1
|
Wu Y, Su K, Zhang Y, Liang L, Wang F, Chen S, Gao L, Zheng Q, Li C, Su Y, Mao Y, Zhu S, Chai C, Lan Q, Zhai M, Jin X, Zhang J, Xu X, Zhang Y, Gao Y, Huang H. A spatiotemporal transcriptomic atlas of mouse placentation. Cell Discov 2024; 10:110. [PMID: 39438452 PMCID: PMC11496649 DOI: 10.1038/s41421-024-00740-6] [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: 01/29/2024] [Accepted: 09/10/2024] [Indexed: 10/25/2024] Open
Abstract
The placenta, a temporary but essential organ for gestational support, undergoes intricate morphological and functional transformations throughout gestation. However, the spatiotemporal patterns of gene expression underlying placentation remain poorly understood. Utilizing Stereo-seq, we constructed a Mouse Placentation Spatiotemporal Transcriptomic Atlas (MPSTA) spanning from embryonic day (E) 7.5 to E14.5, which includes the transcriptomes of large trophoblast cells that were not captured in previous single-cell atlases. We defined four distinct strata of the ectoplacental cone, an early heterogeneous trophectoderm structure, and elucidated the spatial trajectory of trophoblast differentiation during early postimplantation stages before E9.5. Focusing on the labyrinth region, the interface of nutrient exchange in the mouse placenta, our spatiotemporal ligand-receptor interaction analysis unveiled pivotal modulators essential for trophoblast development and placental angiogenesis. We also found that paternally expressed genes are exclusively enriched in the placenta rather than in the decidual regions, including a cluster of genes enriched in endothelial cells that may function in placental angiogenesis. At the invasion front, we identified interface-specific transcription factor regulons, such as Atf3, Jun, Junb, Stat6, Mxd1, Maff, Fos, and Irf7, involved in gestational maintenance. Additionally, we revealed that maternal high-fat diet exposure preferentially affects this interface, exacerbating inflammatory responses and disrupting angiogenic homeostasis. Collectively, our findings furnish a comprehensive, spatially resolved atlas that offers valuable insights and benchmarks for future explorations into placental morphogenesis and pathology.
Collapse
Affiliation(s)
- Yanting Wu
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China.
- Key Laboratory of Reproductive Genetics (Ministry of Education), Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
- Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences (No. 2019RU056), Shanghai, China.
- Shanghai Key Laboratory of Reproduction and Development, Shanghai, China.
| | - Kaizhen Su
- Key Laboratory of Reproductive Genetics (Ministry of Education), Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ying Zhang
- BGI Research, Shenzhen, Guangdong, China
- Shanxi Medical University - BGI Collaborative Center for Future Medicine, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Langchao Liang
- BGI Research, Qingdao, Shandong, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Fei Wang
- BGI Research, Shenzhen, Guangdong, China
| | - Siyue Chen
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China
| | - Ling Gao
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China
| | - Qiutong Zheng
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China
| | - Cheng Li
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China
| | - Yunfei Su
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China
| | - Yiting Mao
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China
| | - Simeng Zhu
- Department of Cardiology, Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chaochao Chai
- BGI Research, Qingdao, Shandong, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Qing Lan
- BGI Research, Shenzhen, Guangdong, China
| | - Man Zhai
- BGI Research, Shenzhen, Guangdong, China
| | - Xin Jin
- BGI Research, Shenzhen, Guangdong, China
| | - Jinglan Zhang
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China
- Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences (No. 2019RU056), Shanghai, China
- Shanghai Key Laboratory of Reproduction and Development, Shanghai, China
| | - Xun Xu
- BGI Research, Shenzhen, Guangdong, China
- Guangdong Provincial Key Laboratory of Genome Read and Write, Shenzhen, Guangdong, China
| | - Yu Zhang
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China.
| | - Ya Gao
- BGI Research, Shenzhen, Guangdong, China.
- Shanxi Medical University - BGI Collaborative Center for Future Medicine, Shanxi Medical University, Taiyuan, Shanxi, China.
- Shenzhen Engineering Laboratory for Birth Defects Screening, BGI Research, Shenzhen, Guangdong, China.
| | - Hefeng Huang
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China.
- Key Laboratory of Reproductive Genetics (Ministry of Education), Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
- Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences (No. 2019RU056), Shanghai, China.
- Shanghai Key Laboratory of Reproduction and Development, Shanghai, China.
- Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
2
|
Wang J, Zhou X, Han T, Zhang H. Epigenetic signatures of trophoblast lineage and their biological functions. Cells Dev 2024; 179:203934. [PMID: 38942294 DOI: 10.1016/j.cdev.2024.203934] [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: 11/15/2023] [Revised: 03/13/2024] [Accepted: 06/13/2024] [Indexed: 06/30/2024]
Abstract
Trophoblasts play a crucial role in embryo implantation and in interacting with the maternal uterus. The trophoblast lineage develops into a substantial part of the placenta, a temporary extra-embryonic organ, capable of undergoing distinctive epigenetic events during development. The critical role of trophoblast-specific epigenetic signatures in regulating placental development has become known, significantly advancing our understanding of trophoblast identity and lineage development. Scientific efforts are revealing how trophoblast-specific epigenetic signatures mediate stage-specific gene regulatory programming during the development of the trophoblast lineage. These epigenetic signatures have a significant impact on blastocyst formation, placental development, as well as the growth and survival of embryos and fetuses. In evolution, DNA hypomethylation in the trophoblast lineage is conserved, and there is a significant disparity in the control of epigenetic dynamics and the landscape of genomic imprinting. Scientists have used murine and human multipotent trophoblast cells as in vitro models to recapitulate the essential epigenetic processes of placental development. Here, we review the epigenetic signatures of the trophoblast lineage and their biological functions to enhance our understanding of placental evolution, development, and function.
Collapse
Affiliation(s)
- Jianqi Wang
- Chongqing Key Laboratory of Maternal and Fetal Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Xiaobo Zhou
- Chongqing Key Laboratory of Maternal and Fetal Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Reproductive Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Tingli Han
- Chongqing Key Laboratory of Maternal and Fetal Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Joint International Research Laboratory of Reproduction and Development of Chinese Ministry of Education, Chongqing Medical University, 400016, China; The Center for Reproductive Medicine, Obstetrics and Gynecology Department, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China.
| | - Hua Zhang
- Chongqing Key Laboratory of Maternal and Fetal Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Joint International Research Laboratory of Reproduction and Development of Chinese Ministry of Education, Chongqing Medical University, 400016, China.
| |
Collapse
|
3
|
Zheng Y, Zha X, Zhang B, Elsabagh M, Wang H, Wang M, Zhang H. The interaction of ER stress and autophagy in trophoblasts: navigating pregnancy outcome†. Biol Reprod 2024; 111:292-311. [PMID: 38678504 DOI: 10.1093/biolre/ioae066] [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: 11/27/2023] [Revised: 04/12/2024] [Accepted: 04/22/2024] [Indexed: 05/01/2024] Open
Abstract
The endoplasmic reticulum is a complex and dynamic organelle that initiates unfolded protein response and endoplasmic reticulum stress in response to the accumulation of unfolded or misfolded proteins within its lumen. Autophagy is a paramount intracellular degradation system that facilitates the transportation of proteins, cytoplasmic components, and organelles to lysosomes for degradation and recycling. Preeclampsia and intrauterine growth retardation are two common complications of pregnancy associated with abnormal trophoblast differentiation and placental dysfunctions and have a major impact on fetal development and maternal health. The intricate interplay between endoplasmic reticulum stress, and autophagy and their impact on pregnancy outcomes, through mediating trophoblast differentiation and placental development, has been highlighted in various reports. Autophagy controls trophoblast regulation through a variety of gene expressions and signaling pathways while excessive endoplasmic reticulum stress triggers downstream apoptotic signaling, culminating in trophoblast apoptosis. This comprehensive review delves into the intricacies of placental development and explores the underlying mechanisms of preeclampsia and intrauterine growth retardation. In addition, this review will elucidate the molecular mechanisms of endoplasmic reticulum stress and autophagy, both individually and in their interplay, in mediating placental development and trophoblast differentiation, particularly highlighting their roles in preeclampsia and intrauterine growth retardation development. This research seeks to the interplay between endoplasmic reticulum stress and impaired autophagy in the placental trophoderm, offering novel insights into their contribution to pregnancy complications.
Collapse
Affiliation(s)
- Yi Zheng
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou, People's Repubic of China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, People's Republic of China
| | - Xia Zha
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou, People's Repubic of China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, People's Republic of China
| | - Bei Zhang
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou, People's Repubic of China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, People's Republic of China
| | - Mabrouk Elsabagh
- Department of Animal Production and Technology, Faculty of Agricultural Sciences and Technologies, Niğde Ömer Halisdemir University, Nigde, Turkey
- Department of Nutrition and Clinical Nutrition, Faculty of Veterinary Medicine, Kafrelsheikh University, KafrelSheikh, Egypt
| | - Hongrong Wang
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou, People's Repubic of China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, People's Republic of China
| | - Mengzhi Wang
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou, People's Repubic of China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, People's Republic of China
- State Key Laboratory of Sheep Genetic Improvement and Healthy Production, Xinjiang Academy of Agricultural Reclamation Science, Shihezi, P. R. China
| | - Hao Zhang
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou, People's Repubic of China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, People's Republic of China
| |
Collapse
|
4
|
Ramesh NA, Box AM, Buttitta LA. Post-eclosion growth in the Drosophila Ejaculatory Duct is driven by Juvenile Hormone signaling and is essential for male fertility. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.12.607650. [PMID: 39185157 PMCID: PMC11343125 DOI: 10.1101/2024.08.12.607650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
The Drosophila Ejaculatory duct (ED) is a secretory tissue of the somatic male reproductive system. The ED is involved in the secretion of seminal fluid components and ED-specific antimicrobial peptides that aid in fertility and the female post-mating response. The ED is composed of secretory epithelial cells surrounded by a layer of innervated contractile muscle. The ED grows in young adult males during the first 24h post-eclosion, but the cell cycle status of the ED secretory cells and the role of post-eclosion ED growth have been unexplored. Here, we show that secretory cells of the adult Drosophila ED undergo variant cell cycles lacking mitosis called the endocycle, that lead to an increase in the cell and organ size of the ED post eclosion. The cells largely exit the endocycle by day 3 of adulthood, when the growth of the ED ceases, resulting in a tissue containing cells of ploidies ranging from 8C-32C. The size of the ED directly correlates with the ploidy of the secretory cells, with additional ectopic endocycles increasing organ size. When endoreplication is compromised in ED secretory cells, it leads to reduced organ size, reduced protein synthesis and compromised fertility. We provide evidence that the growth and endocycling in the young adult male ED is dependent on Juvenile hormone (JH) signaling and we suggest that hormone-induced early adult endocycling is required for optimal fertility and function of the ED tissue. We propose to use the ED as a post-mitotic tissue model to study the role of polyploidy in regulating secretory tissue growth and function.
Collapse
Affiliation(s)
- Navyashree A. Ramesh
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Allison M. Box
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Laura A. Buttitta
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
5
|
Mann KE, Panfilio KA. Tissue-Level Integration Overrides Gradations of Differentiating Cell Identity in Beetle Extraembryonic Tissue. Cells 2024; 13:1211. [PMID: 39056793 PMCID: PMC11274815 DOI: 10.3390/cells13141211] [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: 04/12/2024] [Revised: 07/10/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
During animal embryogenesis, one of the earliest specification events distinguishes extraembryonic (EE) from embryonic tissue fates: the serosa in the case of the insects. While it is well established that the homeodomain transcription factor Zen1 is the critical determinant of the serosa, the subsequent realization of this tissue's identity has not been investigated. Here, we examine serosal differentiation in the beetle Tribolium castaneum based on the quantification of morphological and morphogenetic features, comparing embryos from a Tc-zen1 RNAi dilution series, where complete knockdown results in amnion-only EE tissue identity. We assess features including cell density, tissue boundary morphology, and nuclear size as dynamic readouts for progressive tissue maturation. While some features exhibit an all-or-nothing outcome, other key features show dose-dependent phenotypic responses with trait-specific thresholds. Collectively, these findings provide nuance beyond the known status of Tc-Zen1 as a selector gene for serosal tissue patterning. Overall, our approach illustrates how the analysis of tissue maturation dynamics from live imaging extends but also challenges interpretations based on gene expression data, refining our understanding of tissue identity and when it is achieved.
Collapse
Affiliation(s)
- Katie E. Mann
- School of Life Sciences, University of Warwick, Coventry CV4 7AL, UK
| | - Kristen A. Panfilio
- School of Life Sciences, University of Warwick, Coventry CV4 7AL, UK
- Department of Molecular Genetics, Institute of Biology, University of Hohenheim, Garbenstr. 30, 70599 Stuttgart, Germany
| |
Collapse
|
6
|
Darmasaputra GS, van Rijnberk LM, Galli M. Functional consequences of somatic polyploidy in development. Development 2024; 151:dev202392. [PMID: 38415794 PMCID: PMC10946441 DOI: 10.1242/dev.202392] [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] [Indexed: 02/29/2024]
Abstract
Polyploid cells contain multiple genome copies and arise in many animal tissues as a regulated part of development. However, polyploid cells can also arise due to cell division failure, DNA damage or tissue damage. Although polyploidization is crucial for the integrity and function of many tissues, the cellular and tissue-wide consequences of polyploidy can be very diverse. Nonetheless, many polyploid cell types and tissues share a remarkable similarity in function, providing important information about the possible contribution of polyploidy to cell and tissue function. Here, we review studies on polyploid cells in development, underlining parallel functions between different polyploid cell types, as well as differences between developmentally-programmed and stress-induced polyploidy.
Collapse
Affiliation(s)
- Gabriella S. Darmasaputra
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center Utrecht, Uppsalalaan 8, 3584 CT, Utrecht, the Netherlands
| | - Lotte M. van Rijnberk
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center Utrecht, Uppsalalaan 8, 3584 CT, Utrecht, the Netherlands
| | - Matilde Galli
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center Utrecht, Uppsalalaan 8, 3584 CT, Utrecht, the Netherlands
| |
Collapse
|
7
|
Kinkade JA, Seetharam AS, Sachdev S, Bivens NJ, Phinney BS, Grigorean G, Roberts RM, Tuteja G, Rosenfeld CS. Extracellular vesicles from mouse trophoblast cells: Effects on neural progenitor cells and potential participants in the placenta-brain axis†. Biol Reprod 2024; 110:310-328. [PMID: 37883444 PMCID: PMC10873279 DOI: 10.1093/biolre/ioad146] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 10/12/2023] [Accepted: 10/21/2023] [Indexed: 10/28/2023] Open
Abstract
The fetal brain of the mouse is thought to be dependent upon the placenta as a source of serotonin (5-hydroxytryptamine; 5-HT) and other factors. How factors reach the developing brain remains uncertain but are postulated here to be part of the cargo carried by placental extracellular vesicles (EV). We have analyzed the protein, catecholamine, and small RNA content of EV from mouse trophoblast stem cells (TSC) and TSC differentiated into parietal trophoblast giant cells (pTGC), potential primary purveyors of 5-HT. Current studies examined how exposure of mouse neural progenitor cells (NPC) to EV from either TSC or pTGC affect their transcriptome profiles. The EV from trophoblast cells contained relatively high amounts of 5-HT, as well as dopamine and norepinephrine, but there were no significant differences between EV derived from pTGC and from TSC. Content of miRNA and small nucleolar (sno)RNA, however, did differ according to EV source, and snoRNA were upregulated in EV from pTGC. The primary inferred targets of the microRNA (miRNA) from both pTGC and TSC were mRNA enriched in the fetal brain. NPC readily internalized EV, leading to changes in their transcriptome profiles. Transcripts regulated were mainly ones enriched in neural tissues. The transcripts in EV-treated NPC that demonstrated a likely complementarity with miRNA in EV were mainly up- rather than downregulated, with functions linked to neuronal processes. Our results are consistent with placenta-derived EV providing direct support for fetal brain development and being an integral part of the placenta-brain axis.
Collapse
Affiliation(s)
- Jessica A Kinkade
- Biomedical Sciences, University of Missouri, Columbia, MO, USA
- Division of Animal Sciences, University of Missouri, Columbia, MO, USA
| | - Arun S Seetharam
- Department of Ecology, Evolution and Organismal Biology, Iowa State University, Ames, IA, USA
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, IA, USA
| | - Shrikesh Sachdev
- Division of Animal Sciences, University of Missouri, Columbia, MO, USA
| | - Nathan J Bivens
- Genomics Technology Core Facility, University of Missouri, Columbia, MO, USA
| | - Brett S Phinney
- Proteomics Core UC Davis Genome Center, University of California, Davis, CA, USA
| | - Gabriela Grigorean
- Proteomics Core UC Davis Genome Center, University of California, Davis, CA, USA
| | - R Michael Roberts
- Division of Animal Sciences, University of Missouri, Columbia, MO, USA
| | - Geetu Tuteja
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, IA, USA
| | - Cheryl S Rosenfeld
- Biomedical Sciences, University of Missouri, Columbia, MO, USA
- MU Institute of Data Science and Informatics, University of Missouri, Columbia, MO, USA
- Genetics Area Program, University of Missouri, Columbia, MO, USA
- Thompson Center for Autism and Neurobehavioral Disorders, University of Missouri, Columbia, MO, USA
| |
Collapse
|
8
|
Rakoczy J, Watson ED. Folate-depletion alters mouse trophoblast stem cell regulation in vitro. Placenta 2023; 144:64-68. [PMID: 37995442 DOI: 10.1016/j.placenta.2023.11.009] [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: 09/29/2023] [Revised: 11/09/2023] [Accepted: 11/15/2023] [Indexed: 11/25/2023]
Abstract
Maternal folate deficiency increases risk of congenital malformations, yet its effect on placenta development is unclear. Here, we investigated how folate-depleted culture medium affects the developmental potential of mouse trophoblast stem cells (TSCs). When cultured in stem cell conditions, TSC viability was unaffected by folate depletion, but ectopic differentiation of trophoblast cell subtypes occurred. When cultured in conditions that promote differentiation, folate-depleted TSCs were driven towards a syncytiotrophoblast cell fate potentially at the expense of other lineages. Additionally, trophoblast giant cell nuclei were small implicating folate in the regulation of endoreduplication. Therefore, dietary folate intake likely promotes trophoblast development.
Collapse
Affiliation(s)
- Joanna Rakoczy
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, CB2 3EG, UK
| | - Erica D Watson
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, CB2 3EG, UK.
| |
Collapse
|
9
|
Weier JF, Ferlatte C, Baumgartner A, Nguyen HN, Weier BA, Weier HUG. Analysis of human invasive cytotrophoblasts demonstrates mosaic aneuploidy. PLoS One 2023; 18:e0284317. [PMID: 37478076 PMCID: PMC10361533 DOI: 10.1371/journal.pone.0284317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 03/28/2023] [Indexed: 07/23/2023] Open
Abstract
A total of 24 chromosome-specific fluorescence in situ hybridization probes for interphase nucleus analysis were developed to determine the chromosomal content of individual human invasive cytotrophoblasts derived from in vitro cultured assays. At least 75% of invasive cytotrophoblasts were hyperdiploid and the total number of chromosomes ranged from 47 to 61. The results also demonstrated that these hyperdiploid invasive cytotrophoblasts showed significant heterogeneity. The most copy number gains were observed for chromosomes 13, 14, 15, 19, 21, and 22 with average copy number greater than 2.3. A parallel study using primary invasive cytotrophoblasts also showed a similar trend of copy number changes. Conclusively, 24-chromosome analysis of human non-proliferating cytotrophoblasts (interphase nuclei) was achieved. Hyperdiploidy and chromosomal heterogeneity without endoduplication in invasive cytotrophoblasts may suggest a selective advantage for invasion and short lifespan during normal placental development.
Collapse
Affiliation(s)
- Jingly F Weier
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California (U.C.), San Francisco, California, United States of America
- Life Sciences Division, U.C. E.O. Lawrence Berkeley National Laboratory, Berkeley, California, United States of America
| | - Christy Ferlatte
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California (U.C.), San Francisco, California, United States of America
| | - Adolf Baumgartner
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California (U.C.), San Francisco, California, United States of America
- Life Sciences Division, U.C. E.O. Lawrence Berkeley National Laboratory, Berkeley, California, United States of America
| | - Ha Nam Nguyen
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California (U.C.), San Francisco, California, United States of America
| | - Beatrice A Weier
- Golden State Dermatology, Walnut Creek, California, United States of America
| | - Heinz-Ulrich G Weier
- Life Sciences Division, U.C. E.O. Lawrence Berkeley National Laboratory, Berkeley, California, United States of America
| |
Collapse
|
10
|
Singh VP, Hassan H, Deng F, Tsuchiya D, McKinney S, Ferro K, Gerton JL. Myc promotes polyploidy in murine trophoblast cells and suppresses senescence. Development 2023; 150:dev201581. [PMID: 37278344 PMCID: PMC10309589 DOI: 10.1242/dev.201581] [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: 01/03/2023] [Accepted: 04/27/2023] [Indexed: 05/10/2023]
Abstract
The placenta is essential for reproductive success. The murine placenta includes polyploid giant cells that are crucial for its function. Polyploidy occurs broadly in nature but its regulators and significance in the placenta are unknown. We have discovered that many murine placental cell types are polyploid and have identified factors that license polyploidy using single-cell RNA sequencing. Myc is a key regulator of polyploidy and placental development, and is required for multiple rounds of DNA replication, likely via endocycles, in trophoblast giant cells. Furthermore, MYC supports the expression of DNA replication and nucleotide biosynthesis genes along with ribosomal RNA. Increased DNA damage and senescence occur in trophoblast giant cells without Myc, accompanied by senescence in the neighboring maternal decidua. These data reveal Myc is essential for polyploidy to support normal placental development, thereby preventing premature senescence. Our study, combined with available literature, suggests that Myc is an evolutionarily conserved regulator of polyploidy.
Collapse
Affiliation(s)
| | - Huzaifa Hassan
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Fengyan Deng
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Dai Tsuchiya
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Sean McKinney
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Kevin Ferro
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Jennifer L. Gerton
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| |
Collapse
|
11
|
A differentiation roadmap of murine placentation at single-cell resolution. Cell Discov 2023; 9:30. [PMID: 36928215 PMCID: PMC10020559 DOI: 10.1038/s41421-022-00513-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 12/25/2022] [Indexed: 03/18/2023] Open
Abstract
The placenta is one of the most important yet least understood organs. Due to the limitations of conventional research approaches, we are still far from a comprehensive understanding of mouse placentation, especially regarding the differentiation of trophoblast lineages at the early developmental stage. To decipher cell compositions and developmental processes, we systematically profile the single-cell transcriptomes of trophoblast cells from extraembryonic tissues (embryonic day 7.5 (E7.5) and E8.5) and placentae (E9.5-E14.5) at one-day intervals. We identify distinct trophoblast cell types during mouse placentation, including unreported progenitor cells and intermediate precursor cells. An updated differentiation roadmap of mouse trophoblast lineages is presented following systematic transcriptome analyses. Based on transcriptomic regulatory network inference, we specify transcription factors responsible for the regulation of dynamic developmental processes during lineage diversification. We map lineage differentiation trajectories and find that sinusoid trophoblast giant cells arise from the subpopulation of ectoplacental cone cells. We provide a comprehensive single-cell data resource to shed light on future mechanistic studies of the gene regulatory networks governing hemochorial placentation.
Collapse
|
12
|
Favaron PO, Carter AM. The trophoblast giant cells of cricetid rodents. Front Cell Dev Biol 2023; 10:1097854. [PMID: 36726588 PMCID: PMC9885145 DOI: 10.3389/fcell.2022.1097854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 12/16/2022] [Indexed: 01/19/2023] Open
Abstract
Giant cells are a prominent feature of placentation in cricetid rodents. Once thought to be maternal in origin, they are now known to be trophoblast giant cells (TGCs). The large size of cricetid TGCs and their nuclei reflects a high degree of polyploidy. While some TGCs are found at fixed locations, others migrate throughout the placenta and deep into the uterus where they sometimes survive postpartum. Herein, we review the distribution of TGCs in the placenta of cricetids, including our own data from the New World subfamily Sigmodontinae, and attempt a comparison between the TGCs of cricetid and murid rodents. In both families, parietal TGCs are found in the parietal yolk sac and as a layer between the junctional zone and decidua. In cricetids alone, large numbers of TGCs, likely from the same lineage, accumulate at the edge of the placental disk. Common to murids and cricetids is a haemotrichorial placental barrier where the maternal-facing layer consists of cytotrophoblasts characterized as sinusoidal TGCs. The maternal channels of the labyrinth are supplied by trophoblast-lined canals. Whereas in the mouse these are lined largely by canal TGCs, in cricetids canal TGCs are interspersed with syncytiotrophoblast. Transformation of the uterine spiral arteries occurs in both murids and cricetids and spiral artery TGCs line segments of the arteries that have lost their endothelium and smooth muscle. Since polyploidization of TGCs can amplify selective genomic regions required for specific functions, we argue that the TGCs of cricetids deserve further study and suggest avenues for future research.
Collapse
Affiliation(s)
- Phelipe O. Favaron
- Department of General Biology, Biological Sciences Center, State University of Londrina, Paraná, Brazil
| | - Anthony M. Carter
- Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
13
|
Fischer U, Meese E. Gene Amplification in Tumor Cells: Developed De Novo or Adopted from Stem Cells. Cells 2022; 12:cells12010148. [PMID: 36611942 PMCID: PMC9818554 DOI: 10.3390/cells12010148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/16/2022] [Accepted: 12/28/2022] [Indexed: 12/31/2022] Open
Abstract
Gene amplifications have been known for several decades as physiological processes in amphibian and flies, e.g., during eggshell development in Drosophila and as part of pathological processes in humans, specifically in tumors and drug-resistant cells. The long-held belief that a physiological gene amplification does not occur in humans was, however, fundamental questioned by findings that showed gene amplification in human stem cells. We hypothesis that the physiological and the pathological, i.e., tumor associated processes of gene amplification share at their beginning the same underlying mechanism. Re-replication was reported both in the context of tumor related genome instability and during restricted time windows in Drosophila development causing the known developmental gene amplification in Drosophila. There is also growing evidence that gene amplification and re-replication were present in human stem cells. It appears likely that stem cells utilize a re-replication mechanism that has been developed early in evolution as a powerful tool to increase gene copy numbers very efficiently. Here, we show that, several decades ago, there was already evidence of gene amplification in non-tumor mammalian cells, but that was not recognized at the time and interpreted accordingly. We give an overview on gene amplifications during normal mammalian development, the possible mechanism that enable gene amplification and hypothesize how tumors adopted this capability for gene amplification.
Collapse
|
14
|
Panfilio KA, Chuva de Sousa Lopes SM. The extended analogy of extraembryonic development in insects and amniotes. Philos Trans R Soc Lond B Biol Sci 2022; 377:20210268. [PMID: 36252225 PMCID: PMC9574626 DOI: 10.1098/rstb.2021.0268] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 02/14/2022] [Indexed: 12/22/2022] Open
Abstract
It is fascinating that the amnion and serosa/chorion, two extraembryonic (EE) tissues that are characteristic of the amniote vertebrates (mammals, birds and reptiles), have also independently evolved in insects. In this review, we offer the first detailed, macroevolutionary comparison of EE development and tissue biology across these animal groups. Some commonalities represent independent solutions to shared challenges for protecting the embryo (environmental assaults, risk of pathogens) and supporting its development, including clear links between cellular properties (e.g. polyploidy) and physiological function. Further parallels encompass developmental features such as the early segregation of the serosa/chorion compared to later, progressive differentiation of the amnion and formation of the amniotic cavity from serosal-amniotic folds as a widespread morphogenetic mode across species. We also discuss common developmental roles for orthologous transcription factors and BMP signalling in EE tissues of amniotes and insects, and between EE and cardiac tissues, supported by our exploration of new resources for global and tissue-specific gene expression. This highlights the degree to which general developmental principles and protective tissue features can be deduced from each of these animal groups, emphasizing the value of broad comparative studies to reveal subtle developmental strategies and answer questions that are common across species. This article is part of the theme issue 'Extraembryonic tissues: exploring concepts, definitions and functions across the animal kingdom'.
Collapse
Affiliation(s)
| | - Susana M. Chuva de Sousa Lopes
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 ZC Leiden, The Netherlands
- Department for Reproductive Medicine, Ghent University Hospital, 9000 Ghent, Belgium
| |
Collapse
|
15
|
Buss G, Stratton MB, Milenkovic L, Stearns T. Postmitotic centriole disengagement and maturation leads to centrosome amplification in polyploid trophoblast giant cells. Mol Biol Cell 2022; 33:ar118. [PMID: 36001376 PMCID: PMC9634975 DOI: 10.1091/mbc.e22-05-0182] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
DNA replication is normally coupled with centriole duplication in the cell cycle. Trophoblast giant cells (TGCs) of the placenta undergo endocycles resulting in polyploidy but their centriole state is not known. We used a cell culture model for TGC differentiation to examine centriole and centrosome number and properties. Before differentiation, trophoblast stem cells (TSCs) have either two centrioles before duplication or four centrioles after. We find that the average nuclear area increases approximately eight-fold over differentiation, but most TGCs do not have more than four centrioles. However, these centrioles become disengaged, acquire centrosome proteins, and can nucleate microtubules. In addition, some TGCs undergo further duplication and disengagement of centrioles, resulting in substantially higher numbers. Live imaging revealed that disengagement and separation are centriole autonomous and can occur asynchronously. Centriole amplification, when present, occurs by the standard mechanism of one centriole generating one procentriole. PLK4 inhibition blocks centriole formation in differentiating TGCs but does not affect endocycle progression. In summary, centrioles in TGC endocycles undergo disengagement and conversion to centrosomes. This increases centrosome number but to a limited extent compared with DNA reduplication.
Collapse
Affiliation(s)
- Garrison Buss
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305
| | | | | | - Tim Stearns
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305,Department of Biology, Stanford University, Stanford, CA 94305,*Address correspondence to: Tim Stearns ()
| |
Collapse
|
16
|
A Temporary Pause in the Replication Licensing Restriction Leads to Rereplication during Early Human Cell Differentiation. Cells 2022; 11:cells11061060. [PMID: 35326512 PMCID: PMC8946923 DOI: 10.3390/cells11061060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 03/18/2022] [Accepted: 03/20/2022] [Indexed: 12/10/2022] Open
Abstract
Gene amplifications in amphibians and flies are known to occur during development and have been well characterized, unlike in mammalian cells, where they are predominantly investigated as an attribute of tumors. Recently, we first described gene amplifications in human and mouse neural stem cells, myoblasts, and mesenchymal stem cells during differentiation. The mechanism leading to gene amplifications in amphibians and flies depends on endocycles and multiple origin-firings. So far, there is no knowledge about a comparable mechanism in normal human cells. Here, we describe rereplication during the early myotube differentiation of human skeletal myoblast cells, using fiber combing and pulse-treatment with EdU (5′-Ethynyl-2′-deoxyuridine)/CldU (5-Chlor-2′-deoxyuridine) and IdU (5-Iodo-2′-deoxyuridine)/CldU. We found rereplication during a restricted time window between 2 h and 8 h after differentiation induction. Rereplication was detected in cells simultaneously with the amplification of the MDM2 gene. Our findings support rereplication as a mechanism enabling gene amplification in normal human cells.
Collapse
|
17
|
Richards L, Das S, Nordman JT. Rif1-Dependent Control of Replication Timing. Genes (Basel) 2022; 13:genes13030550. [PMID: 35328102 PMCID: PMC8955891 DOI: 10.3390/genes13030550] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/15/2022] [Accepted: 03/18/2022] [Indexed: 02/01/2023] Open
Abstract
Successful duplication of the genome requires the accurate replication of billions of base pairs of DNA within a relatively short time frame. Failure to accurately replicate the genome results in genomic instability and a host of diseases. To faithfully and rapidly replicate the genome, DNA replication must be tightly regulated and coordinated with many other nuclear processes. These regulations, however, must also be flexible as replication kinetics can change through development and differentiation. Exactly how DNA replication is regulated and how this regulation changes through development is an active field of research. One aspect of genome duplication where much remains to be discovered is replication timing (RT), which dictates when each segment of the genome is replicated during S phase. All organisms display some level of RT, yet the precise mechanisms that govern RT remain are not fully understood. The study of Rif1, a protein that actively regulates RT from yeast to humans, provides a key to unlock the underlying molecular mechanisms controlling RT. The paradigm for Rif1 function is to delay helicase activation within certain regions of the genome, causing these regions to replicate late in S phase. Many questions, however, remain about the intricacies of Rif1 function. Here, we review the current models for the activity of Rif1 with the goal of trying to understand how Rif1 functions to establish the RT program.
Collapse
|
18
|
Bischof C, Mirtschink P, Yuan T, Wu M, Zhu C, Kaur J, Pham MD, Gonzalez-Gonoggia S, Hammer M, Rogg EM, Sharma R, Bottermann K, Gercken B, Hagag E, Berthonneche C, Sossalla S, Stehr SN, Maxeiner J, Duda MA, Latreille M, Zamboni N, Martelli F, Pedrazzini T, Dimmeler S, Krishnan J. Mitochondrial-cell cycle cross-talk drives endoreplication in heart disease. Sci Transl Med 2021; 13:eabi7964. [PMID: 34878823 DOI: 10.1126/scitranslmed.abi7964] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Corinne Bischof
- MRC Clinical Sciences Centre, Imperial College London, London W12 0NN, UK.,Institute of Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Peter Mirtschink
- Institute of Clinical Chemistry and Laboratory Medicine, Department of Clinical Pathobiochemistry, University Hospital Dresden, Fetscherstasse 74, 01307 Dresden, Germany
| | - Ting Yuan
- Institute of Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany.,Department of Medicine III, Division of Cardiology/Nephrology/Angiology, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Meiqian Wu
- Institute of Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany.,Department of Medicine III, Division of Cardiology/Nephrology/Angiology, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Chaonan Zhu
- Institute of Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany.,Department of Medicine III, Division of Cardiology/Nephrology/Angiology, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Jaskiran Kaur
- Institute of Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany.,Department of Medicine III, Division of Cardiology/Nephrology/Angiology, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Minh Duc Pham
- Institute of Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany.,Genome Biologics, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | | | - Marie Hammer
- Institute of Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Eva-Maria Rogg
- Institute of Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Rahul Sharma
- Institute of Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Katharina Bottermann
- Institute of Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Bettina Gercken
- Institute of Clinical Chemistry and Laboratory Medicine, Department of Clinical Pathobiochemistry, University Hospital Dresden, Fetscherstasse 74, 01307 Dresden, Germany
| | - Eman Hagag
- Institute of Clinical Chemistry and Laboratory Medicine, Department of Clinical Pathobiochemistry, University Hospital Dresden, Fetscherstasse 74, 01307 Dresden, Germany
| | - Corinne Berthonneche
- Cardiovascular Assessment Facility, University of Lausanne, CHUV, CH-1011 Lausanne, Switzerland
| | - Samuel Sossalla
- Department of Internal Medicine II, University Medical Center Regensburg, 93053 Regensburg, Germany.,Klinik für Kardiologie und Pneumologie, Georg-August-Universität Goettingen, DZHK (German Centre for Cardiovascular Research), Robert-Koch Str. 40, D-37075 Goettingen, Germany
| | - Sebastian N Stehr
- Department of Anesthesiology and Critical Care Medicine, University Hospital Leipzig, Liebigstrasse 20, D-04103 Leipzig, Germany
| | - Joachim Maxeiner
- Genome Biologics, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Maria Anna Duda
- Genome Biologics, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Mathieu Latreille
- MRC Clinical Sciences Centre, Imperial College London, London W12 0NN, UK
| | - Nicola Zamboni
- Institute of Molecular Systems Biology, ETH Zurich, Zurich 8093, Switzerland
| | - Fabio Martelli
- Molecular Cardiology Laboratory, IRCCS-Policlinico San Donato, 20097, San Donato Milanese, Milan, Italy
| | - Thierry Pedrazzini
- Department of Medicine, University of Lausanne Medical School, CHUV, MP14-220, 1011 Lausanne, Switzerland
| | - Stefanie Dimmeler
- Institute of Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany.,DZHK Partner Site RheinMain, Mainz, Germany.,Cardio-Pulmonary Institute, Giessen, Germany
| | - Jaya Krishnan
- MRC Clinical Sciences Centre, Imperial College London, London W12 0NN, UK.,Institute of Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany.,Department of Medicine III, Division of Cardiology/Nephrology/Angiology, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany.,Cardio-Pulmonary Institute, Giessen, Germany
| |
Collapse
|
19
|
Edwards MM, Zuccaro MV, Sagi I, Ding Q, Vershkov D, Benvenisty N, Egli D, Koren A. Delayed DNA replication in haploid human embryonic stem cells. Genome Res 2021; 31:2155-2169. [PMID: 34810218 PMCID: PMC8647822 DOI: 10.1101/gr.275953.121] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 10/20/2021] [Indexed: 11/25/2022]
Abstract
Haploid human embryonic stem cells (ESCs) provide a powerful genetic system but diploidize at high rates. We hypothesized that diploidization results from aberrant DNA replication. To test this, we profiled DNA replication timing in isogenic haploid and diploid ESCs. The greatest difference was the earlier replication of the X Chromosome in haploids, consistent with the lack of X-Chromosome inactivation. We also identified 21 autosomal regions that had delayed replication in haploids, extending beyond the normal S phase and into G2/M. Haploid-delays comprised a unique set of quiescent genomic regions that are also underreplicated in polyploid placental cells. The same delays were observed in female ESCs with two active X Chromosomes, suggesting that increased X-Chromosome dosage may cause delayed autosomal replication. We propose that incomplete replication at the onset of mitosis could prevent cell division and result in re-entry into the cell cycle and whole genome duplication.
Collapse
Affiliation(s)
- Matthew M Edwards
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York 14853, USA
| | - Michael V Zuccaro
- Department of Pediatrics and Naomi Berrie Diabetes Center, Columbia University, New York, New York 10032, USA
- Columbia University Stem Cell Initiative, New York, New York 10032, USA
| | - Ido Sagi
- The Azrieli Center for Stem Cells and Genetic Research, Department of Genetics, Silberman Institute of Life Sciences, The Hebrew University, Jerusalem 91904, Israel
| | - Qiliang Ding
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York 14853, USA
| | - Dan Vershkov
- The Azrieli Center for Stem Cells and Genetic Research, Department of Genetics, Silberman Institute of Life Sciences, The Hebrew University, Jerusalem 91904, Israel
| | - Nissim Benvenisty
- The Azrieli Center for Stem Cells and Genetic Research, Department of Genetics, Silberman Institute of Life Sciences, The Hebrew University, Jerusalem 91904, Israel
| | - Dieter Egli
- Department of Pediatrics and Naomi Berrie Diabetes Center, Columbia University, New York, New York 10032, USA
- Columbia University Stem Cell Initiative, New York, New York 10032, USA
| | - Amnon Koren
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York 14853, USA
| |
Collapse
|
20
|
Morey R, Farah O, Kallol S, Requena DF, Meads M, Moretto-Zita M, Soncin F, Laurent LC, Parast MM. Transcriptomic Drivers of Differentiation, Maturation, and Polyploidy in Human Extravillous Trophoblast. Front Cell Dev Biol 2021; 9:702046. [PMID: 34540826 PMCID: PMC8446284 DOI: 10.3389/fcell.2021.702046] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 07/27/2021] [Indexed: 12/20/2022] Open
Abstract
During pregnancy, conceptus-derived extravillous trophoblast (EVT) invades the endomyometrium, anchors the placenta to the maternal uterus, and remodels the spiral arteries in order to establish maternal blood supply to the fetoplacental unit. Recent reports have described early gestation EVT as polyploid and senescent. Here, we extend these reports by performing comprehensive profiling of both the genomic organization and transcriptome of first trimester and term EVT. We define pathways and gene regulatory networks involved in both initial differentiation and maturation of this important trophoblast lineage at the maternal-fetal interface. Our results suggest that like first trimester EVT, term EVT undergoes senescence and endoreduplication, is primarily tetraploid, and lacks high rates of copy number variations. Additionally, we have highlighted senescence and polyploidy-related genes, pathways, networks, and transcription factors that appeared to be important in normal EVT differentiation and maturation and validated a key role for the unfolded protein response in this context.
Collapse
Affiliation(s)
- Robert Morey
- Department of Pathology, University of California, San Diego, La Jolla, CA, United States.,Department of Obstetrics, Gynecology, and Reproductive Sciences, Division of Maternal-Fetal Medicine, University of California, San Diego, La Jolla, CA, United States.,Sanford Consortium for Regenerative Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Omar Farah
- Department of Pathology, University of California, San Diego, La Jolla, CA, United States.,Sanford Consortium for Regenerative Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Sampada Kallol
- Department of Pathology, University of California, San Diego, La Jolla, CA, United States.,Sanford Consortium for Regenerative Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Daniela F Requena
- Department of Pathology, University of California, San Diego, La Jolla, CA, United States.,Sanford Consortium for Regenerative Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Morgan Meads
- Department of Pathology, University of California, San Diego, La Jolla, CA, United States.,Sanford Consortium for Regenerative Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Matteo Moretto-Zita
- Department of Pathology, University of California, San Diego, La Jolla, CA, United States.,Sanford Consortium for Regenerative Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Francesca Soncin
- Department of Pathology, University of California, San Diego, La Jolla, CA, United States.,Sanford Consortium for Regenerative Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Louise C Laurent
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Division of Maternal-Fetal Medicine, University of California, San Diego, La Jolla, CA, United States.,Sanford Consortium for Regenerative Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Mana M Parast
- Department of Pathology, University of California, San Diego, La Jolla, CA, United States.,Sanford Consortium for Regenerative Medicine, University of California, San Diego, La Jolla, CA, United States
| |
Collapse
|
21
|
Human HAND1 Inhibits the Conversion of Cholesterol to Steroids in Trophoblasts. J Genet Genomics 2021; 49:350-363. [PMID: 34391879 DOI: 10.1016/j.jgg.2021.07.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 07/11/2021] [Accepted: 07/17/2021] [Indexed: 11/24/2022]
Abstract
Steroidogenesis from cholesterol in placental trophoblasts is fundamentally involved in the establishment and maintenance of pregnancy. The transcription factor gene Heart And Neural crest Derivatives expressed 1 (Hand1) promotes differentiation of mouse trophoblast giant cells. However, the role of HAND1 in human trophoblasts remains unknown. Here, we report that HAND1 inhibits human trophoblastic progesterone (P4) and estradiol (E2) from cholesterol through down-regulation of the expression of steroidogenic enzymes including aromatase, P450 cholesterol side-chain cleavage enzyme (P450scc) and 3β-hydroxysteroid dehydrogenase type 1 (3β-HSD1). Mechanically, while HAND1 inhibits transcription of aromatase by directly binding to aromatase gene promoter, it restrains transcription of P450scc by up-regulation of the methylation status of P450scc gene promoter through its binding to ALKBH1, a demethylase. Unlike aromatase and P450scc, HAND1 decreases 3β-HSD1 mRNA levels by reduction of its RNA stability through binding to and subsequent destabilization of protein HuR. Finally, HAND1 suppresses circulating P4 and E2 levels derived from JEG-3 xenograft, and attenuates uterine response to P4 and E2. Thus, our results uncover a hitherto uncharacterized role of HAND1 in regulation of cholesterol metabolism in human trophoblasts, which may help pinpoint the underlying mechanisms involved in supporting the development and physiological function of the human placenta.
Collapse
|
22
|
Morimoto H, Ueno M, Tanabe H, Kono T, Ogawa H. Progesterone depletion results in Lamin B1 loss and induction of cell death in mouse trophoblast giant cells. PLoS One 2021; 16:e0254674. [PMID: 34260661 PMCID: PMC8279370 DOI: 10.1371/journal.pone.0254674] [Citation(s) in RCA: 2] [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: 04/30/2021] [Accepted: 06/30/2021] [Indexed: 01/04/2023] Open
Abstract
Trophoblast giant cells (TGCs), a mouse trophoblast subtype, have large amounts of cytoplasm and high ploidy levels via endocycles. The diverse functions and gene expression profiles of TGCs have been studied well, but their nuclear structures remain unknown. In this study, we focus on Lamin B1, a nuclear lamina, and clarify its expression dynamics, regulation and roles in TGC functions. TGCs that differentiated from trophoblast stem cells were used. From days 0 to 9 after differentiation, the number of TGCs gradually increased, but the amount of LMNB1 peaked at day 3 and then slightly decreased. An immunostaining experiment showed that LMNB1-depleted TGCs increased after day 6 of differentiation. These LMNB1-depleted TGCs diffused peripheral localization of the heterochromatin marker H3K9me2 in the nuclei. However, LMINB1-knock down was not affected TGCs specific gene expression. We found that the death of TGCs also increased after day 6 of differentiation. Moreover, Lamin B1 loss and the cell death in TGCs were protected by 10-6 M progesterone. Our results conclude that progesterone protects against Lamin B1 loss and prolongs the life and function of TGCs.
Collapse
Affiliation(s)
- Hiromu Morimoto
- Department of Bioscience, Tokyo University of Agriculture, Tokyo, Japan
| | - Misuzu Ueno
- Department of Bioscience, Tokyo University of Agriculture, Tokyo, Japan
| | - Hideyuki Tanabe
- Department of Evolutionary Studies of Biosystems Science, School of Advanced Sciences, The Graduate University for Advanced Studies, SOKENDAI, Shonan Village, Hayama, Kanagawa, Japan
| | - Tomohiro Kono
- Department of Bioscience, Tokyo University of Agriculture, Tokyo, Japan
| | - Hidehiko Ogawa
- Department of Bioscience, Tokyo University of Agriculture, Tokyo, Japan
| |
Collapse
|
23
|
Guernsey MW, van Kruistum H, Reznick DN, Pollux BJA, Baker JC. Molecular Signatures of Placentation and Secretion Uncovered in Poeciliopsis Maternal Follicles. Mol Biol Evol 2021; 37:2679-2690. [PMID: 32421768 PMCID: PMC7475030 DOI: 10.1093/molbev/msaa121] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Placentation evolved many times independently in vertebrates. Although the core functions of all placentas are similar, we know less about how this similarity extends to the molecular level. Here, we study Poeciliopsis, a unique genus of live-bearing fish that have independently evolved complex placental structures at least three times. The maternal follicle is a key component of these structures. It envelops yolk-rich eggs and is morphologically simple in lecithotrophic species but has elaborate villous structures in matrotrophic species. Through sequencing, the follicle transcriptome of a matrotrophic, Poeciliopsis retropinna, and lecithotrophic, P. turrubarensis, species we found genes known to be critical for placenta function expressed in both species despite their difference in complexity. Additionally, when we compare the transcriptome of different river populations of P. retropinna, known to vary in maternal provisioning, we find differential expression of secretory genes expressed specifically in the top layer of villi cells in the maternal follicle. This provides some of the first evidence that the placental structures of Poeciliopsis function using a secretory mechanism rather than direct contact with maternal circulation. Finally, when we look at the expression of placenta proteins at the maternal–fetal interface of a larger sampling of Poeciliopsis species, we find expression of key maternal and fetal placenta proteins in their cognate tissue types of all species, but follicle expression of prolactin is restricted to only matrotrophic species. Taken together, we suggest that all Poeciliopsis follicles are poised for placenta function but require expression of key genes to form secretory villi.
Collapse
Affiliation(s)
- Michael W Guernsey
- Department of Genetics, Stanford University School of Medicine, Stanford, CA
| | - Henri van Kruistum
- Department of Animal Sciences, Wageningen University, Wageningen, The Netherlands
| | - David N Reznick
- Department of Biology, University of California Riverside, Riverside, CA
| | - Bart J A Pollux
- Department of Animal Sciences, Wageningen University, Wageningen, The Netherlands
| | - Julie C Baker
- Department of Genetics, Stanford University School of Medicine, Stanford, CA
| |
Collapse
|
24
|
Sui Y, Peng S. A Mechanism Leading to Changes in Copy Number Variations Affected by Transcriptional Level Might Be Involved in Evolution, Embryonic Development, Senescence, and Oncogenesis Mediated by Retrotransposons. Front Cell Dev Biol 2021; 9:618113. [PMID: 33644055 PMCID: PMC7905054 DOI: 10.3389/fcell.2021.618113] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 01/11/2021] [Indexed: 01/05/2023] Open
Abstract
In recent years, more and more evidence has emerged showing that changes in copy number variations (CNVs) correlated with the transcriptional level can be found during evolution, embryonic development, and oncogenesis. However, the underlying mechanisms remain largely unknown. The success of the induced pluripotent stem cell suggests that genome changes could bring about transformations in protein expression and cell status; conversely, genome alterations generated during embryonic development and senescence might also be the result of genome changes. With rapid developments in science and technology, evidence of changes in the genome affected by transcriptional level has gradually been revealed, and a rational and concrete explanation is needed. Given the preference of the HIV-1 genome to insert into transposons of genes with high transcriptional levels, we propose a mechanism based on retrotransposons facilitated by specific pre-mRNA splicing style and homologous recombination (HR) to explain changes in CNVs in the genome. This mechanism is similar to that of the group II intron that originated much earlier. Under this proposed mechanism, CNVs on genome are dynamically and spontaneously extended in a manner that is positively correlated with transcriptional level or contract as the cell divides during evolution, embryonic development, senescence, and oncogenesis, propelling alterations in them. Besides, this mechanism explains several critical puzzles in these processes. From evidence collected to date, it can be deduced that the message contained in genome is not just three-dimensional but will become four-dimensional, carrying more genetic information.
Collapse
Affiliation(s)
- Yunpeng Sui
- Department of Functional Neurosurgery, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | | |
Collapse
|
25
|
Chattopadhyaya S, Banerjee S. miRNA 146b mediates the regulation of nucleolar size and activity in polyploid megakaryocytes. Biol Cell 2020; 113:118-129. [PMID: 33278308 DOI: 10.1111/boc.202000022] [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: 02/22/2020] [Accepted: 11/25/2020] [Indexed: 02/03/2023]
Abstract
BACKGROUND INFORMATION Megakaryocytes (MKs) follow a unique cell cycle duplication process, called endomitosis, resulting in polyploidisation of cells. It is hypothesised that polyploidy, as well as an increment in cytoplasm volume, allow more efficient platelets generation from MKs. Although polyploidy leads to an increase in the DNA amount, which impacts gene expression, little is known about ribosomal biogenesis in these polylobulated polyploid cells. RESULTS The nucleolus acts as a hub for ribosomal biogenesis, which in turn governs the protein synthesis rate of the cells. We therefore estimated the size and activity of the nucleolus in polyploid cells during megakaryopoiesis in vitro. Polyploid megakaryocytic cell lines and in vitro cultured MKs, which were obtained from human cord blood-derived CD 34+ cells, revealed that miRNA 146b regulated the activity of nucleolar and coiled-body phosphoprotein 1, which plays an integral role in determining nucleolar size and activity. Additionally, miRNA-146b was up-regulated during endomitosis and was found to promote megakaryopoiesis. CONCLUSION We propose that miRNA 146b regulates not only nucleolar size and activity, but also megakaryopoiesis. SIGNIFICANCE This study highlights the importance of nucleolar activity and miRNA in the progression of megakaryopoiesis and thrombopoiesis.
Collapse
Affiliation(s)
- Saran Chattopadhyaya
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, Homi Bhabha National Institute, Bidhannagar, Kolkata, 700064, India
| | - Subrata Banerjee
- School of Biological Sciences, Ramkrishna Mission Vivekananda Educational & Research Institute (RKMVERI), Narendrapur, Kolkata, 700103, India
| |
Collapse
|
26
|
Abstract
The hallmark of most cardiac diseases is the progressive loss of cardiomyocytes. In the perinatal period, cardiomyocytes still proliferate, and the heart shows the capacity to regenerate upon injury. In the adult heart, however, the actual rate of cardiomyocyte renewal is too low to efficiently counteract substantial cell loss caused by cardiac injury. In mammals, cardiac growth by cell number expansion changes to growth by cardiomyocyte enlargement soon after birth, coinciding with a period in which most cardiomyocytes increase their DNA content by multinucleation and nuclear polyploidization. Although cardiomyocyte hypertrophy is often associated with these processes, whether polyploidy is a prerequisite or a consequence of hypertrophic growth is unclear. Both the benefits of cardiomyocyte enlargement over proliferative growth of the heart and the physiological role of polyploidy in cardiomyocytes are enigmatic. Interestingly, hearts in animal species with substantial cardiac regenerative capacity dominantly comprise diploid cardiomyocytes, raising the hypothesis that cardiomyocyte polyploidy poses a barrier for cardiomyocyte proliferation and subsequent heart regeneration. On the contrary, there is also evidence for self-duplication of multinucleated myocytes, suggesting a more complex picture of polyploidy in heart regeneration. Polyploidy is not restricted to the heart but also occurs in other cell types in the body. In this review, we explore the biological relevance of polyploidy in different species and tissues to acquire insight into its specific role in cardiomyocytes. Furthermore, we speculate about the physiological role of polyploidy in cardiomyocytes and how this might relate to renewal and regeneration.
Collapse
Affiliation(s)
- Wouter Derks
- From the Center for Regenerative Therapies Dresden, Technische Universität Dresden, Germany (W.D., O.B.)
| | - Olaf Bergmann
- From the Center for Regenerative Therapies Dresden, Technische Universität Dresden, Germany (W.D., O.B.).,Karolinska Institutet, Cell and Molecular Biology, Stockholm, Sweden (O.B.)
| |
Collapse
|
27
|
Eaton M, Davies AH, Devine J, Zhao X, Simmons DG, Maríusdóttir E, Natale DRC, Matyas JR, Bering EA, Workentine ML, Hallgrimsson B, Cross JC. Complex patterns of cell growth in the placenta in normal pregnancy and as adaptations to maternal diet restriction. PLoS One 2020; 15:e0226735. [PMID: 31917811 PMCID: PMC6952106 DOI: 10.1371/journal.pone.0226735] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 12/03/2019] [Indexed: 02/06/2023] Open
Abstract
The major milestones in mouse placental development are well described, but our understanding is limited to how the placenta can adapt to damage or changes in the environment. By using stereology and expression of cell cycle markers, we found that the placenta grows under normal conditions not just by hyperplasia of trophoblast cells but also through extensive polyploidy and cell hypertrophy. In response to feeding a low protein diet to mothers prior to and during pregnancy, to mimic chronic malnutrition, we found that this normal program was altered and that it was influenced by the sex of the conceptus. Male fetuses showed intrauterine growth restriction (IUGR) by embryonic day (E) 18.5, just before term, whereas female fetuses showed IUGR as early as E16.5. This difference was correlated with differences in the size of the labyrinth layer of the placenta, the site of nutrient and gas exchange. Functional changes were implied based on up-regulation of nutrient transporter genes. The junctional zone was also affected, with a reduction in both glycogen trophoblast and spongiotrophoblast cells. These changes were associated with increased expression of Phlda2 and reduced expression of Egfr. Polyploidy, which results from endoreduplication, is a normal feature of trophoblast giant cells (TGC) but also spongiotrophoblast cells. Ploidy was increased in sinusoidal-TGCs and spongiotrophoblast cells, but not parietal-TGCs, in low protein placentas. These results indicate that the placenta undergoes a range of changes in development and function in response to poor maternal diet, many of which we interpret are aimed at mitigating the impacts on fetal and maternal health.
Collapse
Affiliation(s)
- Malcolm Eaton
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary Alberta
| | - Alastair H. Davies
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary Alberta
| | - Jay Devine
- Department of Anatomy and Cell Biology, Cumming School of Medicine, University of Calgary, Calgary Alberta
| | - Xiang Zhao
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary Alberta
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary Alberta
| | - David G. Simmons
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary Alberta
| | - Elín Maríusdóttir
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary Alberta
| | - David R. C. Natale
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary Alberta
| | - John R. Matyas
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary Alberta
| | - Elizabeth A. Bering
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary Alberta
| | | | - Benedikt Hallgrimsson
- Department of Anatomy and Cell Biology, Cumming School of Medicine, University of Calgary, Calgary Alberta
| | - James C. Cross
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary Alberta
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary Alberta
- * E-mail:
| |
Collapse
|
28
|
Varaden D, Moodley J, Onyangunga OA, Naicker T. Morphometric image analysis of placental C-type lectin domain family 2, member D (CLEC2D) immuno-expression in HIV associated pre-eclampsia. Eur J Obstet Gynecol Reprod Biol X 2019; 3:100039. [PMID: 31403127 PMCID: PMC6687384 DOI: 10.1016/j.eurox.2019.100039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2018] [Revised: 04/11/2019] [Accepted: 04/29/2019] [Indexed: 01/08/2023] Open
Abstract
OBJECTIVE C-type lectin domain family 2, member D (CLEC2D) is implicated in the immune response. Pre-eclampsia and HIV infection have opposing immune responses. In view of the high prevalence of HIV infection and pre-eclampsia in South Africa, this study assessed the placental immuno-expression of CLEC2D in HIV associated pre-eclampsia. METHOD Placental tissue was obtained from 60 pregnancies which were categorized according to pregnancy type (pre-eclamptic or normotensive) and HIV status (positive or negative). Immunohistochemistry and morphometric image analysis were used to evaluate placental CLEC2D immuno-expression. RESULTS CLEC2D expression was significantly decreased in the conducting villi of pre-eclamptic vs normotensive placentae (p = 0.0418) but was increased in the exchange villi, albeit non-significant (p = 0.4948). HIV positive status intensified placental CLEC2D immuno-expression in conducting (p = 0.0312) and exchange (p = 0.0025) villi. CLEC2D expression was significantly different in exchange vs conducting villi (p < 0.0001) and across study groups (p = 0.0003). Normotensive; HIV negative placentae (control) had a non-significant difference in CLEC2D expression across villi types, however significant difference was noted within the remaining groups: normotensive; HIV positive (p < 0.05), pre-eclamptic; HIV positive (p < 0.01 and pre-eclamptic; HIV negative (p < 0.001). CONCLUSION The contrasting expression of CLEC2D in HIV infection and pre-eclampsia is demonstrative of the immunosuppressive and pro-inflammatory roles of the respective pathologies. However, this implication may be confounded by highly active anti-retroviral treatment (HAART).
Collapse
Affiliation(s)
- Deneshree Varaden
- Optics and Imaging Centre, University of KwaZulu-Natal, Durban, KwaZulu-Natal, South Africa
| | - Jagidesa Moodley
- Womens Health and HIV Research Unit, University of KwaZulu-Natal, Durban, KwaZulu-Natal, South Africa
| | - Onankoy A. Onyangunga
- Optics and Imaging Centre, University of KwaZulu-Natal, Durban, KwaZulu-Natal, South Africa
| | - Thajasvarie Naicker
- Optics and Imaging Centre, University of KwaZulu-Natal, Durban, KwaZulu-Natal, South Africa
| |
Collapse
|
29
|
Kalisch-Smith JI, Steane SE, Simmons DG, Pantaleon M, Anderson ST, Akison LK, Wlodek ME, Moritz KM. Periconceptional alcohol exposure causes female-specific perturbations to trophoblast differentiation and placental formation in the rat. Development 2019; 146:dev172205. [PMID: 31182432 DOI: 10.1242/dev.172205] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2018] [Accepted: 04/18/2019] [Indexed: 12/26/2022]
Abstract
The development of pathologies during pregnancy, including pre-eclampsia, hypertension and fetal growth restriction (FGR), often originates from poor functioning of the placenta. In vivo models of maternal stressors, such as nutrient deficiency, and placental insufficiency often focus on inadequate growth of the fetus and placenta in late gestation. These studies rarely investigate the origins of poor placental formation in early gestation, including those affecting the pre-implantation embryo and/or the uterine environment. The current study characterises the impact on blastocyst, uterine and placental outcomes in a rat model of periconceptional alcohol exposure, in which 12.5% ethanol is administered in a liquid diet from 4 days before until 4 days after conception. We show female-specific effects on trophoblast differentiation, embryo-uterine communication, and formation of the placental vasculature, resulting in markedly reduced placental volume at embryonic day 15. Both sexes exhibited reduced trophectoderm pluripotency and global hypermethylation, suggestive of inappropriate epigenetic reprogramming. Furthermore, evidence of reduced placental nutrient exchange and reduced pre-implantation maternal plasma choline levels offers significant mechanistic insight into the origins of FGR in this model.
Collapse
Affiliation(s)
- Jacinta I Kalisch-Smith
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Sarah E Steane
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia, QLD 4072, Australia
| | - David G Simmons
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Marie Pantaleon
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Stephen T Anderson
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Lisa K Akison
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia, QLD 4072, Australia
- Child Health Research Centre, The University of Queensland, South Brisbane, QLD 4101, Australia
| | - Mary E Wlodek
- Department of Physiology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Karen M Moritz
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia, QLD 4072, Australia
- Child Health Research Centre, The University of Queensland, South Brisbane, QLD 4101, Australia
| |
Collapse
|
30
|
Fischer U, Backes C, Fehlmann T, Galata V, Keller A, Meese E. Prospect and challenge of detecting dynamic gene copy number increases in stem cells by whole genome sequencing. J Mol Med (Berl) 2019; 97:1099-1111. [PMID: 31134286 PMCID: PMC6647207 DOI: 10.1007/s00109-019-01792-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 04/29/2019] [Accepted: 05/01/2019] [Indexed: 12/03/2022]
Abstract
Abstract Gene amplification is an evolutionarily well-conserved and highly efficient mechanism to increase the amount of specific proteins. In humans, gene amplification is a hallmark of cancer and has recently been found during stem cell differentiation. Amplifications in stem cells are restricted to specific tissue areas and time windows, rendering their detection difficult. Here, we report on the performance of deep WGS sequencing (average 82-fold depth of coverage) on the BGISEQ with nanoball technology to detect amplifications in human mesenchymal and neural stem cells. As reference technology, we applied array-based comparative genomic hybridization (aCGH), fluorescence in situ hybridization (FISH), and qPCR. Using different in silico strategies for amplification detection, we analyzed the potential of WGS for amplification detection. Our results provide evidence that WGS accurately identifies changes of the copy number profiles in human stem cell differentiation. However, the identified changes are not in all cases consistent between WGS and aCGH. The results between WGS and the validation by qPCR were concordant in 83.3% of all tested 36 cases. In sum, both genome-wide techniques, aCGH and WGS, have unique advantages and specific challenges, calling for locus-specific confirmation by the low-throughput approaches qPCR or FISH. Key messages WGS allows for the identification of dynamic copy number changes in human stem cells. Less stringent threshold setting is crucial for detection of copy number increase. Broad knowledge of dynamic copy number is pivotal to estimate stem cell capabilities.
Electronic supplementary material The online version of this article (10.1007/s00109-019-01792-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ulrike Fischer
- Department of Human Genetics, Saarland University, Building 60, 66421, Homburg/Saar, Germany.
| | - Christina Backes
- Clinical Bioinformatics, Saarland University, Building E2.1, 66123, Saarbrücken, Germany
| | - Tobias Fehlmann
- Clinical Bioinformatics, Saarland University, Building E2.1, 66123, Saarbrücken, Germany
| | - Valentina Galata
- Clinical Bioinformatics, Saarland University, Building E2.1, 66123, Saarbrücken, Germany
| | - Andreas Keller
- Clinical Bioinformatics, Saarland University, Building E2.1, 66123, Saarbrücken, Germany
| | - Eckart Meese
- Department of Human Genetics, Saarland University, Building 60, 66421, Homburg/Saar, Germany
| |
Collapse
|
31
|
Schiebinger G, Shu J, Tabaka M, Cleary B, Subramanian V, Solomon A, Gould J, Liu S, Lin S, Berube P, Lee L, Chen J, Brumbaugh J, Rigollet P, Hochedlinger K, Jaenisch R, Regev A, Lander ES. Optimal-Transport Analysis of Single-Cell Gene Expression Identifies Developmental Trajectories in Reprogramming. Cell 2019; 176:928-943.e22. [PMID: 30712874 PMCID: PMC6402800 DOI: 10.1016/j.cell.2019.01.006] [Citation(s) in RCA: 279] [Impact Index Per Article: 55.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 10/15/2018] [Accepted: 01/02/2019] [Indexed: 12/18/2022]
Abstract
Understanding the molecular programs that guide differentiation during development is a major challenge. Here, we introduce Waddington-OT, an approach for studying developmental time courses to infer ancestor-descendant fates and model the regulatory programs that underlie them. We apply the method to reconstruct the landscape of reprogramming from 315,000 single-cell RNA sequencing (scRNA-seq) profiles, collected at half-day intervals across 18 days. The results reveal a wider range of developmental programs than previously characterized. Cells gradually adopt either a terminal stromal state or a mesenchymal-to-epithelial transition state. The latter gives rise to populations related to pluripotent, extra-embryonic, and neural cells, with each harboring multiple finer subpopulations. The analysis predicts transcription factors and paracrine signals that affect fates and experiments validate that the TF Obox6 and the cytokine GDF9 enhance reprogramming efficiency. Our approach sheds light on the process and outcome of reprogramming and provides a framework applicable to diverse temporal processes in biology.
Collapse
Affiliation(s)
- Geoffrey Schiebinger
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; MIT Center for Statistics, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Jian Shu
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA.
| | - Marcin Tabaka
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Brian Cleary
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Computational and Systems Biology Program, MIT, Cambridge, MA 02142, USA
| | - Vidya Subramanian
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Aryeh Solomon
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Joshua Gould
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Siyan Liu
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Biochemistry Program, Wellesley College, Wellesley, MA 02481, USA
| | - Stacie Lin
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Peter Berube
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Lia Lee
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Jenny Chen
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA 02139, USA
| | - Justin Brumbaugh
- Cancer Center, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Molecular Biology, Center for Regenerative Medicine and Cancer Center, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Philippe Rigollet
- MIT Center for Statistics, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Mathematics, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Konrad Hochedlinger
- Department of Molecular Biology, Center for Regenerative Medicine and Cancer Center, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| | - Rudolf Jaenisch
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA; Computational and Systems Biology Program, MIT, Cambridge, MA 02142, USA
| | - Aviv Regev
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA.
| | - Eric S Lander
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Systems Biology Harvard Medical School, Boston, MA 02125, USA.
| |
Collapse
|
32
|
Gjelsvik KJ, Besen-McNally R, Losick VP. Solving the Polyploid Mystery in Health and Disease. Trends Genet 2019; 35:6-14. [PMID: 30470486 PMCID: PMC6457904 DOI: 10.1016/j.tig.2018.10.005] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 10/12/2018] [Accepted: 10/22/2018] [Indexed: 01/12/2023]
Abstract
Polyploidy (the more than doubling of a cell's genome) frequently arises during organogenesis, tissue repair, and age-associated diseases. Despite its prevalence, major gaps exist in how polyploid cells emerge and affect tissue function. Studies have begun to elucidate the signals required for polyploid cell growth as well as the advantages and disadvantages of polyploidy in health and disease. This review highlights the recent advances on the role and regulation of polyploidy in Drosophila and vertebrate models. The newly discovered versatility of polyploid cells has the potential to provide alternative strategies to promote tissue growth and repair, while limiting disease and dysfunction.
Collapse
Affiliation(s)
- K J Gjelsvik
- MDI Biological Laboratory, 159 Old Bar Harbor Road, Bar Harbor, ME 04609, USA
| | - R Besen-McNally
- MDI Biological Laboratory, 159 Old Bar Harbor Road, Bar Harbor, ME 04609, USA
| | - V P Losick
- MDI Biological Laboratory, 159 Old Bar Harbor Road, Bar Harbor, ME 04609, USA.
| |
Collapse
|
33
|
Munden A, Rong Z, Sun A, Gangula R, Mallal S, Nordman JT. Rif1 inhibits replication fork progression and controls DNA copy number in Drosophila. eLife 2018; 7:e39140. [PMID: 30277458 PMCID: PMC6185109 DOI: 10.7554/elife.39140] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 10/01/2018] [Indexed: 12/22/2022] Open
Abstract
Control of DNA copy number is essential to maintain genome stability and ensure proper cell and tissue function. In Drosophila polyploid cells, the SNF2-domain-containing SUUR protein inhibits replication fork progression within specific regions of the genome to promote DNA underreplication. While dissecting the function of SUUR's SNF2 domain, we identified an interaction between SUUR and Rif1. Rif1 has many roles in DNA metabolism and regulates the replication timing program. We demonstrate that repression of DNA replication is dependent on Rif1. Rif1 localizes to active replication forks in a partially SUUR-dependent manner and directly regulates replication fork progression. Importantly, SUUR associates with replication forks in the absence of Rif1, indicating that Rif1 acts downstream of SUUR to inhibit fork progression. Our findings uncover an unrecognized function of the Rif1 protein as a regulator of replication fork progression.
Collapse
Affiliation(s)
- Alexander Munden
- Department of Biological SciencesVanderbilt UniversityNashvilleUnited States
| | - Zhan Rong
- Department of Biological SciencesVanderbilt UniversityNashvilleUnited States
| | - Amanda Sun
- Department of Biological SciencesVanderbilt UniversityNashvilleUnited States
| | - Rama Gangula
- Department of MedicineVanderbilt University School of MedicineNashvilleUnited States
| | - Simon Mallal
- Department of MedicineVanderbilt University School of MedicineNashvilleUnited States
- Department of Pathology, Microbiology and ImmunologyVanderbilt University School of MedicineNashvilleUnited States
| | - Jared T Nordman
- Department of Biological SciencesVanderbilt UniversityNashvilleUnited States
| |
Collapse
|
34
|
Velicky P, Meinhardt G, Plessl K, Vondra S, Weiss T, Haslinger P, Lendl T, Aumayr K, Mairhofer M, Zhu X, Schütz B, Hannibal RL, Lindau R, Weil B, Ernerudh J, Neesen J, Egger G, Mikula M, Röhrl C, Urban AE, Baker J, Knöfler M, Pollheimer J. Genome amplification and cellular senescence are hallmarks of human placenta development. PLoS Genet 2018; 14:e1007698. [PMID: 30312291 PMCID: PMC6200260 DOI: 10.1371/journal.pgen.1007698] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 10/24/2018] [Accepted: 09/17/2018] [Indexed: 12/16/2022] Open
Abstract
Genome amplification and cellular senescence are commonly associated with pathological processes. While physiological roles for polyploidization and senescence have been described in mouse development, controversy exists over their significance in humans. Here, we describe tetraploidization and senescence as phenomena of normal human placenta development. During pregnancy, placental extravillous trophoblasts (EVTs) invade the pregnant endometrium, termed decidua, to establish an adapted microenvironment required for the developing embryo. This process is critically dependent on continuous cell proliferation and differentiation, which is thought to follow the classical model of cell cycle arrest prior to terminal differentiation. Strikingly, flow cytometry and DNAseq revealed that EVT formation is accompanied with a genome-wide polyploidization, independent of mitotic cycles. DNA replication in these cells was analysed by a fluorescent cell-cycle indicator reporter system, cell cycle marker expression and EdU incorporation. Upon invasion into the decidua, EVTs widely lose their replicative potential and enter a senescent state characterized by high senescence-associated (SA) β-galactosidase activity, induction of a SA secretory phenotype as well as typical metabolic alterations. Furthermore, we show that the shift from endocycle-dependent genome amplification to growth arrest is disturbed in androgenic complete hydatidiform moles (CHM), a hyperplastic pregnancy disorder associated with increased risk of developing choriocarinoma. Senescence is decreased in CHM-EVTs, accompanied by exacerbated endoreduplication and hyperploidy. We propose induction of cellular senescence as a ploidy-limiting mechanism during normal human placentation and unravel a link between excessive polyploidization and reduced senescence in CHM.
Collapse
Affiliation(s)
- Philipp Velicky
- Department of Obstetrics and Gynaecology, Reproductive Biology Unit, Medical University of Vienna, Vienna, Austria
| | - Gudrun Meinhardt
- Department of Obstetrics and Gynaecology, Reproductive Biology Unit, Medical University of Vienna, Vienna, Austria
| | - Kerstin Plessl
- Department of Obstetrics and Gynaecology, Reproductive Biology Unit, Medical University of Vienna, Vienna, Austria
| | - Sigrid Vondra
- Department of Obstetrics and Gynaecology, Reproductive Biology Unit, Medical University of Vienna, Vienna, Austria
| | - Tamara Weiss
- Children's Cancer Research Institute, St. Anna Children´s Hospital, Vienna, Austria
| | - Peter Haslinger
- Department of Obstetrics and Gynaecology, Reproductive Biology Unit, Medical University of Vienna, Vienna, Austria
| | - Thomas Lendl
- Biooptics Facility of Institute of Molecular Pathology, Institute of Molecular Biotechnology and Gregor Mendel Institute, Vienna, Austria
| | - Karin Aumayr
- Biooptics Facility of Institute of Molecular Pathology, Institute of Molecular Biotechnology and Gregor Mendel Institute, Vienna, Austria
| | - Mario Mairhofer
- Department of Gynecological Endocrinology and Reproductive Medicine, Medical University of Vienna, Vienna, Austria
| | - Xiaowei Zhu
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, California, United States of America
| | - Birgit Schütz
- Center for Pathobiochemistry and Genetics, Medical University of Vienna, Vienna, Austria
| | - Roberta L. Hannibal
- Department of Genetics, Stanford University School of Medicine, Stanford, California, United States of America
| | - Robert Lindau
- Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Beatrix Weil
- Center for Pathobiochemistry and Genetics, Medical University of Vienna, Vienna, Austria
| | - Jan Ernerudh
- Department of Clinical Immunology and Transfusion Medicine, and Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Jürgen Neesen
- Center for Pathobiochemistry and Genetics, Medical University of Vienna, Vienna, Austria
| | - Gerda Egger
- Clinical Institute of Pathology, Medical University of Vienna, Vienna, Austria
| | - Mario Mikula
- Center for Pathobiochemistry and Genetics, Medical University of Vienna, Vienna, Austria
| | - Clemens Röhrl
- Center for Pathobiochemistry and Genetics, Medical University of Vienna, Vienna, Austria
| | - Alexander E. Urban
- Department of Psychiatry and Behavioral Sciences, Department of Genetics, Stanford University School of Medicine, Tasha and John Morgridge Faculty Scholar, Stanford Child Health Research Institute, Stanford, California, United States of America
| | - Julie Baker
- Department of Genetics, Stanford University School of Medicine, Stanford, California, United States of America
| | - Martin Knöfler
- Department of Obstetrics and Gynaecology, Reproductive Biology Unit, Medical University of Vienna, Vienna, Austria
| | - Jürgen Pollheimer
- Department of Obstetrics and Gynaecology, Reproductive Biology Unit, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
35
|
Okano-Uchida T, Kent LN, Ouseph MM, McCarty B, Frank JJ, Kladney R, Cuitino MC, Thompson JC, Coppola V, Asano M, Leone G. Endoreduplication of the mouse genome in the absence of ORC1. Genes Dev 2018; 32:978-990. [PMID: 29967292 PMCID: PMC6075035 DOI: 10.1101/gad.311910.118] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 05/09/2018] [Indexed: 01/24/2023]
Abstract
In this study, Okano-Uchida et al. describe the physiological role of ORC1 in mice by generating knock-in mice with LoxP sites flanking exons encoding the critical ATPase domain of ORC1. They show that ORC1 ablation in extraembryonic trophoblasts and hepatocytes failed to impede genome endoreduplication and organ development and function and conclude that ORC1 in mice is essential for mitotic cell divisions but dispensable for endoreduplication. The largest subunit of the origin recognition complex (ORC1) is essential for assembly of the prereplicative complex, firing of DNA replication origins, and faithful duplication of the genome. Here, we generated knock-in mice with LoxP sites flanking exons encoding the critical ATPase domain of ORC1. Global or tissue-specific ablation of ORC1 function in mouse embryo fibroblasts and fetal and adult diploid tissues blocked DNA replication, cell lineage expansion, and organ development. Remarkably, ORC1 ablation in extraembryonic trophoblasts and hepatocytes, two polyploid cell types in mice, failed to impede genome endoreduplication and organ development and function. Thus, ORC1 in mice is essential for mitotic cell divisions but dispensable for endoreduplication. We propose that DNA replication of mammalian polyploid genomes uses a distinct ORC1-independent mechanism.
Collapse
Affiliation(s)
- Takayuki Okano-Uchida
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina 29425, USA
| | - Lindsey N Kent
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina 29425, USA
| | - Madhu M Ouseph
- Solid Tumor Biology Program, Comprehensive Cancer Center, Ohio State University, Columbus, Ohio 43210, USA.,Department of Molecular Genetics, Ohio State University, Columbus, Ohio 43210, USA.,Department of Cancer Biology and Genetics, Ohio State University, Columbus, Ohio 43210, USA
| | - Britney McCarty
- Solid Tumor Biology Program, Comprehensive Cancer Center, Ohio State University, Columbus, Ohio 43210, USA.,Department of Molecular Genetics, Ohio State University, Columbus, Ohio 43210, USA.,Department of Cancer Biology and Genetics, Ohio State University, Columbus, Ohio 43210, USA
| | - Jeffrey J Frank
- Solid Tumor Biology Program, Comprehensive Cancer Center, Ohio State University, Columbus, Ohio 43210, USA.,Department of Molecular Genetics, Ohio State University, Columbus, Ohio 43210, USA.,Department of Cancer Biology and Genetics, Ohio State University, Columbus, Ohio 43210, USA
| | - Raleigh Kladney
- Solid Tumor Biology Program, Comprehensive Cancer Center, Ohio State University, Columbus, Ohio 43210, USA.,Department of Molecular Genetics, Ohio State University, Columbus, Ohio 43210, USA.,Department of Cancer Biology and Genetics, Ohio State University, Columbus, Ohio 43210, USA
| | - Maria C Cuitino
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina 29425, USA
| | - John C Thompson
- Solid Tumor Biology Program, Comprehensive Cancer Center, Ohio State University, Columbus, Ohio 43210, USA.,Department of Molecular Genetics, Ohio State University, Columbus, Ohio 43210, USA.,Department of Cancer Biology and Genetics, Ohio State University, Columbus, Ohio 43210, USA
| | - Vincenzo Coppola
- Solid Tumor Biology Program, Comprehensive Cancer Center, Ohio State University, Columbus, Ohio 43210, USA.,Department of Molecular Genetics, Ohio State University, Columbus, Ohio 43210, USA.,Department of Cancer Biology and Genetics, Ohio State University, Columbus, Ohio 43210, USA
| | - Maki Asano
- Solid Tumor Biology Program, Comprehensive Cancer Center, Ohio State University, Columbus, Ohio 43210, USA.,Department of Molecular Genetics, Ohio State University, Columbus, Ohio 43210, USA.,Department of Molecular Cellular and Biochemistry, Ohio State University, Columbus, Ohio 43210, USA
| | - Gustavo Leone
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina 29425, USA
| |
Collapse
|
36
|
Abstract
Polyploid cells, which contain multiple copies of the typically diploid genome, are widespread in plants and animals. Polyploidization can be developmentally programmed or stress induced, and arises from either cell-cell fusion or a process known as endoreplication, in which cells replicate their DNA but either fail to complete cytokinesis or to progress through M phase entirely. Polyploidization offers cells several potential fitness benefits, including the ability to increase cell size and biomass production without disrupting cell and tissue structure, and allowing improved cell longevity through higher tolerance to genomic stress and apoptotic signals. Accordingly, recent studies have uncovered crucial roles for polyploidization in compensatory cell growth during tissue regeneration in the heart, liver, epidermis and intestine. Here, we review current knowledge of the molecular pathways that generate polyploidy and discuss how polyploidization is used in tissue repair and regeneration.
Collapse
Affiliation(s)
| | - Bruce A Edgar
- Huntsman Cancer Institute, Salt Lake City, UT 84112, USA
| |
Collapse
|
37
|
Nucleosomes of polyploid trophoblast giant cells mostly consist of histone variants and form a loose chromatin structure. Sci Rep 2018; 8:5811. [PMID: 29643413 PMCID: PMC5895725 DOI: 10.1038/s41598-018-23832-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 03/20/2018] [Indexed: 12/31/2022] Open
Abstract
Trophoblast giant cells (TGCs) are one of the cell types that form the placenta and play multiple essential roles in maintaining pregnancy in rodents. TGCs have large, polyploid nuclei resulting from endoreduplication. While previous studies have shown distinct gene expression profiles of TGCs, their chromatin structure remains largely unknown. An appropriate combination of canonical and non-canonical histones, also known as histone variants, allows each cell to exert its cell type-specific functions. Here, we aimed to reveal the dynamics of histone usage and chromatin structure during the differentiation of trophoblast stem cells (TSCs) into TGCs. Although the expression of most genes encoding canonical histones was downregulated, the expression of a few genes encoding histone variants such as H2AX, H2AZ, and H3.3 was maintained at a relatively high level in TGCs. Both the micrococcal nuclease digestion assay and nucleosome stability assay using a microfluidic device indicated that chromatin became increasingly loose as TSCs differentiated. Combinatorial experiments involving H3.3-knockdown and -overexpression demonstrated that variant H3.3 resulted in the formation of loose nucleosomes in TGCs. In conclusion, our study revealed that TGCs possessed loose nucleosomes owing to alterations in their histone composition during differentiation.
Collapse
|
38
|
Fischer U, Kim E, Keller A, Meese E. Specific amplifications and copy number decreases during human neural stem cells differentiation towards astrocytes, neurons and oligodendrocytes. Oncotarget 2018; 8:25872-25884. [PMID: 28415661 PMCID: PMC5432223 DOI: 10.18632/oncotarget.15980] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 02/27/2017] [Indexed: 12/16/2022] Open
Abstract
There is growing evidence that gene amplifications are an attribute of normal cells during development and differentiation. During neural progenitor cell differentiation half of the genome is involved in amplification process. To answer the question how specific amplifications occur at different stages and in different lineages of differentiation we analyzed the genes CDK4, MDM2, EGFR, GINS2, GFAP, TP53, DDB1 and MDM4 in human neural stem cells that were induced to differentiate towards astrocytes, neurons and oligodendrocytes. We found specific amplification pattern for each of the eight analyzed genes both in undifferentiated neural stem and progenitor cells and in cells that were induced for differentiation. Different amplification patterns were also found between adherently grown neural stem cells and cells that were grown as spheres. The most frequently amplified genes were MDM2 and CDK4 with the latter amplified in all three lineages at all analyzed stages. Amplification of the analyzed genes was also found in four glioma stem-like cells. The combined amplification data of stem cells and of tumor stem cells can help to define cell populations at the origin of the tumor. Furthermore, we detected a decrease of gene copies at specific differentiation stages most frequently for MDM4. This study shows specific amplification pattern in defined stem cell populations within specific time windows during differentiation processes indicating that amplifications occur in an orderly sequence during the differentiation of human neural stem and progenitor cells.
Collapse
Affiliation(s)
- Ulrike Fischer
- Department of Human Genetics, Saarland University, Homburg/Saar, Germany
| | - Ella Kim
- Translational Neurooncology Research Group, Johannes Gutenberg University, Mainz, Germany
| | - Andreas Keller
- Clinical Bioinformatics, Saarland University, Saarbrücken, Germany
| | - Eckart Meese
- Department of Human Genetics, Saarland University, Homburg/Saar, Germany
| |
Collapse
|
39
|
Altmayer NC, Galata V, Warschburger N, Keller A, Meese E, Fischer U. Gene amplification in mesenchymal stem cells and during differentiation towards adipocytes or osteoblasts. Oncotarget 2017; 9:1803-1812. [PMID: 29416732 PMCID: PMC5788600 DOI: 10.18632/oncotarget.22804] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2017] [Accepted: 11/01/2017] [Indexed: 12/20/2022] Open
Abstract
Gene amplifications are an attribute of tumor cells and have for long time been overlooked in normal cells. A growing number of investigations describe gene amplifications in normal mammalian cells during development and differentiation. Possibly, tumor cells have rescued the gene amplification mechanism as a physiological attribute of stem cells. Here, we investigated human mesenchymal stem cells (hMSCs) for gene amplification using array-CGH, single cell fluorescence in situ hybridization and qPCR. Gene amplifications were detected in mesenchymal stem cells and in mesenchymal stem cells during differentiation towards adipocytes and osteoblasts. Undifferentiated hMSCs harbor 12 amplified chromosomal regions, hMSCs that differentiated towards adipocytes 18 amplified chromosome regions, and hMSCs that differentiate towards osteoblasts 19 amplified regions. Specifically, hMSCs that differentiated towards adipocytes or osteoblasts harbor CDK4 and MDM2 amplifications both of which frequently occur in osteosarcoma and liposarcoma that are both of same cell origin. Beside the amplifications, we identified 36 under-replicated regions in undifferentiated and in differentiating hMSC cells.
Collapse
Affiliation(s)
| | - Valentina Galata
- Chair of Clinical Bioinformatics, Saarland University, 66123 Saarbrücken, Germany
| | - Nadine Warschburger
- Department of Human Genetics, Saarland University, 66421 Homburg/Saar, Germany
| | - Andreas Keller
- Chair of Clinical Bioinformatics, Saarland University, 66123 Saarbrücken, Germany
| | - Eckart Meese
- Department of Human Genetics, Saarland University, 66421 Homburg/Saar, Germany
| | - Ulrike Fischer
- Department of Human Genetics, Saarland University, 66421 Homburg/Saar, Germany
| |
Collapse
|
40
|
Stormo BM, Fox DT. Polyteny: still a giant player in chromosome research. Chromosome Res 2017; 25:201-214. [PMID: 28779272 PMCID: PMC5768140 DOI: 10.1007/s10577-017-9562-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 07/19/2017] [Accepted: 07/21/2017] [Indexed: 12/17/2022]
Abstract
In this era of high-resolution mapping of chromosome territories, topological interactions, and chromatin states, it is increasingly appreciated that the positioning of chromosomes and their interactions within the nucleus is critical for cellular function. Due to their large size and distinctive structure, polytene chromosomes have contributed a wealth of knowledge regarding chromosome regulation. In this review, we discuss the diversity of polytene chromosomes in nature and in disease, examine the recurring structural features of polytene chromosomes in terms of what they reveal about chromosome biology, and discuss recent advances regarding how polytene chromosomes are assembled and disassembled. After over 130 years of study, these giant chromosomes are still powerful tools to understand chromosome biology.
Collapse
Affiliation(s)
- Benjamin M Stormo
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, 27710, USA
| | - Donald T Fox
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, 27710, USA.
| |
Collapse
|
41
|
Guernsey MW, Chuong EB, Cornelis G, Renfree MB, Baker JC. Molecular conservation of marsupial and eutherian placentation and lactation. eLife 2017; 6. [PMID: 28895534 PMCID: PMC5595433 DOI: 10.7554/elife.27450] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 07/24/2017] [Indexed: 12/18/2022] Open
Abstract
Eutherians are often mistakenly termed 'placental mammals', but marsupials also have a placenta to mediate early embryonic development. Lactation is necessary for both infant and fetal development in eutherians and marsupials, although marsupials have a far more complex milk repertoire that facilitates morphogenesis of developmentally immature young. In this study, we demonstrate that the anatomically simple tammar placenta expresses a dynamic molecular program that is reminiscent of eutherian placentation, including both fetal and maternal signals. Further, we provide evidence that genes facilitating fetal development and nutrient transport display convergent co-option by placental and mammary gland cell types to optimize offspring success.
Collapse
Affiliation(s)
- Michael W Guernsey
- Department of Genetics, Stanford University School of Medicine, Stanford, United States
| | - Edward B Chuong
- Department of Human Genetics, University of Utah School of Medicine, Salt Lake City, United States
| | - Guillaume Cornelis
- Department of Genetics, Stanford University School of Medicine, Stanford, United States
| | - Marilyn B Renfree
- School of BioSciences, University of Melbourne, Melbourne, Australia
| | - Julie C Baker
- Department of Genetics, Stanford University School of Medicine, Stanford, United States
| |
Collapse
|
42
|
John RM. Imprinted genes and the regulation of placental endocrine function: Pregnancy and beyond. Placenta 2017; 56:86-90. [DOI: 10.1016/j.placenta.2017.01.099] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2016] [Revised: 12/20/2016] [Accepted: 01/09/2017] [Indexed: 12/22/2022]
|
43
|
Abstract
In this review, Prioleau and MacAlpine summarize recent advances in our understanding of how primary sequence, chromatin environment, and nuclear architecture contribute to the dynamic selection and activation of replication origins across diverse cell types and developmental stages. For more than three decades, investigators have sought to identify the precise locations where DNA replication initiates in mammalian genomes. The development of molecular and biochemical approaches to identify start sites of DNA replication (origins) based on the presence of defining and characteristic replication intermediates at specific loci led to the identification of only a handful of mammalian replication origins. The limited number of identified origins prevented a comprehensive and exhaustive search for conserved genomic features that were capable of specifying origins of DNA replication. More recently, the adaptation of origin-mapping assays to genome-wide approaches has led to the identification of tens of thousands of replication origins throughout mammalian genomes, providing an unprecedented opportunity to identify both genetic and epigenetic features that define and regulate their distribution and utilization. Here we summarize recent advances in our understanding of how primary sequence, chromatin environment, and nuclear architecture contribute to the dynamic selection and activation of replication origins across diverse cell types and developmental stages.
Collapse
Affiliation(s)
- Marie-Noëlle Prioleau
- Institut Jacques Monod, UMR7592, Centre National de la Recherche Scientifique, Universite Paris Diderot, Equipe Labellisee Association pour la Recherche sur le Cancer, Paris 75013, France
| | - David M MacAlpine
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina 27710. USA
| |
Collapse
|
44
|
Choi SH, Park JH, Nguyen TTN, Shim HJ, Song YH. Initiation of Drosophila chorion gene amplification requires Claspin and mus101, whereas Claspin, but not mus101, plays a major role during elongation. Dev Dyn 2017; 246:466-474. [PMID: 28294450 PMCID: PMC5435936 DOI: 10.1002/dvdy.24499] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Revised: 02/25/2017] [Accepted: 03/02/2017] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Claspin and TopBP1 are checkpoint mediators that are required for the phosphorylation of Chk1 by ATR to maintain genomic stability. Here, we investigated the functions of Drosophila Claspin and mus101 (TopBP1 ortholog) during chorion (eggshell component) gene amplification, which occurs in follicle cells in the absence of global genomic DNA replication. RESULTS Unlike Drosophila mei-41 (ATR ortholog) mutant embryos, Claspin and mus101 mutant embryos showed severe eggshell defects resulting from defects in chorion gene amplification. EdU (5-ethynyl-2'-deoxyuridine) incorporation assay during initiation and elongation stages revealed that Claspin and mus101 were required for initiation, while only Claspin had a major role in the efficient progression of the replication forks. Claspin proteins were enriched in the amplification foci both in the initiation and elongation stage-follicle cell nuclei in a mei-41-independent manner. The focal localization of ORC2, a component of the origin recognition complex, was not significantly affected in the Claspin mutant, whereas it was reduced in the mus101 mutant. CONCLUSIONS Drosophila Claspin plays a major role in the initiation and elongation stages of chorion gene amplification by localizing to the amplification foci in a mei-41-independent manner. Drosophila mus101 is also involved in chorion gene amplification, mostly functioning in initiation, rather than elongation. Developmental Dynamics 246:466-474, 2016. © 2017 The Authors Developmental Dynamics published by Wiley Periodicals, Inc. on behalf of American Association of Anatomists.
Collapse
Affiliation(s)
- Seung Ho Choi
- Ilsong Institute of Life Science, Hallym University, Anyang, Gyeonggi-do, Republic of Korea
| | - Ji-Hong Park
- Department of Biomedical Gerontology, Hallym University, Chuncheon, Gangwon-do, Republic of Korea
| | - Tram Thi Ngoc Nguyen
- Department of Biomedical Gerontology, Hallym University, Chuncheon, Gangwon-do, Republic of Korea
| | - Hee Jin Shim
- Ilsong Institute of Life Science, Hallym University, Anyang, Gyeonggi-do, Republic of Korea
| | - Young-Han Song
- Ilsong Institute of Life Science, Hallym University, Anyang, Gyeonggi-do, Republic of Korea.,Department of Biomedical Gerontology, Hallym University, Chuncheon, Gangwon-do, Republic of Korea
| |
Collapse
|
45
|
Kasak L, Rull K, Sõber S, Laan M. Copy number variation profile in the placental and parental genomes of recurrent pregnancy loss families. Sci Rep 2017; 7:45327. [PMID: 28345611 PMCID: PMC5366903 DOI: 10.1038/srep45327] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 02/23/2017] [Indexed: 12/25/2022] Open
Abstract
We have previously shown an extensive load of somatic copy number variations (CNVs) in the human placental genome with the highest fraction detected in normal term pregnancies. Hereby, we hypothesized that insufficient promotion of CNVs may impair placental development and lead to recurrent pregnancy loss (RPL). RPL affects ~3% of couples aiming at childbirth and idiopathic RPL represents ~50% of cases. We analysed placental and parental CNV profiles of idiopathic RPL trios (mother-father-placenta) and duos (mother-placenta). Consistent with the hypothesis, the placental genomes of RPL cases exhibited 2-fold less CNVs compared to uncomplicated 1st trimester pregnancies (P = 0.02). This difference mainly arose from lower number of duplications. Overall, 1st trimester control placentas shared only 5.3% of identified CNV regions with RPL cases, whereas the respective fraction with term placentas was 35.1% (P = 1.1 × 10−9). Disruption of the genes NUP98 (embryonic stem cell development) and MTRR (folate metabolism) was detected exclusively in RPL placentas, potentially indicative to novel loci implicated in RPL. Interestingly, genes with higher overall expression were prone to deletions (>3-fold higher median expression compared to genes unaffected by CNVs, P = 6.69 × 10−20). Additionally, large pericentromeric and subtelomeric CNVs in parental genomes emerged as a risk factor for RPL.
Collapse
Affiliation(s)
- Laura Kasak
- Human Molecular Genetics Research Group, Institute of Molecular and Cell Biology, University of Tartu, Riia 23 St., Tartu 51010, Estonia
| | - Kristiina Rull
- Human Molecular Genetics Research Group, Institute of Molecular and Cell Biology, University of Tartu, Riia 23 St., Tartu 51010, Estonia.,Department of Obstetrics and Gynaecology, University of Tartu, Puusepa St. 8, Tartu 51014, Estonia.,Women's Clinic of Tartu University Hospital, Puusepa St. 8, Tartu 51014, Estonia
| | - Siim Sõber
- Human Molecular Genetics Research Group, Institute of Molecular and Cell Biology, University of Tartu, Riia 23 St., Tartu 51010, Estonia.,Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila St. 19, Tartu 51014, Estonia
| | - Maris Laan
- Human Molecular Genetics Research Group, Institute of Molecular and Cell Biology, University of Tartu, Riia 23 St., Tartu 51010, Estonia.,Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila St. 19, Tartu 51014, Estonia
| |
Collapse
|
46
|
Abstract
Evolutionary gene duplication, developmental endoreduplication and selective gene amplification are alternative strategies for increasing gene copy number. When these processes occur together, things get really interesting, and new work shows that is the lifestyle of cells in the placenta.
Collapse
Affiliation(s)
- James C Cross
- Department of Comparative Biology and Experimental Medicine, University of Calgary, Calgary, Alberta, T1S 1A2 Canada.
| |
Collapse
|