1
|
Chen D, Zhi Y, Jia X, Ji N, Zhang J, Liang J, Ran M, Liu X, Xu H, Lu Y. Characterization of primordial germ cells from EG&K stage X chicken embryos. Poult Sci 2025; 104:105308. [PMID: 40451075 PMCID: PMC12163151 DOI: 10.1016/j.psj.2025.105308] [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: 02/11/2025] [Revised: 05/15/2025] [Accepted: 05/15/2025] [Indexed: 06/16/2025] Open
Abstract
Primordial germ cells (PGCs) derived from Eyal-Giladi and Kochav (EG&K) stage X embryos (XPGCs) represent the earliest germ cells in poultry. However, little is known about the XPGCs due the difficulty in the isolation and propagation of these cells. In this study, we report a method for the stable isolation and efficient in vitro propagation of XPGCs. We conducted a thorough analysis of the characteristics exhibited by XPGCs. First, we identified an average of 42 PGCs at this stage by using an DAZL-2a-mCherry genetically modified chickens. Three stable cell lines, one male and two female, were established from eight isolates using a filter paper ring and differential adhesion. Germ cell-related protein markers, including DAZL, CVH, and SSEA-1, were detected in the established XPGCs. Re-introduction of the XPGCs into the blastoderms of EG&K stage X embryos or the cardiovascular systems of Hamilton and Hamburger (HH) stage 14 embryos showed that the XPGCs retained the ability to migrate to the gonads after long-term culture. XPGCs demonstrated a lower growth rate in vitro but a higher potential of migrating to the gonads than PGCs derived from the embryonic gonads of HH stage 27-31. The ability to isolate, culture, and characterize XPGCs advances our knowledge of early-stage germ cell development and provides a valuable cell tool for genetic conservation and genome editing in chickens.
Collapse
Affiliation(s)
- Dongyang Chen
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, College of Animal Science and Technology, Guangxi University, Nanning, 530004 PR China
| | - Yifei Zhi
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, College of Animal Science and Technology, Guangxi University, Nanning, 530004 PR China
| | - Xiaoxuan Jia
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, College of Animal Science and Technology, Guangxi University, Nanning, 530004 PR China
| | - Na Ji
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, College of Animal Science and Technology, Guangxi University, Nanning, 530004 PR China
| | - Jiale Zhang
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, College of Animal Science and Technology, Guangxi University, Nanning, 530004 PR China
| | - Jiongming Liang
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, College of Animal Science and Technology, Guangxi University, Nanning, 530004 PR China
| | - Mingxia Ran
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, College of Animal Science and Technology, Guangxi University, Nanning, 530004 PR China
| | - Xingting Liu
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, College of Animal Science and Technology, Guangxi University, Nanning, 530004 PR China
| | - Huiyan Xu
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, College of Animal Science and Technology, Guangxi University, Nanning, 530004 PR China
| | - Yangqing Lu
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, College of Animal Science and Technology, Guangxi University, Nanning, 530004 PR China.
| |
Collapse
|
2
|
Gu Y, Wu K, Niu B, Wang Z, Jie Y, Fan Z, Li J, Sun C, Hou ZC, Shao LW. Hyperthermia suppresses the biological characteristics and migration of chicken primordial germ cells. Front Genome Ed 2025; 6:1512108. [PMID: 39845894 PMCID: PMC11751037 DOI: 10.3389/fgeed.2024.1512108] [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: 10/16/2024] [Accepted: 12/19/2024] [Indexed: 01/24/2025] Open
Abstract
Primordial germ cells (PGCs) play a crucial role in transmitting genetic information to the next-generation. In chickens, genetically edited PGCs can be propagated in vitro and subsequently transplanted into recipient embryos to produce offspring with desired genetic traits. However, during early embryogenesis, the effects of external conditions on PGC migration through the vascular system to the gonads have yet to be explored, which may affect the efficiency of preparing gene-edited chickens. In this study, we investigated the effects of hyperthermia on the biological characteristics and migration of chicken PGCs. A gonad-derived PGC line of White Leghorn (WLH) chicken was established and verified through PAS staining and immunofluorescence of PGC-specific proteins. To visually observe PGC migration in vivo, GFP-positive PGCs were prepared and locations of chimeras were validated. Cell viability, glycogen granule contents, and mRNA expression levels of pluripotency markers (NANOG and POUV), germ cell-specific markers (DAZL and CVH), and telomerase reverse transcriptase (TERT) were reduced in PGCs cultured under high temperatures (43°C for 12, 24, and 48 h). After the heat treatment of donor PGCs (43°C) or recipient embryos (39.5°C), GFP-positive PGCs in gonads were rarely observed. Taken together, our results underscore the negative effects of hyperthermia on the biological characteristics and migration of chicken PGCs, which provides valuable insights for the implementation of PGC-based gene editing techniques in chickens.
Collapse
Affiliation(s)
- Yuzhou Gu
- State Key Laboratory of Animal Biotech Breeding, China Agricultural University, Beijing, China
- National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
- Frontiers Science Center for Molecular Design Breeding (MOE), China Agricultural University, Beijing, China
| | - Kexin Wu
- State Key Laboratory of Animal Biotech Breeding, China Agricultural University, Beijing, China
- National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
- Frontiers Science Center for Molecular Design Breeding (MOE), China Agricultural University, Beijing, China
| | - Bowen Niu
- State Key Laboratory of Animal Biotech Breeding, China Agricultural University, Beijing, China
- National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
- Frontiers Science Center for Molecular Design Breeding (MOE), China Agricultural University, Beijing, China
| | - Zhiting Wang
- State Key Laboratory of Animal Biotech Breeding, China Agricultural University, Beijing, China
- National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
- Frontiers Science Center for Molecular Design Breeding (MOE), China Agricultural University, Beijing, China
| | - Yuchen Jie
- State Key Laboratory of Animal Biotech Breeding, China Agricultural University, Beijing, China
- National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
- Frontiers Science Center for Molecular Design Breeding (MOE), China Agricultural University, Beijing, China
| | - Zixuan Fan
- State Key Laboratory of Animal Biotech Breeding, China Agricultural University, Beijing, China
- National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
- Frontiers Science Center for Molecular Design Breeding (MOE), China Agricultural University, Beijing, China
| | - Junying Li
- State Key Laboratory of Animal Biotech Breeding, China Agricultural University, Beijing, China
- National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Congjiao Sun
- State Key Laboratory of Animal Biotech Breeding, China Agricultural University, Beijing, China
- National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
- Frontiers Science Center for Molecular Design Breeding (MOE), China Agricultural University, Beijing, China
| | - Zhuo-Cheng Hou
- State Key Laboratory of Animal Biotech Breeding, China Agricultural University, Beijing, China
- National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
- Frontiers Science Center for Molecular Design Breeding (MOE), China Agricultural University, Beijing, China
| | - Li-Wa Shao
- State Key Laboratory of Animal Biotech Breeding, China Agricultural University, Beijing, China
- National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
- Frontiers Science Center for Molecular Design Breeding (MOE), China Agricultural University, Beijing, China
| |
Collapse
|
3
|
Carver JJ, Amato CM, Hung-Chang Yao H, Zhu Y. Adamts9 is required for the development of primary ovarian follicles and maintenance of female sex in zebrafish†. Biol Reprod 2024; 111:1107-1128. [PMID: 39180722 PMCID: PMC11565245 DOI: 10.1093/biolre/ioae127] [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: 04/15/2024] [Revised: 07/15/2024] [Accepted: 08/23/2024] [Indexed: 08/26/2024] Open
Abstract
Previous studies have suggested that adamts9 (a disintegrin and metalloprotease with thrombospondin type-1 motifs, member 9), an extracellular matrix (ECM) metalloprotease, participates in primordial germ cell (PGC) migration and is necessary for female fertility. In this study, we found that adamts9 knockout (KO) led to reduced body size, and female-to-male sex conversion in late juvenile or adult zebrafish; however, primary sex determination was not affected in early juveniles of adamts9 KO. Overfeeding and lowering the rearing density rescued growth defects in female adamts9 KO fish but did not rescue defects in ovarian development in adamts9 KO. Delayed PGC proliferation, significantly reduced number and size of Stage IB follicles (equivalent to primary follicles) in early juveniles of adamts9 KO, and arrested development at Stage IB follicles in mid- or late-juveniles of adamts9 KO are likely causes of female infertility and sex conversion. Via RNAseq, we found significant enrichment of differentially expressed genes involved in ECM organization during sexual maturation in ovaries of wildtype fish; and significant dysregulation of these genes in adamts9 KO ovaries. RNAseq analysis also showed enrichment of inflammatory transcriptomic signatures in adult ovaries of these adamts9 KO. Taken together, our results indicate that adamts9 is critical for development of primary ovarian follicles and maintenance of female sex, and loss of adamts9 leads to defects in ovarian follicle development, female infertility, and sex conversion in late juveniles and mature adults. These results show that the ECM and extracellular metalloproteases play major roles in maintaining ovarian follicle development in zebrafish.
Collapse
Affiliation(s)
- Jonathan J Carver
- Department of Biology, East Carolina University, Greenville, NC, USA
| | - Ciro M Amato
- National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO, USA
| | | | - Yong Zhu
- Department of Biology, East Carolina University, Greenville, NC, USA
| |
Collapse
|
4
|
Zhao X, Fan C, Qie T, Fu X, Chen X, Wang Y, Wu Y, Fu X, Shi K, Yan W, Yu H. Diaph1 knockout inhibits mouse primordial germ cell proliferation and affects gonadal development. Reprod Biol Endocrinol 2024; 22:82. [PMID: 39010074 PMCID: PMC11247884 DOI: 10.1186/s12958-024-01257-z] [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: 04/16/2024] [Accepted: 07/09/2024] [Indexed: 07/17/2024] Open
Abstract
BACKGROUND Exploring the molecular mechanisms of primordial germ cell (PGC) migration and the involvement of gonadal somatic cells in gonad development is valuable for comprehending the origins and potential treatments of reproductive-related diseases. METHODS Diaphanous related formin 1 (Diaph1, also known as mDia1) was screened by analyzing publicly available datasets (ATAC-seq, DNase-seq, and RNA-seq). Subsequently, the CRISPR-Cas9 technology was used to construct Diaph1 knockout mice to investigate the role of Diaph1 in gonad development. RESULTS Based on data from public databases, a differentially expressed gene Diaph1, was identified in the migration of mouse PGC. Additionally, the number of PGCs was significantly reduced in Diaph1 knockout mice compared to wild type mice, and the expression levels of genes related to proliferation (Dicer1, Mcm9), adhesion (E-cadherin, Cdh1), and migration (Cxcr4, Hmgcr, Dazl) were significantly decreased. Diaph1 knockout also inhibited Leydig cell proliferation and induced apoptosis in the testis, as well as granulosa cell apoptosis in the ovary. Moreover, the sperm count in the epididymal region and the count of ovarian follicles were significantly reduced in Diaph1 knockout mice, resulting in decreased fertility, concomitant with lowered levels of serum testosterone and estradiol. Further research found that in Diaph1 knockout mice, the key enzymes involved in testosterone synthesis (CYP11A1, 3β-HSD) were decreased in Leydig cells, and the estradiol-associated factor (FSH receptor, AMH) in granulosa cells were also downregulated. CONCLUSIONS Overall, our findings indicate that the knockout of Diaph1 can disrupt the expression of factors that regulate sex hormone production, leading to impaired secretion of sex hormones, ultimately resulting in damage to reproductive function. These results provide a new perspective on the molecular mechanisms underlying PGC migration and gonadal development, and offer valuable insights for further research on the causes, diagnosis, and treatment of related diseases.
Collapse
Affiliation(s)
- Xin Zhao
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, 010020, Inner Mongolia, China
| | - Chunbiao Fan
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, 010020, Inner Mongolia, China
| | - Tongtong Qie
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, 010020, Inner Mongolia, China
| | - Xinrui Fu
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, 010020, Inner Mongolia, China
| | - Xiaoshuang Chen
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, 010020, Inner Mongolia, China
| | - Yujia Wang
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, 010020, Inner Mongolia, China
| | - Yuan Wu
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, 010020, Inner Mongolia, China
| | - Xinyao Fu
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, 010020, Inner Mongolia, China
| | - Kesong Shi
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, 010020, Inner Mongolia, China
| | - Wenlong Yan
- School of Biology and Agriculture, Shaoguan University, Shaoguan, 512005, Guangdong Province, China.
| | - Haiquan Yu
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, 010020, Inner Mongolia, China.
| |
Collapse
|
5
|
Yahiro I, Sato O, Mohapatra S, Mukai K, Toyoda A, Itoh T, Matsuyama M, Chakraborty T, Ohta K. SDF-1/CXCR4 signal is involved in the induction of Primordial Germ Cell migration in a model marine fish, Japanese anchovy (Engraulis japonicus). Gen Comp Endocrinol 2024; 351:114476. [PMID: 38408712 DOI: 10.1016/j.ygcen.2024.114476] [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: 09/05/2023] [Revised: 02/16/2024] [Accepted: 02/19/2024] [Indexed: 02/28/2024]
Abstract
Primordial germ cells (PGCs) are pivotal for gonadal development and reproductive success. Though artificial induction of sterility by targeting PGCs are gaining popularity due to its advantages in fish surrogacy and biodiversity management, it is often skill and time intensive. In this study, we have focused on understanding the role of PGCs and the chemotactic SDF-1/CXCR4 signaling on gonad development of Japanese anchovy (JA, Engraulis japonicus), an upcoming marine model organism with eco-commercial values, with an aim to develop a novel, easy, and versatile gonad sterilization method. Our data showed that PGC migration related genes, i.e., sdf-1a, sdf-1b, cxcr4a, cxcr4b and vasa, are phylogenetically closer relatives of respective herring (Clupea harengus) and zebrafish (Danio rerio) homolog. Subsequently, PGC marking and live tracing experiments confirmed that PGC migration in JA initiates from 16 hours post fertilization (hpf) followed by PGC settlement in the gonadal ridge at 44 hpf. We found that overexpression of zebrafish sdf-1a mRNA in the germ cell suppresses cxcr4a and increases cxcr4b transcription at 8 hpf, dose dependently disrupts PGC migration at 24-48 hpf, induces PGC death and upregulates sdf-1b at 5 days post hatching. 48 h of immersion treatment with CXCR4 antagonist (AMD3100, Abcam) also accelerated PGC mismigration and pushed the PGC away from gonadal ridge in a dose responsive manner, and further when grown to adulthood caused germ cell less gonad formation in some individuals. Cumulatively, our data, for the first time, suggests that JA PGC migration is largely regulated by SDF1/CXCR4 signaling, and modulation of this signaling has strong potential for sterile, germ cell less gonad preparation at a mass scale. However, further in-depth analysis is pertinent to apply this methodology in marine fish species to successfully catapult Japanese anchovy into a true marine fish model.
Collapse
Affiliation(s)
- Issei Yahiro
- Laboratory of Marine Biology, Faculty of Agriculture, Kyushu University, Fukuoka 819-0395, Japan
| | - Oga Sato
- Laboratory of Marine Biology, Faculty of Agriculture, Kyushu University, Fukuoka 819-0395, Japan
| | - Sipra Mohapatra
- Laboratory of Marine Biology, Faculty of Agriculture, Kyushu University, Fukuoka 819-0395, Japan; Aqua-Bioresource Innovation Center, Kyushu University, Saga 847-8511, Japan
| | - Koki Mukai
- Laboratory of Marine Biology, Faculty of Agriculture, Kyushu University, Fukuoka 819-0395, Japan; Fisheries Technology Institute, Japan Fisheries Research and Education Agency, Nagasaki 853-0508, Japan
| | - Atsushi Toyoda
- Advanced Genomics Center, National Institute of Genetics, Shizuoka 411-8540, Japan
| | - Takehiko Itoh
- School of Life Science and Technology, Tokyo Institute of Technology, Tokyo 152-8550, Japan
| | - Michiya Matsuyama
- Aqua-Bioresource Innovation Center, Kyushu University, Saga 847-8511, Japan
| | - Tapas Chakraborty
- Laboratory of Marine Biology, Faculty of Agriculture, Kyushu University, Fukuoka 819-0395, Japan; Aqua-Bioresource Innovation Center, Kyushu University, Saga 847-8511, Japan.
| | - Kohei Ohta
- Laboratory of Marine Biology, Faculty of Agriculture, Kyushu University, Fukuoka 819-0395, Japan; Aqua-Bioresource Innovation Center, Kyushu University, Saga 847-8511, Japan.
| |
Collapse
|
6
|
Barton LJ, Roa-de la Cruz L, Lehmann R, Lin B. The journey of a generation: advances and promises in the study of primordial germ cell migration. Development 2024; 151:dev201102. [PMID: 38607588 PMCID: PMC11165723 DOI: 10.1242/dev.201102] [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: 04/13/2024]
Abstract
The germline provides the genetic and non-genetic information that passes from one generation to the next. Given this important role in species propagation, egg and sperm precursors, called primordial germ cells (PGCs), are one of the first cell types specified during embryogenesis. In fact, PGCs form well before the bipotential somatic gonad is specified. This common feature of germline development necessitates that PGCs migrate through many tissues to reach the somatic gonad. During their journey, PGCs must respond to select environmental cues while ignoring others in a dynamically developing embryo. The complex multi-tissue, combinatorial nature of PGC migration is an excellent model for understanding how cells navigate complex environments in vivo. Here, we discuss recent findings on the migratory path, the somatic cells that shepherd PGCs, the guidance cues somatic cells provide, and the PGC response to these cues to reach the gonad and establish the germline pool for future generations. We end by discussing the fate of wayward PGCs that fail to reach the gonad in diverse species. Collectively, this field is poised to yield important insights into emerging reproductive technologies.
Collapse
Affiliation(s)
- Lacy J. Barton
- Department of Neuroscience, Developmental and Regenerative Biology, The University of Texas at San Antonio, One UTSA Circle, San Antonio, TX 78249, USA
| | - Lorena Roa-de la Cruz
- Department of Neuroscience, Developmental and Regenerative Biology, The University of Texas at San Antonio, One UTSA Circle, San Antonio, TX 78249, USA
| | - Ruth Lehmann
- Whitehead Institute and Department of Biology, MIT, 455 Main Street, Cambridge, MA 02142, USA
| | - Benjamin Lin
- Department of Biochemistry & Cell Biology, Stony Brook University, Stony Brook, NY, 11794, USA
| |
Collapse
|
7
|
Olea GB, Aguirre MV, Lombardo DM. Early gonadogenesis in Columba livia (birds: Columbiformes): Migration, colonization, and differentiation of germ cells. Dev Growth Differ 2024; 66:56-65. [PMID: 37795634 DOI: 10.1111/dgd.12895] [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: 02/23/2023] [Revised: 09/26/2023] [Accepted: 09/27/2023] [Indexed: 10/06/2023]
Abstract
In birds, primordial germ cells (PGCs) use the bloodstream to travel to a specific region, where the cells undergo extravasation followed by intrastromal migration to the gonadal crest for further colonization. Currently, DDX4, SSEA1, and Oct4 are used to identify germ cells. Other germline cell-associated molecules are N-cadherin, GnRHR, and 3β hydroxysteroid dehydrogenase (3βHSD), which have been used in mice and birds during gonadal development; however, its role in early gonadogenesis in birds is poorly described. This study aimed to evaluate the differential immunodetection of N-cadherin binding molecule, Oct4 pluripotency protein, GnRHR receptor, and 3βHSD enzyme in Columba livia embryos during migration colonization of PGCs in the gonadal crest and early gonadogenesis. These markers were revealed by immunohistochemistry in histological preparations of C. livia corresponding to stages (S)15 to S40. Immunodetection of N-cadherin, Oct4, GnRHR, and 3βHSD in the germ line of C. livia allowed the identification of PGCs in the yolk sac membrane at the level of the splanchnic mesoderm during migration to the genital crest and its colonization. In the same way, it was possible to characterize and localize PGCs during early gonadogenesis. This study in C. livia demonstrates that Oct4, N-cadherin, GNRHR, and 3βHSD are immunodetected in PGCs and could be used as potential germline cell markers during cell migration out of blood vessels, colonization in the genital crest, and early gonadogenesis. Furthermore, this study could be used as a novel general model to understand the early gonadogenesis in altricial species.
Collapse
Affiliation(s)
- Gabriela Beatriz Olea
- Cátedra de Histología y Embriología, Facultad de Ciencias Veterinarias, Universidad Nacional del Nordeste, Resistencia, Argentina
- Cátedra de Histología y Embriología. Departamento de Ciencias Básicas, Universidad Nacional del Chaco Austral, Sáenz Peña, Argentina
- Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Buenos Aires, Argentina
| | - María Victoria Aguirre
- Laboratorio de Investigaciones Bioquímicas de la Facultad de Medicina (LIBIM) Facultad de Medicina, Universidad Nacional del Nordeste, Instituto de Química Básica y Aplicada del NEA, (IQUIBA NEA-UNNE- CONICET), Resistencia, Argentina
| | - Daniel Marcelo Lombardo
- Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Buenos Aires, Argentina
- Universidad de Buenos Aires, Facultad de Cs Veterinarias, Instituto de Investigación y Tecnología en Reproducción Animal (INITRA). Cátedra de Histología y Embriología, Viamonte, Argentina
| |
Collapse
|
8
|
Suzuki K, Kwon SJ, Saito D, Atsuta Y. LIN28 is essential for the maintenance of chicken primordial germ cells. Cells Dev 2023; 176:203874. [PMID: 37453484 DOI: 10.1016/j.cdev.2023.203874] [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: 05/09/2023] [Revised: 06/30/2023] [Accepted: 07/10/2023] [Indexed: 07/18/2023]
Abstract
Understanding the mechanism of stem cell maintenance underlies the establishment of long-term and mass culture methods for stem cells that are fundamental for clinical and agricultural applications. In this study, we use chicken primordial germ cell (PGC) as a model to elucidate the molecular mechanisms underlying stem cell maintenance. The PGC is a useful experimental model because it is readily gene-manipulatable and easy to test gene function in vivo using transplantation. Previous studies to establish a long-term culture system have shown that secreted factors such as FGF2 are required to maintain the self-renewal capability of PGC. On the other hand, we know little about intracellular regulators responsible for PGC maintenance. Among representative stem cell factors, we focus on RNA-binding factors LIN28A and LIN28B as possible central regulators for the gene regulatory network essential to PGC maintenance. By taking advantage of the CRISPR/Cas9-mediated gene editing and a clonal culture technique, we find that both LIN28A and LIN28B regulate the proliferation of PGC in vitro. We further showed that colonization efficiency of grafted PGC at the genital ridges, rudiments for the gonads, of chicken embryos were significantly decreased by knockout (KO) of LIN28A or LIN28B. Of note, overexpression of human LIN28 in LIN28-KO PGC was sufficient to restore the low colonization rates, suggesting that LIN28 plays a key role in PGC colonization at the gonads. Transcriptomic analyses of LIN28-KO PGC reveal that several genes related to mesenchymal traits are upregulated, including EGR1, a transcription factor that promotes the differentiation of mesodermal tissues. Finally, we show that the forced expression of human EGR1 deteriorates replication activity and colonization efficiency of PGCs. Taken together, this work demonstrates that LIN28 maintains self-renewal of PGC by suppressing the expression of differentiation genes including EGR1.
Collapse
Affiliation(s)
- Katsuya Suzuki
- Department of Biology, Faculty of Science, Kyushu University, Fukuoka 819-0395, Japan
| | - Seung June Kwon
- Department of Biology, Faculty of Science, Kyushu University, Fukuoka 819-0395, Japan
| | - Daisuke Saito
- Department of Biology, Faculty of Science, Kyushu University, Fukuoka 819-0395, Japan
| | - Yuji Atsuta
- Department of Biology, Faculty of Science, Kyushu University, Fukuoka 819-0395, Japan.
| |
Collapse
|
9
|
Golkar-Narenji A, Dziegiel P, Kempisty B, Petitte J, Mozdziak PE, Bryja A. In vitro culture of reptile PGCS to preserve endangered species. Cell Biol Int 2023; 47:1314-1326. [PMID: 37178380 DOI: 10.1002/cbin.12033] [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: 12/31/2022] [Revised: 04/05/2023] [Accepted: 05/01/2023] [Indexed: 05/15/2023]
Abstract
Primordial germ cells (PGCs), are the source of gametes in vertebrates. There are similarities in the development of PGCs of reptiles with avian and mammalian species PGCs development. PGCs culture has been performed for avian and mammalian species but there is no report for reptilian PGCs culture. In vitro culture of PGCs is needed to produce transgenic animals, preservation of endangered animals and for studies on cell behaviour and research on fertility. Reptiles are traded as exotic pets and a source of food and they are valuable for their skin and they are useful as model for medical research. Transgenic reptile has been suggested to be useful for pet industry and medical research. In this research different aspects of PGCs development was compared in three main classes of vertebrates including mammalian, avian and reptilian species. It is proposed that a discussion on similarities between reptilian PGCs development with avian and mammalian species helps to find clues for studies of reptilian PGCs development details and finding an efficient protocol for in vitro culture of reptilian PG.
Collapse
Affiliation(s)
- Afsaneh Golkar-Narenji
- Prestage Department of Poultry Sciences, North Carolina State University, Raleigh, North Carolina, USA
| | - Piotr Dziegiel
- Department of Human Morphology and Embryology, Division of Histology and Embryology, Wrocław Medical University, Wroclaw, Dolnoslaskie, Poland
| | - Bartosz Kempisty
- Prestage Department of Poultry Sciences, North Carolina State University, Raleigh, North Carolina, USA
- Department of Veterinary Surgery, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, Toruń, Poland
- Graduate Physiology Program NC State University North Carolina State University, Raleigh, North Carolina, USA
- Department of Human Morphology and Embryology, Division of Anatomy, Wroclaw Medical University, Wroclaw, Dolnoslaskie, Poland
| | - James Petitte
- Prestage Department of Poultry Sciences, North Carolina State University, Raleigh, North Carolina, USA
| | - Paul Edward Mozdziak
- Prestage Department of Poultry Sciences, North Carolina State University, Raleigh, North Carolina, USA
- Graduate Physiology Program NC State University North Carolina State University, Raleigh, North Carolina, USA
| | - Artur Bryja
- Department of Human Morphology and Embryology, Division of Anatomy, Wroclaw Medical University, Wroclaw, Dolnoslaskie, Poland
| |
Collapse
|
10
|
Formation, Application, and Significance of Chicken Primordial Germ Cells: A Review. Animals (Basel) 2023; 13:ani13061096. [PMID: 36978637 PMCID: PMC10044044 DOI: 10.3390/ani13061096] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 03/11/2023] [Accepted: 03/13/2023] [Indexed: 03/22/2023] Open
Abstract
Chicken is one of the most widely consumed sources of protein globally. Primordial germ cells (PGCs) are the precursors for ova and sperm. One of the early embryogenesis events in most animals is the segregation of the somatic and germ lineages. PGC cultures occur in the germline, and PGCs are less studied in many species. It is relatively challenging to separate, cultivate, and genetically alter chicken without mutating the basic germline. The present study aims to gather previous research about chicken PGCs and provide a customized review of studies and developments in the field of PGCs, especially for avian species. Furthermore, we show that the propagation of chicken PGCs into embryonic germ cells that contribute to somatic tissues may be produced in vitro. Primordial germ cells offer an ideal system in developmental biology, as these cells play a vital role in the genetic modification and treatment of infertility. Cryopreservation helps to maintain genetic resources and sustainable production in the poultry industry. Keeping in mind the significance of cryopreservation for storage and gametogenesis, we discuss its role in the preservation of primordial germ cells. Transgenesis and genetic modifications in chicken lead to the development of various medicinal chicken varieties and aid in improving their production and quality for consumption purposes. Additionally, these characteristics open up new possibilities for modifying the chicken genome for agricultural and medical purposes.
Collapse
|
11
|
Ichikawa K, Horiuchi H. Fate Decisions of Chicken Primordial Germ Cells (PGCs): Development, Integrity, Sex Determination, and Self-Renewal Mechanisms. Genes (Basel) 2023; 14:genes14030612. [PMID: 36980885 PMCID: PMC10048776 DOI: 10.3390/genes14030612] [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: 02/02/2023] [Revised: 02/24/2023] [Accepted: 02/27/2023] [Indexed: 03/05/2023] Open
Abstract
Primordial germ cells (PGCs) are precursor cells of sperm and eggs. The fate decisions of chicken PGCs in terms of their development, integrity, and sex determination have unique features, thereby providing insights into evolutionary developmental biology. Additionally, fate decisions in the context of a self-renewal mechanism have been applied to establish culture protocols for chicken PGCs, enabling the production of genome-edited chickens and the conservation of genetic resources. Thus, studies on the fate decisions of chicken PGCs have significantly contributed to both academic and industrial development. Furthermore, studies on fate decisions have rapidly advanced owing to the recent development of essential research technologies, such as genome editing and RNA sequencing. Here, we reviewed the status of fate decisions of chicken PGCs and provided insight into other important research issues that require attention.
Collapse
Affiliation(s)
- Kennosuke Ichikawa
- Genome Editing Innovation Center, Hiroshima University, 3-10-23 Kagamiyama, Higashi-Hiroshima 739-0046, Hiroshima, Japan
- Correspondence:
| | - Hiroyuki Horiuchi
- Genome Editing Innovation Center, Hiroshima University, 3-10-23 Kagamiyama, Higashi-Hiroshima 739-0046, Hiroshima, Japan
- Graduate School of Integrated Sciences for Life, Hiroshima University, 1-4-4 Kagamiyama, Higashi-Hiroshima 739-8528, Hiroshima, Japan
| |
Collapse
|
12
|
Himelreich Perić M, Takahashi M, Ježek D, Cunha GR. Early development of the human embryonic testis. Differentiation 2023; 129:4-16. [PMID: 35961887 DOI: 10.1016/j.diff.2022.07.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 07/11/2022] [Accepted: 07/12/2022] [Indexed: 01/25/2023]
Abstract
Human gonadal development culminating in testicular differentiation is described through analysis of histologic sections derived from 33-day to 20-week human embryos/fetuses, focusing on early development (4-8 weeks of gestation). Our study updates the comprehensive studies of Felix (1912), van Wagenen and Simpson (1965), and Juric-Lekic et al. (2013), which were published in books and thus are unsearchable via PubMed. Human gonads develop from the germinal ridge, a thickening of coelomic epithelium on the medial side of the urogenital ridge. The bilateral urogenital ridges contain elements of the mesonephric kidney, namely the mesonephric duct, mesonephric tubules, and mesonephric glomeruli. The germinal ridge, into which primordial germ cells migrate, is initially recognized as a thickening of coelomic epithelium on the urogenital ridge late in the 4th week of gestation. Subsequently, in the 5th week of gestation, a dense mesenchyme develops sub-adjacent to the epithelium of the germinal ridge, and together these elements bulge into the coelomic cavity forming bilateral longitudinal ridges attached to the urogenital ridges. During development, primordial cells migrate into the germinal ridge and subsequently into testicular cords that form within the featureless dense mesenchyme of the germinal ridge at 6-8 weeks of gestation. The initial low density of testicular cords seen at 8 weeks remodels into a dense array of testicular cords surrounded by α-actin-positive myoid cells during the second trimester. Human testicular development shares many features with that of mice being derived from 4 elements: coelomic epithelium, sub-adjacent mesenchyme, primordial germ cells, and the mesonephros.
Collapse
Affiliation(s)
- Marta Himelreich Perić
- Scientific Centre of Excellence for Reproductive and Regenerative Medicine, School of Medicine, University of Zagreb, 10000, Zagreb, Croatia.
| | - Marta Takahashi
- Department of Communication Sciences, Catholic University of Croatia, 10000, Zagreb, Croatia
| | - Davor Ježek
- Scientific Centre of Excellence for Reproductive and Regenerative Medicine, School of Medicine, University of Zagreb, 10000, Zagreb, Croatia; Department of Histology and Embryology, School of Medicine, University of Zagreb, 10000, Zagreb, Croatia
| | - Gerald R Cunha
- Department of Urology, University of California, 400 Parnassus Avenue, San Francisco, CA, 94143, USA
| |
Collapse
|
13
|
Atsuta Y, Suzuki K, Iikawa H, Yaguchi H, Saito D. Prime editing in chicken fibroblasts and primordial germ cells. Dev Growth Differ 2022; 64:548-557. [PMID: 36374008 DOI: 10.1111/dgd.12823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 09/28/2022] [Accepted: 11/01/2022] [Indexed: 11/16/2022]
Abstract
CRISPR/Cas9-based genome editing technologies are revolutionizing developmental biology. One of the advanced CRISPR-based techniques is prime editing (PE), which enables precise gene modification in multiple model organisms. However, there has been no report of taking advantage of the PE system for gene editing in primordial germ cells (PGCs) thus far. In the current study, we describe a method to apply PE to the genome of chicken fibroblasts and PGCs. By combining PE with a transposon-mediated genomic integration, drug selection, and the single-cell culture method, we successfully generated prime-edited chicken fibroblasts and PGCs. The chicken PGC is widely used as an experimental model to study germ cell formation and as a vector for gene transfer to produce transgenic chickens. Such experimental models are useful in the developmental biology field and as potential bioreactors to produce pharmaceutical and nutritious proteins. Thus, the method presented here will provide not only a powerful tool to investigate gene function in germ cell development but also a basis for generating prime-edited transgenic birds.
Collapse
Affiliation(s)
- Yuji Atsuta
- Department of Biology, Faculty of Science, Kyushu University, Fukuoka, Japan
| | - Katsuya Suzuki
- Department of Biology, Faculty of Science, Kyushu University, Fukuoka, Japan
| | - Hiroko Iikawa
- Department of Biology, Faculty of Science, Kyushu University, Fukuoka, Japan
| | - Haruna Yaguchi
- Department of Biology, Faculty of Science, Kyushu University, Fukuoka, Japan
| | - Daisuke Saito
- Department of Biology, Faculty of Science, Kyushu University, Fukuoka, Japan
| |
Collapse
|
14
|
Female Germ Cell Development in Chickens and Humans: The Chicken Oocyte Enriched Genes Convergent and Divergent with the Human Oocyte. Int J Mol Sci 2022; 23:ijms231911412. [PMID: 36232712 PMCID: PMC9570461 DOI: 10.3390/ijms231911412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 09/17/2022] [Accepted: 09/21/2022] [Indexed: 11/16/2022] Open
Abstract
The development of germ cells and other physiological events in the differentiated ovary of humans are highly conserved with several mammalian species, except for the differences in timing. However, comparative knowledge on this topic is very scarce with respect to humans and lower vertebrates, such as chickens. In chickens, female germ cells enter into meiosis around embryonic day (E) 15.5 and are arrested in meiotic prophase I as primary oocytes. The oocytes arrested in meiosis I are accumulated in germ-cell cysts; shortly after hatching, they are enclosed by flattened granulosa cells in order to form primordial follicles. In humans, the process of meiotic recombination in female germ cells begins in the 10–11th week of gestation, and primordial follicles are formed at around week 20. In this review, we comprehensively elucidate both the conservation and the species-specific differences between chickens and humans with respect to germ cell, oocyte, and follicle development. Importantly, we provide functional insights into a set of chicken oocyte enriched genes (from E16 to 1 week post-hatch) that show convergent and divergent expression patterns with respect to the human oocyte (from week 11 to 26).
Collapse
|
15
|
Shams AS, Arpke RW, Gearhart MD, Weiblen J, Mai B, Oyler D, Bosnakovski D, Mahmoud OM, Hassan GM, Kyba M. The chemokine receptor CXCR4 regulates satellite cell activation, early expansion, and self-renewal, in response to skeletal muscle injury. Front Cell Dev Biol 2022; 10:949532. [PMID: 36211464 PMCID: PMC9536311 DOI: 10.3389/fcell.2022.949532] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Accepted: 08/25/2022] [Indexed: 11/13/2022] Open
Abstract
Acute skeletal muscle injury is followed by satellite cell activation, proliferation, and differentiation to replace damaged fibers with newly regenerated muscle fibers, processes that involve satellite cell interactions with various niche signals. Here we show that satellite cell specific deletion of the chemokine receptor CXCR4, followed by suppression of recombination escapers, leads to defects in regeneration and satellite cell pool repopulation in both the transplantation and in situ injury contexts. Mechanistically, we show that endothelial cells and FAPs express the gene for the ligand, SDF1α, and that CXCR4 is principally required for proper activation and for transit through the first cell division, and to a lesser extent the later cell divisions. In the absence of CXCR4, gene expression in quiescent satellite cells is not severely disrupted, but in activated satellite cells a subset of genes normally induced by activation fail to upregulate normally. These data demonstrate that CXCR4 signaling is essential to normal early activation, proliferation, and self-renewal of satellite cells.
Collapse
Affiliation(s)
- Ahmed S. Shams
- Lillehei Heart Institute, Minneapolis, MN, United States
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, United States
- Department of Human Anatomy and Embryology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Robert W. Arpke
- Lillehei Heart Institute, Minneapolis, MN, United States
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, United States
| | - Micah D. Gearhart
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN, United States
| | - Johannes Weiblen
- Lillehei Heart Institute, Minneapolis, MN, United States
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, United States
| | - Ben Mai
- Lillehei Heart Institute, Minneapolis, MN, United States
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, United States
| | - David Oyler
- Lillehei Heart Institute, Minneapolis, MN, United States
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, United States
| | - Darko Bosnakovski
- Lillehei Heart Institute, Minneapolis, MN, United States
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, United States
| | - Omayma M. Mahmoud
- Department of Human Anatomy and Embryology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Gamal M. Hassan
- Department of Human Anatomy and Embryology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Michael Kyba
- Lillehei Heart Institute, Minneapolis, MN, United States
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, United States
- *Correspondence: Michael Kyba,
| |
Collapse
|
16
|
Rengaraj D, Cha DG, Lee HJ, Lee KY, Choi YH, Jung KM, Kim YM, Choi HJ, Choi HJ, Yoo E, Woo SJ, Park JS, Park KJ, Kim JK, Han JY. Dissecting chicken germ cell dynamics by combining a germ cell tracing transgenic chicken model with single-cell RNA sequencing. Comput Struct Biotechnol J 2022; 20:1654-1669. [PMID: 35465157 PMCID: PMC9010679 DOI: 10.1016/j.csbj.2022.03.040] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 03/30/2022] [Accepted: 03/31/2022] [Indexed: 02/02/2023] Open
Abstract
Avian germ cells can be distinguished by certain characteristics during development. On the basis of these characteristics, germ cells can be used for germline transmission. However, the dynamic transcriptional landscape of avian germ cells during development is unknown. Here, we used a novel germ-cell-tracing method to monitor and isolate chicken germ cells at different stages of development. We targeted the deleted in azoospermia like (DAZL) gene, a germ-cell-specific marker, to integrate a green fluorescent protein (GFP) reporter gene without affecting endogenous DAZL expression. The resulting transgenic chickens (DAZL::GFP) were used to uncover the dynamic transcriptional landscape of avian germ cells. Single-cell RNA sequencing of 4,752 male and 13,028 female DAZL::GFP germ cells isolated from embryonic day E2.5 to 1 week post-hatch identified sex-specific developmental stages (4 stages in male and 5 stages in female) and trajectories (apoptosis and meiosis paths in female) of chicken germ cells. The male and female trajectories were characterized by a gradual acquisition of stage-specific transcription factor activities. We also identified evolutionary conserved and species-specific gene expression programs during both chicken and human germ-cell development. Collectively, these novel analyses provide mechanistic insights into chicken germ-cell development.
Collapse
Affiliation(s)
- Deivendran Rengaraj
- Department of Agricultural Biotechnology, and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul 08826, South Korea
| | - Dong Gon Cha
- Department of New Biology, DGIST, Daegu 42988, South Korea
| | - Hong Jo Lee
- Department of Agricultural Biotechnology, and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul 08826, South Korea
| | - Kyung Youn Lee
- Department of Agricultural Biotechnology, and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul 08826, South Korea
| | - Yoon Ha Choi
- Department of New Biology, DGIST, Daegu 42988, South Korea
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang 37673, South Korea
| | - Kyung Min Jung
- Department of Agricultural Biotechnology, and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul 08826, South Korea
| | - Young Min Kim
- Department of Agricultural Biotechnology, and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul 08826, South Korea
| | - Hee Jung Choi
- Department of Agricultural Biotechnology, and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul 08826, South Korea
| | - Hyeon Jeong Choi
- Department of Agricultural Biotechnology, and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul 08826, South Korea
| | - Eunhui Yoo
- Department of Agricultural Biotechnology, and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul 08826, South Korea
| | - Seung Je Woo
- Department of Agricultural Biotechnology, and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul 08826, South Korea
| | - Jin Se Park
- Department of Agricultural Biotechnology, and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul 08826, South Korea
| | - Kyung Je Park
- Department of Agricultural Biotechnology, and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul 08826, South Korea
| | - Jong Kyoung Kim
- Department of New Biology, DGIST, Daegu 42988, South Korea
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang 37673, South Korea
- Corresponding authors at: POSTECH, 77 Cheongam-ro, Nam-gu, Pohang-si, Gyeongsangbuk-do 37673, South Korea (J.K. Kim). Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, South Korea (J.Y. Han).
| | - Jae Yong Han
- Department of Agricultural Biotechnology, and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul 08826, South Korea
- Corresponding authors at: POSTECH, 77 Cheongam-ro, Nam-gu, Pohang-si, Gyeongsangbuk-do 37673, South Korea (J.K. Kim). Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, South Korea (J.Y. Han).
| |
Collapse
|
17
|
Prummel KD, Crowell HL, Nieuwenhuize S, Brombacher EC, Daetwyler S, Soneson C, Kresoja-Rakic J, Kocere A, Ronner M, Ernst A, Labbaf Z, Clouthier DE, Firulli AB, Sánchez-Iranzo H, Naganathan SR, O'Rourke R, Raz E, Mercader N, Burger A, Felley-Bosco E, Huisken J, Robinson MD, Mosimann C. Hand2 delineates mesothelium progenitors and is reactivated in mesothelioma. Nat Commun 2022; 13:1677. [PMID: 35354817 PMCID: PMC8967825 DOI: 10.1038/s41467-022-29311-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 03/04/2022] [Indexed: 01/27/2023] Open
Abstract
The mesothelium lines body cavities and surrounds internal organs, widely contributing to homeostasis and regeneration. Mesothelium disruptions cause visceral anomalies and mesothelioma tumors. Nonetheless, the embryonic emergence of mesothelia remains incompletely understood. Here, we track mesothelial origins in the lateral plate mesoderm (LPM) using zebrafish. Single-cell transcriptomics uncovers a post-gastrulation gene expression signature centered on hand2 in distinct LPM progenitor cells. We map mesothelial progenitors to lateral-most, hand2-expressing LPM and confirm conservation in mouse. Time-lapse imaging of zebrafish hand2 reporter embryos captures mesothelium formation including pericardium, visceral, and parietal peritoneum. We find primordial germ cells migrate with the forming mesothelium as ventral migration boundary. Functionally, hand2 loss disrupts mesothelium formation with reduced progenitor cells and perturbed migration. In mouse and human mesothelioma, we document expression of LPM-associated transcription factors including Hand2, suggesting re-initiation of a developmental program. Our data connects mesothelium development to Hand2, expanding our understanding of mesothelial pathologies.
Collapse
Affiliation(s)
- Karin D Prummel
- Department of Pediatrics, Section of Developmental Biology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
- Department of Molecular Life Sciences, University of Zurich, Zürich, Switzerland
- Structural and Computational Biology Unit, EMBL, Heidelberg, Germany
| | - Helena L Crowell
- Department of Molecular Life Sciences, University of Zurich, Zürich, Switzerland
- SIB Swiss Institute of Bioinformatics, University of Zurich, Zürich, Switzerland
| | - Susan Nieuwenhuize
- Department of Pediatrics, Section of Developmental Biology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
- Department of Molecular Life Sciences, University of Zurich, Zürich, Switzerland
| | - Eline C Brombacher
- Department of Molecular Life Sciences, University of Zurich, Zürich, Switzerland
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Stephan Daetwyler
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX, United States
- Department of Cell Biology, UT Southwestern Medical Center, Dallas, TX, United States
| | - Charlotte Soneson
- Department of Molecular Life Sciences, University of Zurich, Zürich, Switzerland
- SIB Swiss Institute of Bioinformatics, University of Zurich, Zürich, Switzerland
| | - Jelena Kresoja-Rakic
- Department of Pediatrics, Section of Developmental Biology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
- Laboratory of Molecular Oncology, Department of Thoracic Surgery, University Hospital Zurich, Zürich, Switzerland
| | - Agnese Kocere
- Department of Pediatrics, Section of Developmental Biology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
- Department of Molecular Life Sciences, University of Zurich, Zürich, Switzerland
| | - Manuel Ronner
- Laboratory of Molecular Oncology, Department of Thoracic Surgery, University Hospital Zurich, Zürich, Switzerland
| | | | - Zahra Labbaf
- Institute for Cell Biology, ZMBE, Muenster, Germany
| | - David E Clouthier
- Department of Craniofacial Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Anthony B Firulli
- Herman B Wells Center for Pediatric Research, Departments of Pediatrics, Anatomy and Medical and Molecular Genetics, Indiana Medical School, Indianapolis, IN, USA
| | - Héctor Sánchez-Iranzo
- Centro Nacional de Investigaciones Cardiovasculares (CNIC-ISCIII), Madrid, Spain
- Institute of Biological and Chemical System - Biological Information Processing (IBCS-BIP), Karlsruhe Institute of Technology (KIT), Eggenstein-Leopoldshafen, Germany
| | - Sundar R Naganathan
- Institute of Bioengineering, Ecole Polytechnique Federale de Lausanne, Lausanne, Switzerland
| | - Rebecca O'Rourke
- Department of Pediatrics, Section of Developmental Biology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Erez Raz
- Institute for Cell Biology, ZMBE, Muenster, Germany
| | - Nadia Mercader
- Institute of Anatomy, University of Bern, Bern, Switzerland
- Centro Nacional de Investigaciones Cardiovasculares (CNIC-ISCIII), Madrid, Spain
| | - Alexa Burger
- Department of Pediatrics, Section of Developmental Biology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Emanuela Felley-Bosco
- Laboratory of Molecular Oncology, Department of Thoracic Surgery, University Hospital Zurich, Zürich, Switzerland
| | - Jan Huisken
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
- Morgridge Institute for Research, Madison, WI, USA
| | - Mark D Robinson
- Department of Molecular Life Sciences, University of Zurich, Zürich, Switzerland
- SIB Swiss Institute of Bioinformatics, University of Zurich, Zürich, Switzerland
| | - Christian Mosimann
- Department of Pediatrics, Section of Developmental Biology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
18
|
Nakamura H, Iwakawa G, Matsubara K. Activation of Migratory Ability in Male Mouse Primordial Germ Cells by in vitro Organ Culture. JOURNAL OF MEDICAL SCIENCES 2022. [DOI: 10.3923/jms.2022.53.60] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
|
19
|
Huang X, Meng L, Wang S, Man Q, Jiang Y, Zhu G. Transcriptional dynamics of the circulating chicken primordial germ cells revealing key genes in cell adhesion and proliferation prior to gonad colonization. Mol Reprod Dev 2022; 89:214-226. [PMID: 35118723 DOI: 10.1002/mrd.23558] [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: 08/08/2021] [Revised: 11/14/2021] [Accepted: 12/14/2021] [Indexed: 11/07/2022]
Abstract
Primordial germ cells (PGCs), precursors to sperms and oocytes, are responsible for the transfer of genetic information to the next generation. The PGCs arise far away from the developing gonad and thus have to migrate across the embryo to reach their site of function. The migration of PGCs from extraembryonic regions to the genital ridges is accomplished through distinct routes among different species. In particular, the birds PGCs utilized the developing circulation system to travel long distance before settling within the gonad. This study screened the transcriptome profile of chicken PGCs isolated from the bloodstream and the genital ridges to identify the cell intrinsic signals that could guide the unique migration path through circulation. We found cell adhesion and extracellular matrix (ECM) associated pathways were highly enriched in the PGCs from blood but not gonads. The platelet-derived growth factor receptors (PDGFRA and PDGFRB) were downregulated during gonad colonization and knockdown of either PDGFRA or PDGFRB inhibit the proliferation of blood PGCs. Furthermore, the migration of blood PGCs was impaired by the suppression of PDGFRA but not PDGFRB. Hence, the chicken PGCs show dynamic transcriptional remodeling during the blood-to-gonad migration and colonization. The free-floating PGCs in the circulation already express genes associated with cell-cell and cell-ECM interactions and therefore prepare for gonadal colonization.
Collapse
Affiliation(s)
- Xiaochen Huang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of the Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Lu Meng
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of the Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Sheng Wang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of the Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Qiu Man
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of the Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Yunqi Jiang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of the Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Guiyu Zhu
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of the Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
20
|
Rengaraj D, Cha DG, Park KJ, Lee KY, Woo SJ, Han JY. Finer resolution analysis of transcriptional programming during the active migration of chicken primordial germ cells. Comput Struct Biotechnol J 2022; 20:5911-5924. [PMID: 36382185 PMCID: PMC9636429 DOI: 10.1016/j.csbj.2022.10.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 10/04/2022] [Accepted: 10/21/2022] [Indexed: 12/01/2022] Open
Abstract
Primordial germ cells (PGCs) in chickens polarize and move passively toward the anterior region by the morphogenetic movement of the embryo. Further migration of PGCs towards the genital ridge via the germinal crescent region and blood vessels occurs actively through the chemoattractive signals. The mechanisms of initiation of PGCs migration, lodging the PGCs in the vascular system, and colonization of PGCs in the gonads are well-studied. However, transcriptome sequencing-based cues directing the migration of the PGCs towards gonads, some of the relevant molecules, biological processes, and transcription factors (TFs) are less studied in chickens. The current study comprehensively interprets the transcriptional programming of PGCs during their active migration (E2.5 to E8). Current results revealed several vital understandings, including a set of genes that upregulated male-specifically (XPA, GNG10, RPL17, RPS23, and NDUFS4) or female-specifically (HINTW, NIPBL, TERAL2, ATP5F1AW, and SMAD2W) in migrating PGCs, and transcriptionally distinct PGCs, particularly in the gonadal environment. We identified DNA methylation and histone modification-associated genes that are novel in chicken PGCs and show a time-dependent enrichment in migrating PGCs. We further identified a large number of differentially expressed genes (DEGs, including TFs) in blood PGCs (at E2.5) compared to gonadal PGCs (at E8) in both sexes; however, this difference was greater in males. We also revealed the enriched biological processes and signaling pathways of significant DEGs identified commonly, male-specifically, or female-specifically between the PGCs isolated at E2.5, E6, and E8. Collectively, these analyses provide molecular insights into chicken PGCs during their active migration phase.
Collapse
Affiliation(s)
- Deivendran Rengaraj
- Department of Agricultural Biotechnology, and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul 08826, Korea
| | - Dong Gon Cha
- Department of New Biology, DGIST, Daegu 42988, Korea
| | - Kyung Je Park
- Department of Agricultural Biotechnology, and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul 08826, Korea
| | - Kyung Youn Lee
- Department of Agricultural Biotechnology, and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul 08826, Korea
| | - Seung Je Woo
- Department of Agricultural Biotechnology, and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul 08826, Korea
| | - Jae Yong Han
- Department of Agricultural Biotechnology, and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul 08826, Korea
- Corresponding author at: Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Korea.
| |
Collapse
|
21
|
An avian embryo patient-derived xenograft model for preclinical studies of human breast cancers. iScience 2021; 24:103423. [PMID: 34849474 PMCID: PMC8608609 DOI: 10.1016/j.isci.2021.103423] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 09/30/2021] [Accepted: 11/08/2021] [Indexed: 11/26/2022] Open
Abstract
Lack of preclinical patient-derived xenograft cancer models in which to conduct large-scale molecular studies seriously impairs the development of effective personalized therapies. We report here an in vivo concept consisting of implanting human tumor cells in targeted tissues of an avian embryo, delivering therapeutics, evaluating their efficacy by measuring tumors using light sheet confocal microscopy, and conducting large-scale RNA-seq analysis to characterize therapeutic-induced changes in gene expression. The model was established to recapitulate triple-negative breast cancer (TNBC) and validated using TNBC standards of care and an investigational therapeutic agent. Human TNBC cells implanted in the avian embryo survive and efficiently form tumors Anti-tumoral therapies intravenously injected are assessed by 3D imaging Post-graft large-scale analyses allow studying the mechanism of action of drugs The AVI-PDX allows preclinical evaluation of therapies and patient stratification
Collapse
|
22
|
Aalto A, Olguin-Olguin A, Raz E. Zebrafish Primordial Germ Cell Migration. Front Cell Dev Biol 2021; 9:684460. [PMID: 34249937 PMCID: PMC8260996 DOI: 10.3389/fcell.2021.684460] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 05/17/2021] [Indexed: 01/03/2023] Open
Abstract
Similar to many other organisms, zebrafish primordial germ cells (PGCs) are specified at a location distinct from that of gonadal somatic cells. Guided by chemotactic cues, PGCs migrate through embryonic tissues toward the region where the gonad develops. In this process, PGCs employ a bleb-driven amoeboid migration mode, characterized by low adhesion and high actomyosin contractility, a strategy used by other migrating cells, such as leukocytes and certain types of cancer cells. The mechanisms underlying the motility and the directed migration of PGCs should be robust to ensure arrival at the target, thereby contributing to the fertility of the organism. These features make PGCs an excellent model for studying guided single-cell migration in vivo. In this review, we present recent findings regarding the establishment and maintenance of cell polarity that are essential for motility and discuss the mechanisms by which cell polarization and directed migration are controlled by chemical and physical cues.
Collapse
Affiliation(s)
- Anne Aalto
- Institute of Cell Biology, Center for Molecular Biology of Inflammation, University of Münster, Münster, Germany
| | - Adan Olguin-Olguin
- Institute of Cell Biology, Center for Molecular Biology of Inflammation, University of Münster, Münster, Germany
| | - Erez Raz
- Institute of Cell Biology, Center for Molecular Biology of Inflammation, University of Münster, Münster, Germany
| |
Collapse
|
23
|
Chang CT, Lee YH, HuangFu WC, Liu IH. Cell-intrinsic Fgf signaling contributes to primordial germ cell homing in zebrafish. Theriogenology 2020; 158:424-431. [PMID: 33039926 DOI: 10.1016/j.theriogenology.2020.09.037] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 09/26/2020] [Accepted: 09/28/2020] [Indexed: 12/24/2022]
Abstract
Primordial germ cells (PGCs) are specified before gastrulation and migrate toward the developing gonads. Previous in vitro studies have demonstrated a cell-intrinsic requirement of fibroblast growth factors (FGFs) by PGCs; however, no evidence suggests FGFs signal directly to PGCs in vivo. Here, using zebrafish as the animal model, we identified the mRNA expressions of Fgf receptors (Fgfrs) and determined the roles of Fgf signaling in migrating PGCs. To clarify the functions of Fgf signaling, we manipulated Fgf signaling specifically in PGCs using dominant-negative (dn) and constitutively-active (ca) Fgfrs and revealed a requirement of a basal Fgf signaling level for the robust arrival of PGCs. Repression of Fgf signaling in PGCs swayed the marginal positioning of PGCs as early as 6 h post-fertilization (6 hpf) and disrupted their arrival at the gonadal ridge at 24 hpf. On the other hand, the ectopic PGC phenotypes caused by the dn-Fgfrs could be alleviated by constitutive activation of Fgf signaling. In addition, we carefully ruled out the somatic effects in mosaic embryos by injecting RNA materials into one blastomere of the four- or eight-cell stage embryos. Injection of dn-Fgfrs into one of eight blastomeres hampered the arrival of only the treated PGCs, while the other PGCs remained unaffected. Furthermore, mosaic treatment of ca-Fgfrs rescued the ectopic rates of dn-Fgfr treated PGCs, while the other PGCs remained more ectopic within the same embryos. Interestingly, PGC-specific repression of Fgf signaling did not compromise the PGC number. To our knowledge, this is the first in vivo evidence to show that Fgf signaling plays a cell-intrinsic role in the migration of vertebrate PGCs.
Collapse
Affiliation(s)
- Chia-Teng Chang
- Department of Animal Science and Technology, National Taiwan University, Taipei, 106, Taiwan
| | - Yen-Hua Lee
- Department of Animal Science and Technology, National Taiwan University, Taipei, 106, Taiwan
| | - Wei-Chun HuangFu
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, 11031, Taiwan
| | - I-Hsuan Liu
- Department of Animal Science and Technology, National Taiwan University, Taipei, 106, Taiwan; Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, 106, Taiwan; School of Veterinary Medicine, National Taiwan University, Taipei, 106, Taiwan.
| |
Collapse
|
24
|
Zhang T, Chi XX, Kong FX, Chu XL. Effect of genistein on the gene and protein expressions of CXCL-12 and EGR-1 in the rat ovary. J Anim Physiol Anim Nutr (Berl) 2020; 105:191-197. [PMID: 32981128 DOI: 10.1111/jpn.13448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 08/20/2020] [Indexed: 11/28/2022]
Abstract
The effect of genistein (GEN) on the gene expression level of stromal cell-derived factor-1/CXCL-12 and early growth response gene-1 was studied in ovarian tissue of young and initially ageing (early stages in the ageing process) female rats. Forty, young female Sprague Dawley (SD) rats of 2-3 months old (200 ±20 g) and forty, initially ageing female SD rats of 10-12 months (490 ± 20 g) old were selected. According to the weight, rats were divided into control group, low-dose group (L), medium-dose group (M) and a high-dose group (H) and were given 15, 30 and 60 mg/kg GEN respectively. The positive control (Oestrogen) group was given 0.5 mg/kg diethylstilbestrol. The treatment lasted for 30 days. The mRNA expression of C-X-C motif chemokine ligand 12 (CXCL-12) and early growth response factor-1 (EGR-1) was measured by real-time PCR, and protein expression of EGR-1 was detected by Western blot. When compared to the negative control group (NC), the ovary/body weight ratio in the young rats decreased in the GEN group, but the difference was not significant. Similarly, compared with NC, the ovary/body weight ratio in the initially ageing rats also decreased with the increase in GEN concentration, but the decrease was significant in M and H groups (p < .01). The administration of GEN enhanced both the gene and protein expression levels of CXCL-12 and EGR-1 in the ovary. Pearson's correlation analysis showed a synergistic effect between CXCL-12 and EGR-1. Thus, we conclude that the effect of GEN on CXCL-12 and EGR-1 in the initially ageing group was obvious than that in the younger group.
Collapse
Affiliation(s)
- Tao Zhang
- College of Medical Laboratory Science and Technology, Harbin Medical University-Daqing, Daqing, Heilongjiang Province, China
| | - Xiao-Xing Chi
- College of Food Science, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang Province, China.,Key Laboratory of Agro-Products Processing and Quality Safety of Heilongjiang Province, Daqing, Heilongjiang Province, China
| | - Fan-Xiu Kong
- College of Food Science, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang Province, China
| | - Xiao-Li Chu
- College of Food Science, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang Province, China
| |
Collapse
|
25
|
Effects of HSP90 inhibition on primordial germ cells migration: A study in the gonad of the chick embryo. Morphologie 2020; 104:228-236. [PMID: 32896470 DOI: 10.1016/j.morpho.2020.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 07/21/2020] [Accepted: 07/22/2020] [Indexed: 11/24/2022]
Abstract
INTRODUCTION Primordial Germ Cells (PGCs) differentiate into spermatozoa or oocytes. They appear early during embryonic development before migrating to the gonadal ridges. Because of their long migration, PGCs have been proposed as a valuable model to study long distance cell migration. Some species also present a vascular phase in the migration of the germline and could therefore be compared to metastatic migration. HSP90 is a heat shock protein involved in the stabilization of several client-proteins, including oncoproteins. HSP90 inhibition has been proved to decrease PGCs migration in mouse and zebrafish. MATERIAL AND METHODS We investigated the effect of geldanamycin on PGCs migration in a species with a vascular phase, the chicken. Geldanamycin was injected in the egg at 48h of incubation, PGC's were detected in blood using of blood smears, and in the embryo by immunohistochemistry using anti-HSP90 antibody. RESULTS The effects of the treatment were similar to those observed in mouse and zebrafish. We show the presence of ectopic germs cells in the vasculature and in the dorsal mesentery, and some deformities of the gonads. CONCLUSION Inhibition of HSP90 decreases the migration of PGCs and proposed the migration of PGCs in the chick embryo as an interesting model to study metastatic invasion.
Collapse
|
26
|
Nagy N, Busalt F, Halasy V, Kohn M, Schmieder S, Fejszak N, Kaspers B, Härtle S. In and Out of the Bursa-The Role of CXCR4 in Chicken B Cell Development. Front Immunol 2020; 11:1468. [PMID: 32765509 PMCID: PMC7381227 DOI: 10.3389/fimmu.2020.01468] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 06/05/2020] [Indexed: 11/13/2022] Open
Abstract
In contrast to mammals, early B cell differentiation and diversification of the antibody repertoire in chickens do not take place in the bone marrow but in a specialized gut associated lymphoid tissue (GALT), the bursa of Fabricius. During embryonic development, B cell precursors migrate to the bursa anlage, where they proliferate and diversify their B cell receptor repertoire. Around hatch these diversified B cells start to emigrate from the bursa of Fabricius and populate peripheral lymphoid organs, but very little is known how the migratory processes are regulated. As CXCL12 (syn. SDF-1) and CXCR4 were shown to be essential for the control of B cell migration during the development of lymphoid tissues in mammals, we analyzed expression and function of this chemokine/chemokine-receptor pair in the chicken bursa. We found a strong variation of mRNA abundance of CXCL12 and CXCR4 in different stages of bursa development, with high abundance of CXCL12 mRNA in the bursa anlage at embryonic day 10 (ED10). In situ hybridization demonstrated disseminated CXCL12 expression in the early bursa anlage, which condensed in the developing follicles and was mainly restricted to the follicle cortex post-hatch. Flow cytometric analysis detected CXCR4 protein already on early B cell stages, increasing during bursal development. Post-hatch, a subpopulation with the hallmarks of emigrating B cells became detectable, which had lower CXCR4 expression, suggesting that downregulation of CXCR4 is necessary to leave the CXCL12-high bursal environment. In vivo blockade of CXCR4 using AMD3100 at the time of B cell precursor immigration strongly inhibited follicle development, demonstrating that CXCL12 attracts pre-bursal B cells into the bursal anlage. Altogether, we show that CXCL12 and its receptor CXCR4 are important for both populating the bursa with B cells and emigration of mature B cells into the periphery post hatch, and that CXCR4 function in primary B cell organs is conserved between mammals and birds.
Collapse
Affiliation(s)
- Nandor Nagy
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Florian Busalt
- Department of Veterinary Sciences, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Viktoria Halasy
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Marina Kohn
- Department of Veterinary Sciences, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Stefan Schmieder
- Department of Veterinary Sciences, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Nora Fejszak
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Bernd Kaspers
- Department of Veterinary Sciences, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Sonja Härtle
- Department of Veterinary Sciences, Ludwig-Maximilians-Universität München, Munich, Germany
| |
Collapse
|
27
|
Serrano Nájera G, Weijer CJ. Cellular processes driving gastrulation in the avian embryo. Mech Dev 2020; 163:103624. [PMID: 32562871 PMCID: PMC7511600 DOI: 10.1016/j.mod.2020.103624] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 05/18/2020] [Accepted: 05/28/2020] [Indexed: 01/18/2023]
Abstract
Gastrulation consists in the dramatic reorganisation of the epiblast, a one-cell thick epithelial sheet, into a multilayered embryo. In chick, the formation of the internal layers requires the generation of a macroscopic convection-like flow, which involves up to 50,000 epithelial cells in the epiblast. These cell movements locate the mesendoderm precursors into the midline of the epiblast to form the primitive streak. There they acquire a mesenchymal phenotype, ingress into the embryo and migrate outward to populate the inner embryonic layers. This review covers what is currently understood about how cell behaviours ultimately cause these morphogenetic events and how they are regulated. We discuss 1) how the biochemical patterning of the embryo before gastrulation creates compartments of differential cell behaviours, 2) how the global epithelial flows arise from the coordinated actions of individual cells, 3) how the cells delaminate individually from the epiblast during the ingression, and 4) how cells move after the ingression following stereotypical migration routes. We conclude by exploring new technical advances that will facilitate future research in the chick model system.
Collapse
Affiliation(s)
- Guillermo Serrano Nájera
- Division of Cell and Developmental Biology, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Cornelis J Weijer
- Division of Cell and Developmental Biology, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK.
| |
Collapse
|
28
|
Müller MR, Skowron MA, Albers P, Nettersheim D. Molecular and epigenetic pathogenesis of germ cell tumors. Asian J Urol 2020; 8:144-154. [PMID: 33996469 PMCID: PMC8099689 DOI: 10.1016/j.ajur.2020.05.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 01/24/2020] [Accepted: 04/22/2020] [Indexed: 12/18/2022] Open
Abstract
The development of germ cell tumors (GCTs) is a unique pathogenesis occurring at an early developmental stage during specification, migration or colonization of primordial germ cells (PGCs) in the genital ridge. Since driver mutations could not be identified so far, the involvement of the epigenetic machinery during the pathogenesis seems to play a crucial role. Currently, it is investigated whether epigenetic modifications occurring between the omnipotent two-cell stage and the pluripotent implanting PGCs might result in disturbances eventually leading to GCTs. Although progress in understanding epigenetic mechanisms during PGC development is ongoing, little is known about the complete picture of its involvement during GCT development and eventual classification into clinical subtypes. This review will shed light into the current knowledge of the complex epigenetic and molecular contribution during pathogenesis of GCTs by emphasizing on early developmental stages until arrival of late PGCs in the gonads. We questioned how misguided migrating and/or colonizing PGCs develop to either type I or type II GCTs. Additionally, we asked how pluripotency can be regulated during PGC development and which epigenetic changes contribute to GCT pathogenesis. We propose that SOX2 and SOX17 determine either embryonic stem cell-like (embryonal carcinoma) or PGC-like cell fate (seminoma). Finally, we suggest that factors secreted by the microenvironment, i.e. BMPs and BMP inhibiting molecules, dictate the fate decision of germ cell neoplasia in situ (into seminoma and embryonal carcinoma) and seminomas (into embryonal carcinoma or extraembryonic lineage), indicating an important role of the microenvironment on GCT plasticity.
Collapse
Affiliation(s)
- Melanie R Müller
- Department of Urology, Urological Research Lab, Translational UroOncology, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Margaretha A Skowron
- Department of Urology, Urological Research Lab, Translational UroOncology, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Peter Albers
- Department of Urology, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Daniel Nettersheim
- Department of Urology, Urological Research Lab, Translational UroOncology, University Hospital Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
29
|
Development of a transposon-based technology for transfection of day 0 chicken embryos. Gene 2020; 730:144318. [DOI: 10.1016/j.gene.2019.144318] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 12/18/2019] [Accepted: 12/20/2019] [Indexed: 12/12/2022]
|
30
|
Yaparla A, Reeves P, Grayfer L. Myelopoiesis of the Amphibian Xenopus laevis Is Segregated to the Bone Marrow, Away From Their Hematopoietic Peripheral Liver. Front Immunol 2020; 10:3015. [PMID: 32038608 PMCID: PMC6987381 DOI: 10.3389/fimmu.2019.03015] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 12/09/2019] [Indexed: 01/21/2023] Open
Abstract
Across vertebrates, hematopoiesis takes place within designated tissues, wherein committed myeloid progenitors further differentiate toward cells with megakaryocyte/erythroid potential (MEP) or those with granulocyte/macrophage potential (GMP). While the liver periphery (LP) of the Xenopus laevis amphibian functions as a principal site of hematopoiesis and contains MEPs, cells with GMP potential are instead segregated to the bone marrow (BM) of this animal. Presently, using gene expression and western blot analyses of blood cell lineage-specific transcription factors, we confirmed that while the X. laevis LP hosts hematopoietic stem cells and MEPs, their BM contains GMPs. In support of our hypothesis that cells bearing GMP potential originate from the frog LP and migrate through blood circulation to the BM in response to chemical cues; we demonstrated that medium conditioned by the X. laevis BM chemoattracts LP and peripheral blood cells. Compared to LP and by examining a comprehensive panel of chemokine genes, we showed that the X. laevis BM possessed greater expression of a single chemokine, CXCL12, the recombinant form of which was chemotactic to LP and peripheral blood cells and appeared to be a major chemotactic component within BM-conditioned medium. In confirmation of the hepatic origin of the cells that give rise to these frogs' GMPs, we also demonstrated that the X. laevis BM supported the growth of their LP-derived cells.
Collapse
Affiliation(s)
- Amulya Yaparla
- Department of Biological Sciences, George Washington University, Washington, DC, United States
| | - Phillip Reeves
- School Without Walls High School, Washington, DC, United States
| | - Leon Grayfer
- Department of Biological Sciences, George Washington University, Washington, DC, United States
| |
Collapse
|
31
|
Grimaldi C, Raz E. Germ cell migration-Evolutionary issues and current understanding. Semin Cell Dev Biol 2019; 100:152-159. [PMID: 31864795 DOI: 10.1016/j.semcdb.2019.11.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 11/25/2019] [Accepted: 11/27/2019] [Indexed: 11/19/2022]
Abstract
In many organisms, primordial germ cells (PGCs) are specified at a different location than where the gonad forms, meaning that PGCs must migrate toward the gonad within the early developing embryo. Following species-specific paths, PGCs can be passively carried by surrounding tissues and also perform active migration. When PGCs actively migrate through and along a variety of embryonic structures in different organisms, they adopt an ancestral robust migration mode termed "amoeboid motility", which allows cells to migrate within diverse environments. In this review, we discuss the possible significance of the PGC migration process in facilitating the evolution of animal body shape. In addition, we summarize the latest findings relevant for the molecular and cellular mechanisms controlling the movement and the directed migration of PGCs in different species.
Collapse
Affiliation(s)
- Cecilia Grimaldi
- Institute of Cell Biology, Center for Molecular Biology of Inflammation, University of Münster, Münster, 48149, Germany
| | - Erez Raz
- Institute of Cell Biology, Center for Molecular Biology of Inflammation, University of Münster, Münster, 48149, Germany.
| |
Collapse
|
32
|
Yoshino T, Saito D. Epithelial-to-mesenchymal transition–based morphogenesis of dorsal mesentery and gonad. Semin Cell Dev Biol 2019; 92:105-112. [DOI: 10.1016/j.semcdb.2018.09.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 08/30/2018] [Accepted: 09/02/2018] [Indexed: 12/26/2022]
|
33
|
Chen YC, Lin SP, Chang YY, Chang WP, Wei LY, Liu HC, Huang JF, Pain B, Wu SC. In vitro culture and characterization of duck primordial germ cells. Poult Sci 2019; 98:1820-1832. [PMID: 30462334 PMCID: PMC6414036 DOI: 10.3382/ps/pey515] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 10/15/2018] [Indexed: 12/15/2022] Open
Abstract
This study aimed to isolate, culture, and characterize duck primordial germ cells (PGCs) and to compare these cells with chicken PGCs. We first cultured Muscovy duck (Cairina moschata) circulating PGCs and gonadal PGCs (gPGCs) in the modified serum-containing medium used to amplify chicken PGCs. gPGCs were found to proliferate better in serum-free chemically defined medium than in serum-containing medium. Thereafter, gPGCs were similarly isolated from 2 other duck breeds, the Pekin duck (Anas platyrhynchos) and the hybrid mule duck (C. moschata × A. platyrhynchos), and amplified for a limited period of time in the chemically defined culture condition, but sufficiently to be characterized and transplanted. Cultured gPGCs of all 3 duck breeds were characterized by Periodic acid-Schiff staining, immunocytochemical staining, and expression analysis of germline-specific and pluripotency genes. Cultured duck gPGCs colonized the gonads after being genetically labeled and injected into recipient embryos. Taken together, these results demonstrate that duck PGCs retain their germline characteristics after being isolated, expanded in vitro, and genetically modified. Further studies are required to establish the optimal conditions for long-term culture of duck PGCs, which may involve supplementing the culture medium with other growth factors or compounds.
Collapse
Affiliation(s)
- Yi-Chen Chen
- Institute of Biotechnology, National Taiwan University, Taipei 10672, Taiwan.,Univ Lyon, Université Lyon 1, Stem Cell and Brain Research Institute, U1208, USC1361, INSERM, INRA, Bron 69500, France
| | - Shau-Ping Lin
- Institute of Biotechnology, National Taiwan University, Taipei 10672, Taiwan.,Agricultural Biotechnology Research Center, Academia Sinica, Taipei 11529, Taiwan
| | - Yi-Ying Chang
- Ilan Branch, Livestock Research Institute, Council of Agriculture, Executive Yuan, Ilan 26846, Taiwan
| | - Wei-Peng Chang
- Ilan Branch, Livestock Research Institute, Council of Agriculture, Executive Yuan, Ilan 26846, Taiwan
| | - Liang-Yuan Wei
- Ilan Branch, Livestock Research Institute, Council of Agriculture, Executive Yuan, Ilan 26846, Taiwan
| | - Hsiu-Chou Liu
- Ilan Branch, Livestock Research Institute, Council of Agriculture, Executive Yuan, Ilan 26846, Taiwan
| | - Jeng-Fang Huang
- Ilan Branch, Livestock Research Institute, Council of Agriculture, Executive Yuan, Ilan 26846, Taiwan.,Livestock Research Institute, Council of Agriculture, Executive Yuan, Tainan 71246, Taiwan
| | - Bertrand Pain
- Univ Lyon, Université Lyon 1, Stem Cell and Brain Research Institute, U1208, USC1361, INSERM, INRA, Bron 69500, France
| | - Shinn-Chih Wu
- Institute of Biotechnology, National Taiwan University, Taipei 10672, Taiwan.,Department of Animal Science and Technology, National Taiwan University, Taipei 10672, Taiwan
| |
Collapse
|
34
|
Yang Y, Workman S, Wilson MJ. The molecular pathways underlying early gonadal development. J Mol Endocrinol 2019; 62:R47-R64. [PMID: 30042122 DOI: 10.1530/jme-17-0314] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 07/18/2018] [Accepted: 07/24/2018] [Indexed: 12/30/2022]
Abstract
The body of knowledge surrounding reproductive development spans the fields of genetics, anatomy, physiology and biomedicine, to build a comprehensive understanding of the later stages of reproductive development in humans and animal models. Despite this, there remains much to learn about the bi-potential progenitor structure that the ovary and testis arise from, known as the genital ridge (GR). This tissue forms relatively late in embryonic development and has the potential to form either the ovary or testis, which in turn produce hormones required for the development of the rest of the reproductive tract. It is imperative that we understand the genetic networks underpinning GR development if we are to begin to understand abnormalities in the adult. This is particularly relevant in the contexts of disorders of sex development (DSDs) and infertility, two conditions that many individuals struggle with worldwide, with often no answers as to their aetiology. Here, we review what is known about the genetics of GR development. Investigating the genetic networks required for GR formation will not only contribute to our understanding of the genetic regulation of reproductive development, it may in turn open new avenues of investigation into reproductive abnormalities and later fertility issues in the adult.
Collapse
Affiliation(s)
- Yisheng Yang
- Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Stephanie Workman
- Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Megan J Wilson
- Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| |
Collapse
|
35
|
Sid H, Schusser B. Applications of Gene Editing in Chickens: A New Era Is on the Horizon. Front Genet 2018; 9:456. [PMID: 30356667 PMCID: PMC6189320 DOI: 10.3389/fgene.2018.00456] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 09/18/2018] [Indexed: 01/15/2023] Open
Abstract
The chicken represents a valuable model for research in the area of immunology, infectious diseases as well as developmental biology. Although it was the first livestock species to have its genome sequenced, there was no reverse genetic technology available to help understanding specific gene functions. Recently, homologous recombination was used to knockout the chicken immunoglobulin genes. Subsequent studies using immunoglobulin knockout birds helped to understand different aspects related to B cell development and antibody production. Furthermore, the latest advances in the field of genome editing including the CRISPR/Cas9 system allowed the introduction of site specific gene modifications in various animal species. Thus, it may provide a powerful tool for the generation of genetically modified chickens carrying resistance for certain pathogens. This was previously demonstrated by targeting the Trp38 region which was shown to be effective in the control of avian leukosis virus in chicken DF-1 cells. Herein we review the current and future prospects of gene editing and how it possibly contributes to the development of resistant chickens against infectious diseases.
Collapse
Affiliation(s)
| | - Benjamin Schusser
- Department of Animal Sciences, Reproductive Biotechnology, School of Life Sciences Weihenstephan, Technical University Munich, Freising, Germany
| |
Collapse
|
36
|
Lejong M, Choa-Duterre M, Vanmuylder N, Louryan S. Geldanamycin administration reduces the amount of primordial germ cells in the mouse embryo. Morphologie 2018; 102:219-224. [PMID: 29807855 DOI: 10.1016/j.morpho.2018.05.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 04/01/2018] [Accepted: 05/03/2018] [Indexed: 06/08/2023]
Abstract
INTRODUCTION Heat shock proteins (HSPs) are expressed or overexpressed in response to exposure to stress. They act as molecular chaperones, ensuring the correct folding of numerous client proteins. HSP90 is one of the most conserved HSPs. Its role extends beyond stress tolerance. HSP90 also contributes to development, differenciation, apoptosis and oncogenesis. Numerous tumors are associated with an overexpression of HSP90 and this expression can be used to evaluate its metastatic capacity. Primordial germ cells (PGCs) exhibit HSP90 expression under normal conditions. PGCs arise early in development and migrate by a combination of passive and active movements towards the gonads. The aim of this work was to study the impact of an inhibition of HSP90 on the migration of the PGCs. Geldanamycin, a well established HSP90 inhibitor with potent antitumor properties was used to achieve this inhibition. MATERIEL AND METHODS 5mg of Geldanamycin were administered to E8 pregnant mice. E17 embryos were removed and fixed for staining and Immunohistochemistry with anti-HSP90 and anti-VASA antibodies. RESULTS Geldanamycin-treated mouse embryos exhibited less VASA-immunopositive cells compared to the non-treated ones. These results suggest that geldanamycin administration at the time of PGCs migration reduces the number of PGCs in the gonads. HSP90 and VASA stainings were identical. We therefore expressed the idea that HSP90 could be used as a reliable marker for PGCs.
Collapse
Affiliation(s)
- M Lejong
- Laboratory of anatomy, biomechanics and organogenesis, faculty of medicine, Université Libre de Bruxelles, campus Erasme, route de Lennick, 808, 1070 Anderlecht, Belgium
| | - M Choa-Duterre
- Laboratory of anatomy, biomechanics and organogenesis, faculty of medicine, Université Libre de Bruxelles, campus Erasme, route de Lennick, 808, 1070 Anderlecht, Belgium
| | - N Vanmuylder
- Laboratory of anatomy, biomechanics and organogenesis, faculty of medicine, Université Libre de Bruxelles, campus Erasme, route de Lennick, 808, 1070 Anderlecht, Belgium
| | - S Louryan
- Laboratory of anatomy, biomechanics and organogenesis, faculty of medicine, Université Libre de Bruxelles, campus Erasme, route de Lennick, 808, 1070 Anderlecht, Belgium.
| |
Collapse
|
37
|
Jung HG, Hwang YS, Park YH, Cho HY, Rengaraj D, Han JY. Role of Epigenetic Regulation by the REST/CoREST/HDAC Corepressor Complex of Moderate NANOG Expression in Chicken Primordial Germ Cells. Stem Cells Dev 2018; 27:1215-1225. [DOI: 10.1089/scd.2018.0059] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Affiliation(s)
- Hyun Gyo Jung
- Department of Agricultural Biotechnology, Research Institute of Agriculture and Life Sciences, College of Agriculture and Life Sciences, Seoul National University, Seoul, Korea
| | - Young Sun Hwang
- Department of Agricultural Biotechnology, Research Institute of Agriculture and Life Sciences, College of Agriculture and Life Sciences, Seoul National University, Seoul, Korea
| | - Young Hyun Park
- Department of Agricultural Biotechnology, Research Institute of Agriculture and Life Sciences, College of Agriculture and Life Sciences, Seoul National University, Seoul, Korea
| | - Ho Yeon Cho
- Department of Agricultural Biotechnology, Research Institute of Agriculture and Life Sciences, College of Agriculture and Life Sciences, Seoul National University, Seoul, Korea
| | - Deivendran Rengaraj
- Department of Agricultural Biotechnology, Research Institute of Agriculture and Life Sciences, College of Agriculture and Life Sciences, Seoul National University, Seoul, Korea
| | - Jae Yong Han
- Department of Agricultural Biotechnology, Research Institute of Agriculture and Life Sciences, College of Agriculture and Life Sciences, Seoul National University, Seoul, Korea
| |
Collapse
|
38
|
Hen G, Sela-Donenfeld D. "A narrow bridge home": The dorsal mesentery in primordial germ cell migration. Semin Cell Dev Biol 2018; 92:97-104. [PMID: 30153479 DOI: 10.1016/j.semcdb.2018.08.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2018] [Revised: 08/23/2018] [Accepted: 08/23/2018] [Indexed: 01/08/2023]
Abstract
Specification of primordial germ cells (PGCs) in all vertebrates takes place in extragonadal sites. This requires migration of PGCs through embryonic tissues towards the genital ridges by both passive and active types of migration. Commonly, colonization in the genital ridges follows migration of the PGCs along the thin tissue of the dorsal mesentery. Here we review the anatomy of the dorsal mesentery, the role it plays in migration of PGCs, and the interactions of PGCs with different cell types, extracellular matrix and signaling pathways that are all essential for attraction and orientation of PGCs along the dorsal mesentery towards the gonad anlage.
Collapse
Affiliation(s)
- Gideon Hen
- Koret School of Veterinary Medicine, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Dalit Sela-Donenfeld
- Koret School of Veterinary Medicine, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel.
| |
Collapse
|
39
|
Lee JH, Park JW, Kim SW, Park J, Park TS. C-X-C chemokine receptor type 4 (CXCR4) is a key receptor for chicken primordial germ cell migration. J Reprod Dev 2017; 63:555-562. [PMID: 28867677 PMCID: PMC5735266 DOI: 10.1262/jrd.2017-067] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
In mammals, germ cells originate outside of the developing gonads and follow a unique migration pattern through the embryonic tissue toward the genital ridges. Many studies have attempted to identify critical receptors and
factors involved in germ cell migration. However, relatively few reports exist on germ cell receptors and chemokines that are involved in germ cell migration in avian species. In the present study, we investigated the specific
migratory function of C-X-C chemokine receptor type 4 (CXCR4) in chicken primordial germ cells (PGCs). We induced loss-of-function via a frameshift mutation in the CXCR4 gene in chicken PGCs using clustered
regularly interspaced short palindromic repeat-CRISPR-associated protein 9 (CRISPR/Cas9) genome editing. The migratory capacity of CXCR4 knockout PGCs was significantly reduced in vivo after
transplantation into recipient embryos. However, CXCR4-expressing somatic cell lines, such as chicken DT40 and DF1, failed to migrate into the developing gonads, suggesting that another key factor(s) is necessary for targeting and
settlement of PGCs into the genital ridges. In conclusion, we show that CXCR4 plays a critical role in the migration of chicken germ cells.
Collapse
Affiliation(s)
- Jeong Hyo Lee
- Graduate School of International Agricultural Technology and Institute of Green-Bio Science and Technology, Seoul National University, Gangwon-do 232-916, Korea
| | - Jeong-Woong Park
- Graduate School of International Agricultural Technology and Institute of Green-Bio Science and Technology, Seoul National University, Gangwon-do 232-916, Korea
| | - Si Won Kim
- Graduate School of International Agricultural Technology and Institute of Green-Bio Science and Technology, Seoul National University, Gangwon-do 232-916, Korea
| | - Joonghoon Park
- Graduate School of International Agricultural Technology and Institute of Green-Bio Science and Technology, Seoul National University, Gangwon-do 232-916, Korea
| | - Tae Sub Park
- Graduate School of International Agricultural Technology and Institute of Green-Bio Science and Technology, Seoul National University, Gangwon-do 232-916, Korea
| |
Collapse
|
40
|
A pilgrim's progress: Seeking meaning in primordial germ cell migration. Stem Cell Res 2017; 24:181-187. [PMID: 28754603 DOI: 10.1016/j.scr.2017.07.017] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 06/08/2017] [Accepted: 07/15/2017] [Indexed: 01/08/2023] Open
Abstract
Comparative studies of primordial germ cell (PGC) development across organisms in many phyla reveal surprising diversity in the route of migration, timing and underlying molecular mechanisms, suggesting that the process of migration itself is conserved. However, beyond the perfunctory transport of cellular precursors to their later arising home of the gonads, does PGC migration serve a function? Here we propose that the process of migration plays an additional role in quality control, by eliminating PGCs incapable of completing migration as well as through mechanisms that favor PGCs capable of responding appropriately to migration cues. Focusing on PGCs in mice, we explore evidence for a selective capacity of migration, considering the tandem regulation of proliferation and migration, cell-intrinsic and extrinsic control, the potential for tumors derived from failed PGC migrants, the potential mechanisms by which migratory PGCs vary in their cellular behaviors, and corresponding effects on development. We discuss the implications of a selective role of PGC migration for in vitro gametogenesis.
Collapse
|
41
|
Jung KM, Kim YM, Ono T, Han JY. Size-dependent isolation of primordial germ cells from avian species. Mol Reprod Dev 2017; 84:508-516. [DOI: 10.1002/mrd.22802] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 02/28/2017] [Accepted: 03/20/2017] [Indexed: 11/10/2022]
Affiliation(s)
- Kyung M. Jung
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences; College of Agriculture and Life Sciences; Seoul National University; Seoul Korea
| | - Young M. Kim
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences; College of Agriculture and Life Sciences; Seoul National University; Seoul Korea
| | - Tamao Ono
- Division of Animal Science; Faculty of Agriculture; Shinshu University; Minamiminowa Nagano Japan
| | - Jae Y. Han
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences; College of Agriculture and Life Sciences; Seoul National University; Seoul Korea
- Institute for Biomedical Sciences; Shinshu University; Minamiminowa Nagano Japan
| |
Collapse
|
42
|
Sasado T, Kondoh H, Furutani-Seiki M, Naruse K. Mutation in cpsf6/CFIm68 (Cleavage and Polyadenylation Specificity Factor Subunit 6) causes short 3'UTRs and disturbs gene expression in developing embryos, as revealed by an analysis of primordial germ cell migration using the medaka mutant naruto. PLoS One 2017; 12:e0172467. [PMID: 28253363 PMCID: PMC5333813 DOI: 10.1371/journal.pone.0172467] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 02/06/2017] [Indexed: 02/02/2023] Open
Abstract
Our previous studies analyzing medaka mutants defective in primordial germ cell (PGC) migration identified cxcr4b and cxcr7, which are both receptors of the chemokine sdf1/cxcl12, as key regulators of PGC migration. Among PGC migration mutants, naruto (nar) is unique in that the mutant phenotype includes gross morphological abnormalities of embryos, suggesting that the mutation affects a broader range of processes. A fine genetic linkage mapping and genome sequencing showed the nar gene encodes Cleavage and Polyadenylation Specificity Factor subunit 6 (CPSF6/CFIm68). CPSF6 is a component of the Cleavage Factor Im complex (CFIm) which plays a key role in pre-mRNA 3'-cleavage and polyadenylation. 3'RACE of sdf1a/b and cxcr7 transcripts in the mutant embryos indicated shorter 3'UTRs with poly A additions occurring at more upstream positions than wild-type embryos, suggesting CPSF6 functions to prevent premature 3'UTR cleavage. In addition, expression of the coding region sequences of sdf1a/b in nar mutants was more anteriorly extended in somites than wild-type embryos, accounting for the abnormally extended distribution of PGCs in nar mutants. An expected consequence of shortening 3'UTR is the escape from the degradation mechanism mediated by microRNAs interacting with distal 3'UTR sequence. The abnormal expression pattern of sdf1a coding sequence may be at least partially accounted for by this mechanism. Given the pleiotropic effects of nar mutation, further analysis using the nar mutant will reveal processes in which CPSF6 plays essential regulatory roles in poly A site selection and involvement of 3'UTRs in posttranscriptional gene regulation in various genes in vivo.
Collapse
Affiliation(s)
- Takao Sasado
- Laboratory of Bioresources, National Institute for Basic Biology, Aichi, Japan
| | - Hisato Kondoh
- Faculty of Life Sciences, Kyoto Sangyo University, Kyoto, Japan
| | | | - Kiyoshi Naruse
- Laboratory of Bioresources, National Institute for Basic Biology, Aichi, Japan
| |
Collapse
|
43
|
Loveland KL, Klein B, Pueschl D, Indumathy S, Bergmann M, Loveland BE, Hedger MP, Schuppe HC. Cytokines in Male Fertility and Reproductive Pathologies: Immunoregulation and Beyond. Front Endocrinol (Lausanne) 2017; 8:307. [PMID: 29250030 PMCID: PMC5715375 DOI: 10.3389/fendo.2017.00307] [Citation(s) in RCA: 131] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Accepted: 10/23/2017] [Indexed: 12/22/2022] Open
Abstract
Germline development in vivo is dependent on the environment formed by somatic cells and the differentiation cues they provide; hence, the impact of local factors is highly relevant to the production of sperm. Knowledge of how somatic and germline cells interact is central to achieving biomedical goals relating to restoring, preserving or restricting fertility in humans. This review discusses the growing understanding of how cytokines contribute to testicular function and maintenance of male reproductive health, and to the pathologies associated with their abnormal activity in this organ. Here we consider both cytokines that signal through JAKs and are regulated by SOCS, and those utilizing other pathways, such as the MAP kinases and SMADs. The importance of cytokines in the establishment and maintenance of the testis as an immune-privilege site are described. Current research relating to the involvement of immune cells in testis development and disease is highlighted. This includes new data relating to testicular cancer which reinforce the understanding that tumorigenic cells shape their microenvironment through cytokine actions. Clinical implications in pathologies relating to local inflammation and to immunotherapies are discussed.
Collapse
Affiliation(s)
- Kate L. Loveland
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Molecular and Translational Sciences, School of Clinical Sciences, Monash Medical Centre, Monash University, Clayton, VIC, Australia
- Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia
- *Correspondence: Kate L. Loveland,
| | - Britta Klein
- Institute of Veterinary Anatomy, Histology and Embryology, Justus Liebig University Giessen, Giessen, Germany
- Institute of Anatomy and Cell Biology, Justus Liebig University Giessen, Giessen, Germany
| | - Dana Pueschl
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Molecular and Translational Sciences, School of Clinical Sciences, Monash Medical Centre, Monash University, Clayton, VIC, Australia
- Institute of Veterinary Anatomy, Histology and Embryology, Justus Liebig University Giessen, Giessen, Germany
| | - Sivanjah Indumathy
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Molecular and Translational Sciences, School of Clinical Sciences, Monash Medical Centre, Monash University, Clayton, VIC, Australia
- Institute of Veterinary Anatomy, Histology and Embryology, Justus Liebig University Giessen, Giessen, Germany
| | - Martin Bergmann
- Institute of Veterinary Anatomy, Histology and Embryology, Justus Liebig University Giessen, Giessen, Germany
| | | | - Mark P. Hedger
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Molecular and Translational Sciences, School of Clinical Sciences, Monash Medical Centre, Monash University, Clayton, VIC, Australia
| | - Hans-Christian Schuppe
- Department of Urology, Pediatric Urology and Andrology, Justus Liebig University Giessen, Giessen, Germany
| |
Collapse
|
44
|
Tagami T, Miyahara D, Nakamura Y. Avian Primordial Germ Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1001:1-18. [PMID: 28980226 DOI: 10.1007/978-981-10-3975-1_1] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
Germ cells transmit genetic information to the next generation through gametogenesis. Primordial germ cells (PGCs) are the first germ-cell population established during development, and are the common origins of both oocytes and spermatogonia. Unlike in other species, PGCs in birds undergo blood circulation to migrate toward the genital ridge, and are one of the major biological properties of avian PGCs. Germ cells enter meiosis and arrest at prophase I during embryogenesis in females, whereas in males they enter mitotic arrest during embryogenesis and enter meiosis only after birth. In chicken, gonadal sex differentiation occurs as early as embryonic day 6, but meiotic initiation of female germ cells starts from a relatively late stage (embryonic day 15.5). Retinoic acid controls meiotic entry in developing chicken gonads through the expressions of retinaldehyde dehydrogenase 2, a major retinoic acid synthesizing enzyme, and cytochrome P450 family 26, subfamily B member 1, a major retinoic acid-degrading enzyme. The other major biological property of avian PGCs is that they can be propagated in vitro for the long term, and this technique is useful for investigating proliferation mechanisms. The main factor involved in chicken PGC proliferation is fibroblast growth factor 2, which activates the signaling of MEK/ERK and thus promotes the cell cycle and anti-apoptosis. Furthermore, the activation of PI3K/Akt signaling is indispensable for the proliferation and survival of chicken PGCs.
Collapse
Affiliation(s)
- Takahiro Tagami
- Institute of Livestock Grassland Science, NARO, Ibaraki, Japan.
| | - Daichi Miyahara
- Institute of Livestock Grassland Science, NARO, Ibaraki, Japan
- Shinshu University, Ueda, Japan
| | | |
Collapse
|
45
|
Zaman P, Wang J, Blau A, Wang W, Li T, Kohane DS, Loscalzo J, Zhang YY. Incorporation of heparin-binding proteins into preformed dextran sulfate-chitosan nanoparticles. Int J Nanomedicine 2016; 11:6149-6159. [PMID: 27920522 PMCID: PMC5125769 DOI: 10.2147/ijn.s119174] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Incorporation of proteins into dextran sulfate (DS)-chitosan (CS) nanoparticles (DSCS NPs) is commonly performed using entrapment procedures, in which protein molecules are mixed with DS and CS until particle formation occurs. As DS is an analog of heparin, the authors examined whether proteins could be directly incorporated into preformed DSCS NPs through a heparin binding domain-mediated interaction. The authors formulated negatively-charged DSCS NPs, and quantified the amount of charged DS in the outer shell of the particles. The authors then mixed the DSCS NPs with heparin-binding proteins (SDF-1α, VEGF, FGF-2, BMP-2, or lysozyme) to achieve incorporation. Data show that for DSCS NPs containing 100 nmol charged glucose sulfate units in DS, up to ~1.5 nmol of monomeric or ~0.75 nmol of dimeric heparin-binding proteins were incorporated without significantly altering the size or zeta potential of the particles. Incorporation efficiencies of these proteins were 95%–100%. In contrast, serum albumin or serum globulin showed minimal incorporation (8% and 4%, respectively) in 50% physiological saline, despite their large adsorption in water (80% and 92%, respectively). The NP-incorporated SDF-1α and VEGF exhibited full activity and sustained thermal stability. An in vivo aerosolization study showed that NP-incorporated SDF-1α persisted in rat lungs for 72 h (~34% remaining), while free SDF-1α was no longer detectable after 16 h. As many growth factors and cytokines contain heparin-binding sites/domains, incorporation into preformed DSCS NPs could facilitate in vivo applications of these proteins.
Collapse
Affiliation(s)
- Paula Zaman
- Department of Medicine, Brigham and Women's Hospital
| | - Julia Wang
- Department of Medicine, Brigham and Women's Hospital
| | - Adam Blau
- Department of Medicine, Brigham and Women's Hospital
| | - Weiping Wang
- Department of Anesthesiology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Tina Li
- Department of Medicine, Brigham and Women's Hospital
| | - Daniel S Kohane
- Department of Anesthesiology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Ying-Yi Zhang
- Department of Medicine, Brigham and Women's Hospital
| |
Collapse
|
46
|
Yoshino T, Murai H, Saito D. Hedgehog-BMP signalling establishes dorsoventral patterning in lateral plate mesoderm to trigger gonadogenesis in chicken embryos. Nat Commun 2016; 7:12561. [PMID: 27558761 PMCID: PMC5007334 DOI: 10.1038/ncomms12561] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2015] [Accepted: 07/11/2016] [Indexed: 12/31/2022] Open
Abstract
The gonad appears in the early embryo after several events: cells at the lateral plate mesoderm (LPM) undergo ingression, begin gonadal differentiation and then retain primordial germ cells (PGCs). Here we show that in the chicken embryo, these events are triggered on the basis of dorsoventral patterning at the medial LPM. Gonadal progenitor cells (GPCs) at the ventromedial LPM initiate gonadogenesis by undergoing ingression, whereas mesonephric capsule progenitor cells (MCPCs) at the dorsomedial LPM do not. These contrasting behaviours are caused by Hedgehog signalling, which is activated in GPCs but not in MCPCs. Inhibiting Hedgehog signalling prevents GPCs from forming gonadal structures and collecting PGCs. When activated by Hedgehog signalling, MCPCs form an ectopic gonad. This Hedgehog signalling is mediated by BMP4. These findings provide insight into embryonic patterning and gonadal initiation in the chicken embryo. Ingression of cells from the lateral plate mesoderm (LPM) initiates gonad differentiation but how these events are triggered is unclear. Here, the authors show that gonadal progenitor cells at the ventromedial LPM initiate gonadogenesis, and are activated by Hedgehog and BMP4 signalling.
Collapse
Affiliation(s)
- Takashi Yoshino
- Department of Zoology, Graduate School of Science, Kyoto University, Kitashirakawa, Sakyo-ku, Kyoto 606-8502, Japan
| | - Hidetaka Murai
- Frontier Research Institute for Interdisciplinary Sciences (FRIS), Tohoku University, Aoba-ku, Sendai 980-8578, Japan
| | - Daisuke Saito
- Frontier Research Institute for Interdisciplinary Sciences (FRIS), Tohoku University, Aoba-ku, Sendai 980-8578, Japan
| |
Collapse
|
47
|
Barton LJ, LeBlanc MG, Lehmann R. Finding their way: themes in germ cell migration. Curr Opin Cell Biol 2016; 42:128-137. [PMID: 27484857 DOI: 10.1016/j.ceb.2016.07.007] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 06/15/2016] [Accepted: 07/08/2016] [Indexed: 11/26/2022]
Abstract
Embryonic germ cell migration is a vital component of the germline lifecycle. The translocation of germ cells from the place of origin to the developing somatic gonad involves several processes including passive movements with underlying tissues, transepithelial migration, cell adhesion dynamics, the establishment of environmental guidance cues and the ability to sustain directed migration. How germ cells accomplish these feats in established model organisms will be discussed in this review, with a focus on recent discoveries and themes conserved across species.
Collapse
Affiliation(s)
- Lacy J Barton
- HHMI and Skirball Institute at NYU School of Medicine, 540 First Avenue, New York, NY 10016, United States
| | - Michelle G LeBlanc
- HHMI and Skirball Institute at NYU School of Medicine, 540 First Avenue, New York, NY 10016, United States
| | - Ruth Lehmann
- HHMI and Skirball Institute at NYU School of Medicine, 540 First Avenue, New York, NY 10016, United States.
| |
Collapse
|
48
|
NAKAJIMA Y, FUKUDA H, ONUMA M, MURATA K, UEDA M, SUNAGA E, SHIRAISHI T, TAJIMA A. Migratory ability of gonadal germ cells (GGCs) isolated from Ciconia boyciana and Geronticus eremita embryos into the gonad of developing chicken embryos. J Vet Med Sci 2016; 78:1055-8. [PMID: 26922915 PMCID: PMC4937143 DOI: 10.1292/jvms.15-0664] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 02/12/2016] [Indexed: 11/29/2022] Open
Abstract
We conducted experiments to evaluate the ability of gonadal germ cells (GGCs), isolated from the embryonic gonads of Ciconia boyciana or Geronticus eremita, to migrate into the gonads of developing chicken embryos. Fluorescently labeled GGCs, isolated by the PBS (-) method, were transferred into the dorsal aorta of 2-day-old chicken embryos. Five days after transfer, fluorescent GGCs were detected in the gonads of recipient embryos. Our results indicate that GGCs from Ciconia boyciana and Geronticus eremita are capable of migrating into the gonads of developing chicken embryos.
Collapse
Affiliation(s)
- Yuki NAKAJIMA
- Faculty of Life and Environmental Sciences, University of Tsukuba, Ten-noh Dai
1–1–1, Tsukuba, Ibaraki 305–8572, Japan
- Fellow of the Japanese Society for the Promotion of Science, Japan
- Current address: Advanced Biological Information Research Division, INAMORI
Frontier Research Center, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819–0395, Japan
| | - Haruka FUKUDA
- Graduate School of Life and Environmental Sciences, University of Tsukuba,
Ten-noh Dai 1–1–1, Tsukuba, Ibaraki 305–8572, Japan
| | - Manabu ONUMA
- Center for Environmental Biology and Ecosystem Studies, National Institute for
Environmental Studies, 16–2 Onogawa, Tsukuba, Ibaraki 305–8506, Japan
| | - Koichi MURATA
- Zoorasia Yokohama Zoological Gardens, Kamishiranecho 1175–1, Yokohama Asahi-ku,
Kanagawa 241–0001, Japan
- Preservation and Research Center/The City of Yokohama, Kawaishukucho 155–1,
Yokohama Asahi-ku, Kanagawa 241–0804, Japan
- College of Bioresource Sciences, Nihon University, Kameino 1866, Fujisawa,
Kanagawa, 252–0880, Japan
| | - Miya UEDA
- Zoorasia Yokohama Zoological Gardens, Kamishiranecho 1175–1, Yokohama Asahi-ku,
Kanagawa 241–0001, Japan
| | - Emi SUNAGA
- Preservation and Research Center/The City of Yokohama, Kawaishukucho 155–1,
Yokohama Asahi-ku, Kanagawa 241–0804, Japan
| | - Toshirou SHIRAISHI
- Preservation and Research Center/The City of Yokohama, Kawaishukucho 155–1,
Yokohama Asahi-ku, Kanagawa 241–0804, Japan
| | - Atsushi TAJIMA
- Faculty of Life and Environmental Sciences, University of Tsukuba, Ten-noh Dai
1–1–1, Tsukuba, Ibaraki 305–8572, Japan
| |
Collapse
|
49
|
Paksa A, Bandemer J, Hoeckendorf B, Razin N, Tarbashevich K, Minina S, Meyen D, Biundo A, Leidel SA, Peyrieras N, Gov NS, Keller PJ, Raz E. Repulsive cues combined with physical barriers and cell-cell adhesion determine progenitor cell positioning during organogenesis. Nat Commun 2016; 7:11288. [PMID: 27088892 PMCID: PMC4837475 DOI: 10.1038/ncomms11288] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 03/09/2016] [Indexed: 01/15/2023] Open
Abstract
The precise positioning of organ progenitor cells constitutes an essential, yet poorly understood step during organogenesis. Using primordial germ cells that participate in gonad formation, we present the developmental mechanisms maintaining a motile progenitor cell population at the site where the organ develops. Employing high-resolution live-cell microscopy, we find that repulsive cues coupled with physical barriers confine the cells to the correct bilateral positions. This analysis revealed that cell polarity changes on interaction with the physical barrier and that the establishment of compact clusters involves increased cell–cell interaction time. Using particle-based simulations, we demonstrate the role of reflecting barriers, from which cells turn away on contact, and the importance of proper cell–cell adhesion level for maintaining the tight cell clusters and their correct positioning at the target region. The combination of these developmental and cellular mechanisms prevents organ fusion, controls organ positioning and is thus critical for its proper function. The precise positioning of organ progenitor cells is essential for organ development and function. Here the authors use live imaging and mathematical modelling to show that the confinement of a motile progenitor cell population results from coupled physical barriers and cell-cell interactions.
Collapse
Affiliation(s)
- Azadeh Paksa
- Institute for Cell Biology, ZMBE, Von-Esmarch-Street 56, 48149 Muenster, Germany
| | - Jan Bandemer
- Institute for Cell Biology, ZMBE, Von-Esmarch-Street 56, 48149 Muenster, Germany
| | | | - Nitzan Razin
- Department of Chemical Physics, Weizmann Institute of Science, Rehovot 76100, Israel
| | | | - Sofia Minina
- Germ Cell Development, Max-Planck Institute of Biophysical Chemistry, Am Fassberg 11, 37070 Göttingen, Germany
| | - Dana Meyen
- Institute for Cell Biology, ZMBE, Von-Esmarch-Street 56, 48149 Muenster, Germany
| | - Antonio Biundo
- Max Planck Research Group for RNA Biology, Max Planck Institute for Molecular Biomedicine, Von-Esmarch-Strasse 54, 48149 Muenster, Germany
| | - Sebastian A Leidel
- Max Planck Research Group for RNA Biology, Max Planck Institute for Molecular Biomedicine, Von-Esmarch-Strasse 54, 48149 Muenster, Germany
| | - Nadine Peyrieras
- USR3695 BioEmergences, CNRS, Université Paris-Saclay, Avenue de la Terrasse, 91190 Gif-sur-Yvette, France
| | - Nir S Gov
- Department of Chemical Physics, Weizmann Institute of Science, Rehovot 76100, Israel
| | | | - Erez Raz
- Institute for Cell Biology, ZMBE, Von-Esmarch-Street 56, 48149 Muenster, Germany
| |
Collapse
|
50
|
Es-Haghi M, Bassami M, Dehghani H. Construction and Quantitative Validation of Chicken CXCR4 Expression Reporter. Mol Biotechnol 2016; 58:202-11. [PMID: 26809356 DOI: 10.1007/s12033-016-9917-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Site directional migration is an important biological event and an essential behavior for latent migratory cells. A migratory cell maintains its motility, survival, and proliferation abilities by a network of signaling pathways where CXCR4/SDF signaling route plays crucial role for directed homing of a polarized cell. The chicken embryo due to its specific vasculature modality has been used as a valuable model for organogenesis, migration, cancer, and metastasis. In this research, the regulatory regions of chicken CXCR4 gene have been characterized in a chicken hematopoietic lymphoblast cell line (MSB1). A region extending from -2000 bp upstream of CXCR4 gene to +68 after its transcriptional start site, in addition to two other mutant fragments were constructed and cloned in a promoter-less reporter vector. Promoter activity was analyzed by quantitative real-time RT-PCR and flow cytometry techniques. Our findings show that the full sequence from -2000 to +68 bp of CXCR4 regulatory region is required for maximum promoter functionality, while the mutant CXCR4 promoter fragments show a partial promoter activity. The chicken CXCR4 promoter validated in this study could be used for characterization of directed migratory cells in chicken development and disease models.
Collapse
Affiliation(s)
- Masoumeh Es-Haghi
- Division of Biotechnology, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Mohammadreza Bassami
- Division of Biotechnology, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Hesam Dehghani
- Division of Biotechnology, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran.
- Embryonic and Stem Cell Biology and Biotechnology Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran.
| |
Collapse
|