1
|
Rulli SB, Ahtiainen P, Ratner LD, Jonas K, Calandra RS, Poutanen M, Huhtaniemi I. Elevated chorionic gonadotropic hormone in transgenic mice induces parthenogenetic activation and ovarian teratomas. Mol Cell Endocrinol 2024; 587:112214. [PMID: 38537882 DOI: 10.1016/j.mce.2024.112214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 02/25/2024] [Accepted: 03/19/2024] [Indexed: 04/13/2024]
Abstract
Both male and female reproductive functions are impacted by altered gonadotrophin secretion and action, which may also influence the development of endocrine tumours. To ascertain if chronic hypersecretion of human chorionic gonadotropin (hCG) contributes to the development of gonadal tumours, double transgenic (TG) mice that overexpress hCGα- and β-subunits were analysed. By the age of two months, ovarian tumours with characteristics of teratomas developed with 100% penetrance. Teratomas were also seen in wild-type ovaries orthotopically transplanted into TG mice, demonstrating an endocrine/paracrine mechanism for the hCG-induced ovarian tumorigenesis. Both in vitro and in vivo experiments showed oocyte parthenogenetic activation in TG females. In addition, ovaries showed reduced ovulatory gene expression, inhibited ERK1/2 phosphorylation, and impaired cumulus cell expansion. Hence, persistently high endocrine hCG activity causes parthenogenetic activation and development of ovarian teratomas, along with altered follicle development and impaired ERK1/2 signalling, offering a novel mechanism associated with the molecular pathogenesis of ovarian teratomas.
Collapse
Affiliation(s)
- Susana B Rulli
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, Kiinamyllynkatu 10, FIN-20520, Turku, Finland; Instituto de Biología y Medicina Experimental-CONICET, Vuelta de Obligado 2490, 1428, Buenos Aires, Argentina; Centro de Estudios Biomédicos Básicos, Aplicados y Desarrollo (CEBBAD), Universidad Maimónides, Hidalgo 775, C1405BCK, Ciudad Autónoma de Buenos Aires, Argentina.
| | - Petteri Ahtiainen
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, Kiinamyllynkatu 10, FIN-20520, Turku, Finland
| | - Laura D Ratner
- Instituto de Biología y Medicina Experimental-CONICET, Vuelta de Obligado 2490, 1428, Buenos Aires, Argentina
| | - Kim Jonas
- Department of Digestion, Metabolism and Reproduction, Institute of Reproductive and Developmental Biology, Hammersmith Campus, Imperial College London, London, W12 0NN, UK; Department of Women and Children's Health, School of Population and Life Course Sciences, King's College London, London, SE1 1UL, UK
| | - Ricardo S Calandra
- Instituto de Biología y Medicina Experimental-CONICET, Vuelta de Obligado 2490, 1428, Buenos Aires, Argentina
| | - Matti Poutanen
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, Kiinamyllynkatu 10, FIN-20520, Turku, Finland; Turku Center for Disease Modeling, University of Turku, Kiinamyllynkatu 10, FIN-20520, Turku, Finland
| | - Ilpo Huhtaniemi
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, Kiinamyllynkatu 10, FIN-20520, Turku, Finland; Department of Digestion, Metabolism and Reproduction, Institute of Reproductive and Developmental Biology, Hammersmith Campus, Imperial College London, London, W12 0NN, UK
| |
Collapse
|
2
|
Liu Y, Fan H, Kang X, Hao Y, Wang N, Zheng H, Li Y, Kang S. A rare germline BMP15 missense mutation causes hereditary ovarian immature teratoma in human. Proc Natl Acad Sci U S A 2024; 121:e2310409121. [PMID: 38427603 DOI: 10.1073/pnas.2310409121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 01/11/2024] [Indexed: 03/03/2024] Open
Abstract
Ovarian immature teratomas (OITs) are malignant tumors originating from the ovarian germ cells that mainly occur during the first 30 y of a female's life. Early age of onset strongly suggests the presence of susceptibility gene mutations for the disease yet to be discovered. Whole exon sequencing was used to screen pathogenic mutations from pedigrees with OITs. A rare missense germline mutation (C262T) in the first exon of the BMP15 gene was identified. In silico calculation suggested that the mutation could impair the formation of mature peptides. In vitro experiments on cell lines confirmed that the mutation caused an 84.7% reduction in the secretion of mature BMP15. Clinical samples from OIT patients also showed a similar pattern of decrease in the BMP15 expression. In the transgenic mouse model, the spontaneous parthenogenetic activation significantly increased in oocytes carrying the T allele. Remarkably, a mouse carrying the T allele developed the phenotype of OIT. Oocyte-specific RNA sequencing revealed that abnormal activation of the H-Ras/MAPK pathway might contribute to the development of OIT. BMP15 was identified as a pathogenic gene for OIT which improved our understanding of the etiology of OIT and provided a potential biomarker for genetic screening of this disorder.
Collapse
Affiliation(s)
- Yakun Liu
- Department of Gynecology, Fourth Hospital, Hebei Medical University, Shijiazhuang 050011, China
| | - Hongwei Fan
- Department of Gynecology, Fourth Hospital, Hebei Medical University, Shijiazhuang 050011, China
| | - Xi Kang
- Department of Surgery, Fourth Hospital, Hebei Medical University, Shijiazhuang 050011, China
| | - Yuntao Hao
- Department of Gynecology, Fourth Hospital, Hebei Medical University, Shijiazhuang 050011, China
| | - Na Wang
- Department of Molecular Biology, Fourth Hospital, Hebei Medical University, Shijiazhuang 050011, China
| | - Hui Zheng
- Nanjing Personal Oncology Biotechnology Co., Ltd., Nanjing, Jiangsu 211103, China
| | - Yan Li
- Department of Molecular Biology, Fourth Hospital, Hebei Medical University, Shijiazhuang 050011, China
| | - Shan Kang
- Department of Gynecology, Fourth Hospital, Hebei Medical University, Shijiazhuang 050011, China
| |
Collapse
|
3
|
Naser AA, Miyazaki T, Wang J, Takabayashi S, Pachoensuk T, Tokumoto T. MC4R mutant mice develop ovarian teratomas. Sci Rep 2021; 11:3483. [PMID: 33568756 PMCID: PMC7876032 DOI: 10.1038/s41598-021-83001-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Accepted: 01/19/2021] [Indexed: 01/05/2023] Open
Abstract
Teratomas in mice, composed of different tissue types, are derived from primordial germ cells (PGCs) in the foetal gonads. The strongest candidate gene in the testicular teratoma locus (Ter) responsible for testicular teratoma formation was identified as mutation in Dnd1, Dnd1R178*. However, the phenotype of mice with a mutated Dnd1 gene was germ cell loss. This suggests that other genes are involved in teratoma formation. Testicular teratomas can also be induced experimentally (experimentally testicular teratomas: ETTs) in 129/Sv mice by transplanting E12.5 foetal testes into adult testes. Previously, we mapped the ett1 locus, which is the locus responsible for ETT formation on chromosome 18. By exome sequence analysis of the 129 and LTXBJ (LT) strains, we identified a missense mutation in the melanocortin 4 receptor (MC4R) gene among 8 genes in the ett1 region. The missense mutation causes a substitution of glycine 25 by serine. Thus, this gene is a candidate for ETT formation. We established the LT-ett1 congenic strain, which introduced the locus responsible for ETT formation genetically into the genomes of a testicular teratoma non-susceptible strain. In this study, we crossed LT-ett1 and a previously established LT-Ter strain to establish the double congenic strain LT-Ter-ett1. Also, we established a strain with a point mutation in the MC4R gene of the LT strain by genome editing, LT-MC4RG25S. Furthermore, double genetically modified strain LT-Ter-MC4RG25S was established to address the relation between Ter and MC4R. Surprisingly, highly developed ovarian teratomas (OTs), instead of testicular teratomas, appeared not only in the LT-Ter-MC4RG25S and LT-MC4RG25S strains but also in the LT-ett1 and LT-Ter-ett1 strains. The incidence of OT formation was high in double genetically modified strains. The results demonstrated that MC4R is one of the genes responsible for OT formation. It was suggested that the effect of the missense mutation in MC4R on teratoma formation was promoted by abnormal germ cell formation by the mutation in DND1.
Collapse
Affiliation(s)
- Abdullah An Naser
- Integrated Bioscience Section, Graduate School of Science and Technology, National University Corporation Shizuoka University, Ohya 836, Suruga-ku, Shizuoka, 422-8529, Japan
| | - Takehiro Miyazaki
- Integrated Bioscience Section, Graduate School of Science and Technology, National University Corporation Shizuoka University, Ohya 836, Suruga-ku, Shizuoka, 422-8529, Japan
- Department of Molecular Genetics, Graduate School of Medicine, Kyoto University, Yoshida Konoe, Sakyo, Kyoto, 606-8501, Japan
| | - Jun Wang
- Department of Bioscience, Faculty of Science, Shizuoka University, Shizuoka, 422, Japan
| | - Shuji Takabayashi
- Laboratory Animal Facilities & Services, Preeminent Medical Photonics Education & Research Center, Hamamatsu University School of Medicine, 1-20-1, Handayama, Higashi-ku, Hamamatsu, Shizuoka, 431-3192, Japan
| | - Theeranukul Pachoensuk
- Integrated Bioscience Section, Graduate School of Science and Technology, National University Corporation Shizuoka University, Ohya 836, Suruga-ku, Shizuoka, 422-8529, Japan
| | - Toshinobu Tokumoto
- Integrated Bioscience Section, Graduate School of Science and Technology, National University Corporation Shizuoka University, Ohya 836, Suruga-ku, Shizuoka, 422-8529, Japan.
- Department of Bioscience, Faculty of Science, Shizuoka University, Shizuoka, 422, Japan.
| |
Collapse
|
4
|
Molecular characterization and expression analysis of foxo3l in response to exogenous hormones in black rockfish (Sebastes schlegelii). Gene 2020; 753:144777. [PMID: 32428695 DOI: 10.1016/j.gene.2020.144777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 05/07/2020] [Accepted: 05/14/2020] [Indexed: 11/22/2022]
Abstract
As a crucial member of the Forkhead Box family, class O (FoxO) plays an essential role in growth, cell differentiation, metabolism, immunization, and apoptosis. Meanwhile, FoxO3 is the primary regulator and effective inhibitor of primordial follicle activation. In this study, seven foxo genes were identified in black rockfish (Sebastes schlegelii), including two foxo1 genes (foxo1a, foxo1b), two foxo3 genes (foxo3, foxo3l), one foxo4 gene, and two foxo6 genes (foxo6a, foxo6b). foxo3l was derived from teleost-specific whole-genome duplication events. Evaluation of tissue expression pattern revealed that foxo3l displayed sexually dimorphic expression with a high level in the ovary and spatial expression only in the cytoplasm of follicle cells and oocytes. When the ovaries were stimulated by estrogen and gonadotropin, foxo3l expression was remarkably reduced, and the effect of androgen was completely different. We considered that foxo3l lost its ability to inhibit follicular precocity because of mass ovulation by hormone stimulation, resulting in its decreased expression. Such evidence indicated that foxo3l is an important regulator of reproduction-related functions in black rockfish. This study provides new insights into foxo3l genes for further functional research in teleost.
Collapse
|
5
|
Bertozzi TM, Ferguson-Smith AC. Metastable epialleles and their contribution to epigenetic inheritance in mammals. Semin Cell Dev Biol 2020; 97:93-105. [PMID: 31551132 DOI: 10.1016/j.semcdb.2019.08.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 08/15/2019] [Accepted: 08/20/2019] [Indexed: 02/02/2023]
Abstract
Many epigenetic differences between individuals are driven by genetic variation. Mammalian metastable epialleles are unusual in that they show variable DNA methylation states between genetically identical individuals. The occurrence of such states across generations has resulted in their consideration by many as strong evidence for epigenetic inheritance in mammals, with the classic Avy and AxinFu mouse models - each products of repeat element insertions - being the most widely accepted examples. Equally, there has been interest in exploring their use as epigenetic biosensors given their susceptibility to environmental compromise. Here we review the classic murine metastable epialleles as well as more recently identified candidates, with the aim of providing a more holistic understanding of their biology. We consider the extent to which epigenetic inheritance occurs at metastable epialleles and explore the limited mechanistic insights into the establishment of their variable epigenetic states. We discuss their environmental modulation and their potential relevance in genome regulation. In light of recent whole-genome screens for novel metastable epialleles, we point out the need to reassess their biological relevance in multi-generational studies and we highlight their value as a model to study repeat element silencing as well as the mechanisms and consequences of mammalian epigenetic stochasticity.
Collapse
Affiliation(s)
- Tessa M Bertozzi
- Department of Genetics, University of Cambridge, Cambridge CB2 3EH, UK
| | | |
Collapse
|
6
|
Gu W, Mochizuki K, Otsuka K, Hamada R, Takehara A, Matsui Y. Dnd1-mediated epigenetic control of teratoma formation in mouse. Biol Open 2018; 7:bio032318. [PMID: 29378702 PMCID: PMC5829515 DOI: 10.1242/bio.032318] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Accepted: 01/02/2018] [Indexed: 01/16/2023] Open
Abstract
Spontaneous testicular teratoma develops from primordial germ cells (PGCs) in embryos; however, the molecular mechanisms underlying teratoma formation are not fully understood. Mutation of the dead-end 1 (Dnd1) gene, which encodes an RNA-binding protein, drastically enhances teratoma formation in the 129/Sv mouse strain. To elucidate the mechanism of Dnd1 mutation-induced teratoma formation, we focused on histone H3 lysine 27 (H3K27) trimethylation (me3), and found that the levels of H3K27me3 and its responsible methyltransferase, enhancer of zeste homolog 2 (Ezh2), were decreased in the teratoma-forming cells of Dnd1 mutant embryos. We also showed that Dnd1 suppressed miR-26a-mediated inhibition of Ezh2 expression, and that Dnd1 deficiency resulted in decreased H3K27me3 of a cell-cycle regulator gene, Ccnd1 In addition, Ezh2 expression or Ccnd1 deficiency repressed the reprogramming of PGCs into pluripotent stem cells, which mimicked the conversion of embryonic germ cells into teratoma-forming cells. These results revealed an epigenetic molecular linkage between Dnd1 and the suppression of testicular teratoma formation.
Collapse
Affiliation(s)
- Wei Gu
- Cell Resource Center for Biomedical Research, Institute of Development, Aging and Cancer (IDAC), Tohoku University, 4-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
- Laboratory of Germ Cell Development, Graduate School of Life Sciences, Tohoku University, 2-1-1 Katahira, Aoba-ku, Sendai, Miyagi 980-8577, Japan
| | - Kentaro Mochizuki
- Cell Resource Center for Biomedical Research, Institute of Development, Aging and Cancer (IDAC), Tohoku University, 4-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
- Laboratory of Germ Cell Development, Graduate School of Life Sciences, Tohoku University, 2-1-1 Katahira, Aoba-ku, Sendai, Miyagi 980-8577, Japan
- The Japan Agency for Medical Research and Development-Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo 100-0004, Japan
| | - Kei Otsuka
- Cell Resource Center for Biomedical Research, Institute of Development, Aging and Cancer (IDAC), Tohoku University, 4-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| | - Ryohei Hamada
- Cell Resource Center for Biomedical Research, Institute of Development, Aging and Cancer (IDAC), Tohoku University, 4-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| | - Asuka Takehara
- Cell Resource Center for Biomedical Research, Institute of Development, Aging and Cancer (IDAC), Tohoku University, 4-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
- The Japan Agency for Medical Research and Development-Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo 100-0004, Japan
| | - Yasuhisa Matsui
- Cell Resource Center for Biomedical Research, Institute of Development, Aging and Cancer (IDAC), Tohoku University, 4-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
- Laboratory of Germ Cell Development, Graduate School of Life Sciences, Tohoku University, 2-1-1 Katahira, Aoba-ku, Sendai, Miyagi 980-8577, Japan
- The Japan Agency for Medical Research and Development-Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo 100-0004, Japan
- Center for Regulatory Epigenome and Diseases, Tohoku University School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| |
Collapse
|
7
|
Investigation of the clinical features of lower uterine segment carcinoma: association with advanced stage disease and indication of poorer prognosis. Arch Gynecol Obstet 2017; 297:193-198. [PMID: 29116461 DOI: 10.1007/s00404-017-4576-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 10/10/2017] [Indexed: 12/16/2022]
Abstract
PURPOSE We retrospectively analyzed the differential clinical features and prognosis of endometrial carcinomas arising from the lower uterus, which are reported to have a poorer prognosis than those arising from the upper uterus. METHODS 246 patients with endometrial carcinoma who underwent surgery were entered as subjects. RESULTS Twenty-three were classified as having lower uterine segment carcinomas (LUSC); the remaining 223 were upper uterine segment carcinomas (UUSC). LUSC cases were associated with a more advanced FIGO stage than UUSC (p < 0.001). Deep myometrial invasion and lymph node metastasis were more common in LUSC than in UUSC (p = 0.006 and p < 0.001, respectively). LUSC cases demonstrated significantly shorter overall survival (OS) and progression-free survival (PFS) than UUSC (p = 0.02 and p < 0.001, respectively). Multivariate cox proportional hazards analysis demonstrated that the hazard ratio for LUSC was 1.769 for OS and 3.479 for PFS. For endometrial carcinoma survival, FIGO stage and histological type were extracted as independent variables. CONCLUSIONS LUSC is a high-risk indicator for poorer prognosis for endometrial carcinoma because it is associated with more advanced stage disease, deep myometrial invasion and lymph node metastasis, and indicates a significantly worsened PFS probability. Our analysis concludes that LUSC is FIGO stage-dependent and an important factor for OS.
Collapse
|
8
|
Khawajkie Y, Buckett W, Nguyen NMP, Mechtouf N, Ao A, Arseneau J, Slim R. Recurrent triploid digynic conceptions and mature ovarian teratomas: Are they different manifestations of the same genetic defect? Genes Chromosomes Cancer 2017; 56:832-840. [DOI: 10.1002/gcc.22484] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 07/17/2017] [Accepted: 07/17/2017] [Indexed: 11/05/2022] Open
Affiliation(s)
| | - William Buckett
- Department of Obstetrics and Gynecology; McGill University; Montreal Canada
| | - Ngoc Minh Phuong Nguyen
- Department of Obstetrics and Gynecology; McGill University; Montreal Canada
- Department of Human Genetics; McGill University; Montreal Canada
| | - Nawel Mechtouf
- Department of Human Genetics; McGill University; Montreal Canada
| | - Asangla Ao
- Department of Obstetrics and Gynecology; McGill University; Montreal Canada
- Department of Human Genetics; McGill University; Montreal Canada
| | - Jocelyne Arseneau
- Department of Pathology; McGill University; Montreal Canada
- Gynecologic Oncology Unit, McGill University Health Centre; Montreal Canada
| | - Rima Slim
- Division of Experimental Medicine; McGill University; Montreal Canada
- Department of Obstetrics and Gynecology; McGill University; Montreal Canada
- Department of Human Genetics; McGill University; Montreal Canada
| |
Collapse
|
9
|
Viatte S, Lee JC, Fu B, Espéli M, Lunt M, De Wolf JNE, Wheeler L, Reynolds JA, Castelino M, Symmons DPM, Lyons PA, Barton A, Smith KGC. Association Between Genetic Variation in FOXO3 and Reductions in Inflammation and Disease Activity in Inflammatory Polyarthritis. Arthritis Rheumatol 2017; 68:2629-2636. [PMID: 27214848 PMCID: PMC5091631 DOI: 10.1002/art.39760] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 05/12/2016] [Indexed: 12/25/2022]
Abstract
Objective Genetic variation in FOXO3 (tagged by rs12212067) has been associated with a milder course of rheumatoid arthritis (RA) and shown to limit monocyte‐driven inflammation through a transforming growth factor β1–dependent pathway. This genetic association, however, has not been consistently observed in other RA cohorts. We sought to clarify the contribution of FOXO3 to prognosis in RA by combining detailed analysis of nonradiographic disease severity measures with an in vivo model of arthritis. Methods Collagen‐induced arthritis, the most commonly used mouse model of RA, was used to assess how Foxo3 contributes to arthritis severity. Using clinical, serologic, and biochemical methods, the arthritis that developed in mice carrying a loss‐of‐function mutation in Foxo3 was compared with that which occurred in littermate controls. The association of rs12212067 with nonradiographic measures of RA severity, including the C‐reactive protein level, the swollen joint count, the tender joint count, the Disease Activity Score in 28 joints, and the Health Assessment Questionnaire score, were modeled longitudinally in a large prospective cohort of patients with early RA. Results Loss of Foxo3 function resulted in more severe arthritis in vivo (both clinically and histologically) and was associated with higher titers of anticollagen antibodies and interleukin‐6 in the blood. Similarly, rs12212067 (a single‐nucleotide polymorphism that increases FOXO3 transcription) was associated with reduced inflammation, both biochemically and clinically, and with lower RA activity scores. Conclusion Consistent with its known role in restraining inflammatory responses, FOXO3 limits the severity of in vivo arthritis and, through genetic variation that increases its transcription, is associated with reduced inflammation and disease activity in RA patients, effects that result in less radiographic damage.
Collapse
Affiliation(s)
| | - James C Lee
- University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Bo Fu
- University of Manchester, Manchester, UK, and University College London, London, UK
| | - Marion Espéli
- UMR 996, Inflammation, Chemokines, and Immunopathology, INSERM, Université Paris-Sud, Université Paris-Saclay, Clamart, France
| | - Mark Lunt
- University of Manchester, Manchester, UK
| | | | | | | | | | - Deborah P M Symmons
- NIHR Manchester Musculoskeletal Biomedical Research Unit, Central Manchester NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - Paul A Lyons
- University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Anne Barton
- NIHR Manchester Musculoskeletal Biomedical Research Unit, Central Manchester NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK.
| | | |
Collapse
|
10
|
FOXO3 is differentially required for CD8 + T-cell death during tolerance versus immunity. Immunol Cell Biol 2016; 94:895-899. [PMID: 27323690 DOI: 10.1038/icb.2016.53] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2015] [Revised: 05/24/2016] [Accepted: 05/25/2016] [Indexed: 12/19/2022]
Abstract
Peripheral tolerance mechanisms limit autoimmunity by constitutively eliminating self-reactive CD8+ T cells from the periphery in a process called deletion. Previous work has demonstrated that this deletion process is mediated by BIM-dependent apoptotic death due to transcriptional induction of the Bim gene. Currently, the transcriptional pathways responsible for Bim induction during peripheral deletion remain unclear. We speculated that the transcriptional regulator FOXO3 may induce BIM-dependent death during peripheral deletion, as it has been implicated in Bim induction and cell death during effector CD8+ T-cell differentiation. Despite observing less Akt-dependent inactivation of FOXO transcription factors in tolerised cells relative to effector cells, we demonstrate that FOXO3-deficient CD8+ T cells induce Bim and die normally during peripheral deletion. These data thus demonstrate that BIM-dependent death during CD8+ T-cell deletion is FOXO3 independent. Furthermore, these data provide the first evidence that the pathways responsible for Bim induction and cell death during effector differentiation versus tolerance of CD8+ T cells are molecularly distinct.
Collapse
|
11
|
Gonadal development and germ cell tumors in mouse and humans. Semin Cell Dev Biol 2015; 45:114-23. [DOI: 10.1016/j.semcdb.2015.10.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 10/01/2015] [Indexed: 12/12/2022]
|
12
|
Yang QE, Nagaoka SI, Gwost I, Hunt PA, Oatley JM. Inactivation of Retinoblastoma Protein (Rb1) in the Oocyte: Evidence That Dysregulated Follicle Growth Drives Ovarian Teratoma Formation in Mice. PLoS Genet 2015; 11:e1005355. [PMID: 26176933 PMCID: PMC4503754 DOI: 10.1371/journal.pgen.1005355] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 06/12/2015] [Indexed: 02/02/2023] Open
Abstract
The origin of most ovarian tumors is undefined. Here, we report development of a novel mouse model in which conditional inactivation of the tumor suppressor gene Rb1 in oocytes leads to the formation of ovarian teratomas (OTs). While parthenogenetically activated ooctyes are a known source of OT in some mutant mouse models, enhanced parthenogenetic propensity in vitro was not observed for Rb1-deficient oocytes. Further analyses revealed that follicle recruitment and growth is disrupted in ovaries of mice with conditional inactivation of Rb1, leading to abnormal accumulation of secondary/preantral follicles. These findings underpin the concept that miscues between the germ cell and somatic compartments cause premature oocyte activation and the formation of OTs. Furthermore, these results suggest that defects in folliculogenesis and a permissive genetic background are sufficient to drive OT development, even in the absence of enhanced parthenogenetic activation. Thus, we have discovered a novel role of Rb1 in regulating the entry of primordial oocytes into the pool of growing follicles and signaling between the oocyte and granulosa cells during the protracted process of oocyte growth. Our findings, coupled with data from studies of other OT models, suggest that defects in the coordinated regulation between growth of the oocyte and somatic components in follicles are an underlying cause of OT formation.
Collapse
Affiliation(s)
- Qi-En Yang
- Center for Reproductive Biology, School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, Washington, United States of America
| | - So I. Nagaoka
- Center for Reproductive Biology, School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, Washington, United States of America
| | - Ivy Gwost
- Center for Reproductive Biology, School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, Washington, United States of America
| | - Patricia A. Hunt
- Center for Reproductive Biology, School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, Washington, United States of America
| | - Jon M. Oatley
- Center for Reproductive Biology, School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, Washington, United States of America
- * E-mail:
| |
Collapse
|
13
|
The first mouse mutants of D14Abb1e (Fam208a) show that it is critical for early development. Mamm Genome 2014; 25:293-303. [PMID: 24781204 PMCID: PMC4105592 DOI: 10.1007/s00335-014-9516-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2013] [Accepted: 04/01/2014] [Indexed: 01/09/2023]
Abstract
An ENU mutagenesis screen to identify novel epigenetic modifiers was established in mice carrying a multi-copy GFP transgene, which is expressed in a variegated manner in erythrocytes and is highly sensitive to epigenetic silencing. The screen has produced mouse mutants of both known modifiers of epigenetic state, such as Dnmt1 and Smarca5, and novel modifiers, such as Smchd1 and Rlf. Here we report two mouse lines generated from the screen, MommeD6 and MommeD20, with point mutations in D14Abb1e. These are the first mouse mutants of D14Abb1e (alsoknownasFam208a), a gene about which little is known. Heterozygous intercrosses show that homozygous mutants from both the MommeD6 and MommeD20 lines are not viable beyond gastrulation, demonstrating an important role for D14Abb1e in development. We demonstrate that haploinsufficiency for D14Abb1e effects transgene expression at the RNA level. Analysis of the predicted D14Abb1e protein sequence reveals that it contains putative nuclear localisation signals and a domain of unknown function, DUF3715. Our studies reveal that D14Abb1e is localised to the nucleus and is expressed in skin and testes.
Collapse
|
14
|
Abstract
Much of what we know about the role of epigenetics in the determination of phenotype has come from studies of inbred mice. Some unusual expression patterns arising from endogenous and transgenic murine alleles, such as the Agouti coat color alleles, have allowed the study of variegation, variable expressivity, transgenerational epigenetic inheritance, parent-of-origin effects, and position effects. These phenomena have taught us much about gene silencing and the probabilistic nature of epigenetic processes. Based on some of these alleles, large-scale mutagenesis screens have broadened our knowledge of epigenetic control by identifying and characterizing novel genes involved in these processes.
Collapse
Affiliation(s)
- Marnie Blewitt
- Walter and Eliza Hall Institute, Melbourne, 3052 Victoria, Australia
| | | |
Collapse
|
15
|
Lee J, Espéli M, Anderson C, Linterman M, Pocock J, Williams N, Roberts R, Viatte S, Fu B, Peshu N, Hien T, Phu N, Wesley E, Edwards C, Ahmad T, Mansfield J, Gearry R, Dunstan S, Williams T, Barton A, Vinuesa C, Parkes M, Lyons PA, Smith KG. Human SNP links differential outcomes in inflammatory and infectious disease to a FOXO3-regulated pathway. Cell 2013; 155:57-69. [PMID: 24035192 PMCID: PMC3790457 DOI: 10.1016/j.cell.2013.08.034] [Citation(s) in RCA: 181] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2013] [Revised: 07/08/2013] [Accepted: 08/19/2013] [Indexed: 12/19/2022]
Abstract
The clinical course and eventual outcome, or prognosis, of complex diseases varies enormously between affected individuals. This variability critically determines the impact a disease has on a patient’s life but is very poorly understood. Here, we exploit existing genome-wide association study data to gain insight into the role of genetics in prognosis. We identify a noncoding polymorphism in FOXO3A (rs12212067: T > G) at which the minor (G) allele, despite not being associated with disease susceptibility, is associated with a milder course of Crohn’s disease and rheumatoid arthritis and with increased risk of severe malaria. Minor allele carriage is shown to limit inflammatory responses in monocytes via a FOXO3-driven pathway, which through TGFβ1 reduces production of proinflammatory cytokines, including TNFα, and increases production of anti-inflammatory cytokines, including IL-10. Thus, we uncover a shared genetic contribution to prognosis in distinct diseases that operates via a FOXO3-driven pathway modulating inflammatory responses. PaperClip
Reanalysis of GWAS data identifies a SNP associated with outcome in Crohn’s disease This SNP modulates inflammatory responses in monocytes via a FOXO3-driven pathway The mild disease-associated allele reduces TNFα and increases IL-10 production Prognosis in RA and malaria (also TNFα-related diseases) is also linked to this SNP
Collapse
Affiliation(s)
- James C. Lee
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0XY, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke’s Hospital, Cambridge CB2 0QQ, UK
| | - Marion Espéli
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0XY, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke’s Hospital, Cambridge CB2 0QQ, UK
| | - Carl A. Anderson
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SA, UK
| | - Michelle A. Linterman
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0XY, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke’s Hospital, Cambridge CB2 0QQ, UK
| | - Joanna M. Pocock
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0XY, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke’s Hospital, Cambridge CB2 0QQ, UK
| | - Naomi J. Williams
- Department of Pathogens and Immunity, John Curtin School of Medical Research, Australian National University, Canberra ACT 2601, Australia
| | - Rebecca Roberts
- University of Otago, Department of Medicine, Christchurch 8011, New Zealand
| | - Sebastien Viatte
- Arthritis Research UK Epidemiology Unit, Manchester Academic Health Science Center, University of Manchester, Manchester M13 9PT, UK
| | - Bo Fu
- Arthritis Research UK Epidemiology Unit, Manchester Academic Health Science Center, University of Manchester, Manchester M13 9PT, UK
- Centre for Biostatistics, Institute of Population Health, University of Manchester, Manchester M13 9PL, UK
| | - Norbert Peshu
- Kenya Medical Research Institute/Wellcome Trust Research Program, Centre for Geographic Medicine Research, Kilifi P.O. Box 230-80108, Kenya
| | - Tran Tinh Hien
- Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Program, Hospital for Tropical Diseases, District 5 Ho Chi Minh City, Vietnam
| | - Nguyen Hoan Phu
- The Hospital for Tropical Diseases, District 5 Ho Chi Minh City, Vietnam
| | - Emma Wesley
- Peninsula College of Medicine and Dentistry, University of Exeter, Exeter EX2 5DW, UK
| | - Cathryn Edwards
- Department of Gastroenterology, Torbay Hospital, Torquay TQ2 7AA, UK
| | - Tariq Ahmad
- Peninsula College of Medicine and Dentistry, University of Exeter, Exeter EX2 5DW, UK
| | - John C. Mansfield
- Institute of Genetic Medicine, Newcastle University, Newcastle NE1 3BZ, UK
| | - Richard Gearry
- University of Otago, Department of Medicine, Christchurch 8011, New Zealand
| | - Sarah Dunstan
- Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Program, Hospital for Tropical Diseases, District 5 Ho Chi Minh City, Vietnam
- Centre for Tropical Medicine, Nuffield Department of Clinical Medicine, University of Oxford, Oxford OX3 7LJ, UK
| | - Thomas N. Williams
- Kenya Medical Research Institute/Wellcome Trust Research Program, Centre for Geographic Medicine Research, Kilifi P.O. Box 230-80108, Kenya
- Department of Medicine, Imperial College, London SW7 2AZ, UK
| | - Anne Barton
- Arthritis Research UK Epidemiology Unit, Manchester Academic Health Science Center, University of Manchester, Manchester M13 9PT, UK
| | - Carola G. Vinuesa
- Department of Pathogens and Immunity, John Curtin School of Medical Research, Australian National University, Canberra ACT 2601, Australia
| | | | - Miles Parkes
- Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke’s Hospital, Cambridge CB2 0QQ, UK
| | - Paul A. Lyons
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0XY, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke’s Hospital, Cambridge CB2 0QQ, UK
| | - Kenneth G.C. Smith
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0XY, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke’s Hospital, Cambridge CB2 0QQ, UK
- Corresponding author
| |
Collapse
|
16
|
Youngson NA, Epp T, Roberts AR, Daxinger L, Ashe A, Huang E, Lester KL, Harten SK, Kay GF, Cox T, Matthews JM, Chong S, Whitelaw E. No evidence for cumulative effects in a Dnmt3b hypomorph across multiple generations. Mamm Genome 2013; 24:206-17. [PMID: 23636699 DOI: 10.1007/s00335-013-9451-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Accepted: 03/05/2013] [Indexed: 12/31/2022]
Abstract
Observations of inherited phenotypes that cannot be explained solely through genetic inheritance are increasing. Evidence points to transmission of non-DNA molecules in the gamete as mediators of the phenotypes. However, in most cases it is unclear what the molecules are, with DNA methylation, chromatin proteins, and small RNAs being the most prominent candidates. From a screen to generate novel mouse mutants of genes involved in epigenetic reprogramming, we produced a DNA methyltransferase 3b allele that is missing exon 13. Mice that are homozygous for the mutant allele have smaller stature and reduced viability, with particularly high levels of female post-natal death. Reduced DNA methylation was also detected at telocentric repeats and the X-linked Hprt gene. However, none of the abnormal phenotypes or DNA methylation changes worsened with multiple generations of homozygous mutant inbreeding. This suggests that in our model the abnormalities are reset each generation and the processes of transgenerational epigenetic reprogramming are effective in preventing their inheritance.
Collapse
Affiliation(s)
- Neil A Youngson
- Queensland Institute of Medical Research, Herston, Brisbane, QLD 4006, Australia.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Ovine forkhead box class O 3 (FOXO3) gene variation and its association with lifespan. Mol Biol Rep 2013; 40:3829-34. [PMID: 23299403 DOI: 10.1007/s11033-012-2461-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2012] [Accepted: 12/18/2012] [Indexed: 10/27/2022]
Abstract
FOXO3 is a member of the FOXO (forkhead box class O) transcription factor family and has roles in cell cycle control, apoptosis, neural and hematopoietic cell differentiation and DNA repair among other functions. Several human studies provide evidence for an association between aging, longevity and variation in FOXO3. Recently variation has been identified in exon 2 of ovine FOXO3. This exon encodes the C-terminus of the DNA-binding domain and a transcription activation domain that is the key regulator of transcriptional activity in target genes. The association of genetic variation in exon 2 with lifespan was investigated in 1,732 New Zealand (NZ) sheep. Of the seven haplotypes detected, the presence of the D haplotype, which codes for an amino acid substitution (a conserved methionine residue is replaced by a valine) at residue 407, is associated with a decrease of 0.39 years in mean age (P = 0.034). Significant differences in mean age were also detected between genotypes containing D (AD: 4.7 ± 0.21; BD: 4.7 ± 0.25) and genotypes that did not contain D (AA: 5.1 ± 0.14; AB: 4.9 ± 0.18; BB: 5.3 ± 0.25). Other genotypes were rare in the sheep investigated and were not analyzed. This suggests that genetic variation in ovine FOXO3 influences the lifespan of sheep either directly or indirectly by impacting on factors that lead to reduced productivity and increased likelihood of culling.
Collapse
|
18
|
Balakrishnan A, Chaillet JR. Role of the inositol polyphosphate-4-phosphatase type II Inpp4b in the generation of ovarian teratomas. Dev Biol 2012; 373:118-29. [PMID: 23078915 DOI: 10.1016/j.ydbio.2012.10.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2012] [Revised: 09/14/2012] [Accepted: 10/09/2012] [Indexed: 10/27/2022]
Abstract
Teratomas are a unique class of tumors composed of ecto-, meso- and endodermal tissues, all foreign to the site of origin. In humans, the most common teratoma is the ovarian teratoma. Not much is known about the molecular and genetic etiologies of these tumors. Female carriers of the Tgkd transgene are highly susceptible to developing teratomas. Ovaries of Tgkd/+ hemizygous female mice exhibit defects in luteinization, with numerous corpora lutea, some of which contain central trapped, fully-grown oocytes. Genetically, Tgkd teratomas originate from mature oocytes that have completed meiosis I, suggesting that Tgkd teratomas originate from these trapped oocytes. The insertion of Tgkd 3' of the Inpp4b gene is associated with decreased expression of Inpp4b and changes in intracellular PI3 Kinase/AKT signaling in follicular granulosa cells. Because Inpp4b is not expressed in fully-grown wild-type or Tgkd oocytes, these findings suggest that hyperactivation of the PI3K/AKT pathway caused by the decrease in INPP4B in granulosa cells promotes an ovarian environment defective in folliculogenesis and conducive to teratoma formation.
Collapse
Affiliation(s)
- Ashwini Balakrishnan
- Department of Microbiology and Molecular Genetics, Magee-Womens Research Institute, University of Pittsburgh, Pittsburgh, PA 15208, USA
| | | |
Collapse
|
19
|
Abstract
Modifier genes are an integral part of the genetic landscape in both humans and experimental organisms, but have been less well explored in mammals than other systems. A growing number of modifier genes in mouse models of disease nonetheless illustrate the potential for novel findings, while new technical advances promise many more to come. Modifier genes in mouse models include induced mutations and spontaneous or wild-derived variations captured in inbred strains. Identification of modifiers among wild-derived variants in particular should detect disease modifiers that have been shaped by selection and might therefore be compatible with high fitness and function. Here we review selected examples and argue that modifier genes derived from natural variation may provide a bias for nodes in genetic networks that have greater intrinsic plasticity and whose therapeutic manipulation may therefore be more resilient to side effects than conventional targets.
Collapse
Affiliation(s)
- Bruce A Hamilton
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, California, United States of America.
| | | |
Collapse
|
20
|
Roberts AR, Blewitt ME, Youngson NA, Whitelaw E, Chong S. Reduced dosage of the modifiers of epigenetic reprogramming Dnmt1, Dnmt3L, SmcHD1 and Foxo3a has no detectable effect on mouse telomere length in vivo. Chromosoma 2011; 120:377-85. [PMID: 21553025 PMCID: PMC3140923 DOI: 10.1007/s00412-011-0318-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2011] [Accepted: 03/18/2011] [Indexed: 12/18/2022]
Abstract
Studies carried out in cultured cells have implicated modifiers of epigenetic reprogramming in the regulation of telomere length, reporting elongation in cells that were null for DNA methyltransferase DNA methyltransferase 1 (Dnmt1), both de novo DNA methyltransferases, Dnmt3a and Dnmt3b or various histone methyltransferases. To investigate this further, we assayed telomere length in whole embryos or adult tissue from mice carrying mutations in four different modifiers of epigenetic reprogramming: Dnmt1, DNA methyltransferase 3-like, structural maintenance of chromosomes hinge domain containing 1, and forkhead box O3a. Terminal restriction fragment analysis was used to compare telomere length in homozygous mutants, heterozygous mutants and wild-type littermates. Contrary to expectation, we did not detect overall lengthening in the mutants, raising questions about the role of epigenetic processes in telomere length in vivo.
Collapse
Affiliation(s)
- Amity R Roberts
- Epigenetics Laboratory, Queensland Institute of Medical Research, Herston, QLD, Australia
| | | | | | | | | |
Collapse
|