1
|
Kim J, Vanrobaeys Y, Davatolhagh MF, Kelvington B, Chatterjee S, Ferri SL, Angelakos C, Mills AA, Fuccillo MV, Nickl-Jockschat T, Abel T. A chromosome region linked to neurodevelopmental disorders acts in distinct neuronal circuits in males and females to control locomotor behavior. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.17.594746. [PMID: 38952795 PMCID: PMC11216371 DOI: 10.1101/2024.05.17.594746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/03/2024]
Abstract
Biological sex shapes the manifestation and progression of neurodevelopmental disorders (NDDs). These disorders often demonstrate male-specific vulnerabilities; however, the identification of underlying mechanisms remains a significant challenge in the field. Hemideletion of the 16p11.2 region (16p11.2 del/+) is associated with NDDs, and mice modeling 16p11.2 del/+ exhibit sex-specific striatum-related phenotypes relevant to NDDs. Striatal circuits, crucial for locomotor control, consist of two distinct pathways: the direct and indirect pathways originating from D1 dopamine receptor (D1R) and D2 dopamine receptor (D2R) expressing spiny projection neurons (SPNs), respectively. In this study, we define the impact of 16p11.2 del/+ on striatal circuits in male and female mice. Using snRNA-seq, we identify sex- and cell type-specific transcriptomic changes in the D1- and D2-SPNs of 16p11.2 del/+ mice, indicating distinct transcriptomic signatures in D1-SPNs and D2-SPNs in males and females, with a ∼5-fold greater impact in males. Further pathway analysis reveals differential gene expression changes in 16p11.2 del/+ male mice linked to synaptic plasticity in D1- and D2-SPNs and GABA signaling pathway changes in D1-SPNs. Consistent with our snRNA-seq study revealing changes in GABA signaling pathways, we observe distinct changes in miniature inhibitory postsynaptic currents (mIPSCs) in D1- and D2-SPNs from 16p11.2 del/+ male mice. Behaviorally, we utilize conditional genetic approaches to introduce the hemideletion selectively in either D1- or D2-SPNs and find that conditional hemideletion of genes in the 16p11.2 region in D2-SPNs causes hyperactivity in male mice, but hemideletion in D1-SPNs does not. Within the striatum, hemideletion of genes in D2-SPNs in the dorsal lateral striatum leads to hyperactivity in males, demonstrating the importance of this striatal region. Interestingly, conditional 16p11.2 del/+ within the cortex drives hyperactivity in both sexes. Our work reveals that a locus linked to NDDs acts in different striatal circuits, selectively impacting behavior in a sex- and cell type-specific manner, providing new insight into male vulnerability for NDDs. Highlights - 16p11.2 hemideletion (16p11.2 del/+) induces sex- and cell type-specific transcriptomic signatures in spiny projection neurons (SPNs). - Transcriptomic changes in GABA signaling in D1-SPNs align with changes in inhibitory synapse function. - 16p11.2 del/+ in D2-SPNs causes hyperactivity in males but not females. - 16p11.2 del/+ in D2-SPNs in the dorsal lateral striatum drives hyperactivity in males. - 16p11.2 del/+ in cortex drives hyperactivity in both sexes. Graphic abstract
Collapse
|
2
|
Lim R, Sugino T, Nolte H, Andrade J, Zimmermann B, Shi C, Doddaballapur A, Ong YT, Wilhelm K, Fasse JWD, Ernst A, Kaulich M, Husnjak K, Boettger T, Guenther S, Braun T, Krüger M, Benedito R, Dikic I, Potente M. Deubiquitinase USP10 regulates Notch signaling in the endothelium. SCIENCE (NEW YORK, N.Y.) 2019; 364:188-193. [PMID: 30975888 DOI: 10.1126/science.aat0778] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 03/14/2019] [Indexed: 12/15/2022]
Abstract
Notch signaling is a core patterning module for vascular morphogenesis that codetermines the sprouting behavior of endothelial cells (ECs). Tight quantitative and temporal control of Notch activity is essential for vascular development, yet the details of Notch regulation in ECs are incompletely understood. We found that ubiquitin-specific peptidase 10 (USP10) interacted with the NOTCH1 intracellular domain (NICD1) to slow the ubiquitin-dependent turnover of this short-lived form of the activated NOTCH1 receptor. Accordingly, inactivation of USP10 reduced NICD1 abundance and stability and diminished Notch-induced target gene expression in ECs. In mice, the loss of endothelial Usp10 increased vessel sprouting and partially restored the patterning defects caused by ectopic expression of NICD1. Thus, USP10 functions as an NICD1 deubiquitinase that fine-tunes endothelial Notch responses during angiogenic sprouting.
Collapse
Affiliation(s)
- R Lim
- Angiogenesis and Metabolism Laboratory, Max Planck Institute for Heart and Lung Research, D-61231 Bad Nauheim, Germany
| | - T Sugino
- Angiogenesis and Metabolism Laboratory, Max Planck Institute for Heart and Lung Research, D-61231 Bad Nauheim, Germany
| | - H Nolte
- Institute for Genetics and Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, D-50931 Cologne, Germany
| | - J Andrade
- Angiogenesis and Metabolism Laboratory, Max Planck Institute for Heart and Lung Research, D-61231 Bad Nauheim, Germany
| | - B Zimmermann
- Angiogenesis and Metabolism Laboratory, Max Planck Institute for Heart and Lung Research, D-61231 Bad Nauheim, Germany
| | - C Shi
- Angiogenesis and Metabolism Laboratory, Max Planck Institute for Heart and Lung Research, D-61231 Bad Nauheim, Germany
| | - A Doddaballapur
- Angiogenesis and Metabolism Laboratory, Max Planck Institute for Heart and Lung Research, D-61231 Bad Nauheim, Germany
| | - Y T Ong
- Angiogenesis and Metabolism Laboratory, Max Planck Institute for Heart and Lung Research, D-61231 Bad Nauheim, Germany
| | - K Wilhelm
- Angiogenesis and Metabolism Laboratory, Max Planck Institute for Heart and Lung Research, D-61231 Bad Nauheim, Germany
| | - J W D Fasse
- Angiogenesis and Metabolism Laboratory, Max Planck Institute for Heart and Lung Research, D-61231 Bad Nauheim, Germany
| | - A Ernst
- Institute of Biochemistry II, Faculty of Medicine, Goethe University, D-60590 Frankfurt am Main, Germany.,Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Project Group Translational Medicine and Pharmacology TMP, D-60590 Frankfurt am Main, Germany
| | - M Kaulich
- Institute of Biochemistry II, Faculty of Medicine, Goethe University, D-60590 Frankfurt am Main, Germany
| | - K Husnjak
- Institute of Biochemistry II, Faculty of Medicine, Goethe University, D-60590 Frankfurt am Main, Germany
| | - T Boettger
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, D-61231 Bad Nauheim, Germany
| | - S Guenther
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, D-61231 Bad Nauheim, Germany
| | - T Braun
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, D-61231 Bad Nauheim, Germany
| | - M Krüger
- Institute for Genetics and Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, D-50931 Cologne, Germany
| | - R Benedito
- Molecular Genetics of Angiogenesis Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain
| | - I Dikic
- Institute of Biochemistry II, Faculty of Medicine, Goethe University, D-60590 Frankfurt am Main, Germany.,Buchmann Institute for Molecular Life Sciences, Goethe University, D-60438 Frankfurt am Main, Germany
| | - M Potente
- Angiogenesis and Metabolism Laboratory, Max Planck Institute for Heart and Lung Research, D-61231 Bad Nauheim, Germany. .,DZHK (German Center for Cardiovascular Research), partner site Frankfurt Rhine-Main, D-13347 Berlin, Germany.,International Institute of Molecular and Cell Biology, 02-109 Warsaw, Poland
| |
Collapse
|
3
|
Gurumurthy CB, Lloyd KCK. Generating mouse models for biomedical research: technological advances. Dis Model Mech 2019; 12:dmm029462. [PMID: 30626588 PMCID: PMC6361157 DOI: 10.1242/dmm.029462] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Over the past decade, new methods and procedures have been developed to generate genetically engineered mouse models of human disease. This At a Glance article highlights several recent technical advances in mouse genome manipulation that have transformed our ability to manipulate and study gene expression in the mouse. We discuss how conventional gene targeting by homologous recombination in embryonic stem cells has given way to more refined methods that enable allele-specific manipulation in zygotes. We also highlight advances in the use of programmable endonucleases that have greatly increased the feasibility and ease of editing the mouse genome. Together, these and other technologies provide researchers with the molecular tools to functionally annotate the mouse genome with greater fidelity and specificity, as well as to generate new mouse models using faster, simpler and less costly techniques.
Collapse
Affiliation(s)
- Channabasavaiah B Gurumurthy
- Developmental Neuroscience, Munroe Meyer Institute for Genetics and Rehabilitation, University of Nebraska Medical Center, Omaha, NE 68106-5915, USA
- Mouse Genome Engineering Core Facility, Vice Chancellor for Research Office, University of Nebraska Medical Center, Omaha, NE 68106-5915, USA
| | - Kevin C Kent Lloyd
- Department of Surgery, School of Medicine, University of California, Davis, CA 95618, USA
- Mouse Biology Program, University of California, Davis, CA 95618, USA
| |
Collapse
|
4
|
Selective killing of spinal cord neural stem cells impairs locomotor recovery in a mouse model of spinal cord injury. J Neuroinflammation 2018; 15:58. [PMID: 29475438 PMCID: PMC5824446 DOI: 10.1186/s12974-018-1085-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 02/01/2018] [Indexed: 01/19/2023] Open
Abstract
Background Spinal cord injury (SCI) is a devastating condition mainly deriving from a traumatic damage of the spinal cord (SC). Immune cells and endogenous SC-neural stem cells (SC-NSCs) play a critical role in wound healing processes, although both are ineffective to completely restore tissue functioning. The role of SC-NSCs in SCI and, in particular, whether such cells can interplay with the immune response are poorly investigated issues, although mechanisms governing such interactions might open new avenues to develop novel therapeutic approaches. Methods We used two transgenic mouse lines to trace as well as to kill SC-NSCs in mice receiving SCI. We used Nestin CreERT2 mice to trace SC-NSCs descendants in the spinal cord of mice subjected to SCI. While mice carrying the suicide gene thymidine kinase (TK) along with the GFP reporter, under the control of the Nestin promoter regions (NestinTK mice) were used to label and selectively kill SC-NSCs. Results We found that SC-NSCs are capable to self-activate after SCI. In addition, a significant worsening of clinical and pathological features of SCI was observed in the NestinTK mice, upon selective ablation of SC-NSCs before the injury induction. Finally, mice lacking in SC-NSCs and receiving SCI displayed reduced levels of different neurotrophic factors in the SC and significantly higher number of M1-like myeloid cells. Conclusion Our data show that SC-NSCs undergo cell proliferation in response to traumatic spinal cord injury. Mice lacking SC-NSCs display overt microglia activation and exaggerate expression of pro-inflammatory cytokines. The absence of SC-NSCs impaired functional recovery as well as neuronal and oligodendrocyte cell survival. Collectively our data indicate that SC-NSCs can interact with microglia/macrophages modulating their activation/responses and that such interaction is importantly involved in mechanisms leading tissue recovery. Electronic supplementary material The online version of this article (10.1186/s12974-018-1085-9) contains supplementary material, which is available to authorized users.
Collapse
|
5
|
Pessentheiner AR, Huber K, Pelzmann HJ, Prokesch A, Radner FPW, Wolinski H, Lindroos-Christensen J, Hoefler G, Rülicke T, Birner-Gruenberger R, Bilban M, Bogner-Strauss JG. APMAP interacts with lysyl oxidase-like proteins, and disruption of Apmap leads to beneficial visceral adipose tissue expansion. FASEB J 2017; 31:4088-4103. [PMID: 28559441 PMCID: PMC5566180 DOI: 10.1096/fj.201601337r] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 05/15/2017] [Indexed: 01/18/2023]
Abstract
Adipocyte plasma membrane-associated protein (APMAP) has been described as an adipogenic factor in 3T3-L1 cells with unknown biochemical function; we therefore aimed to investigate the physiologic function of APMAP in vivo We generated Apmap-knockout mice and challenged them with an obesogenic diet to investigate their metabolic phenotype. We identified a novel truncated adipocyte-specific isoform of APMAP in mice that is produced by alternative transcription. Mice lacking the full-length APMAP protein, the only isoform that is expressed in humans, have an improved metabolic phenotype upon diet-induced obesity, indicated by enhanced insulin sensitivity, preserved glucose tolerance, increased respiratory exchange ratio, decreased inflammatory marker gene expression, and reduced adipocyte size. At the molecular level, APMAP interacts with the extracellular collagen cross-linking matrix proteins lysyl oxidase-like 1 and 3. On a high-fat diet, the expression of lysyl oxidase-like 1 and 3 is strongly decreased in Apmap-knockout mice, paralleled by reduced expression of profibrotic collagens and total collagen content in epididymal white adipose tissue, indicating decreased fibrotic potential. Together, our data suggest that APMAP is a novel regulator of extracellular matrix components, and establish that APMAP is a potential target to mitigate obesity-associated insulin resistance.-Pessentheiner, A. R., Huber, K., Pelzmann, H. J., Prokesch, A., Radner, F. P. W., Wolinski, H., Lindroos-Christensen, J., Hoefler, G., Rülicke, T., Birner-Gruenberger, R., Bilban, M., Bogner-Strauss, J. G. APMAP interacts with lysyl oxidase-like proteins, and disruption of Apmap leads to beneficial visceral adipose tissue expansion.
Collapse
Affiliation(s)
| | - Katharina Huber
- Institute of Biochemistry, Graz University of Technology, Graz, Austria
| | - Helmut J Pelzmann
- Institute of Biochemistry, Graz University of Technology, Graz, Austria
| | - Andreas Prokesch
- Institute of Cell Biology, Histology, and Embryology, Medical University of Graz, Graz, Austria
| | - Franz P W Radner
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Heimo Wolinski
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
- BioTechMed-Graz, University of Graz, Graz, Austria
| | | | - Gerald Hoefler
- Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Thomas Rülicke
- Institute of Laboratory Animal Science, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Ruth Birner-Gruenberger
- Institute of Pathology, Medical University of Graz, Graz, Austria
- Omics Center Graz, BioTechMed-Graz, University of Graz, Graz, Austria
| | - Martin Bilban
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | | |
Collapse
|
6
|
Schweisgut J, Schutt C, Wüst S, Wietelmann A, Ghesquière B, Carmeliet P, Dröse S, Korach KS, Braun T, Boettger T. Sex-specific, reciprocal regulation of ERα and miR-22 controls muscle lipid metabolism in male mice. EMBO J 2017; 36:1199-1214. [PMID: 28314781 DOI: 10.15252/embj.201695988] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 02/14/2017] [Accepted: 02/16/2017] [Indexed: 11/09/2022] Open
Abstract
Control of energy homeostasis and metabolism is achieved by integrating numerous pathways, and miRNAs are involved in this process by regulating expression of multiple target genes. However, relatively little is known about the posttranscriptional processing of miRNAs and a potential role for the precursors they derive from. Here, we demonstrate that mature miRNA-22 is more abundant in muscle from male mice relative to females and that this enables sex-specific regulation of muscular lipid metabolism and body weight by repressing estrogen receptor alpha (ERα) expression. We found that the ERα adjusts its own activity by preventing processing of miR-22 via direct binding to a conserved ERα-binding element within the primary miR-22 precursor. Mutation of the ERα binding site within the pri-miR-22 in vivo eliminates sex-specific differences in miR-22 expression. We reason that the resulting tissue selective negative feedback regulation is essential to establish sex-specific differences in muscle metabolism and body weight development.
Collapse
Affiliation(s)
- Judith Schweisgut
- Department of Cardiac Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Christian Schutt
- Department of Cardiac Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Stas Wüst
- Department of Cardiac Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Astrid Wietelmann
- MRI Service Group, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Bart Ghesquière
- Vesalius Research Center (VRC), VIB-KULeuven, Campus Gasthuisberg, Leuven, Belgium
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, Leuven, Belgium.,Department of Oncology, Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, University of Leuven, Leuven, Belgium
| | - Stefan Dröse
- Department of Anesthesiology, Intensive-Care Medicine and Pain Therapy, University Hospital Frankfurt, Frankfurt, Germany
| | - Kenneth S Korach
- Receptor Biology Group, Reproductive and Developmental Biology Laboratory, Research Triangle Park, NC, USA
| | - Thomas Braun
- Department of Cardiac Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Thomas Boettger
- Department of Cardiac Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| |
Collapse
|
7
|
Hekmatnejad B, Yu VWC, Addison W, Mandic V, Pellicelli M, Arabian A, St-Arnaud R. FIAT Deletion Increases Bone Mass But Does Not Prevent High-Fat-Diet-Induced Metabolic Complications. Endocrinology 2017; 158:264-276. [PMID: 27906582 PMCID: PMC5413077 DOI: 10.1210/en.2016-1867] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Accepted: 11/23/2016] [Indexed: 12/28/2022]
Abstract
Factor inhibiting activating transcription factor 4 (ATF4)-mediated transcription (FIAT) interacts with ATF4 to repress its transcriptional activity. We performed a phenotypic analysis of Fiat-deficient male mice (Fiat-/Y) at 8 and 16 weeks of age. Microcomputed tomography analysis of the distal femur demonstrated 46% and 13% age-dependent increases in trabecular bone volume and thickness, respectively, in Fiat-/Y mice. Cortical bone measurements at the femoral midshaft revealed a substantial increase in cortical thickness in older Fiat-/Y mice. Bone gain was related to increased mineral apposition rate and increased osteoblast function. Femoral stiffness and strength were substantially increased in Fiat-/Y compared with wild-type (WT) mice. We also investigated whether FIAT contributes to metabolic function. When fed standard mouse chow, Fiat-/Y animals were glucose-tolerant. However, when fed a high-fat diet (HFD) for 8 weeks, Fiat-/Y mice gained more weight than control mice, with a specific increase in white adipose tissue fat mass. The increase in fat mass was due to reduced energy expenditure, which correlated with reduced fatty acid oxidation and lipolysis in the adipose tissue of mutant mice. The expression of the Scd1 gene, involved in lipogenesis, was upregulated in the subcutaneous adipose tissue of Fiat-/Y mice. Moreover, HFD-fed Fiat-/Y mice exhibited increased circulating leptin and insulin levels relative to WT mice, demonstrating that endocrine abnormalities are associated with the disturbance in energy balance. We conclude that Fiat-/Y mice exhibited an anabolic bone phenotype but displayed increased susceptibility to developing metabolic-related disorders when consuming an HFD.
Collapse
Affiliation(s)
- Bahareh Hekmatnejad
- Research Centre, Shriners Hospital for Children-Canada, Montreal, Quebec H4A 0A9, Canada;
- Human Genetics,
| | - Vionnie W. C. Yu
- Research Centre, Shriners Hospital for Children-Canada, Montreal, Quebec H4A 0A9, Canada;
- Human Genetics,
| | - William Addison
- Research Centre, Shriners Hospital for Children-Canada, Montreal, Quebec H4A 0A9, Canada;
| | - Vice Mandic
- Research Centre, Shriners Hospital for Children-Canada, Montreal, Quebec H4A 0A9, Canada;
| | - Martin Pellicelli
- Research Centre, Shriners Hospital for Children-Canada, Montreal, Quebec H4A 0A9, Canada;
- Human Genetics,
| | - Alice Arabian
- Research Centre, Shriners Hospital for Children-Canada, Montreal, Quebec H4A 0A9, Canada;
| | - René St-Arnaud
- Research Centre, Shriners Hospital for Children-Canada, Montreal, Quebec H4A 0A9, Canada;
- Human Genetics,
- Surgery, and
- Medicine, McGill University, Montreal, Quebec H3A 2T5, Canada; and
- Research Institute, McGill University Health Centre, Montreal, Quebec H4A 3J1, Canada
| |
Collapse
|
8
|
Uchtmann K, Park ER, Bergsma A, Segula J, Edick MJ, Miranti CK. Homozygous loss of mouse tetraspanin CD82 enhances integrin αIIbβ3 expression and clot retraction in platelets. Exp Cell Res 2015; 339:261-9. [PMID: 26562164 DOI: 10.1016/j.yexcr.2015.11.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 11/03/2015] [Accepted: 11/07/2015] [Indexed: 11/15/2022]
Abstract
Integrin αIIbβ3 is critical for platelet-mediated blood clotting. Tetraspanins are well-established regulators of integrins and genetic loss of tetraspanin CD151 or TSSC6 in mice leads to increased bleeding due to inadequate integrin αIIbβ3 outside-in signaling. Conversely, mild but enhanced integrin αIIbβ3 activation and hyperaggregation is observed in CD9 and CD63 null mice respectively. CD82 is reportedly expressed in platelets; however its function is unknown. Using genetically engineered CD82 null mice, we investigated the role of the tetraspanin CD82 in platelet activation. Loss of CD82 resulted in reduced bleed times in vivo. CD82 was present on the surface of both human and mouse platelets, and its levels did not change upon platelet activation or degranulation. No differences in platelet activation, degranulation, or aggregation in response to ADP or collagen were detected in CD82 null mice. However, the kinetics of clot retraction was enhanced, which was intrinsic to the CD82-null platelets. Integrin αIIbβ3 surface expression was elevated on the platelets from CD82 null mice and they displayed enhanced adhesion and tyrosine kinase signaling on fibrinogen. This is the first report on CD82 function in platelets; which we found intrinsically modulates clot retraction, integrin αIIbβ3 expression, cell adhesion, and tyrosine signaling.
Collapse
Affiliation(s)
- Kristen Uchtmann
- Lab of Integrin Signaling, Van Andel Research Institute, Grand Rapids, MI 49503 United States
| | - Electa R Park
- Lab of Integrin Signaling, Van Andel Research Institute, Grand Rapids, MI 49503 United States
| | - Alexis Bergsma
- Lab of Integrin Signaling, Van Andel Research Institute, Grand Rapids, MI 49503 United States
| | - Justin Segula
- Lab of Integrin Signaling, Van Andel Research Institute, Grand Rapids, MI 49503 United States
| | - Mathew J Edick
- Lab of Integrin Signaling, Van Andel Research Institute, Grand Rapids, MI 49503 United States
| | - Cindy K Miranti
- Lab of Integrin Signaling, Van Andel Research Institute, Grand Rapids, MI 49503 United States.
| |
Collapse
|
9
|
Larroquette F, Seto L, Gaub PL, Kamal B, Wallis D, Larivière R, Vallée J, Robitaille R, Tsuda H. Vapb/Amyotrophic lateral sclerosis 8 knock-in mice display slowly progressive motor behavior defects accompanying ER stress and autophagic response. Hum Mol Genet 2015; 24:6515-29. [PMID: 26362257 DOI: 10.1093/hmg/ddv360] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Accepted: 09/01/2015] [Indexed: 12/12/2022] Open
Abstract
Missense mutations (P56S) in Vapb are associated with autosomal dominant motor neuron diseases: amyotrophic lateral sclerosis and lower motor neuron disease. Although transgenic mice overexpressing the mutant vesicle-associated membrane protein-associated protein B (VAPB) protein with neuron-specific promoters have provided some insight into the toxic properties of the mutant proteins, their role in pathogenesis remains unclear. To identify pathological defects in animals expressing the P56S mutant VAPB protein at physiological levels in the appropriate tissues, we have generated Vapb knock-in mice replacing wild-type Vapb gene with P56S mutant Vapb gene and analyzed the resulting pathological phenotypes. Heterozygous P56S Vapb knock-in mice show mild age-dependent defects in motor behaviors as characteristic features of the disease. The homozygous P56S Vapb knock-in mice show more severe defects compared with heterozygous mice reflecting the dominant and dose-dependent effects of P56S mutation. Significantly, the knock-in mice demonstrate accumulation of P56S VAPB protein and ubiquitinated proteins in cytoplasmic inclusions, selectively in motor neurons. The mutant mice demonstrate induction of ER stress and autophagic response in motor neurons before obvious onset of behavioral defects, suggesting that these cellular biological defects might contribute to the initiation of the disease. The P56S Vapb knock-in mice could be a valuable tool to gain a better understanding of the mechanisms by which the disease arises.
Collapse
Affiliation(s)
- Frédérique Larroquette
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada H3A 2B4
| | - Lesley Seto
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada H3A 2B4
| | - Perrine L Gaub
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada H3A 2B4
| | - Brishna Kamal
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada H3A 2B4
| | - Deeann Wallis
- Department of Biochemistry and Biophysics, Texas A&M University, Cypress, TX, USA and
| | - Roxanne Larivière
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada H3A 2B4
| | - Joanne Vallée
- Departement of Neurosciences, University of Montreal, Montreal, Quebec, Canada
| | - Richard Robitaille
- Departement of Neurosciences, University of Montreal, Montreal, Quebec, Canada
| | - Hiroshi Tsuda
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada H3A 2B4,
| |
Collapse
|
10
|
Besser J, Malan D, Wystub K, Bachmann A, Wietelmann A, Sasse P, Fleischmann BK, Braun T, Boettger T. MiRNA-1/133a clusters regulate adrenergic control of cardiac repolarization. PLoS One 2014; 9:e113449. [PMID: 25415383 PMCID: PMC4240597 DOI: 10.1371/journal.pone.0113449] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Accepted: 10/24/2014] [Indexed: 12/24/2022] Open
Abstract
The electrical properties of the heart are primarily determined by the activity of ion channels and the activity of these molecules is permanently modulated and adjusted to the physiological needs by adrenergic signaling. miRNAs are known to control the expression of many proteins and to fulfill distinct functions in the mammalian heart, though the in vivo effects of miRNAs on the electrical activity of the heart are poorly characterized. The miRNAs miR-1 and miR-133a are the most abundant miRNAs of the heart and are expressed from two miR-1/133a genomic clusters. Genetic modulation of miR-1/133a cluster expression without concomitant severe disturbance of general cardiomyocyte physiology revealed that these miRNA clusters govern cardiac muscle repolarization. Reduction of miR-1/133a dosage induced a longQT phenotype in mice especially at low heart rates. Longer action potentials in cardiomyocytes are caused by modulation of the impact of β-adrenergic signaling on the activity of the depolarizing L-type calcium channel. Pharmacological intervention to attenuate β-adrenergic signaling or L-type calcium channel activity in vivo abrogated the longQT phenotype that is caused by modulation of miR-1/133a activity. Thus, we identify the miR-1/133a miRNA clusters to be important to prevent a longQT-phenotype in the mammalian heart.
Collapse
Affiliation(s)
- Johannes Besser
- Department of Cardiac Development and Remodeling, Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Daniela Malan
- Institut für Physiologie I, Life & Brain Center, Universität Bonn, Bonn, Germany
| | - Katharina Wystub
- Department of Cardiac Development and Remodeling, Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Angela Bachmann
- Department of Cardiac Development and Remodeling, Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Astrid Wietelmann
- Department of Cardiac Development and Remodeling, Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Philipp Sasse
- Institut für Physiologie I, Life & Brain Center, Universität Bonn, Bonn, Germany
| | - Bernd K. Fleischmann
- Institut für Physiologie I, Life & Brain Center, Universität Bonn, Bonn, Germany
| | - Thomas Braun
- Department of Cardiac Development and Remodeling, Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany
- * E-mail: (TB); (TB)
| | - Thomas Boettger
- Department of Cardiac Development and Remodeling, Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany
- * E-mail: (TB); (TB)
| |
Collapse
|
11
|
Ryder E, Doe B, Gleeson D, Houghton R, Dalvi P, Grau E, Habib B, Miklejewska E, Newman S, Sethi D, Sinclair C, Vyas S, Wardle-Jones H, Bottomley J, Bussell J, Galli A, Salisbury J, Ramirez-Solis R. Rapid conversion of EUCOMM/KOMP-CSD alleles in mouse embryos using a cell-permeable Cre recombinase. Transgenic Res 2014; 23:177-85. [PMID: 24197666 PMCID: PMC3890051 DOI: 10.1007/s11248-013-9764-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Accepted: 10/22/2013] [Indexed: 10/31/2022]
Abstract
We describe here use of a cell-permeable Cre to efficiently convert the EUCOMM/KOMP-CSD tm1a allele to the tm1b form in preimplantation mouse embryos in a high-throughput manner, consistent with the requirements of the International Mouse Phenotyping Consortium-affiliated NIH KOMP2 project. This method results in rapid allele conversion and minimizes the use of experimental animals when compared to conventional Cre transgenic mouse breeding, resulting in a significant reduction in costs and time with increased welfare benefits.
Collapse
Affiliation(s)
- Edward Ryder
- The Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, CB10 1SA UK
| | - Brendan Doe
- The Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, CB10 1SA UK
| | - Diane Gleeson
- The Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, CB10 1SA UK
| | - Richard Houghton
- The Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, CB10 1SA UK
| | - Priya Dalvi
- The Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, CB10 1SA UK
| | - Evelyn Grau
- The Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, CB10 1SA UK
| | - Bishoy Habib
- The Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, CB10 1SA UK
| | | | - Stuart Newman
- The Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, CB10 1SA UK
| | - Debarati Sethi
- The Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, CB10 1SA UK
| | - Caroline Sinclair
- The Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, CB10 1SA UK
| | - Sapna Vyas
- The Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, CB10 1SA UK
| | | | | | - Joanna Bottomley
- The Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, CB10 1SA UK
| | - James Bussell
- The Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, CB10 1SA UK
| | - Antonella Galli
- The Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, CB10 1SA UK
| | - Jennifer Salisbury
- The Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, CB10 1SA UK
| | | |
Collapse
|
12
|
Hekmatnejad B, Mandic V, Yu VWC, Akhouayri O, Arabian A, St-Arnaud R. Altered gene dosage confirms the genetic interaction between FIAT and αNAC. Gene 2014; 538:328-33. [PMID: 24440290 DOI: 10.1016/j.gene.2014.01.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Accepted: 01/04/2014] [Indexed: 10/25/2022]
Abstract
Factor inhibiting ATF4-mediated transcription (FIAT) interacts with Nascent polypeptide associated complex and coregulator alpha (αNAC). In cultured osteoblastic cells, this interaction contributes to maximal FIAT-mediated inhibition of Osteocalcin (Ocn) gene transcription. We set out to demonstrate the physiological relevance of this interaction by altering gene dosage in compound Fiat and Naca (encoding αNAC) heterozygous mice. Compound Naca(+/-); Fiat(+/-) heterozygous animals were viable, developed normally, and exhibited no significant difference in body weight compared with control littermate genotypes. Animals with a single Fiat allele had reduced Fiat mRNA expression without changes in the expression of related family members. Expression of the osteocyte differentiation marker Dmp1 was elevated in compound heterozygotes. Static histomorphometry parameters were assessed at 8weeks of age using microcomputed tomography (μCT). Trabecular measurements were not different between genotypes. Cortical thickness and area were not affected by gene dosage, but we measured a significant increase in cortical porosity in compound heterozygous mice, without changes in biomechanical parameters. The bone phenotype of compound Naca(+/-); Fiat(+/-) heterozygotes confirms that FIAT and αNAC are part of a common genetic pathway and support a role for the FIAT/αNAC interaction in normal bone physiology.
Collapse
Affiliation(s)
- Bahareh Hekmatnejad
- Research Unit, Shriners Hospital for Children - Canada, Montreal, Quebec H3G 1A6, Canada; Department of Human Genetics, McGill University, Montreal, Quebec H3A 2T5, Canada
| | - Vice Mandic
- Research Unit, Shriners Hospital for Children - Canada, Montreal, Quebec H3G 1A6, Canada; Department of Human Genetics, McGill University, Montreal, Quebec H3A 2T5, Canada
| | - Vionnie W C Yu
- Research Unit, Shriners Hospital for Children - Canada, Montreal, Quebec H3G 1A6, Canada; Department of Human Genetics, McGill University, Montreal, Quebec H3A 2T5, Canada
| | - Omar Akhouayri
- Research Unit, Shriners Hospital for Children - Canada, Montreal, Quebec H3G 1A6, Canada; Department of Human Genetics, McGill University, Montreal, Quebec H3A 2T5, Canada
| | - Alice Arabian
- Research Unit, Shriners Hospital for Children - Canada, Montreal, Quebec H3G 1A6, Canada
| | - René St-Arnaud
- Research Unit, Shriners Hospital for Children - Canada, Montreal, Quebec H3G 1A6, Canada; Department of Human Genetics, McGill University, Montreal, Quebec H3A 2T5, Canada; Department of Surgery, McGill University, Montreal, Quebec H3A 2T5, Canada; Department of Medicine, McGill University, Montreal, Quebec H3A 2T5, Canada.
| |
Collapse
|
13
|
Chang C, Yeh S, Lee SO, Chang TM. Androgen receptor (AR) pathophysiological roles in androgen-related diseases in skin, bone/muscle, metabolic syndrome and neuron/immune systems: lessons learned from mice lacking AR in specific cells. NUCLEAR RECEPTOR SIGNALING 2013; 11:e001. [PMID: 24653668 PMCID: PMC3960937 DOI: 10.1621/nrs.11001] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/07/2013] [Accepted: 05/28/2013] [Indexed: 12/19/2022]
Abstract
The androgen receptor (AR) is expressed ubiquitously and plays a variety of roles in a vast number of physiological and pathophysiological processes. Recent studies of AR knockout (ARKO) mouse models, particularly the cell type- or tissue-specific ARKO models, have uncovered many AR cell type- or tissue-specific pathophysiological roles in mice, which otherwise would not be delineated from conventional castration and androgen insensitivity syndrome studies. Thus, the AR in various specific cell types plays pivotal roles in production and maturation of immune cells, bone mineralization, and muscle growth. In metabolism, the ARs in brain, particularly in the hypothalamus, and the liver appear to participate in regulation of insulin sensitivity and glucose homeostasis. The AR also plays key roles in cutaneous wound healing and cardiovascular diseases, including atherosclerosis and abdominal aortic aneurysm. This article will discuss the results obtained from the total, cell type-, or tissue-specific ARKO models. The understanding of AR cell type- or tissue-specific physiological and pathophysiological roles using these in
vivo mouse models will provide useful information in uncovering AR roles in humans and eventually help us to develop better therapies via targeting the AR or its downstream signaling molecules to combat androgen/AR-related diseases.
Collapse
Affiliation(s)
- Chawnshang Chang
- George Whipple Lab for Cancer Research, Departments of Pathology, Urology, Radiation Oncology, and the Wilmot Cancer Center, University of Rochester Medical Center, Rochester, New York, USA (CC, SY, SOL, T-MC) and Sex Hormone Research Center, China Medical University/Hospital, Taichung, Taiwan (CC)
| | - Shuyuan Yeh
- George Whipple Lab for Cancer Research, Departments of Pathology, Urology, Radiation Oncology, and the Wilmot Cancer Center, University of Rochester Medical Center, Rochester, New York, USA (CC, SY, SOL, T-MC) and Sex Hormone Research Center, China Medical University/Hospital, Taichung, Taiwan (CC)
| | - Soo Ok Lee
- George Whipple Lab for Cancer Research, Departments of Pathology, Urology, Radiation Oncology, and the Wilmot Cancer Center, University of Rochester Medical Center, Rochester, New York, USA (CC, SY, SOL, T-MC) and Sex Hormone Research Center, China Medical University/Hospital, Taichung, Taiwan (CC)
| | - Ta-Min Chang
- George Whipple Lab for Cancer Research, Departments of Pathology, Urology, Radiation Oncology, and the Wilmot Cancer Center, University of Rochester Medical Center, Rochester, New York, USA (CC, SY, SOL, T-MC) and Sex Hormone Research Center, China Medical University/Hospital, Taichung, Taiwan (CC)
| |
Collapse
|
14
|
Schönheit J, Kuhl C, Gebhardt M, Klett F, Riemke P, Scheller M, Huang G, Naumann R, Leutz A, Stocking C, Priller J, Andrade-Navarro M, Rosenbauer F. PU.1 Level-Directed Chromatin Structure Remodeling at the Irf8 Gene Drives Dendritic Cell Commitment. Cell Rep 2013; 3:1617-28. [DOI: 10.1016/j.celrep.2013.04.007] [Citation(s) in RCA: 95] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2012] [Revised: 11/23/2012] [Accepted: 04/08/2013] [Indexed: 01/22/2023] Open
|
15
|
Impaired in vitro erythropoiesis following deletion of the Scl (Tal1) +40 enhancer is largely compensated for in vivo despite a significant reduction in expression. Mol Cell Biol 2013; 33:1254-66. [PMID: 23319051 DOI: 10.1128/mcb.01525-12] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
The Scl (Tal1) gene encodes a helix-loop-helix transcription factor essential for hematopoietic stem cell and erythroid development. The Scl +40 enhancer is situated downstream of Map17, the 3' flanking gene of Scl, and is active in transgenic mice during primitive and definitive erythropoiesis. To analyze the in vivo function of the Scl +40 enhancer within the Scl/Map17 transcriptional domain, we deleted this element in the germ line. Scl(Δ40/Δ40) mice were viable with reduced numbers of erythroid CFU in both bone marrow and spleen yet displayed a normal response to stress hematopoiesis. Analysis of Scl(Δ40/Δ40) embryonic stem (ES) cells revealed impaired erythroid differentiation, which was accompanied by a failure to upregulate Scl when erythropoiesis was initiated. Map17 expression was also reduced in hematopoietic tissues and differentiating ES cells, and the Scl +40 element was able to enhance activity of the Map17 promoter. However, only Scl but not Map17 could rescue the Scl(Δ40/Δ40) ES phenotype. Together, these data demonstrate that the Scl +40 enhancer is an erythroid cell-specific enhancer that regulates the expression of both Scl and Map17. Moreover, deletion of the +40 enhancer causes a novel erythroid phenotype, which can be rescued by ectopic expression of Scl but not Map17.
Collapse
|
16
|
Butti E, Bacigaluppi M, Rossi S, Cambiaghi M, Bari M, Cebrian Silla A, Brambilla E, Musella A, De Ceglia R, Teneud L, De Chiara V, D'Adamo P, Garcia-Verdugo JM, Comi G, Muzio L, Quattrini A, Leocani L, Maccarrone M, Centonze D, Martino G. Subventricular zone neural progenitors protect striatal neurons from glutamatergic excitotoxicity. ACTA ACUST UNITED AC 2012; 135:3320-35. [PMID: 23008234 DOI: 10.1093/brain/aws194] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The functional significance of adult neural stem and progenitor cells in hippocampal-dependent learning and memory has been well documented. Although adult neural stem and progenitor cells in the subventricular zone are known to migrate to, maintain and reorganize the olfactory bulb, it is less clear whether they are functionally required for other processes. Using a conditional transgenic mouse model, selective ablation of adult neural stem and progenitor cells in the subventricular zone induced a dramatic increase in morbidity and mortality of central nervous system disorders characterized by excitotoxicity-induced cell death accompanied by reactive inflammation, such as 4-aminopyridine-induced epilepsy and ischaemic stroke. To test the role of subventricular zone adult neural stem and progenitor cells in protecting central nervous system tissue from glutamatergic excitotoxicity, neurophysiological recordings of spontaneous excitatory postsynaptic currents from single medium spiny striatal neurons were measured on acute brain slices. Indeed, lipopolysaccharide-stimulated, but not unstimulated, subventricular zone adult neural stem and progenitor cells reverted the increased frequency and duration of spontaneous excitatory postsynaptic currents by secreting the endocannabinod arachidonoyl ethanolamide, a molecule that regulates glutamatergic tone through type 1 cannabinoid receptor (CB(1)) binding. In vivo restoration of cannabinoid levels, either by administration of the type 1 cannabinoid receptor agonist HU210 or the inhibitor of the principal catabolic enzyme fatty acid amide hydrolase, URB597, completely reverted the increased morbidity and mortality of adult neural stem and progenitor cell-ablated mice suffering from epilepsy and ischaemic stroke. Our results provide the first evidence that adult neural stem and progenitor cells located within the subventricular zone exert an 'innate' homeostatic regulatory role by protecting striatal neurons from glutamate-mediated excitotoxicity.
Collapse
Affiliation(s)
- Erica Butti
- Neuroimmunology Unit, INSPE, Division of Neuroscience, San Raffaele Scientific Institute Via Olgettina 58, 20132 Milan, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Drp2 and periaxin form Cajal bands with dystroglycan but have distinct roles in Schwann cell growth. J Neurosci 2012; 32:9419-28. [PMID: 22764250 DOI: 10.1523/jneurosci.1220-12.2012] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Cajal bands are cytoplasmic channels flanked by appositions where the abaxonal surface of Schwann cell myelin apposes and adheres to the overlying plasma membrane. These appositions contain a dystroglycan complex that includes periaxin and dystrophin-related protein 2 (Drp2). Loss of periaxin disrupts appositions and Cajal bands in Schwann cells and causes a severe demyelinating neuropathy in mouse and human. Here, we investigated the role of mouse Drp2 in apposition assembly and Cajal band function and compared it with periaxin. We show that periaxin and Drp2 are not only both required to form appositions, but they must also interact. Periaxin-Drp2 interaction is also required for Drp2 phosphorylation, but phosphorylation is not required for the assembly of appositions. Drp2 loss causes corresponding increases in Dystrophin family members, utrophin and dystrophin Dp116, although dystroglycan remains unchanged. We also show that all dystroglycan complexes in Schwann cells use the uncleaved form of β-dystroglycan. Drp2-null Schwann cells have disrupted appositions and Cajal bands, and they undergo focal hypermyelination and concomitant demyelination. Nevertheless, they do not have the short internodal lengths and associated reduced nerve conduction velocity seen in the absence of periaxin, showing that periaxin regulates Schwann cell elongation independent of its role in the dystroglycan complex. We conclude that the primary role of the dystroglycan complex in appositions is to stabilize and limit the radial growth of myelin.
Collapse
|
18
|
Migdalska AM, van der Weyden L, Ismail O, Rust AG, Rashid M, White JK, Sánchez-Andrade G, Lupski JR, Logan DW, Arends MJ, Adams DJ. Generation of the Sotos syndrome deletion in mice. Mamm Genome 2012; 23:749-57. [PMID: 22926222 PMCID: PMC3510424 DOI: 10.1007/s00335-012-9416-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2012] [Accepted: 07/16/2012] [Indexed: 11/28/2022]
Abstract
Haploinsufficiency of the human 5q35 region spanning the NSD1 gene results in a rare genomic disorder known as Sotos syndrome (Sotos), with patients displaying a variety of clinical features, including pre- and postnatal overgrowth, intellectual disability, and urinary/renal abnormalities. We used chromosome engineering to generate a segmental monosomy, i.e., mice carrying a heterozygous 1.5-Mb deletion of 36 genes on mouse chromosome 13 (4732471D19Rik-B4galt7), syntenic with 5q35.2–q35.3 in humans (Df(13)Ms2Dja+/− mice). Surprisingly Df(13)Ms2Dja+/− mice were significantly smaller for their gestational age and also showed decreased postnatal growth, in contrast to Sotos patients. Df(13)Ms2Dja+/− mice did, however, display deficits in long-term memory retention and dilation of the pelvicalyceal system, which in part may model the learning difficulties and renal abnormalities observed in Sotos patients. Thus, haploinsufficiency of genes within the mouse 4732471D19Rik–B4galt7 deletion interval play important roles in growth, memory retention, and the development of the renal pelvicalyceal system.
Collapse
Affiliation(s)
- Anna M Migdalska
- Experimental Cancer Genetics, Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, CB10 1HH, UK
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Nijnik A, Clare S, Hale C, Chen J, Raisen C, Mottram L, Lucas M, Estabel J, Ryder E, Adissu H, Adams NC, Ramirez-Solis R, White JK, Steel KP, Dougan G, Hancock REW. The role of sphingosine-1-phosphate transporter Spns2 in immune system function. THE JOURNAL OF IMMUNOLOGY 2012; 189:102-11. [PMID: 22664872 DOI: 10.4049/jimmunol.1200282] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Sphingosine-1-phosphate (S1P) is lipid messenger involved in the regulation of embryonic development, immune system functions, and many other physiological processes. However, the mechanisms of S1P transport across cellular membranes remain poorly understood, with several ATP-binding cassette family members and the spinster 2 (Spns2) member of the major facilitator superfamily known to mediate S1P transport in cell culture. Spns2 was also shown to control S1P activities in zebrafish in vivo and to play a critical role in zebrafish cardiovascular development. However, the in vivo roles of Spns2 in mammals and its involvement in the different S1P-dependent physiological processes have not been investigated. In this study, we characterized Spns2-null mouse line carrying the Spns2(tm1a(KOMP)Wtsi) allele (Spns2(tm1a)). The Spns2(tm1a/tm1a) animals were viable, indicating a divergence in Spns2 function from its zebrafish ortholog. However, the immunological phenotype of the Spns2(tm1a/tm1a) mice closely mimicked the phenotypes of partial S1P deficiency and impaired S1P-dependent lymphocyte trafficking, with a depletion of lymphocytes in circulation, an increase in mature single-positive T cells in the thymus, and a selective reduction in mature B cells in the spleen and bone marrow. Spns2 activity in the nonhematopoietic cells was critical for normal lymphocyte development and localization. Overall, Spns2(tm1a/tm1a) resulted in impaired humoral immune responses to immunization. This study thus demonstrated a physiological role for Spns2 in mammalian immune system functions but not in cardiovascular development. Other components of the S1P signaling network are investigated as drug targets for immunosuppressive therapy, but the selective action of Spns2 may present an advantage in this regard.
Collapse
Affiliation(s)
- Anastasia Nijnik
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, United Kingdom.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Wilson CH, Crombie C, van der Weyden L, Poulogiannis G, Rust AG, Pardo M, Gracia T, Yu L, Choudhary J, Poulin GB, McIntyre RE, Winton DJ, March HN, Arends MJ, Fraser AG, Adams DJ. Nuclear receptor binding protein 1 regulates intestinal progenitor cell homeostasis and tumour formation. EMBO J 2012; 31:2486-97. [PMID: 22510880 PMCID: PMC3365428 DOI: 10.1038/emboj.2012.91] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2011] [Accepted: 03/06/2012] [Indexed: 01/02/2023] Open
Abstract
Genetic screens in simple model organisms have identified many of the key components of the conserved signal transduction pathways that are oncogenic when misregulated. Here, we identify H37N21.1 as a gene that regulates vulval induction in let-60(n1046gf), a strain with a gain-of-function mutation in the Caenorhabditis elegans Ras orthologue, and show that somatic deletion of Nrbp1, the mouse orthologue of this gene, results in an intestinal progenitor cell phenotype that leads to profound changes in the proliferation and differentiation of all intestinal cell lineages. We show that Nrbp1 interacts with key components of the ubiquitination machinery and that loss of Nrbp1 in the intestine results in the accumulation of Sall4, a key mediator of stem cell fate, and of Tsc22d2. We also reveal that somatic loss of Nrbp1 results in tumourigenesis, with haematological and intestinal tumours predominating, and that nuclear receptor binding protein 1 (NRBP1) is downregulated in a range of human tumours, where low expression correlates with a poor prognosis. Thus NRBP1 is a conserved regulator of cell fate, that plays an important role in tumour suppression.
Collapse
Affiliation(s)
- Catherine H Wilson
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, UK
| | - Catriona Crombie
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, UK
| | | | - George Poulogiannis
- Division of Signal Transduction, Beth Israel Deaconess Medical Center and Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - Alistair G Rust
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, UK
| | - Mercedes Pardo
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, UK
| | - Tannia Gracia
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, UK
| | - Lu Yu
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, UK
| | - Jyoti Choudhary
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, UK
| | - Gino B Poulin
- Faculty of Life Sciences, University of Manchester, Manchester, UK
| | - Rebecca E McIntyre
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, UK
| | | | - H Nikki March
- Cancer Research UK Cambridge Research Institute, Cambridge, UK
| | - Mark J Arends
- Department of Pathology, Addenbrookes Hospital, University of Cambridge, Cambridge, UK
| | - Andrew G Fraser
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, UK
- The Donnelly Centre, University of Toronto, Toronto, Canada
| | - David J Adams
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, UK
| |
Collapse
|
21
|
Chiu HS, York JP, Wilkinson L, Zhang P, Little MH, Pennisi DJ. Production of a mouse line with a conditional Crim1 mutant allele. Genesis 2012; 50:711-6. [PMID: 22511315 DOI: 10.1002/dvg.22032] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2011] [Revised: 02/27/2012] [Accepted: 04/12/2012] [Indexed: 11/09/2022]
Abstract
Crim1 is a developmentally expressed, transmembrane protein essential for normal embryonic development. We generated mice engineered to contain a Crim1 conditional null allele by flanking exons three and four of Crim1 with unidirectional LoxP sites. After crossing Crim1+/FLOX mice with a CMV-Cre line, a Crim1+/Δflox colony was established after germline transmission of the deleted allele. We then analyzed genomic DNA, mRNA transcripts, and protein expression from Crim1Δflox/Δflox null mice to confirm the nature of the genomic lesion. Crim1Δflox/Δflox mice displayed phenotypes similar to those previously described for a Crim1 gene-trap mutant, Crim1KST264/KST264, including perinatal lethality, digit syndactyly, eye, and kidney abnormalities, with varying penetrance and severity. The production of a conditional mutant allele represents a valuable resource for the study of the tissue-specific roles for Crim1, and for understanding the pleimorphic phenotypes associated with Crim1 mutation.
Collapse
Affiliation(s)
- Han Sheng Chiu
- School of Biomedical Sciences, The University of Queensland, Brisbane, Australia
| | | | | | | | | | | |
Collapse
|
22
|
Bruscoli S, Velardi E, Di Sante M, Bereshchenko O, Venanzi A, Coppo M, Berno V, Mameli MG, Colella R, Cavaliere A, Riccardi C. Long glucocorticoid-induced leucine zipper (L-GILZ) protein interacts with ras protein pathway and contributes to spermatogenesis control. J Biol Chem 2011; 287:1242-51. [PMID: 22110132 DOI: 10.1074/jbc.m111.316372] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Correct function of spermatogonia is critical for the maintenance of spermatogenesis throughout life, but the cellular pathways regulating undifferentiated spermatogonia proliferation, differentiation, and survival are only partially known. We show here that long glucocorticoid-induced leucine zipper (L-GILZ) is highly expressed in spermatogonia and primary spermatocytes and controls spermatogenesis. Gilz deficiency in knock-out (gilz KO) mice leads to a complete loss of germ cell lineage within first cycles of spermatogenesis, resulting in male sterility. Spermatogenesis failure is intrinsic to germ cells and is associated with increased proliferation and aberrant differentiation of undifferentiated spermatogonia and with hyperactivity of Ras signaling pathway as indicated by an increase of ERK and Akt phosphorylation. Spermatogonia differentiation does not proceed beyond the prophase of the first meiotic division due to massive apoptosis associated with accumulation of unrepaired chromosomal damage. These results identify L-GILZ as a novel important factor for undifferentiated spermatogonia function and spermatogenesis.
Collapse
Affiliation(s)
- Stefano Bruscoli
- Department of Clinical and Experimental Medicine, Section of Pharmacology, Toxicology, and Chemotherapy, Medical School, University of Perugia, Via del Giochetto, 06122 Perugia, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Abstract
Rett syndrome is a neurodevelopmental disorder caused by mutations in methyl-CpG-binding protein 2 (MECP2), a transcriptional regulator. In addition to cognitive, communication, and motor problems, affected individuals have abnormalities in autonomic function and respiratory control that may contribute to premature lethality. Mice lacking Mecp2 die early and recapitulate the autonomic and respiratory phenotypes seen in humans. The association of autonomic and respiratory deficits with premature death suggests that Mecp2 is critical within autonomic and respiratory control centers for survival. To test this, we compared the autonomic and respiratory phenotypes of mice with a null allele of Mecp2 to mice with Mecp2 removed from their brainstem and spinal cord. We found that MeCP2 is necessary within the brainstem and spinal cord for normal lifespan, normal control of heart rate, and respiratory response to hypoxia. Restoration of MeCP2 in a subset of the cells in this same region is sufficient to rescue abnormal heart rate and abnormal respiratory response to hypoxia. Furthermore, restoring MeCP2 function in neural centers critical for autonomic and respiratory function alleviates the lethality associated with loss of MeCP2 function, supporting the notion of targeted therapy toward treating Rett syndrome.
Collapse
|
24
|
Deng Q, Andersson E, Hedlund E, Alekseenko Z, Coppola E, Panman L, Millonig JH, Brunet JF, Ericson J, Perlmann T. Specific and integrated roles of Lmx1a, Lmx1b and Phox2a in ventral midbrain development. Development 2011; 138:3399-408. [PMID: 21752929 DOI: 10.1242/dev.065482] [Citation(s) in RCA: 96] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The severe disorders associated with a loss or dysfunction of midbrain dopamine neurons (DNs) have intensified research aimed at deciphering developmental programs controlling midbrain development. The homeodomain proteins Lmx1a and Lmx1b are important for the specification of DNs during embryogenesis, but it is unclear to what degree they may mediate redundant or specific functions. Here, we provide evidence showing that DN progenitors in the ventral midbrain can be subdivided into molecularly distinct medial and lateral domains, and these subgroups show different sensitivity to the loss of Lmx1a and Lmx1b. Lmx1a is specifically required for converting non-neuronal floor-plate cells into neuronal DN progenitors, a process that involves the establishment of Notch signaling in ventral midline cells. On the other hand, lateral DN progenitors that do not appear to originate from the floor plate are selectively ablated in Lmx1b mutants. In addition, we also reveal an unanticipated role for Lmx1b in regulating Phox2a expression and the sequential specification of ocular motor neurons (OMNs) and red nucleus neurons (RNNs) from progenitors located lateral to DNs in the midbrain. Our data therefore establish that Lmx1b influences the differentiation of multiple neuronal subtypes in the ventral midbrain, whereas Lmx1a appears to be exclusively devoted to the differentiation of the DN lineage.
Collapse
Affiliation(s)
- Qiaolin Deng
- Karolinska Institutet, Department of Cell and Molecular Biology, von Eulers väg 3, 171 77 Stockholm, Sweden
- Ludwig Institute for Cancer Research, Nobels väg 3, Karolinska Institutet, 71 77 Stockholm, Sweden
| | - Elisabet Andersson
- Karolinska Institutet, Department of Cell and Molecular Biology, von Eulers väg 3, 171 77 Stockholm, Sweden
| | - Eva Hedlund
- Ludwig Institute for Cancer Research, Nobels väg 3, Karolinska Institutet, 71 77 Stockholm, Sweden
| | - Zhanna Alekseenko
- Karolinska Institutet, Department of Cell and Molecular Biology, von Eulers väg 3, 171 77 Stockholm, Sweden
| | - Eva Coppola
- Institut de Biologie de l'Ecole Normale Supérieure (IBENS), CNRS UMR8197, INSERM U1024, 75005, Paris, France
| | - Lia Panman
- Ludwig Institute for Cancer Research, Nobels väg 3, Karolinska Institutet, 71 77 Stockholm, Sweden
| | - James H. Millonig
- UMDNJ, Neuroscience and Cell Biology, CABM, 679 Hoes Lane, Piscataway, NJ 08854, USA
| | - Jean-Francois Brunet
- Institut de Biologie de l'Ecole Normale Supérieure (IBENS), CNRS UMR8197, INSERM U1024, 75005, Paris, France
| | - Johan Ericson
- Karolinska Institutet, Department of Cell and Molecular Biology, von Eulers väg 3, 171 77 Stockholm, Sweden
| | - Thomas Perlmann
- Karolinska Institutet, Department of Cell and Molecular Biology, von Eulers väg 3, 171 77 Stockholm, Sweden
- Ludwig Institute for Cancer Research, Nobels väg 3, Karolinska Institutet, 71 77 Stockholm, Sweden
| |
Collapse
|
25
|
Sylva M, Li VSW, Buffing AAA, van Es JH, van den Born M, van der Velden S, Gunst Q, Koolstra JH, Moorman AFM, Clevers H, van den Hoff MJB. The BMP antagonist follistatin-like 1 is required for skeletal and lung organogenesis. PLoS One 2011; 6:e22616. [PMID: 21826198 PMCID: PMC3149603 DOI: 10.1371/journal.pone.0022616] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2011] [Accepted: 06/26/2011] [Indexed: 11/19/2022] Open
Abstract
Follistatin-like 1 (Fstl1) is a secreted protein of the BMP inhibitor class. During development, expression of Fstl1 is already found in cleavage stage embryos and becomes gradually restricted to mesenchymal elements of most organs during subsequent development. Knock down experiments in chicken and zebrafish demonstrated a role as a BMP antagonist in early development. To investigate the role of Fstl1 during mouse development, a conditional Fstl1 KO allele as well as a Fstl1-GFP reporter mouse were created. KO mice die at birth from respiratory distress and show multiple defects in lung development. Also, skeletal development is affected. Endochondral bone development, limb patterning as well as patterning of the axial skeleton are perturbed in the absence of Fstl1. Taken together, these observations show that Fstl1 is a crucial regulator in BMP signalling during mouse development.
Collapse
Affiliation(s)
- Marc Sylva
- Heart Failure Research Center, Academic Medical Center, Amsterdam, The Netherlands
| | - Vivian S. W. Li
- Hubrecht Institute, KNAW and University Medical Center, Utrecht, The Netherlands
| | - Anita A. A. Buffing
- Heart Failure Research Center, Academic Medical Center, Amsterdam, The Netherlands
| | - Johan H. van Es
- Hubrecht Institute, KNAW and University Medical Center, Utrecht, The Netherlands
| | - Maaike van den Born
- Hubrecht Institute, KNAW and University Medical Center, Utrecht, The Netherlands
| | | | - Quinn Gunst
- Heart Failure Research Center, Academic Medical Center, Amsterdam, The Netherlands
| | - Jan Harm Koolstra
- Academic Centre of Dentistry Amsterdam (ACTA), Amsterdam, The Netherlands
| | - Antoon F. M. Moorman
- Heart Failure Research Center, Academic Medical Center, Amsterdam, The Netherlands
| | - Hans Clevers
- Hubrecht Institute, KNAW and University Medical Center, Utrecht, The Netherlands
| | | |
Collapse
|
26
|
Liu H, Wang W, Chew SK, Lee SC, Li J, Vassiliou GS, Green T, Futreal PA, Bradley A, Zhang S, Liu P. Stella-Cre mice are highly efficient Cre deleters. Genesis 2011; 49:689-95. [PMID: 21786403 PMCID: PMC3674523 DOI: 10.1002/dvg.20741] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2010] [Accepted: 02/10/2011] [Indexed: 11/12/2022]
Abstract
Cre-loxP recombination is widely used for genetic manipulation of the mouse genome. Here, we report generation and characterization of a new Cre line, Stella-Cre, where Cre expression cassette was targeted to the 3' UTR of the Stella locus. Stella is specifically expressed in preimplantation embryos and in the germline. Cre-loxP recombination efficiency in Stella-Cre mice was investigated at several genomic loci including Rosa26, Jak2, and Npm1. At all the loci examined, we observed 100% Cre-loxP recombination efficiency in the embryos and in the germline. Thus, Stella-Cre mice serve as a very efficient deleter line.
Collapse
Affiliation(s)
- Hui Liu
- College of Animal Science and Technology, Huazhong Agriculture University, Wuhan, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Xu B, Chen C, Chen H, Zheng SG, Bringas P, Xu M, Zhou X, Chen D, Umans L, Zwijsen A, Shi W. Smad1 and its target gene Wif1 coordinate BMP and Wnt signaling activities to regulate fetal lung development. Development 2011; 138:925-35. [PMID: 21270055 DOI: 10.1242/dev.062687] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Bone morphogenetic protein 4 (Bmp4) is essential for lung development. To define the intracellular signaling mechanisms by which Bmp4 regulates lung development, BMP-specific Smad1 or Smad5 was selectively knocked out in fetal mouse lung epithelial cells. Abrogation of lung epithelial-specific Smad1, but not Smad5, resulted in retardation of lung branching morphogenesis and reduced sacculation, accompanied by altered distal lung epithelial cell proliferation and differentiation and, consequently, severe neonatal respiratory failure. By combining cDNA microarray with ChIP-chip analyses, Wnt inhibitory factor 1 (Wif1) was identified as a novel target gene of Smad1 in the developing mouse lung epithelial cells. Loss of Smad1 transcriptional activation of Wif1 was associated with reduced Wif1 expression and increased Wnt/β-catenin signaling activity in lung epithelia, resulting in specific fetal lung abnormalities. This suggests a novel regulatory loop of Bmp4-Smad1-Wif1-Wnt/β-catenin in coordinating BMP and Wnt pathways to control fetal lung development.
Collapse
Affiliation(s)
- Bing Xu
- Developmental Biology and Regenerative Medicine Program, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, CA 90027, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Wakker V, Brons JF, Aanhaanen WTJ, van Roon MA, Moorman AFM, Christoffels VM. Generation of mice with a conditional null allele for Tbx2. Genesis 2010; 48:195-9. [PMID: 20095052 DOI: 10.1002/dvg.20596] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The T-box transcription factor Tbx2 plays important roles in patterning and development, and has been implicated in cell-cycle regulation and cancer. Conventional disruption of Tbx2 results in abnormalities of the heart, limbs, eye and other structures, and early fetal lethality. To gain insight into the role of Tbx2 in different tissues and at different stages of development, we have generated a conditional null allele of Tbx2 by flanking Exon 2 with loxP sites (Tbx2(fl2)). Homozygous Tbx2(fl2) mice are viable and fertile, indicating that the Tbx2(fl2) allele is a fully functional Tbx2 allele. Cre-mediated recombination, using a ubiquitously active CMV-Cre line, results in deletion of Exon 2 and loss of protein expression. Embryos homozygous for the recombined allele (Tbx2(Delta2)) show the same heart and limb defects as conventional Tbx2-deficient embryos. This Tbx2 conditional null allele will be a valuable tool to uncover tissue-specific roles of Tbx2 in development and disease.
Collapse
Affiliation(s)
- Vincent Wakker
- Heart Failure Research Center, Academic Medical Center, Amsterdam, The Netherlands
| | | | | | | | | | | |
Collapse
|
29
|
The activating mutation R201C in GNAS promotes intestinal tumourigenesis in Apc(Min/+) mice through activation of Wnt and ERK1/2 MAPK pathways. Oncogene 2010; 29:4567-75. [PMID: 20531296 PMCID: PMC2923080 DOI: 10.1038/onc.2010.202] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Somatically acquired, activating mutations of GNAS, the gene encoding the stimulatory G-protein Gsα subunit, have been identified in kidney, thyroid, pituitary, leydig cell, adrenocortical and more recently, in colorectal tumours, suggesting that mutations such as R201C may be oncogenic in these tissues. To study the role of GNAS in intestinal tumourigenesis, we placed GNAS R201C under the control of the A33-antigen promoter (Gpa33), which is almost exclusively expressed in the intestines. The GNAS R201C mutation has been shown to result in the constitutive activation of Gsα and adenylate cyclase and to lead to the autonomous synthesis of cAMP. Gpa33tm1(GnasR201C)Wtsi/+ mice showed significantly elevated cAMP levels and a compensatory upregulation of cAMP-specific phosphodiesterases in the intestinal epithelium. GNAS R201C alone was not sufficient to induce tumourigenesis by 12 months but there was a significant increase in adenoma formation when Gpa33tm1(GnasR201C)Wtsi/+ mice were bred onto an ApcMin/+ background. GNAS R201C expression was associated with elevated expression of Wnt and extracellular signal-regulated kinase 1/2 mitogen-activated protein kinase (ERK1/2 MAPK) pathway target genes, increased phosphorylation of ERK1/2 MAPK, and increased immunostaining for the proliferation marker Ki67. Furthermore, the effects of GNAS R201C on the Wnt pathway were additive to inactivation of Apc. Our data strongly suggest that activating mutations of GNAS cooperate with inactivation of APC and are likely to contribute to colorectal tumourigenesis.
Collapse
|
30
|
Bildsoe H, Loebel DAF, Jones VJ, Chen YT, Behringer RR, Tam PPL. Requirement for Twist1 in frontonasal and skull vault development in the mouse embryo. Dev Biol 2009; 331:176-88. [PMID: 19414008 DOI: 10.1016/j.ydbio.2009.04.034] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2008] [Revised: 04/27/2009] [Accepted: 04/27/2009] [Indexed: 01/10/2023]
Abstract
Using a Cre-mediated conditional deletion approach, we have dissected the function of Twist1 in the morphogenesis of the craniofacial skeleton. Loss of Twist1 in neural crest cells and their derivatives impairs skeletogenic differentiation and leads to the loss of bones of the snout, upper face and skull vault. While no anatomically recognizable maxilla is formed, a malformed mandible is present. Since Twist1 is expressed in the tissues of the maxillary eminence and the mandibular arch, this finding suggests that the requirement for Twist1 is not the same in all neural crest derivatives. The effect of the loss of Twist1 function is not restricted to neural crest-derived bones, since the predominantly mesoderm-derived parietal and interparietal bones are also affected, presumably as a consequence of lost interactions with neural crest-derived tissues. In contrast, the formation of other mesodermal skeletal derivatives such as the occipital bones and most of the chondrocranium are not affected by the loss of Twist1 in the neural crest cells.
Collapse
Affiliation(s)
- Heidi Bildsoe
- Children's Medical Research Institute, The University of Sydney, Wentworthville, NSW, Australia
| | | | | | | | | | | |
Collapse
|
31
|
Schliekelman M, Cowley DO, O'Quinn R, Oliver TG, Lu L, Salmon ED, Van Dyke T. Impaired Bub1 function in vivo compromises tension-dependent checkpoint function leading to aneuploidy and tumorigenesis. Cancer Res 2009; 69:45-54. [PMID: 19117986 DOI: 10.1158/0008-5472.can-07-6330] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Bub1 is a serine/threonine kinase originally described as a core component of the spindle assembly checkpoint (SAC) mechanism in yeast. Bub1 binding at kinetochores has been reported to be required for SAC function and localization of other SAC components. A proper SAC is believed to be essential for murine embryonic development, as all previously described null mutations in SAC components in mice cause embryonic lethality. We produced mice harboring a Bub1 mutant allele lacking exons 2 and 3, resulting in a hypomorphic mutant expressed at <5% of wild-type levels. Despite this significant reduction, homozygous mutant animals are viable on a mixed 129P2/B6 or FVB background but display increased tumorigenesis with aging, whereas mice with a C57Bl/6J background die perinatally. Bub1 mutant murine embryonic fibroblasts (MEFs) display defects in chromosome congression to the metaphase plate, severe chromosome missegregation, and aneuploidy accompanied by high levels of premature senescence. Mutant MEFs have a robust SAC in response to nocodazole treatment but an impaired response to Taxol. Mutant MEFs also show reduced kinetochore localization of BubR1, but not of Mad2. The significant reduction in SAC response to Taxol, but not nocodazole, coupled with the reduced binding of BubR1, but not Mad2, indicates that Bub1 is particularly critical for the SAC response to a lack of tension on kinetochores. Thus, Bub1 is essential for proper chromosome segregation, a defect that can lead to severe phenotypes, including perinatal lethality and a predisposition to cancer.
Collapse
Affiliation(s)
- Mark Schliekelman
- Curriculum in Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA
| | | | | | | | | | | | | |
Collapse
|
32
|
Whitehead KJ, Chan AC, Navankasattusas S, Koh W, London NR, Ling J, Mayo AH, Drakos SG, Jones CA, Zhu W, Marchuk DA, Davis GE, Li DY. The cerebral cavernous malformation signaling pathway promotes vascular integrity via Rho GTPases. Nat Med 2009; 15:177-84. [PMID: 19151728 PMCID: PMC2767168 DOI: 10.1038/nm.1911] [Citation(s) in RCA: 284] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2008] [Accepted: 12/02/2008] [Indexed: 11/21/2022]
Abstract
Cerebral cavernous malformation (CCM) is a common vascular dysplasia that affects both systemic and CNS blood vessels. Loss of function mutations in the CCM2 gene cause CCM. Here we show that targeted disruption of Ccm2 in mice results in failed lumen formation and early embryonic death through an endothelial cell autonomous mechanism. We demonstrate that CCM2 regulates endothelial cytoskeletal architecture, cell-cell interactions and lumen formation. Heterozygosity at Ccm2, a genotype equivalent to human CCM, results in impaired endothelial barrier function. Because our biochemical studies indicate that loss of CCM2 results in activation of RHOA GTPase, we rescued the cellular phenotype and barrier function in heterozygous mice using simvastatin, a drug known to inhibit Rho GTPases. These data offer the prospect for pharmacologic treatment of a human vascular dysplasia using a widely available and safe drug.
Collapse
Affiliation(s)
- Kevin J Whitehead
- Division of Cardiology, Department of Medicine, 30 North 1900 East, Room 4A100, University of Utah, Salt Lake City, Utah 84132, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Abstract
Chromosomal rearrangements, such as deletions, duplications, inversions and translocations, occur frequently in humans and can be disease-associated or phenotypically neutral. To understand the genetic consequences of such genomic changes, these mutations need to be modelled in experimentally tractable systems. The mouse is an excellent organism for this analysis because of its biological and genetic similarity to humans, the ease with which its genome can be manipulated and the similarity of observed affects. Through chromosome engineering, defined rearrangements can be introduced into the mouse genome. The resulting mouse models are leading to a better understanding of the molecular and cellular basis of dosage alterations in human disease phenotypes, in turn opening new diagnostic and therapeutic opportunities.
Collapse
Affiliation(s)
- Louise van der Weyden
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridgeshire, UK
| | | | | |
Collapse
|
34
|
Aurora-A kinase is essential for bipolar spindle formation and early development. Mol Cell Biol 2008; 29:1059-71. [PMID: 19075002 DOI: 10.1128/mcb.01062-08] [Citation(s) in RCA: 99] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Aurora-A is a conserved kinase implicated in mitotic regulation and carcinogenesis. Aurora-A was previously implicated in mitotic entry and spindle assembly, although contradictory results prevented a clear understanding of the roles of Aurora-A in mammals. We developed a conditional null mutation in the mouse Aurora-A gene to investigate Aurora-A functions in primary cells ex vivo and in vivo. We show here that conditional Aurora-A ablation in cultured embryonic fibroblasts causes impaired mitotic entry and mitotic arrest with a profound defect in bipolar spindle formation. Germ line Aurora-A deficiency causes embryonic death at the blastocyst stage with pronounced cell proliferation failure, mitotic arrest, and monopolar spindle formation. Aurora-A deletion in mid-gestation embryos causes an increase in mitotic and apoptotic cells. These results indicate that murine Aurora-A facilitates, but is not absolutely required for, mitotic entry in murine embryonic fibroblasts and is essential for centrosome separation and bipolar spindle formation in vitro and in vivo. Aurora-A deletion increases apoptosis, suggesting that molecular therapies targeting Aurora-A may be effective in inducing tumor cell apoptosis. Aurora-A conditional mutant mice provide a valuable system for further defining Aurora-A functions and for predicting effects of Aurora-A therapeutic intervention.
Collapse
|
35
|
Mao CA, Kiyama T, Pan P, Furuta Y, Hadjantonakis AK, Klein WH. Eomesodermin, a target gene of Pou4f2, is required for retinal ganglion cell and optic nerve development in the mouse. Development 2007; 135:271-80. [PMID: 18077589 DOI: 10.1242/dev.009688] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The mechanisms regulating retinal ganglion cell (RGC) development are crucial for retinogenesis and for the establishment of normal vision. However, these mechanisms are only vaguely understood. RGCs are the first neuronal lineage to segregate from pluripotent progenitors in the developing retina. As output neurons, RGCs display developmental features very distinct from those of the other retinal cell types. To better understand RGC development, we have previously constructed a gene regulatory network featuring a hierarchical cascade of transcription factors that ultimately controls the expression of downstream effector genes. This has revealed the existence of a Pou domain transcription factor, Pou4f2, that occupies a key node in the RGC gene regulatory network and that is essential for RGC differentiation. However, little is known about the genes that connect upstream regulatory genes, such as Pou4f2 with downstream effector genes responsible for RGC differentiation. The purpose of this study was to characterize the retinal function of eomesodermin (Eomes), a T-box transcription factor with previously unsuspected roles in retinogenesis. We show that Eomes is expressed in developing RGCs and is a mediator of Pou4f2 function. Pou4f2 directly regulates Eomes expression through a cis-regulatory element within a conserved retinal enhancer. Deleting Eomes in the developing retina causes defects reminiscent of those in Pou4f2(-/-) retinas. Moreover, myelin ensheathment in the optic nerves of Eomes(-/-) embryos is severely impaired, suggesting that Eomes regulates this process. We conclude that Eomes is a crucial regulator positioned immediately downstream of Pou4f2 and is required for RGC differentiation and optic nerve development.
Collapse
Affiliation(s)
- Chai-An Mao
- Department of Biochemistry and Molecular Biology, The University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA
| | | | | | | | | | | |
Collapse
|
36
|
Badaloni A, Bonanomi D, Albieri I, Givogri I, Bongarzone E, Valtorta F, Consalez GG. Transgenic mice expressing a dual, CRE-inducible reporter for the analysis of axon guidance and synaptogenesis. Genesis 2007; 45:405-12. [PMID: 17554764 DOI: 10.1002/dvg.20307] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Improved and modular tools are needed for the neuroanatomical dissection of CNS axonal tracts, and to study the cell-intrinsic and cell-extrinsic cues that govern their assembly and plasticity. Here we describe a general purpose transgenic tracer that can be used to visualize axonal tracts and synaptic terminals in any region of the embryonic neural tube or postnatal CNS, on any wild type or mutant genetic background. The construct permits CRE-inducible expression of a dicistronic axonal marker encoding two surface reporter proteins: a farnesylated GFP and the human Placental Alkaline Phosphatase (PLAP). Both proteins localize alongside the neuronal surface, permitting the concomitant detection of cell body, neurites, and presynaptic and postsynaptic sites in the same neuron. This provides a CRE-inducible dual system for imaging neural circuits in vivo, and to study their assembly and remodeling in cultured neurons, neural stem cells, and tissue explants derived from the reporter line. Unlike existing lines, this reporter does not encode a ubiquitously expressed, floxable LacZ gene, permitting the simultaneous analysis of beta galactosidase activity in mutant lines.
Collapse
Affiliation(s)
- Aurora Badaloni
- San Raffaele Scientific Institute and Università Vita-Salute San Raffaele, Milano, Italy
| | | | | | | | | | | | | |
Collapse
|
37
|
Abstract
Genetic mosaics produced by FLP/FRT induced mitotic recombination have been widely used in Drosophila to study gene function in development. Recently, the Cre/loxP system has been applied to induce mitotic recombination in mouse embryonic stem cells and in many adult mouse tissues. We have used this strategy to generate a previously undescribed p53 mouse model in which expression of a ubiquitously expressed recombinase in a heterozygous p53 knockout animal produces mitotic recombinant clones homozygous for the p53 mutation. The induction of loss of heterozygosity in a few cells in an otherwise normal tissue mimics genetic aspects of tumorigenesis more closely than existing models and has revealed the possible cell autonomous nature of Wnt3. Our results suggest that inducible mitotic recombination can be used for clonal analysis of mutants in the mouse.
Collapse
Affiliation(s)
- Wei Wang
- *Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SA, United Kingdom; and
| | - Madhuri Warren
- *Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SA, United Kingdom; and
- Department of Pathology, University of Cambridge, Addenbrookes Hospital, Hills Road, Cambridge CB2 2QQ, United Kingdom
| | - Allan Bradley
- *Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SA, United Kingdom; and
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|
38
|
Ismat FA, Xu J, Lu MM, Epstein JA. The neurofibromin GAP-related domain rescues endothelial but not neural crest development in Nf1 mice. J Clin Invest 2006; 116:2378-84. [PMID: 16906226 PMCID: PMC1533876 DOI: 10.1172/jci28341] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2006] [Accepted: 06/13/2006] [Indexed: 11/17/2022] Open
Abstract
Neurofibromatosis type I (NF1; also known as von Recklinghausen's disease) is a common autosomal-dominant condition primarily affecting neural crest-derived tissues. The disease gene, NF1, encodes neurofibromin, a protein of over 2,800 amino acids that contains a 216-amino acid domain with Ras-GTPase-activating protein (Ras-GAP) activity. Potential therapies for NF1 currently in development and being tested in clinical trials are designed to modify NF1 Ras-GAP activity or target downstream effectors of Ras signaling. Mice lacking the murine homolog (Nf1) have mid-gestation lethal cardiovascular defects due to a requirement for neurofibromin in embryonic endothelium. We sought to determine whether the GAP activity of neurofibromin is sufficient to rescue complete loss of function or whether other as yet unidentified functions of neurofibromin might also exist. Using cre-inducible ubiquitous and tissue-specific expression, we demonstrate that the isolated GAP-related domain (GRD) rescued cardiovascular development in Nf1(-/-) embryos, but overgrowth of neural crest-derived tissues persisted, leading to perinatal lethality. These results suggest that neurofibromin may possess activities outside of the GRD that modulate neural crest homeostasis and that therapeutic approaches solely aimed at targeting Ras activity may not be sufficient to treat tumors of neural crest origin in NF1.
Collapse
Affiliation(s)
- Fraz A Ismat
- Division of Cardiology, Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA.
| | | | | | | |
Collapse
|
39
|
Charles MA, Saunders TL, Wood WM, Owens K, Parlow AF, Camper SA, Ridgway EC, Gordon DF. Pituitary-specific Gata2 knockout: effects on gonadotrope and thyrotrope function. Mol Endocrinol 2006; 20:1366-77. [PMID: 16543408 DOI: 10.1210/me.2005-0378] [Citation(s) in RCA: 116] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
GATA2 is expressed in the pituitary during development and in adult gonadotropes and thyrotropes. It is proposed to be important for gonadotrope and thyrotrope cell fate choice and for TSH production. To test this idea, we produced a pituitary-specific knockout of Gata2, designed so that the DNA-binding zinc-finger region is deleted in the presence of a pituitary-specific recombinase transgene. These mice have reduced secretion of gonadotropins basally and in response to castration challenge, although the mice are fertile. GATA2 deficiency also compromises thyrotrope function. Mutants have fewer thyrotrope cells at birth, male Gata2-deficient mice exhibit growth delay from 3-9 wk of age, and adult mutants produce less TSH in response to severe hypothyroidism after radiothyroidectomy. Therefore, Gata2 appears to be dispensable for gonadotrope and thyrotrope cell fate and maintenance, but important for optimal gonadotrope and thyrotrope function. Gata2-deficient mice exhibit elevated levels of Gata3 transcripts in the pituitary gland, suggesting that GATA3 can compensate for GATA2.
Collapse
Affiliation(s)
- Michael A Charles
- Department of Human Genetics, University of Michigan, Ann Arbor, Michigan 48109-0618, USA
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Eckardt D, Theis M, Döring B, Speidel D, Willecke K, Ott T. Spontaneous ectopic recombination in cell-type-specific Cre mice removes loxP-flanked marker cassettes in vivo. Genesis 2005; 38:159-65. [PMID: 15083516 DOI: 10.1002/gene.20011] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Conditional gene targeting using the Cre/loxP technology generally includes integration of a selection marker cassette flanked by loxP recognition sites (floxed) in the target gene locus. Subsequent marker removal avoids possible impairment of gene expression or mosaicism due to partial and total deletions after Cre-mediated recombination in vivo. The use of deleter Cre mice for in vivo marker removal in floxed connexin43 mice revealed considerable mosaicism, but no selective marker removal. In addition, we noted that several Cre transgenic lines displayed spontaneous ectopic activity, reminiscent of deleter Cre mice, and required the confirmation of cell type-specific deletion in every individual mouse. When we used myosin heavy chain promoter Cre (alphaMyHC-Cre) mice for cardiomyocyte specific deletion, we observed, in addition to cardiomyocyte-restricted or complete excision, selective marker removal in a subgroup of mice as well. Thus, selective marker removal can be achieved as a byproduct of cell-type restricted deletion.
Collapse
|
41
|
Salmena L, Lemmers B, Hakem A, Matysiak-Zablocki E, Murakami K, Au PYB, Berry DM, Tamblyn L, Shehabeldin A, Migon E, Wakeham A, Bouchard D, Yeh WC, McGlade JC, Ohashi PS, Hakem R. Essential role for caspase 8 in T-cell homeostasis and T-cell-mediated immunity. Genes Dev 2003; 17:883-95. [PMID: 12654726 PMCID: PMC196031 DOI: 10.1101/gad.1063703] [Citation(s) in RCA: 370] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2002] [Accepted: 02/18/2003] [Indexed: 01/18/2023]
Abstract
Defects in death receptor-mediated apoptosis have been linked to cancer and autoimmune disease in humans. The in vivo role of caspase 8, a component of this pathway, has eluded analysis in postnatal tissues because of the lack of an appropriate animal model. Targeted disruption of caspase 8 is lethal in utero. We generated mice with a targeted caspase 8 mutation that is restricted to the T-cell lineage. Despite normal thymocyte development in the absence of caspase 8, we observed a marked decrease in the number of peripheral T-cells and impaired T-cell response ex vivo to activation stimuli. caspase 8 ablation protected thymocytes and activated T-cells from CD95 ligand but not anti-CD3-induced apoptosis, or apoptosis activated by agents that are known to act through the mitochondria. caspase 8 mutant mice were unable to mount an immune response to viral infection, indicating that caspase 8 deletion in T-cells leads to immunodeficiency. These findings identify an essential, cell-stage-specific role for caspase 8 in T-cell homeostasis and T-cell-mediated immunity. This is consistent with the recent identification of caspase 8 mutations in human immunodeficiency.
Collapse
Affiliation(s)
- Leonardo Salmena
- The Advanced Medical Discovery Institute (AMDI), Ontario Cancer Institute, and Department of Medical Biophysics, University of Toronto, Toronto, Ontario M5G 2C1, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Leneuve P, Colnot S, Hamard G, Francis F, Niwa-Kawakita M, Giovannini M, Holzenberger M. Cre-mediated germline mosaicism: a new transgenic mouse for the selective removal of residual markers from tri-lox conditional alleles. Nucleic Acids Res 2003; 31:e21. [PMID: 12595570 PMCID: PMC149843 DOI: 10.1093/nar/gng021] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The binary Cre-lox conditional knockout system requires an essential part of the target gene to be flanked by loxP sites, enabling excision in vivo upon Cre expression. LoxP sites are introduced by homologous recombination, together with a selectable marker. However, this marker can disturb gene expression and should be removed. The marker is therefore often prepared with a third, flanking loxP site (tri-lox construct), facilitating its selective removal by partial Cre-lox recombination. We have shown that this excision can be achieved in vivo in the germline using EIIaCre transgenic mice, and have described the advantages of in vivo over in vitro removal. We show here that MeuCre40, a new transgenic mouse, more reliably and reproducibly generates an optimal partial mosaic Cre-lox recombination pattern in the early embryo. This mosaicism was transmitted to the germline and to many other tissues. Alleles with partial deletions, in particular floxed alleles from which the selectable marker was removed, were readily recovered in the next generation, after segregation from the transgene. Segregation via paternal or maternal transmission led to successful recovery of the alleles of interest. We also obtained total deletion of the floxed regions in the same experiment, making this transgene a polyvalent Cre-lox tool. We rigorously tested the ability of MeuCre40 to solve tri-lox problems, by using it for the in vivo removal of neo(R)- and hprt-expression cassettes from three different tri-lox mutants.
Collapse
Affiliation(s)
- Patricia Leneuve
- Inserm U515, Hôpital Saint-Antoine, 184 r Fbg St-Antoine, 75571 Paris 12, France
| | | | | | | | | | | | | |
Collapse
|
43
|
Affiliation(s)
- Alea A Mills
- Cold Spring Harbor Laboratory, One Bungtown Road, Cold Spring Harbor, New York 11724, USA.
| | | | | |
Collapse
|