University Drive West, Lethbridge, AB, T1K 6T5, Canada
| | - Robbin L Gibb
- Canadian Centre for Behavioural Neuroscience, Department of Neuroscience, University of Lethbridge, 4401 University Drive West, Lethbridge, AB, T1K 6T5, Canada
| | - Aaron J Gruber
- Canadian Centre for Behavioural Neuroscience, Department of Neuroscience, University of Lethbridge, 4401 University Drive West, Lethbridge, AB, T1K 6T5, Canada.
| |
Collapse
|
52
|
Pigna E, Renzini A, Greco E, Simonazzi E, Fulle S, Mancinelli R, Moresi V, Adamo S. HDAC4 preserves skeletal muscle structure following long-term denervation by mediating distinct cellular responses. Skelet Muscle 2018; 8:6. [PMID: 29477142 PMCID: PMC6389241 DOI: 10.1186/s13395-018-0153-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 02/18/2018] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Denervation triggers numerous molecular responses in skeletal muscle, including the activation of catabolic pathways and oxidative stress, leading to progressive muscle atrophy. Histone deacetylase 4 (HDAC4) mediates skeletal muscle response to denervation, suggesting the use of HDAC inhibitors as a therapeutic approach to neurogenic muscle atrophy. However, the effects of HDAC4 inhibition in skeletal muscle in response to long-term denervation have not been described yet. METHODS To further study HDAC4 functions in response to denervation, we analyzed mutant mice in which HDAC4 is specifically deleted in skeletal muscle. RESULTS After an initial phase of resistance to neurogenic muscle atrophy, skeletal muscle with a deletion of HDAC4 lost structural integrity after 4 weeks of denervation. Deletion of HDAC4 impaired the activation of the ubiquitin-proteasome system, delayed the autophagic response, and dampened the OS response in skeletal muscle. Inhibition of the ubiquitin-proteasome system or the autophagic response, if on the one hand, conferred resistance to neurogenic muscle atrophy; on the other hand, induced loss of muscle integrity and inflammation in mice lacking HDAC4 in skeletal muscle. Moreover, treatment with the antioxidant drug Trolox prevented loss of muscle integrity and inflammation in in mice lacking HDAC4 in skeletal muscle, despite the resistance to neurogenic muscle atrophy. CONCLUSIONS These results reveal new functions of HDAC4 in mediating skeletal muscle response to denervation and lead us to propose the combined use of HDAC inhibitors and antioxidant drugs to treat neurogenic muscle atrophy.
Collapse
Affiliation(s)
- Eva Pigna
- DAHFMO Unit of Histology and Medical Embryology, Interuniversity Institute of Myology, Sapienza University of Rome, Rome, Italy
| | - Alessandra Renzini
- DAHFMO Unit of Histology and Medical Embryology, Interuniversity Institute of Myology, Sapienza University of Rome, Rome, Italy
| | - Emanuela Greco
- DAHFMO Unit of Histology and Medical Embryology, Interuniversity Institute of Myology, Sapienza University of Rome, Rome, Italy
| | - Elena Simonazzi
- DAHFMO Unit of Histology and Medical Embryology, Interuniversity Institute of Myology, Sapienza University of Rome, Rome, Italy
| | - Stefania Fulle
- Department of Neuroscience Imaging and Clinical Sciences-Section of Physiology and Physiopathology, University "G. d'Annunzio" Chieti-Pescara, Chieti, Italy
| | - Rosa Mancinelli
- Department of Neuroscience Imaging and Clinical Sciences-Section of Physiology and Physiopathology, University "G. d'Annunzio" Chieti-Pescara, Chieti, Italy
| | - Viviana Moresi
- DAHFMO Unit of Histology and Medical Embryology, Interuniversity Institute of Myology, Sapienza University of Rome, Rome, Italy.
| | - Sergio Adamo
- DAHFMO Unit of Histology and Medical Embryology, Interuniversity Institute of Myology, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
53
|
Peterson SE, Garitaonandia I, Loring JF. The tumorigenic potential of pluripotent stem cells: What can we do to minimize it? Bioessays 2017; 38 Suppl 1:S86-95. [PMID: 27417126 DOI: 10.1002/bies.201670915] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Revised: 02/05/2016] [Accepted: 02/09/2016] [Indexed: 12/14/2022]
Abstract
Human pluripotent stem cells (hPSCs) have the potential to fundamentally change the way that we go about treating and understanding human disease. Despite this extraordinary potential, these cells also have an innate capability to form tumors in immunocompromised individuals when they are introduced in their pluripotent state. Although current therapeutic strategies involve transplantation of only differentiated hPSC derivatives, there is still a concern that transplanted cell populations could contain a small percentage of cells that are not fully differentiated. In addition, these cells have been frequently reported to acquire genetic alterations that, in some cases, are associated with certain types of human cancers. Here, we try to separate the panic from reality and rationally evaluate the true tumorigenic potential of these cells. We also discuss a recent study examining the effect of culture conditions on the genetic integrity of hPSCs. Finally, we present a set of sensible guidelines for minimizing the tumorigenic potential of hPSC-derived cells. © 2016 The Authors. Inside the Cell published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Suzanne E Peterson
- Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California, USA.,Center for Regenerative Medicine, The Scripps Research Institute, La Jolla, California, USA
| | - Ibon Garitaonandia
- Department of Neurogenetics, International Stem Cell Corporation, Oceanside, CA, USA
| | - Jeanne F Loring
- Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California, USA.,Center for Regenerative Medicine, The Scripps Research Institute, La Jolla, California, USA
| |
Collapse
|
54
|
Mouse Genetic Analysis of Bone Marrow Stem Cell Niches: Technological Pitfalls, Challenges, and Translational Considerations. Stem Cell Reports 2017; 9:1343-1358. [PMID: 29056332 PMCID: PMC5829346 DOI: 10.1016/j.stemcr.2017.09.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 09/18/2017] [Accepted: 09/19/2017] [Indexed: 01/08/2023] Open
Abstract
The development of mouse genetic tools has made a significant contribution to the understanding of skeletal and hematopoietic stem cell niches in bone marrow (BM). However, many experimental designs (e.g., selections of marker genes, target vector constructions, and choices of reporter murine strains) have unavoidable technological limitations and bias, which lead to experimental discrepancies, data reproducibility issues, and frequent data misinterpretation. Consequently, there are a number of conflicting views relating to fundamental biological questions, including origins and locations of skeletal and hematopoietic stem cells in the BM. In this report, we systematically unravel complicated data interpretations via comprehensive analyses of technological benefits, pitfalls, and challenges in frequently used mouse models and discuss their translational relevance to human stem cell biology. Particularly, we emphasize the important roles of using large human genomic data-informatics in facilitating genetic analyses of mouse models and resolving existing controversies in mouse and human BM stem cell biology.
Collapse
|
55
|
Tobita T, Kiyozumi D, Ikawa M. Placenta-specific gene manipulation using lentiviral vector and its application. Placenta 2017; 59 Suppl 1:S37-S43. [PMID: 28988726 DOI: 10.1016/j.placenta.2017.09.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Revised: 09/20/2017] [Accepted: 09/25/2017] [Indexed: 12/16/2022]
Abstract
The placenta is an essential organ for embryo development in the uterus of eutherian mammals. Large contributions in unveiling molecular mechanisms and physiological functions underlying placental formation were made by analyzing mutant and transgenic animals. However, it had been difficult to elucidate whether the placental defects observed in such animals originate from the placenta itself or from the fetus, as both placental and fetal genomes are modified. Therefore strategies to modify the placental genome without affecting the "fetal genome" had been needed. Through the ingenious use of lentiviral (LV) vectors, placenta-specific modification is now possible. Lentivirus is a genus of retroviruses that use reverse-transcriptase to convert its single-strand RNA genome to double-strand DNA and integrate into the host genome. Previous studies showed that when LV vectors were used to transduce embryos at the 2-cell stage, the viral genome is systemically introduced into host genome. Interestingly, by delaying the timing of transduction to the blastocyst stage, the transgene is expressed specifically in the placenta as a consequence of trophectoderm-specific viral transduction. This review summarizes the development of the LV vector-mediated placenta-specific gene manipulation technology and its application in placental research over the past decade. A perspective for future application of LV vectors to further placenta research, especially in combination with next generation genome editing technologies, is also presented.
Collapse
Affiliation(s)
- Tomohiro Tobita
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan; Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Daiji Kiyozumi
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Masahito Ikawa
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan; Graduate School of Medicine, Osaka University, Osaka, Japan.
| |
Collapse
|
56
|
Singh RK, Mallela RK, Hayes A, Dunham NR, Hedden ME, Enke RA, Fariss RN, Sternberg H, West MD, Nasonkin IO. Dnmt1, Dnmt3a and Dnmt3b cooperate in photoreceptor and outer plexiform layer development in the mammalian retina. Exp Eye Res 2017; 159:132-146. [PMID: 27865785 DOI: 10.1016/j.exer.2016.11.014] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Revised: 10/21/2016] [Accepted: 11/14/2016] [Indexed: 11/18/2022]
Abstract
Characterizing the role of epigenetic regulation in the mammalian retina is critical for understanding fundamental mechanisms of retinal development and disease. DNA methylation, an epigenetic modifier of genomic DNA, plays an important role in modulating networks of tissue and cell-specific gene expression. However, the impact of DNA methylation on retinal development and homeostasis of retinal neurons remains unclear. Here, we have created a tissue-specific DNA methyltransferase (Dnmt) triple mutant mouse in an effort to characterize the impact of DNA methylation on retinal development and homeostasis. An Rx-Cre transgene was used to drive targeted mutation of all three murine Dnmt genes in the mouse retina encoding major DNA methylation enzymes DNMT1, DNMT3A and DNMT3B. The triple mutant mice represent a hypomorph model since Dnmt1 catalytic activity was still present and excision of Dnmt3a and Dnmt3b had only about 90% efficiency. Mutation of all three Dnmts resulted in global genomic hypomethylation and dramatic reorganization of the photoreceptor and synaptic layers within retina. Transcriptome and proteomic analyses demonstrated enrichment of dysregulated phototransduction and synaptic genes. The 5 mC signal in triple mutant retina was confined to the central heterochromatin but reduced in the peripheral heterochromatin region of photoreceptor nuclei. In addition, we found a reduction of the 5 mC signal in ganglion cell nuclei. Collectively, this data suggests cooperation of all three Dnmts in the formation and homeostasis of photoreceptors and other retinal neurons within the mammalian retina, and highlight the relevance of epigenetic regulation to sensory retinal disorders and vision loss.
Collapse
Affiliation(s)
- Ratnesh K Singh
- Department of Ophthalmology, University of Pittsburgh Medical School, USA.
| | - Ramya K Mallela
- Department of Ophthalmology, University of Pittsburgh Medical School, USA
| | - Abigail Hayes
- Department of Ophthalmology, West Virginia University, USA
| | | | | | - Raymond A Enke
- Department of Biology, James Madison University, USA; Center for Genome and Metagenome Studies, James Madison University, USA
| | - Robert N Fariss
- Biological Imaging Core, National Eye Institute, Bethesda, MD 20892, USA
| | - Hal Sternberg
- BioTime, 1010 Atlantic Avenue, Alameda, CA 94501, USA
| | | | - Igor O Nasonkin
- Department of Ophthalmology, University of Pittsburgh Medical School, USA.
| |
Collapse
|
57
|
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
|
58
|
Harris SL, Kazmierczak M, Pangršič T, Shah P, Chuchvara N, Barrantes-Freer A, Moser T, Schwander M. Conditional deletion of pejvakin in adult outer hair cells causes progressive hearing loss in mice. Neuroscience 2017; 344:380-393. [PMID: 28089576 DOI: 10.1016/j.neuroscience.2016.12.055] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 12/27/2016] [Accepted: 12/29/2016] [Indexed: 10/20/2022]
Abstract
Mutations in the Pejvakin (Pjvk) gene cause autosomal recessive hearing loss DFNB59 with audiological features of auditory neuropathy spectrum disorder (ANSD) or cochlear dysfunction. The precise mechanisms underlying the variable clinical phenotypes of DFNB59 remain unclear. Here, we demonstrate that mice with conditional ablation of the Pjvk gene in all sensory hair cells or only in outer hair cells (OHCs) show similar auditory phenotypes with early-onset profound hearing loss. By contrast, loss of Pjvk in adult OHCs causes a slowly progressive hearing loss associated with OHC degeneration and delayed loss of inner hair cells (IHCs), indicating a primary role for pejvakin in regulating OHC function and survival. Consistent with this model, synaptic transmission at the IHC ribbon synapse is largely unaffected in sirtaki mice that carry a C-terminal deletion mutation in Pjvk. Using the C-terminal domain of pejvakin as bait, we identified in a cochlear cDNA library ROCK2, an effector for the small GTPase Rho, and the scaffold protein IQGAP1, involved in modulating actin dynamics. Both ROCK2 and IQGAP1 associate via their coiled-coil domains with pejvakin. We conclude that pejvakin is required to sustain OHC activity and survival in a cell-autonomous manner likely involving regulation of Rho signaling.
Collapse
Affiliation(s)
- Suzan L Harris
- Department of Cell Biology and Neuroscience, Rutgers the State University of New Jersey, Piscataway, NJ 08854, United States
| | - Marcin Kazmierczak
- Department of Cell Biology and Neuroscience, Rutgers the State University of New Jersey, Piscataway, NJ 08854, United States
| | - Tina Pangršič
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, 37099 Göttingen, Germany; Collaborative Research Center 889, University of Göttingen, 37099 Göttingen, Germany
| | - Prahar Shah
- Department of Cell Biology and Neuroscience, Rutgers the State University of New Jersey, Piscataway, NJ 08854, United States
| | - Nadiya Chuchvara
- Department of Cell Biology and Neuroscience, Rutgers the State University of New Jersey, Piscataway, NJ 08854, United States
| | - Alonso Barrantes-Freer
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, 37099 Göttingen, Germany; Collaborative Research Center 889, University of Göttingen, 37099 Göttingen, Germany
| | - Tobias Moser
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, 37099 Göttingen, Germany; Collaborative Research Center 889, University of Göttingen, 37099 Göttingen, Germany
| | - Martin Schwander
- Department of Cell Biology and Neuroscience, Rutgers the State University of New Jersey, Piscataway, NJ 08854, United States.
| |
Collapse
|
59
|
Lent-On-Plus Lentiviral vectors for conditional expression in human stem cells. Sci Rep 2016; 6:37289. [PMID: 27853296 PMCID: PMC5112523 DOI: 10.1038/srep37289] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 10/28/2016] [Indexed: 12/25/2022] Open
Abstract
Conditional transgene expression in human stem cells has been difficult to achieve due to the low efficiency of existing delivery methods, the strong silencing of the transgenes and the toxicity of the regulators. Most of the existing technologies are based on stem cells clones expressing appropriate levels of tTA or rtTA transactivators (based on the TetR-VP16 chimeras). In the present study, we aim the generation of Tet-On all-in-one lentiviral vectors (LVs) that tightly regulate transgene expression in human stem cells using the original TetR repressor. By using appropriate promoter combinations and shielding the LVs with the Is2 insulator, we have constructed the Lent-On-Plus Tet-On system that achieved efficient transgene regulation in human multipotent and pluripotent stem cells. The generation of inducible stem cell lines with the Lent-ON-Plus LVs did not require selection or cloning, and transgene regulation was maintained after long-term cultured and upon differentiation toward different lineages. To our knowledge, Lent-On-Plus is the first all-in-one vector system that tightly regulates transgene expression in bulk populations of human pluripotent stem cells and its progeny.
Collapse
|
60
|
Runegaard AH, Jensen KL, Fitzpatrick CM, Dencker D, Weikop P, Gether U, Rickhag M. Preserved dopaminergic homeostasis and dopamine-related behaviour in hemizygous TH-Cre mice. Eur J Neurosci 2016; 45:121-128. [PMID: 27453291 DOI: 10.1111/ejn.13347] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 07/14/2016] [Accepted: 07/18/2016] [Indexed: 02/06/2023]
Abstract
Cre-driver mouse lines have been extensively used as genetic tools to target and manipulate genetically defined neuronal populations by expression of Cre recombinase under selected gene promoters. This approach has greatly advanced neuroscience but interpretations are hampered by the fact that most Cre-driver lines have not been thoroughly characterized. Thus, a phenotypic characterization is of major importance to reveal potential aberrant phenotypes prior to implementation and usage to selectively inactivate or induce transgene expression. Here, we present a biochemical and behavioural assessment of the dopaminergic system in hemizygous tyrosine hydroxylase (TH)-Cre mice in comparison to wild-type (WT) controls. Our data show that TH-Cre mice display preserved dopaminergic homeostasis with unaltered levels of TH and dopamine as well as unaffected dopamine turnover in striatum. TH-Cre mice also show preserved dopamine transporter expression and function supporting sustained dopaminergic transmission. In addition, TH-Cre mice demonstrate normal responses in basic behavioural paradigms related to dopaminergic signalling including locomotor activity, reward preference and anxiolytic behaviour. Our results suggest that TH-Cre mice represent a valid tool to study the dopamine system, though careful characterization must always be performed to prevent false interpretations following Cre-dependent transgene expression and manipulation of selected neuronal pathways.
Collapse
Affiliation(s)
- Annika H Runegaard
- Molecular Neuropharmacology and Genetics Laboratory, Lundbeck Foundation Center for Biomembranes in Nanomedicine, Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kathrine L Jensen
- Molecular Neuropharmacology and Genetics Laboratory, Lundbeck Foundation Center for Biomembranes in Nanomedicine, Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ciarán M Fitzpatrick
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Ditte Dencker
- Laboratory of Neuropsychiatry, Psychiatric Center Copenhagen and Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Pia Weikop
- Laboratory of Neuropsychiatry, Psychiatric Center Copenhagen and Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Ulrik Gether
- Molecular Neuropharmacology and Genetics Laboratory, Lundbeck Foundation Center for Biomembranes in Nanomedicine, Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mattias Rickhag
- Molecular Neuropharmacology and Genetics Laboratory, Lundbeck Foundation Center for Biomembranes in Nanomedicine, Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
61
|
Carow B, Gao Y, Coquet J, Reilly M, Rottenberg ME. lck-Driven Cre Expression Alters T Cell Development in the Thymus and the Frequencies and Functions of Peripheral T Cell Subsets. THE JOURNAL OF IMMUNOLOGY 2016; 197:2261-8. [PMID: 27503210 DOI: 10.4049/jimmunol.1600827] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 07/05/2016] [Indexed: 11/19/2022]
Abstract
Conditional gene targeting using the bacteriophage-derived Cre recombinase is widely applied for functional gene studies in mice. Mice transgenic for Cre under the control of the lck gene promoter are used to study the role of loxP-targeted genes in T cell development and function. In this article, we show a striking 65% reduction in cellularity, preferential development of γδ versus αβ T cells, and increased expression of IL-7R in the thymus of mice expressing Cre under the proximal lck promoter (lck-cre(+) mice). The transition from CD4/CD8 double-negative to double-positive cells was blocked, and lck-cre(+) double-positive cells were more prone to apoptosis and showed higher levels of Cre expression. Importantly, numbers of naive T cells were reduced in spleens and lymph nodes of lck-cre(+) mice. In contrast, frequencies of γδ T cells, CD44(+)CD62L(-) effector T cells, and Foxp3(+) regulatory T cells were elevated, as was the frequency of IFN-γ-secreting CD4(+) and CD8(+) T cells. A literature survey of 332 articles that used lck-cre(+) mice for deletion of floxed genes indicated that results are statistically influenced by the control used (lck-cre(+) or lck-cre(-)), more frequently resembling the lck-cre(+) phenotype described in this article if lck-cre(-) controls were used. Altogether, care should be taken when interpreting published results and to properly control targeted gene deletions using the lck-cre(+) strain.
Collapse
Affiliation(s)
- Berit Carow
- Department of Microbiology, Tumor, and Cell Biology, Karolinska Institutet, S 171 77 Stockholm, Sweden; and
| | - Yu Gao
- Department of Microbiology, Tumor, and Cell Biology, Karolinska Institutet, S 171 77 Stockholm, Sweden; and
| | - Jonathan Coquet
- Department of Microbiology, Tumor, and Cell Biology, Karolinska Institutet, S 171 77 Stockholm, Sweden; and
| | - Marie Reilly
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, S 171 77 Stockholm, Sweden
| | - Martin E Rottenberg
- Department of Microbiology, Tumor, and Cell Biology, Karolinska Institutet, S 171 77 Stockholm, Sweden; and
| |
Collapse
|
62
|
Abraham AD, Neve KA, Lattal KM. Effects of D1 receptor knockout on fear and reward learning. Neurobiol Learn Mem 2016; 133:265-273. [PMID: 27423521 DOI: 10.1016/j.nlm.2016.07.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 07/08/2016] [Accepted: 07/11/2016] [Indexed: 01/11/2023]
Abstract
Dopamine signaling is involved in a variety of neurobiological processes that contribute to learning and memory. D1-like dopamine receptors (including D1 and D5 receptors) are thought to be involved in memory and reward processes, but pharmacological approaches have been limited in their ability to distinguish between D1 and D5 receptors. Here, we examine the effects of a specific knockout of D1 receptors in associative learning tasks involving aversive (shock) or appetitive (cocaine) unconditioned stimuli. We find that D1 knockout mice show similar levels of cued and contextual fear conditioning to WT controls following conditioning protocols involving one, two, or four shocks. D1 knockout mice show increased generalization of fear conditioning and extinction across contexts, revealed as increased freezing to a novel context following conditioning and decreased freezing to an extinguished cue during a contextual renewal test. Further, D1 knockout mice show mild enhancements in extinction following an injection of SKF81297, a D1/D5 receptor agonist, suggesting a role for D5 receptors in extinction enhancements induced by nonspecific pharmacological agonists. Finally, although D1 knockout mice show decreased locomotion induced by cocaine, they are able to form a cocaine-induced conditioned place preference. We discuss these findings in terms of the role of dopamine D1 receptors in general learning and memory processes.
Collapse
Affiliation(s)
- Antony D Abraham
- Department of Behavioral Neuroscience, Oregon Health & Science University, United States
| | - Kim A Neve
- Department of Behavioral Neuroscience, Oregon Health & Science University, United States; Research Service, VA Portland Health Care System, Portland, OR 97239, United States
| | - K Matthew Lattal
- Department of Behavioral Neuroscience, Oregon Health & Science University, United States.
| |
Collapse
|
63
|
Rocha-Martins M, Cavalheiro GR, Matos-Rodrigues GE, Martins RAP. From Gene Targeting to Genome Editing: Transgenic animals applications and beyond. AN ACAD BRAS CIENC 2016; 87:1323-48. [PMID: 26397828 DOI: 10.1590/0001-3765201520140710] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Genome modification technologies are powerful tools for molecular biology and related areas. Advances in animal transgenesis and genome editing technologies during the past three decades allowed systematic interrogation of gene function that can help model how the genome influences cellular physiology. Genetic engineering via homologous recombination (HR) has been the standard method to modify genomic sequences. Nevertheless, nuclease-guided genome editing methods that were developed recently, such as ZFN, TALEN and CRISPR/Cas, opened new perspectives for biomedical research. Here, we present a brief historical perspective of genome modification methods, focusing on transgenic mice models. Moreover, we describe how new techniques were discovered and improved, present the paradigm shifts and discuss their limitations and applications for biomedical research as well as possible future directions.
Collapse
Affiliation(s)
- Maurício Rocha-Martins
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, BR
| | - Gabriel R Cavalheiro
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, BR
| | | | - Rodrigo A P Martins
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, BR
| |
Collapse
|
64
|
Cancer-specific binary expression system activated in mice by bacteriophage HK022 Integrase. Sci Rep 2016; 6:24971. [PMID: 27117628 PMCID: PMC4846993 DOI: 10.1038/srep24971] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 04/07/2016] [Indexed: 12/01/2022] Open
Abstract
Binary systems based on site-specific recombination have been used for tumor specific transcription targeting of suicide genes in animal models. In these binary systems a site specific recombinase or integrase that is expressed from a tumor specific promoter drives tumor specific expression of a cytotoxic gene. In the present study we developed a new cancer specific binary expression system activated by the Integrase (Int) of the lambdoid phage HK022. We demonstrate the validity of this system by the specific expression of a luciferase (luc) reporter in human embryonic kidney 293T (HEK293T) cells and in a lung cancer mouse model. Due to the absence viral vectors and of cytotoxicity the Int based binary system offers advantages over previously described counterparts and may therefore be developed into a safer cancer cell killing system.
Collapse
|
65
|
Lee HB, Sundberg BN, Sigafoos AN, Clark KJ. Genome Engineering with TALE and CRISPR Systems in Neuroscience. Front Genet 2016; 7:47. [PMID: 27092173 PMCID: PMC4821859 DOI: 10.3389/fgene.2016.00047] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 03/16/2016] [Indexed: 12/26/2022] Open
Abstract
Recent advancement in genome engineering technology is changing the landscape of biological research and providing neuroscientists with an opportunity to develop new methodologies to ask critical research questions. This advancement is highlighted by the increased use of programmable DNA-binding agents (PDBAs) such as transcription activator-like effector (TALE) and RNA-guided clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR associated (Cas) systems. These PDBAs fused or co-expressed with various effector domains allow precise modification of genomic sequences and gene expression levels. These technologies mirror and extend beyond classic gene targeting methods contributing to the development of novel tools for basic and clinical neuroscience. In this Review, we discuss the recent development in genome engineering and potential applications of this technology in the field of neuroscience.
Collapse
Affiliation(s)
- Han B Lee
- Neurobiology of Disease Graduate Program, Mayo Graduate School Rochester, MN, USA
| | - Brynn N Sundberg
- Department of Biochemistry and Molecular Biology, Mayo Clinic Rochester, MN, USA
| | - Ashley N Sigafoos
- Department of Biochemistry and Molecular Biology, Mayo Clinic Rochester, MN, USA
| | - Karl J Clark
- Neurobiology of Disease Graduate Program, Mayo Graduate SchoolRochester, MN, USA; Department of Biochemistry and Molecular Biology, Mayo ClinicRochester, MN, USA
| |
Collapse
|
66
|
Chen CV, Brummet JL, Jordan CL, Breedlove SM. Down, But Not Out: Partial Elimination of Androgen Receptors in the Male Mouse Brain Does Not Affect Androgenic Regulation of Anxiety or HPA Activity. Endocrinology 2016; 157:764-73. [PMID: 26562258 PMCID: PMC5393364 DOI: 10.1210/en.2015-1417] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
We previously found that androgen receptor (AR) activity mediates two effects of T in adult male mice: reduction of anxiety-like behaviors and dampening of the hypothalamic-pituitary-adrenal response to stress. To determine whether brain ARs mediate these effects, we used the Cre/loxP technology seeking to disable AR throughout the central nervous system (CNS). Female mice carrying the floxed AR allele (ARlox) were crossed with males carrying cre recombinase transgene controlled by the nestin promoter (NesCre), producing cre in developing neurons and glia. Among male offspring, four genotypes resulted: males carrying ARlox and NesCre (NesARko), and three control groups (wild types, NesCre, and ARlox). Reporter mice indicated ubiquitous Cre expression throughout the CNS. Nevertheless, AR immunocytochemistry in NesARko mice revealed efficient knockout (KO) of AR in some brain regions (hippocampus and medial prefrontal cortex [mPFC]), but not others. Substantial AR protein was seen in the amygdala and hypothalamus among other regions, whereas negligible AR remained in others like the bed nucleus of the stria terminalis and dorsal periaqueductal gray. This selective KO allowed for testing the role of AR in hippocampus and mPFC. Males were castrated and implanted with T at postnatal day 60 before testing on postnatal day 90-100. In contrast with males with global KO of AR, T still modulated anxiety-related behavior and hypothalamic-pituitary-adrenal activity in NesARko males. These results leave open the possibility that AR acting in the CNS mediates these effects of T, but demonstrate that AR is not required in the hippocampus or mPFC for T's anxiolytic effects.
Collapse
Affiliation(s)
- Chieh V Chen
- Psychology Department (C.V.C., J.L.B., C.L.J., S.M.B.) and Neuroscience Program (C.L.J., S.M.B.), Michigan State University, East Lansing, Michigan 48824
| | - Jennifer L Brummet
- Psychology Department (C.V.C., J.L.B., C.L.J., S.M.B.) and Neuroscience Program (C.L.J., S.M.B.), Michigan State University, East Lansing, Michigan 48824
| | - Cynthia L Jordan
- Psychology Department (C.V.C., J.L.B., C.L.J., S.M.B.) and Neuroscience Program (C.L.J., S.M.B.), Michigan State University, East Lansing, Michigan 48824
| | - S Marc Breedlove
- Psychology Department (C.V.C., J.L.B., C.L.J., S.M.B.) and Neuroscience Program (C.L.J., S.M.B.), Michigan State University, East Lansing, Michigan 48824
| |
Collapse
|
67
|
Assmann JC, Körbelin J, Schwaninger M. Genetic manipulation of brain endothelial cells in vivo. Biochim Biophys Acta Mol Basis Dis 2015; 1862:381-94. [PMID: 26454206 DOI: 10.1016/j.bbadis.2015.10.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Revised: 09/30/2015] [Accepted: 10/05/2015] [Indexed: 12/19/2022]
Affiliation(s)
- Julian C Assmann
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany
| | - Jakob Körbelin
- University Medical Center Hamburg-Eppendorf, Hubertus Wald Cancer Center, Department of Oncology and Hematology, Martinistr. 52, 20246 Hamburg, Germany
| | - Markus Schwaninger
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany.
| |
Collapse
|
68
|
Ceizar M, Dhaliwal J, Xi Y, Smallwood M, Kumar KL, Lagace DC. Bcl-2 is required for the survival of doublecortin-expressing immature neurons. Hippocampus 2015; 26:211-9. [DOI: 10.1002/hipo.22504] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/06/2015] [Indexed: 12/13/2022]
Affiliation(s)
- Maheen Ceizar
- Department of Neuroscience, Faculty of Medicine; University of Ottawa; Ottawa Ontario K1H 8M5 Canada
| | - Jagroop Dhaliwal
- Department of Neuroscience, Faculty of Medicine; University of Ottawa; Ottawa Ontario K1H 8M5 Canada
| | - Yanwei Xi
- Department of Neuroscience, Faculty of Medicine; University of Ottawa; Ottawa Ontario K1H 8M5 Canada
| | - Megan Smallwood
- Department of Neuroscience, Faculty of Medicine; University of Ottawa; Ottawa Ontario K1H 8M5 Canada
| | - Keren Leviel Kumar
- Department of Neuroscience, Faculty of Medicine; University of Ottawa; Ottawa Ontario K1H 8M5 Canada
| | - Diane C. Lagace
- Department of Neuroscience, Faculty of Medicine; University of Ottawa; Ottawa Ontario K1H 8M5 Canada
| |
Collapse
|
69
|
Tillack K, Aboutalebi H, Kramer ER. An Efficient and Versatile System for Visualization and Genetic Modification of Dopaminergic Neurons in Transgenic Mice. PLoS One 2015; 10:e0136203. [PMID: 26291828 PMCID: PMC4546329 DOI: 10.1371/journal.pone.0136203] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 07/30/2015] [Indexed: 11/18/2022] Open
Abstract
Background & Aims The brain dopaminergic (DA) system is involved in fine tuning many behaviors and several human diseases are associated with pathological alterations of the DA system such as Parkinson’s disease (PD) and drug addiction. Because of its complex network integration, detailed analyses of physiological and pathophysiological conditions are only possible in a whole organism with a sophisticated tool box for visualization and functional modification. Methods & Results Here, we have generated transgenic mice expressing the tetracycline-regulated transactivator (tTA) or the reverse tetracycline-regulated transactivator (rtTA) under control of the tyrosine hydroxylase (TH) promoter, TH-tTA (tet-OFF) and TH-rtTA (tet-ON) mice, to visualize and genetically modify DA neurons. We show their tight regulation and efficient use to overexpress proteins under the control of tet-responsive elements or to delete genes of interest with tet-responsive Cre. In combination with mice encoding tet-responsive luciferase, we visualized the DA system in living mice progressively over time. Conclusion These experiments establish TH-tTA and TH-rtTA mice as a powerful tool to generate and monitor mouse models for DA system diseases.
Collapse
Affiliation(s)
- Karsten Tillack
- Development and Maintenance of the Nervous System, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Helia Aboutalebi
- Development and Maintenance of the Nervous System, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Edgar R. Kramer
- Development and Maintenance of the Nervous System, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- * E-mail:
| |
Collapse
|
70
|
Declercq J, Brouwers B, Pruniau VPEG, Stijnen P, de Faudeur G, Tuand K, Meulemans S, Serneels L, Schraenen A, Schuit F, Creemers JWM. Metabolic and Behavioural Phenotypes in Nestin-Cre Mice Are Caused by Hypothalamic Expression of Human Growth Hormone. PLoS One 2015; 10:e0135502. [PMID: 26275221 PMCID: PMC4537087 DOI: 10.1371/journal.pone.0135502] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 07/22/2015] [Indexed: 11/19/2022] Open
Abstract
The Nestin-Cre driver mouse line has mild hypopituitarism, reduced body weight, a metabolic phenotype and reduced anxiety. Although several causes have been suggested, a comprehensive explanation is still lacking. In this study we examined the molecular mechanisms leading to this compound phenotype. Upon generation of the Nestin-Cre mice, the human growth hormone (hGH) minigene was inserted downstream of the Cre recombinase to ensure efficient transgene expression. As a result, hGH is expressed in the hypothalamus. This results in the auto/paracrine activation of the GH receptor as demonstrated by the increased phosphorylation of signal transducer and activator of transcription 5 (STAT5) and reduced expression of growth hormone releasing hormone (Ghrh). Low Ghrh levels cause hypopituitarism consistent with the observed mouse growth hormone (mGH) deficiency. mGH deficiency caused reduced activation of the GH receptor and hence reduced phosphorylation of STAT5 in the liver. This led to decreased levels of hepatic Igf-1 mRNA and consequently postnatal growth retardation. Furthermore, genes involved in lipid uptake and synthesis, such as CD36 and very low-density lipoprotein receptor were upregulated, resulting in liver steatosis. In conclusion, this study demonstrates the unexpected expression of hGH in the hypothalamus of Nestin-Cre mice which is able to activate both the GH receptor and the prolactin receptor. Increased hypothalamic GH receptor signaling explains the observed hypopituitarism, reduced growth and metabolic phenotype of Nestin-Cre mice. Activation of either receptor is consistent with reduced anxiety.
Collapse
Affiliation(s)
- Jeroen Declercq
- Laboratory for Biochemical Neuroendocrinology, Department of Human Genetics, KU Leuven, Leuven 3000, Belgium
| | - Bas Brouwers
- Laboratory for Biochemical Neuroendocrinology, Department of Human Genetics, KU Leuven, Leuven 3000, Belgium
| | - Vincent P. E. G. Pruniau
- Laboratory for Biochemical Neuroendocrinology, Department of Human Genetics, KU Leuven, Leuven 3000, Belgium
| | - Pieter Stijnen
- Laboratory for Biochemical Neuroendocrinology, Department of Human Genetics, KU Leuven, Leuven 3000, Belgium
| | - Geoffroy de Faudeur
- Gene Expression Unit, Department of Cellular and Molecular Medicine, KU Leuven, Leuven 3000, Belgium
| | - Krizia Tuand
- Laboratory for Biochemical Neuroendocrinology, Department of Human Genetics, KU Leuven, Leuven 3000, Belgium
| | - Sandra Meulemans
- Laboratory for Biochemical Neuroendocrinology, Department of Human Genetics, KU Leuven, Leuven 3000, Belgium
| | - Lutgarde Serneels
- Laboratory for the Research of Degenerative Diseases, KU Leuven, Leuven 3000, Belgium
| | - Anica Schraenen
- Gene Expression Unit, Department of Cellular and Molecular Medicine, KU Leuven, Leuven 3000, Belgium
| | - Frans Schuit
- Gene Expression Unit, Department of Cellular and Molecular Medicine, KU Leuven, Leuven 3000, Belgium
| | - John W. M. Creemers
- Laboratory for Biochemical Neuroendocrinology, Department of Human Genetics, KU Leuven, Leuven 3000, Belgium
- * E-mail:
| |
Collapse
|
71
|
Fritzsch B, Pan N, Jahan I, Elliott KL. Inner ear development: building a spiral ganglion and an organ of Corti out of unspecified ectoderm. Cell Tissue Res 2015; 361:7-24. [PMID: 25381571 PMCID: PMC4426086 DOI: 10.1007/s00441-014-2031-5] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Accepted: 10/09/2014] [Indexed: 01/21/2023]
Abstract
The mammalian inner ear develops from a placodal thickening into a complex labyrinth of ducts with five sensory organs specialized to detect position and movement in space. The mammalian ear also develops a spiraled cochlear duct containing the auditory organ, the organ of Corti (OC), specialized to translate sound into hearing. Development of the OC from a uniform sheet of ectoderm requires unparalleled precision in the topological developmental engineering of four different general cell types, namely sensory neurons, hair cells, supporting cells, and general otic epithelium, into a mosaic of ten distinctly recognizable cell types in and around the OC, each with a unique distribution. Moreover, the OC receives unique innervation by ear-derived spiral ganglion afferents and brainstem-derived motor neurons as efferents and requires neural-crest-derived Schwann cells to form myelin and neural-crest-derived cells to induce the stria vascularis. This transformation of a sheet of cells into a complicated interdigitating set of cells necessitates the orchestrated expression of multiple transcription factors that enable the cellular transformation from ectoderm into neurosensory cells forming the spiral ganglion neurons (SGNs), while simultaneously transforming the flat epithelium into a tube, the cochlear duct, housing the OC. In addition to the cellular and conformational changes forming the cochlear duct with the OC, changes in the surrounding periotic mesenchyme form passageways for sound to stimulate the OC. We review molecular developmental data, generated predominantly in mice, in order to integrate the well-described expression changes of transcription factors and their actions, as revealed in mutants, in the formation of SGNs and OC in the correct position and orientation with suitable innervation. Understanding the molecular basis of these developmental changes leading to the formation of the mammalian OC and highlighting the gaps in our knowledge might guide in vivo attempts to regenerate this most complicated cellular mosaic of the mammalian body for the reconstitution of hearing in a rapidly growing population of aging people suffering from hearing loss.
Collapse
Affiliation(s)
- Bernd Fritzsch
- College of Liberal Arts and Sciences, Department of Biology, University of Iowa, 143 BB, 123 Jefferson Avenue, Iowa City, IA 52242, USA,
| | | | | | | |
Collapse
|
72
|
Tomlinson JAP, Caplin B, Boruc O, Bruce-Cobbold C, Cutillas P, Dormann D, Faull P, Grossman RC, Khadayate S, Mas VR, Nitsch DD, Wang Z, Norman JT, Wilcox CS, Wheeler DC, Leiper J. Reduced Renal Methylarginine Metabolism Protects against Progressive Kidney Damage. J Am Soc Nephrol 2015; 26:3045-59. [PMID: 25855779 DOI: 10.1681/asn.2014030280] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Accepted: 02/16/2015] [Indexed: 01/10/2023] Open
Abstract
Nitric oxide (NO) production is diminished in many patients with cardiovascular and renal disease. Asymmetric dimethylarginine (ADMA) is an endogenous inhibitor of NO synthesis, and elevated plasma levels of ADMA are associated with poor outcomes. Dimethylarginine dimethylaminohydrolase-1 (DDAH1) is a methylarginine-metabolizing enzyme that reduces ADMA levels. We reported previously that a DDAH1 gene variant associated with increased renal DDAH1 mRNA transcription and lower plasma ADMA levels, but counterintuitively, a steeper rate of renal function decline. Here, we test the hypothesis that reduced renal-specific ADMA metabolism protects against progressive renal damage. Renal DDAH1 is expressed predominately within the proximal tubule. A novel proximal tubule-specific Ddah1 knockout (Ddah1(PT-/-)) mouse demonstrated tubular cell accumulation of ADMA and lower NO concentrations, but unaltered plasma ADMA concentrations. Ddah1(PT-/-) mice were protected from reduced kidney tissue mass, collagen deposition, and profibrotic cytokine expression in two independent renal injury models: folate nephropathy and unilateral ureteric obstruction. Furthermore, a study of two independent kidney transplant cohorts revealed higher levels of human renal allograft methylarginine-metabolizing enzyme gene expression associated with steeper function decline. We also report an association among DDAH1 expression, NO activity, and uromodulin expression supported by data from both animal and human studies, raising the possibility that kidney DDAH1 expression exacerbates renal injury through uromodulin-related mechanisms. Together, these data demonstrate that reduced renal tubular ADMA metabolism protects against progressive kidney function decline. Thus, circulating ADMA may be an imprecise marker of renal methylarginine metabolism, and therapeutic ADMA reduction may even be deleterious to kidney function.
Collapse
Affiliation(s)
- James A P Tomlinson
- Medical Research Council Clinical Sciences Centre, Imperial College, London, United Kingdom;
| | - Ben Caplin
- Centre for Nephrology, UCL Medical School Royal Free, London, United Kingdom
| | - Olga Boruc
- Medical Research Council Clinical Sciences Centre, Imperial College, London, United Kingdom
| | - Claire Bruce-Cobbold
- Medical Research Council Clinical Sciences Centre, Imperial College, London, United Kingdom
| | - Pedro Cutillas
- Medical Research Council Clinical Sciences Centre, Imperial College, London, United Kingdom
| | - Dirk Dormann
- Medical Research Council Clinical Sciences Centre, Imperial College, London, United Kingdom
| | - Peter Faull
- Medical Research Council Clinical Sciences Centre, Imperial College, London, United Kingdom
| | - Rebecca C Grossman
- Centre for Nephrology, UCL Medical School Royal Free, London, United Kingdom
| | - Sanjay Khadayate
- Medical Research Council Clinical Sciences Centre, Imperial College, London, United Kingdom
| | - Valeria R Mas
- Translational Genomics Transplant Laboratory, Transplant Division, Department of Surgery, University of Virginia, Charlottesville, Virginia
| | - Dorothea D Nitsch
- Department of Non-communicable Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, United Kingdom; and
| | - Zhen Wang
- Medical Research Council Clinical Sciences Centre, Imperial College, London, United Kingdom
| | - Jill T Norman
- Centre for Nephrology, UCL Medical School Royal Free, London, United Kingdom
| | - Christopher S Wilcox
- Hypertension, Kidney and Vascular Research Center, Georgetown University, Washington, DC
| | - David C Wheeler
- Centre for Nephrology, UCL Medical School Royal Free, London, United Kingdom
| | - James Leiper
- Medical Research Council Clinical Sciences Centre, Imperial College, London, United Kingdom
| |
Collapse
|
73
|
Wang L, Mishina Y, Liu F. Osterix-Cre transgene causes craniofacial bone development defect. Calcif Tissue Int 2015; 96:129-37. [PMID: 25550101 PMCID: PMC4320656 DOI: 10.1007/s00223-014-9945-5] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Accepted: 12/12/2014] [Indexed: 12/25/2022]
Abstract
The Cre/loxP system has been widely used to generate tissue-specific gene knockout mice. Inducible (Tet-off) Osx-GFP::Cre (Osx-Cre) mouse line that targets osteoblasts is widely used in the bone research field. In this study, we investigated the effect of Osx-Cre on craniofacial bone development. We found that newborn Osx-Cre mice showed severe hypomineralization in parietal, frontal, and nasal bones as well as the coronal sutural area when compared to control mice. As the mice matured, the intramembranous bone hypomineralization phenotype became less severe. The major hypomineralization defect in parietal, frontal, and nasal bones had mostly disappeared by postnatal day 21, but the defect in sutural areas persisted. Importantly, Doxycycline treatment eliminated cranial bone defects at birth which indicates that Cre expression may be responsible for the phenotype. In addition, we showed that the primary calvarial osteoblasts isolated from neonatal Osx-Cre mice had comparable differentiation ability compared to their littermate controls. This study reinforces the idea that Cre-positive litter mates are indispensable controls in studies using conditional gene deletion.
Collapse
Affiliation(s)
- Li Wang
- Department of Biologic and Materials Sciences, University of Michigan School of Dentistry, Ann Arbor, MI 48109, USA
| | - Yuji Mishina
- Department of Biologic and Materials Sciences, University of Michigan School of Dentistry, Ann Arbor, MI 48109, USA
| | - Fei Liu
- Department of Biologic and Materials Sciences, University of Michigan School of Dentistry, Ann Arbor, MI 48109, USA
- Corresponding author. , Phone: 734-936-0911, Fax: 734-647-2805
| |
Collapse
|
74
|
Posttraining ablation of adult-generated olfactory granule cells degrades odor-reward memories. J Neurosci 2015; 34:15793-803. [PMID: 25411506 DOI: 10.1523/jneurosci.2336-13.2014] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Proliferation of neural progenitor cells in the subventricular zone leads to the continuous generation of new olfactory granule cells (OGCs) throughout life. These cells synaptically integrate into olfactory bulb circuits after ∼2 weeks and transiently exhibit heightened plasticity and responses to novel odors. Although these observations suggest that adult-generated OGCs play important roles in olfactory-related memories, global suppression of olfactory neurogenesis does not typically prevent the formation of odor-reward memories, perhaps because residual OGCs can compensate. Here, we used a transgenic strategy to selectively ablate large numbers of adult-generated OGCs either before or after learning in mice. Consistent with previous studies, pretraining ablation of adult-generated OGCs did not prevent the formation of an odor-reward memory, presumably because existing OGCs can support memory formation in their absence. However, ablation of a similar cohort of adult-generated OGCs after training impaired subsequent memory expression, indicating that if these cells are available at the time of training, they play an essential role in subsequent expression of odor-reward memories. Memory impairment was associated with the loss of adult-generated OGCs that were >10 d in age and did not depend on the developmental stage in which they were generated, suggesting that, once sufficiently mature, OGCs generated during juvenility and adulthood play similar roles in the expression of odor-reward memories. Finally, ablation of adult-generated OGCs 1 month after training did not produce amnesia, indicating that adult-generated OGCs play a time-limited role in the expression of odor-reward memories.
Collapse
|
75
|
Dorà NJ, Collinson JM, Hill RE, West JD. Hemizygous Le-Cre transgenic mice have severe eye abnormalities on some genetic backgrounds in the absence of LoxP sites. PLoS One 2014; 9:e109193. [PMID: 25272013 PMCID: PMC4182886 DOI: 10.1371/journal.pone.0109193] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Accepted: 08/29/2014] [Indexed: 11/18/2022] Open
Abstract
Eye phenotypes were investigated in Le-CreTg/−; Pax6fl/+ mice, which were expected to show tissue-specific reduction of Pax6 in surface ectoderm derivatives. To provide a better comparison with our previous studies of Pax6+/− eye phenotypes, hemizygous Le-CreTg/− and heterozygous Pax6fl/+mice were crossed onto the CBA/Ca genetic background. After the Le-Cre transgene had been backcrossed to CBA/Ca for seven generations, significant eye abnormalities occurred in some hemizygous Le-CreTg/−; Pax6+/+ controls (without a floxed Pax6fl allele) as well as experimental Le-CreTg/−; Pax6fl/+ mice. However, no abnormalities were seen in Le-Cre−/−; Pax6fl/+ or Le-Cre−/−; Pax6+/+ controls (without the Le-Cre transgene). The severity and frequency of the eye abnormalities in Le-CreTg/−; Pax6+/+ control mice diminished after backcrossing Le-CreTg/− mice to the original FVB/N strain for two generations, showing that the effect was reversible. This genetic background effect suggests that the eye abnormalities are a consequence of an interaction between the Le-Cre transgene and alleles of unknown modifier genes present in certain genetic backgrounds. The abnormalities were also ameliorated by introducing additional Pax6 gene copies on a CBA/Ca background, suggesting involvement of Pax6 depletion in Le-CreTg/−; Pax6+/+ mice rather than direct action of Cre recombinase on cryptic pseudo-loxP sites. One possibility is that expression of Cre recombinase from the Pax6-Le regulatory sequences in the Le-Cre transgene depletes cofactors required for endogenous Pax6 gene expression. Our observation that eye abnormalities can occur in hemizygous Le-CreTg/−; Pax6+/+ mice, in the absence of a floxed allele, demonstrates the importance of including all the relevant genetic controls in Cre-loxP experiments.
Collapse
Affiliation(s)
- Natalie J. Dorà
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, United Kingdom
| | - J. Martin Collinson
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Robert E. Hill
- Medical Research Council Human Genetics Unit, Medical Research Council Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - John D. West
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, United Kingdom
- * E-mail:
| |
Collapse
|
76
|
Li Y, Choi PS, Casey SC, Felsher DW. Activation of Cre recombinase alone can induce complete tumor regression. PLoS One 2014; 9:e107589. [PMID: 25208064 PMCID: PMC4160265 DOI: 10.1371/journal.pone.0107589] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Accepted: 08/13/2014] [Indexed: 01/10/2023] Open
Abstract
The Cre/loxP system is a powerful tool for generating conditional genomic recombination and is often used to examine the mechanistic role of specific genes in tumorigenesis. However, Cre toxicity due to its non-specific endonuclease activity has been a concern. Here, we report that tamoxifen-mediated Cre activation in vivo induced the regression of primary lymphomas in p53−/− mice. Our findings illustrate that Cre activation alone can induce the regression of established tumors.
Collapse
Affiliation(s)
- Yulin Li
- Department of Medicine, Division of Oncology, School of Medicine, Stanford University, Stanford, California, United States of America
| | - Peter S. Choi
- Department of Medicine, Division of Oncology, School of Medicine, Stanford University, Stanford, California, United States of America
| | - Stephanie C. Casey
- Department of Medicine, Division of Oncology, School of Medicine, Stanford University, Stanford, California, United States of America
| | - Dean W. Felsher
- Department of Medicine, Division of Oncology, School of Medicine, Stanford University, Stanford, California, United States of America
- * E-mail:
| |
Collapse
|
77
|
Gierut JJ, Jacks TE, Haigis KM. Strategies to achieve conditional gene mutation in mice. Cold Spring Harb Protoc 2014; 2014:339-49. [PMID: 24692485 DOI: 10.1101/pdb.top069807] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The laboratory mouse is an ideal model organism for studying disease because it is physiologically similar to human and also because its genome is readily manipulated. Genetic engineering allows researchers to introduce specific loss-of-function or gain-of-function mutations into genes and then to study the resulting phenotypes in an in vivo context. One drawback of using traditional transgenic and knockout mice to study human diseases is that many mutations passed through the germline can profoundly affect development, thus impeding the study of disease phenotypes in adults. New technology has made it possible to generate conditional mutations that can be introduced in a spatially and/or temporally restricted manner. Mouse strains carrying conditional mutations represent valuable experimental models for the study of human diseases and they can be used to develop strategies for prevention and treatment of these diseases. In this article, we will describe the most widely used DNA recombinase systems used to achieve conditional gene mutation in mouse models and discuss how these systems can be employed in vivo.
Collapse
Affiliation(s)
- Jessica J Gierut
- Molecular Pathology Unit and Center for Cancer Research, Massachusetts General Hospital, Department of Pathology, Harvard Medical School, Charlestown, Massachusetts 02129
| | | | | |
Collapse
|
78
|
Storbeck M, Hupperich K, Gaspar JA, Meganathan K, Martínez Carrera L, Wirth R, Sachinidis A, Wirth B. Neuronal-specific deficiency of the splicing factor Tra2b causes apoptosis in neurogenic areas of the developing mouse brain. PLoS One 2014; 9:e89020. [PMID: 24586484 PMCID: PMC3929626 DOI: 10.1371/journal.pone.0089020] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Accepted: 01/13/2014] [Indexed: 02/07/2023] Open
Abstract
Alternative splicing (AS) increases the informational content of the genome and is more prevalent in the brain than in any other tissue. The splicing factor Tra2b (Sfrs10) can modulate splicing inclusion of exons by specifically detecting GAA-rich binding motifs and its absence causes early embryonic lethality in mice. TRA2B has been shown to be involved in splicing processes of Nasp (nuclear autoantigenic sperm protein), MAPT (microtubule associated protein tau) and SMN (survival motor neuron), and is therefore implicated in spermatogenesis and neurological diseases like Alzheimer’s disease, dementia, Parkinson’s disease and spinal muscular atrophy. Here we generated a neuronal-specific Tra2b knock-out mouse that lacks Tra2b expression in neuronal and glial precursor cells by using the Nestin-Cre. Neuronal-specific Tra2b knock-out mice die immediately after birth and show severe abnormalities in cortical development, which are caused by massive apoptotic events in the ventricular layers of the cortex, demonstrating a pivotal role of Tra2b for the developing central nervous system. Using whole brain RNA on exon arrays we identified differentially expressed alternative exons of Tubulinδ1 and Shugoshin-like2 as in vivo targets of Tra2b. Most interestingly, we found increased expression of the cyclin dependent kinase inhibitor 1a (p21) which we could functionally link to neuronal precursor cells in the affected brain regions. We provide further evidence that the absence of Tra2b causes p21 upregulation and ultimately cell death in NSC34 neuronal-like cells. These findings demonstrate that Tra2b regulates splicing events essential for maintaining neuronal viability during development. Apoptotic events triggered via p21 might not be restricted to the developing brain but could possibly be generalized to the whole organism and explain early embryonic lethality in Tra2b-depleted mice.
Collapse
Affiliation(s)
- Markus Storbeck
- Institute of Human Genetics, University of Cologne, Cologne, Germany
- Institute of Genetics, University of Cologne, Cologne, Germany
- Center for Molecular Medicine, University of Cologne, Cologne, Germany
| | - Kristina Hupperich
- Institute of Human Genetics, University of Cologne, Cologne, Germany
- Institute of Genetics, University of Cologne, Cologne, Germany
- Center for Molecular Medicine, University of Cologne, Cologne, Germany
| | | | | | - Lilian Martínez Carrera
- Institute of Human Genetics, University of Cologne, Cologne, Germany
- Institute of Genetics, University of Cologne, Cologne, Germany
- Center for Molecular Medicine, University of Cologne, Cologne, Germany
| | - Radu Wirth
- Institute of Human Genetics, University of Cologne, Cologne, Germany
| | | | - Brunhilde Wirth
- Institute of Human Genetics, University of Cologne, Cologne, Germany
- Institute of Genetics, University of Cologne, Cologne, Germany
- Center for Molecular Medicine, University of Cologne, Cologne, Germany
- * E-mail:
| |
Collapse
|
79
|
Caspase-6 activity in the CA1 region of the hippocampus induces age-dependent memory impairment. Cell Death Differ 2014; 21:696-706. [PMID: 24413155 DOI: 10.1038/cdd.2013.194] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Revised: 11/29/2013] [Accepted: 12/03/2013] [Indexed: 12/11/2022] Open
Abstract
Active Caspase-6 is abundant in the neuropil threads, neuritic plaques and neurofibrillary tangles of Alzheimer disease brains. However, its contribution to the pathophysiology of Alzheimer disease is unclear. Here, we show that higher levels of Caspase-6 activity in the CA1 region of aged human hippocampi correlate with lower cognitive performance. To determine whether Caspase-6 activity, in the absence of plaques and tangles, is sufficient to cause memory deficits, we generated a transgenic knock-in mouse that expresses a self-activated form of human Caspase-6 in the CA1. This Caspase-6 mouse develops age-dependent spatial and episodic memory impairment. Caspase-6 induces neuronal degeneration and inflammation. We conclude that Caspase-6 activation in mouse CA1 neurons is sufficient to induce neuronal degeneration and age-dependent memory impairment. These results indicate that Caspase-6 activity in CA1 could be responsible for the lower cognitive performance of aged humans. Consequently, preventing or inhibiting Caspase-6 activity in the aged may provide an efficient novel therapeutic approach against Alzheimer disease.
Collapse
|
80
|
de Leeuw CN, Dyka FM, Boye SL, Laprise S, Zhou M, Chou AY, Borretta L, McInerny SC, Banks KG, Portales-Casamar E, Swanson MI, D’Souza CA, Boye SE, Jones SJM, Holt RA, Goldowitz D, Hauswirth WW, Wasserman WW, Simpson EM. Targeted CNS Delivery Using Human MiniPromoters and Demonstrated Compatibility with Adeno-Associated Viral Vectors. Mol Ther Methods Clin Dev 2014; 1:5. [PMID: 24761428 PMCID: PMC3992516 DOI: 10.1038/mtm.2013.5] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Accepted: 11/05/2013] [Indexed: 01/21/2023]
Abstract
Critical for human gene therapy is the availability of small promoter tools to drive gene expression in a highly specific and reproducible manner. We tackled this challenge by developing human DNA MiniPromoters using computational biology and phylogenetic conservation. MiniPromoters were tested in mouse as single-copy knock-ins at the Hprt locus on the X Chromosome, and evaluated for lacZ reporter expression in CNS and non-CNS tissue. Eighteen novel MiniPromoters driving expression in mouse brain were identified, two MiniPromoters for driving pan-neuronal expression, and 17 MiniPromoters for the mouse eye. Key areas of therapeutic interest were represented in this set: the cerebral cortex, embryonic hypothalamus, spinal cord, bipolar and ganglion cells of the retina, and skeletal muscle. We also demonstrated that three retinal ganglion cell MiniPromoters exhibit similar cell-type specificity when delivered via adeno-associated virus (AAV) vectors intravitreally. We conclude that our methodology and characterization has resulted in desirable expression characteristics that are intrinsic to the MiniPromoter, not dictated by copy number effects or genomic location, and results in constructs predisposed to success in AAV. These MiniPromoters are immediately applicable for pre-clinical studies towards gene therapy in humans, and are publicly available to facilitate basic and clinical research, and human gene therapy.
Collapse
Affiliation(s)
- Charles N de Leeuw
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Frank M Dyka
- Department of Ophthalmology, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Sanford L Boye
- Department of Ophthalmology, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Stéphanie Laprise
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Michelle Zhou
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Alice Y Chou
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Lisa Borretta
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Simone C McInerny
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Kathleen G Banks
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Elodie Portales-Casamar
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Magdalena I Swanson
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Cletus A D’Souza
- Canada’s Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, British Columbia, Canada
| | - Shannon E Boye
- Department of Ophthalmology, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Steven JM Jones
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
- Canada’s Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, British Columbia, Canada
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Robert A Holt
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
- Canada’s Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, British Columbia, Canada
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
- Department of Psychiatry, University of British Columbia, Vancouver, British Columbia, Canada
| | - Daniel Goldowitz
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
| | - William W Hauswirth
- Department of Ophthalmology, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Wyeth W Wasserman
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Elizabeth M Simpson
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Psychiatry, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
81
|
Lee HS, McCarty JH. Inducible gene deletion in glial cells to study angiogenesis in the central nervous system. Methods Mol Biol 2014; 1135:261-74. [PMID: 24510871 DOI: 10.1007/978-1-4939-0320-7_22] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Most organs and tissues of the vertebrate body harbor elaborate network of blood vessels with diverse functions that are determined, in part, by cues within the local environment (Warren and Iruela-Arispe, Curr Opin Hematol 17:213-218, 2010). How vascular endothelial cells decipher these cues to promote normal blood vessel development and physiology remains largely uncharacterized. In this review, we will focus on genetic strategies to analyze glial regulation of blood vessel growth and sprouting within the microenvironment of the retina, a component of the central nervous system (CNS) that contains a complex web of blood vessels with many unique features, including a blood-retinal barrier (Abbott et al., Nat Rev Neurosci 7:41-53, 2006). Blood vessels promote retinal development and homeostasis and alterations in vascular functions can lead to various developmental and adult-onset retinal pathologies (Fruttiger, Angiogenesis 10:77-88, 2007). How glial cells control retinal endothelial cell growth and sprouting remains largely uncharacterized. We will detail methodologies involving inducible Cre-lox technologies to acutely ablate genes of interest in CNS glial cells. These methods allow for precise spatial and temporal regulation of gene expression to study how glial cells in the retinal microenvironment control angiogenesis and blood-retinal barrier development.
Collapse
Affiliation(s)
- Hye Shin Lee
- Department of Cancer Biology, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | | |
Collapse
|
82
|
Peterson SE, Loring JF. Genomic instability in pluripotent stem cells: implications for clinical applications. J Biol Chem 2013; 289:4578-84. [PMID: 24362040 DOI: 10.1074/jbc.r113.516419] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Human pluripotent stem cells (hPSCs) are known to acquire genomic changes as they proliferate and differentiate. Despite concerns that these changes will compromise the safety of hPSC-derived cell therapy, there is currently scant evidence linking the known hPSC genomic abnormalities with malignancy. For the successful use of hPSCs for clinical applications, we will need to learn to distinguish between innocuous genomic aberrations and those that may cause tumors. To minimize any effects of acquired mutations on cell therapy, we strongly recommend that cells destined for transplant be monitored throughout their preparation using a high-resolution method such as SNP genotyping.
Collapse
Affiliation(s)
- Suzanne E Peterson
- From the Department of Chemical Physiology and Center for Regenerative Medicine, The Scripps Research Institute, La Jolla, California 92037
| | | |
Collapse
|
83
|
Zhang J, Dublin P, Griemsmann S, Klein A, Brehm R, Bedner P, Fleischmann BK, Steinhäuser C, Theis M. Germ-line recombination activity of the widely used hGFAP-Cre and nestin-Cre transgenes. PLoS One 2013; 8:e82818. [PMID: 24349371 PMCID: PMC3857304 DOI: 10.1371/journal.pone.0082818] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2012] [Accepted: 11/07/2013] [Indexed: 12/30/2022] Open
Abstract
Herein we demonstrate with PCR, immunodetection and reporter gene approaches that the widely used human Glial Fibrillary Acidic Protein (hGFAP)-Cre transgene exhibits spontaneous germ-line recombination activity in leading to deletion in brain, heart and tail tissue with high frequency. The ectopic activity of hGFAP-Cre requires a rigorous control. We likewise observed that a second widely used nestin-Cre transgene shows germ-line deletion. Here we describe procedures to identify mice with germ-line recombination mediated by the hGFAP-Cre and nestin-Cre transgenes. Such control is essential to avoid pleiotropic effects due to germ-line deletion of loxP-flanked target genes and to maintain the CNS-restricted deletion status in transgenic mouse colonies.
Collapse
Affiliation(s)
- Jiong Zhang
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany
| | - Pavel Dublin
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany
| | - Stephanie Griemsmann
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany
| | - Alexandra Klein
- Institute of Physiology I, Medical Faculty, Life and Brain Center, Bonn, Germany
| | - Ralph Brehm
- Anatomisches Institut, Tierärztliche Hochschule Hannover, Hannover, Germany
| | - Peter Bedner
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany
| | - Bernd K. Fleischmann
- Institute of Physiology I, Medical Faculty, Life and Brain Center, Bonn, Germany
| | - Christian Steinhäuser
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany
- * E-mail: (MT); (CS)
| | - Martin Theis
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany
- * E-mail: (MT); (CS)
| |
Collapse
|
84
|
Bersell K, Choudhury S, Mollova M, Polizzotti BD, Ganapathy B, Walsh S, Wadugu B, Arab S, Kühn B. Moderate and high amounts of tamoxifen in αMHC-MerCreMer mice induce a DNA damage response, leading to heart failure and death. Dis Model Mech 2013; 6:1459-69. [PMID: 23929941 PMCID: PMC3820268 DOI: 10.1242/dmm.010447] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Numerous mouse models have utilized Cre-loxP technology to modify gene expression. Adverse effects of Cre recombinase activity have been reported, including in the heart. However, the mechanisms associated with cardiac Cre toxicity are largely unknown. Here, we show that expression of Cre in cardiomyocytes induces a DNA damage response, resulting in cardiomyocyte apoptosis, cardiac fibrosis and cardiac dysfunction. In an effort to increase the recombination efficiency of a widely used tamoxifen-sensitive Cre transgene under control of the α-myosin-heavy-chain promoter (αMHC-MerCreMer), we observed myocardial dysfunction and decreased survival, which were dependent on the dose of tamoxifen injected. After excluding a Cre-independent contribution by tamoxifen, we found that Cre induced myocardial fibrosis, activation of pro-fibrotic genes and cardiomyocyte apoptosis. Examination of the molecular mechanisms showed activation of DNA damage response signaling and p53 stabilization in the absence of loxP sites, suggesting that Cre induced illegitimate DNA breaks. Cardiomyocyte apoptosis was also induced by expressing Cre using adenoviral transduction, indicating that the effect was not dependent on genomic integration of the transgene. Cre-mediated homologous recombination at loxP sites was dose-dependent and had a ceiling effect at ∼80% of cardiomyocytes showing recombination. By titrating the amount of tamoxifen to maximize recombination while minimizing animal lethality, we determined that 30 μg tamoxifen/g body weight/day injected on three consecutive days is the optimal condition for the αMHC-MerCreMer system to induce recombination in the Rosa26-lacZ strain. Our results further highlight the importance of experimental design, including the use of appropriate genetic controls for Cre expression.
Collapse
Affiliation(s)
- Kevin Bersell
- Department of Cardiology, Boston Children's Hospital, MA 02115, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
85
|
Abstract
Nestin-Cre mice have a significant metabolic phenotype that is hard to discern from current literature. Indeed, the Cre-lox system has numerous problems that can affect physiological parameters, and these are missed when the correct control strains are not used. Despite the increasing use of the Cre-lox system, these issues were not visible to the scientific community previously and may have affected published work. This makes it important to highlight the issues and raise awareness of the pitfalls of the Cre-lox system. Therefore, this perspective will discuss the impact of CNS and peripheral "off-target" Cre recombination on metabolic systems and describe the development of new approaches to obviate the difficulties.
Collapse
Affiliation(s)
- Erika Harno
- Neuroscience Research Group, Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, UK
| | | | | |
Collapse
|
86
|
Karaca M, Maechler P. Development of Mice with Brain-Specific Deletion of Floxed Glud1 (Glutamate Dehydrogenase 1) Using Cre Recombinase Driven by the Nestin Promoter. Neurochem Res 2013; 39:456-9. [DOI: 10.1007/s11064-013-1041-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Revised: 04/08/2013] [Accepted: 04/09/2013] [Indexed: 12/20/2022]
|
87
|
Non-cell-autonomous tumor suppression by p53. Cell 2013; 153:449-60. [PMID: 23562644 PMCID: PMC3702034 DOI: 10.1016/j.cell.2013.03.020] [Citation(s) in RCA: 622] [Impact Index Per Article: 51.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2012] [Revised: 01/16/2013] [Accepted: 03/14/2013] [Indexed: 12/15/2022]
Abstract
The p53 tumor suppressor can restrict malignant transformation by triggering cell-autonomous programs of cell-cycle arrest or apoptosis. p53 also promotes cellular senescence, a tumor-suppressive program that involves stable cell-cycle arrest and secretion of factors that modify the tissue microenvironment. In the presence of chronic liver damage, we show that ablation of a p53-dependent senescence program in hepatic stellate cells increases liver fibrosis and cirrhosis associated with reduced survival and enhances the transformation of adjacent epithelial cells into hepatocellular carcinoma. p53-expressing senescent stellate cells release factors that skew macrophage polarization toward a tumor-inhibiting M1-state capable of attacking senescent cells in culture, whereas proliferating p53-deficient stellate cells secrete factors that stimulate polarization of macrophages into a tumor-promoting M2-state and enhance the proliferation of premalignant cells. Hence, p53 can act non-cell autonomously to suppress tumorigenesis by promoting an antitumor microenvironment, in part, through secreted factors that modulate macrophage function.
Collapse
|
88
|
Inactivation of Socs3 in the hypothalamus enhances the hindbrain response to endogenous satiety signals via oxytocin signaling. J Neurosci 2013. [PMID: 23197703 DOI: 10.1523/jneurosci.1669-12.2012] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Leptin is an adipocyte-derived hormone that controls energy balance by acting primarily in the CNS, but its action is lost in common forms of obesity due to central leptin resistance. One potential mechanism for such leptin resistance is an increased hypothalamic expression of Suppressor of cytokine signaling 3 (Socs3), a feedback inhibitor of the Jak-Stat pathway that prevents Stat3 activation. Ample studies have confirmed the important role of Socs3 in leptin resistance and obesity. However, the degree to which Socs3 participates in the regulation of energy homeostasis in nonobese conditions remains largely undetermined. In this study, using adult mice maintained under standard diet, we demonstrate that Socs3 deficiency in the mediobasal hypothalamus (MBH) reduces food intake, protects against body weight gain, and limits adiposity, suggesting that Socs3 is necessary for normal body weight maintenance. Mechanistically, MBH Socs3-deficient mice display increased hindbrain sensitivity to endogenous, meal-related satiety signals, mediated by oxytocin signaling. Thus, oxytocin signaling likely mediates the effect of hypothalamic leptin on satiety circuits of the caudal brainstem. This provides an anatomical substrate for the effect of leptin on meal size, and more generally, a mechanism for how the brain controls short-term food intake as a function of the energetic stores available in the organism to maintain energy homeostasis. Any dysfunction in this pathway could potentially lead to overeating and obesity.
Collapse
|
89
|
Takeda H, Koso H, Tessarollo L, Copeland NG, Jenkins NA. Musashi1-CreER(T2) : a new cre line for conditional mutagenesis in neural stem cells. Genesis 2013; 51:128-34. [PMID: 23132814 PMCID: PMC3577968 DOI: 10.1002/dvg.22357] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2012] [Revised: 10/25/2012] [Accepted: 10/28/2012] [Indexed: 11/12/2022]
Abstract
The RNA-binding protein Musashi1 (Msi1) is one of two mammalian homologues of Drosophila Musashi, which is required for the asymmetric cell division of sensory organ precursor cells. In the mouse central nervous system (CNS), Msi1 is preferentially expressed in mitotically active progenitor cells in the ventricular zone (VZ) of the neural tube during embryonic development and in the subventricular zone (SVZ) of the postnatal brain. Previous studies showed that cells in the SVZ can contribute to long-term neurogenesis in the olfactory bulb (OB), but it remains unclear whether Msi1-expressing cells have self-renewing potential and can contribute to neurogenesis in the adult. Here, we describe the generation of Msi1-CreER(T2) knock-in mice and show by cell lineage tracing that Msi1-CreER(T2) -expressing cells mark neural stem cells (NSCs) in both the embryonic and adult brain. Msi1-CreER(T2) mice thus represent a new tool in our arsenal for genetically manipulating NSCs, which will be essential for understanding the molecular mechanisms underlying neural development.
Collapse
Affiliation(s)
- Haruna Takeda
- Division of Genomics and Genetics, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore 138673
- Department of Microbiology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Hideto Koso
- Division of Genomics and Genetics, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore 138673
- Division of Molecular and Developmental Biology, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639, Japan
| | - Lino Tessarollo
- Neural Development Group, Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Frederick, Maryland, 21702, USA
| | - Neal G. Copeland
- Division of Genomics and Genetics, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore 138673
- Cancer Research Program, The Methodist Hospital Research Institute, Houston, TX 77030, USA
| | - Nancy A. Jenkins
- Division of Genomics and Genetics, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore 138673
- Cancer Research Program, The Methodist Hospital Research Institute, Houston, TX 77030, USA
| |
Collapse
|
90
|
Karimova M, Abi-Ghanem J, Berger N, Surendranath V, Pisabarro MT, Buchholz F. Vika/vox, a novel efficient and specific Cre/loxP-like site-specific recombination system. Nucleic Acids Res 2012; 41:e37. [PMID: 23143104 PMCID: PMC3553980 DOI: 10.1093/nar/gks1037] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Targeted genome engineering has become an important research area for diverse disciplines, with site-specific recombinases (SSRs) being among the most popular genome engineering tools. Their ability to trigger excision, integration, inversion and translocation has made SSRs an invaluable tool to manipulate DNA in vitro and in vivo. However, sophisticated strategies that combine different SSR systems are ever increasing. Hence, the demand for additional precise and efficient recombinases is dictated by the increasing complexity of the genetic studies. Here, we describe a novel site-specific recombination system designated Vika/vox. Vika originates from a degenerate bacteriophage of Vibrio coralliilyticus and shares low sequence similarity to other tyrosine recombinases, but functionally carries out a similar type of reaction. We demonstrate that Vika is highly specific in catalyzing vox recombination without recombining target sites from other SSR systems. We also compare the recombination activity of Vika/vox with other SSR systems, providing a guideline for deciding on the most suitable enzyme for a particular application and demonstrate that Vika expression does not cause cytotoxicity in mammalian cells. Our results show that Vika/vox is a novel powerful and safe instrument in the 'genetic toolbox' that can be used alone or in combination with other SSRs in heterologous hosts.
Collapse
Affiliation(s)
- Madina Karimova
- Medical Systems Biology, Medical Faculty and University Hospital Carl Gustav Carus, University of Technology, Fetscherstrasse 74, 01307 Dresden, Germany
| | | | | | | | | | | |
Collapse
|
91
|
Strain background influences neurotoxicity and behavioral abnormalities in mice expressing the tetracycline transactivator. J Neurosci 2012; 32:10574-86. [PMID: 22855807 DOI: 10.1523/jneurosci.0893-12.2012] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The tet-off system has been widely used to create transgenic models of neurological disorders including Alzheimer's, Parkinson's, Huntington's, and prion disease. The utility of this system lies in the assumption that the tetracycline transactivator (TTA) acts as an inert control element and does not contribute to phenotypes under study. Here we report that neuronal expression of TTA can affect hippocampal cytoarchitecture and behavior in a strain-dependent manner. While studying neurodegeneration in two tet-off Alzheimer's disease models, we unexpectedly discovered neuronal loss within the dentate gyrus of single transgenic TTA controls. Granule neurons appeared most sensitive to TTA exposure during postnatal development, and doxycycline treatment during this period was neuroprotective. TTA-induced degeneration could be rescued by moving the transgene onto a congenic C57BL/6J background and recurred on reintroduction of either CBA or C3H/He backgrounds. Quantitative trait analysis of B6C3 F2 TTA mice identified a region on Chromosome 14 that contains a major modifier of the neurodegenerative phenotype. Although B6 mice were resistant to degeneration, they were not ideal for cognitive testing. F1 offspring of TTA C57BL/6J and 129X1/SvJ, FVB/NJ, or DBA/1J showed improved spatial learning, but TTA expression caused subtle differences in contextual fear conditioning on two of these backgrounds, indicating that strain and genotype can interact independently under different behavioral settings. All model systems have limitations that should be recognized and mitigated where possible; our findings stress the importance of mapping the effects caused by TTA alone when working with tet-off models.
Collapse
|
92
|
Murray SA, Eppig JT, Smedley D, Simpson EM, Rosenthal N. Beyond knockouts: cre resources for conditional mutagenesis. Mamm Genome 2012; 23:587-99. [PMID: 22926223 PMCID: PMC3655717 DOI: 10.1007/s00335-012-9430-2] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2012] [Accepted: 08/07/2012] [Indexed: 12/12/2022]
Abstract
With the effort of the International Phenotyping Consortium to produce thousands of strains with conditional potential gathering steam, there is growing recognition that it must be supported by a rich toolbox of cre driver strains. The approaches to build cre strains have evolved in both sophistication and reliability, replacing first-generation strains with tools that can target individual cell populations with incredible precision and specificity. The modest set of cre drivers generated by individual labs over the past 15+ years is now growing rapidly, thanks to a number of large-scale projects to produce new cre strains for the community. The power of this growing resource, however, depends upon the proper deep characterization of strain function, as even the best designed strain can display a variety of undesirable features that must be considered in experimental design. This must be coupled with the parallel development of informatics tools to provide functional data to the user and facilitated access to the strains through public repositories. We discuss the current progress on all of these fronts and the challenges that remain to ensure the scientific community can capitalize on the tremendous number of mouse resources at their disposal.
Collapse
Affiliation(s)
- Stephen A Murray
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA.
| | | | | | | | | |
Collapse
|
93
|
Burrill DR, Inniss MC, Boyle PM, Silver PA. Synthetic memory circuits for tracking human cell fate. Genes Dev 2012; 26:1486-97. [PMID: 22751502 DOI: 10.1101/gad.189035.112] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
A variety of biological phenomena, from disease progression to stem cell differentiation, are typified by a prolonged cellular response to a transient environmental cue. While biologically relevant, heterogeneity in these long-term responses is difficult to assess at the population level, necessitating the development of biological tools to track cell fate within subpopulations. Here we present a novel synthetic biology approach for identifying and tracking mammalian cell subpopulations. We constructed three genomically integrated circuits that use bistable autoregulatory transcriptional feedback to retain memory of exposure to brief stimuli. These "memory devices" are used to isolate and track the progeny of cells that responded differentially to doxycycline, hypoxia, or DNA-damaging agents. Following hypoxic or ultraviolet radiation exposure, strongly responding cells activate the memory device and exhibit changes in gene expression, growth rates, and viability for multiple generations after the initial stimulus. Taken together, these results indicate that a heritable memory of hypoxia and DNA damage exists in subpopulations that differ in long-term cell behavior.
Collapse
Affiliation(s)
- Devin R Burrill
- Department of Systems Biology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | | | |
Collapse
|
94
|
Cell lineage tracing techniques for the study of brain development and regeneration. Int J Dev Neurosci 2012; 30:560-9. [PMID: 22944528 DOI: 10.1016/j.ijdevneu.2012.08.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2012] [Revised: 08/12/2012] [Accepted: 08/12/2012] [Indexed: 11/22/2022] Open
Abstract
Characterization of the means by which cells are generated and organized to make an organ as complex as the brain is a formidable task. Understanding how adult stem cells give rise to progeny that integrate into the existing structures during regeneration or in response to injury is equally challenging. Lineage tracing techniques are essential to studying cell behaviors such as proliferation, migration and differentiation, since they allow stem or precursor cells to be marked and their descendants followed and characterized over time. Here, we describe some of the key lineage tracing techniques available to date, highlighting advantages and drawbacks and focusing on their application in neural fate mapping. The more traditional methods are now joined by exciting new approaches to provide a vast array of tools at the disposal of neurobiologists.
Collapse
|
95
|
A role for presenilins in autophagy revisited: normal acidification of lysosomes in cells lacking PSEN1 and PSEN2. J Neurosci 2012; 32:8633-48. [PMID: 22723704 DOI: 10.1523/jneurosci.0556-12.2012] [Citation(s) in RCA: 98] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Presenilins 1 and 2 (PS1 and PS2) are the catalytic subunits of the γ-secretase complex, and genes encoding mutant PS1 and PS2 variants cause familial forms of Alzheimer's disease. Lee et al. (2010) recently reported that loss of PS1 activity lead to impairments in autophagosomal function as a consequence of lysosomal alkalinization, caused by failed maturation of the proton translocating V0a1 subunit of the vacuolar (H+)-ATPase and targeting to the lysosome. We have reexamined these issues in mammalian cells and in brains of mice lacking PS (PScdko) and have been unable to find evidence that the turnover of autophagic substrates, vesicle pH, V0a1 maturation, or lysosome function is altered compared with wild-type counterparts. Collectively, our studies fail to document a role for presenilins in regulating cellular autophagosomal function. On the other hand, our transcriptome studies of PScdko mouse brains reveal, for the first time, a role for PS in regulating lysosomal biogenesis.
Collapse
|
96
|
Thanos A, Morizane Y, Murakami Y, Giani A, Mantopoulos D, Kayama M, Roh MI, Michaud N, Pawlyk B, Sandberg M, Young LH, Miller JW, Vavvas DG. Evidence for baseline retinal pigment epithelium pathology in the Trp1-Cre mouse. THE AMERICAN JOURNAL OF PATHOLOGY 2012; 180:1917-27. [PMID: 22429967 DOI: 10.1016/j.ajpath.2012.01.017] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2011] [Revised: 12/28/2011] [Accepted: 01/20/2012] [Indexed: 01/14/2023]
Abstract
The increasing popularity of the Cre/loxP recombination system has led to the generation of numerous transgenic mouse lines in which Cre recombinase is expressed under the control of organ- or cell-specific promoters. Alterations in retinal pigment epithelium (RPE), a multifunctional cell monolayer that separates the retinal photoreceptors from the choroid, are prevalent in the pathogenesis of a number of ocular disorders, including age-related macular degeneration. To date, six transgenic mouse lines have been developed that target Cre to the RPE under the control of various gene promoters. However, multiple lines of evidence indicate that high levels of Cre expression can be toxic to mammalian cells. In this study, we report that in the Trp1-Cre mouse, a commonly used transgenic Cre strain for RPE gene function studies, Cre recombinase expression alone leads to RPE dysfunction and concomitant disorganization of RPE layer morphology, large areas of RPE atrophy, retinal photoreceptor dysfunction, and microglial cell activation in the affected areas. The phenotype described herein is similar to previously published reports of conditional gene knockouts that used the Trp1-Cre mouse, suggesting that Cre toxicity alone could account for some of the reported phenotypes and highlighting the importance of the inclusion of Cre-expressing mice as controls in conditional gene targeting studies.
Collapse
Affiliation(s)
- Aristomenis Thanos
- Retina Service, Angiogenesis Laboratory, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts 02114, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
97
|
[Progress in Cre/lox site-specific recombination system in higher eukaryotes]. YI CHUAN = HEREDITAS 2012; 34:177-89. [PMID: 22382059 DOI: 10.3724/sp.j.1005.2012.00177] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Cre/lox system derived from P1 bacteriaphage can quickly and effectively achieve gene insertion, deletion, replacement, and inversion by means of site-specific recombination. As one of the most important tools for gene targeting at present, Cre/lox system has been widely used in Arabidopsis thaliana, Oryza sativa L., Mus musculus, Drosophila melanogaster, Danio rerio, and other higher eukaryotic organisms. This review roundly described the basic profile of Cre/lox system, and its application in higher eukaryotes. In addition, we also discussed the main problems and developmental trend of the Cre/lox system in this review, which can be a good reference for using Cre/lox system to realize the gene manipulations of the different high eukaryotic organisms.
Collapse
|
98
|
Transgenic techniques for cell ablation or molecular deletion to investigate functions of astrocytes and other GFAP-expressing cell types. Methods Mol Biol 2012; 814:531-44. [PMID: 22144330 DOI: 10.1007/978-1-61779-452-0_35] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Genetic tools are enabling the molecular dissection of the functions and mechanisms of many biological processes. Transgenic manipulations provide powerful tools with which to test hypotheses regarding functions of specific cell types and molecules in vivo in combination with different types of experimental models. Various techniques are available that can target genetic manipulations specifically to astrocytes and that are enabling the molecular dissection of astrocyte biology in vivo. This article summarizes procedures and experience from our laboratory using transgenic strategies that enable either the ablation of proliferating astrocytes and related cells, or the deletion of specific molecules selectively from astrocytes, to study the functions of astrocytes and related cell types in health and disease in vivo using different experimental mouse models.
Collapse
|
99
|
Abstract
Astrocytes communicate with the vascular endothelium via direct cell-cell contacts as well as a variety of secreted growth factors and extracellular matrix (ECM) proteins. Integrins are heterodimeric cell surface receptors for ECM protein ligands, and many integrin subunits are expressed in astrocytes. Here, we will discuss gene deletion strategies in mice that have deciphered functions for specific integrins in astrocyte-endothelial cell adhesion and signaling. Specifically, we will detail how Cre-lox molecular genetic techniques have revealed important roles for integrin αvβ8 in regulating cerebral blood vessel development and homeostasis. First, we will detail how to generate Cre-lox mutant mouse models that our group and others have used to study αvβ8 integrin in embryonic astroglial progenitors and postnatal astrocytes. Second, we will discuss how viral-delivered Cre can be used to acutely delete integrin genes in astrocytes within defined anatomic regions of the brain. Third, detailed in vivo methods to verify Cre-mediated gene recombination in astrocytes will be presented. Lastly, we will present one experimental strategy to determine how integrin gene deletion affects astrocyte-endothelial cell coupling in the CNS. While this review focuses on the generation and characterization of mice lacking αvβ8 integrin, these experimental strategies can be expanded to analyze other cell adhesion and signaling genes important for astroglial-mediated regulation of blood vessel development and homeostasis.
Collapse
Affiliation(s)
- Aaron K Mobley
- Department of Cancer Biology, University of Texas M. D. Anderson Cancer Center, Houston, TX, USA
| | | |
Collapse
|
100
|
Davey RA, Clarke MV, Sastra S, Skinner JP, Chiang C, Anderson PH, Zajac JD. Decreased body weight in young Osterix-Cre transgenic mice results in delayed cortical bone expansion and accrual. Transgenic Res 2011; 21:885-93. [PMID: 22160436 DOI: 10.1007/s11248-011-9581-z] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2011] [Accepted: 11/30/2011] [Indexed: 12/18/2022]
Abstract
Conditional gene inactivation using the Cre/loxP system has lead to significant advances in our understanding of the function of genes in a wide range of disciplines. It is becoming increasingly apparent in the literature, that Cre transgenic mice may themselves have a phenotype. In the following study we describe the bone phenotype of a commonly used Cre transgenic mouse line to study osteoblasts, the Osx-GFP::Cre (Osx-Cre) mice. Cortical and trabecular bone parameters were determined in the femurs of Osx-Cre mice at 6 and 12 weeks of age by microtomography (μCT). At 6 weeks of age, Osx-Cre mice had reduced body weight by 22% (P < 0.0001) and delayed cortical bone expansion and accrual, characterized by decreases in periosteal circumference by 7% (P < 0.05) and cortical thickness by 11% (P < 0.01), compared to wild type controls. Importantly, the cortical bone phenotype of the skeletally immature Osx-Cre mice at 6 weeks of age could be accounted for by their low body weight. The delayed weight gain and cortical growth of Osx-Cre mice was overcome by 12 weeks of age, with no differences observed between Osx-Cre and wild type controls. In conclusion, Osx-Cre expressing mice display a delayed growth phenotype in the absence of doxycycline treatment, evidenced by decreased cortical bone expansion and accrual at 6 weeks of age, as an indirect result of decreased body weight. While this delay in growth is overcome by adulthood at 12 weeks of age, caution together with appropriate data analysis must be considered when assessing the experimental data from skeletally immature Cre/loxP knockout mice generated using the Osx-Cre mouse line to avoid misinterpretation.
Collapse
Affiliation(s)
- Rachel A Davey
- Department of Medicine, Austin Health, University of Melbourne, Studley Road, Heidelberg, VIC 3084, Australia.
| | | | | | | | | | | | | |
Collapse
|