51
|
Tsai TS, Johnson J, White Y, John JC. The molecular characterization of porcine egg precursor cells. Oncotarget 2017; 8:63484-63505. [PMID: 28969006 PMCID: PMC5609938 DOI: 10.18632/oncotarget.18833] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 06/03/2017] [Indexed: 12/12/2022] Open
Abstract
Female-factor infertility can be caused by poor oocyte quality and depleted ovarian reserves. Egg precursor cells (EPCs), isolated from the ovarian cortex, have the potential to be used to overcome female infertility. We aimed to define the origins of EPCs by analyzing their gene expression profiles and mtDNA content using a mini-pig model. We characterized FAC-sorted DDX4+-derived porcine EPCs by performing RNA-sequencing and determined that they utilize pathways important for cell cycle and proliferation, which supports the existence of adult mitotically active oogonial cells. Expression of the pluripotent markers Sox2 and Oct4, and the primitive germ cell markers Blimp1 and Stella were not detected. However, Nanog and Ddx4 were expressed, as were the primitive germ cell markers Fragilis, c-Kit and Tert. Moreover, porcine EPCs expressed self-renewal and proliferation markers including Myc, Esrrb, Id2, Klf4, Klf5, Stat3, Fgfr1, Fgfr2 and Il6st. The presence of Zp1, Zp2, Zp3 and Nobox were not detected, indicating that porcine EPCs are not indicative of mature primordial oocytes. We performed mitochondrial DNA Next Generation Sequencing and determined that one mtDNA variant harbored by EPCs was present in oocytes, preimplantation embryos and somatic tissues over three generations in our mini-pig model indicating the potential germline origin of EPCs.
Collapse
Affiliation(s)
- Te-Sha Tsai
- Centre for Genetic Diseases, Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia.,Centre for Genetic Diseases, Department of Molecular and Translational Science, Monash University, Clayton, Victoria 3168, Australia
| | - Jacqueline Johnson
- Centre for Genetic Diseases, Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia
| | | | - Justin C John
- Centre for Genetic Diseases, Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia.,Centre for Genetic Diseases, Department of Molecular and Translational Science, Monash University, Clayton, Victoria 3168, Australia
| |
Collapse
|
52
|
Esfandiari F, Ashtiani MK, Sharifi‐Tabar M, Saber M, Daemi H, Ghanian MH, Shahverdi A, Baharvand H. Microparticle‐Mediated Delivery of BMP4 for Generation of Meiosis‐Competent Germ Cells from Embryonic Stem Cells. Macromol Biosci 2017; 17. [DOI: 10.1002/mabi.201600284] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Revised: 08/17/2016] [Indexed: 01/13/2023]
Abstract
Producing meiosis‐competent germ cells (GCs) from embryonic stem cells (ESCs) is essential for developing advanced therapies for infertility. Here, a novel approach is presented for generation of GCs from ESCs. In this regard, microparticles (MPs) have been developed from alginate sulfate loaded with bone morphogenetic protein 4 (BMP4). The results here show that BMP4 release from alginate sulfate MPs is significantly retarded by the sulfated groups compared to neat alginate. Then, BMP4‐laden MPs are incorporated within the aggregates during differentiation of GCs from ESCs. It is observed that BMP4‐laden MPs increase GC differentiation from ESCs at least twofold compared to the conventional soluble delivery method. Interestingly, following meiosis induction,Dazl, an intrinsic factor that enables GCs to enter meiosis, and two essential meiosis genes (Stra8andSmc1b) are upregulated significantly in MP‐induced aggregates compared to aggregates, which are formed by the conventional method. Together, these data show that controlled delivery of BMP4 during ESC differentiation into GC establish meiosis‐competent GCs which can serve as an attractive GC source for reproductive medicine.image
Collapse
Affiliation(s)
- Fereshteh Esfandiari
- Department of Stem Cells and Developmental Biology Cell Science Research Center Royan Institute for Stem Cell Biology and Technology ACECR Tehran 1665659911 Iran
- Department of Developmental Biology University of Science and Culture Tehran 1461968151 Iran
| | - Mohammad Kazemi Ashtiani
- Department of Stem Cells and Developmental Biology Cell Science Research Center Royan Institute for Stem Cell Biology and Technology ACECR Tehran 1665659911 Iran
| | - Mehdi Sharifi‐Tabar
- Department of Molecular Systems Biology Cell Science Research Center Royan Institute for Stem Cell Biology and Technology ACECR Tehran 1665659911 Iran
| | - Maryam Saber
- Department of Stem Cells and Developmental Biology Cell Science Research Center Royan Institute for Stem Cell Biology and Technology ACECR Tehran 1665659911 Iran
| | - Hamed Daemi
- Department of Stem Cells and Developmental Biology Cell Science Research Center Royan Institute for Stem Cell Biology and Technology ACECR Tehran 1665659911 Iran
| | - Mohammad Hossein Ghanian
- Department of Stem Cells and Developmental Biology Cell Science Research Center Royan Institute for Stem Cell Biology and Technology ACECR Tehran 1665659911 Iran
| | - Abdolhossein Shahverdi
- Department of Embryology Reproductive Biomedicine Research Center Royan Institute for Reproductive Biomedicine ACECR Tehran 1665659911 Iran
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology Cell Science Research Center Royan Institute for Stem Cell Biology and Technology ACECR Tehran 1665659911 Iran
- Department of Developmental Biology University of Science and Culture Tehran 1461968151 Iran
| |
Collapse
|
53
|
Hassan W, Viebahn C. A correlative study of the allantois in pig and rabbit highlighting the diversity of extraembryonic tissues in four mammalian species, including mouse and man. J Morphol 2017; 278:600-620. [PMID: 28165148 DOI: 10.1002/jmor.20657] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 12/30/2016] [Accepted: 01/04/2017] [Indexed: 12/25/2022]
Abstract
Despite its conserved role in placenta and umbilical cord formation, the mammalian allantois shows remarkable diversity in size and form as well as in the timing of its appearance and attachment to the chorion. In the mouse, the common allantoic diverticulum is lacking; instead, the allantoic core domain is defined as a progenitor center for allantoic development. In this study, the allantoises of the pig and the rabbit as two nonrodent mammals of increasing significance in biomedical research are compared (1) morphologically using high resolution light and electron microscopy and (2) molecularly using brachyury mRNA expression as a mesodermal marker. Multiple small allantoic diverticula in the rabbit contrast with a single large cavity filling the entire allantois of the pig, but neither pig nor rabbit allantois expresses brachyury. The mesothelium on the allantois surface shows regional variability of cell contacts and microvilli, while blood vessels appear randomly around the allantoic diverticula in a mesodermal layer of variable thickness. Primordial germ cell-like cells are found in the allantois of the pig but not of the rabbit. To understand further the relevance of this developmental and morphological diversity, we compare the allantois development of pig and rabbit with early developmental landmarks of mouse and man. Our findings suggest that (1) tissue interaction between endoderm and mesoderm is important for allantoic development and vascular differentiation in species with a rudimentary allantoic diverticulum, (2) allantoic mesothelium plays a specific role in chorioallantoic attachment, allantoic differentiation and vascularization, and (3) there is a pronounced diversity in the extraembryonic migratory pathways of primordial germ cells among mammals. Finally, the phylogenetically basal characteristics of the pig allantois are suggestive of a functional similarity in mammals with a large allantois before placentation and in (aplacental) sauropsids with a chorioallantoic membrane well-adjusted to material exchange function.
Collapse
Affiliation(s)
- Waad Hassan
- Institute of Anatomy and Embryology, University Medical Center Göttingen, Göttingen, Germany
| | - Christoph Viebahn
- Institute of Anatomy and Embryology, University Medical Center Göttingen, Göttingen, Germany
| |
Collapse
|
54
|
Abstract
The ovary, the female gonad, serves as the source for the germ cells as well as the major supplier of steroid sex hormones. During embryonic development, the primordial germ cells (PGCs) are specified, migrate to the site of the future gonad, and proliferate, forming structures of germ cells nests, which will eventually break down to generate the primordial follicles (PMFs). Each PMF contains an oocyte arrested at the first prophase of meiosis, surrounded by a flattened layer of somatic pre-granulosa cells. Most of the PMFs are kept dormant and only a selected population is activated to join the growing pool of follicles in a process regulated by both intra- and extra-oocyte factors. The PMFs will further develop into secondary pre-antral follicles, a stage which depends on bidirectional communication between the oocyte and the surrounding somatic cells. Many of the signaling molecules involved in this dialog belong to the transforming growth factor β (TGF-β) superfamily. As the follicle continues to develop, a cavity called antrum is formed. The resulting antral follicles relay on the pituitary gonadotropins, follicle-stimulating hormone (FSH), and luteinizing hormone (LH) for their development. Most of the follicles undergo atretic degeneration and only a subset of the antral follicles, known as the dominant follicles, will reach the preovulatory stage at each reproductive cycle, respond to LH, and subsequently ovulate, releasing a fertilizable oocyte. The remaining somatic cells in the raptured follicle will undergo terminal differentiation and form the corpus luteum, which secretes progesterone necessary to maintain pregnancy.
Collapse
|
55
|
Ratajczak MZ, Ratajczak J, Suszynska M, Miller DM, Kucia M, Shin DM. A Novel View of the Adult Stem Cell Compartment From the Perspective of a Quiescent Population of Very Small Embryonic-Like Stem Cells. Circ Res 2017; 120:166-178. [PMID: 28057792 PMCID: PMC5221475 DOI: 10.1161/circresaha.116.309362] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2016] [Revised: 11/16/2016] [Accepted: 11/17/2016] [Indexed: 12/12/2022]
Abstract
Evidence has accumulated that adult hematopoietic tissues and other organs contain a population of dormant stem cells (SCs) that are more primitive than other, already restricted, monopotent tissue-committed SCs (TCSCs). These observations raise several questions, such as the developmental origin of these cells, their true pluripotent or multipotent nature, which surface markers they express, how they can be efficiently isolated from adult tissues, and what role they play in the adult organism. The phenotype of these cells and expression of some genes characteristic of embryonic SCs, epiblast SCs, and primordial germ cells suggests their early-embryonic deposition in developing tissues as precursors of adult SCs. In this review, we will critically discuss all these questions and the concept that small dormant SCs related to migratory primordial germ cells, described as very small embryonic-like SCs, are deposited during embryogenesis in bone marrow and other organs as a backup population for adult tissue-committed SCs and are involved in several processes related to tissue or organ rejuvenation, aging, and cancerogenesis. The most recent results on successful ex vivo expansion of human very small embryonic-like SC in chemically defined media free from feeder-layer cells open up new and exciting possibilities for their application in regenerative medicine.
Collapse
Affiliation(s)
- Mariusz Z Ratajczak
- From the Department of Medicine, Stem Cell Biology Program at the James Graham Brown Cancer Center, University of Louisville, KY (M.Z.R., J.R., M.S., D.M.M., M.K.); Department of Regenerative Medicine, Warsaw Medical University, Poland (M.Z.R., M.K.); and Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul, South Korea (D.-M.S.).
| | - Janina Ratajczak
- From the Department of Medicine, Stem Cell Biology Program at the James Graham Brown Cancer Center, University of Louisville, KY (M.Z.R., J.R., M.S., D.M.M., M.K.); Department of Regenerative Medicine, Warsaw Medical University, Poland (M.Z.R., M.K.); and Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul, South Korea (D.-M.S.)
| | - Malwina Suszynska
- From the Department of Medicine, Stem Cell Biology Program at the James Graham Brown Cancer Center, University of Louisville, KY (M.Z.R., J.R., M.S., D.M.M., M.K.); Department of Regenerative Medicine, Warsaw Medical University, Poland (M.Z.R., M.K.); and Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul, South Korea (D.-M.S.)
| | - Donald M Miller
- From the Department of Medicine, Stem Cell Biology Program at the James Graham Brown Cancer Center, University of Louisville, KY (M.Z.R., J.R., M.S., D.M.M., M.K.); Department of Regenerative Medicine, Warsaw Medical University, Poland (M.Z.R., M.K.); and Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul, South Korea (D.-M.S.)
| | - Magda Kucia
- From the Department of Medicine, Stem Cell Biology Program at the James Graham Brown Cancer Center, University of Louisville, KY (M.Z.R., J.R., M.S., D.M.M., M.K.); Department of Regenerative Medicine, Warsaw Medical University, Poland (M.Z.R., M.K.); and Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul, South Korea (D.-M.S.)
| | - Dong-Myung Shin
- From the Department of Medicine, Stem Cell Biology Program at the James Graham Brown Cancer Center, University of Louisville, KY (M.Z.R., J.R., M.S., D.M.M., M.K.); Department of Regenerative Medicine, Warsaw Medical University, Poland (M.Z.R., M.K.); and Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul, South Korea (D.-M.S.)
| |
Collapse
|
56
|
Mechanisms of Vertebrate Germ Cell Determination. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 953:383-440. [PMID: 27975276 DOI: 10.1007/978-3-319-46095-6_8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Two unique characteristics of the germ line are the ability to persist from generation to generation and to retain full developmental potential while differentiating into gametes. How the germ line is specified that allows it to retain these characteristics within the context of a developing embryo remains unknown and is one focus of current research. Germ cell specification proceeds through one of two basic mechanisms: cell autonomous or inductive. Here, we discuss how germ plasm driven germ cell specification (cell autonomous) occurs in both zebrafish and the frog Xenopus. We describe the segregation of germ cells during embryonic development of solitary and colonial ascidians to provide an evolutionary context to both mechanisms. We conclude with a discussion of the inductive mechanism as exemplified by both the mouse and axolotl model systems. Regardless of mechanism, several general themes can be recognized including the essential role of repression and posttranscriptional regulation of gene expression.
Collapse
|
57
|
Irie N, Surani MA. Efficient Induction and Isolation of Human Primordial Germ Cell-Like Cells from Competent Human Pluripotent Stem Cells. Methods Mol Biol 2017; 1463:217-226. [PMID: 27734359 DOI: 10.1007/978-1-4939-4017-2_16] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
We recently reported a robust and defined culture system for the specification of human primordial germ cell-like cells (hPGCLCs) from human pluripotent stem cells (hPSCs), both embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) in vitro (Irie et al. Cell 160: 253-268, 2015). Similar attempts previously produced hPGCLCs from hPSCs at a very low efficiency, and the resulting cells were not fully characterized. A key step, which facilitated efficient hPGCLC specification from hPSCs, was the induction of a "competent" state for PGC fate via the medium containing a cocktail of four inhibitors. The competency of hPSCs can be maintained indefinitely and interchangeably with the conventional/low-competent hPSCs. Specification of hPGCLC occurs following sequential expression of key germ cell fate regulators, notably SOX17 and BLIMP1, as well as initiation of epigenetic resetting over 5 days. The hPGCLCs can be isolated using specific cell surface markers without the need for generating germ cell-specific reporter hPSC lines. This powerful method for the induction and isolation of hPGCLCs can be applied to both hESCs and iPSCs, which can be used for advances in human germ line biology.
Collapse
Affiliation(s)
- Naoko Irie
- Wellcome Trust Cancer Research UK Gurdon Institute, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QN, UK
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge, CB2 3EG, UK
| | - M Azim Surani
- Wellcome Trust Cancer Research UK Gurdon Institute, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QN, UK.
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge, CB2 3EG, UK.
| |
Collapse
|
58
|
Eslami-Arshaghi T, Vakilian S, Seyedjafari E, Ardeshirylajimi A, Soleimani M, Salehi M. Primordial germ cell differentiation of nuclear transfer embryonic stem cells using surface modified electroconductive scaffolds. In Vitro Cell Dev Biol Anim 2017; 53:371-380. [PMID: 28039620 DOI: 10.1007/s11626-016-0113-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2016] [Accepted: 11/01/2016] [Indexed: 01/26/2023]
Abstract
A combination of nanotopographical cues and surface modification of collagen and fibronectin is a potential platform in primordial germ cells (PGCs) differentiation. In the present study, the synergistic effect of nanotopography and surface modification on differentiation of nuclear transfer embryonic stem cells (nt-ESCs) toward PGC lineage was investigated. In order to achieve this goal, poly-anyline (PANi) was mix within poly-L-lactic acid (PLLA). Afterward, the random composite mats were fabricated using PLLA and PANi mix solution. The nanofiber topography notably upregulated the expressions of prdm14, mvh and c-kit compared with tissue culture polystyrene (TCP). Moreover, the combination of nanofiber topography and surface modification resulted in more enhancement of PGCs differentiation compared with non-modified nanofibrous scaffold. Additionally, gene expression results showed that mvh and c-kit were expressed at higher intensity in cells exposed to collagen and fibronectin rather than collagen or fibronectin solitary. These results demonstrated the importance of combined effect of collagen and fibronectin in order to develop a functional extracellular matrix (ECM) mimic in directing stem cell fate and the potential of such biofunctional scaffolds for treatment of infertility.
Collapse
Affiliation(s)
| | | | - Ehsan Seyedjafari
- Department of Biotechnology, College of Science, University of Tehran, Tehran, Iran
| | - Abdolreza Ardeshirylajimi
- Stem Cells Technology Research Center, Tehran, Iran.,Department of Tissue Engeneering and Regenerative Medicine, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Masoud Soleimani
- Department of Hematology and Blood Banking, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mohammad Salehi
- Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran. .,Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
59
|
Sun R, Sun YC, Ge W, Tan H, Cheng SF, Yin S, Sun XF, Li L, Dyce P, Li J, Yang X, Shi QH, Shen W. The crucial role of Activin A on the formation of primordial germ cell-like cells from skin-derived stem cells in vitro. Cell Cycle 2016; 14:3016-29. [PMID: 26406115 DOI: 10.1080/15384101.2015.1078031] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Primordial germ cells (PGCs) are founder cells of the germ cell lineage, and can be differentiated from stem cells in an induced system in vitro. However, the induction conditions need to be optimized in order to improve the differentiation efficiency. Activin A (ActA) is a member of the TGF-β super family and plays an important role in oogenesis and folliculogenesis. In the present study, we found that ActA promoted PGC-like cells (PGCLCs) formation from mouse skin-derived stem cells (SDSCs) in both embryoid body-like structure (EBLS) differentiation and the co-culture stage in a dose dependent manner. ActA treatment (100 ng/ml) during EBLS differentiation stage and further co-cultured for 6 days without ActA significantly increased PGCLCs from 53.2% to 82.8%, and as well as EBLS differentiation without ActA followed by co-cultured with 100 ng/ml ActA for 4 to 12 days with the percentage of PGCLCs increasing markedly in vitro. Moreover, mice treated with ActA at 100 ng/kg body weight from embryonic day (E) 5.5-12.5 led to more PGCs formation. However, the stimulating effects of ActA were interrupted by Smad3 RNAi, and in an in vitro cultured Smad3(-/-) mouse skin cells scenario. SMAD3 is thus likely a key effecter molecule in the ActA signaling pathway. In addition, we found that the expression of some epiblast cell markers, Fgf5, Dnmt3a, Dnmt3b and Wnt3, was increased in EBLSs cultured for 4 days or PGCLCs co-cultured for 12 days with ActA treatment. Interestingly, at 16 days of differentiation, the percentage of PGCLCs was decreased in the presence of ActA, but the expression of meiosis-relative genes, such as Stra8, Dmc1, Sycp3 and Sycp1, was increased. In conclusion, our data here demonstrated that ActA can promote PGCLC formation from SDSCs in vitro, at early stages of differentiation, and affect meiotic initiation of PGCLCs in later stages.
Collapse
Affiliation(s)
- Rui Sun
- a Molecular and Cell Genetics Laboratory; The CAS Key Laboratory of Innate Immunity and Chronic Disease; Hefei National Laboratory for Physical Sciences at Microscale; School of Life Sciences; University of Science and Technology of China ; Hefei , Anhui , China
| | - Yuan-Chao Sun
- b Institute of Reproductive Sciences; Qingdao Agricultural University , Qingdao ; Shandong , China.,c Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong; College of Animal Science and Technology; Qingdao Agricultural University , Qingdao ; Shandong , China
| | - Wei Ge
- b Institute of Reproductive Sciences; Qingdao Agricultural University , Qingdao ; Shandong , China.,c Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong; College of Animal Science and Technology; Qingdao Agricultural University , Qingdao ; Shandong , China
| | - Hui Tan
- b Institute of Reproductive Sciences; Qingdao Agricultural University , Qingdao ; Shandong , China.,c Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong; College of Animal Science and Technology; Qingdao Agricultural University , Qingdao ; Shandong , China
| | - Shun-Feng Cheng
- b Institute of Reproductive Sciences; Qingdao Agricultural University , Qingdao ; Shandong , China.,c Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong; College of Animal Science and Technology; Qingdao Agricultural University , Qingdao ; Shandong , China
| | - Shen Yin
- b Institute of Reproductive Sciences; Qingdao Agricultural University , Qingdao ; Shandong , China.,c Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong; College of Animal Science and Technology; Qingdao Agricultural University , Qingdao ; Shandong , China
| | - Xiao-Feng Sun
- b Institute of Reproductive Sciences; Qingdao Agricultural University , Qingdao ; Shandong , China.,c Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong; College of Animal Science and Technology; Qingdao Agricultural University , Qingdao ; Shandong , China
| | - Lan Li
- b Institute of Reproductive Sciences; Qingdao Agricultural University , Qingdao ; Shandong , China.,c Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong; College of Animal Science and Technology; Qingdao Agricultural University , Qingdao ; Shandong , China
| | - Paul Dyce
- d Department of Animal and Poultry Science ; University of Guelph ; Guelph ; Ontario , Canada
| | - Julang Li
- d Department of Animal and Poultry Science ; University of Guelph ; Guelph ; Ontario , Canada
| | - Xiao Yang
- e Genetic Laboratory of Development and Diseases; Beijing Institute of Biotechnology ; Beijing , China
| | - Qing-Hua Shi
- a Molecular and Cell Genetics Laboratory; The CAS Key Laboratory of Innate Immunity and Chronic Disease; Hefei National Laboratory for Physical Sciences at Microscale; School of Life Sciences; University of Science and Technology of China ; Hefei , Anhui , China.,f Collaborative Innovation Center of Genetics and Development; Fudan University ; Shanghai , China
| | - Wei Shen
- b Institute of Reproductive Sciences; Qingdao Agricultural University , Qingdao ; Shandong , China.,c Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong; College of Animal Science and Technology; Qingdao Agricultural University , Qingdao ; Shandong , China
| |
Collapse
|
60
|
Afsartala Z, Rezvanfar MA, Hodjat M, Tanha S, Assadollahi V, Bijangi K, Abdollahi M, Ghasemzadeh-Hasankolaei M. Amniotic membrane mesenchymal stem cells can differentiate into germ cells in vitro. In Vitro Cell Dev Biol Anim 2016; 52:1060-1071. [PMID: 27503516 DOI: 10.1007/s11626-016-0073-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 07/14/2016] [Indexed: 11/28/2022]
Abstract
This is the first report on differentiation of mouse amniotic membrane mesenchymal stem cells (AM-MSCs) into male germ cells (GCs). AM-MSCs have the multipotent differentiation capacity and can be differentiated into various cell types. In the present study, AM-MSCs were induced for differentiation into GCs. AM-MSCs were isolated from mouse embryonic membrane by enzymatic digestion. AM-MSCs were characterized with osteogenic and adipogenic differentiation test and flow cytometric analysis of some CD-markers. AM-MSCs were induced to differentiate into GCs using a creative two-step method. Passage-3 AM-MSCs were firstly treated with 25 ng/ml bone morphogenetic protein 4 (BMP4) for 5 d and in continuing with 1 μM retinoic acid (RA) for 12 d (total treatment time was 17 d). At the end of the treatment period, real-time reverse transcription (RT)-PCR was performed to evaluate the expression of GC-specific markers-Itgb1, Dazl, Stra8, Piwil2, Mvh, Oct4, and c-Kit- in the cells. Moreover, flow cytometry and immunofluorescence staining were performed to evaluate the expression of Mvh and Dazl at protein level. Real-time RT-PCR showed that most of the tested markers were upregulated in the treated AM-MSCs. Furthermore, flow cytometric and immunofluorescence analyses both revealed that a considerable part of the treated cells expressed GC-specific markers. The percentage of positive cells for Mvh and Dazl was about 23 and 46%, respectively. Our results indicated that a number of AM-MSCs successfully differentiated into the GCs. Finally, it seems that AM-MSCs would be a potential source of adult pluripotent stem cells for in vitro generation of GCs and cell-based therapies for treatment of infertility.
Collapse
Affiliation(s)
- Zohreh Afsartala
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Mohammad Amin Rezvanfar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy and Pharmaceutical Sciences Research Center (PSRC), Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Mahshid Hodjat
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Shima Tanha
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Vahideh Assadollahi
- Cellular and Molecular Research Center, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | | | - Mohammad Abdollahi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy and Pharmaceutical Sciences Research Center (PSRC), Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Mohammad Ghasemzadeh-Hasankolaei
- Infertility and Reproductive Health Research Center, Health Research Institute, Babol University of Medical Sciences, P.O. Box: 47318-38711, Amirkola, Babol, Iran.
| |
Collapse
|
61
|
FACS-sorted putative oogonial stem cells from the ovary are neither DDX4-positive nor germ cells. Sci Rep 2016; 6:27991. [PMID: 27301892 PMCID: PMC4908409 DOI: 10.1038/srep27991] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 05/26/2016] [Indexed: 12/11/2022] Open
Abstract
Whether the adult mammalian ovary contains oogonial stem cells (OSCs) is controversial. They have been isolated by a live-cell sorting method using the germ cell marker DDX4, which has previously been assumed to be cytoplasmic, not surface-bound. Furthermore their stem cell and germ cell characteristics remain disputed. Here we show that although OSC-like cells can be isolated from the ovary using an antibody to DDX4, there is no good in silico modelling to support the existence of a surface-bound DDX4. Furthermore these cells when isolated were not expressing DDX4, and did not initially possess germline identity. Despite these unremarkable beginnings, they acquired some pre-meiotic markers in culture, including DDX4, but critically never expressed oocyte-specific markers, and furthermore were not immortal but died after a few months. Our results suggest that freshly isolated OSCs are not germ stem cells, and are not being isolated by their DDX4 expression. However it may be that culture induces some pre-meiotic markers. In summary the present study offers weight to the dogma that the adult ovary is populated by a fixed number of oocytes and that adult de novo production is a rare or insignificant event.
Collapse
|
62
|
Tran ND, Kissner M, Subramanyam D, Parchem RJ, Laird DJ, Blelloch RH. A miR-372/let-7 Axis Regulates Human Germ Versus Somatic Cell Fates. Stem Cells 2016; 34:1985-91. [PMID: 27066911 DOI: 10.1002/stem.2378] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Revised: 02/23/2016] [Accepted: 03/13/2016] [Indexed: 11/06/2022]
Abstract
The embryonic stem cell cycle (ESCC) and let-7 families of miRNAs function antagonistically in the switch between mouse embryonic stem cell self-renewal and somatic differentiation. Here, we report that the human ESCC miRNA miR-372 and let-7 act antagonistically in germline differentiation from human embryonic stem cells (hESCs) and human induced pluripotent stem cells (iPSCs). hESC and iPSC-derived primordial germ cell-like cells (PGCLCs) expressed high levels of miR-372 and conversely, somatic cells expressed high levels of let-7. Manipulation of miRNA levels by introduction of miRNA mimics or knockdown with miRNA sponges demonstrated that miR-372 promotes whereas let-7 antagonizes PGCLC differentiation. Knockdown of the individual miR-372 targets SMARCC1, MECP2, CDKN1, RBL2, RHOC, and TGFBR2 increased PGCLC production, whereas knockdown of the let-7 targets CMYC and NMYC suppressed PGCLC differentiation. These findings uncover a miR-372/let-7 axis regulating human primordial germ cell (PGC) specification. Stem Cells 2016;34:1985-1991.
Collapse
Affiliation(s)
- Nam D Tran
- Departments of OB/Gyn and Urology, Center for Reproductive Sciences, and the Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, California, USA
| | - Michael Kissner
- Departments of OB/Gyn and Urology, Center for Reproductive Sciences, and the Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, California, USA
| | - Deepa Subramanyam
- Departments of OB/Gyn and Urology, Center for Reproductive Sciences, and the Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, California, USA
| | - Ronald J Parchem
- Departments of OB/Gyn and Urology, Center for Reproductive Sciences, and the Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, California, USA
| | - Diana J Laird
- Departments of OB/Gyn and Urology, Center for Reproductive Sciences, and the Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, California, USA
| | - Robert H Blelloch
- Departments of OB/Gyn and Urology, Center for Reproductive Sciences, and the Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, California, USA
| |
Collapse
|
63
|
Vaithiyanathan K, Liew SH, Zerafa N, Gamage T, Cook M, O’Reilly LA, Bouillet P, Scott CL, Strasser A, Findlay JK, Hutt KJ. BCL2-modifying factor promotes germ cell loss during murine oogenesis. Reproduction 2016; 151:553-62. [DOI: 10.1530/rep-15-0561] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 02/25/2016] [Indexed: 11/08/2022]
Abstract
Abstract
Apoptosis plays a prominent role during ovarian development by eliminating large numbers of germ cells from the female germ line. However, the precise mechanisms and regulatory proteins involved in germ cell death are yet to be determined. In this study, we characterised the role of the pro-apoptotic BH3-only protein, BCL2-modifying factor (BMF), in germ cell apoptosis in embryonic and neonatal mouse ovaries. BMF protein was immunohistochemically localised to germ cells at embryonic days 15.5 (E15.5) and E17.5 and postnatal day 1 (PN1), coincident with entry into the meiotic prophase, but was undetectable at E13.5, and only present at low levels at PN3 and PN5. Consistent with this expression pattern, loss of BMF in female mice was associated with a decrease in apoptosis at E15.5 and E17.5. Furthermore, increased numbers of germ cells were found in ovaries from Bmf−/− mice compared with WT animals at E15.5 and PN1. However, germ cell numbers were comparable between Bmf−/− and WT ovaries at PN3, PN5 and PN10. Collectively, these data indicate that BMF mediates foetal oocyte loss and its action limits the maximal number of germ cells attained in the developing ovary, but does not influence the number of primordial follicles initially established in ovarian reserve.
Collapse
|
64
|
Liu C, Liu W, Fan L, Liu J, Li P, Zhang W, Gao J, Li Z, Zhang Q, Wang X. Sequences analyses and expression profiles in tissues and embryos of Japanese flounder (Paralichthys olivaceus) PRDM1. FISH PHYSIOLOGY AND BIOCHEMISTRY 2016; 42:467-482. [PMID: 26508172 DOI: 10.1007/s10695-015-0152-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 10/20/2015] [Indexed: 06/05/2023]
Abstract
PRDM1 (PRDI-BF1-RIZ1 homologous domain containing 1) appears to be a pleiotropic regulatory factor in various processes. It contains a PR (PRDI-BF1-RIZ1 homologous) domain protein and five zinc fingers. In the present study, a gene coding the homolog of prdm1 and the 5' regulatory region of prdm1 was identified from the Paralichthys olivaceus (denoted Po-prdm1). Results of real-time quantitative polymerase chain reaction amplification (RT-qPCR) and in situ hybridization (ISH) in embryos revealed that Po-prdm1 was highly expressed between the early gastrula and tail bud stages, with its expression peaking in the mid-gastrula stage, whereas the results of RT-qPCR and ISH in tissues demonstrated that Po-prdm1 transcripts were ubiquitously detected in all tissues, which indicates its pleiotropic function in multiple processes. ISH of gonadal tissues revealed that the transcripts were located in the nucleus and cytoplasm of the oocytes in the ovaries but only in the spermatogonia and not in the spermatocytes in the testes. The Po-prdm1 transcription factor binding sites and their conserved binding region among vertebrates were analyzed in this study. The combined results suggest that Po-PRDM1 has a conserved function in teleosts and mammals.
Collapse
Affiliation(s)
- Conghui Liu
- Key Laboratory of Marine Genetics and Breeding (Ocean University of China), Ministry of Education, Qingdao, 266003, China
| | - Wei Liu
- Key Laboratory of Marine Genetics and Breeding (Ocean University of China), Ministry of Education, Qingdao, 266003, China
| | - Lin Fan
- Key Laboratory of Marine Genetics and Breeding (Ocean University of China), Ministry of Education, Qingdao, 266003, China
| | - Jinxiang Liu
- Key Laboratory of Marine Genetics and Breeding (Ocean University of China), Ministry of Education, Qingdao, 266003, China
| | - Peizhen Li
- Key Laboratory of Marine Genetics and Breeding (Ocean University of China), Ministry of Education, Qingdao, 266003, China
| | - Wei Zhang
- Key Laboratory of Marine Genetics and Breeding (Ocean University of China), Ministry of Education, Qingdao, 266003, China
| | - Jinning Gao
- Key Laboratory of Marine Genetics and Breeding (Ocean University of China), Ministry of Education, Qingdao, 266003, China
| | - Zan Li
- Key Laboratory of Marine Genetics and Breeding (Ocean University of China), Ministry of Education, Qingdao, 266003, China
| | - Quanqi Zhang
- Key Laboratory of Marine Genetics and Breeding (Ocean University of China), Ministry of Education, Qingdao, 266003, China
| | - Xubo Wang
- Key Laboratory of Marine Genetics and Breeding (Ocean University of China), Ministry of Education, Qingdao, 266003, China.
- College of Marine Life Science, Ocean University of China, No. 5 Yushan Road, Qingdao, 266003, China.
| |
Collapse
|
65
|
Pluripotent stem cells and livestock genetic engineering. Transgenic Res 2016; 25:289-306. [PMID: 26894405 DOI: 10.1007/s11248-016-9929-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Accepted: 01/06/2016] [Indexed: 01/12/2023]
Abstract
The unlimited proliferative ability and capacity to contribute to germline chimeras make pluripotent embryonic stem cells (ESCs) perfect candidates for complex genetic engineering. The utility of ESCs is best exemplified by the numerous genetic models that have been developed in mice, for which such cells are readily available. However, the traditional systems for mouse genetic engineering may not be practical for livestock species, as it requires several generations of mating and selection in order to establish homozygous founders. Nevertheless, the self-renewal and pluripotent characteristics of ESCs could provide advantages for livestock genetic engineering such as ease of genetic manipulation and improved efficiency of cloning by nuclear transplantation. These advantages have resulted in many attempts to isolate livestock ESCs, yet it has been generally concluded that the culture conditions tested so far are not supportive of livestock ESCs self-renewal and proliferation. In contrast, there are numerous reports of derivation of livestock induced pluripotent stem cells (iPSCs), with demonstrated capacity for long term proliferation and in vivo pluripotency, as indicated by teratoma formation assay. However, to what extent these iPSCs represent fully reprogrammed PSCs remains controversial, as most livestock iPSCs depend on continuous expression of reprogramming factors. Moreover, germline chimerism has not been robustly demonstrated, with only one successful report with very low efficiency. Therefore, even 34 years after derivation of mouse ESCs and their extensive use in the generation of genetic models, the livestock genetic engineering field can stand to gain enormously from continued investigations into the derivation and application of ESCs and iPSCs.
Collapse
|
66
|
Abstract
Pluripotency is a unique developmental state that lays the foundation upon which the entire embryo is built. Pluripotent cells possess the unique capacity to generate, in an exquisitely defined sequence, all the distinct cell types comprising the fetal and adult organism. The discovery of pluripotent stem cells and now the ability to generate them from differentiated cells has had a profound impact on a vast array of scientific disciplines. In addition to their clinical potential as a source of therapeutic cell types, pluripotent stem cells provide scalable access to otherwise experimentally inaccessible development- and disease-associated biology. Here I provide my perspective on the continuum of pluripotency in the early mammalian embryo. I also discuss how novel genomic technologies are now enabling the capture of molecular “snapshots” of the several distinct pluripotent states that stem cells undergo during this pivotal developmental period. Pluripotency is a developmental continuum Snapshots of pluripotency can be captured in derivative stem cell lines Two dominant attractor states of pluripotency can be maintained in vitro Synergistic studies of both states have provided a new understanding of development
Collapse
Affiliation(s)
- Paul J Tesar
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, 10900 Euclid Avenue, Cleveland, OH 44106, USA.
| |
Collapse
|
67
|
Lovasco LA, Gustafson EA, Seymour KA, de Rooij DG, Freiman RN. TAF4b is required for mouse spermatogonial stem cell development. Stem Cells 2016; 33:1267-76. [PMID: 25727968 DOI: 10.1002/stem.1914] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Revised: 10/23/2014] [Accepted: 11/28/2014] [Indexed: 12/22/2022]
Abstract
Long-term mammalian spermatogenesis requires proper development of spermatogonial stem cells (SSCs) that replenish the testis with germ cell progenitors during adult life. TAF4b is a gonadal-enriched component of the general transcription factor complex, TFIID, which is required for the maintenance of spermatogenesis in the mouse. Successful germ cell transplantation assays into adult TAF4b-deficient host testes suggested that TAF4b performs an essential germ cell autonomous function in SSC establishment and/or maintenance. To elucidate the SSC function of TAF4b, we characterized the initial gonocyte pool and rounds of spermatogenic differentiation in the context of the Taf4b-deficient mouse testis. Here, we demonstrate a significant reduction in the late embryonic gonocyte pool and a deficient expansion of this pool soon after birth. Resulting from this reduction of germ cell progenitors is a developmental delay in meiosis initiation, as compared to age-matched controls. While GFRα1+ spermatogonia are appropriately present as Asingle and Apaired in wild-type testes, TAF4b-deficient testes display an increased proportion of long and clustered chains of GFRα1+ cells. In the absence of TAF4b, seminiferous tubules in the adult testis either lack germ cells altogether or are found to have missing generations of spermatogenic progenitor cells. Together these data indicate that TAF4b-deficient spermatogenic progenitor cells display a tendency for differentiation at the expense of self-renewal and a renewing pool of SSCs fail to establish during the critical window of SSC development.
Collapse
Affiliation(s)
- Lindsay A Lovasco
- Department of Molecular and Cellular Biology and Biochemistry, Brown University, Providence, Rhode Island, USA
| | | | | | | | | |
Collapse
|
68
|
Abstract
Primordial germ cells are usually made early in the development of an organism. These are the mother of all stem cells that are necessary for propagation of the species, yet use highly diverse mechanisms between organisms. How they are specified, and when and where they form, are central to developmental biology. Using diverse organisms to study this development is illuminating for understanding the mechanics these cells use in this essential function and for identifying the breadth of evolutionary changes that have occurred between species. This essay emphasizes how echinoderms may contribute to the patchwork quilt of our understanding of germ line formation during embryogenesis.
Collapse
Affiliation(s)
- Gary M Wessel
- Department of Molecular and Cellular Biology, Brown University, Providence, Rhode Island, USA.
| |
Collapse
|
69
|
De Felici M. The Formation and Migration of Primordial Germ Cells in Mouse and Man. Results Probl Cell Differ 2016; 58:23-46. [DOI: 10.1007/978-3-319-31973-5_2] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/09/2023]
|
70
|
Esfandiari F, Mashinchian O, Ashtiani MK, Ghanian MH, Hayashi K, Saei AA, Mahmoudi M, Baharvand H. Possibilities in Germ Cell Research: An Engineering Insight. Trends Biotechnol 2015; 33:735-746. [DOI: 10.1016/j.tibtech.2015.09.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 09/06/2015] [Accepted: 09/08/2015] [Indexed: 01/05/2023]
|
71
|
Nikolic A, Volarevic V, Armstrong L, Lako M, Stojkovic M. Primordial Germ Cells: Current Knowledge and Perspectives. Stem Cells Int 2015; 2016:1741072. [PMID: 26635880 PMCID: PMC4655300 DOI: 10.1155/2016/1741072] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 05/17/2015] [Indexed: 01/11/2023] Open
Abstract
Infertility is a condition that occurs very frequently and understanding what defines normal fertility is crucial to helping patients. Causes of infertility are numerous and the treatment often does not lead to desired pregnancy especially when there is a lack of functional gametes. In humans, the primordial germ cell (PGC) is the primary undifferentiated stem cell type that will differentiate towards gametes: spermatozoa or oocytes. With the development of stem cell biology and differentiation protocols, PGC can be obtained from pluripotent stem cells providing a new therapeutic possibility to treat infertile couples. Recent studies demonstrated that viable mouse pups could be obtained from in vitro differentiated stem cells suggesting that translation of these results to human is closer. Therefore, the aim of this review is to summarize current knowledge about PGC indicating the perspective of their use in both research and medical application for the treatment of infertility.
Collapse
Affiliation(s)
- Aleksandar Nikolic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, 69 Svetozara Markovica Street, 34000 Kragujevac, Serbia
| | - Vladislav Volarevic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, 69 Svetozara Markovica Street, 34000 Kragujevac, Serbia
| | - Lyle Armstrong
- Institute of Genetic Medicine, Newcastle University, The International Centre for Life, Central Parkway, Newcastle upon Tyne NE1 3BZ, UK
| | - Majlinda Lako
- Institute of Genetic Medicine, Newcastle University, The International Centre for Life, Central Parkway, Newcastle upon Tyne NE1 3BZ, UK
| | - Miodrag Stojkovic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, 69 Svetozara Markovica Street, 34000 Kragujevac, Serbia
- Spebo Medical, Norvezanska 16, 16 000 Leskovac, Serbia
| |
Collapse
|
72
|
Yakovlev KV. Localization of germ plasm-related structures during sea urchin oogenesis. Dev Dyn 2015; 245:56-66. [PMID: 26385846 DOI: 10.1002/dvdy.24348] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 09/12/2015] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Animal germ cells have specific organelles that are similar to ribonucleoprotein complex, called germ plasm, which is accumulated in eggs. Germ plasm is essential for inherited mechanism of germ line segregation in early embryogenesis. Sea urchins have early germ line segregation in early embryogenesis. Nevertheless, organization of germ plasm-related organelles and their molecular composition are still unclear. Another issue is whether maternally accumulated germ plasm exists in the sea urchin eggs. RESULTS I analyzed intracellular localization of germ plasm during oogenesis in sea urchin Strongylocentrotus intermedius by using morphological approach and immunocytochemical detection of Vasa, a germ plasm marker. All ovarian germ cells have germ plasm-related organelles in the form of germ granules, Balbiani bodies, and perinuclear nuage found previously in germ cells in other animals. Maternal germ plasm is accumulated in late oogenesis at the cell periphery. Cytoskeletal drug treatment showed an association of Vasa-positive granules with actin filaments in the egg cortex. CONCLUSIONS All female germ cells of sea urchins have germ plasm-related organelles. Eggs have a maternally accumulated germ plasm associated with cortical cytoskeleton. These findings correlate with early segregation of germ line in sea urchins.
Collapse
Affiliation(s)
- Konstantin V Yakovlev
- Laboratory of Cytotechnology, A.V. Zhirmunsky Institute of Marine Biology of the Far Eastern Branch of the Russian Academy of Sciences, Vladivostok, Russia
| |
Collapse
|
73
|
Jedrusik A. Making the first decision: lessons from the mouse. Reprod Med Biol 2015; 14:135-150. [PMID: 29259411 PMCID: PMC5715835 DOI: 10.1007/s12522-015-0206-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2014] [Accepted: 03/31/2015] [Indexed: 01/06/2023] Open
Abstract
Pre-implantation development encompasses a period of 3-4 days over which the mammalian embryo has to make its first decision: to separate the pluripotent inner cell mass (ICM) from the extra-embryonic epithelial tissue, the trophectoderm (TE). The ICM gives rise to tissues mainly building the body of the future organism, while the TE contributes to the extra-embryonic tissues that support embryo development after implantation. This review provides an overview of the cellular and molecular mechanisms that control the critical aspects of this first decision, and highlights the role of critical events, namely zytotic genome activation, compaction, polarization, asymmetric cell divisions, formation of the blastocyst cavity and expression of key transcription factors.
Collapse
Affiliation(s)
- Agnieszka Jedrusik
- Wellcome Trust/CR UK Gurdon InstituteTennis Court RoadCB2 1QNCambridgeUK
- Department of Physiology, Development and NeuroscienceUniversity of CambridgeDowning StreetCB2 3DYCambridgeUK
| |
Collapse
|
74
|
Martínez-Arroyo AM, Míguez-Forján JM, Remohí J, Pellicer A, Medrano JV. Understanding Mammalian Germ Line Development with In Vitro Models. Stem Cells Dev 2015; 24:2101-13. [DOI: 10.1089/scd.2015.0060] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Affiliation(s)
- Ana M. Martínez-Arroyo
- Fundación Instituto Valenciano de Infertilidad (FIVI), Valencia University, INCLIVA, Valencia, Spain
| | - Jose M. Míguez-Forján
- Fundación Instituto Valenciano de Infertilidad (FIVI), Valencia University, INCLIVA, Valencia, Spain
| | - Jose Remohí
- Fundación Instituto Valenciano de Infertilidad (FIVI), Valencia University, INCLIVA, Valencia, Spain
| | - Antonio Pellicer
- Fundación Instituto de Investigación Sanitaria La Fe, Valencia, Spain
| | - Jose V. Medrano
- Fundación Instituto Valenciano de Infertilidad (FIVI), Valencia University, INCLIVA, Valencia, Spain
- Fundación Instituto de Investigación Sanitaria La Fe, Valencia, Spain
| |
Collapse
|
75
|
Guillot PV. Induced pluripotent stem (iPS) cells from human fetal stem cells. Best Pract Res Clin Obstet Gynaecol 2015; 31:112-20. [PMID: 26427551 DOI: 10.1016/j.bpobgyn.2015.08.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Accepted: 08/31/2015] [Indexed: 12/14/2022]
Abstract
Pluripotency defines the ability of stem cells to differentiate into all the lineages of the three germ layers and self-renew indefinitely. Somatic cells can regain the developmental potential of embryonic stem cells following ectopic expression of a set of transcription factors or, in certain circumstances, via modulation of culture conditions and supplementation with small molecule, that is, induced pluripotent stem (iPS) cells. Here, we discuss the use of fetal tissues for reprogramming, focusing in particular on stem cells derived from human amniotic fluid, and the development of chemical reprogramming. We next address the advantages and disadvantages of deriving pluripotent cells from fetal tissues and the potential clinical applications.
Collapse
Affiliation(s)
- Pascale V Guillot
- UCL Institute for Women's Health, University College London, London, UK.
| |
Collapse
|
76
|
Easley CA, Simerly CR, Schatten G. Gamete derivation from embryonic stem cells, induced pluripotent stem cells or somatic cell nuclear transfer-derived embryonic stem cells: state of the art. Reprod Fertil Dev 2015; 27:89-92. [PMID: 25472048 DOI: 10.1071/rd14317] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Generating gametes from pluripotent stem cells (PSCs) has many scientific justifications and several biomedical rationales. Here, we consider several strategies for deriving gametes from PSCs from mice and primates (human and non-human) and their anticipated strengths, challenges and limitations. Although the 'Weismann barrier', which separates the mortal somatic cell lineages from the potentially immortal germline, has long existed, breakthroughs first in mice and now in humans are artificially creating germ cells from somatic cells. Spermatozoa with full reproductive viability establishing multiple generations of seemingly normal offspring have been reported in mice and, in humans, haploid spermatids with correct parent-of-origin imprints have been obtained. Similar progress with making oocytes has been published using mouse PSCs differentiated in vitro into primordial germ cells, which are then cultured after xenografting reconstructed artificial ovaries. Progress in making human oocytes artificially is proving challenging. The usefulness of these artificial gametes, from assessing environmental exposure toxicity to optimising medical treatments to prevent negative off-target effects on fertility, may prove invaluable, as may basic discoveries on the fundamental mechanisms of gametogenesis.
Collapse
Affiliation(s)
- Charles A Easley
- Laboratory of Translational Cell Biology, Department of Cell Biology, Emory University School of Medicine, 615 Michael Street, Atlanta, GA 30322, USA
| | - Calvin R Simerly
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, PA 15108, USA
| | - Gerald Schatten
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, PA 15108, USA
| |
Collapse
|
77
|
Lee DM, Wilk R, Hu J, Krause HM, Harris TJC. Germ Cell Segregation from the Drosophila Soma Is Controlled by an Inhibitory Threshold Set by the Arf-GEF Steppke. Genetics 2015; 200:863-72. [PMID: 25971667 PMCID: PMC4512548 DOI: 10.1534/genetics.115.176867] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 05/10/2015] [Indexed: 12/11/2022] Open
Abstract
Germline cells segregate from the soma to maintain their totipotency, but the cellular mechanisms of this segregation are unclear. The Drosophila melanogaster embryo forms a posterior group of primordial germline cells (PGCs) by their division from the syncytial soma. Extended plasma membrane furrows enclose the PGCs in response to the germ plasm protein Germ cell-less (Gcl) and Rho1-actomyosin activity. Recently, we found that loss of the Arf-GEF Steppke (Step) leads to similar Rho1-dependent plasma membrane extensions but from pseudocleavage furrows of the soma. Here, we report that the loss of step also leads to premature formation of a large cell group at the anterior pole of the embryo . These anterior cells lacked germ plasm, but budded and formed at the same time as posterior PGCs, and then divided asynchronously as PGCs also do. With genetic analyses we found that Step normally activates Arf small G proteins and antagonizes Rho1-actomyosin pathways to inhibit anterior cell formation. A uniform distribution of step mRNA around the one-cell embryo cortex suggested that Step restricts cell formation through a global control mechanism. Thus, we examined the effect of Step on PGC formation at the posterior pole. Reducing Gcl or Rho1 levels decreased PGC numbers, but additional step RNAi restored their numbers. Reciprocally, GFP-Step overexpression induced dosage- and Arf-GEF-dependent loss of PGCs, an effect worsened by reducing Gcl or actomyosin pathway activity. We propose that a global distribution of Step normally sets an inhibitory threshold for Rho1 activity to restrict early cell formation to the posterior.
Collapse
Affiliation(s)
- Donghoon M Lee
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario M5S 3G5, Canada
| | - Ronit Wilk
- Department of Molecular Genetics and The Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario M5S 3E1, Canada
| | - Jack Hu
- Department of Molecular Genetics and The Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario M5S 3E1, Canada
| | - Henry M Krause
- Department of Molecular Genetics and The Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario M5S 3E1, Canada
| | - Tony J C Harris
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario M5S 3G5, Canada
| |
Collapse
|
78
|
Borg M, Berger F. Chromatin remodelling during male gametophyte development. THE PLANT JOURNAL : FOR CELL AND MOLECULAR BIOLOGY 2015; 83:177-188. [PMID: 25892182 DOI: 10.1111/tpj.12856] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Revised: 04/10/2015] [Accepted: 04/14/2015] [Indexed: 05/28/2023]
Abstract
The plant life cycle alternates between a diploid sporophytic phase and haploid gametophytic phase, with the latter giving rise to the gametes. Male gametophyte development encompasses two mitotic divisions that results in a simple three-celled structure knows as the pollen grain, in which two sperm cells are encased within a larger vegetative cell. Both cell types exhibit a very different type of chromatin organization - highly condensed in sperm cell nuclei and highly diffuse in the vegetative cell. Distinct classes of histone variants have dynamic and differential expression in the two cell lineages of the male gametophyte. Here we review how the dynamics of histone variants are linked to reprogramming of chromatin activities in the male gametophyte, compaction of the sperm cell genome and zygotic transitions post-fertilization.
Collapse
Affiliation(s)
- Michael Borg
- Gregor Mendel Institute, Vienna Biocenter, Dr. Bohr-Gasse 3, 1030, Vienna, Austria
| | - Frédéric Berger
- Gregor Mendel Institute, Vienna Biocenter, Dr. Bohr-Gasse 3, 1030, Vienna, Austria
| |
Collapse
|
79
|
A perspective on the evolution of germ-cell development and germinal mosaics of deleterious mutations. Genetica 2015; 143:563-9. [PMID: 26113303 DOI: 10.1007/s10709-015-9854-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Accepted: 06/22/2015] [Indexed: 10/23/2022]
Abstract
In many animals a small number of primordial germ cells (PGCs) are set aside early in development, mitosis and mitochondrial DNA syntheses are arrested, transcription is stopped or reduced, and the PGCs migrate later to the emerging gonads and become germ cells. What could be the evolutionary advantage of sequestering non-dividing PGCs early in development? A commonly cited advantage is a reduction in the number of new deleterious mutations that would occur if there were additional divisions in PGCs early in development. We would like to add to this advantage the fact that these additional mutations in PGCs give rise to germinal mosaics (i.e., premeiotic clusters of mutation) in multiple progeny of the same individual, thus having a larger detrimental effect on the evolutionary fitness of their carriers. Here, we reviewed published studies providing evidence that germinal mosaics of deleterious mutant alleles are not rare, occur for all types of genetic damage, and have been observed in all tested organisms and in nature. We propose the hypothesis that PGC sequestration during early animal development may have evolved in part in response to selection for preventing the occurrence of premeiotic clusters of deleterious mutant alleles, and describe a series of predictions that would allow the assessment of the potential role of germinal mosaics on the evolution of PGC sequestration.
Collapse
|
80
|
Stopa N, Krebs JE, Shechter D. The PRMT5 arginine methyltransferase: many roles in development, cancer and beyond. Cell Mol Life Sci 2015; 72:2041-59. [PMID: 25662273 PMCID: PMC4430368 DOI: 10.1007/s00018-015-1847-9] [Citation(s) in RCA: 370] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2014] [Revised: 01/10/2015] [Accepted: 01/29/2015] [Indexed: 10/24/2022]
Abstract
Post-translational arginine methylation is responsible for regulation of many biological processes. The protein arginine methyltransferase 5 (PRMT5, also known as Hsl7, Jbp1, Skb1, Capsuleen, or Dart5) is the major enzyme responsible for mono- and symmetric dimethylation of arginine. An expanding literature demonstrates its critical biological function in a wide range of cellular processes. Histone and other protein methylation by PRMT5 regulate genome organization, transcription, stem cells, primordial germ cells, differentiation, the cell cycle, and spliceosome assembly. Metazoan PRMT5 is found in complex with the WD-repeat protein MEP50 (also known as Wdr77, androgen receptor coactivator p44, or Valois). PRMT5 also directly associates with a range of other protein factors, including pICln, Menin, CoPR5 and RioK1 that may alter its subcellular localization and protein substrate selection. Protein substrate and PRMT5-MEP50 post-translation modifications induce crosstalk to regulate PRMT5 activity. Crystal structures of C. elegans PRMT5 and human and frog PRMT5-MEP50 complexes provide substantial insight into the mechanisms of substrate recognition and procession to dimethylation. Enzymological studies of PRMT5 have uncovered compelling insights essential for future development of specific PRMT5 inhibitors. In addition, newly accumulating evidence implicates PRMT5 and MEP50 expression levels and their methyltransferase activity in cancer tumorigenesis, and, significantly, as markers of poor clinical outcome, marking them as potential oncogenes. Here, we review the substantial new literature on PRMT5 and its partners to highlight the significance of understanding this essential enzyme in health and disease.
Collapse
Affiliation(s)
- Nicole Stopa
- Department of Biological Sciences, University of Alaska Anchorage, 3211 Providence Drive, Anchorage, AK 99508, USA
| | - Jocelyn E. Krebs
- Department of Biological Sciences, University of Alaska Anchorage, 3211 Providence Drive, Anchorage, AK 99508, USA
| | - David Shechter
- Department of Biochemistry, Albert Einstein College of Medicine of Yeshiva University, 1300 Morris Park Avenue, Bronx, NY 10461, USA
| |
Collapse
|
81
|
Surani MA. Human Germline: A New Research Frontier. Stem Cell Reports 2015; 4:955-60. [PMID: 26028529 PMCID: PMC4471826 DOI: 10.1016/j.stemcr.2015.04.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Revised: 04/28/2015] [Accepted: 04/29/2015] [Indexed: 11/16/2022] Open
Abstract
We recently elucidated the mechanism of human primordial germ cell (hPGC) specification and resetting of the epigenome for totipotency. The regulators of hPGC specification also initiate resetting of the epigenome, leading to a comprehensive erasure of DNA methylation, erasure of imprints and X reactivation in early hPGCs in vivo. These studies reveal differences with the mouse model, which are probably due to differences in the regulation of human pluripotency, and in postimplantation development at gastrulation, which indicates the importance of non-rodent models for investigations. Within the extreme hypomethylated environment of the early human germline are loci that are resistant to DNA demethylation, with subsequent predominant expression in neural cells. These loci provide a model for studies on the mechanism of transgenerational epigenetic inheritance, and their response to environmental factors. Such epigenetic mechanism of inheritance could potentially provide greater phenotypic plasticity, with significant consequences for human development and disease.
Collapse
Affiliation(s)
- M Azim Surani
- Wellcome Trust Cancer Research UK Gurdon Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, UK; Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3EG, UK; Wellcome Trust-Medical Research Council Stem Cell Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 3EG, UK.
| |
Collapse
|
82
|
Monk D. Germline-derived DNA methylation and early embryo epigenetic reprogramming: The selected survival of imprints. Int J Biochem Cell Biol 2015; 67:128-38. [PMID: 25966912 DOI: 10.1016/j.biocel.2015.04.014] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Accepted: 04/27/2015] [Indexed: 12/27/2022]
Abstract
DNA methylation is an essential epigenetic mechanism involved in many essential cellular processes. During development epigenetic reprograming takes place during gametogenesis and then again in the pre-implantation embryo. These two reprograming windows ensure genome-wide removal of methylation in the primordial germ cells so that sex-specific signatures can be acquired in the sperm and oocyte. Following fertilization the majority of this epigenetic information is erased to give the developing embryo an epigenetic profile coherent with pluripotency. It is estimated that ∼65% of the genome is differentially methylated between the gametes, however following embryonic reprogramming only parent-of-origin methylation at known imprinted loci remains. This suggests that trans-acting factors such as Zfp57 can discriminate imprinted differentially methylated regions (DMRs) from the thousands of CpG rich regions that are differentially marked in the gametes. Recently transient imprinted DMRs have been identified suggesting that these loci are also protected from pre-implantation reprograming but succumb to de novo remethylation at the implantation stage. This highlights that "ubiquitous" imprinted loci are also resilient to gaining methylation by protecting their unmethylated alleles. In this review I examine the processes involved in epigenetic reprograming and the mechanisms that ensure allelic methylation at imprinted loci is retained throughout the life of the organism, discussing the critical differences between mouse and humans. This article is part of a Directed Issue entitled: Epigenetics Dynamics in development and disease.
Collapse
Affiliation(s)
- David Monk
- Imprinting and Cancer group, Cancer Epigenetic and Biology Program, Institut d'Investigació Biomedica de Bellvitge, Hospital Duran i Reynals, Barcelona 08908, Spain.
| |
Collapse
|
83
|
Hu YC, Nicholls PK, Soh YQS, Daniele JR, Junker JP, van Oudenaarden A, Page DC. Licensing of primordial germ cells for gametogenesis depends on genital ridge signaling. PLoS Genet 2015; 11:e1005019. [PMID: 25739037 PMCID: PMC4349450 DOI: 10.1371/journal.pgen.1005019] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 01/22/2015] [Indexed: 01/07/2023] Open
Abstract
In mouse embryos at mid-gestation, primordial germ cells (PGCs) undergo licensing to become gametogenesis-competent cells (GCCs), gaining the capacity for meiotic initiation and sexual differentiation. GCCs then initiate either oogenesis or spermatogenesis in response to gonadal cues. Germ cell licensing has been considered to be a cell-autonomous and gonad-independent event, based on observations that some PGCs, having migrated not to the gonad but to the adrenal gland, nonetheless enter meiosis in a time frame parallel to ovarian germ cells -- and do so regardless of the sex of the embryo. Here we test the hypothesis that germ cell licensing is cell-autonomous by examining the fate of PGCs in Gata4 conditional mutant (Gata4 cKO) mouse embryos. Gata4, which is expressed only in somatic cells, is known to be required for genital ridge initiation. PGCs in Gata4 cKO mutants migrated to the area where the genital ridge, the precursor of the gonad, would ordinarily be formed. However, these germ cells did not undergo licensing and instead retained characteristics of PGCs. Our results indicate that licensing is not purely cell-autonomous but is induced by the somatic genital ridge. During embryonic development, stem cell-like primordial germ cells travel across the developing embryo to the genital ridge, which gives rise to the gonad. Around the time of their arrival, the primordial germ cells gain the capacity to undertake sexual specialization and meiosis—a process called germ cell licensing. Based on the observation that meiosis and sexual differentiation can occur when primordial germ cells stray into the area of the adrenal gland, the primordial germ cell has been thought to be responsible for its own licensing. We tested this notion by examining the licensing process in mutant mouse embryos that did not form a genital ridge. We discovered that in the absence of the genital ridge, primordial germ cells migrate across the developing embryo properly, but instead of undergoing licensing, these cells retain their primordial germ cell characteristics. We conclude that licensing of embryonic primordial germ cells for gametogenesis is dependent on signaling from the genital ridge.
Collapse
Affiliation(s)
- Yueh-Chiang Hu
- Whitehead Institute, Cambridge, Massachusetts, United States of America
| | - Peter K. Nicholls
- Whitehead Institute, Cambridge, Massachusetts, United States of America
| | - Y. Q. Shirleen Soh
- Whitehead Institute, Cambridge, Massachusetts, United States of America
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Joseph R. Daniele
- Whitehead Institute, Cambridge, Massachusetts, United States of America
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Jan Philipp Junker
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
- Department of Physics, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
- Hubrecht Institute—KNAW (Royal Netherlands Academy of Arts and Sciences) and University Medical Center Utrecht, Utrecht, Netherlands
| | - Alexander van Oudenaarden
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
- Department of Physics, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
- Hubrecht Institute—KNAW (Royal Netherlands Academy of Arts and Sciences) and University Medical Center Utrecht, Utrecht, Netherlands
| | - David C. Page
- Whitehead Institute, Cambridge, Massachusetts, United States of America
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
- Howard Hughes Medical Institute, Whitehead Institute, Cambridge, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
84
|
Scaldaferri ML, Klinger FG, Farini D, Di Carlo A, Carsetti R, Giorda E, De Felici M. Hematopoietic activity in putative mouse primordial germ cell populations. Mech Dev 2015; 136:53-63. [PMID: 25684074 DOI: 10.1016/j.mod.2015.02.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Revised: 01/19/2015] [Accepted: 02/10/2015] [Indexed: 01/07/2023]
Abstract
In the present paper, starting from the observation of heterogeneous expression of the GOF-18ΔPE-GFP Pou5f1 (Oct3/4) transgene in putative mouse PGC populations settled in the aorta-gonad-mesonephros (AGM) region, we identified various OCT3/4 positive populations showing distinct expression of PGC markers (BLIMP-1, AP, TG-1, STELLA) and co-expressing several proteins (CD-34, CD-41, FLK-1) and genes (Brachyury, Hox-B4, Scl/Tal-1 and Gata-2) of hematopoietic precursors. Moreover, we found that Oct3/4-GFP(weak) CD-34(weak/high) cells possess robust hematopoietic colony forming activity (CFU) in vitro. These data indicate that the cell population usually considered PGCs moving toward the gonadal ridges encompasses a subset of cells co-expressing several germ cell and hematopoietic markers and possessing hematopoietic activity. These results are discussed within of the current model of germline segregation.
Collapse
Affiliation(s)
- Maria Lucia Scaldaferri
- Department of Biomedicine and Prevention, Section of Histology and Embryology, University of Rome "Tor Vergata", Rome, Italy
| | - Francesca Gioia Klinger
- Department of Biomedicine and Prevention, Section of Histology and Embryology, University of Rome "Tor Vergata", Rome, Italy
| | - Donatella Farini
- Department of Biomedicine and Prevention, Section of Histology and Embryology, University of Rome "Tor Vergata", Rome, Italy
| | - Anna Di Carlo
- Department of Biomedicine and Prevention, Section of Histology and Embryology, University of Rome "Tor Vergata", Rome, Italy
| | - Rita Carsetti
- Research Center Ospedale Pediatrico Bambino Gesù, IRCSS, Laboratory of Flow-Cytometry and B Cell Development, Rome, Italy
| | - Ezio Giorda
- Research Center Ospedale Pediatrico Bambino Gesù, IRCSS, Laboratory of Flow-Cytometry and B Cell Development, Rome, Italy
| | - Massimo De Felici
- Department of Biomedicine and Prevention, Section of Histology and Embryology, University of Rome "Tor Vergata", Rome, Italy.
| |
Collapse
|
85
|
Abstract
Pluripotent stem cells have great potential for regenerative medicine; however, their clinical use is associated with a risk of tumor formation. We utilized pluripotent cells expressing green fluorescent protein and puromycin resistance under control of the Oct4 promoter to study the persistence of potential pluripotent cells under embryoid body (EB) culture conditions, which are commonly used to obtain organotypic cells. We found that i.) OCT4-expressing cells dramatically decrease during the first week of differentiation, ii.) the number of OCT4-expressing cells recovers from day 7 on, iii.) the OCT4-expressing cells are similar to embryonic stem cells grown in the presence of leukemia inhibitory factor LIF but express several markers associated with germ cell formation, such as DAZL and STRA-8 and iv.) the persistence of potentially pluripotent cells is independent of supportive cells in EBs. Finally, OCT4-expressing cells, isolated from EBs after 2-month of culture, were further maintained under feeder-free conditions in absence of LIF and continued to express OCT4 in 95 % of the population for at least 36 days. These findings point to an alternative state of stable OCT4 expression. In the frame of the landscape model of differentiation two attractors of pluripotency might be defined based on their different characteristics.
Collapse
|
86
|
Irie N, Weinberger L, Tang WWC, Kobayashi T, Viukov S, Manor YS, Dietmann S, Hanna JH, Surani MA. SOX17 is a critical specifier of human primordial germ cell fate. Cell 2014; 160:253-68. [PMID: 25543152 PMCID: PMC4310934 DOI: 10.1016/j.cell.2014.12.013] [Citation(s) in RCA: 599] [Impact Index Per Article: 54.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Revised: 11/13/2014] [Accepted: 12/04/2014] [Indexed: 12/20/2022]
Abstract
Specification of primordial germ cells (PGCs) marks the beginning of the totipotent state. However, without a tractable experimental model, the mechanism of human PGC (hPGC) specification remains unclear. Here, we demonstrate specification of hPGC-like cells (hPGCLCs) from germline competent pluripotent stem cells. The characteristics of hPGCLCs are consistent with the embryonic hPGCs and a germline seminoma that share a CD38 cell-surface marker, which collectively defines likely progression of the early human germline. Remarkably, SOX17 is the key regulator of hPGC-like fate, whereas BLIMP1 represses endodermal and other somatic genes during specification of hPGCLCs. Notable mechanistic differences between mouse and human PGC specification could be attributed to their divergent embryonic development and pluripotent states, which might affect other early cell-fate decisions. We have established a foundation for future studies on resetting of the epigenome in hPGCLCs and hPGCs for totipotency and the transmission of genetic and epigenetic information. A defined model for hPGCLC specification from germline-competent hESCs Expression profiles of hPGCLCs match with authentic hPGCs SOX17 is the key regulator of hPGCLC CD38 glycoprotein is a cell-surface marker of the human germline
Collapse
Affiliation(s)
- Naoko Irie
- Wellcome Trust Cancer Research UK Gurdon Institute, Tennis Court Road, University of Cambridge, Cambridge CB2 1QN, UK; Department of Physiology, Development and Neuroscience, Downing Street, University of Cambridge, Cambridge CB2 3EG, UK; Wellcome Trust-Medical Research Council Stem Cell Institute, Tennis Court Road, University of Cambridge, Cambridge CB2 3EG, UK
| | - Leehee Weinberger
- The Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Walfred W C Tang
- Wellcome Trust Cancer Research UK Gurdon Institute, Tennis Court Road, University of Cambridge, Cambridge CB2 1QN, UK; Department of Physiology, Development and Neuroscience, Downing Street, University of Cambridge, Cambridge CB2 3EG, UK; Wellcome Trust-Medical Research Council Stem Cell Institute, Tennis Court Road, University of Cambridge, Cambridge CB2 3EG, UK
| | - Toshihiro Kobayashi
- Wellcome Trust Cancer Research UK Gurdon Institute, Tennis Court Road, University of Cambridge, Cambridge CB2 1QN, UK; Department of Physiology, Development and Neuroscience, Downing Street, University of Cambridge, Cambridge CB2 3EG, UK; Wellcome Trust-Medical Research Council Stem Cell Institute, Tennis Court Road, University of Cambridge, Cambridge CB2 3EG, UK
| | - Sergey Viukov
- The Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Yair S Manor
- The Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Sabine Dietmann
- Wellcome Trust-Medical Research Council Stem Cell Institute, Tennis Court Road, University of Cambridge, Cambridge CB2 3EG, UK
| | - Jacob H Hanna
- The Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 76100, Israel.
| | - M Azim Surani
- Wellcome Trust Cancer Research UK Gurdon Institute, Tennis Court Road, University of Cambridge, Cambridge CB2 1QN, UK; Department of Physiology, Development and Neuroscience, Downing Street, University of Cambridge, Cambridge CB2 3EG, UK; Wellcome Trust-Medical Research Council Stem Cell Institute, Tennis Court Road, University of Cambridge, Cambridge CB2 3EG, UK.
| |
Collapse
|
87
|
Kim Y, Jeong J, Kang H, Lim J, Heo J, Ratajczak J, Ratajczak MZ, Shin DM. The molecular nature of very small embryonic-like stem cells in adult tissues. Int J Stem Cells 2014; 7:55-62. [PMID: 25473442 PMCID: PMC4249904 DOI: 10.15283/ijsc.2014.7.2.55] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/09/2014] [Indexed: 11/26/2022] Open
Abstract
Pluripotent stem cells (PSCs) have been considered as the most important cells in regenerative medicine as they are able to differentiate into all types of cells in the human body. PSCs have been established from several sources of embryo tissue or by reprogramming of terminally differentiated adult tissue by transduction of so-called Yamanaka factors (Oct4, Sox2, Klf4, and cMyc). Interestingly, accumulating evidence has demonstrated the residence of PSCs in adult tissue and with the ability to differentiate into multiple types of tissue-committed stem cells (TCSCs). We also recently demonstrated that a population of pluripotent Oct4+ SSEA-1+Sca-1+Lin−CD45− very small embryonic-like stem cells (VSELs) resides in the adult murine bone marrow (BM) and in other murine tissue. These very small (∼3–6 μm) cells express pluripotent markers such as Oct4, Nanog, and SSEA-1. VSELs could be specified into several tissue-residing TCSCs in response to tissue/organ injury, and thus suggesting that these cells have a physiological role in the rejuvenation of a pool of TCSCs under steady-state conditions. In this review article, we discuss the molecular nature of the rare population of VSELs which have a crucial role in regulating the pluripotency, proliferation, differentiation, and aging of these cells.
Collapse
Affiliation(s)
- YongHwan Kim
- Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul, Korea ; Department of Physiology, University of Ulsan College of Medicine, Seoul, Korea
| | - Jaeho Jeong
- Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul, Korea ; Department of Physiology, University of Ulsan College of Medicine, Seoul, Korea
| | - Hyunsook Kang
- Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul, Korea ; Department of Physiology, University of Ulsan College of Medicine, Seoul, Korea
| | - Jisun Lim
- Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul, Korea ; Department of Physiology, University of Ulsan College of Medicine, Seoul, Korea
| | - Jinbeom Heo
- Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul, Korea ; Department of Physiology, University of Ulsan College of Medicine, Seoul, Korea
| | - Janina Ratajczak
- Stem Cell Institute at the James Graham Brown Cancer Center, University of Louisville, KY, USA
| | - Mariusz Z Ratajczak
- Stem Cell Institute at the James Graham Brown Cancer Center, University of Louisville, KY, USA
| | - Dong-Myung Shin
- Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul, Korea ; Department of Physiology, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
88
|
Ratajczak MZ, Marycz K, Poniewierska-Baran A, Fiedorowicz K, Zbucka-Kretowska M, Moniuszko M. Very small embryonic-like stem cells as a novel developmental concept and the hierarchy of the stem cell compartment. Adv Med Sci 2014; 59:273-280. [PMID: 25170822 DOI: 10.1016/j.advms.2014.08.001] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Revised: 07/06/2014] [Accepted: 08/04/2014] [Indexed: 01/14/2023]
Abstract
Our current understanding of stem cells suffers from a lack of precision, as the stem cell compartment is a broad continuum between early stages of development and adult postnatal tissues, and it is not fully understood how this transition occurs. The definition of stem cell pluripotency is adapted from embryology and excludes the possibility that some early-development stem cells with pluri- and/or multipotential differentiation potential may reside in postnatal tissues in a dormant state in which they are protected from uncontrolled proliferation and thus do not form teratomas or have the ability to complement blastocyst development. We will discuss the concept that a population of very small embryonic-like stem cells (VSELs) could be a link between early-development stages and adult stem cell compartments and reside in a quiescent state in adult tissues. The epigenetic mechanism identified that changes expression of certain genes involved in insulin/insulin-like growth factor signaling (IIS) in VSELs, on the one hand, keeps these cells quiescent in adult tissues and, on the other hand, provides a novel view of the stem cell compartment, IIS, tissue/organ rejuvenation, aging, and cancerogenesis.
Collapse
Affiliation(s)
- Mariusz Z Ratajczak
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA; Department of Physiology, Pomeranian Medical University, Szczecin, Poland.
| | - Krzysztof Marycz
- University of Environmental and Life Sciences, Electron Microscopy Laboratory, Wroclaw, Poland; Wroclaw Research Centre EIT+, Wroclaw, Poland
| | - Agata Poniewierska-Baran
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA; Department of Physiology, Pomeranian Medical University, Szczecin, Poland
| | | | - Monika Zbucka-Kretowska
- Department of Reproduction and Gynecological Endocrinology, Medical University of Bialystok, Bialystok, Poland
| | - Marcin Moniuszko
- Department of Regenerative Medicine and Immune Regulation, Medical University of Bialystok, Bialystok, Poland; Department of Allergology and Internal Medicine, Medical University of Bialystok, Bialystok, Poland
| |
Collapse
|
89
|
Ghasemzadeh-Hasankolaei M, Eslaminejad MB, Batavani R, Ghasemzadeh-Hasankolaei M. Male and female rat bone marrow-derived mesenchymal stem cells are different in terms of the expression of germ cell specific genes. Anat Sci Int 2014; 90:187-96. [DOI: 10.1007/s12565-014-0250-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Accepted: 07/17/2014] [Indexed: 11/24/2022]
|
90
|
Messerschmidt DM, Knowles BB, Solter D. DNA methylation dynamics during epigenetic reprogramming in the germline and preimplantation embryos. Genes Dev 2014; 28:812-28. [PMID: 24736841 PMCID: PMC4003274 DOI: 10.1101/gad.234294.113] [Citation(s) in RCA: 467] [Impact Index Per Article: 42.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Methylation of DNA is an essential epigenetic control mechanism in mammals. Messerschmidt et al. review the current understanding of epigenetic dynamics regulating the molecular processes that prepare the mammalian embryo for normal development. Methylation of DNA is an essential epigenetic control mechanism in mammals. During embryonic development, cells are directed toward their future lineages, and DNA methylation poses a fundamental epigenetic barrier that guides and restricts differentiation and prevents regression into an undifferentiated state. DNA methylation also plays an important role in sex chromosome dosage compensation, the repression of retrotransposons that threaten genome integrity, the maintenance of genome stability, and the coordinated expression of imprinted genes. However, DNA methylation marks must be globally removed to allow for sexual reproduction and the adoption of the specialized, hypomethylated epigenome of the primordial germ cell and the preimplantation embryo. Recent technological advances in genome-wide DNA methylation analysis and the functional description of novel enzymatic DNA demethylation pathways have provided significant insights into the molecular processes that prepare the mammalian embryo for normal development.
Collapse
Affiliation(s)
- Daniel M Messerschmidt
- Developmental Epigenetics and Disease, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology, and Research (A*STAR), 138673 Singapore,
| | | | | |
Collapse
|
91
|
Attari F, Sepehri H, Ansari H, Hassani SN, Esfandiari F, Asgari B, Shahverdi A, Baharvand H. Efficient induction of pluripotency in primordial germ cells by dual inhibition of TGF-β and ERK signaling pathways. Stem Cells Dev 2014; 23:1050-1061. [PMID: 24382167 DOI: 10.1089/scd.2013.0438] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Primordial germ cells (PGCs) have the ability to be reprogrammed into a pluripotent state and are defined as embryonic germ cells (EGCs) in vitro. EGC formation is more efficient, has a shorter culture period than somatic cell reprogramming, and does not require exogenous genetic manipulation. Therefore, EGCs are a good model to analyze mechanisms by which committed cells acquire a pluripotent state. In the present study we have attempted to elucidate a more defined and robust protocol that promotes EGC generation through the suppression of transforming growth factor-β (TGF-β) and extracellular signal-regulated kinase (ERK) signaling pathways by SB431542 (SB) and PD0325901 (PD), respectively. Under this condition the efficiency of transformation of PGCs into EGCs was more than the inhibition of glycogen synthase kinase 3 and ERK signaling pathways. Pluripotency of the resultant-derived EGC lines were further confirmed by gene expression, immunofluorescent staining, directed differentiation ability, teratoma formation, and their contribution to chimeric mice and germ-line transmission. These results showed that PGCs from different embryonic stages (E8.5 and E12.5) could be reprogrammed, maintained, and expanded efficiently under feeder- and serum-free chemically defined conditions by dual inhibition of TGF-β and ERK signaling pathways, regardless of the animal's genetic background.
Collapse
Affiliation(s)
- Farnoosh Attari
- 1 Department of Animal Biology, School of Biology and Center of Excellence in Phylogeny of Living Organisms, College of Science, University of Tehran , Tehran, Iran
| | | | | | | | | | | | | | | |
Collapse
|
92
|
Wessel GM, Fresques T, Kiyomoto M, Yajima M, Zazueta V. Origin and development of the germ line in sea stars. Genesis 2014; 52:367-77. [PMID: 24648114 PMCID: PMC4116737 DOI: 10.1002/dvg.22772] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2014] [Accepted: 03/14/2014] [Indexed: 12/17/2022]
Abstract
This review summarizes and integrates our current understanding of how sea stars make gametes. Although little is known of the mechanism of germ line formation in these animals, recent results point to specific cells and to cohorts of molecules in the embryos and larvae that may lay the ground work for future research efforts. A coelomic outpocketing forms in the posterior of the gut in larvae, referred to as the posterior enterocoel (PE), that when removed, significantly reduces the number of germ cell later in larval growth. This same PE structure also selectively accumulates several germ-line associated factors-vasa, nanos, piwi-and excludes factors involved in somatic cell fate. Since its formation is relatively late in development, these germ cells may form by inductive mechanisms. When integrated into the morphological observations of germ cells and gonad development in larvae, juveniles, and adults, the field of germ line determination appears to have a good model system to study inductive germ line determination to complement the recent work on the molecular mechanisms in mice. We hope this review will also guide investigators interested in germ line determination and regulation of the germ line into how these animals can help in this research field. The review is not intended to be comprehensive-sea star reproduction has been studied for over 100 years and many reviews are comprehensive in their coverage of, for example, seasonal growth of the gonads in response to light, nutrient, and temperature. Rather the intent of this review is to help the reader focus on new experimental results attached to the historical underpinnings of how the germ cell functions in sea stars with particular emphasis to clarify the important areas of priority for future research.
Collapse
Affiliation(s)
- Gary M. Wessel
- Department of Molecular and Cellular Biology, Brown University, Providence RI USA
| | - Tara Fresques
- Department of Molecular and Cellular Biology, Brown University, Providence RI USA
| | | | - Mamiko Yajima
- Department of Molecular and Cellular Biology, Brown University, Providence RI USA
| | - Vanesa Zazueta
- Department of Molecular and Cellular Biology, Brown University, Providence RI USA
| |
Collapse
|
93
|
In mouse oocytes the mitochondrion-originated germinal body-like structures accumulate mouse Vasa homologue (MVH) protein. ZYGOTE 2014; 23:501-6. [DOI: 10.1017/s0967199414000124] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
SummaryMouse Vasa homologue (MVH) antibodies were applied to mouse Graafian oocytes to clarify if mitochondrion-originated germinal body-like structures, described previously by conventional electron microscopy, were associated with the germ plasm. It was found that both the mitochondrion-like structures with cristae and the germinal body-like structures that lacked any signs of cristae were labelled specifically by the anti-MVH antibody. Moreover, some granules were MVH-positive ultrastructural hybrids of the mitochondria and germinal body-like structures, the presence of which clearly supported the idea of a mitochondrial origin for the germinal body-like structures. This finding is the first evidence that mitochondrion-originated germinal body-like granules represent mouse germ plasm.
Collapse
|
94
|
Suszynska M, Zuba-Surma EK, Maj M, Mierzejewska K, Ratajczak J, Kucia M, Ratajczak MZ. The proper criteria for identification and sorting of very small embryonic-like stem cells, and some nomenclature issues. Stem Cells Dev 2014; 23:702-713. [PMID: 24299281 PMCID: PMC3967357 DOI: 10.1089/scd.2013.0472] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Accepted: 11/27/2013] [Indexed: 01/14/2023] Open
Abstract
Evidence has accumulated that both murine and human adult tissues contain early-development stem cells with a broader differentiation potential than other adult monopotent stem cells. These cells, being pluripotent or multipotent, exist at different levels of specification and most likely represent overlapping populations of cells that, depending on the isolation strategy, ex vivo expansion protocol, and markers employed for their identification, have been given different names. In this review, we will discuss a population of very small embryonic-like stem cells (VSELs) in the context of other stem cells that express pluripotent/multipotent markers isolated from adult tissues as well as review the most current, validated working criteria on how to properly identify and isolate these very rare cells. VSELs have been successfully purified in several laboratories; however, a few have failed to isolate them, which has raised some unnecessary controversy in the field. Therefore, in this short review, we will address the most important reasons that some investigators have experienced problems in isolating these very rare cells and discuss some still unresolved challenges which should be overcome before these cells can be widely employed in the clinic.
Collapse
Affiliation(s)
- Malwina Suszynska
- Stem Cell Institute at the James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky
- Department of Physiology Pomeranian Medial University, Szczecin, Poland
| | - Ewa K. Zuba-Surma
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Magdalena Maj
- Stem Cell Institute at the James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky
| | | | - Janina Ratajczak
- Stem Cell Institute at the James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky
- Department of Physiology Pomeranian Medial University, Szczecin, Poland
| | - Magda Kucia
- Stem Cell Institute at the James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky
- Department of Physiology Pomeranian Medial University, Szczecin, Poland
| | - Mariusz Z. Ratajczak
- Stem Cell Institute at the James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky
- Department of Physiology Pomeranian Medial University, Szczecin, Poland
| |
Collapse
|
95
|
Ratajczak MZ, Zuba-Surma E, Wojakowski W, Suszynska M, Mierzejewska K, Liu R, Ratajczak J, Shin DM, Kucia M. Very small embryonic-like stem cells (VSELs) represent a real challenge in stem cell biology: recent pros and cons in the midst of a lively debate. Leukemia 2014; 28:473-484. [PMID: 24018851 PMCID: PMC3948156 DOI: 10.1038/leu.2013.255] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2013] [Accepted: 08/28/2013] [Indexed: 02/07/2023]
Abstract
The concept that adult tissue, including bone marrow (BM), contains early-development cells with broader differentiation potential has again been recently challenged. In response, we would like to review the accumulated evidence from several independent laboratories that adult tissues, including BM, harbor a population of very rare stem cells that may cross germ layers in their differentiation potential. Thus, the BM stem cell compartment hierarchy needs to be revisited. These dormant, early-development cells that our group described as very small embryonic-like stem cells (VSELs) most likely overlap with similar populations of stem cells that have been identified in adult tissues by other investigators as the result of various experimental strategies and have been given various names. As reported, murine VSELs have some pluripotent stem cell characteristics. Moreover, they display several epiblast/germline markers that suggest their embryonic origin and developmental deposition in adult BM. Moreover, at the molecular level, changes in expression of parentally imprinted genes (for example, Igf2-H19) and resistance to insulin/insulin-like growth factor signaling (IIS) regulates their quiescent state in adult tissues. In several emergency situations related to organ damage, VSELs can be activated and mobilized into peripheral blood, and in appropriate animal models they contribute to tissue organ/regeneration. Interestingly, their number correlates with lifespan in mice, and they may also be involved in some malignancies. VSELs have been successfully isolated in several laboratories; however, some investigators experience problems with their isolation.
Collapse
Affiliation(s)
- M Z Ratajczak
- Stem Cell Institute at the James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA
| | - E Zuba-Surma
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - W Wojakowski
- Third Department of Cardiology, Silesian Medical University, Katowice, Poland
| | - M Suszynska
- Stem Cell Institute at the James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA
| | - K Mierzejewska
- Department of Physiology, Pomeranian Medical University, Szczecin, Poland
| | - R Liu
- Stem Cell Institute at the James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA
| | - J Ratajczak
- Stem Cell Institute at the James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA
- Department of Physiology, Pomeranian Medical University, Szczecin, Poland
| | - D M Shin
- Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul, Korea
| | - M Kucia
- Stem Cell Institute at the James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA
- Department of Physiology, Pomeranian Medical University, Szczecin, Poland
| |
Collapse
|
96
|
|
97
|
Abstract
Primordial germ cells (PGCs) are the precursors of sperm and eggs, which generate a new organism that is capable of creating endless new generations through germ cells. PGCs are specified during early mammalian postimplantation development, and are uniquely programmed for transmission of genetic and epigenetic information to subsequent generations. In this Primer, we summarise the establishment of the fundamental principles of PGC specification during early development and discuss how it is now possible to make mouse PGCs from pluripotent embryonic stem cells, and indeed somatic cells if they are first rendered pluripotent in culture.
Collapse
Affiliation(s)
- Erna Magnúsdóttir
- Wellcome Trust, Cancer Research UK, Gurdon Institute, University of Cambridge, Cambridge CB2 1QN, UK
| | | |
Collapse
|
98
|
Durruthy Durruthy J, Ramathal C, Sukhwani M, Fang F, Cui J, Orwig KE, Reijo Pera RA. Fate of induced pluripotent stem cells following transplantation to murine seminiferous tubules. Hum Mol Genet 2014; 23:3071-84. [PMID: 24449759 PMCID: PMC4030765 DOI: 10.1093/hmg/ddu012] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Studies of human germ cell development are limited in large part by inaccessibility of germ cells during development. Moreover, although several studies have reported differentiation of mouse and human germ cells from pluripotent stem cells (PSCs) in vitro, differentiation of human germ cells from PSCs in vivo has not been reported. Here, we tested whether mRNA reprogramming in combination with xeno-transplantation may provide a viable system to probe the genetics of human germ cell development via use of induced pluripotent stem cells (iPSCs). For this purpose, we derived integration-free iPSCs via mRNA-based reprogramming with OCT3/4, SOX2, KLF4 and cMYC alone (OSKM) or in combination with the germ cell-specific mRNA, VASA (OSKMV). All iPSC lines met classic criteria of pluripotency. Moreover, global gene expression profiling did not distinguish large differences between undifferentiated OSKM and OSKMV iPSCs; however, some differences were observed in expression of pluripotency factors and germ cell-specific genes, and in epigenetic profiles and in vitro differentiation studies. In contrast, transplantation of undifferentiated iPSCs directly into the seminiferous tubules of germ cell-depleted immunodeficient mice revealed divergent fates of iPSCs produced with different factors. Transplantation resulted in morphologically and immunohistochemically recognizable germ cells in vivo, particularly in the case of OSKMV cells. Significantly, OSKMV cells also did not form tumors while OSKM cells that remained outside the seminiferous tubule proliferated extensively and formed tumors. Results indicate that mRNA reprogramming in combination with transplantation may contribute to tools for genetic analysis of human germ cell development.
Collapse
Affiliation(s)
- Jens Durruthy Durruthy
- Department of Genetics and Department of Obstetrics and Gynecology, Institute for Stem Cell Biology and Regenerative Medicine, Center for Reproductive and Stem Cell Biology, Stanford University, Stanford, CA 94305, USA and
| | - Cyril Ramathal
- Department of Genetics and Department of Obstetrics and Gynecology, Institute for Stem Cell Biology and Regenerative Medicine, Center for Reproductive and Stem Cell Biology, Stanford University, Stanford, CA 94305, USA and
| | - Meena Sukhwani
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Pittsburgh School of Medicine, Magee-Womens Research Institute, Pittsburgh, PA 15213, USA
| | - Fang Fang
- Department of Genetics and Department of Obstetrics and Gynecology, Institute for Stem Cell Biology and Regenerative Medicine, Center for Reproductive and Stem Cell Biology, Stanford University, Stanford, CA 94305, USA and
| | - Jun Cui
- Department of Genetics and Department of Obstetrics and Gynecology, Institute for Stem Cell Biology and Regenerative Medicine, Center for Reproductive and Stem Cell Biology, Stanford University, Stanford, CA 94305, USA and
| | - Kyle E Orwig
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Pittsburgh School of Medicine, Magee-Womens Research Institute, Pittsburgh, PA 15213, USA
| | - Renee A Reijo Pera
- Department of Genetics and Department of Obstetrics and Gynecology, Institute for Stem Cell Biology and Regenerative Medicine, Center for Reproductive and Stem Cell Biology, Stanford University, Stanford, CA 94305, USA and
| |
Collapse
|
99
|
Wang Q, Liu X, Tang N, Archambeault DR, Li J, Song H, Tang C, He B, Matzuk MM, Wang Y. GASZ promotes germ cell derivation from embryonic stem cells. Stem Cell Res 2013; 11:845-60. [DOI: 10.1016/j.scr.2013.05.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2012] [Revised: 04/30/2013] [Accepted: 05/24/2013] [Indexed: 10/26/2022] Open
|
100
|
Naeemipour M, Dehghani H, Bassami M, Bahrami A. Expression dynamics of pluripotency genes in chicken primordial germ cells before and after colonization of the genital ridges. Mol Reprod Dev 2013; 80:849-61. [PMID: 23877993 DOI: 10.1002/mrd.22216] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2013] [Accepted: 07/12/2013] [Indexed: 01/24/2023]
Abstract
Mammalian species utilize an inductive mechanism of germ cell specification, diverting the fate of some of somatic cells toward pluripotency and germ-cell totipotency. It is not known if avian species utilize a similar mechanism nor if, analogous to mammalian primordial germ cells (PGCs), pluripotency genes are continuously upregulated in migrating and genital ridge-colonizing avian PGCs. Thus, this study was conducted to quantify and to analyze the expression profile of pluripotency genes at different stages of chicken PGCs development at Hamburger and Hamilton (HH) stage 14, when the majority of PGCs have entered into the bloodstream; at HH stage 18, when PGCs have resided for 8-12 hr in the bloodstream; and at HH stage 28, when the majority of PGCs are found in the genital ridge. The transcription for Oct4, Sox2, and Nanog continuously decreased from HH stage 14 to HH stage 28. In addition, equal amounts of total RNA could be isolated from chicken PGCs at each stage of development, indicating that the observed drop of transcription of pluripotency genes is not a consequence of transcriptional repression in general. Decreased expression for all three proteins was also observed at HH stage 28. Furthermore, in comparison to blood PGCs, those residing in the gonad have lost their full capacity for colony formation. Our results indicate that, in contrast to mammalian PGCs, chicken PGCs continuously downregulate the expression of pluripotency genes and show a progressive loss of pluripotency-associated features during different stages of germ-line migration.
Collapse
Affiliation(s)
- Mohsen Naeemipour
- Department of Basic Science, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| | | | | | | |
Collapse
|