1
|
McEvoy CT, MacDonald KD, Shorey-Kendrick LE, Davies MH, Lund KC, Lam R, Dozier BL, Martin LD, Corcoran F, Schelonka RL, Tepper RS, Spindel ER. Nasal CPAP increases alveolar number in a rhesus monkey model of moderate prematurity. Eur Respir J 2025; 65:2400727. [PMID: 39819570 DOI: 10.1183/13993003.00727-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 12/22/2024] [Indexed: 01/19/2025]
Abstract
BACKGROUND Most premature human infants are born in the moderate to late preterm (MLP) range, ≥30 to <37 weeks gestation, and demonstrate increased incidence of wheeze and respiratory illness as they age. Animal models suggest that mechanical lung distention stimulates lung growth and alveolar development. To determine if nasal continuous positive airway pressure (nCPAP) influences MLP infant lung development, we developed a rhesus monkey model of moderate prematurity, randomised to 9 days of nCPAP or sham nCPAP. METHODS Timed-pregnant fetuses were delivered by elective hysterotomy at gestational age (GA) 140±1 days (85% gestation; term=165 days; human equivalent of 32-34 weeks) or at GA-149±1 days as a relative gestational age reference (GA-control). The day after delivery, the GA-140 animals were treated with nCPAP or sham nCPAP for 9 days, 12 consecutive hours each day. Pulmonary function testing followed by necropsy for analysis of lung structure and gene expression was performed on the equivalent of GA-150 for all animals. RESULTS The nCPAP and sham groups were clinically similar but distinct from the GA-control group. Stereological analysis of lung structure showed significantly increased numbers of alveoli in the nCPAP group compared to the sham group. Other functional and anatomical changes were consistent with increased alveolarisation. Gene expression between the nCPAP and sham groups remained highly similar and distinct from GA-control animals. CONCLUSIONS We show that nCPAP in MLP infants stimulates alveolarisation with relatively few other changes. How this may benefit subsequent infant respiratory health requires further study.
Collapse
Affiliation(s)
- Cindy T McEvoy
- Department of Pediatrics, Oregon Health and Science University, Portland, OR, USA
| | - Kelvin D MacDonald
- Department of Pediatrics, Oregon Health and Science University, Portland, OR, USA
| | | | - Michael H Davies
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR, USA
| | - Kelli C Lund
- Department of Pediatrics, University of Utah Health, Salt Lake City, UT, USA
| | - Ryan Lam
- Department of Neonatology, Salem Hospital, Salem, OR, USA
| | - Brandy L Dozier
- Division of Comparative Medicine, Oregon National Primate Research Center, Beaverton, OR, USA
| | - Lauren Drew Martin
- Division of Comparative Medicine, Oregon National Primate Research Center, Beaverton, OR, USA
| | - Fiona Corcoran
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR, USA
| | - Robert L Schelonka
- Department of Pediatrics, Oregon Health and Science University, Portland, OR, USA
| | - Robert S Tepper
- Department of Pediatrics, H.B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
- R.S. Tepper and E.R. Spindel made equal contributions as senior authors
| | - Eliot R Spindel
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR, USA
- R.S. Tepper and E.R. Spindel made equal contributions as senior authors
| |
Collapse
|
2
|
Pierson Smela MD, Kramme CC, Fortuna PRJ, Adams JL, Su R, Dong E, Kobayashi M, Brixi G, Kavirayuni VS, Tysinger E, Kohman RE, Shioda T, Chatterjee P, Church GM. Directed differentiation of human iPSCs to functional ovarian granulosa-like cells via transcription factor overexpression. eLife 2023; 12:e83291. [PMID: 36803359 PMCID: PMC9943069 DOI: 10.7554/elife.83291] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 01/18/2023] [Indexed: 02/22/2023] Open
Abstract
An in vitro model of human ovarian follicles would greatly benefit the study of female reproduction. Ovarian development requires the combination of germ cells and several types of somatic cells. Among these, granulosa cells play a key role in follicle formation and support for oogenesis. Whereas efficient protocols exist for generating human primordial germ cell-like cells (hPGCLCs) from human induced pluripotent stem cells (hiPSCs), a method of generating granulosa cells has been elusive. Here, we report that simultaneous overexpression of two transcription factors (TFs) can direct the differentiation of hiPSCs to granulosa-like cells. We elucidate the regulatory effects of several granulosa-related TFs and establish that overexpression of NR5A1 and either RUNX1 or RUNX2 is sufficient to generate granulosa-like cells. Our granulosa-like cells have transcriptomes similar to human fetal ovarian cells and recapitulate key ovarian phenotypes including follicle formation and steroidogenesis. When aggregated with hPGCLCs, our cells form ovary-like organoids (ovaroids) and support hPGCLC development from the premigratory to the gonadal stage as measured by induction of DAZL expression. This model system will provide unique opportunities for studying human ovarian biology and may enable the development of therapies for female reproductive health.
Collapse
Affiliation(s)
- Merrick D Pierson Smela
- Wyss Institute, Harvard UniversityBostonUnited States
- Department of Genetics, Harvard Medical SchoolBostonUnited States
| | - Christian C Kramme
- Wyss Institute, Harvard UniversityBostonUnited States
- Department of Genetics, Harvard Medical SchoolBostonUnited States
| | - Patrick RJ Fortuna
- Wyss Institute, Harvard UniversityBostonUnited States
- Department of Genetics, Harvard Medical SchoolBostonUnited States
| | - Jessica L Adams
- Wyss Institute, Harvard UniversityBostonUnited States
- Department of Genetics, Harvard Medical SchoolBostonUnited States
| | - Rui Su
- Wyss Institute, Harvard UniversityBostonUnited States
- Department of Genetics, Harvard Medical SchoolBostonUnited States
| | - Edward Dong
- Wyss Institute, Harvard UniversityBostonUnited States
- Department of Genetics, Harvard Medical SchoolBostonUnited States
| | - Mutsumi Kobayashi
- Massachusetts General Hospital Center for Cancer Research, Harvard Medical SchoolCharlestownUnited States
| | - Garyk Brixi
- Wyss Institute, Harvard UniversityBostonUnited States
- Department of Genetics, Harvard Medical SchoolBostonUnited States
- Department of Biomedical Engineering, Duke UniversityDurhamUnited States
- Department of Computer Science, Duke UniversityDurhamUnited States
| | - Venkata Srikar Kavirayuni
- Department of Biomedical Engineering, Duke UniversityDurhamUnited States
- Department of Computer Science, Duke UniversityDurhamUnited States
| | - Emma Tysinger
- Department of Biomedical Engineering, Duke UniversityDurhamUnited States
- Department of Computer Science, Duke UniversityDurhamUnited States
| | - Richie E Kohman
- Wyss Institute, Harvard UniversityBostonUnited States
- Department of Genetics, Harvard Medical SchoolBostonUnited States
| | - Toshi Shioda
- Massachusetts General Hospital Center for Cancer Research, Harvard Medical SchoolCharlestownUnited States
| | - Pranam Chatterjee
- Wyss Institute, Harvard UniversityBostonUnited States
- Department of Genetics, Harvard Medical SchoolBostonUnited States
- Department of Biomedical Engineering, Duke UniversityDurhamUnited States
- Department of Computer Science, Duke UniversityDurhamUnited States
| | - George M Church
- Wyss Institute, Harvard UniversityBostonUnited States
- Department of Genetics, Harvard Medical SchoolBostonUnited States
| |
Collapse
|
3
|
Munyoki SK, Orwig KE. Perspectives: Methods for Evaluating Primate Spermatogonial Stem Cells. Methods Mol Biol 2023; 2656:341-364. [PMID: 37249880 DOI: 10.1007/978-1-0716-3139-3_18] [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: 05/31/2023]
Abstract
Mammalian spermatogenesis is a complex, highly productive process generating millions of sperm per day. Spermatogonial stem cells (SSCs) are at the foundation of spermatogenesis and can either self-renew, producing more SSCs, or differentiate to initiate spermatogenesis and produce sperm. The biological potential of SSCs to produce and maintain spermatogenesis makes them a promising tool for the treatment of male infertility. However, translating knowledge from rodents to higher primates (monkeys and humans) is challenged by different vocabularies that are used to describe stem cells and spermatogenic lineage development in those species. Furthermore, while rodent SSCs are defined by their biological potential to produce and maintain spermatogenesis in a transplant assay, there is no equivalent routine and accessible bioassay to test monkey and human SSCs or replicate their functions in vitro. This chapter describes progress characterizing, isolating, culturing, and transplanting SSCs in higher primates.
Collapse
Affiliation(s)
- Sarah K Munyoki
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Integrative Systems Biology Graduate Program, Magee-Women's Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Kyle E Orwig
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Integrative Systems Biology Graduate Program, Magee-Women's Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
4
|
Cheng H, Shang D, Zhou R. Germline stem cells in human. Signal Transduct Target Ther 2022; 7:345. [PMID: 36184610 PMCID: PMC9527259 DOI: 10.1038/s41392-022-01197-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 09/06/2022] [Accepted: 09/14/2022] [Indexed: 12/02/2022] Open
Abstract
The germline cells are essential for the propagation of human beings, thus essential for the survival of mankind. The germline stem cells, as a unique cell type, generate various states of germ stem cells and then differentiate into specialized cells, spermatozoa and ova, for producing offspring, while self-renew to generate more stem cells. Abnormal development of germline stem cells often causes severe diseases in humans, including infertility and cancer. Primordial germ cells (PGCs) first emerge during early embryonic development, migrate into the gentile ridge, and then join in the formation of gonads. In males, they differentiate into spermatogonial stem cells, which give rise to spermatozoa via meiosis from the onset of puberty, while in females, the female germline stem cells (FGSCs) retain stemness in the ovary and initiate meiosis to generate oocytes. Primordial germ cell-like cells (PGCLCs) can be induced in vitro from embryonic stem cells or induced pluripotent stem cells. In this review, we focus on current advances in these embryonic and adult germline stem cells, and the induced PGCLCs in humans, provide an overview of molecular mechanisms underlying the development and differentiation of the germline stem cells and outline their physiological functions, pathological implications, and clinical applications.
Collapse
Affiliation(s)
- Hanhua Cheng
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Renmin Hospital of Wuhan University, Wuhan University, 430072, Wuhan, China.
| | - Dantong Shang
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Renmin Hospital of Wuhan University, Wuhan University, 430072, Wuhan, China
| | - Rongjia Zhou
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Renmin Hospital of Wuhan University, Wuhan University, 430072, Wuhan, China.
| |
Collapse
|
5
|
Mitsunaga S, Shioda K, Hanna JH, Isselbacher KJ, Shioda T. Production and Analysis of Human Primordial Germ Cell-Like Cells. Methods Mol Biol 2021; 2195:125-145. [PMID: 32852762 PMCID: PMC11804835 DOI: 10.1007/978-1-0716-0860-9_10] [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: 07/31/2023]
Abstract
Primordial germ cells (PGCs) are common ancestors of all germline cells. In mammals, PGCs emerge in early-stage embryos around the timing of gastrulation at or near epiblast, and specification of PGCs from their precursor cells involves multiple growth factors secreted by adjacent cells. Recent advancements in germline stem cell biology have made it possible to generate PGC-like cell culture models (PGCLCs for PGC-like cells) from human and mouse pluripotent stem cells by mimicking the embryonic growth factor environment in vitro. Here we describe a method of producing human PGCLCs from primed-pluripotency induced pluripotent stem cells (iPSCs) via temporal conversion to naive pluripotency followed by formation of embryoid bodies (EBs) using the spin-EB method.
Collapse
Affiliation(s)
- Shino Mitsunaga
- Center for Cancer Research, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Keiko Shioda
- Center for Cancer Research, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Jacob H Hanna
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Kurt J Isselbacher
- Center for Cancer Research, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Toshi Shioda
- Center for Cancer Research, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA.
| |
Collapse
|
6
|
Abdyyev VK, Sant DW, Kiseleva EV, Spangenberg VE, Kolomiets OL, Andrade NS, Dashinimaev EB, Vorotelyak EA, Vasiliev AV. In vitro derived female hPGCLCs are unable to complete meiosis in embryoid bodies. Exp Cell Res 2020; 397:112358. [PMID: 33160998 DOI: 10.1016/j.yexcr.2020.112358] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 10/27/2020] [Accepted: 10/31/2020] [Indexed: 11/19/2022]
Abstract
The fundamental question about the functionality of in vitro derived human primordial germ cell-like cells remains unanswered, despite ongoing research in this area. Attempts have been made to imitate the differentiation of human primordial germ cells (hPGCs) and meiocytes in vitro from human pluripotent stem cells (hPSCs). A defined system for developing human haploid cells in vitro is the challenge that scientists face to advance the knowledge of human germ cell development. To develop human primordial germ cell-like cells (hPGCLCs) from human pluripotent stem cells (hPSCs) that are capable of giving rise to haploid cells, we applied a sequential induction protocol via the early mesodermal push of female human embryonic and induced pluripotent stem cells. BMP4-induced early mesoderm-like cells showed significant alterations in their expression profiles toward early (PRDM1 and NANOS3) and late (VASA and DAZL) germ cell markers. Furthermore, using retinoic acid (RA), we induced hPGCLCs in embryoid bodies and identified positive staining for the meiotic initiation marker STRA8. Efforts to find the cells exhibiting progression to meiosis were unsuccessful. The validation by the expression of SCP3 did not correspond to the natural pattern. Regarding the 20-day meiotic induction, the derived hPGCLCs containing two X-chromosomes were unable to complete the meiotic division. We observed the expression of the oocyte marker PIWIL1 and PIWIL4. RNAseq analysis and cluster dendrogram showed a similar clustering of hPGCLC groups and meiotic like cell groups as compared to previously published data. This reproducible in vitro model for deriving hPGCLCs provides opportunities for studying the molecular mechanisms involved in the specification of hPGCs. Moreover, our results will support a further elucidation of gametogenesis and meiosis of female hPGCs.
Collapse
Affiliation(s)
- Vepa K Abdyyev
- Koltzov Institute of Developmental Biology of the Russian Academy of Sciences, Moscow, Russia; Department of Biology, Lomonosov Moscow State University, Moscow, Russia.
| | - David W Sant
- Department of Biomedical Informatics, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Ekaterina V Kiseleva
- Koltzov Institute of Developmental Biology of the Russian Academy of Sciences, Moscow, Russia
| | - Victor E Spangenberg
- Vavilov Institute of General Genetics, The Russian Academy of Sciences, Moscow, Russia
| | - Oksana L Kolomiets
- Vavilov Institute of General Genetics, The Russian Academy of Sciences, Moscow, Russia
| | - Nadja S Andrade
- Department of Psychiatry and Behavioral Studies, Center for Therapeutic Innovation, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Erdem B Dashinimaev
- Koltzov Institute of Developmental Biology of the Russian Academy of Sciences, Moscow, Russia; Pirogov Russian National Research Medical University (RNRMU), Moscow, Russia
| | - Ekaterina A Vorotelyak
- Koltzov Institute of Developmental Biology of the Russian Academy of Sciences, Moscow, Russia; Department of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Andrei V Vasiliev
- Koltzov Institute of Developmental Biology of the Russian Academy of Sciences, Moscow, Russia; Department of Biology, Lomonosov Moscow State University, Moscow, Russia
| |
Collapse
|
7
|
Mall EM, Rotte N, Yoon J, Sandhowe-Klaverkamp R, Röpke A, Wistuba J, Hübner K, Schöler HR, Schlatt S. A novel xeno-organoid approach: exploring the crosstalk between human iPSC-derived PGC-like and rat testicular cells. Mol Hum Reprod 2020; 26:879-893. [PMID: 33049038 DOI: 10.1093/molehr/gaaa067] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 09/11/2020] [Indexed: 02/06/2023] Open
Abstract
Specification of germ cell-like cells from induced pluripotent stem cells has become a clinically relevant tool for research. Research on initial embryonic processes is often limited by the access to foetal tissue, and in humans, the molecular events resulting in primordial germ cell (PGC) specification and sex determination remain to be elucidated. A deeper understanding of the underlying processes is crucial to describe pathomechanisms leading to impaired reproductive function. Several protocols have been established for the specification of human pluripotent stem cell towards early PGC-like cells (PGCLC), currently representing the best model to mimic early human germline developmental processes in vitro. Further sex determination towards the male lineage depends on somatic gonadal cells providing the necessary molecular cues. By establishing a culture system characterized by the re-organization of somatic cells from postnatal rat testes into cord-like structures and optimizing efficient PGCLC specification protocols, we facilitated the co-culture of human germ cell-like cells within a surrogate testicular microenvironment. Specified conditions allowed the survival of rat somatic testicular and human PGCLCs for 14 days. Human cells maintained the characteristic expression of octamer-binding transcription factor 4, SRY-box transcription factor 17, and transcription factor AP-2 gamma and were recovered from the xeno-organoids by cell sorting. This novel xeno-organoid approach will allow the in vitro exploration of early sex determination of human PGCLCs.
Collapse
Affiliation(s)
- E M Mall
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - N Rotte
- Centre of Reproductive Medicine and Andrology, University of Münster, Münster, Germany.,Institute of Reproductive Genetics, University of Münster, Münster, Germany
| | - J Yoon
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - R Sandhowe-Klaverkamp
- Centre of Reproductive Medicine and Andrology, University of Münster, Münster, Germany
| | - A Röpke
- Institute of Human Genetics, University of Münster, Münster, Germany
| | - J Wistuba
- Centre of Reproductive Medicine and Andrology, University of Münster, Münster, Germany
| | - K Hübner
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - H R Schöler
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Münster, Germany.,Medical Faculty, University of Münster, Münster, Germany
| | - S Schlatt
- Centre of Reproductive Medicine and Andrology, University of Münster, Münster, Germany
| |
Collapse
|
8
|
Rohani L, Borys BS, Razian G, Naghsh P, Liu S, Johnson AA, Machiraju P, Holland H, Lewis IA, Groves RA, Toms D, Gordon PMK, Li JW, So T, Dang T, Kallos MS, Rancourt DE. Stirred suspension bioreactors maintain naïve pluripotency of human pluripotent stem cells. Commun Biol 2020; 3:492. [PMID: 32895477 PMCID: PMC7476926 DOI: 10.1038/s42003-020-01218-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 08/03/2020] [Indexed: 11/11/2022] Open
Abstract
Due to their ability to standardize key physiological parameters, stirred suspension bioreactors can potentially scale the production of quality-controlled pluripotent stem cells (PSCs) for cell therapy application. Because of differences in bioreactor expansion efficiency between mouse (m) and human (h) PSCs, we investigated if conversion of hPSCs, from the conventional "primed" pluripotent state towards the "naïve" state prevalent in mPSCs, could be used to enhance hPSC production. Through transcriptomic enrichment of mechano-sensing signaling, the expression of epigenetic regulators, metabolomics, and cell-surface protein marker analyses, we show that the stirred suspension bioreactor environment helps maintain a naïve-like pluripotent state. Our research corroborates that converting hPSCs towards a naïve state enhances hPSC manufacturing and indicates a potentially important role for the stirred suspension bioreactor's mechanical environment in maintaining naïve-like pluripotency.
Collapse
Affiliation(s)
- Leili Rohani
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Breanna S Borys
- Pharmaceutical Production Research Facility, Schulich School of Engineering, University of Calgary, Calgary, AB, Canada
- Biomedical Engineering Graduate Program, University of Calgary, Calgary, AB, Canada
| | - Golsa Razian
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Pooyan Naghsh
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Shiying Liu
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | | | - Pranav Machiraju
- Department of Paediatrics and Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Heidrun Holland
- Saxonian Incubator for Clinical Translation (SIKT), University of Leipzig, Leipzig, Germany
| | - Ian A Lewis
- Department of Biological Sciences, University of Calgary, Calgary, AB, Canada
| | - Ryan A Groves
- Department of Biological Sciences, University of Calgary, Calgary, AB, Canada
| | - Derek Toms
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - Paul M K Gordon
- CSM Center for Health Genomic and Informatics, University of Calgary, Calgary, AB, Canada
| | - Joyce W Li
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Tania So
- Pharmaceutical Production Research Facility, Schulich School of Engineering, University of Calgary, Calgary, AB, Canada
- Biomedical Engineering Graduate Program, University of Calgary, Calgary, AB, Canada
| | - Tiffany Dang
- Pharmaceutical Production Research Facility, Schulich School of Engineering, University of Calgary, Calgary, AB, Canada
- Biomedical Engineering Graduate Program, University of Calgary, Calgary, AB, Canada
| | - Michael S Kallos
- Pharmaceutical Production Research Facility, Schulich School of Engineering, University of Calgary, Calgary, AB, Canada
- Biomedical Engineering Graduate Program, University of Calgary, Calgary, AB, Canada
| | - Derrick E Rancourt
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
9
|
Chen D, Liu W, Zimmerman J, Pastor WA, Kim R, Hosohama L, Ho J, Aslanyan M, Gell JJ, Jacobsen SE, Clark AT. The TFAP2C-Regulated OCT4 Naive Enhancer Is Involved in Human Germline Formation. Cell Rep 2019; 25:3591-3602.e5. [PMID: 30590035 PMCID: PMC6342560 DOI: 10.1016/j.celrep.2018.12.011] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 11/15/2018] [Accepted: 12/03/2018] [Indexed: 12/21/2022] Open
Abstract
Human primordial germ cells (hPGCs) are the first embryonic progenitors in the germ cell lineage, yet the molecular mechanisms required for hPGC formation are not well characterized. To identify regulatory regions in hPGC development, we used the assay for transposase-accessible chromatin using sequencing (ATAC-seq) to systematically characterize regions of open chromatin in hPGCs and hPGC-like cells (hPGCLCs) differentiated from human embryonic stem cells (hESCs). We discovered regions of open chromatin unique to hPGCs and hPGCLCs that significantly overlap with TFAP2C-bound enhancers identified in the naive ground state of pluripotency. Using CRISPR/Cas9, we show that deleting the TFAP2C-bound naive enhancer at the OCT4 locus (also called POU5F1) results in impaired OCT4 expression and a negative effect on hPGCLC identity. Combining genomics and functional studies, Chen et al. identify the open chromatin state of human primordial germ cells (hPGCs), leading to the discovery that TFAP2C regulates hPGC development through the opening of naive enhancers.
Collapse
Affiliation(s)
- Di Chen
- Department of Molecular Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Wanlu Liu
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, USA
| | - Jill Zimmerman
- Department of Molecular Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA, USA
| | - William A Pastor
- Department of Molecular Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Rachel Kim
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, USA
| | - Linzi Hosohama
- Department of Molecular Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Jamie Ho
- Department of Molecular Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Marianna Aslanyan
- Department of Molecular Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Joanna J Gell
- Department of Pediatrics, Division of Hematology-Oncology, Los Angeles, CA 90095, USA; David Geffen School of Medicine, Los Angeles, CA 90095, USA
| | - Steven E Jacobsen
- Department of Molecular Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, USA; Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, USA; Howard Hughes Medical Institute, University of California, Los Angeles, Los Angeles, CA, USA
| | - Amander T Clark
- Department of Molecular Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, USA; Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
10
|
Niu Y, Sun N, Li C, Lei Y, Huang Z, Wu J, Si C, Dai X, Liu C, Wei J, Liu L, Feng S, Kang Y, Si W, Wang H, Zhang E, Zhao L, Li Z, Luo X, Cui G, Peng G, Izpisúa Belmonte JC, Ji W, Tan T. Dissecting primate early post-implantation development using long-term in vitro embryo culture. Science 2019; 366:science.aaw5754. [DOI: 10.1126/science.aaw5754] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 10/16/2019] [Indexed: 12/12/2022]
Abstract
The transition from peri-implantation to gastrulation in mammals entails the specification and organization of the lineage progenitors into a body plan. Technical and ethical challenges have limited understanding of the cellular and molecular mechanisms that underlie this transition. We established a culture system that enabled the development of cynomolgus monkey embryos in vitro for up to 20 days. Cultured embryos underwent key primate developmental stages, including lineage segregation, bilaminar disc formation, amniotic and yolk sac cavitation, and primordial germ cell–like cell (PGCLC) differentiation. Single-cell RNA-sequencing analysis revealed development trajectories of primitive endoderm, trophectoderm, epiblast lineages, and PGCLCs. Analysis of single-cell chromatin accessibility identified transcription factors specifying each cell type. Our results reveal critical developmental events and complex molecular mechanisms underlying nonhuman primate embryogenesis in the early postimplantation period, with possible relevance to human development.
Collapse
Affiliation(s)
- Yuyu Niu
- Yunnan Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
| | - Nianqin Sun
- Yunnan Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
| | - Chang Li
- Yunnan Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
| | - Ying Lei
- BGI-Shenzhen, Shenzhen, Guangdong 518083, China
- China National GeneBank, BGI-Shenzhen, Shenzhen, Guangdong 518120, China
| | - Zhihao Huang
- BGI-Shenzhen, Shenzhen, Guangdong 518083, China
- China National GeneBank, BGI-Shenzhen, Shenzhen, Guangdong 518120, China
| | - Jun Wu
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Chenyang Si
- Yunnan Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
| | - Xi Dai
- BGI-Shenzhen, Shenzhen, Guangdong 518083, China
- China National GeneBank, BGI-Shenzhen, Shenzhen, Guangdong 518120, China
| | - Chuanyu Liu
- BGI-Shenzhen, Shenzhen, Guangdong 518083, China
- China National GeneBank, BGI-Shenzhen, Shenzhen, Guangdong 518120, China
| | - Jingkuan Wei
- Yunnan Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
| | - Longqi Liu
- BGI-Shenzhen, Shenzhen, Guangdong 518083, China
- China National GeneBank, BGI-Shenzhen, Shenzhen, Guangdong 518120, China
| | - Su Feng
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences (CAS), University of Chinese Academy of Sciences, Shanghai 200032, China
| | - Yu Kang
- Yunnan Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
| | - Wei Si
- Yunnan Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
| | - Hong Wang
- Yunnan Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
| | - E. Zhang
- Yunnan Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
| | - Lu Zhao
- Yunnan Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
| | - Ziwei Li
- Yunnan Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
| | - Xi Luo
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, CAS, Guangzhou 510530, China
| | - Guizhong Cui
- Center of Cell Lineage and Atlas, Guangzhou Regenerative Medicine and Health Guangdong Laboratory (GRMH-GDL), Guangzhou 510530, China
| | - Guangdun Peng
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, CAS, Guangzhou 510530, China
- Center of Cell Lineage and Atlas, Guangzhou Regenerative Medicine and Health Guangdong Laboratory (GRMH-GDL), Guangzhou 510530, China
| | | | - Weizhi Ji
- Yunnan Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
- CAS Center for Excellence in Brain Science and Intelligence Technology, CAS, Shanghai 200032, China
| | - Tao Tan
- Yunnan Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
| |
Collapse
|
11
|
Sosa E, Chen D, Rojas EJ, Hennebold JD, Peters KA, Wu Z, Lam TN, Mitchell JM, Sukhwani M, Tailor RC, Meistrich ML, Orwig KE, Shetty G, Clark AT. Differentiation of primate primordial germ cell-like cells following transplantation into the adult gonadal niche. Nat Commun 2018; 9:5339. [PMID: 30559363 PMCID: PMC6297357 DOI: 10.1038/s41467-018-07740-7] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 11/16/2018] [Indexed: 11/17/2022] Open
Abstract
A major challenge in stem cell differentiation is the availability of bioassays to prove cell types generated in vitro are equivalent to cells in vivo. In the mouse, differentiation of primordial germ cell-like cells (PGCLCs) from pluripotent cells was validated by transplantation, leading to the generation of spermatogenesis and to the birth of offspring. Here we report the use of xenotransplantation (monkey to mouse) and homologous transplantation (monkey to monkey) to validate our in vitro protocol for differentiating male rhesus (r) macaque PGCLCs (rPGCLCs) from induced pluripotent stem cells (riPSCs). Specifically, transplantation of aggregates containing rPGCLCs into mouse and nonhuman primate testicles overcomes a major bottleneck in rPGCLC differentiation. These findings suggest that immature rPGCLCs once transplanted into an adult gonadal niche commit to differentiate towards late rPGCs that initiate epigenetic reprogramming but do not complete the conversion into ENO2-positive spermatogonia. Human embryonic stem cells can be differentiated in vitro into primordial germ cell-like cells (PGCLCs) that resemble early primordial germ cells (PGCs). Here the authors transplant PGCLCs generated from rhesus macaque iPSCs into mouse and rhesus macaque seminiferous tubules, which matures these into late PGCs and spermatogonia-like cells.
Collapse
Affiliation(s)
- Enrique Sosa
- Department of Molecular, Cell and Developmental Biology, Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Di Chen
- Department of Molecular, Cell and Developmental Biology, Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Ernesto J Rojas
- Department of Molecular, Cell and Developmental Biology, Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Jon D Hennebold
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Beaverton, OR, 97006, USA.,Department of Obstetrics and Gynecology, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Karen A Peters
- Department of Obstetrics, Gynecology and Reproductive Sciences and Magee Women's Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Zhuang Wu
- Department of Experimental Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Truong N Lam
- Department of Experimental Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Jennifer M Mitchell
- Department of Veterinary Medicine and Surgery, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Meena Sukhwani
- Department of Obstetrics, Gynecology and Reproductive Sciences and Magee Women's Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Ramesh C Tailor
- Department of Radiation Physics, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Marvin L Meistrich
- Department of Experimental Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Kyle E Orwig
- Department of Obstetrics, Gynecology and Reproductive Sciences and Magee Women's Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Gunapala Shetty
- Department of Experimental Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Amander T Clark
- Department of Molecular, Cell and Developmental Biology, Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
| |
Collapse
|
12
|
Mitsunaga S, Shioda T. Evolutionarily diverse mechanisms of germline specification among mammals: what about us? Stem Cell Investig 2018; 5:12. [PMID: 29782583 PMCID: PMC5945857 DOI: 10.21037/sci.2018.04.03] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 03/31/2018] [Indexed: 01/03/2023]
Affiliation(s)
- Shino Mitsunaga
- Massachusetts General Hospital Center for Cancer Research, Charlestown, MA, USA
| | - Toshi Shioda
- Massachusetts General Hospital Center for Cancer Research, Charlestown, MA, USA
- Harvard Medical School, Boston, MA, USA
| |
Collapse
|
13
|
Fayomi AP, Orwig KE. Spermatogonial stem cells and spermatogenesis in mice, monkeys and men. Stem Cell Res 2018; 29:207-214. [PMID: 29730571 PMCID: PMC6010318 DOI: 10.1016/j.scr.2018.04.009] [Citation(s) in RCA: 212] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 04/10/2018] [Accepted: 04/13/2018] [Indexed: 12/12/2022] Open
Abstract
Continuous spermatogenesis in post-pubertal mammals is dependent on spermatogonial stem cells (SSCs), which balance self-renewing divisions that maintain stem cell pool with differentiating divisions that sustain continuous sperm production. Rodent stem and progenitor spermatogonia are described by their clonal arrangement in the seminiferous epithelium (e.g., Asingle, Apaired or Aaligned spermatogonia), molecular markers (e.g., ID4, GFRA1, PLZF, SALL4 and others) and most importantly by their biological potential to produce and maintain spermatogenesis when transplanted into recipient testes. In contrast, stem cells in the testes of higher primates (nonhuman and human) are defined by description of their nuclear morphology and staining with hematoxylin as Adark and Apale spermatogonia. There is limited information about how dark and pale descriptions of nuclear morphology in higher primates correspond with clone size, molecular markers or transplant potential. Do the apparent differences in stem cells and spermatogenic lineage development between rodents and primates represent true biological differences or simply differences in the volume of research and the vocabulary that has developed over the past half century? This review will provide an overview of stem, progenitor and differentiating spermatogonia that support spermatogenesis; identifying parallels between rodents and primates where they exist as well as features unique to higher primates.
Collapse
Affiliation(s)
- Adetunji P Fayomi
- Molecular Genetics and Developmental Biology Graduate Program, Department of Obstetrics, Gynecology and Reproductive Sciences, Magee-Womens Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, United States
| | - Kyle E Orwig
- Molecular Genetics and Developmental Biology Graduate Program, Department of Obstetrics, Gynecology and Reproductive Sciences, Magee-Womens Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, United States.
| |
Collapse
|
14
|
Deriving Dorsal Spinal Sensory Interneurons from Human Pluripotent Stem Cells. Stem Cell Reports 2018; 10:390-405. [PMID: 29337120 PMCID: PMC5832443 DOI: 10.1016/j.stemcr.2017.12.012] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 12/14/2017] [Accepted: 12/15/2017] [Indexed: 12/28/2022] Open
Abstract
Cellular replacement therapies for neurological conditions use human embryonic stem cell (hESC)- or induced pluripotent stem cell (hiPSC)-derived neurons to replace damaged or diseased populations of neurons. For the spinal cord, significant progress has been made generating the in-vitro-derived motor neurons required to restore coordinated movement. However, there is as yet no protocol to generate in-vitro-derived sensory interneurons (INs), which permit perception of the environment. Here, we report on the development of a directed differentiation protocol to derive sensory INs for both hESCs and hiPSCs. Two developmentally relevant factors, retinoic acid in combination with bone morphogenetic protein 4, can be used to generate three classes of sensory INs: the proprioceptive dI1s, the dI2s, and mechanosensory dI3s. Critical to this protocol is the competence state of the neural progenitors, which changes over time. This protocol will facilitate developing cellular replacement therapies to reestablish sensory connections in injured patients. Robust protocol to generate spinal sensory neurons from human pluripotent cells RA ± BMP4 direct hPSCs toward the dI1, dI2, and dI3 classes of dorsal interneurons Only neural progenitors in the correct competence state respond to RA/BMP4 signals
Collapse
|
15
|
Chen D, Liu W, Lukianchikov A, Hancock GV, Zimmerman J, Lowe MG, Kim R, Galic Z, Irie N, Surani MA, Jacobsen SE, Clark AT. Germline competency of human embryonic stem cells depends on eomesodermin. Biol Reprod 2017; 97:850-861. [PMID: 29091993 PMCID: PMC5803789 DOI: 10.1093/biolre/iox138] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 10/27/2017] [Indexed: 12/11/2022] Open
Abstract
In humans, germline competency and the specification of primordial germ cells (PGCs) are thought to occur in a restricted developmental window during early embryogenesis. Despite the importance of specifying the appropriate number of PGCs for human reproduction, the molecular mechanisms governing PGC formation remain largely unexplored. Here, we compared PGC-like cell (PGCLC) differentiation from 18 independently derived human embryonic stem cell (hESC) lines, and discovered that the expression of primitive streak genes were positively associated with hESC germline competency. Furthermore, we show that chemical inhibition of TGFβ and WNT signaling, which are required for primitive streak formation and CRISPR/Cas9 deletion of Eomesodermin (EOMES), significantly impacts PGCLC differentiation from hESCs. Taken together, our results suggest that human PGC formation involves signaling and transcriptional programs associated with somatic germ layer induction and expression of EOMES.
Collapse
Affiliation(s)
- Di Chen
- Department of Molecular Cell and Developmental Biology, University of California, Los Angeles, California, USA
| | - Wanlu Liu
- Molecular Biology Institute, University of California, Los Angeles, California, USA
| | - Anastasia Lukianchikov
- Department of Molecular Cell and Developmental Biology, University of California, Los Angeles, California, USA
| | - Grace V Hancock
- Department of Molecular Cell and Developmental Biology, University of California, Los Angeles, California, USA
- Molecular Biology Institute, University of California, Los Angeles, California, USA
| | - Jill Zimmerman
- Department of Molecular Cell and Developmental Biology, University of California, Los Angeles, California, USA
| | - Matthew G Lowe
- Department of Molecular Cell and Developmental Biology, University of California, Los Angeles, California, USA
- Molecular Biology Institute, University of California, Los Angeles, California, USA
| | - Rachel Kim
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, California, USA
| | - Zoran Galic
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, California, USA
- Department of Medicine, University of California, Los Angeles, California, USA
| | - Naoko Irie
- Wellcome Trust Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge, UK
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Wellcome Trust-Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - M Azim Surani
- Wellcome Trust Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge, UK
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Wellcome Trust-Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Steven E Jacobsen
- Department of Molecular Cell and Developmental Biology, University of California, Los Angeles, California, USA
- Molecular Biology Institute, University of California, Los Angeles, California, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, California, USA
- Department of Biological Chemistry, University of California, Los Angeles, California, USA
- Howard Hughes Medical Institute, University of California, Los Angeles, California, USA
| | - Amander T Clark
- Department of Molecular Cell and Developmental Biology, University of California, Los Angeles, California, USA
- Molecular Biology Institute, University of California, Los Angeles, California, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, California, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, California, USA
| |
Collapse
|