1
|
Wang J, Gao G, Wang D. Developing AAV-delivered nonsense suppressor tRNAs for neurological disorders. Neurotherapeutics 2024; 21:e00391. [PMID: 38959711 PMCID: PMC11269797 DOI: 10.1016/j.neurot.2024.e00391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 05/29/2024] [Accepted: 06/19/2024] [Indexed: 07/05/2024] Open
Abstract
Adeno-associated virus (AAV)-based gene therapy is a clinical stage therapeutic modality for neurological disorders. A common genetic defect in myriad monogenic neurological disorders is nonsense mutations that account for about 11% of all human pathogenic mutations. Stop codon readthrough by suppressor transfer RNA (sup-tRNA) has long been sought as a potential gene therapy approach to target nonsense mutations, but hindered by inefficient in vivo delivery. The rapid advances in AAV delivery technology have not only powered gene therapy development but also enabled in vivo preclinical assessment of a range of nucleic acid therapeutics, such as sup-tRNA. Compared with conventional AAV gene therapy that delivers a transgene to produce therapeutic proteins, AAV-delivered sup-tRNA has several advantages, such as small gene sizes and operating within the endogenous gene expression regulation, which are important considerations for treating some neurological disorders. This review will first examine sup-tRNA designs and delivery by AAV vectors. We will then analyze how AAV-delivered sup-tRNA can potentially address some neurological disorders that are challenging to conventional gene therapy, followed by discussing available mouse models of neurological diseases for in vivo preclinical testing. Potential challenges for AAV-delivered sup-tRNA to achieve therapeutic efficacy and safety will also be discussed.
Collapse
Affiliation(s)
- Jiaming Wang
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA; Department of Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Guangping Gao
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA; Department of Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA.
| | - Dan Wang
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA; RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA.
| |
Collapse
|
2
|
Taylor A, Prasad A, Mueller RL. Amphibian Segmentation Clock Models Suggest How Large Genome and Cell Sizes Slow Developmental Rate. Integr Org Biol 2024; 6:obae021. [PMID: 39006893 PMCID: PMC11245677 DOI: 10.1093/iob/obae021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 05/20/2024] [Accepted: 06/18/2024] [Indexed: 07/16/2024] Open
Abstract
Evolutionary increases in genome size, cell volume, and nuclear volume have been observed across the tree of life, with positive correlations documented between all three traits. Developmental tempo slows as genomes, nuclei, and cells increase in size, yet the driving mechanisms are poorly understood. To bridge this gap, we use a mathematical model of the somitogenesis clock to link slowed developmental tempo with changes in intra-cellular gene expression kinetics induced by increasing genome size and nuclear volume. We adapt a well-known somitogenesis clock model to two model amphibian species that vary 10-fold in genome size: Xenopus laevis (3.1 Gb) and Ambystoma mexicanum (32 Gb). Based on simulations and backed by analytical derivations, we identify parameter changes originating from increased genome and nuclear size that slow gene expression kinetics. We simulate biological scenarios for which these parameter changes mathematically recapitulate slowed gene expression in A. mexicanum relative to X. laevis, and we consider scenarios for which additional alterations in gene product stability and chromatin packing are necessary. Results suggest that slowed degradation rates as well as changes induced by increasing nuclear volume and intron length, which remain relatively unexplored, are significant drivers of slowed developmental tempo.
Collapse
Affiliation(s)
- A Taylor
- Department of Biology, Colorado State University, Fort Collins, CO 80523, USA
| | - A Prasad
- Department of Chemical and Biological Engineering, Colorado State University, Fort Collins, CO 80523, USA
| | - R Lockridge Mueller
- Department of Biology, Colorado State University, Fort Collins, CO 80523, USA
| |
Collapse
|
3
|
Grønbæk-Thygesen M, Hartmann-Petersen R. Cellular and molecular mechanisms of aspartoacylase and its role in Canavan disease. Cell Biosci 2024; 14:45. [PMID: 38582917 PMCID: PMC10998430 DOI: 10.1186/s13578-024-01224-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 03/24/2024] [Indexed: 04/08/2024] Open
Abstract
Canavan disease is an autosomal recessive and lethal neurological disorder, characterized by the spongy degeneration of the white matter in the brain. The disease is caused by a deficiency of the cytosolic aspartoacylase (ASPA) enzyme, which catalyzes the hydrolysis of N-acetyl-aspartate (NAA), an abundant brain metabolite, into aspartate and acetate. On the physiological level, the mechanism of pathogenicity remains somewhat obscure, with multiple, not mutually exclusive, suggested hypotheses. At the molecular level, recent studies have shown that most disease linked ASPA gene variants lead to a structural destabilization and subsequent proteasomal degradation of the ASPA protein variants, and accordingly Canavan disease should in general be considered a protein misfolding disorder. Here, we comprehensively summarize the molecular and cell biology of ASPA, with a particular focus on disease-linked gene variants and the pathophysiology of Canavan disease. We highlight the importance of high-throughput technologies and computational prediction tools for making genotype-phenotype predictions as we await the results of ongoing trials with gene therapy for Canavan disease.
Collapse
Affiliation(s)
- Martin Grønbæk-Thygesen
- The Linderstrøm-Lang Centre for Protein Science, Department of Biology, University of Copenhagen, Ole Maaløes Vej 5, 2200N, Copenhagen, Denmark.
| | - Rasmus Hartmann-Petersen
- The Linderstrøm-Lang Centre for Protein Science, Department of Biology, University of Copenhagen, Ole Maaløes Vej 5, 2200N, Copenhagen, Denmark.
| |
Collapse
|
4
|
Papaioannou VE, Behringer RR. The "No Phenotype" Challenge in Analyzing Mutant Mice. Cold Spring Harb Protoc 2024; 2024:107977. [PMID: 37932097 DOI: 10.1101/pdb.over107977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2023]
Abstract
If homozygous mutant mice survive to adulthood, are fertile, and have no visible phenotypes attributable to mutation of the relevant gene, there are a number of possible reasons why an effect of the mutation is not evident. Technical errors that might have occurred during gene targeting or genotyping must first be eliminated. Variable penetrance of the mutation should be considered as well as the possibility of age-related or late-onset phenotypes, such as tumor formation or other pathologies. The gene expression pattern and nature of the protein product of the gene could provide clues. A number of simple tests can be applied to uncover cryptic phenotypes that are not easily seen on casual inspection (e.g., tests of the senses and of balance and coordination). Genetic and environmental challenges can be applied to overtly normal mutant mice to reveal deviations from normal.
Collapse
Affiliation(s)
- Virginia E Papaioannou
- Department of Genetics and Development, Columbia University Medical Center, New York, New York 10032, USA
| | - Richard R Behringer
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| |
Collapse
|
5
|
Yu L, Lemay P, Ludlow A, Guyot MC, Jones M, Mohamed FF, Saroya GA, Panaretos C, Schneider E, Wang Y, Myers G, Khoriaty R, Li Q, Franceschi R, Engel JD, Kaartinen V, Rothstein TL, Justice MJ, Kibar Z, Singh SA. A new murine Rpl5 (uL18) mutation provides a unique model of variably penetrant Diamond-Blackfan anemia. Blood Adv 2021; 5:4167-4178. [PMID: 34464976 PMCID: PMC8945612 DOI: 10.1182/bloodadvances.2021004658] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 05/04/2021] [Indexed: 01/28/2023] Open
Abstract
Ribosome dysfunction is implicated in multiple abnormal developmental and disease states in humans. Heterozygous germline mutations in genes encoding ribosomal proteins are found in most individuals with Diamond-Blackfan anemia (DBA), whereas somatic mutations have been implicated in a variety of cancers and other disorders. Ribosomal protein-deficient animal models show variable phenotypes and penetrance, similar to human patients with DBA. In this study, we characterized a novel ENU mouse mutant (Skax23m1Jus) with growth and skeletal defects, cardiac malformations, and increased mortality. After genetic mapping and whole-exome sequencing, we identified an intronic Rpl5 mutation, which segregated with all affected mice. This mutation was associated with decreased ribosome generation, consistent with Rpl5 haploinsufficiency. Rpl5Skax23-Jus/+ animals had a profound delay in erythroid maturation and increased mortality at embryonic day (E) 12.5, which improved by E14.5. Surviving mutant animals had macrocytic anemia at birth, as well as evidence of ventricular septal defect (VSD). Surviving adult and aged mice exhibited no hematopoietic defect or VSD. We propose that this novel Rpl5Skax23-Jus/+ mutant mouse will be useful in studying the factors influencing the variable penetrance that is observed in DBA.
Collapse
Affiliation(s)
- Lei Yu
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI
| | - Philippe Lemay
- Department of Neurosciences, CHU Sainte Justine Research Center, University of Montréal, Montreal, QC, Canada
| | - Alexander Ludlow
- Center for Immunobiology and Department of Investigative Medicine,Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI
| | - Marie-Claude Guyot
- Department of Neurosciences, CHU Sainte Justine Research Center, University of Montréal, Montreal, QC, Canada
| | - Morgan Jones
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI
| | - Fatma F. Mohamed
- Department of Biologic and Materials Sciences, University of Michigan School of Dentistry, Ann Arbor, MI
| | - Ghazi-Abdullah Saroya
- Department of Biologic and Materials Sciences, University of Michigan School of Dentistry, Ann Arbor, MI
| | - Christopher Panaretos
- Department of Biologic and Materials Sciences, University of Michigan School of Dentistry, Ann Arbor, MI
| | - Emily Schneider
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI
| | - Yu Wang
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI
| | - Greggory Myers
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI
| | - Rami Khoriaty
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI
| | - Qing Li
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI
| | - Renny Franceschi
- Department of Biologic and Materials Sciences, University of Michigan School of Dentistry, Ann Arbor, MI
| | - James Douglas Engel
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI
| | - Vesa Kaartinen
- Department of Biologic and Materials Sciences, University of Michigan School of Dentistry, Ann Arbor, MI
| | - Thomas L. Rothstein
- Center for Immunobiology and Department of Investigative Medicine,Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI
| | - Monica J. Justice
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada; and
| | - Zoha Kibar
- Department of Neurosciences, CHU Sainte Justine Research Center, University of Montréal, Montreal, QC, Canada
| | - Sharon A. Singh
- Center for Immunobiology and Department of Investigative Medicine,Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI
| |
Collapse
|
6
|
Nešuta O, Thomas AG, Alt J, Hin N, Neužilová A, Long S, Tsukamoto T, Rojas C, Wei H, Slusher BS. High Throughput Screening Cascade To Identify Human Aspartate N-Acetyltransferase (ANAT) Inhibitors for Canavan Disease. ACS Chem Neurosci 2021; 12:3445-3455. [PMID: 34477360 DOI: 10.1021/acschemneuro.1c00455] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Canavan disease (CD) is a progressive, fatal neurological disorder that begins in infancy resulting from a mutation in aspartoacyclase (ASPA), an enzyme that catalyzes the deacetylation of N-acetyl aspartate (NAA) into acetate and aspartate. Increased NAA levels in the brains of affected children are one of the hallmarks of CD. Interestingly, genetic deletion of N-acetyltransferase-8-like (NAT8L), which encodes aspartate N-aceyltransferase (ANAT), an enzyme responsible for the synthesis of NAA from l-aspartate and acetyl-CoA, leads to normalization of NAA levels and improvement of symptoms in several genetically engineered mouse models of CD. Therefore, pharmacological inhibition of ANAT presents a promising therapeutic strategy for treating CD. Currently, however, there are no clinically viable ANAT inhibitors. Herein we describe the development of fluorescence-based high throughput screening (HTS) and radioactive-based orthogonal assays using recombinant human ANAT expressed in E. coli. In the fluorescence-based assay, ANAT activity was linear with respect to time of incubation up to 30 min and protein concentration up to 97.5 ng/μL with Km values for l-aspartate and acetyl-CoA of 237 μM and 11 μM, respectively. Using this optimized assay, we conducted a pilot screening of a 10 000-compound library. Hits from the fluorescence-based assay were subjected to an orthogonal radioactive-based assay using L-[U-14C] aspartate as a substrate. Two compounds were confirmed to have dose-dependent inhibition in both assays. Inhibitory kinetics studies of the most potent compound revealed an uncompetitive inhibitory mechanism with respect to l-aspartate and a noncompetitive inhibitory mechanism against acetyl-CoA. The screening cascade developed herein will enable large-scale compound library screening to identify novel ANAT inhibitors as leads for further medicinal chemistry optimization.
Collapse
Affiliation(s)
- Ondřej Nešuta
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, 855 N. Wolfe Street, Baltimore, Maryland 21205, United States
| | - Ajit G. Thomas
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, 855 N. Wolfe Street, Baltimore, Maryland 21205, United States
| | - Jesse Alt
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, 855 N. Wolfe Street, Baltimore, Maryland 21205, United States
| | - Niyada Hin
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, 855 N. Wolfe Street, Baltimore, Maryland 21205, United States
| | - Anna Neužilová
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, 855 N. Wolfe Street, Baltimore, Maryland 21205, United States
| | - Shunyou Long
- ChemBioCORE, High Throughput Screening Facility, Johns Hopkins University, 733 N. Broadway, Baltimore, Maryland 21205, United States
| | - Takashi Tsukamoto
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, 855 N. Wolfe Street, Baltimore, Maryland 21205, United States
- Department of Neurology, Johns Hopkins University, Baltimore, Maryland 21205, United States
| | - Camilo Rojas
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, 855 N. Wolfe Street, Baltimore, Maryland 21205, United States
| | - Huijun Wei
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, 855 N. Wolfe Street, Baltimore, Maryland 21205, United States
- Department of Neurology, Johns Hopkins University, Baltimore, Maryland 21205, United States
| | - Barbara S. Slusher
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, 855 N. Wolfe Street, Baltimore, Maryland 21205, United States
- Department of Neurology, Johns Hopkins University, Baltimore, Maryland 21205, United States
| |
Collapse
|
7
|
Ju C, Zhang M, Guan M, Li S, Zhang Y, Zhao J, Gao X. Fast and efficient generation of a full-length balancer chromosome by a single Cre/loxP recombination event. Mamm Genome 2021; 33:169-180. [PMID: 34386878 DOI: 10.1007/s00335-021-09897-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 07/14/2021] [Indexed: 11/28/2022]
Abstract
Balancer chromosomes, primarily discovered and used in Drosophila melanogaster, are valuable tools to maintain lethal mutations in a particular genomic segment. Full-length balancer chromosomes would be particularly useful because of the capacity to maintain whole genomic traits. However, murine full-length balancer chromosomes generated via a single Cre/loxP recombination are still not demonstrated. In this study, we developed a novel mouse strain with full-length inverted chromosome 17 (Ch17Inv balancer) via a single Cre/loxP recombination event in mES cells. The Ch17Inv balancer mice are viable and phenotypically normal. When bred with other strains, the haplotype of chromosome 17 can be stably maintained as determined by the high throughput SNPs assay. Interestingly, we found that the recombination events were efficiently reduced within the inverted region but not eliminated. The method established in this study can be applied to generate other full-length balancer chromosomes. Moreover, the Ch17Inv balancer strain would be a valuable resource to maintain the entire chromosome 17 from different donor strains.
Collapse
Affiliation(s)
- Cunxiang Ju
- GemPharmatech Co., Ltd., Xuefu Rd. 12#, Jiangbei New Area, Nanjing, China.
| | - Mingkun Zhang
- GemPharmatech Co., Ltd., Xuefu Rd. 12#, Jiangbei New Area, Nanjing, China
| | - Min Guan
- GemPharmatech Co., Ltd., Xuefu Rd. 12#, Jiangbei New Area, Nanjing, China
| | - Song Li
- GemPharmatech Co., Ltd., Xuefu Rd. 12#, Jiangbei New Area, Nanjing, China
| | - Yuxi Zhang
- GemPharmatech Co., Ltd., Xuefu Rd. 12#, Jiangbei New Area, Nanjing, China
| | - Jing Zhao
- GemPharmatech Co., Ltd., Xuefu Rd. 12#, Jiangbei New Area, Nanjing, China
| | - Xiang Gao
- GemPharmatech Co., Ltd., Xuefu Rd. 12#, Jiangbei New Area, Nanjing, China.
| |
Collapse
|
8
|
Iwata S, Sasaki T, Nagahara M, Iwamoto T. An efficient i-GONAD method for creating and maintaining lethal mutant mice using an inversion balancer identified from the C3H/HeJJcl strain. G3 (BETHESDA, MD.) 2021; 11:6291665. [PMID: 34849815 PMCID: PMC8496231 DOI: 10.1093/g3journal/jkab194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Accepted: 05/27/2021] [Indexed: 06/13/2023]
Abstract
As the efficiency of the clustered regularly interspaced short palindromic repeats/Cas system is extremely high, creation and maintenance of homozygous lethal mutants are often difficult. Here, we present an efficient in vivo electroporation method called improved genome editing via oviductal nucleic acid delivery (i-GONAD), wherein one of two alleles in the lethal gene was selectively edited in the presence of a non-targeted B6.C3H-In(6)1J inversion identified from the C3H/HeJJcl strain. This method did not require isolation, culture, transfer, or other in vitro handling of mouse embryos. The edited lethal genes were stably maintained in heterozygotes, as recombination is strongly suppressed within this inversion interval. Using this strategy, we successfully generated the first Tprkb null knockout strain with an embryonic lethal mutation and showed that B6.C3H-In(6)1J can efficiently suppress recombination. As B6.C3H-In(6)1J was tagged with a gene encoding the visible coat color marker, Mitf, the Tprkb mutation could be visually recognized. We listed the stock balancer strains currently available as public bioresources to create these lethal gene knockouts. This method will allow for more efficient experiments for further analysis of lethal mutants.
Collapse
Affiliation(s)
- Satoru Iwata
- Center for Education in Laboratory Animal Research, Chubu University, Kasugai, Aichi 487-8501, Japan
- Department of Biomedical Sciences, College of Life and Health Sciences, Chubu University, Kasugai, Aichi 487-8501, Japan
- College of Bioscience and Biotechnology, Chubu University, Kasugai, Aichi 487-8501, Japan
| | - Takahisa Sasaki
- Department of Biomedical Sciences, College of Life and Health Sciences, Chubu University, Kasugai, Aichi 487-8501, Japan
| | - Miki Nagahara
- Center for Education in Laboratory Animal Research, Chubu University, Kasugai, Aichi 487-8501, Japan
| | - Takashi Iwamoto
- Center for Education in Laboratory Animal Research, Chubu University, Kasugai, Aichi 487-8501, Japan
- Department of Biomedical Sciences, College of Life and Health Sciences, Chubu University, Kasugai, Aichi 487-8501, Japan
| |
Collapse
|
9
|
Lotun A, Gessler DJ, Gao G. Canavan Disease as a Model for Gene Therapy-Mediated Myelin Repair. Front Cell Neurosci 2021; 15:661928. [PMID: 33967698 PMCID: PMC8102781 DOI: 10.3389/fncel.2021.661928] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 03/23/2021] [Indexed: 11/13/2022] Open
Abstract
In recent years, the scientific and therapeutic fields for rare, genetic central nervous system (CNS) diseases such as leukodystrophies, or white matter disorders, have expanded significantly in part due to technological advancements in cellular and clinical screenings as well as remedial therapies using novel techniques such as gene therapy. However, treatments aimed at normalizing the pathological changes associated with leukodystrophies have especially been complicated due to the innate and variable effects of glial abnormalities, which can cause large-scale functional deficits in developmental myelination and thus lead to downstream neuronal impairment. Emerging research in the past two decades have depicted glial cells, particularly oligodendrocytes and astrocytes, as key, regulatory modulators in constructing and maintaining myelin function and neuronal viability. Given the significance of myelin formation in the developing brain, myelin repair in a time-dependent fashion is critical in restoring homeostatic functionality to the CNS of patients diagnosed with white matter disorders. Using Canavan Disease (CD) as a leukodystrophy model, here we review the hypothetical roles of N-acetylaspartate (NAA), one of the brain's most abundant amino acid derivatives, in Canavan disease's CNS myelinating pathology, as well as discuss the possible functions astrocytes serve in both CD and other leukodystrophies' time-sensitive disease correction. Through this analysis, we also highlight the potential remyelinating benefits of gene therapy for other leukodystrophies in which alternative CNS cell targeting for white matter disorders may be an applicable path for reparative treatment.
Collapse
Affiliation(s)
- Anoushka Lotun
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, United States.,Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, United States
| | - Dominic J Gessler
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, United States.,Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, United States.,Department of Neurosurgery, University of Minnesota, Minneapolis, MN, United States
| | - Guangping Gao
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, United States.,Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, United States.,Li Weibo Institute for Rare Diseases Research, University of Massachusetts Medical School, Worcester, MA, United States
| |
Collapse
|
10
|
Harding P, Cunha DL, Moosajee M. Animal and cellular models of microphthalmia. THERAPEUTIC ADVANCES IN RARE DISEASE 2021; 2:2633004021997447. [PMID: 37181112 PMCID: PMC10032472 DOI: 10.1177/2633004021997447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 02/02/2021] [Indexed: 05/16/2023]
Abstract
Microphthalmia is a rare developmental eye disorder affecting 1 in 7000 births. It is defined as a small (axial length ⩾2 standard deviations below the age-adjusted mean) underdeveloped eye, caused by disruption of ocular development through genetic or environmental factors in the first trimester of pregnancy. Clinical phenotypic heterogeneity exists amongst patients with varying levels of severity, and associated ocular and systemic features. Up to 11% of blind children are reported to have microphthalmia, yet currently no treatments are available. By identifying the aetiology of microphthalmia and understanding how the mechanisms of eye development are disrupted, we can gain a better understanding of the pathogenesis. Animal models, mainly mouse, zebrafish and Xenopus, have provided extensive information on the genetic regulation of oculogenesis, and how perturbation of these pathways leads to microphthalmia. However, differences exist between species, hence cellular models, such as patient-derived induced pluripotent stem cell (iPSC) optic vesicles, are now being used to provide greater insights into the human disease process. Progress in 3D cellular modelling techniques has enhanced the ability of researchers to study interactions of different cell types during eye development. Through improved molecular knowledge of microphthalmia, preventative or postnatal therapies may be developed, together with establishing genotype-phenotype correlations in order to provide patients with the appropriate prognosis, multidisciplinary care and informed genetic counselling. This review summarises some key discoveries from animal and cellular models of microphthalmia and discusses how innovative new models can be used to further our understanding in the future. Plain language summary Animal and Cellular Models of the Eye Disorder, Microphthalmia (Small Eye) Microphthalmia, meaning a small, underdeveloped eye, is a rare disorder that children are born with. Genetic changes or variations in the environment during the first 3 months of pregnancy can disrupt early development of the eye, resulting in microphthalmia. Up to 11% of blind children have microphthalmia, yet currently no treatments are available. By understanding the genes necessary for eye development, we can determine how disruption by genetic changes or environmental factors can cause this condition. This helps us understand why microphthalmia occurs, and ensure patients are provided with the appropriate clinical care and genetic counselling advice. Additionally, by understanding the causes of microphthalmia, researchers can develop treatments to prevent or reduce the severity of this condition. Animal models, particularly mice, zebrafish and frogs, which can also develop small eyes due to the same genetic/environmental changes, have helped us understand the genes which are important for eye development and can cause birth eye defects when disrupted. Studying a patient's own cells grown in the laboratory can further help researchers understand how changes in genes affect their function. Both animal and cellular models can be used to develop and test new drugs, which could provide treatment options for patients living with microphthalmia. This review summarises the key discoveries from animal and cellular models of microphthalmia and discusses how innovative new models can be used to further our understanding in the future.
Collapse
Affiliation(s)
| | | | - Mariya Moosajee
- UCL Institute of Ophthalmology, 11-43 Bath
Street, London, EC1V 9EL, UK
- Moorfields Eye Hospital NHS Foundation Trust,
London, UK
- Great Ormond Street Hospital for Children NHS
Foundation Trust, London, UK
- The Francis Crick Institute, London, UK
| |
Collapse
|
11
|
Abstract
The mouse is one of the most widely used model organisms for genetic study. The tools available to alter the mouse genome have developed over the preceding decades from forward screens to gene targeting in stem cells to the recent influx of CRISPR approaches. In this review, we first consider the history of mice in genetic study, the development of classic approaches to genome modification, and how such approaches have been used and improved in recent years. We then turn to the recent surge of nuclease-mediated techniques and how they are changing the field of mouse genetics. Finally, we survey common classes of alleles used in mice and discuss how they might be engineered using different methods.
Collapse
Affiliation(s)
- James F Clark
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mt. Sinai, New York, New York 10029, USA
| | - Colin J Dinsmore
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mt. Sinai, New York, New York 10029, USA
| | - Philippe Soriano
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mt. Sinai, New York, New York 10029, USA
| |
Collapse
|
12
|
A missense mutation of ErbB2 produces a novel mouse model of stillbirth associated with a cardiac abnormality but lacking abnormalities of placental structure. PLoS One 2020; 15:e0233007. [PMID: 32492036 PMCID: PMC7269201 DOI: 10.1371/journal.pone.0233007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 04/28/2020] [Indexed: 12/02/2022] Open
Abstract
Background In humans, stillbirth describes the death of a fetus before birth after 28 weeks gestation, and accounts for approximately 2.6 million deaths worldwide annually. In high-income countries, up to half of stillbirths have an unknown cause and are described as “unexplained stillbirths”; this lack of understanding impairs efforts to prevent stillbirth. There are also few animal models of stillbirth, but those that have been described usually have significant placental abnormalities. This study describes a novel mutant murine model of fetal death with atrial conduction block due to an ErbB2 missense mutation which is not associated with abnormal placental morphology. Methods Phenotypic characterisation and histological analysis of the mutant mouse model was conducted. The mRNA distribution of the early cardiomyocyte marker Nkx2-5 was assessed via in situ hybridisation. Cardiac structure was quantified and cellular morphology evaluated by electron microscopy. Immunostaining was employed to quantify placental structure and cell characteristics on matched heterozygous and homozygous mutant placental samples. Results There were no structural abnormalities observed in hearts of mutant embryos. Comparable Nkx2-5 expression was observed in hearts of mutants and controls, suggesting normal cardiac specification. Additionally, there was no significant difference in the weight, placenta dimensions, giant cell characteristics, labyrinth tissue composition, levels of apoptosis, proliferation or vascularisation between placentas of homozygous mutant mice and controls. Conclusion Embryonic lethality in the ErbB2 homozygous mutant mouse cannot be attributed to placental pathology. As such, we conclude the ErbB2M802R mutant is a model of stillbirth with a non-placental cause of death. The mechanism of the atrial block resulting from ErbB2 mutation and its role in embryonic death is still unclear. Studying this mutant mouse model could identify candidate genes involved in stillbirth associated with structural or functional cardiac defects.
Collapse
|
13
|
Enikanolaiye A, Ruston J, Zeng R, Taylor C, Schrock M, Buchovecky CM, Shendure J, Acar E, Justice MJ. Suppressor mutations in Mecp2-null mice implicate the DNA damage response in Rett syndrome pathology. Genome Res 2020; 30:540-552. [PMID: 32317254 PMCID: PMC7197480 DOI: 10.1101/gr.258400.119] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 03/20/2020] [Indexed: 12/31/2022]
Abstract
Mutations in X-linked methyl-CpG-binding protein 2 (MECP2) cause Rett syndrome (RTT). To identify functional pathways that could inform therapeutic entry points, we carried out a genetic screen for secondary mutations that improved phenotypes in Mecp2/Y mice after mutagenesis with N-ethyl-N-nitrosourea (ENU). Here, we report the isolation of 106 founder animals that show suppression of Mecp2-null traits from screening 3177 Mecp2/Y genomes. Whole-exome sequencing, genetic crosses, and association analysis identified 22 candidate genes. Additional lesions in these candidate genes or pathway components associate variant alleles with phenotypic improvement in 30 lines. A network analysis shows that 63% of the genes cluster into the functional categories of transcriptional repression, chromatin modification, or DNA repair, delineating a pathway relationship with MECP2. Many mutations lie in genes that modulate synaptic signaling or lipid homeostasis. Mutations in genes that function in the DNA damage response (DDR) also improve phenotypes in Mecp2/Y mice. Association analysis was successful in resolving combinatorial effects of multiple loci. One line, which carries a suppressor mutation in a gene required for cholesterol synthesis, Sqle, carries a second mutation in retinoblastoma binding protein 8, endonuclease (Rbbp8, also known as CtIP), which regulates a DDR choice in double-stranded break (DSB) repair. Cells from Mecp2/Y mice have increased DSBs, so this finding suggests that the balance between homology-directed repair and nonhomologous end joining is important for neuronal cells. In this and other lines, two suppressor mutations confer greater improvement than one alone, suggesting that combination therapies could be effective in RTT.
Collapse
Affiliation(s)
- Adebola Enikanolaiye
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario, M5G 0A4, Canada
| | - Julie Ruston
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario, M5G 0A4, Canada
| | - Rong Zeng
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario, M5G 0A4, Canada
| | - Christine Taylor
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario, M5G 0A4, Canada
| | - Marijke Schrock
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Christie M Buchovecky
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Jay Shendure
- Department of Genome Sciences, University of Washington, Seattle, Washington 98195, USA
- Brotman Baty Institute for Precision Medicine, Seattle, Washington 98195, USA
- Allen Discovery Center for Cell Lineage Tracing, Seattle, Washington 98195, USA
- Howard Hughes Medical Institute, Seattle, Washington 98195, USA
| | - Elif Acar
- The Centre for Phenogenomics, Toronto, Ontario, M5T 3H7, Canada
- Department of Statistics, University of Manitoba, Winnipeg, Manitoba, R3T 2N2, Canada
| | - Monica J Justice
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario, M5G 0A4, Canada
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, USA
- The Centre for Phenogenomics, Toronto, Ontario, M5T 3H7, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, M5S 1A8, Canada
| |
Collapse
|
14
|
García-García MJ. A History of Mouse Genetics: From Fancy Mice to Mutations in Every Gene. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1236:1-38. [PMID: 32304067 DOI: 10.1007/978-981-15-2389-2_1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The laboratory mouse has become the model organism of choice in numerous areas of biological and biomedical research, including the study of congenital birth defects. The appeal of mice for these experimental studies stems from the similarities between the physiology, anatomy, and reproduction of these small mammals with our own, but it is also based on a number of practical reasons: mice are easy to maintain in a laboratory environment, are incredibly prolific, and have a relatively short reproductive cycle. Another compelling reason for choosing mice as research subjects is the number of tools and resources that have been developed after more than a century of working with these small rodents in laboratory environments. As will become obvious from the reading of the different chapters in this book, research in mice has already helped uncover many of the genes and processes responsible for congenital birth malformations and human diseases. In this chapter, we will provide an overview of the methods, scientific advances, and serendipitous circumstances that have made these discoveries possible, with a special emphasis on how the use of genetics has propelled scientific progress in mouse research and paved the way for future discoveries.
Collapse
|
15
|
Yin W, Kim HT, Wang S, Gunawan F, Li R, Buettner C, Grohmann B, Sengle G, Sinner D, Offermanns S, Stainier DYR. Fibrillin-2 is a key mediator of smooth muscle extracellular matrix homeostasis during mouse tracheal tubulogenesis. Eur Respir J 2019; 53:13993003.00840-2018. [PMID: 30578393 DOI: 10.1183/13993003.00840-2018] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 11/28/2018] [Indexed: 12/16/2022]
Abstract
Epithelial tubes, comprised of polarised epithelial cells around a lumen, are crucial for organ function. However, the molecular mechanisms underlying tube formation remain largely unknown. Here, we report on the function of fibrillin (FBN)2, an extracellular matrix (ECM) glycoprotein, as a critical regulator of tracheal tube formation.We performed a large-scale forward genetic screen in mouse to identify regulators of respiratory organ development and disease. We identified Fbn2 mutants which exhibit shorter and narrowed tracheas as well as defects in tracheal smooth muscle cell alignment and polarity.We found that FBN2 is essential for elastic fibre formation and Fibronectin accumulation around tracheal smooth muscle cells. These processes appear to be regulated at least in part through inhibition of p38-mediated upregulation of matrix metalloproteinases (MMPs), as pharmacological decrease of p38 phosphorylation or MMP activity partially attenuated the Fbn2 mutant tracheal phenotypes. Analysis of human tracheal tissues indicates that a decrease in ECM proteins, including FBN2 and Fibronectin, is associated with tracheomalacia.Our findings provide novel insights into the role of ECM homeostasis in mesenchymal cell polarisation during tracheal tubulogenesis.
Collapse
Affiliation(s)
- Wenguang Yin
- Max Planck Institute for Heart and Lung Research, Dept of Developmental Genetics, Bad Nauheim, Germany.,W. Yin and D.Y.R. Stainier are joint senior authors
| | - Hyun-Taek Kim
- Max Planck Institute for Heart and Lung Research, Dept of Developmental Genetics, Bad Nauheim, Germany
| | - ShengPeng Wang
- Max Planck Institute for Heart and Lung Research, Dept of Pharmacology, Bad Nauheim, Germany.,Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China
| | - Felix Gunawan
- Max Planck Institute for Heart and Lung Research, Dept of Developmental Genetics, Bad Nauheim, Germany
| | - Rui Li
- Max Planck Institute for Heart and Lung Research, Dept of Pharmacology, Bad Nauheim, Germany
| | - Carmen Buettner
- Max Planck Institute for Heart and Lung Research, Dept of Developmental Genetics, Bad Nauheim, Germany
| | - Beate Grohmann
- Max Planck Institute for Heart and Lung Research, Dept of Developmental Genetics, Bad Nauheim, Germany
| | - Gerhard Sengle
- Center for Biochemistry, Medical Faculty, University of Cologne, Cologne, Germany.,Center for Molecular Medicine Cologne (CMMC), Cologne, Germany
| | - Debora Sinner
- Division of Neonatology and Pulmonary Biology, CCHMC, University of Cincinnati, College of Medicine Cincinnati, OH, USA
| | - Stefan Offermanns
- Max Planck Institute for Heart and Lung Research, Dept of Pharmacology, Bad Nauheim, Germany.,Center for Molecular Medicine, Goethe University, Frankfurt, Germany
| | - Didier Y R Stainier
- Max Planck Institute for Heart and Lung Research, Dept of Developmental Genetics, Bad Nauheim, Germany.,W. Yin and D.Y.R. Stainier are joint senior authors
| |
Collapse
|
16
|
Tian D, Wenlock S, Kabir M, Tzotzos G, Doig AJ, Hentges KE. Identifying mouse developmental essential genes using machine learning. Dis Model Mech 2018; 11:11/12/dmm034546. [PMID: 30563825 PMCID: PMC6307915 DOI: 10.1242/dmm.034546] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 10/19/2018] [Indexed: 12/20/2022] Open
Abstract
The genes that are required for organismal survival are annotated as ‘essential genes’. Identifying all the essential genes of an animal species can reveal critical functions that are needed during the development of the organism. To inform studies on mouse development, we developed a supervised machine learning classifier based on phenotype data from mouse knockout experiments. We used this classifier to predict the essentiality of mouse genes lacking experimental data. Validation of our predictions against a blind test set of recent mouse knockout experimental data indicated a high level of accuracy (>80%). We also validated our predictions for other mouse mutagenesis methodologies, demonstrating that the predictions are accurate for lethal phenotypes isolated in random chemical mutagenesis screens and embryonic stem cell screens. The biological functions that are enriched in essential and non-essential genes have been identified, showing that essential genes tend to encode intracellular proteins that interact with nucleic acids. The genome distribution of predicted essential and non-essential genes was analysed, demonstrating that the density of essential genes varies throughout the genome. A comparison with human essential and non-essential genes was performed, revealing conservation between human and mouse gene essentiality status. Our genome-wide predictions of mouse essential genes will be of value for the planning of mouse knockout experiments and phenotyping assays, for understanding the functional processes required during mouse development, and for the prioritisation of disease candidate genes identified in human genome and exome sequence datasets. Summary: Here, we used computer-based machine learning methodology to predict which genes in the mouse genome are essential for development, and present a database of mouse essential and non-essential genes.
Collapse
Affiliation(s)
- David Tian
- Division of Evolution and Genomic Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Stephanie Wenlock
- Division of Evolution and Genomic Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Mitra Kabir
- Division of Evolution and Genomic Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - George Tzotzos
- Department of Agriculture, Food and Environmental Sciences, Marche Polytechnic University, Ancona 60121, Italy
| | - Andrew J Doig
- Manchester Institute of Biotechnology, The University of Manchester, 131 Princess Street, Manchester M1 7DN, UK .,Division of Neuroscience and Experimental Psychology, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK
| | - Kathryn E Hentges
- Division of Evolution and Genomic Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Oxford Road, Manchester M13 9PT, UK
| |
Collapse
|
17
|
Uli N, Michelen-Gomez E, Ramos EI, Druley TE. Age-specific changes in genome-wide methylation enrich for Foxa2 and estrogen receptor alpha binding sites. PLoS One 2018; 13:e0203147. [PMID: 30256791 PMCID: PMC6157835 DOI: 10.1371/journal.pone.0203147] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 08/15/2018] [Indexed: 12/26/2022] Open
Abstract
The role of DNA methylation patterns in complex phenotypes remains unclear. To explore this question, we adapted our methods for rare variant analysis to characterize genome-wide murine DNA hybridization array to investigate methylation at CpG islands, shores, and regulatory elements. We have applied this platform to compare age and tissue- specific methylation differences in the brain and spleen of young and aged mice. As expected from prior studies, there are clear global differences in organ-specific, but not age-specific, methylation due mostly to changes at repetitive elements. Surprisingly, out of 200,000 loci there were only 946 differentially methylated cytosines (DMCs) between young and old samples (529 hypermethylated, 417 hypomethylated in aged mice) compared to thousands of tissue-specific DMCs. Hypermethylated loci were clustered around the promoter region of Sfi1, exon 2 of Slc11a2, Drg1, Esr1 and Foxa2 transcription factor binding sites. In particular, there were 75 hypermethylated Foxa2 binding sites across a 2.7 Mb region of chromosome 11. Hypomethylated loci were clustered around Mid1, Isoc2b and genome-wide loci with binding sites for Foxa2 and Esr1, which are known to play important roles in development and aging. These data suggest discreet tissue-independent methylation changes associated with aging processes such as cell division (Sfi1, Mid1), energy production (Drg1, Isoc2b) and cell death (Foxa2, Esr1).
Collapse
Affiliation(s)
- Nishanth Uli
- Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Eduardo Michelen-Gomez
- Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Enrique I. Ramos
- Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Todd E. Druley
- Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- * E-mail:
| |
Collapse
|
18
|
Yin W, Kim HT, Wang S, Gunawan F, Wang L, Kishimoto K, Zhong H, Roman D, Preussner J, Guenther S, Graef V, Buettner C, Grohmann B, Looso M, Morimoto M, Mardon G, Offermanns S, Stainier DYR. The potassium channel KCNJ13 is essential for smooth muscle cytoskeletal organization during mouse tracheal tubulogenesis. Nat Commun 2018; 9:2815. [PMID: 30022023 PMCID: PMC6052067 DOI: 10.1038/s41467-018-05043-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 05/25/2018] [Indexed: 12/22/2022] Open
Abstract
Tubulogenesis is essential for the formation and function of internal organs. One such organ is the trachea, which allows gas exchange between the external environment and the lungs. However, the cellular and molecular mechanisms underlying tracheal tube development remain poorly understood. Here, we show that the potassium channel KCNJ13 is a critical modulator of tracheal tubulogenesis. We identify Kcnj13 in an ethylnitrosourea forward genetic screen for regulators of mouse respiratory organ development. Kcnj13 mutants exhibit a shorter trachea as well as defective smooth muscle (SM) cell alignment and polarity. KCNJ13 is essential to maintain ion homeostasis in tracheal SM cells, which is required for actin polymerization. This process appears to be mediated, at least in part, through activation of the actin regulator AKT, as pharmacological increase of AKT phosphorylation ameliorates the Kcnj13-mutant trachea phenotypes. These results provide insight into the role of ion homeostasis in cytoskeletal organization during tubulogenesis.
Collapse
Affiliation(s)
- Wenguang Yin
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, 61231, Germany.
| | - Hyun-Taek Kim
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, 61231, Germany
| | - ShengPeng Wang
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, 61231, Germany
- Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Felix Gunawan
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, 61231, Germany
| | - Lei Wang
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, 61231, Germany
| | - Keishi Kishimoto
- Laboratory for Lung Development, RIKEN Center for Developmental Biology, Kobe, 650-0047, Japan
| | - Hua Zhong
- Departments of Pathology and Immunology and Molecular and Human Genetics, Integrative Molecular and Biomedical Sciences Program, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Dany Roman
- Departments of Pathology and Immunology and Molecular and Human Genetics, Integrative Molecular and Biomedical Sciences Program, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Jens Preussner
- Max Planck Institute for Heart and Lung Research, ECCPS Bioinformatics and Deep Sequencing Platform, Bad Nauheim, 61231, Germany
| | - Stefan Guenther
- Max Planck Institute for Heart and Lung Research, ECCPS Bioinformatics and Deep Sequencing Platform, Bad Nauheim, 61231, Germany
| | - Viola Graef
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, 61231, Germany
| | - Carmen Buettner
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, 61231, Germany
| | - Beate Grohmann
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, 61231, Germany
| | - Mario Looso
- Max Planck Institute for Heart and Lung Research, ECCPS Bioinformatics and Deep Sequencing Platform, Bad Nauheim, 61231, Germany
| | - Mitsuru Morimoto
- Laboratory for Lung Development, RIKEN Center for Developmental Biology, Kobe, 650-0047, Japan
| | - Graeme Mardon
- Departments of Pathology and Immunology and Molecular and Human Genetics, Integrative Molecular and Biomedical Sciences Program, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Stefan Offermanns
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, 61231, Germany
- Center for Molecular Medicine, Goethe University, Frankfurt, 60590, Germany
| | - Didier Y R Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, 61231, Germany.
| |
Collapse
|
19
|
Ridge LA, Mitchell K, Al-Anbaki A, Shaikh Qureshi WM, Stephen LA, Tenin G, Lu Y, Lupu IE, Clowes C, Robertson A, Barnes E, Wright JA, Keavney B, Ehler E, Lovell SC, Kadler KE, Hentges KE. Non-muscle myosin IIB (Myh10) is required for epicardial function and coronary vessel formation during mammalian development. PLoS Genet 2017; 13:e1007068. [PMID: 29084269 PMCID: PMC5697871 DOI: 10.1371/journal.pgen.1007068] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Revised: 11/21/2017] [Accepted: 10/11/2017] [Indexed: 01/01/2023] Open
Abstract
The coronary vasculature is an essential vessel network providing the blood supply to the heart. Disruptions in coronary blood flow contribute to cardiac disease, a major cause of premature death worldwide. The generation of treatments for cardiovascular disease will be aided by a deeper understanding of the developmental processes that underpin coronary vessel formation. From an ENU mutagenesis screen, we have isolated a mouse mutant displaying embryonic hydrocephalus and cardiac defects (EHC). Positional cloning and candidate gene analysis revealed that the EHC phenotype results from a point mutation in a splice donor site of the Myh10 gene, which encodes NMHC IIB. Complementation testing confirmed that the Myh10 mutation causes the EHC phenotype. Characterisation of the EHC cardiac defects revealed abnormalities in myocardial development, consistent with observations from previously generated NMHC IIB null mouse lines. Analysis of the EHC mutant hearts also identified defects in the formation of the coronary vasculature. We attribute the coronary vessel abnormalities to defective epicardial cell function, as the EHC epicardium displays an abnormal cell morphology, reduced capacity to undergo epithelial-mesenchymal transition (EMT), and impaired migration of epicardial-derived cells (EPDCs) into the myocardium. Our studies on the EHC mutant demonstrate a requirement for NMHC IIB in epicardial function and coronary vessel formation, highlighting the importance of this protein in cardiac development and ultimately, embryonic survival.
Collapse
Affiliation(s)
- Liam A. Ridge
- Division of Evolution and Genome Sciences, School of Biological Sciences, Faculty of Biology, Medicine, and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| | - Karen Mitchell
- Division of Evolution and Genome Sciences, School of Biological Sciences, Faculty of Biology, Medicine, and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| | - Ali Al-Anbaki
- Division of Evolution and Genome Sciences, School of Biological Sciences, Faculty of Biology, Medicine, and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| | - Wasay Mohiuddin Shaikh Qureshi
- Division of Evolution and Genome Sciences, School of Biological Sciences, Faculty of Biology, Medicine, and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| | - Louise A. Stephen
- Division of Evolution and Genome Sciences, School of Biological Sciences, Faculty of Biology, Medicine, and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| | - Gennadiy Tenin
- Division of Evolution and Genome Sciences, School of Biological Sciences, Faculty of Biology, Medicine, and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| | - Yinhui Lu
- Wellcome Trust Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine, and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| | - Irina-Elena Lupu
- Division of Evolution and Genome Sciences, School of Biological Sciences, Faculty of Biology, Medicine, and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| | - Christopher Clowes
- Division of Evolution and Genome Sciences, School of Biological Sciences, Faculty of Biology, Medicine, and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| | - Abigail Robertson
- Division of Evolution and Genome Sciences, School of Biological Sciences, Faculty of Biology, Medicine, and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| | - Emma Barnes
- Syngenta Ltd, Jealott’s Hill International Research Centre, Bracknell, United Kingdom
| | - Jayne A. Wright
- Syngenta Ltd, Jealott’s Hill International Research Centre, Bracknell, United Kingdom
| | - Bernard Keavney
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine, and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
- Manchester Heart Centre, Central Manchester University Hospitals NHS Foundation Trust, Manchester, United Kingdom
| | - Elisabeth Ehler
- The Randall Division of Cell and Molecular Biophysics and the Cardiovascular Division, Kings College London, London, United Kingdom
| | - Simon C. Lovell
- Division of Evolution and Genome Sciences, School of Biological Sciences, Faculty of Biology, Medicine, and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| | - Karl E. Kadler
- Wellcome Trust Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine, and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| | - Kathryn E. Hentges
- Division of Evolution and Genome Sciences, School of Biological Sciences, Faculty of Biology, Medicine, and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
- * E-mail:
| |
Collapse
|
20
|
Nguyen AT, Prado MA, Schmidt PJ, Sendamarai AK, Wilson-Grady JT, Min M, Campagna DR, Tian G, Shi Y, Dederer V, Kawan M, Kuehnle N, Paulo JA, Yao Y, Weiss MJ, Justice MJ, Gygi SP, Fleming MD, Finley D. UBE2O remodels the proteome during terminal erythroid differentiation. Science 2017; 357:eaan0218. [PMID: 28774900 PMCID: PMC5812729 DOI: 10.1126/science.aan0218] [Citation(s) in RCA: 110] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 06/08/2017] [Indexed: 12/13/2022]
Abstract
During terminal differentiation, the global protein complement is remodeled, as epitomized by erythrocytes, whose cytosol is ~98% globin. The erythroid proteome undergoes a rapid transition at the reticulocyte stage; however, the mechanisms driving programmed elimination of preexisting cytosolic proteins are unclear. We found that a mutation in the murine Ube2o gene, which encodes a ubiquitin-conjugating enzyme induced during erythropoiesis, results in anemia. Proteomic analysis suggested that UBE2O is a broad-spectrum ubiquitinating enzyme that remodels the erythroid proteome. In particular, ribosome elimination, a hallmark of reticulocyte differentiation, was defective in Ube2o-/- mutants. UBE2O recognized ribosomal proteins and other substrates directly, targeting them to proteasomes for degradation. Thus, in reticulocytes, the induction of ubiquitinating factors may drive the transition from a complex to a simple proteome.
Collapse
Affiliation(s)
- Anthony T Nguyen
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Miguel A Prado
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Paul J Schmidt
- Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Anoop K Sendamarai
- Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | | | - Mingwei Min
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Dean R Campagna
- Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Geng Tian
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Yuan Shi
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Verena Dederer
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Mona Kawan
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Nathalie Kuehnle
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Joao A Paulo
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Yu Yao
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Mitchell J Weiss
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Monica J Justice
- Genetics and Genome Biology Program, Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, Toronto, Ontario M5G 0A4, Canada
| | - Steven P Gygi
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Mark D Fleming
- Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA.
| | - Daniel Finley
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
21
|
Hong CJ, Hamilton BA. Zfp423 Regulates Sonic Hedgehog Signaling via Primary Cilium Function. PLoS Genet 2016; 12:e1006357. [PMID: 27727273 PMCID: PMC5065120 DOI: 10.1371/journal.pgen.1006357] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2016] [Accepted: 09/09/2016] [Indexed: 11/17/2022] Open
Abstract
Zfp423 encodes a 30-zinc finger transcription factor that intersects several canonical signaling pathways. Zfp423 mutations result in ciliopathy-related phenotypes, including agenesis of the cerebellar vermis in mice and Joubert syndrome (JBTS19) and nephronophthisis (NPHP14) in humans. Unlike most ciliopathy genes, Zfp423 encodes a nuclear protein and its developmental expression is complex, leading to alternative proposals for cellular mechanisms. Here we show that Zfp423 is expressed by cerebellar granule cell precursors, that loss of Zfp423 in these precursors leads to cell-intrinsic reduction in proliferation, loss of response to Shh, and primary cilia abnormalities that include diminished frequency of both Smoothened and IFT88 localization. Loss of Zfp423 alters expression of several genes encoding key cilium components, including increased expression of Tulp3. Tulp3 is a direct binding target of Zfp423 and reducing the overexpression of Tulp3 in Zfp423-deficient cells suppresses Smoothened translocation defects. These results define Zfp423 deficiency as a bona fide ciliopathy, acting upstream of Shh signaling, and indicate a mechanism intrinsic to granule cell precursors for the resulting cerebellar hypoplasia.
Collapse
Affiliation(s)
- Chen-Jei Hong
- Department of Cellular & Molecular Medicine, University of California, San Diego School of Medicine, La Jolla, California, United States of America.,Department of Medicine, University of California, San Diego School of Medicine, La Jolla, California, United States of America.,Moores UCSD Cancer Center, University of California, San Diego School of Medicine, La Jolla, California, United States of America.,Institute for Genomic Medicine, University of California, San Diego School of Medicine, La Jolla, California, United States of America
| | - Bruce A Hamilton
- Department of Cellular & Molecular Medicine, University of California, San Diego School of Medicine, La Jolla, California, United States of America.,Department of Medicine, University of California, San Diego School of Medicine, La Jolla, California, United States of America.,Moores UCSD Cancer Center, University of California, San Diego School of Medicine, La Jolla, California, United States of America.,Institute for Genomic Medicine, University of California, San Diego School of Medicine, La Jolla, California, United States of America
| |
Collapse
|
22
|
Abstract
Metabolic disorders comprise a large group of heterogeneous diseases ranging from very prevalent diseases such as diabetes mellitus to rare genetic disorders like Canavan Disease. Whether either of these diseases is amendable by gene therapy depends to a large degree on the knowledge of their pathomechanism, availability of the therapeutic gene, vector selection, and availability of suitable animal models. In this book chapter, we review three metabolic disorders of the central nervous system (CNS; Canavan Disease, Niemann-Pick disease and Phenylketonuria) to give examples for primary and secondary metabolic disorders of the brain and the attempts that have been made to use adeno-associated virus (AAV) based gene therapy for treatment. Finally, we highlight commonalities and obstacles in the development of gene therapy for metabolic disorders of the CNS exemplified by those three diseases.
Collapse
Affiliation(s)
- Dominic J Gessler
- University of Massachusetts Medical School, 368 Plantation Street, AS6-2049, Worcester, MA, 01605, USA
| | - Guangping Gao
- University of Massachusetts Medical School, 368 Plantation Street, AS6-2049, Worcester, MA, 01605, USA.
| |
Collapse
|
23
|
Spontaneous malformations of the cerebellar vermis: Prevalence, inheritance, and relationship to lobule/fissure organization in the C57BL/6 lineage. Neuroscience 2015; 310:242-51. [PMID: 26383253 DOI: 10.1016/j.neuroscience.2015.09.025] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 09/09/2015] [Indexed: 11/22/2022]
Abstract
The complex neuronal circuitry of the cerebellum is embedded within its lamina, folia, and lobules, which together play an important role in sensory and motor function. Studies in mouse models have demonstrated that both cerebellar lamination and lobule/fissure development are under genetic control. The cerebellar vermis of C57BL/6 mice exhibits spontaneous malformations of neuronal migration of posterior lobules (VIII-IX; molecular layer heterotopia); however, the extent to which other inbred mice also exhibit these malformations is unknown. Using seven different inbred mouse strains and two first filial generation (F1) hybrids, we show that only the C57BL/6 strain exhibits heterotopia. Furthermore, we observed heterotopia in consomic and recombinant inbred strains. These data indicate that heterotopia formation is a weakly penetrant trait requiring homozygosity of one or more C57BL/6 alleles outside of chromosome 1 and the sex chromosomes. Additional morphological analyses showed no relationship between heterotopia formation and other features of lobule/fissure organization. These data are relevant toward understanding normal cerebellar development and disorders affecting cerebellar foliation and lamination.
Collapse
|
24
|
Ohno Y, Shimizu S, Tatara A, Imaoku T, Ishii T, Sasa M, Serikawa T, Kuramoto T. Hcn1 is a tremorgenic genetic component in a rat model of essential tremor. PLoS One 2015; 10:e0123529. [PMID: 25970616 PMCID: PMC4430019 DOI: 10.1371/journal.pone.0123529] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Accepted: 02/18/2015] [Indexed: 01/12/2023] Open
Abstract
Genetic factors are thought to play a major role in the etiology of essential tremor (ET); however, few genetic changes that induce ET have been identified to date. In the present study, to find genes responsible for the development of ET, we employed a rat model system consisting of a tremulous mutant strain, TRM/Kyo (TRM), and its substrain TRMR/Kyo (TRMR). The TRM rat is homozygous for the tremor (tm) mutation and shows spontaneous tremors resembling human ET. The TRMR rat also carries a homozygous tm mutation but shows no tremor, leading us to hypothesize that TRM rats carry one or more genes implicated in the development of ET in addition to the tm mutation. We used a positional cloning approach and found a missense mutation (c. 1061 C>T, p. A354V) in the hyperpolarization-activated cyclic nucleotide-gated 1 channel (Hcn1) gene. The A354V HCN1 failed to conduct hyperpolarization-activated currents in vitro, implicating it as a loss-of-function mutation. Blocking HCN1 channels with ZD7288 in vivo evoked kinetic tremors in nontremulous TRMR rats. We also found neuronal activation of the inferior olive (IO) in both ZD7288-treated TRMR and non-treated TRM rats and a reduced incidence of tremor in the IO-lesioned TRM rats, suggesting a critical role of the IO in tremorgenesis. A rat strain carrying the A354V mutation alone on a genetic background identical to that of the TRM rats showed no tremor. Together, these data indicate that body tremors emerge when the two mutant loci, tm and Hcn1A354V, are combined in a rat model of ET. In this model, HCN1 channels play an important role in the tremorgenesis of ET. We propose that oligogenic, most probably digenic, inheritance is responsible for the genetic heterogeneity of ET.
Collapse
Affiliation(s)
- Yukihiro Ohno
- Laboratory of Pharmacology, Osaka University of Pharmaceutical Sciences, Takatsuki, 569–1094, Japan
| | - Saki Shimizu
- Laboratory of Pharmacology, Osaka University of Pharmaceutical Sciences, Takatsuki, 569–1094, Japan
| | - Ayaka Tatara
- Laboratory of Pharmacology, Osaka University of Pharmaceutical Sciences, Takatsuki, 569–1094, Japan
| | - Takuji Imaoku
- Laboratory of Pharmacology, Osaka University of Pharmaceutical Sciences, Takatsuki, 569–1094, Japan
| | - Takahiro Ishii
- Department of Physiology and Neurobiology, Graduate School of Medicine, Kyoto University, Kyoto, 606–8501, Japan
| | | | - Tadao Serikawa
- Laboratory of Pharmacology, Osaka University of Pharmaceutical Sciences, Takatsuki, 569–1094, Japan
- Institute of Laboratory Animals, Graduate School of Medicine, Kyoto University, Kyoto, 606–8501, Japan
| | - Takashi Kuramoto
- Institute of Laboratory Animals, Graduate School of Medicine, Kyoto University, Kyoto, 606–8501, Japan
| |
Collapse
|
25
|
Abstract
With the wide availability of massively parallel sequencing technologies, genetic mapping has become the rate limiting step in mammalian forward genetics. Here we introduce a method for real-time identification of N-ethyl-N-nitrosourea-induced mutations that cause phenotypes in mice. All mutations are identified by whole exome G1 progenitor sequencing and their zygosity is established in G2/G3 mice before phenotypic assessment. Quantitative and qualitative traits, including lethal effects, in single or multiple combined pedigrees are then analyzed with Linkage Analyzer, a software program that detects significant linkage between individual mutations and aberrant phenotypic scores and presents processed data as Manhattan plots. As multiple alleles of genes are acquired through mutagenesis, pooled "superpedigrees" are created to analyze the effects. Our method is distinguished from conventional forward genetic methods because it permits (1) unbiased declaration of mappable phenotypes, including those that are incompletely penetrant (2), automated identification of causative mutations concurrent with phenotypic screening, without the need to outcross mutant mice to another strain and backcross them, and (3) exclusion of genes not involved in phenotypes of interest. We validated our approach and Linkage Analyzer for the identification of 47 mutations in 45 previously known genes causative for adaptive immune phenotypes; our analysis also implicated 474 genes not previously associated with immune function. The method described here permits forward genetic analysis in mice, limited only by the rates of mutant production and screening.
Collapse
|
26
|
microRNA profiling in three main stages during porcine spermatogenesis. J Assist Reprod Genet 2015; 32:451-60. [PMID: 25563581 DOI: 10.1007/s10815-014-0406-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Accepted: 12/11/2014] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Spermatogenesis is an intricate biological event wherein an undifferentiated spermatogonium develops into mature sperms. MicroRNAs are a type of single strand small non-coding RNA molecule and are implicated in the regulation of many crucial pathways during cell proliferation, apoptosis, and differentiation. METHOD Here, we present a comprehensive comparison of miRNA expression profiling in three main stages during porcine spermatogenesis using high-throughput sequencing. RESULTS We built three small RNA libraries for the testis, the epididymis and the ejaculated sperm from a Landrace boar, and in total obtained 3821 precursor hairpins encoding for 4761 mature miRNAs, of which 23 are miRNA*. Notably, 940 precursor miRNAs produced both the 5'- and 3'- strands as sister pairs, indicating the distinctive expression patterns of germ cell miRNAs. Additionally, 418 out of 710 co-expressed miRNAs were identified as being differentially expressed between libraries (P < 0.001). Apart from the sexual specific X chromosome, many miRNAs were found to be located on chromosome 12, which may play potential roles in spermatogenesis according to the result of synteny analysis with human and mouse. The Gene Ontology and KEGG pathway analysis revealed that the target genes of co-expressed miRNAs were highly involved in the cell cycle process, metal ion binding, modification of plasma membrane, and the p53 signal pathway.
Collapse
|
27
|
Tenin G, Clowes C, Wolton K, Krejci E, Wright JA, Lovell SC, Sedmera D, Hentges KE. Erbb2 is required for cardiac atrial electrical activity during development. PLoS One 2014; 9:e107041. [PMID: 25269082 PMCID: PMC4182046 DOI: 10.1371/journal.pone.0107041] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Accepted: 08/13/2014] [Indexed: 01/16/2023] Open
Abstract
The heart is the first organ required to function during embryonic development and is absolutely necessary for embryo survival. Cardiac activity is dependent on both the sinoatrial node (SAN), which is the pacemaker of heart's electrical activity, and the cardiac conduction system which transduces the electrical signal though the heart tissue, leading to heart muscle contractions. Defects in the development of cardiac electrical function may lead to severe heart disorders. The Erbb2 (Epidermal Growth Factor Receptor 2) gene encodes a member of the EGF receptor family of receptor tyrosine kinases. The Erbb2 receptor lacks ligand-binding activity but forms heterodimers with other EGF receptors, stabilising their ligand binding and enhancing kinase-mediated activation of downstream signalling pathways. Erbb2 is absolutely necessary in normal embryonic development and homozygous mouse knock-out Erbb2 embryos die at embryonic day (E)10.5 due to severe cardiac defects. We have isolated a mouse line, l11Jus8, from a random chemical mutagenesis screen, which carries a hypomorphic missense mutation in the Erbb2 gene. Homozygous mutant embryos exhibit embryonic lethality by E12.5-13. The l11Jus8 mutants display cardiac haemorrhage and a failure of atrial function due to defects in atrial electrical signal propagation, leading to an atrial-specific conduction block, which does not affect ventricular conduction. The l11Jus8 mutant phenotype is distinct from those reported for Erbb2 knockout mouse mutants. Thus, the l11Jus8 mouse reveals a novel function of Erbb2 during atrial conduction system development, which when disrupted causes death at mid-gestation.
Collapse
Affiliation(s)
- Gennadiy Tenin
- Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom
| | - Christopher Clowes
- Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom
| | - Kathryn Wolton
- Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom
| | - Eliska Krejci
- Institute of Anatomy, First Faculty of Medicine, Charles University, Prague, and Institute of Physiology, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | | | - Simon C. Lovell
- Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom
| | - David Sedmera
- Institute of Anatomy, First Faculty of Medicine, Charles University, Prague, and Institute of Physiology, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Kathryn E. Hentges
- Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom
- * E-mail:
| |
Collapse
|
28
|
Stottmann R, Beier DR. ENU Mutagenesis in the Mouse. CURRENT PROTOCOLS IN HUMAN GENETICS 2014; 82:15.4.1-15.4.10. [PMID: 25042716 PMCID: PMC4113905 DOI: 10.1002/0471142905.hg1504s82] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
This unit describes the treatment of laboratory mice with the mutagen N-ethyl-N-nitrosourea (ENU) to induce very highly increased rates of mutation throughout the genome. Further, it describes several popular mating schemes designed to produce animals displaying phenotypes associated with the induced mutations.
Collapse
Affiliation(s)
- Rolf Stottmann
- Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - David R. Beier
- Center for Developmental Biology and Regenerative Medicine, Seattle Children’s Research Institute
| |
Collapse
|
29
|
Peterson RT. Discovery of therapeutic targets by phenotype-based zebrafish screens. DRUG DISCOVERY TODAY. TECHNOLOGIES 2014; 1:49-54. [PMID: 24981267 DOI: 10.1016/j.ddtec.2004.07.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
The easy identification of phenotypes in the transparent zebrafish embryo has enabled numerous genetic, antisense morpholino oligonucleotide, and small molecule screens. Can zebrafish screens also be used for unbiased discovery of novel drug targets?:
Collapse
Affiliation(s)
- Randall T Peterson
- Developmental Biology Laboratory, Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, 149 13th Street, Charlestown, MA 02129, USA.
| |
Collapse
|
30
|
Clowes C, Boylan MGS, Ridge LA, Barnes E, Wright JA, Hentges KE. The functional diversity of essential genes required for mammalian cardiac development. Genesis 2014; 52:713-37. [PMID: 24866031 PMCID: PMC4141749 DOI: 10.1002/dvg.22794] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Revised: 05/22/2014] [Accepted: 05/23/2014] [Indexed: 01/04/2023]
Abstract
Genes required for an organism to develop to maturity (for which no other gene can compensate) are considered essential. The continuing functional annotation of the mouse genome has enabled the identification of many essential genes required for specific developmental processes including cardiac development. Patterns are now emerging regarding the functional nature of genes required at specific points throughout gestation. Essential genes required for development beyond cardiac progenitor cell migration and induction include a small and functionally homogenous group encoding transcription factors, ligands and receptors. Actions of core cardiogenic transcription factors from the Gata, Nkx, Mef, Hand, and Tbx families trigger a marked expansion in the functional diversity of essential genes from midgestation onwards. As the embryo grows in size and complexity, genes required to maintain a functional heartbeat and to provide muscular strength and regulate blood flow are well represented. These essential genes regulate further specialization and polarization of cell types along with proliferative, migratory, adhesive, contractile, and structural processes. The identification of patterns regarding the functional nature of essential genes across numerous developmental systems may aid prediction of further essential genes and those important to development and/or progression of disease. genesis 52:713–737, 2014.
Collapse
Affiliation(s)
- Christopher Clowes
- Faculty of Life Sciences, University of Manchester, Michael Smith Building, Oxford Road, Manchester, United Kingdom
| | | | | | | | | | | |
Collapse
|
31
|
Abstract
This article describes the treatment of laboratory mice with the mutagen N-ethyl-N-nitrosourea (ENU) to induce very highly increased rates of mutation throughout the genome. Further, it describes several popular mating schemes designed to produce animals displaying phenotypes associated with the induced mutations.
Collapse
Affiliation(s)
- Rolf Stottmann
- Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | | |
Collapse
|
32
|
Francis JS, Markov V, Leone P. Dietary triheptanoin rescues oligodendrocyte loss, dysmyelination and motor function in the nur7 mouse model of Canavan disease. J Inherit Metab Dis 2014; 37:369-81. [PMID: 24288037 DOI: 10.1007/s10545-013-9663-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Revised: 10/24/2013] [Accepted: 11/11/2013] [Indexed: 12/11/2022]
Abstract
The inherited pediatric leukodystrophy Canavan disease is characterized by dysmyelination and severe spongiform degeneration, and is currently refractory to treatment. A definitive understanding of core disease mechanisms is lacking, but pathology is believed to result at least in part compromised fatty acid synthesis during myelination. Recent evidence generated in an animal model suggests that the breakdown of N-acetylaspartate metabolism in CD results in a heightened coupling of fatty acid synthesis to oligodendrocyte oxidative metabolism during the early stages of myelination, thereby causing acute oxidative stress. We present here the results of a dietary intervention designed to support oxidative integrity during developmental myelination in the nur7 mouse model of Canavan disease. Provision of the odd carbon triglyceride triheptanoin to neonatal nur7 mice reduced oxidative stress, promoted long-term oligodendrocyte survival, and increased myelin in the brain. Improvements in oligodendrocyte survival and myelination were associated with a highly significant reduction in spongiform degeneration and improved motor function in triheptanoin treated mice. Initiation of triheptanoin treatment in older animals resulted in markedly more modest effects on these same pathological indices, indicating a window of therapeutic intervention that corresponds with developmental myelination. These results support the targeting of oxidative integrity at early stages of Canavan disease, and provide a foundation for the clinical development of a non-invasive dietary triheptanoin treatment regimen.
Collapse
Affiliation(s)
- Jeremy S Francis
- Cell and Gene Therapy Center, Department of Cell Biology, Rowan University School of Osteopathic Medicine, 40 East Laurel Rd, Stratford, NJ, USA,
| | | | | |
Collapse
|
33
|
Probst FJ, Corrigan RR, del Gaudio D, Salinger AP, Lorenzo I, Gao SS, Chiu I, Xia A, Oghalai JS, Justice MJ. A point mutation in the gene for asparagine-linked glycosylation 10B (Alg10b) causes nonsyndromic hearing impairment in mice (Mus musculus). PLoS One 2013; 8:e80408. [PMID: 24303013 PMCID: PMC3841196 DOI: 10.1371/journal.pone.0080408] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Accepted: 10/02/2013] [Indexed: 01/10/2023] Open
Abstract
The study of mouse hearing impairment mutants has led to the identification of a number of human hearing impairment genes and has greatly furthered our understanding of the physiology of hearing. The novel mouse mutant neurological/sensory 5 (nse5) demonstrates a significantly reduced or absent startle response to sound and is therefore a potential murine model of human hearing impairment. Genetic analysis of 500 intercross progeny localized the mutant locus to a 524 kilobase (kb) interval on mouse chromosome 15. A missense mutation in a highly-conserved amino acid was found in the asparagine-linked glycosylation 10B gene (Alg10b), which is within the critical interval for the nse5 mutation. A 20.4 kb transgene containing a wildtype copy of the Alg10b gene rescued the mutant phenotype in nse5/nse5 homozygous animals, confirming that the mutation in Alg10b is responsible for the nse5/nse5 mutant phenotype. Homozygous nse5/nse5 mutants had abnormal auditory brainstem responses (ABRs), distortion product otoacoustic emissions (DPOAEs), and cochlear microphonics (CMs). Endocochlear potentials (EPs), on the other hand, were normal. ABRs and DPOAEs also confirmed the rescue of the mutant nse5/nse5 phenotype by the wildtype Alg10b transgene. These results suggested a defect in the outer hair cells of mutant animals, which was confirmed by histologic analysis. This is the first report of mutation in a gene involved in the asparagine (N)-linked glycosylation pathway causing nonsyndromic hearing impairment, and it suggests that the hearing apparatus, and the outer hair cells in particular, are exquisitely sensitive to perturbations of the N-linked glycosylation pathway.
Collapse
Affiliation(s)
- Frank J. Probst
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Rebecca R. Corrigan
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Daniela del Gaudio
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Andrew P. Salinger
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Isabel Lorenzo
- Genetically Engineered Mouse Shared Resource, Baylor College of Medicine, Houston, Texas, United States of America
| | - Simon S. Gao
- Department of Bioengineering, Rice University, Houston, Texas, United States of America
| | - Ilene Chiu
- Department of Otolaryngology-Head and Neck Surgery, Baylor College of Medicine, Houston, Texas, United States of America
| | - Anping Xia
- Department of Otolaryngology-Head and Neck Surgery, Stanford University, Stanford, California, United States of America
| | - John S. Oghalai
- Department of Otolaryngology-Head and Neck Surgery, Stanford University, Stanford, California, United States of America
| | - Monica J. Justice
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
- * E-mail:
| |
Collapse
|
34
|
A suppressor screen in Mecp2 mutant mice implicates cholesterol metabolism in Rett syndrome. Nat Genet 2013; 45:1013-20. [PMID: 23892605 PMCID: PMC3837522 DOI: 10.1038/ng.2714] [Citation(s) in RCA: 155] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2013] [Accepted: 06/24/2013] [Indexed: 12/13/2022]
Abstract
Mutations in methyl CpG binding protein 2 (MECP2) cause Rett Syndrome, the most severe autism spectrum disorder. Re-expressing Mecp2 in symptomatic Mecp2 null mice dramatically improves function and longevity, providing hope that therapeutic intervention is possible in humans. To identify pathways in disease pathology for therapeutic intervention, a dominant ENU mutagenesis suppressor screen was carried out in Mecp2 null mice. Five suppressors that ameliorate symptoms of Mecp2 loss were isolated. Here we show that a stop codon mutation in squalene epoxidase (Sqle), a rate-limiting enzyme in cholesterol biosynthesis underlies suppression in one line. Subsequently, we show that lipid metabolism is perturbed in the brain and liver of Mecp2 null males. Consistently, statin drugs improve systemic perturbations of lipid metabolism, alleviate motor symptoms and confer increased longevity in Mecp2 mutant mice. The genetic screen therefore points to cholesterol homeostasis as a potential target for the treatment of Rett patients.
Collapse
|
35
|
Abstract
The gene encoding the WD repeat-containing protein 81 (WDR81) has recently been described as the disease locus in a consanguineous family that suffers from cerebellar ataxia, mental retardation, and quadrupedal locomotion syndrome (CAMRQ2). Adult mice from the N-ethyl-N-nitrosourea-induced mutant mouse line nur5 display tremor and an abnormal gait, as well as Purkinje cell degeneration and photoreceptor cell loss. We have used polymorphic marker mapping to demonstrate that affected nur5 mice carry a missense mutation, L1349P, in the Wdr81 gene. Moreover, homozygous nur5 mice that carry a wild-type Wdr81 transgene are rescued from the abnormal phenotype, indicating that Wdr81 is the causative gene in nur5. WDR81 is expressed in Purkinje cells and photoreceptor cells, among other CNS neurons, and like the human mutation, the nur5 modification lies in the predicted major facilitator superfamily domain of the WDR81 protein. Electron microscopy analysis revealed that a subset of mitochondria in Purkinje cell dendrites of the mutant animals displayed an aberrant, large spheroid-like structure. Moreover, immunoelectron microscopy and analysis of mitochondrial-enriched cerebellum fractions indicate that WDR81 is localized in mitochondria of Purkinje cell neurons. Because the nur5 mouse mutant demonstrates phenotypic similarities to the human disease, it provides a valuable genetic model for elucidating the pathogenic mechanism of the WDR81 mutation in CAMRQ2.
Collapse
|
36
|
van der Weyden L, Adams DJ. Cancer of mice and men: old twists and new tails. J Pathol 2013; 230:4-16. [PMID: 23436574 DOI: 10.1002/path.4184] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2013] [Revised: 01/28/2013] [Accepted: 02/16/2013] [Indexed: 12/18/2022]
Abstract
In this review we set out to celebrate the contribution that mouse models of human cancer have made to our understanding of the fundamental mechanisms driving tumourigenesis. We take the opportunity to look forward to how the mouse will be used to model cancer and the tools and technologies that will be applied, and indulge in looking back at the key advances the mouse has made possible.
Collapse
|
37
|
Yuan S, Zaidi S, Brueckner M. Congenital heart disease: emerging themes linking genetics and development. Curr Opin Genet Dev 2013; 23:352-9. [PMID: 23790954 PMCID: PMC4154700 DOI: 10.1016/j.gde.2013.05.004] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Revised: 05/14/2013] [Accepted: 05/16/2013] [Indexed: 12/13/2022]
Abstract
Although congenital heart disease (CHD) is the most common survivable birth defect, the etiology of most CHD remains unclear. Several lines of evidence from humans and vertebrate models have supported a genetic component for CHD, yet the extreme locus heterogeneity and lack of a distinct genotype-phenotype correlation have limited causative gene discovery. However, recent advances in genomic technologies are permitting detailed evaluation of the genetic abnormalities in large cohorts of CHD patients. This has led to the identification of copy-number variation and de novo mutations together accounting for up to 15% of CHD. Further, new strategies coupling human genetics with model organisms have provided mechanistic insights into the molecular and developmental pathways underlying CHD pathogenesis, notably chromatin remodeling and ciliary signaling.
Collapse
Affiliation(s)
- Shiaulou Yuan
- Department of Pediatrics, Yale University School of Medicine, 15 York St., New Haven, CT 06520
- Department of Genetics, Yale University School of Medicine, 15 York St., New Haven, CT 06520
| | - Samir Zaidi
- Department of Genetics, Yale University School of Medicine, 15 York St., New Haven, CT 06520
| | - Martina Brueckner
- Department of Pediatrics, Yale University School of Medicine, 15 York St., New Haven, CT 06520
- Department of Genetics, Yale University School of Medicine, 15 York St., New Haven, CT 06520
| |
Collapse
|
38
|
Beck TF, Shchelochkov OA, Yu Z, Kim BJ, Hernández-García A, Zaveri HP, Bishop C, Overbeek PA, Stockton DW, Justice MJ, Scott DA. Novel frem1-related mouse phenotypes and evidence of genetic interactions with gata4 and slit3. PLoS One 2013; 8:e58830. [PMID: 23536828 PMCID: PMC3594180 DOI: 10.1371/journal.pone.0058830] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2012] [Accepted: 02/07/2013] [Indexed: 11/27/2022] Open
Abstract
The FRAS1-related extracellular matrix 1 (FREM1) gene encodes an extracellular matrix protein that plays a critical role in the development of multiple organ systems. In humans, recessive mutations in FREM1 cause eye defects, congenital diaphragmatic hernia, renal anomalies and anorectal malformations including anteriorly placed anus. A similar constellation of findings-microphthalmia, cryptophthalmos, congenital diaphragmatic hernia, renal agenesis and rectal prolapse-have been described in FREM1-deficient mice. In this paper, we identify a homozygous Frem1 missense mutation (c.1687A>T, p.Ile563Phe) in an N-ethyl-N-nitrosourea (ENU)-derived mouse strain, crf11, with microphthalmia, cryptophthalmos, renal agenesis and rectal prolapse. This mutation affects a highly conserved residue in FREM1's third CSPG domain. The p.Ile563Phe change is predicted to be deleterious and to cause decreased FREM1 protein stability. The crf11 allele also fails to complement the previously described eyes2 allele of Frem1 (p.Lys826*) providing further evidence that the crf11 phenotype is due to changes affecting Frem1 function. We then use mice bearing the crf11 and eyes2 alleles to identify lung lobulation defects and decreased anogenital distance in males as novel phenotypes associated with FREM1 deficiency in mice. Due to phenotypic overlaps between FREM1-deficient mice and mice that are deficient for the retinoic acid-responsive transcription factor GATA4 and the extracellular matrix protein SLIT3, we also perform experiments to look for in vivo genetic interactions between the genes that encode these proteins. These experiments reveal that Frem1 interacts genetically with Gata4 in the development of lung lobulation defects and with Slit3 in the development of renal agenesis. These results demonstrate that FREM1-deficient mice faithfully recapitulate many of the phenotypes seen in individuals with FREM1 deficiency and that variations in GATA4 and SLIT3 expression modulate some FREM1-related phenotypes in mice.
Collapse
Affiliation(s)
- Tyler F. Beck
- Departments of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Oleg A. Shchelochkov
- Department of Pediatrics, The University of Iowa, Iowa City, Iowa, United States of America
| | - Zhiyin Yu
- Departments of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Bum Jun Kim
- Departments of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Andrés Hernández-García
- Departments of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Hitisha P. Zaveri
- Departments of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Colin Bishop
- The Wake Forest Institute for Regenerative Medicine, Winston Salem, North Carolina, United States of America
| | - Paul A. Overbeek
- Departments of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
- Molecular and Cell Biology, Baylor College of Medicine, Houston, Texas, United States of America
| | - David W. Stockton
- Departments of Pediatrics and Internal Medicine, Wayne State University, Detroit, Michigan, United States of America
| | - Monica J. Justice
- Departments of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Daryl A. Scott
- Departments of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
- Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas, United States of America
| |
Collapse
|
39
|
Genome-wide ENU mutagenesis in combination with high density SNP analysis and exome sequencing provides rapid identification of novel mouse models of developmental disease. PLoS One 2013; 8:e55429. [PMID: 23469164 PMCID: PMC3585849 DOI: 10.1371/journal.pone.0055429] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2012] [Accepted: 12/22/2012] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Mice harbouring gene mutations that cause phenotypic abnormalities during organogenesis are invaluable tools for linking gene function to normal development and human disorders. To generate mouse models harbouring novel alleles that are involved in organogenesis we conducted a phenotype-driven, genome-wide mutagenesis screen in mice using the mutagen N-ethyl-N-nitrosourea (ENU). METHODOLOGY/PRINCIPAL FINDINGS ENU was injected into male C57BL/6 mice and the mutations transmitted through the germ-line. ENU-induced mutations were bred to homozygosity and G3 embryos screened at embryonic day (E) 13.5 and E18.5 for abnormalities in limb and craniofacial structures, skin, blood, vasculature, lungs, gut, kidneys, ureters and gonads. From 52 pedigrees screened 15 were detected with anomalies in one or more of the structures/organs screened. Using single nucleotide polymorphism (SNP)-based linkage analysis in conjunction with candidate gene or next-generation sequencing (NGS) we identified novel recessive alleles for Fras1, Ift140 and Lig1. CONCLUSIONS/SIGNIFICANCE In this study we have generated mouse models in which the anomalies closely mimic those seen in human disorders. The association between novel mutant alleles and phenotypes will lead to a better understanding of gene function in normal development and establish how their dysfunction causes human anomalies and disease.
Collapse
|
40
|
Bartnikas TB, Steinbicker AU, Campagna DR, Blevins S, Woodward LS, Herrera C, Bloch KD, Justice MJ, Fleming MD. Identification and characterization of a novel murine allele of Tmprss6. Haematologica 2013; 98:854-61. [PMID: 23300183 DOI: 10.3324/haematol.2012.074617] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Mutagenesis screens can establish mouse models of utility for the study of critical biological processes such as iron metabolism. Such screens can produce mutations in novel genes or establish novel alleles of known genes, both of which can be useful tools for study. In order to identify genes of relevance to hematologic as well as other phenotypes, we performed N-ethyl-N-nitrosourea mutagenesis in C57BL/6J mice. An anemic mouse was identified and a putative mutation was characterized by mapping, sequencing and in vitro activity analysis. The mouse strain was backcrossed for ten generations then phenotypically characterized with respect to a previously established null mouse strain. Potential modifying loci were identified by quantitative trait locus analysis. Mapping and sequencing in an anemic mouse termed hem8 identified an I286F substitution in Tmprss6, a serine protease essential for iron metabolism; this substitution impaired in vitro protease activity. After backcrossing to C57BL6/J for ten generations, the hem8(-/-) strain exhibited a phenotype similar in some but not all aspects to that of Tmprss6(-/-) mice. The hem8 and Tmprss6-null mutations were allelic. Both hem8(-/-) and Tmprss6(-/-) mice responded similarly to pharmacological modulators of bone morphogenetic protein signaling, a key regulator of iron metabolism. Quantitative trait locus analysis in the hem8 strain identified potential modifying loci on chromosomes 2, 4, 7 and 10. In conclusion, the hem8 mouse model carries a novel allele of Tmprss6. Potential uses for this strain in the study of iron metabolism are discussed.
Collapse
Affiliation(s)
- Thomas B Bartnikas
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Lossie AC, Lo CL, Baumgarner KM, Cramer MJ, Garner JP, Justice MJ. ENU mutagenesis reveals that Notchless homolog 1 (Drosophila) affects Cdkn1a and several members of the Wnt pathway during murine pre-implantation development. BMC Genet 2012; 13:106. [PMID: 23231322 PMCID: PMC3558363 DOI: 10.1186/1471-2156-13-106] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2012] [Accepted: 11/24/2012] [Indexed: 01/20/2023] Open
Abstract
Background Our interests lie in determining the genes and genetic pathways that are important for establishing and maintaining maternal-fetal interactions during pregnancy. Mutation analysis targeted to a 34 Mb domain flanked by Trp53 and Wnt3 demonstrates that this region of mouse chromosome 11 contains a large number of essential genes. Two mutant alleles (l11Jus1 and l11Jus4), which fall into the same complementation group, survive through implantation but fail prior to gastrulation. Results Through a positional cloning strategy, we discovered that these homozygous mutant alleles contain non-conservative missense mutations in the Notchless homolog 1 (Drosophila) (Nle1) gene. NLE1 is a member of the large WD40-repeat protein family, and is thought to signal via the canonical NOTCH pathway in vertebrates. However, the phenotype of the Nle1 mutant mice is much more severe than single Notch receptor mutations or even in animals in which NOTCH signaling is blocked. To test the hypothesis that NLE1 functions in multiple signaling pathways during pre-implantation development, we examined expression of multiple Notch downstream target genes, as well as select members of the Wnt pathway in wild-type and mutant embryos. We did not detect altered expression of any primary members of the Notch pathway or in Notch downstream target genes. However, our data reveal that Cdkn1a, a NOTCH target, was upregulated in Nle1 mutants, while several members of the Wnt pathway are downregulated. In addition, we found that Nle1 mutant embryos undergo caspase-mediated apoptosis as hatched blastocysts, but not as morulae or blastocysts. Conclusions Taken together, these results uncover potential novel functions for NLE1 in the WNT and CDKN1A pathways during embryonic development in mammals.
Collapse
Affiliation(s)
- Amy C Lossie
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907, USA.
| | | | | | | | | | | |
Collapse
|
42
|
Ayadi A, Birling MC, Bottomley J, Bussell J, Fuchs H, Fray M, Gailus-Durner V, Greenaway S, Houghton R, Karp N, Leblanc S, Lengger C, Maier H, Mallon AM, Marschall S, Melvin D, Morgan H, Pavlovic G, Ryder E, Skarnes WC, Selloum M, Ramirez-Solis R, Sorg T, Teboul L, Vasseur L, Walling A, Weaver T, Wells S, White JK, Bradley A, Adams DJ, Steel KP, Hrabě de Angelis M, Brown SD, Herault Y. Mouse large-scale phenotyping initiatives: overview of the European Mouse Disease Clinic (EUMODIC) and of the Wellcome Trust Sanger Institute Mouse Genetics Project. Mamm Genome 2012; 23:600-10. [PMID: 22961258 PMCID: PMC3463797 DOI: 10.1007/s00335-012-9418-y] [Citation(s) in RCA: 103] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2012] [Accepted: 07/23/2012] [Indexed: 12/17/2022]
Abstract
Two large-scale phenotyping efforts, the European Mouse Disease Clinic (EUMODIC) and the Wellcome Trust Sanger Institute Mouse Genetics Project (SANGER-MGP), started during the late 2000s with the aim to deliver a comprehensive assessment of phenotypes or to screen for robust indicators of diseases in mouse mutants. They both took advantage of available mouse mutant lines but predominantly of the embryonic stem (ES) cells resources derived from the European Conditional Mouse Mutagenesis programme (EUCOMM) and the Knockout Mouse Project (KOMP) to produce and study 799 mouse models that were systematically analysed with a comprehensive set of physiological and behavioural paradigms. They captured more than 400 variables and an additional panel of metadata describing the conditions of the tests. All the data are now available through EuroPhenome database (www.europhenome.org) and the WTSI mouse portal (http://www.sanger.ac.uk/mouseportal/), and the corresponding mouse lines are available through the European Mouse Mutant Archive (EMMA), the International Knockout Mouse Consortium (IKMC), or the Knockout Mouse Project (KOMP) Repository. Overall conclusions from both studies converged, with at least one phenotype scored in at least 80% of the mutant lines. In addition, 57% of the lines were viable, 13% subviable, 30% embryonic lethal, and 7% displayed fertility impairments. These efforts provide an important underpinning for a future global programme that will undertake the complete functional annotation of the mammalian genome in the mouse model.
Collapse
Affiliation(s)
- Abdel Ayadi
- Institut Clinique de la Souris, PHENOMIN, IGBMC/ICS-MCI, CNRS, INSERM, Université de Strasbourg, UMR7104, UMR964, 1 rue Laurent Fries, 67404 Illkirch, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Smith CL, Eppig JT. The Mammalian Phenotype Ontology as a unifying standard for experimental and high-throughput phenotyping data. Mamm Genome 2012; 23:653-68. [PMID: 22961259 PMCID: PMC3463787 DOI: 10.1007/s00335-012-9421-3] [Citation(s) in RCA: 126] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2012] [Accepted: 07/24/2012] [Indexed: 01/16/2023]
Abstract
The Mammalian Phenotype Ontology (MP) is a structured vocabulary for describing mammalian phenotypes and serves as a critical tool for efficient annotation and comprehensive retrieval of phenotype data. Importantly, the ontology contains broad and specific terms, facilitating annotation of data from initial observations or screens and detailed data from subsequent experimental research. Using the ontology structure, data are retrieved inclusively, i.e., data annotated to chosen terms and to terms subordinate in the hierarchy. Thus, searching for "abnormal craniofacial morphology" also returns annotations to "megacephaly" and "microcephaly," more specific terms in the hierarchy path. The development and refinement of the MP is ongoing, with new terms and modifications to its organization undergoing continuous assessment as users and expert reviewers propose expansions and revisions. A wealth of phenotype data on mouse mutations and variants annotated to the MP already exists in the Mouse Genome Informatics database. These data, along with data curated to the MP by many mouse mutagenesis programs and mouse repositories, provide a platform for comparative analyses and correlative discoveries. The MP provides a standard underpinning to mouse phenotype descriptions for existing and future experimental and large-scale phenotyping projects. In this review we describe the MP as it presently exists, its application to phenotype annotations, the relationship of the MP to other ontologies, and the integration of the MP within large-scale phenotyping projects. Finally we discuss future application of the MP in providing standard descriptors of the phenotype pipeline test results from the International Mouse Phenotype Consortium projects.
Collapse
|
44
|
Abstract
ENU mutagenesis is a forward genetics strategy in which random mutagenesis and phenotypic screening is used to identify genes based on the phenotype induced when they are mutated. A modifier screen is a type of screen in which mice with a pre-existing phenotype are utilized to identify mutations that can enhance or suppress this phenotype. This approach has the potential to uncover missing pathway members, reveal novel genetic interactions, and pinpoint new drug targets. Considerations when planning a suppressor screen include current knowledge, genomic footprint, penetrance, variance, robustness, latency of the starting phenotype, viability, fertility, genetic background and ENU tolerance of starting strain, screening assay, mouse numbers required, and mutation identification strategy. Practical advice on each of these is provided in this review. Curr. Protoc. Mouse Biol. 2:75-87 © 2012 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Marina R Carpinelli
- Molecular Medicine Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, Australia.,Hearing Co-Operative Research Centre, Melbourne, Australia.,Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Australia
| | - Rachel A Burt
- Molecular Medicine Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, Australia.,Hearing Co-Operative Research Centre, Melbourne, Australia.,Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Australia
| | - Benjamin T Kile
- Cancer and Hematology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, Australia
| |
Collapse
|
45
|
ENU mutagenesis screen to establish motor phenotypes in wild-type mice and modifiers of a pre-existing motor phenotype in tau mutant mice. J Biomed Biotechnol 2011; 2011:130947. [PMID: 22219655 PMCID: PMC3246812 DOI: 10.1155/2011/130947] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2011] [Accepted: 11/04/2011] [Indexed: 11/20/2022] Open
Abstract
Modifier screening is a powerful genetic tool. While not widely used in the vertebrate system, we applied these tools to transgenic mouse strains that recapitulate key aspects of Alzheimer's disease (AD), such as tau-expressing mice. These are characterized by a robust pathology including both motor and memory impairment. The phenotype can be modulated by ENU mutagenesis, which results in novel mutant mouse strains and allows identifying the underlying gene/mutation. Here we discuss this strategy in detail. We firstly obtained pedigrees that modify the tau-related motor phenotype, with mapping ongoing. We further obtained transgene-independent motor pedigrees: (i) hyperactive, circling ENU 37 mice with a causal mutation in the Tbx1 gene—the complete knock-out of Tbx1 models DiGeorge Syndrome; (ii) ENU12/301 mice that show sudden jerky movements and tremor constantly; they have a causal mutation in the Kcnq1 gene, modelling aspects of the Romano-Ward and Jervell and Lange-Nielsen syndromes; and (iii) ENU16/069 mice with tremor and hypermetric gait that have a causal mutation in the Mpz (Myelin Protein Zero) gene, modelling Charcot-Marie-Tooth disease type 1 (CMT1B). Together, we provide evidence for a real potential of an ENU mutagenesis to dissect motor functions in wild-type and tau mutant mice.
Collapse
|
46
|
Lo CL, Shen F, Baumgarner K, Cramer MJ, Lossie AC. Identification of 129S6/SvEvTac-specific polymorphisms on mouse chromosome 11. DNA Cell Biol 2011; 31:402-14. [PMID: 21988490 DOI: 10.1089/dna.2011.1353] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Polymorphisms such as single-nucleotide polymorphisms (SNPs) and insertions/deletions (Indels) can be associated with phenotypic traits and be used as markers for disease diagnosis. Identification of these genetic variations within laboratory mice is crucial to improve our understanding of the genetic background of the mice used for research. As part of a positional cloning project, we sequenced six genes (Mettl16, Evi2a, Psmd11, Cct6d, Rffl, and Ap2b1) within a 6.8-Mb domain of mmu chr 11 in the C57BL/6J and 129S6/SvEvTac inbred strains. Although 129S6/SvEvTac is widely used in the mouse community, there is very little current (or projected future) sequence information available for this strain. We identified 6 Indels and 21 novel SNPs and confirmed genotype information for 114 additional SNPs in these 6 genes. Mettl16 and Ap2b1 contained the largest numbers of variants between the C57BL/6J and 129S6/SvEvTac strains. In addition, we found five new SNPs between 129S6/SvEvTac and 129S1/SvImJ within the Ap2b1 locus. Although we did not detect differences between C57BL/6J and 129S6/SvEvTac within Evi2a, this locus contains a relatively high SNP density compared with the surrounding sequence. Our study highlights the genetic differences among three inbred mouse strains (C57BL/6J, 129S6/SvEvTac, and 129S1/SvImJ) and provides valuable sequence information that can be used to track alleles in genomics-based studies.
Collapse
Affiliation(s)
- Chiao-Ling Lo
- Department of Animal Sciences, Purdue University, West Lafayette, Indiana 47907, USA
| | | | | | | | | |
Collapse
|
47
|
Fairfield H, Gilbert GJ, Barter M, Corrigan RR, Curtain M, Ding Y, D'Ascenzo M, Gerhardt DJ, He C, Huang W, Richmond T, Rowe L, Probst FJ, Bergstrom DE, Murray SA, Bult C, Richardson J, Kile BT, Gut I, Hager J, Sigurdsson S, Mauceli E, Di Palma F, Lindblad-Toh K, Cunningham ML, Cox TC, Justice MJ, Spector MS, Lowe SW, Albert T, Donahue LR, Jeddeloh J, Shendure J, Reinholdt LG. Mutation discovery in mice by whole exome sequencing. Genome Biol 2011; 12:R86. [PMID: 21917142 PMCID: PMC3308049 DOI: 10.1186/gb-2011-12-9-r86] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2011] [Revised: 08/04/2011] [Accepted: 09/14/2011] [Indexed: 01/18/2023] Open
Abstract
We report the development and optimization of reagents for in-solution, hybridization-based capture of the mouse exome. By validating this approach in a multiple inbred strains and in novel mutant strains, we show that whole exome sequencing is a robust approach for discovery of putative mutations, irrespective of strain background. We found strong candidate mutations for the majority of mutant exomes sequenced, including new models of orofacial clefting, urogenital dysmorphology, kyphosis and autoimmune hepatitis.
Collapse
Affiliation(s)
| | | | - Mary Barter
- The Jackson Laboratory, 600 Main St, Bar Harbor, ME 04609, USA
| | - Rebecca R Corrigan
- Baylor College of Medicine, Department of Molecular and Human Genetics, One Baylor Plaza R804, Houston, Texas 77030, USA
| | | | - Yueming Ding
- Cold Spring Harbor Laboratory, One Bungtown Road, Cold Spring Harbor, NY 11724, USA
| | | | | | - Chao He
- National Center for Genome Analysis (CNAG), Parc Científic de Barcelona, Torre I, Baldiri Reixac, 408028 Barcelona, Spain
| | - Wenhui Huang
- Walter and Eliza Hall Institute, 1G Royal Parade, Parkville, Victoria 3052, Australia
| | | | - Lucy Rowe
- The Jackson Laboratory, 600 Main St, Bar Harbor, ME 04609, USA
| | - Frank J Probst
- Baylor College of Medicine, Department of Molecular and Human Genetics, One Baylor Plaza R804, Houston, Texas 77030, USA
| | | | | | - Carol Bult
- The Jackson Laboratory, 600 Main St, Bar Harbor, ME 04609, USA
| | - Joel Richardson
- The Jackson Laboratory, 600 Main St, Bar Harbor, ME 04609, USA
| | - Benjamin T Kile
- University of Washington, Department of Pediatrics, Division of Craniofacial Medicine and Seattle Children's Craniofacial Center, 4800 Sand Point Way NE, Seattle, WA 98105, USA
| | - Ivo Gut
- Regeneron Pharmaceuticals Inc., 777 Old Saw Mill River Road, Tarrytown, NY 10591, USA
| | - Jorg Hager
- Regeneron Pharmaceuticals Inc., 777 Old Saw Mill River Road, Tarrytown, NY 10591, USA
| | - Snaevar Sigurdsson
- Broad Institute of Massachusetts Institute of Technology and Harvard, 5 Cambridge Center, Cambridge, MA 02142, USA
| | - Evan Mauceli
- Broad Institute of Massachusetts Institute of Technology and Harvard, 5 Cambridge Center, Cambridge, MA 02142, USA
| | - Federica Di Palma
- Broad Institute of Massachusetts Institute of Technology and Harvard, 5 Cambridge Center, Cambridge, MA 02142, USA
| | - Kerstin Lindblad-Toh
- Broad Institute of Massachusetts Institute of Technology and Harvard, 5 Cambridge Center, Cambridge, MA 02142, USA
| | - Michael L Cunningham
- University of Washington, Department of Genome Sciences, Foege Building S-250, Box 355065, 3720 15th Ave NE, Seattle, WA 98195-5065, USA
| | - Timothy C Cox
- University of Washington, Department of Genome Sciences, Foege Building S-250, Box 355065, 3720 15th Ave NE, Seattle, WA 98195-5065, USA
| | - Monica J Justice
- Baylor College of Medicine, Department of Molecular and Human Genetics, One Baylor Plaza R804, Houston, Texas 77030, USA
| | - Mona S Spector
- National Center for Genome Analysis (CNAG), Parc Científic de Barcelona, Torre I, Baldiri Reixac, 408028 Barcelona, Spain
| | - Scott W Lowe
- National Center for Genome Analysis (CNAG), Parc Científic de Barcelona, Torre I, Baldiri Reixac, 408028 Barcelona, Spain
| | | | | | | | - Jay Shendure
- University of Washington, Department of Genome Sciences, Foege Building S-250, Box 355065, 3720 15th Ave NE, Seattle, WA 98195-5065, USA
| | | |
Collapse
|
48
|
Abstract
We describe an experimental approach for generating mutant alleles in rat spermatogonial stem cells (SSCs) using Sleeping Beauty (SB) transposon-mediated insertional mutagenesis. The protocol is based on mobilization of mutagenic gene-trap transposons from transfected plasmid vectors into the genomes of cultured stem cells. Cells with transposon insertions in expressed genes are selected on the basis of activation of an antibiotic-resistance gene encoded by the transposon. These gene-trap clones are transplanted into the testes of recipient males (either as monoclonal or polyclonal libraries); crossing of these founders with wild-type females allows the insertions to be passed to F(1) progeny. This simple, economic and user-friendly methodological pipeline enables screens for functional gene annotation in the rat, with applicability in other vertebrate models where germ line-competent stem cells have been established. The complete protocol from transfection of SSCs to the genotyping of heterozygous F(1) offspring that harbor genomic SB gene-trap insertions takes 5-6 months.
Collapse
|
49
|
Parsons TE, Schmidt EJ, Boughner JC, Jamniczky HA, Marcucio RS, Hallgrímsson B. Epigenetic integration of the developing brain and face. Dev Dyn 2011; 240:2233-44. [PMID: 21901785 DOI: 10.1002/dvdy.22729] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/19/2011] [Indexed: 12/13/2022] Open
Abstract
The integration of the brain and face and to what extent this relationship constrains or enables evolutionary change in the craniofacial complex is an issue of long-standing interest in vertebrate evolution. To investigate brain-face integration, we studied the covariation between the forebrain and midface at gestational days 10-10.5 in four strains of laboratory mice. We found that phenotypic variation in the forebrain is highly correlated with that of the face during face formation such that variation in the size of the forebrain correlates with the degree of prognathism and orientation of the facial prominences. This suggests strongly that the integration of the brain and face is relevant to the etiology of midfacial malformations such as orofacial clefts. This axis of integration also has important implications for the evolutionary developmental biology of the mammalian craniofacial complex.
Collapse
Affiliation(s)
- Trish E Parsons
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, Alberta, Canada
| | | | | | | | | | | |
Collapse
|
50
|
Sheridan R, Lampe K, Shanmukhappa SK, Putnam P, Keddache M, Divanovic S, Bezerra J, Hoebe K. Lampe1: an ENU-germline mutation causing spontaneous hepatosteatosis identified through targeted exon-enrichment and next-generation sequencing. PLoS One 2011; 6:e21979. [PMID: 21760938 PMCID: PMC3131302 DOI: 10.1371/journal.pone.0021979] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2011] [Accepted: 06/14/2011] [Indexed: 01/22/2023] Open
Abstract
Using a small scale ENU mutagenesis approach we identified a recessive germline mutant, designated Lampe1 that exhibited growth retardation and spontaneous hepatosteatosis. Low resolution mapping based on 20 intercrossed Lampe1 mice revealed linkage to a ∼14 Mb interval on the distal site of chromosome 11 containing a total of 285 genes. Exons and 50 bp flanking sequences within the critical region were enriched with sequence capture microarrays and subsequently analyzed by next-generation sequencing. Using this approach 98.1 percent of the targeted DNA was covered with a depth of 10 or more reads per nucleotide and 3 homozygote mutations were identified. Two mutations represented intronic nucleotide changes whereas one mutation affected a splice donor site in intron 11–12 of Palmitoyl Acetyl-coenzyme A oxygenase-1 (Acox1), causing skipping of exon 12. Phenotyping of Acox1Lampe1 mutants revealed a progression from hepatosteatosis to steatohepatitis, and ultimately hepatocellular carcinoma. The current approach provides a highly efficient and affordable method to identify causative mutations induced by ENU mutagenesis and animal models relevant to human pathology.
Collapse
Affiliation(s)
- Rachel Sheridan
- Department of Pathology and Laboratory Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Kristin Lampe
- Department of Molecular Immunology, Cincinnati Children's Hospital Research Foundation, Cincinnati, Ohio, United States of America
| | - Shiva Kumar Shanmukhappa
- Division of Comparative Medicine and Pathology, New England Primate Research Center, Harvard Medical School, Southborough, Massachusetts, United States of America
| | - Patrick Putnam
- Department of Human Genetics, Cincinnati Children's Hospital Research Foundation, Cincinnati, Ohio, United States of America
| | - Mehdi Keddache
- Department of Human Genetics, Cincinnati Children's Hospital Research Foundation, Cincinnati, Ohio, United States of America
| | - Senad Divanovic
- Department of Molecular Immunology, Cincinnati Children's Hospital Research Foundation, Cincinnati, Ohio, United States of America
| | - Jorge Bezerra
- Department of Gastroenterology, Hepatology and Nutrition, Cincinnati Children's Hospital Research Foundation, Cincinnati, Ohio, United States of America
| | - Kasper Hoebe
- Department of Molecular Immunology, Cincinnati Children's Hospital Research Foundation, Cincinnati, Ohio, United States of America
- * E-mail:
| |
Collapse
|