1
|
Baudot AD, Wang VMY, Leach JD, O’Prey J, Long JS, Paulus-Hock V, Lilla S, Thomson DM, Greenhorn J, Ghaffar F, Nixon C, Helfrich MH, Strathdee D, Pratt J, Marchesi F, Zanivan S, Ryan KM. Glycan degradation promotes macroautophagy. Proc Natl Acad Sci U S A 2022; 119:e2111506119. [PMID: 35737835 PMCID: PMC9245654 DOI: 10.1073/pnas.2111506119] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 04/22/2022] [Indexed: 11/18/2022] Open
Abstract
Macroautophagy promotes cellular homeostasis by delivering cytoplasmic constituents to lysosomes for degradation [Mizushima, Nat. Cell Biol. 20, 521-527 (2018)]. However, while most studies have focused on the mechanisms of protein degradation during this process, we report here that macroautophagy also depends on glycan degradation via the glycosidase, α-l-fucosidase 1 (FUCA1), which removes fucose from glycans. We show that cells lacking FUCA1 accumulate lysosomal glycans, which is associated with impaired autophagic flux. Moreover, in a mouse model of fucosidosis-a disease characterized by inactivating mutations in FUCA1 [Stepien et al., Genes (Basel) 11, E1383 (2020)]-glycan and autophagosome/autolysosome accumulation accompanies tissue destruction. Mechanistically, using lectin capture and mass spectrometry, we identified several lysosomal enzymes with altered fucosylation in FUCA1-null cells. Moreover, we show that the activity of some of these enzymes in the absence of FUCA1 can no longer be induced upon autophagy stimulation, causing retardation of autophagic flux, which involves impaired autophagosome-lysosome fusion. These findings therefore show that dysregulated glycan degradation leads to defective autophagy, which is likely a contributing factor in the etiology of fucosidosis.
Collapse
Affiliation(s)
- Alice D. Baudot
- Tumour Cell Death and Autophagy Laboratory, Cancer Research UK Beatson Institute, Glasgow G61 1BD, United Kingdom
| | - Victoria M.-Y. Wang
- Tumour Cell Death and Autophagy Laboratory, Cancer Research UK Beatson Institute, Glasgow G61 1BD, United Kingdom
| | - Josh D. Leach
- Tumour Cell Death and Autophagy Laboratory, Cancer Research UK Beatson Institute, Glasgow G61 1BD, United Kingdom
- School of Veterinary Medicine, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow G61 1QH, United Kingdom
| | - Jim O’Prey
- Tumour Cell Death and Autophagy Laboratory, Cancer Research UK Beatson Institute, Glasgow G61 1BD, United Kingdom
| | - Jaclyn S. Long
- Tumour Cell Death and Autophagy Laboratory, Cancer Research UK Beatson Institute, Glasgow G61 1BD, United Kingdom
| | - Viola Paulus-Hock
- Tumour Cell Death and Autophagy Laboratory, Cancer Research UK Beatson Institute, Glasgow G61 1BD, United Kingdom
| | - Sergio Lilla
- Tumour Cell Death and Autophagy Laboratory, Cancer Research UK Beatson Institute, Glasgow G61 1BD, United Kingdom
| | - David M. Thomson
- Strathclyde Institute of Pharmacy and Biomedical Science, University of Strathclyde, Glasgow G4 0RE, United Kingdom
| | - John Greenhorn
- Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, United Kingdom
| | - Farah Ghaffar
- Tumour Cell Death and Autophagy Laboratory, Cancer Research UK Beatson Institute, Glasgow G61 1BD, United Kingdom
| | - Colin Nixon
- Tumour Cell Death and Autophagy Laboratory, Cancer Research UK Beatson Institute, Glasgow G61 1BD, United Kingdom
| | - Miep H. Helfrich
- Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, United Kingdom
| | - Douglas Strathdee
- Tumour Cell Death and Autophagy Laboratory, Cancer Research UK Beatson Institute, Glasgow G61 1BD, United Kingdom
| | - Judith Pratt
- Strathclyde Institute of Pharmacy and Biomedical Science, University of Strathclyde, Glasgow G4 0RE, United Kingdom
| | - Francesco Marchesi
- School of Veterinary Medicine, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow G61 1QH, United Kingdom
| | - Sara Zanivan
- Tumour Cell Death and Autophagy Laboratory, Cancer Research UK Beatson Institute, Glasgow G61 1BD, United Kingdom
- Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, United Kingdom
| | - Kevin M. Ryan
- Tumour Cell Death and Autophagy Laboratory, Cancer Research UK Beatson Institute, Glasgow G61 1BD, United Kingdom
- Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, United Kingdom
| |
Collapse
|
2
|
Humpton TJ, Nomura K, Weber J, Magnussen HM, Hock AK, Nixon C, Dhayade S, Stevenson D, Huang DT, Strathdee D, Blyth K, Vousden KH. Differential requirements for MDM2 E3 activity during embryogenesis and in adult mice. Genes Dev 2021; 35:117-132. [PMID: 33334825 PMCID: PMC7778261 DOI: 10.1101/gad.341875.120] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 11/06/2020] [Indexed: 12/17/2022]
Abstract
The p53 tumor suppressor protein is a potent activator of proliferative arrest and cell death. In normal cells, this pathway is restrained by p53 protein degradation mediated by the E3-ubiquitin ligase activity of MDM2. Oncogenic stress releases p53 from MDM2 control, so activating the p53 response. However, many tumors that retain wild-type p53 inappropriately maintain the MDM2-p53 regulatory loop in order to continuously suppress p53 activity. We have shown previously that single point mutations in the human MDM2 RING finger domain prevent the interaction of MDM2 with the E2/ubiquitin complex, resulting in the loss of MDM2's E3 activity without preventing p53 binding. Here, we show that an analogous mouse MDM2 mutant (MDM2 I438K) restrains p53 sufficiently for normal growth but exhibits an enhanced stress response in vitro. In vivo, constitutive expression of MDM2 I438K leads to embryonic lethality that is rescued by p53 deletion, suggesting MDM2 I438K is not able to adequately control p53 function through development. However, the switch to I438K expression is tolerated in adult mice, sparing normal cells but allowing for an enhanced p53 response to DNA damage. Viewed as a proof of principle model for therapeutic development, our findings support an approach that would inhibit MDM2 E3 activity without preventing MDM2/p53 binding as a promising avenue for development of compounds to activate p53 in tumors with reduced on-target toxicities.
Collapse
Affiliation(s)
- Timothy J Humpton
- The Francis Crick Institute, London NW1 1AT, United Kingdom
- Cancer Research UK Beatson Institute, Glasgow G61 1BD, United Kingdom
| | - Koji Nomura
- Cancer Research UK Beatson Institute, Glasgow G61 1BD, United Kingdom
| | - Julia Weber
- The Francis Crick Institute, London NW1 1AT, United Kingdom
| | - Helge M Magnussen
- Cancer Research UK Beatson Institute, Glasgow G61 1BD, United Kingdom
| | - Andreas K Hock
- Cancer Research UK Beatson Institute, Glasgow G61 1BD, United Kingdom
| | - Colin Nixon
- Cancer Research UK Beatson Institute, Glasgow G61 1BD, United Kingdom
| | - Sandeep Dhayade
- Cancer Research UK Beatson Institute, Glasgow G61 1BD, United Kingdom
| | - David Stevenson
- Cancer Research UK Beatson Institute, Glasgow G61 1BD, United Kingdom
| | - Danny T Huang
- Cancer Research UK Beatson Institute, Glasgow G61 1BD, United Kingdom
- Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, United Kingdom
| | - Douglas Strathdee
- Cancer Research UK Beatson Institute, Glasgow G61 1BD, United Kingdom
| | - Karen Blyth
- Cancer Research UK Beatson Institute, Glasgow G61 1BD, United Kingdom
- Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, United Kingdom
| | | |
Collapse
|
3
|
Lindahl M, Chalazonitis A, Palm E, Pakarinen E, Danilova T, Pham TD, Setlik W, Rao M, Võikar V, Huotari J, Kopra J, Andressoo JO, Piepponen PT, Airavaara M, Panhelainen A, Gershon MD, Saarma M. Cerebral dopamine neurotrophic factor-deficiency leads to degeneration of enteric neurons and altered brain dopamine neuronal function in mice. Neurobiol Dis 2019; 134:104696. [PMID: 31783118 PMCID: PMC7000201 DOI: 10.1016/j.nbd.2019.104696] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 11/07/2019] [Accepted: 11/24/2019] [Indexed: 12/13/2022] Open
Abstract
Cerebral dopamine neurotrophic factor (CDNF) is neuroprotective for nigrostriatal dopamine neurons and restores dopaminergic function in animal models of Parkinson’s disease (PD). To understand the role of CDNF in mammals, we generated CDNF knockout mice (Cdnf−/−), which are viable, fertile, and have a normal life-span. Surprisingly, an age-dependent loss of enteric neurons occurs selectively in the submucosal but not in the myenteric plexus. This neuronal loss is a consequence not of increased apoptosis but of neurodegeneration and autophagy. Quantitatively, the neurodegeneration and autophagy found in the submucosal plexus in duodenum, ileum and colon of the Cdnf−/− mouse are much greater than in those of Cdnf+/+ mice. The selective vulnerability of submucosal neurons to the absence of CDNF is reminiscent of the tendency of pathological abnormalities to occur in the submucosal plexus in biopsies of patients with PD. In contrast, the number of substantia nigra dopamine neurons and dopamine and its metabolite concentrations in the striatum are unaltered in Cdnf−/− mice; however, there is an age-dependent deficit in the function of the dopamine system in Cdnf−/− male mice analyzed. This is observed as D-amphetamine-induced hyperactivity, aberrant dopamine transporter function, and as increased D-amphetamine-induced dopamine release demonstrating that dopaminergic axon terminal function in the striatum of the Cdnf−/− mouse brain is altered. The deficiencies of Cdnf−/− mice, therefore, are reminiscent of those seen in early stages of Parkinson’s disease.
Collapse
Affiliation(s)
- Maria Lindahl
- Institute of Biotechnology, HiLIFE Unit, Viikinkaari 5D, FI-00014, University of Helsinki, Finland.
| | | | - Erik Palm
- Institute of Biotechnology, HiLIFE Unit, Viikinkaari 5D, FI-00014, University of Helsinki, Finland
| | - Emmi Pakarinen
- Institute of Biotechnology, HiLIFE Unit, Viikinkaari 5D, FI-00014, University of Helsinki, Finland
| | - Tatiana Danilova
- Institute of Biotechnology, HiLIFE Unit, Viikinkaari 5D, FI-00014, University of Helsinki, Finland
| | - Tuan D Pham
- Department of Pathology & Cell Biology, Columbia University, NY, New York, USA
| | - Wanda Setlik
- Department of Pathology & Cell Biology, Columbia University, NY, New York, USA
| | - Meenakshi Rao
- Department of Pathology & Cell Biology, Columbia University, NY, New York, USA
| | - Vootele Võikar
- Neuroscience Center/Laboratory Animal Center, Mustialankatu 1, FI-00014, University of Helsinki, Finland
| | - Jatta Huotari
- Institute of Biotechnology, HiLIFE Unit, Viikinkaari 5D, FI-00014, University of Helsinki, Finland
| | - Jaakko Kopra
- Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, Viikinkaari 5E, FI-00014, University of Helsinki, Finland
| | - Jaan-Olle Andressoo
- Institute of Biotechnology, HiLIFE Unit, Viikinkaari 5D, FI-00014, University of Helsinki, Finland
| | - Petteri T Piepponen
- Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, Viikinkaari 5E, FI-00014, University of Helsinki, Finland
| | - Mikko Airavaara
- Institute of Biotechnology, HiLIFE Unit, Viikinkaari 5D, FI-00014, University of Helsinki, Finland
| | - Anne Panhelainen
- Institute of Biotechnology, HiLIFE Unit, Viikinkaari 5D, FI-00014, University of Helsinki, Finland
| | - Michael D Gershon
- Department of Pathology & Cell Biology, Columbia University, NY, New York, USA
| | - Mart Saarma
- Institute of Biotechnology, HiLIFE Unit, Viikinkaari 5D, FI-00014, University of Helsinki, Finland
| |
Collapse
|
4
|
Attwood KM, Salsman J, Chung D, Mathavarajah S, Van Iderstine C, Dellaire G. PML isoform expression and DNA break location relative to PML nuclear bodies impacts the efficiency of homologous recombination. Biochem Cell Biol 2019; 98:314-326. [PMID: 31671275 DOI: 10.1139/bcb-2019-0115] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Promyelocytic leukemia nuclear bodies (PML NBs) are nuclear subdomains that respond to genotoxic stress by increasing in number via changes in chromatin structure. However, the role of the PML protein and PML NBs in specific mechanisms of DNA repair has not been fully characterized. Here, we have directly examined the role of PML in homologous recombination (HR) using I-SceI extrachromosomal and chromosome-based homology-directed repair (HDR) assays, and in HDR by CRISPR/Cas9-mediated gene editing. We determined that PML loss can inhibit HR in an extrachromosomal HDR assay but had less of an effect on CRISPR/Cas9-mediated chromosomal HDR. Overexpression of PML also inhibited both CRISPR HDR and I-SceI-induced HDR using a chromosomal reporter, and in an isoform-specific manner. However, the impact of PML overexpression on the chromosomal HDR reporter was dependent on the intranuclear chromosomal positioning of the reporter. Specifically, HDR at the TAP1 gene locus, which is associated with PML NBs, was reduced compared with a locus not associated with a PML NB; yet, HDR could be reduced at the non-PML NB-associated locus by PML overexpression. Thus, both loss and overexpression of PML isoforms can inhibit HDR, and proximity of a chromosomal break to a PML NB can impact HDR efficiency.
Collapse
Affiliation(s)
- Kathleen M Attwood
- Department of Biochemistry & Molecular Biology, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Jayme Salsman
- Department of Pathology, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Dudley Chung
- Department of Pathology, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | | | | | - Graham Dellaire
- Department of Biochemistry & Molecular Biology, Dalhousie University, Halifax, NS B3H 4R2, Canada.,Department of Pathology, Dalhousie University, Halifax, NS B3H 4R2, Canada
| |
Collapse
|
5
|
Woo AJ, Patry CAA, Ghamari A, Pregernig G, Yuan D, Zheng K, Piers T, Hibbs M, Li J, Fidalgo M, Wang JY, Lee JH, Leedman PJ, Wang J, Fraenkel E, Cantor AB. Zfp281 (ZBP-99) plays a functionally redundant role with Zfp148 (ZBP-89) during erythroid development. Blood Adv 2019; 3:2499-2511. [PMID: 31455666 PMCID: PMC6712527 DOI: 10.1182/bloodadvances.2018030551] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Accepted: 06/11/2019] [Indexed: 12/17/2022] Open
Abstract
Erythroid maturation requires the concerted action of a core set of transcription factors. We previously identified the Krüppel-type zinc finger transcription factor Zfp148 (also called ZBP-89) as an interacting partner of the master erythroid transcription factor GATA1. Here we report the conditional knockout of Zfp148 in mice. Global loss of Zfp148 results in perinatal lethality from nonhematologic causes. Selective Zfp148 loss within the hematopoietic system results in a mild microcytic and hypochromic anemia, mildly impaired erythroid maturation, and delayed recovery from phenylhydrazine-induced hemolysis. Based on the mild erythroid phenotype of these mice compared with GATA1-deficient mice, we hypothesized that additional factor(s) may complement Zfp148 function during erythropoiesis. We show that Zfp281 (also called ZBP-99), another member of the Zfp148 transcription factor family, is highly expressed in murine and human erythroid cells. Zfp281 knockdown by itself results in partial erythroid defects. However, combined deficiency of Zfp148 and Zfp281 causes a marked erythroid maturation block. Zfp281 physically associates with GATA1, occupies many common chromatin sites with GATA1 and Zfp148, and regulates a common set of genes required for erythroid cell differentiation. These findings uncover a previously unknown role for Zfp281 in erythroid development and suggest that it functionally overlaps with that of Zfp148 during erythropoiesis.
Collapse
Affiliation(s)
- Andrew J Woo
- Division of Pediatric Hematology-Oncology, Boston Children's Hospital/Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
- Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands and Centre for Medical Research, University of Western Australia, Perth, WA, Australia
| | - Chelsea-Ann A Patry
- Division of Pediatric Hematology-Oncology, Boston Children's Hospital/Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Alireza Ghamari
- Division of Pediatric Hematology-Oncology, Boston Children's Hospital/Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Gabriela Pregernig
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA
| | - Daniel Yuan
- Division of Pediatric Hematology-Oncology, Boston Children's Hospital/Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Kangni Zheng
- Division of Pediatric Hematology-Oncology, Boston Children's Hospital/Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Taylor Piers
- Division of Pediatric Hematology-Oncology, Boston Children's Hospital/Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Moira Hibbs
- Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands and Centre for Medical Research, University of Western Australia, Perth, WA, Australia
| | - Ji Li
- Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands and Centre for Medical Research, University of Western Australia, Perth, WA, Australia
| | - Miguel Fidalgo
- Department of Cell, Developmental and Regenerative Biology, Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY
- Center for Research in Molecular Medicine and Chronic Diseases, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Jenny Y Wang
- Children's Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, Sydney, NSW, Australia
| | - Joo-Hyeon Lee
- Wellcome Trust/Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom; and
| | - Peter J Leedman
- Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands and Centre for Medical Research, University of Western Australia, Perth, WA, Australia
| | - Jianlong Wang
- Department of Cell, Developmental and Regenerative Biology, Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Ernest Fraenkel
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA
| | - Alan B Cantor
- Division of Pediatric Hematology-Oncology, Boston Children's Hospital/Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA
| |
Collapse
|
6
|
Morphogenesis of extra-embryonic tissues directs the remodelling of the mouse embryo at implantation. Nat Commun 2019; 10:3557. [PMID: 31391456 PMCID: PMC6686005 DOI: 10.1038/s41467-019-11482-5] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 07/03/2019] [Indexed: 11/09/2022] Open
Abstract
Mammalian embryos change shape dramatically upon implantation. The cellular and molecular mechanism underlying this transition are largely unknown. Here, we show that this transition is directed by cross talk between the embryonic epiblast and the first extra-embryonic tissue, the trophectoderm. Specifically, we show via visualisation of a Cdx2-GFP reporter line and pharmacologically mediated loss and gain of function experiments that the epiblast provides FGF signal that results in differential fate acquisition in the multipotent trophectoderm leading to the formation of a tissue boundary within this tissue. The trophectoderm boundary becomes essential for expansion of the tissue into a multi-layered epithelium. Folding of this multi-layered trophectoderm induces spreading of the second extra-embryonic tissue, the primitive endoderm. Together, these events remodel the pre-implantation embryo into its post-implantation cylindrical shape. Our findings uncover how communication between embryonic and extra-embryonic tissues provides positional cues to drive shape changes in mammalian development during implantation.
Collapse
|
7
|
Vest KE, Phillips BL, Banerjee A, Apponi LH, Dammer EB, Xu W, Zheng D, Yu J, Tian B, Pavlath GK, Corbett AH. Novel mouse models of oculopharyngeal muscular dystrophy (OPMD) reveal early onset mitochondrial defects and suggest loss of PABPN1 may contribute to pathology. Hum Mol Genet 2017; 26:3235-3252. [PMID: 28575395 PMCID: PMC5886286 DOI: 10.1093/hmg/ddx206] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 05/14/2017] [Accepted: 05/23/2017] [Indexed: 01/09/2023] Open
Abstract
Oculopharyngeal muscular dystrophy (OPMD) is a late onset disease caused by polyalanine expansion in the poly(A) binding protein nuclear 1 (PABPN1). Several mouse models have been generated to study OPMD; however, most of these models have employed transgenic overexpression of alanine-expanded PABPN1. These models do not recapitulate the OPMD patient genotype and PABPN1 overexpression could confound molecular phenotypes. We have developed a knock-in mouse model of OPMD (Pabpn1+/A17) that contains one alanine-expanded Pabpn1 allele under the control of the native promoter and one wild-type Pabpn1 allele. This mouse is the closest available genocopy of OPMD patients. We show that Pabpn1+/A17 mice have a mild myopathic phenotype in adult and aged animals. We examined early molecular and biochemical phenotypes associated with expressing native levels of A17-PABPN1 and detected shorter poly(A) tails, modest changes in poly(A) signal (PAS) usage, and evidence of mitochondrial damage in these mice. Recent studies have suggested that a loss of PABPN1 function could contribute to muscle pathology in OPMD. To investigate a loss of function model of pathology, we generated a heterozygous Pabpn1 knock-out mouse model (Pabpn1+/Δ). Like the Pabpn1+/A17 mice, Pabpn1+/Δ mice have mild histologic defects, shorter poly(A) tails, and evidence of mitochondrial damage. However, the phenotypes detected in Pabpn1+/Δ mice only partially overlap with those detected in Pabpn1+/A17 mice. These results suggest that loss of PABPN1 function could contribute to but may not completely explain the pathology detected in Pabpn1+/A17 mice.
Collapse
Affiliation(s)
- Katherine E. Vest
- Department of Pharmacology, Emory University School of Medicine, Atlanta, GA, USA
- Department of Biology, Emory University, Atlanta, GA, USA
| | - Brittany L. Phillips
- Department of Pharmacology, Emory University School of Medicine, Atlanta, GA, USA
- Department of Biology, Emory University, Atlanta, GA, USA
| | - Ayan Banerjee
- Department of Biology, Emory University, Atlanta, GA, USA
| | - Luciano H. Apponi
- Department of Pharmacology, Emory University School of Medicine, Atlanta, GA, USA
| | - Eric B. Dammer
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Weiting Xu
- Department of Microbiology, Biochemistry, and Molecular Genetics, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Dinghai Zheng
- Department of Microbiology, Biochemistry, and Molecular Genetics, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Julia Yu
- Department of Biology, Emory University, Atlanta, GA, USA
| | - Bin Tian
- Department of Microbiology, Biochemistry, and Molecular Genetics, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Grace K. Pavlath
- Department of Pharmacology, Emory University School of Medicine, Atlanta, GA, USA
| | | |
Collapse
|
8
|
Kumar A, Kopra J, Varendi K, Porokuokka LL, Panhelainen A, Kuure S, Marshall P, Karalija N, Härma MA, Vilenius C, Lilleväli K, Tekko T, Mijatovic J, Pulkkinen N, Jakobson M, Jakobson M, Ola R, Palm E, Lindahl M, Strömberg I, Võikar V, Piepponen TP, Saarma M, Andressoo JO. GDNF Overexpression from the Native Locus Reveals its Role in the Nigrostriatal Dopaminergic System Function. PLoS Genet 2015; 11:e1005710. [PMID: 26681446 PMCID: PMC4682981 DOI: 10.1371/journal.pgen.1005710] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2015] [Accepted: 11/06/2015] [Indexed: 12/14/2022] Open
Abstract
Degeneration of nigrostriatal dopaminergic system is the principal lesion in Parkinson’s disease. Because glial cell line-derived neurotrophic factor (GDNF) promotes survival of dopamine neurons in vitro and in vivo, intracranial delivery of GDNF has been attempted for Parkinson’s disease treatment but with variable success. For improving GDNF-based therapies, knowledge on physiological role of endogenous GDNF at the sites of its expression is important. However, due to limitations of existing genetic model systems, such knowledge is scarce. Here, we report that prevention of transcription of Gdnf 3’UTR in Gdnf endogenous locus yields GDNF hypermorphic mice with increased, but spatially unchanged GDNF expression, enabling analysis of postnatal GDNF function. We found that increased level of GDNF in the central nervous system increases the number of adult dopamine neurons in the substantia nigra pars compacta and the number of dopaminergic terminals in the dorsal striatum. At the functional level, GDNF levels increased striatal tissue dopamine levels and augmented striatal dopamine release and re-uptake. In a proteasome inhibitor lactacystin-induced model of Parkinson’s disease GDNF hypermorphic mice were protected from the reduction in striatal dopamine and failure of dopaminergic system function. Importantly, adverse phenotypic effects associated with spatially unregulated GDNF applications were not observed. Enhanced GDNF levels up-regulated striatal dopamine transporter activity by at least five fold resulting in enhanced susceptibility to 6-OHDA, a toxin transported into dopamine neurons by DAT. Further, we report how GDNF levels regulate kidney development and identify microRNAs miR-9, miR-96, miR-133, and miR-146a as negative regulators of GDNF expression via interaction with Gdnf 3’UTR in vitro. Our results reveal the role of GDNF in nigrostriatal dopamine system postnatal development and adult function, and highlight the importance of correct spatial expression of GDNF. Furthermore, our results suggest that 3’UTR targeting may constitute a useful tool in analyzing gene function. Intracranial delivery of GDNF has been attempted for Parkinson’s disease (PD) treatment but with variable success. For improving GDNF-based therapies, knowledge on physiological role of endogenous GDNF at the sites of its expression is important. However, due to limitations of existing genetic model systems, such knowledge is scarce. Here, we utilize an innovative genetic approach by targeting the 3’UTR regulation of Gdnf in mice. Such animals express elevated levels of Gdnf exclusively in natively Gdnf-expressing cells, enabling dissection of endogenous GDNF functions in vivo. We show that endogenous GDNF regulates dopamine system development and function and protects mice in a rodent PD model without side effects associated with ectopic GDNF applications. Further, we report how GDNF levels regulate kidney development and identify microRNAs which control GDNF expression. Our study highlights the importance of correct spatial expression of GDNF and opens a novel approach to study gene function in mice.
Collapse
Affiliation(s)
- Anmol Kumar
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Jaakko Kopra
- Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Kärt Varendi
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | | | - Anne Panhelainen
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Satu Kuure
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Pepin Marshall
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Nina Karalija
- Department of Histology and Cell Biology, Umeå University, Umeå, Sweden
| | - Mari-Anne Härma
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Carolina Vilenius
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Kersti Lilleväli
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Triin Tekko
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Jelena Mijatovic
- Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Nita Pulkkinen
- Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Madis Jakobson
- Department of Biochemistry and Developmental Biology, Institute of Biomedicine, University of Helsinki, Helsinki, Finland
| | - Maili Jakobson
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Roxana Ola
- Department of Biochemistry and Developmental Biology, Institute of Biomedicine, University of Helsinki, Helsinki, Finland
| | - Erik Palm
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Maria Lindahl
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Ingrid Strömberg
- Department of Histology and Cell Biology, Umeå University, Umeå, Sweden
| | - Vootele Võikar
- Neuroscience Center and Department of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | - T. Petteri Piepponen
- Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Mart Saarma
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Jaan-Olle Andressoo
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
- * E-mail:
| |
Collapse
|
9
|
Cardaci S, Zheng L, MacKay G, van den Broek NJ, MacKenzie ED, Nixon C, Stevenson D, Tumanov S, Bulusu V, Kamphorst JJ, Vazquez A, Fleming S, Schiavi F, Kalna G, Blyth K, Strathdee D, Gottlieb E. Pyruvate carboxylation enables growth of SDH-deficient cells by supporting aspartate biosynthesis. Nat Cell Biol 2015; 17:1317-26. [PMID: 26302408 PMCID: PMC4591470 DOI: 10.1038/ncb3233] [Citation(s) in RCA: 216] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Accepted: 07/29/2015] [Indexed: 12/13/2022]
Abstract
Succinate dehydrogenase (SDH) is a heterotetrameric nuclear-encoded complex responsible for the oxidation of succinate to fumarate in the tricarboxylic acid cycle. Loss-of-function mutations in any of the SDH genes are associated with cancer formation. However, the impact of SDH loss on cell metabolism and the mechanisms enabling growth of SDH-defective cells are largely unknown. Here, we generated Sdhb-ablated kidney mouse cells and used comparative metabolomics and stable-isotope-labelling approaches to identify nutritional requirements and metabolic adaptations to SDH loss. We found that lack of SDH activity commits cells to consume extracellular pyruvate, which sustains Warburg-like bioenergetic features. We further demonstrated that pyruvate carboxylation diverts glucose-derived carbons into aspartate biosynthesis, thus sustaining cell growth. By identifying pyruvate carboxylase as essential for the proliferation and tumorigenic capacity of SDH-deficient cells, this study revealed a metabolic vulnerability for potential future treatment of SDH-associated malignancies.
Collapse
MESH Headings
- Animals
- Aspartic Acid/biosynthesis
- Carboxylic Acids/metabolism
- Carcinoma, Renal Cell/genetics
- Carcinoma, Renal Cell/metabolism
- Cell Line, Transformed
- Cell Proliferation
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Neoplastic/metabolism
- Cells, Cultured
- Humans
- Immunoblotting
- Kidney/cytology
- Kidney/metabolism
- Kidney Neoplasms/genetics
- Kidney Neoplasms/metabolism
- Male
- Metabolomics/methods
- Mice, 129 Strain
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Nude
- Pyruvate Carboxylase/metabolism
- Pyruvic Acid/metabolism
- RNA Interference
- Succinate Dehydrogenase/genetics
- Succinate Dehydrogenase/metabolism
Collapse
Affiliation(s)
- Simone Cardaci
- Cancer Research UK, Beatson Institute, Switchback Rd, Glasgow, G61 1BD, UK
| | - Liang Zheng
- Cancer Research UK, Beatson Institute, Switchback Rd, Glasgow, G61 1BD, UK
| | - Gillian MacKay
- Cancer Research UK, Beatson Institute, Switchback Rd, Glasgow, G61 1BD, UK
| | | | | | - Colin Nixon
- Cancer Research UK, Beatson Institute, Switchback Rd, Glasgow, G61 1BD, UK
| | - David Stevenson
- Cancer Research UK, Beatson Institute, Switchback Rd, Glasgow, G61 1BD, UK
| | - Sergey Tumanov
- Cancer Research UK, Beatson Institute, Switchback Rd, Glasgow, G61 1BD, UK
- Institute of Cancer Sciences, University of Glasgow, Glasgow, G61 1BD, UK
| | - Vinay Bulusu
- Cancer Research UK, Beatson Institute, Switchback Rd, Glasgow, G61 1BD, UK
- Institute of Cancer Sciences, University of Glasgow, Glasgow, G61 1BD, UK
| | - Jurre J. Kamphorst
- Cancer Research UK, Beatson Institute, Switchback Rd, Glasgow, G61 1BD, UK
- Institute of Cancer Sciences, University of Glasgow, Glasgow, G61 1BD, UK
| | - Alexei Vazquez
- Cancer Research UK, Beatson Institute, Switchback Rd, Glasgow, G61 1BD, UK
| | - Stewart Fleming
- Department of Pathology, University of Dundee, Ninewells Hospital, Dundee, DD1 9SY, UK
| | - Francesca Schiavi
- Veneto Institute of Oncology IRCCS, Familial cancer clinic and oncoendocrinology, Via Gattamelata 64, 35128 Padova, Italy
| | - Gabriela Kalna
- Cancer Research UK, Beatson Institute, Switchback Rd, Glasgow, G61 1BD, UK
| | - Karen Blyth
- Cancer Research UK, Beatson Institute, Switchback Rd, Glasgow, G61 1BD, UK
| | - Douglas Strathdee
- Cancer Research UK, Beatson Institute, Switchback Rd, Glasgow, G61 1BD, UK
| | - Eyal Gottlieb
- Cancer Research UK, Beatson Institute, Switchback Rd, Glasgow, G61 1BD, UK
| |
Collapse
|
10
|
Cadalbert LC, Ghaffar FN, Stevenson D, Bryson S, Vaz FM, Gottlieb E, Strathdee D. Mouse Tafazzin Is Required for Male Germ Cell Meiosis and Spermatogenesis. PLoS One 2015; 10:e0131066. [PMID: 26114544 PMCID: PMC4483168 DOI: 10.1371/journal.pone.0131066] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Accepted: 05/27/2015] [Indexed: 01/08/2023] Open
Abstract
Barth syndrome is an X-linked mitochondrial disease, symptoms of which include neutropenia and cardiac myopathy. These symptoms are the most significant clinical consequences of a disease, which is increasingly recognised to have a variable presentation. Mutation in the Taz gene in Xq28 is thought to be responsible for the condition, by altering mitochondrial lipid content and mitochondrial function. Male chimeras carrying a targeted mutation of Taz on their X-chromosome were infertile. Testes from the Taz knockout chimeras were smaller than their control counterparts and this was associated with a disruption of the progression of spermatocytes through meiosis to spermiogenesis. Taz knockout ES cells also showed a defect when differentiated to germ cells in vitro. Mutant spermatocytes failed to progress past the pachytene stage of meiosis and had higher levels of DNA double strand damage and increased levels of endogenous retrotransposon activity. Altogether these data revealed a novel role for Taz in helping to maintain genome integrity in meiosis and facilitating germ cell differentiation. We have unravelled a novel function for the Taz protein, which should contribute to an understanding of how a disruption of the Taz gene results in the complex symptoms underlying Barth Syndrome.
Collapse
Affiliation(s)
- Laurence C. Cadalbert
- Transgenic Technology Laboratory, Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, United Kingdom
| | - Farah Naz Ghaffar
- Transgenic Technology Laboratory, Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, United Kingdom
| | - David Stevenson
- Transgenic Technology Laboratory, Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, United Kingdom
| | - Sheila Bryson
- Transgenic Technology Laboratory, Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, United Kingdom
| | - Frédéric M. Vaz
- Laboratory Genetic Metabolic Diseases, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Eyal Gottlieb
- Cell Death and Metabolism Laboratory, Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, United Kingdom
| | - Douglas Strathdee
- Transgenic Technology Laboratory, Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, United Kingdom
| |
Collapse
|
11
|
Ketema M, Secades P, Kreft M, Nahidiazar L, Janssen H, Jalink K, de Pereda JM, Sonnenberg A. The rod domain is not essential for the function of plectin in maintaining tissue integrity. Mol Biol Cell 2015; 26:2402-17. [PMID: 25971800 PMCID: PMC4571296 DOI: 10.1091/mbc.e15-01-0043] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 05/06/2015] [Indexed: 11/22/2022] Open
Abstract
Plectin is a cytoskeletal linker protein that consists of a central rod domain connecting two globular domains. Rodless plectin is able to functionally compensate for the loss of full-length plectin in mice and, like full-length plectin, is able to form dimers. Epidermolysis bullosa simplex associated with late-onset muscular dystrophy (EBS-MD) is an autosomal recessive disorder resulting from mutations in the plectin gene. The majority of these mutations occur within the large exon 31 encoding the central rod domain and leave the production of a low-level rodless plectin splice variant unaffected. To investigate the function of the rod domain, we generated rodless plectin mice through conditional deletion of exon 31. Rodless plectin mice develop normally without signs of skin blistering or muscular dystrophy. Plectin localization and hemidesmosome organization are unaffected in rodless plectin mice. However, superresolution microscopy revealed a closer juxtaposition of the C-terminus of plectin to the integrin β4 subunit in rodless plectin keratinocytes. Wound healing occurred slightly faster in rodless plectin mice than in wild-type mice, and keratinocytes migration was increased in the absence of the rod domain. The faster migration of rodless plectin keratinocytes is not due to altered biochemical properties because, like full-length plectin, rodless plectin is a dimeric protein. Our data demonstrate that rodless plectin can functionally compensate for the loss of full-length plectin in mice. Thus the low expression level of plectin rather than the absence of the rod domain dictates the development of EBS-MD.
Collapse
Affiliation(s)
- Mirjam Ketema
- Division of Cell Biology, Netherlands Cancer Institute, 1066 CX Amsterdam, Netherlands
| | - Pablo Secades
- Division of Cell Biology, Netherlands Cancer Institute, 1066 CX Amsterdam, Netherlands
| | - Maaike Kreft
- Division of Cell Biology, Netherlands Cancer Institute, 1066 CX Amsterdam, Netherlands
| | - Leila Nahidiazar
- Division of Cell Biology, Netherlands Cancer Institute, 1066 CX Amsterdam, Netherlands
| | - Hans Janssen
- Division of Cell Biology, Netherlands Cancer Institute, 1066 CX Amsterdam, Netherlands
| | - Kees Jalink
- Division of Cell Biology, Netherlands Cancer Institute, 1066 CX Amsterdam, Netherlands
| | - Jose M de Pereda
- Instituto de Biología Molecular y Celular del Cancer, University of Salamanca-CSIC, E-37007 Salamanca, Spain
| | - Arnoud Sonnenberg
- Division of Cell Biology, Netherlands Cancer Institute, 1066 CX Amsterdam, Netherlands
| |
Collapse
|
12
|
Bilodeau M, Shojaie S, Ackerley C, Post M, Rossant J. Identification of a proximal progenitor population from murine fetal lungs with clonogenic and multilineage differentiation potential. Stem Cell Reports 2014; 3:634-49. [PMID: 25358791 PMCID: PMC4223706 DOI: 10.1016/j.stemcr.2014.07.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Revised: 07/22/2014] [Accepted: 07/24/2014] [Indexed: 11/22/2022] Open
Abstract
Lung development-associated diseases are major causes of morbidity and lethality in preterm infants and children. Access to the lung progenitor/stem cell populations controlling pulmonary development during embryogenesis and early postnatal years is essential to understand the molecular basis of such diseases. Using a Nkx2-1mCherry reporter mouse, we have identified and captured Nkx2-1-expressing lung progenitor cells from the proximal lung epithelium during fetal development. These cells formed clonal spheres in semisolid culture that could be maintained in vitro and demonstrated self-renewal and expansion capabilities over multiple passages. In-vitro-derived Nkx2-1-expressing clonal spheres differentiated into a polarized epithelium comprised of multiple cell lineages, including basal and secretory cells, that could repopulate decellularized lung scaffolds. Nkx2-1 expression thus defines a fetal lung epithelial progenitor cell population that can be used as a model system to study pulmonary development and associated pediatric diseases. Nkx2-1 expression can be used to isolate proximal lung progenitors The fetal proximal lung progenitors are distinct from currently known progenitors The fetal proximal lung progenitors are clonogenic and self-renewing The fetal proximal lung progenitors are multipotent for airway lineages
Collapse
Affiliation(s)
- Mélanie Bilodeau
- Program in Developmental and Stem Cell Biology, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto ON M5G 0A4, Canada
| | - Sharareh Shojaie
- Program in Physiology and Experimental Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto ON M5G 0A4, Canada; Department of Physiology, University of Toronto, Toronto ON M5S 1A8, Canada
| | - Cameron Ackerley
- Program in Physiology and Experimental Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto ON M5G 0A4, Canada; Department of Paediatric Laboratory Medicine, The Hospital for Sick Children, Toronto ON M5G 1X8, Canada
| | - Martin Post
- Program in Physiology and Experimental Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto ON M5G 0A4, Canada; Department of Physiology, University of Toronto, Toronto ON M5S 1A8, Canada
| | - Janet Rossant
- Program in Developmental and Stem Cell Biology, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto ON M5G 0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto ON M5S 1A8, Canada.
| |
Collapse
|
13
|
Lancini C, van den Berk PCM, Vissers JHA, Gargiulo G, Song JY, Hulsman D, Serresi M, Tanger E, Blom M, Vens C, van Lohuizen M, Jacobs H, Citterio E. Tight regulation of ubiquitin-mediated DNA damage response by USP3 preserves the functional integrity of hematopoietic stem cells. ACTA ACUST UNITED AC 2014; 211:1759-77. [PMID: 25113974 PMCID: PMC4144738 DOI: 10.1084/jem.20131436] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
In vivo deletion of USP3, a deubiquitinating enzyme involved in DNA damage repair, increases the incidence of spontaneous cancer and impairs the proliferation and repopulation ability of HSCs. Histone ubiquitination at DNA breaks is required for activation of the DNA damage response (DDR) and DNA repair. How the dynamic removal of this modification by deubiquitinating enzymes (DUBs) impacts genome maintenance in vivo is largely unknown. To address this question, we generated mice deficient for Ub-specific protease 3 (USP3; Usp3Δ/Δ), a histone H2A DUB which negatively regulates ubiquitin-dependent DDR signaling. Notably, USP3 deletion increased the levels of histone ubiquitination in adult tissues, reduced the hematopoietic stem cell (HSC) reserves over time, and shortened animal life span. Mechanistically, our data show that USP3 is important in HSC homeostasis, preserving HSC self-renewal, and repopulation potential in vivo and proliferation in vitro. A defective DDR and unresolved spontaneous DNA damage contribute to cell cycle restriction of Usp3Δ/Δ HSCs. Beyond the hematopoietic system, Usp3Δ/Δ animals spontaneously developed tumors, and primary Usp3Δ/Δ cells failed to preserve chromosomal integrity. These findings broadly support the regulation of chromatin ubiquitination as a key pathway in preserving tissue function through modulation of the response to genotoxic stress.
Collapse
Affiliation(s)
- Cesare Lancini
- Division of Molecular Genetics, Division of Biological Stress Response, and Division of Experimental Animal Pathology, The Netherlands Cancer Institute, 1066 CX Amsterdam, Netherlands
| | - Paul C M van den Berk
- Division of Molecular Genetics, Division of Biological Stress Response, and Division of Experimental Animal Pathology, The Netherlands Cancer Institute, 1066 CX Amsterdam, Netherlands
| | - Joseph H A Vissers
- Division of Molecular Genetics, Division of Biological Stress Response, and Division of Experimental Animal Pathology, The Netherlands Cancer Institute, 1066 CX Amsterdam, Netherlands
| | - Gaetano Gargiulo
- Division of Molecular Genetics, Division of Biological Stress Response, and Division of Experimental Animal Pathology, The Netherlands Cancer Institute, 1066 CX Amsterdam, Netherlands
| | - Ji-Ying Song
- Division of Molecular Genetics, Division of Biological Stress Response, and Division of Experimental Animal Pathology, The Netherlands Cancer Institute, 1066 CX Amsterdam, Netherlands
| | - Danielle Hulsman
- Division of Molecular Genetics, Division of Biological Stress Response, and Division of Experimental Animal Pathology, The Netherlands Cancer Institute, 1066 CX Amsterdam, Netherlands
| | - Michela Serresi
- Division of Molecular Genetics, Division of Biological Stress Response, and Division of Experimental Animal Pathology, The Netherlands Cancer Institute, 1066 CX Amsterdam, Netherlands
| | - Ellen Tanger
- Division of Molecular Genetics, Division of Biological Stress Response, and Division of Experimental Animal Pathology, The Netherlands Cancer Institute, 1066 CX Amsterdam, Netherlands
| | - Marleen Blom
- Division of Molecular Genetics, Division of Biological Stress Response, and Division of Experimental Animal Pathology, The Netherlands Cancer Institute, 1066 CX Amsterdam, Netherlands
| | - Conchita Vens
- Division of Molecular Genetics, Division of Biological Stress Response, and Division of Experimental Animal Pathology, The Netherlands Cancer Institute, 1066 CX Amsterdam, Netherlands
| | - Maarten van Lohuizen
- Division of Molecular Genetics, Division of Biological Stress Response, and Division of Experimental Animal Pathology, The Netherlands Cancer Institute, 1066 CX Amsterdam, Netherlands
| | - Heinz Jacobs
- Division of Molecular Genetics, Division of Biological Stress Response, and Division of Experimental Animal Pathology, The Netherlands Cancer Institute, 1066 CX Amsterdam, Netherlands
| | - Elisabetta Citterio
- Division of Molecular Genetics, Division of Biological Stress Response, and Division of Experimental Animal Pathology, The Netherlands Cancer Institute, 1066 CX Amsterdam, Netherlands
| |
Collapse
|
14
|
Igawa K, Kokubu C, Yusa K, Horie K, Yoshimura Y, Yamauchi K, Suemori H, Yokozeki H, Toyoda M, Kiyokawa N, Okita H, Miyagawa Y, Akutsu H, Umezawa A, Katayama I, Takeda J. Removal of reprogramming transgenes improves the tissue reconstitution potential of keratinocytes generated from human induced pluripotent stem cells. Stem Cells Transl Med 2014; 3:992-1001. [PMID: 25024429 DOI: 10.5966/sctm.2013-0179] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Human induced pluripotent stem cell (hiPSC) lines have a great potential for therapeutics because customized cells and organs can be induced from such cells. Assessment of the residual reprogramming factors after the generation of hiPSC lines is required, but an ideal system has been lacking. Here, we generated hiPSC lines from normal human dermal fibroblasts with piggyBac transposon bearing reprogramming transgenes followed by removal of the transposon by the transposase. Under this condition, we compared the phenotypes of transgene-residual and -free hiPSCs of the same genetic background. The transgene-residual hiPSCs, in which the transcription levels of the reprogramming transgenes were eventually suppressed, were quite similar to the transgene-free hiPSCs in a pluripotent state. However, after differentiation into keratinocytes, clear differences were observed. Morphological, functional, and molecular analyses including single-cell gene expression profiling revealed that keratinocytes from transgene-free hiPSC lines were more similar to normal human keratinocytes than those from transgene-residual hiPSC lines, which may be partly explained by reactivation of residual transgenes upon induction of keratinocyte differentiation. These results suggest that transgene-free hiPSC lines should be chosen for therapeutic purposes.
Collapse
Affiliation(s)
- Ken Igawa
- Department of Dermatology and Department of Social and Environmental Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan; Department of Dermatology, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan; Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, United Kingdom; Department of Embryonic Stem Cell Research, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan; Department of Reproductive Biology, Center for Regenerative Medicine, National Center for Child Health and Development, Tokyo, Japan
| | - Chikara Kokubu
- Department of Dermatology and Department of Social and Environmental Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan; Department of Dermatology, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan; Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, United Kingdom; Department of Embryonic Stem Cell Research, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan; Department of Reproductive Biology, Center for Regenerative Medicine, National Center for Child Health and Development, Tokyo, Japan
| | - Kosuke Yusa
- Department of Dermatology and Department of Social and Environmental Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan; Department of Dermatology, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan; Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, United Kingdom; Department of Embryonic Stem Cell Research, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan; Department of Reproductive Biology, Center for Regenerative Medicine, National Center for Child Health and Development, Tokyo, Japan
| | - Kyoji Horie
- Department of Dermatology and Department of Social and Environmental Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan; Department of Dermatology, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan; Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, United Kingdom; Department of Embryonic Stem Cell Research, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan; Department of Reproductive Biology, Center for Regenerative Medicine, National Center for Child Health and Development, Tokyo, Japan
| | - Yasuhide Yoshimura
- Department of Dermatology and Department of Social and Environmental Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan; Department of Dermatology, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan; Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, United Kingdom; Department of Embryonic Stem Cell Research, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan; Department of Reproductive Biology, Center for Regenerative Medicine, National Center for Child Health and Development, Tokyo, Japan
| | - Kaori Yamauchi
- Department of Dermatology and Department of Social and Environmental Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan; Department of Dermatology, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan; Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, United Kingdom; Department of Embryonic Stem Cell Research, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan; Department of Reproductive Biology, Center for Regenerative Medicine, National Center for Child Health and Development, Tokyo, Japan
| | - Hirofumi Suemori
- Department of Dermatology and Department of Social and Environmental Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan; Department of Dermatology, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan; Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, United Kingdom; Department of Embryonic Stem Cell Research, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan; Department of Reproductive Biology, Center for Regenerative Medicine, National Center for Child Health and Development, Tokyo, Japan
| | - Hiroo Yokozeki
- Department of Dermatology and Department of Social and Environmental Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan; Department of Dermatology, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan; Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, United Kingdom; Department of Embryonic Stem Cell Research, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan; Department of Reproductive Biology, Center for Regenerative Medicine, National Center for Child Health and Development, Tokyo, Japan
| | - Masashi Toyoda
- Department of Dermatology and Department of Social and Environmental Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan; Department of Dermatology, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan; Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, United Kingdom; Department of Embryonic Stem Cell Research, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan; Department of Reproductive Biology, Center for Regenerative Medicine, National Center for Child Health and Development, Tokyo, Japan
| | - Nobutaka Kiyokawa
- Department of Dermatology and Department of Social and Environmental Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan; Department of Dermatology, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan; Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, United Kingdom; Department of Embryonic Stem Cell Research, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan; Department of Reproductive Biology, Center for Regenerative Medicine, National Center for Child Health and Development, Tokyo, Japan
| | - Hajime Okita
- Department of Dermatology and Department of Social and Environmental Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan; Department of Dermatology, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan; Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, United Kingdom; Department of Embryonic Stem Cell Research, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan; Department of Reproductive Biology, Center for Regenerative Medicine, National Center for Child Health and Development, Tokyo, Japan
| | - Yoshitaka Miyagawa
- Department of Dermatology and Department of Social and Environmental Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan; Department of Dermatology, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan; Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, United Kingdom; Department of Embryonic Stem Cell Research, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan; Department of Reproductive Biology, Center for Regenerative Medicine, National Center for Child Health and Development, Tokyo, Japan
| | - Hidenori Akutsu
- Department of Dermatology and Department of Social and Environmental Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan; Department of Dermatology, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan; Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, United Kingdom; Department of Embryonic Stem Cell Research, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan; Department of Reproductive Biology, Center for Regenerative Medicine, National Center for Child Health and Development, Tokyo, Japan
| | - Akihiro Umezawa
- Department of Dermatology and Department of Social and Environmental Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan; Department of Dermatology, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan; Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, United Kingdom; Department of Embryonic Stem Cell Research, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan; Department of Reproductive Biology, Center for Regenerative Medicine, National Center for Child Health and Development, Tokyo, Japan
| | - Ichiro Katayama
- Department of Dermatology and Department of Social and Environmental Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan; Department of Dermatology, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan; Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, United Kingdom; Department of Embryonic Stem Cell Research, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan; Department of Reproductive Biology, Center for Regenerative Medicine, National Center for Child Health and Development, Tokyo, Japan
| | - Junji Takeda
- Department of Dermatology and Department of Social and Environmental Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan; Department of Dermatology, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan; Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, United Kingdom; Department of Embryonic Stem Cell Research, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan; Department of Reproductive Biology, Center for Regenerative Medicine, National Center for Child Health and Development, Tokyo, Japan
| |
Collapse
|
15
|
Ketema M, Kreft M, Secades P, Janssen H, Sonnenberg A. Nesprin-3 connects plectin and vimentin to the nuclear envelope of Sertoli cells but is not required for Sertoli cell function in spermatogenesis. Mol Biol Cell 2013; 24:2454-66. [PMID: 23761073 PMCID: PMC3727937 DOI: 10.1091/mbc.e13-02-0100] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Nesprin-3 regulates perinuclear localization of plectin and vimentin in Sertoli cells but is dispensable for Sertoli cell function in spermatogenesis. In addition, nuclear positioning and anchorage are not disturbed in nesprin-3–knockout mice. Nesprin-3 is a nuclear envelope protein that connects the nucleus to intermediate filaments by interacting with plectin. To investigate the role of nesprin-3 in the perinuclear localization of plectin, we generated nesprin-3–knockout mice and examined the effects of nesprin-3 deficiency in different cell types and tissues. Nesprin-3 and plectin are coexpressed in a variety of tissues, including peripheral nerve and muscle. The expression level of nesprin-3 in skeletal muscle is very low and decreases during myoblast differentiation in vitro. Of interest, plectin was concentrated at the nuclear envelope in only a few cell types. This was most prominent in Sertoli cells of the testis, in which nesprin-3 is required for the localization of both plectin and vimentin at the nuclear perimeter. Testicular morphology and the position of the nucleus in Sertoli cells were normal, however, in the nesprin-3–knockout mice and the mice were fertile. Furthermore, nesprin-3 was not required for the polarization and migration of mouse embryonic fibroblasts. Thus, although nesprin-3 is critical for the localization of plectin to the nuclear perimeter of Sertoli cells, the resulting link between the nuclear envelope and the intermediate filament system seems to be dispensable for normal testicular morphology and spermatogenesis.
Collapse
Affiliation(s)
- Mirjam Ketema
- Division of Cell Biology, Netherlands Cancer Institute, 1066 CX Amsterdam, Netherlands
| | | | | | | | | |
Collapse
|
16
|
The piggyBac transposon displays local and distant reintegration preferences and can cause mutations at noncanonical integration sites. Mol Cell Biol 2013; 33:1317-30. [PMID: 23358416 DOI: 10.1128/mcb.00670-12] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The DNA transposon piggyBac is widely used as a tool in mammalian experimental systems for transgenesis, mutagenesis, and genome engineering. We have characterized genome-wide insertion site preferences of piggyBac by sequencing a large set of integration sites arising from transposition from two separate genomic loci and a plasmid donor in mouse embryonic stem cells. We found that piggyBac preferentially integrates locally to the excision site when mobilized from a chromosomal location and identified other nonlocal regions of the genome with elevated insertion frequencies. piggyBac insertions were associated with expressed genes and markers of open chromatin structure and were excluded from heterochromatin. At the nucleotide level, piggyBac prefers to insert into TA-rich regions within a broader GC-rich context. We also found that piggyBac can insert into sites other than its known TTAA insertion site at a low frequency (2%). Such insertions introduce mismatches that are repaired with signatures of host cell repair pathways. Transposons could be mobilized from plasmids with the observed noncanonical flanking regions, indicating that piggyBac could generate point mutations in the genome.
Collapse
|
17
|
SOX4 transcriptionally regulates multiple SEMA3/plexin family members and promotes tumor growth in pancreatic cancer. PLoS One 2012; 7:e48637. [PMID: 23251334 PMCID: PMC3520963 DOI: 10.1371/journal.pone.0048637] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2012] [Accepted: 10/01/2012] [Indexed: 01/13/2023] Open
Abstract
Semaphorin signaling through Plexin frequently participates in tumorigenesis and malignant progression in various types of cancer. In particular, the role of semaphorin signaling in pancreatic ductal adenocarcinoma (PDAC) remains unexplored, despite a high likelihood of metastasis and mortality. Unlike other epithelial malignancies that often express a small number of specific genes in the Semaphorin/Plexin family, five or more are often expressed in human PDAC. Such concomitant expression of these SEMA3/Plexin family members is not a result of gene amplification, but (at least partially) from increased gene transcription activated by SOX4 de novo expressed in PDAC. Via chromatin-immunoprecipitation, luciferase promoter activity assay and electrophoresis mobility shift assay, SOX4 is demonstrated to bind to the consensus site at the promoter of each SEMA3 and Plexin gene to enhance transcription activity. Conversely, RNAi-knockdown of SOX4 in PDAC cell lines results in decreased expression of SEMA3/Plexin family members and is associated with restricted tumor growth both in vitro and in SCID mice. We further demonstrate that SOX4 levels parallel with the summed expression of SEMA3/Plexin family members (P = 0.033, NPar Kruskal-Wallis one-way analysis), which also correlates with poor survival in human PDAC (P = 0.0409, Kaplan-Meier analysis). Intriguingly, miR-129-2 and miR-335, both of which target SOX4 for degradation, are co-repressed in human PDAC cases associated with up-regulated SOX4 in a statistically significant way. In conclusion, we disclose a miR-129-2(miR-335)/SOX4/Semaphorin-Plexin regulatory axis in the tumorigenesis of pancreatic cancer.
Collapse
|
18
|
Su CY, Bay SN, Mariani LE, Hillman MJ, Caspary T. Temporal deletion of Arl13b reveals that a mispatterned neural tube corrects cell fate over time. Development 2012; 139:4062-71. [PMID: 23014696 DOI: 10.1242/dev.082321] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Cilia are necessary for sonic hedgehog (Shh) signaling, which is required to pattern the neural tube. We know that ventral neural cell fates are defined by a specific cohort of transcription factors that are induced by distinct thresholds of Shh activity mediated by opposing gradients of Gli activator (GliA) and Gli repressor (GliR). Despite this understanding, the role of Shh as an instructive morphogen is viewed as increasingly complex, with current models integrating positive inputs in terms of ligand concentration and time, along with negative feedback via the downstream gene regulatory network. To investigate the relative contributions of the positive and negative inputs from Shh signaling in neural patterning, we took advantage of a protein that uncouples the regulation of GliA and GliR: the cilia protein ADP-ribosylation factor-like 13b (Arl13b). By deleting Arl13b in mouse, we induced low-level constitutive GliA function at specific developmental stages and defined a crucial period prior to E10.5 when shifts in the level of GliA cause cells to change their fate. Strikingly, we found that improperly patterned cells in these mice converted to the wild-type pattern by E12.5. We further showed that the recovery of patterning did not occur when we also deleted Gli3, the primary GliR in the neural tube, revealing a crucial role of Gli3 in the maintenance of neural patterning.
Collapse
Affiliation(s)
- Chen-Ying Su
- Graduate Program in Genetics and Molecular Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | | | | | | | | |
Collapse
|
19
|
Dutton SB, Makinson CD, Papale LA, Shankar A, Balakrishnan B, Nakazawa K, Escayg A. Preferential inactivation of Scn1a in parvalbumin interneurons increases seizure susceptibility. Neurobiol Dis 2012; 49:211-20. [PMID: 22926190 DOI: 10.1016/j.nbd.2012.08.012] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2012] [Revised: 07/30/2012] [Accepted: 08/16/2012] [Indexed: 01/29/2023] Open
Abstract
Voltage-gated sodium channels (VGSCs) are essential for the generation and propagation of action potentials in electrically excitable cells. Dominant mutations in SCN1A, which encodes the Nav1.1 VGSC α-subunit, underlie several forms of epilepsy, including Dravet syndrome (DS) and genetic epilepsy with febrile seizures plus (GEFS+). Electrophysiological analyses of DS and GEFS+ mouse models have led to the hypothesis that SCN1A mutations reduce the excitability of inhibitory cortical and hippocampal interneurons. To more directly examine the relative contribution of inhibitory interneurons and excitatory pyramidal cells to SCN1A-derived epilepsy, we first compared the expression of Nav1.1 in inhibitory parvalbumin (PV) interneurons and excitatory neurons from P22 mice using fluorescent immunohistochemistry. In the hippocampus and neocortex, 69% of Nav1.1 immunoreactive neurons were also positive for PV. In contrast, 13% and 5% of Nav1.1 positive cells in the hippocampus and neocortex, respectively, were found to co-localize with excitatory cells identified by CaMK2α immunoreactivity. Next, we reduced the expression of Scn1a in either a subset of interneurons (mainly PV interneurons) or excitatory cells by crossing mice heterozygous for a floxed Scn1a allele to either the Ppp1r2-Cre or EMX1-Cre transgenic lines, respectively. The inactivation of one Scn1a allele in interneurons of the neocortex and hippocampus was sufficient to reduce thresholds to flurothyl- and hyperthermia-induced seizures, whereas thresholds were unaltered following inactivation in excitatory cells. Reduced interneuron Scn1a expression also resulted in the generation of spontaneous seizures. These findings provide direct evidence for an important role of PV interneurons in the pathogenesis of Scn1a-derived epilepsies.
Collapse
Affiliation(s)
- Stacey B Dutton
- Department of Human Genetics, Emory University, Atlanta, GA, 30022, USA
| | | | - Ligia A Papale
- Department of Human Genetics, Emory University, Atlanta, GA, 30022, USA
| | - Anupama Shankar
- Department of Human Genetics, Emory University, Atlanta, GA, 30022, USA
| | | | - Kazu Nakazawa
- Unit on Genetics of Cognition and Behavior, National Institute of Mental Health, Bethesda, MD, USA
| | - Andrew Escayg
- Department of Human Genetics, Emory University, Atlanta, GA, 30022, USA.
| |
Collapse
|
20
|
Sun X, Fu X, Li J, Xing C, Martin DW, Zhang HH, Chen Z, Dong JT. Heterozygous deletion of Atbf1 by the Cre-loxP system in mice causes preweaning mortality. Genesis 2012; 50:819-27. [PMID: 22644989 DOI: 10.1002/dvg.22041] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2011] [Revised: 05/02/2012] [Accepted: 05/21/2012] [Indexed: 11/10/2022]
Abstract
ATBF1 is a large nuclear protein that contains multiple zinc-finger motifs and four homeodomains. In mammals, ATBF1 regulates differentiation, and its mutation and/or downregulation is involved in tumorigenesis in several organs. To gain more insight into the physiological functions of ATBF1, we generated and validated a conditional allele of mouse Atbf1 in which exons 7 and 8 were flanked by loxP sites (Atbf1(flox) ). Germline deletion of a single Atbf1 allele was achieved by breeding to EIIa-cre transgenic mice, and Atbf1 heterozygous mice displayed reduced body weight, preweaning mortality, increased cell proliferation, and attenuated cytokeratin 18 expression, indicating haploinsufficiency of Atbf1. Floxed Atbf1 mice will help us understand such biological processes as neuronal differentiation and tumorigenesis.
Collapse
Affiliation(s)
- Xiaodong Sun
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Winship Cancer Institute, Atlanta, GA 30322, USA
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Lu TX, Hartner J, Lim EJ, Fabry V, Mingler MK, Cole ET, Orkin SH, Aronow BJ, Rothenberg ME. MicroRNA-21 limits in vivo immune response-mediated activation of the IL-12/IFN-gamma pathway, Th1 polarization, and the severity of delayed-type hypersensitivity. THE JOURNAL OF IMMUNOLOGY 2011; 187:3362-73. [PMID: 21849676 DOI: 10.4049/jimmunol.1101235] [Citation(s) in RCA: 285] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
An altered balance between Th1 and Th2 cytokines is responsible for a variety of immunoinflammatory disorders such as asthma, yet the role of posttranscriptional mechanisms, such as those mediated by microRNAs (miRs), in adjusting the relative magnitude and balance of Th cytokine expression have been largely unexplored. In this study, we show that miR-21 has a central role in setting a balance between Th1 and Th2 responses to Ags. Targeted ablation of miR-21 in mice led to reduced lung eosinophilia after allergen challenge, with a broadly reprogrammed immunoactivation transcriptome and significantly increased levels of the Th1 cytokine IFN-γ. Biological network-based transcriptome analysis of OVA-challenged miR-21(-/-) mice identified an unexpected prominent dysregulation of IL-12/IFN-γ pathways as the most significantly affected in the lungs, with a key role for miR-21 in IFN-γ signaling and T cell polarization, consistent with a functional miR-21 binding site in IL-12p35. In support of these hypotheses, miR-21 deficiency led dendritic cells to produce more IL-12 after LPS stimulation and OVA-challenged CD4(+) T lymphocytes to produce increased IFN-γ and decreased IL-4. Further, loss of miR-21 significantly enhanced the Th1-associated delayed-type hypersensitivity cutaneous responses. Thus, our results define miR-21 as a major regulator of Th1 versus Th2 responses, defining a new mechanism for regulating polarized immunoinflammatory responses.
Collapse
Affiliation(s)
- Thomas X Lu
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
van der Weyden L, Giotopoulos G, Rust AG, Matheson LS, van Delft FW, Kong J, Corcoran AE, Greaves MF, Mullighan CG, Huntly BJ, Adams DJ. Modeling the evolution of ETV6-RUNX1-induced B-cell precursor acute lymphoblastic leukemia in mice. Blood 2011; 118:1041-51. [PMID: 21628403 PMCID: PMC3622520 DOI: 10.1182/blood-2011-02-338848] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The t(12;21) translocation that generates the ETV6-RUNX1 (TEL-AML1) fusion gene, is the most common chromosomal rearrangement in childhood cancer and is exclusively associated with B-cell precursor acute lymphoblastic leukemia (BCP-ALL). The translocation arises in utero and is necessary but insufficient for the development of leukemia. Single-nucleotide polymorphism array analysis of ETV6-RUNX1 patient samples has identified multiple additional genetic alterations; however, the role of these lesions in leukemogenesis remains undetermined. Moreover, murine models of ETV6-RUNX1 ALL that faithfully recapitulate the human disease are lacking. To identify novel genes that cooperate with ETV6-RUNX1 in leukemogenesis, we generated a mouse model that uses the endogenous Etv6 locus to coexpress the Etv6-RUNX1 fusion and Sleeping Beauty transposase. An insertional mutagenesis screen was performed by intercrossing these mice with those carrying a Sleeping Beauty transposon array. In contrast to previous models, a substantial proportion (20%) of the offspring developed BCP-ALL. Isolation of the transposon insertion sites identified genes known to be associated with BCP-ALL, including Ebf1 and Epor, in addition to other novel candidates. This is the first mouse model of ETV6-RUNX1 to develop BCP-ALL and provides important insight into the cooperating genetic alterations in ETV6-RUNX1 leukemia.
Collapse
|
23
|
Wit N, Krijger PHL, van den Berk PCM, Jacobs H. Lysine residue 185 of Rad1 is a topological but not a functional counterpart of lysine residue 164 of PCNA. PLoS One 2011; 6:e16669. [PMID: 21304913 PMCID: PMC3031632 DOI: 10.1371/journal.pone.0016669] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2010] [Accepted: 01/10/2011] [Indexed: 12/11/2022] Open
Abstract
Monoubiquitylation of the homotrimeric DNA sliding clamp PCNA at lysine residue 164 (PCNAK164) is a highly conserved, DNA damage-inducible process that is mediated by the E2/E3 complex Rad6/Rad18. This ubiquitylation event recruits translesion synthesis (TLS) polymerases capable of replicating across damaged DNA templates. Besides PCNA, the Rad6/Rad18 complex was recently shown in yeast to ubiquitylate also 9-1-1, a heterotrimeric DNA sliding clamp composed of Rad9, Rad1, and Hus1 in a DNA damage-inducible manner. Based on the highly similar crystal structures of PCNA and 9-1-1, K185 of Rad1 (Rad1K185) was identified as the only topological equivalent of PCNAK164. To investigate a potential role of posttranslational modifications of Rad1K185 in DNA damage management, we here generated a mouse model with a conditional deletable Rad1K185R allele. The Rad1K185 residue was found to be dispensable for Chk1 activation, DNA damage survival, and class switch recombination of immunoglobulin genes as well as recruitment of TLS polymerases during somatic hypermutation of immunoglobulin genes. Our data indicate that Rad1K185 is not a functional counterpart of PCNAK164.
Collapse
Affiliation(s)
- Niek Wit
- Division of Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Peter H. L. Krijger
- Division of Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | - Heinz Jacobs
- Division of Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- * E-mail:
| |
Collapse
|
24
|
Tachibana-Konwalski K, Godwin J, van der Weyden L, Champion L, Kudo NR, Adams DJ, Nasmyth K. Rec8-containing cohesin maintains bivalents without turnover during the growing phase of mouse oocytes. Genes Dev 2010; 24:2505-16. [PMID: 20971813 PMCID: PMC2975927 DOI: 10.1101/gad.605910] [Citation(s) in RCA: 189] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2010] [Accepted: 09/23/2010] [Indexed: 12/13/2022]
Abstract
During female meiosis, bivalent chromosomes are thought to be held together from birth until ovulation by sister chromatid cohesion mediated by cohesin complexes whose ring structure depends on kleisin subunits, either Rec8 or Scc1. Because cohesion is established at DNA replication in the embryo, its maintenance for such a long time may require cohesin turnover. To address whether Rec8- or Scc1-containing cohesin holds bivalents together and whether it turns over, we created mice whose kleisin subunits can be cleaved by TEV protease. We show by microinjection experiments and confocal live-cell imaging that Rec8 cleavage triggers chiasmata resolution during meiosis I and sister centromere disjunction during meiosis II, while Scc1 cleavage triggers sister chromatid disjunction in the first embryonic mitosis, demonstrating a dramatic transition from Rec8- to Scc1-containing cohesin at fertilization. Crucially, activation of an ectopic Rec8 transgene during the growing phase of Rec8(TEV)(/TEV) oocytes does not prevent TEV-mediated bivalent destruction, implying little or no cohesin turnover for ≥2 wk during oocyte growth. We suggest that the inability of oocytes to regenerate cohesion may contribute to age-related meiosis I errors.
Collapse
Affiliation(s)
| | - Jonathan Godwin
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, United Kingdom
| | | | - Lysie Champion
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, United Kingdom
| | - Nobuaki R. Kudo
- Institute of Reproductive and Developmental Biology, Imperial College London, London W12 0NN, United Kingdom
| | - David J. Adams
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, United Kingdom
| | - Kim Nasmyth
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, United Kingdom
| |
Collapse
|
25
|
Adams DJ, van der Weyden L. Contemporary approaches for modifying the mouse genome. Physiol Genomics 2008; 34:225-38. [PMID: 18559964 PMCID: PMC2519963 DOI: 10.1152/physiolgenomics.90242.2008] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2008] [Accepted: 06/11/2008] [Indexed: 12/05/2022] Open
Abstract
The mouse is a premiere experimental organism that has contributed significantly to our understanding of vertebrate biology. Manipulation of the mouse genome via embryonic stem (ES) cell technology makes it possible to engineer an almost limitless repertoire of mutations to model human disease and assess gene function. In this review we outline recent advances in mouse experimental genetics and provide a "how-to" guide for those people wishing to access this technology. We also discuss new technologies, such as transposon-mediated mutagenesis, and resources of targeting vectors and ES cells, which are likely to dramatically accelerate the pace with which we can assess gene function in vivo, and the progress of forward and reverse genetic screens in mice.
Collapse
Affiliation(s)
- David J Adams
- Experimental Cancer Genetics, The Wellcome Trust Sanger Institute, Hinxton, United Kingdom
| | | |
Collapse
|
26
|
Neufeld G, Kessler O. The semaphorins: versatile regulators of tumour progression and tumour angiogenesis. Nat Rev Cancer 2008; 8:632-45. [PMID: 18580951 DOI: 10.1038/nrc2404] [Citation(s) in RCA: 301] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The semaphorins and their receptors, the neuropilins and the plexins, were originally characterized as constituents of the complex regulatory system responsible for the guidance of axons during the development of the central nervous system. However, a growing body of evidence indicates that various semaphorins can either promote or inhibit tumour progression through the promotion or inhibition of processes such as tumour angiogenesis, tumour metastasis and tumour cell survival. This Review focuses on the emerging role of the semaphorins in cancer.
Collapse
Affiliation(s)
- Gera Neufeld
- Cancer Research and Vascular Biology Center, The Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, P.O. Box 9679, 1 Efron Street, Haifa, 31096, Israel.
| | | |
Collapse
|
27
|
Normal germ line establishment in mice carrying a deletion of the Ifitm/Fragilis gene family cluster. Mol Cell Biol 2008; 28:4688-96. [PMID: 18505827 DOI: 10.1128/mcb.00272-08] [Citation(s) in RCA: 111] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The family of interferon-inducible transmembrane proteins (Ifitm) consists of five highly sequence-related cell surface proteins, which are implicated in diverse cellular processes. Ifitm genes are conserved, widely expressed, and characteristically found in genomic clusters, such as the 67-kb Ifitm family locus on mouse chromosome 7. Recently, Ifitm1 and Ifitm3 have been suggested to mediate migration of early primordial germ cells (PGCs), a process that is little understood. To investigate Ifitm function during germ cell development, we used targeted chromosome engineering to generate mutants which either lack the entire Ifitm locus or carry a disrupted Ifitm3 gene only. Here we show that the mutations have no detectable effects on development of the germ line or on the generation of live young. Hence, contrary to previous reports, Ifitm genes are not essential for PGC migration. The Ifitm family is a striking example of a conserved gene cluster which appears to be functionally redundant during development.
Collapse
|
28
|
Neufeld G, Lange T, Varshavsky A, Kessler O. Semaphorin signaling in vascular and tumor biology. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2007; 600:118-31. [PMID: 17607951 DOI: 10.1007/978-0-387-70956-7_10] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/16/2023]
Abstract
The neuropilins were originally characterized as cell membrane receptors that bind axon guidance factors belonging to the class-3 semaphorin subfamily. To transduce semaphorin signals, they form complexes with members of the plexin receptor family in which neuropilins serve as the ligand binding components and the plexins as the signal transducing components. The neuropilins were subsequently found to double as receptors for specific heparin binding splice forms of vascular endothelial growth factor (VEGF), and to be expressed on endothelial cells. This finding suggested that semaphorins may function as modulators of angiogenesis. It was recently found that several types of semaphorins such as semaphorin-3F function as inhibitors of angiogenesis while others, most notably semaphorin-4D, function as angiogenic factors. Furthermore, semaphorins such as semaphorin-3F and semaphorin-3B have been characterized as tumor suppressors and have been found to exert direct effects upon tumor cells. In this chapter we cover recent developments in this rapidly developing field of research.
Collapse
Affiliation(s)
- Gera Neufeld
- Cancer and Vascular Biology Research Center, Rappaport Research Institute in the Medical Sciences, The Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, 1 Efron St., P.O. Box 9679, Haifa, 31096, Israel.
| | | | | | | |
Collapse
|
29
|
Perez-Campo FM, Spencer HL, Elder RH, Stern PL, Ward CM. Novel vectors for homologous recombination strategies in mouse embryonic stem cells: an ES cell line expressing EGFP under control of the 5T4 promoter. Exp Cell Res 2007; 313:3604-15. [PMID: 17765223 DOI: 10.1016/j.yexcr.2007.07.021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2007] [Revised: 07/12/2007] [Accepted: 07/13/2007] [Indexed: 01/28/2023]
Abstract
The use of gene mutation/knock-out strategies in mouse embryonic stem (ES) cells has revolutionized the study of gene function in ES cells and embryonic development. However, the construction of vectors for homologous recombination strategies requires considerable expertise and time. We describe two novel vectors that can generate site specific knock-out or EGFP knock-in ES cells within 6 weeks from construct design to identification of positive ES cell clones. As proof-of-principle, we have utilized the knock-out targeting vector to modify the NEIL2 locus in ES cells. In addition, using the knock-in vector, we have inserted EGFP downstream of the 5T4 oncofetal antigen promoter in ES cells (5T4-GFP ES cells). Undifferentiated 5T4-GFP ES cells lack EGFP and maintain expression of the pluripotent markers OCT-4 and NANOG. Upon differentiation, EGFP expression is increased in 5T4-GFP ES cells and this correlates with 5T4 transcript expression of the unmodified allele, loss of Nanog and Oct-4 transcripts and upregulation of differentiation-associated transcripts. Furthermore, we demonstrate that fluorescent activated cell sorting of 5T4-GFP ES cells allows isolation of pluripotent or differentiated cells from a heterogeneous population. These vectors provide researchers with a rapid method of modifying specific ES cell genes to study cellular differentiation and embryonic development.
Collapse
Affiliation(s)
- Flor M Perez-Campo
- Stem Cell Biology Group, Paterson Institute for Cancer Research, Christie Hospital NHS Trust, Wilmslow Road, Manchester, UK
| | | | | | | | | |
Collapse
|
30
|
Cadiñanos J, Bradley A. Generation of an inducible and optimized piggyBac transposon system. Nucleic Acids Res 2007; 35:e87. [PMID: 17576687 PMCID: PMC1919496 DOI: 10.1093/nar/gkm446] [Citation(s) in RCA: 217] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2007] [Revised: 05/14/2007] [Accepted: 05/18/2007] [Indexed: 01/12/2023] Open
Abstract
Genomic studies in the mouse have been slowed by the lack of transposon-mediated mutagenesis. However, since the resurrection of Sleeping Beauty (SB), the possibility of performing forward genetics in mice has been reinforced. Recently, piggyBac (PB), a functional transposon from insects, was also described to work in mammals. As the activity of PB is higher than that of SB11 and SB12, two hyperactive SB transposases, we have characterized and improved the PB system in mouse ES cells. We have generated a mouse codon-optimized version of the PB transposase coding sequence (CDS) which provides transposition levels greater than the original. We have also found that the promoter sequence predicted in the 5'-terminal repeat of the PB transposon is active in the mammalian context. Finally, we have engineered inducible versions of the optimized piggyBac transposase fused with ERT2. One of them, when induced, provides higher levels of transposition than the native piggyBac CDS, whereas in the absence of induction its activity is indistinguishable from background. We expect that these tools, adaptable to perform mouse-germline mutagenesis, will facilitate the identification of genes involved in pathological and physiological processes, such as cancer or ES cell differentiation.
Collapse
Affiliation(s)
| | - Allan Bradley
- The Wellcome Trust Sanger Institute. Wellcome Trust Genome Campus, Hinxton, Cambridgeshire, CB10 1SA, UK
| |
Collapse
|
31
|
van Meeteren LA, Ruurs P, Stortelers C, Bouwman P, van Rooijen MA, Pradère JP, Pettit TR, Wakelam MJO, Saulnier-Blache JS, Mummery CL, Moolenaar WH, Jonkers J. Autotaxin, a secreted lysophospholipase D, is essential for blood vessel formation during development. Mol Cell Biol 2006; 26:5015-22. [PMID: 16782887 PMCID: PMC1489177 DOI: 10.1128/mcb.02419-05] [Citation(s) in RCA: 441] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Autotaxin (ATX), or nucleotide pyrophosphatase-phosphodiesterase 2, is a secreted lysophospholipase D that promotes cell migration, metastasis, and angiogenesis. ATX generates lysophosphatidic acid (LPA), a lipid mitogen and motility factor that acts on several G protein-coupled receptors. Here we report that ATX-deficient mice die at embryonic day 9.5 (E9.5) with profound vascular defects in yolk sac and embryo resembling the Galpha13 knockout phenotype. Furthermore, at E8.5, ATX-deficient embryos showed allantois malformation, neural tube defects, and asymmetric headfolds. The onset of these abnormalities coincided with increased expression of ATX and LPA receptors in normal embryos. ATX heterozygous mice appear healthy but show half-normal ATX activity and plasma LPA levels. Our results reveal a critical role for ATX in vascular development, indicate that ATX is the major LPA-producing enzyme in vivo, and suggest that the vascular defects in ATX-deficient embryos may be explained by loss of LPA signaling through Galpha13.
Collapse
Affiliation(s)
- Laurens A van Meeteren
- Division of Cellular Biochemistry, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Piechotta K, Dudanova I, Missler M. The resilient synapse: insights from genetic interference of synaptic cell adhesion molecules. Cell Tissue Res 2006; 326:617-42. [PMID: 16855838 DOI: 10.1007/s00441-006-0267-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2006] [Accepted: 05/31/2006] [Indexed: 01/05/2023]
Abstract
Synaptic cell adhesion molecules (SCAMs) are mostly membrane-anchored molecules with extracellular domains that extend into the synaptic cleft. Prototypical SCAMs interact with homologous or heterologous molecules on the surface of adjacent cells, ensuring the precise apposition of pre- and postsynaptic elements. More recent definitions of SCAMs often include molecules involved in axon pathfinding, cell recognition and synaptic differentiation events, making SCAMs functionally and molecularly a highly diverse group. In this review, we summarize the proposed in vivo functions of a large variety of SCAMs. We mainly focus on results obtained from analyses of genetic model organisms, mostly mouse knockout mutants, lacking expression of the respective candidate genes. In contrast to the substantial effect yielded by some knockouts of molecules involved in synaptic vesicle release, no SCAM mutant has been reported thus far that shows a prominently altered structure of the majority of synapses or even lacks synapses altogether. This surprising resilience of synaptic structure might be explained by a high redundancy between different SCAMs, by the assumption that the crucial molecular players in synapse structure have yet to be discovered or by a grand variability in the mechanisms of synapse formation that underlies the diversity of synapses. Whatever the final answer turns out to be, the genetic dissection of the SCAM superfamilies has led to a much better understanding of the different steps required to form, differentiate and modify a synapse.
Collapse
Affiliation(s)
- Kerstin Piechotta
- Center for Physiology and Pathophysiology, Georg-August University, Humboldtallee 23, 37073 Göttingen, Germany
| | | | | |
Collapse
|
33
|
van der Weyden L, Arends MJ, Chausiaux OE, Ellis PJ, Lange UC, Surani MA, Affara N, Murakami Y, Adams DJ, Bradley A. Loss of TSLC1 causes male infertility due to a defect at the spermatid stage of spermatogenesis. Mol Cell Biol 2006; 26:3595-609. [PMID: 16611999 PMCID: PMC1447413 DOI: 10.1128/mcb.26.9.3595-3609.2006] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2005] [Revised: 10/01/2005] [Accepted: 01/30/2006] [Indexed: 12/22/2022] Open
Abstract
Tumor suppressor of lung cancer 1 (TSLC1), also known as SgIGSF, IGSF4, and SynCAM, is strongly expressed in spermatogenic cells undergoing the early and late phases of spermatogenesis (spermatogonia to zygotene spermatocytes and elongating spermatids to spermiation). Using embryonic stem cell technology to generate a null mutation of Tslc1 in mice, we found that Tslc1 null male mice were infertile. Tslc1 null adult testes showed that spermatogenesis had arrested at the spermatid stage, with degenerating and apoptotic spermatids sloughing off into the lumen. In adult mice, Tslc1 null round spermatids showed evidence of normal differentiation (an acrosomal cap and F-actin polarization indistinguishable from that of wild-type spermatids); however, the surviving spermatozoa were immature, malformed, found at very low levels in the epididymis, and rarely motile. Analysis of the first wave of spermatogenesis in Tslc1 null mice showed a delay in maturation by day 22 and degeneration of round spermatids by day 28. Expression profiling of the testes revealed that Tslc1 null mice showed increases in the expression levels of genes involved in apoptosis, adhesion, and the cytoskeleton. Taken together, these data show that Tslc1 is essential for normal spermatogenesis in mice.
Collapse
Affiliation(s)
- Louise van der Weyden
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SA, United Kingdom
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Langerak P, Nygren AOH, Schouten JP, Jacobs H. Rapid and quantitative detection of homologous and non-homologous recombination events using three oligonucleotide MLPA. Nucleic Acids Res 2005; 33:e188. [PMID: 16340005 PMCID: PMC1310898 DOI: 10.1093/nar/gni187] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Embryonic stem (ES) cell technology allows modification of the mouse germline from large deletions and insertions to single nucleotide substitutions by homologous recombination. Identification of these rare events demands an accurate and fast detection method. Current methods for detection rely on Southern blotting and/or conventional PCR. Both the techniques have major drawbacks, Southern blotting is time-consuming and PCR can generate false positives. As an alternative, we here demonstrate a novel approach of Multiplex Ligation-dependent Probe Amplification (MLPA) as a quick, quantitative and reliable method for the detection of homologous, non-homologous and incomplete recombination events in ES cell clones. We have adapted MLPA to detect homologous recombinants in ES cell clones targeted with two different constructs: one introduces a single nucleotide change in the PCNA gene and the other allows for a conditional inactivation of the wild-type PCNA allele. By using MLPA probes consisting of three oligonucleotides we were able to simultaneously detect and quantify both wild-type and mutant alleles.
Collapse
Affiliation(s)
| | | | | | - Heinz Jacobs
- To whom correspondence should be addressed. Tel: +31 20 512 2066; Fax: +31 20 512 2057;
| |
Collapse
|
35
|
van der Weyden L, Tachibana KK, Gonzalez MA, Adams DJ, Ng BL, Petty R, Venkitaraman AR, Arends MJ, Bradley A. The RASSF1A isoform of RASSF1 promotes microtubule stability and suppresses tumorigenesis. Mol Cell Biol 2005; 25:8356-67. [PMID: 16135822 PMCID: PMC1234312 DOI: 10.1128/mcb.25.18.8356-8367.2005] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The RASSF1A isoform of RASSF1 is frequently inactivated by epigenetic alterations in human cancers, but it remains unclear if and how it acts as a tumor suppressor. RASSF1A overexpression reduces in vitro colony formation and the tumorigenicity of cancer cell lines in vivo. Conversely, RASSF1A knockdown causes multiple mitotic defects that may promote genomic instability. Here, we have used a genetic approach to address the function of RASSF1A as a tumor suppressor in vivo by targeted deletion of Rassf1A in the mouse. Rassf1A null mice were viable and fertile and displayed no pathological abnormalities. Rassf1A null embryonic fibroblasts displayed an increased sensitivity to microtubule depolymerizing agents. No overtly altered cell cycle parameters or aberrations in centrosome number were detected in Rassf1A null fibroblasts. Rassf1A null fibroblasts did not show increased sensitivity to microtubule poisons or DNA-damaging agents and showed no evidence of gross genomic instability, suggesting that cellular responses to genotoxins were unaffected. Rassf1A null mice showed an increased incidence of spontaneous tumorigenesis and decreased survival rate compared with wild-type mice. Irradiated Rassf1A null mice also showed increased tumor susceptibility, particularly to tumors associated with the gastrointestinal tract, compared with wild-type mice. Thus, our results demonstrate that Rassf1A acts as a tumor suppressor gene.
Collapse
Affiliation(s)
- L van der Weyden
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SA, United Kingdom
| | | | | | | | | | | | | | | | | |
Collapse
|