1
|
Hasegawa K, Zhao Y, Garbuzov A, Corces MR, Neuhöfer P, Gillespie VM, Cheung P, Belk JA, Huang YH, Wei Y, Chen L, Chang HY, Artandi SE. Clonal inactivation of TERT impairs stem cell competition. Nature 2024; 632:201-208. [PMID: 39020172 PMCID: PMC11291281 DOI: 10.1038/s41586-024-07700-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 06/11/2024] [Indexed: 07/19/2024]
Abstract
Telomerase is intimately associated with stem cells and cancer, because it catalytically elongates telomeres-nucleoprotein caps that protect chromosome ends1. Overexpression of telomerase reverse transcriptase (TERT) enhances the proliferation of cells in a telomere-independent manner2-8, but so far, loss-of-function studies have provided no evidence that TERT has a direct role in stem cell function. In many tissues, homeostasis is shaped by stem cell competition, a process in which stem cells compete on the basis of inherent fitness. Here we show that conditional deletion of Tert in the spermatogonial stem cell (SSC)-containing population in mice markedly impairs competitive clone formation. Using lineage tracing from the Tert locus, we find that TERT-expressing SSCs yield long-lived clones, but that clonal inactivation of TERT promotes stem cell differentiation and a genome-wide reduction in open chromatin. This role for TERT in competitive clone formation occurs independently of both its reverse transcriptase activity and the canonical telomerase complex. Inactivation of TERT causes reduced activity of the MYC oncogene, and transgenic expression of MYC in the TERT-deleted pool of SSCs efficiently rescues clone formation. Together, these data reveal a catalytic-activity-independent requirement for TERT in enhancing stem cell competition, uncover a genetic connection between TERT and MYC and suggest that a selective advantage for stem cells with high levels of TERT contributes to telomere elongation in the male germline during homeostasis and ageing.
Collapse
Affiliation(s)
- Kazuteru Hasegawa
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
| | - Yang Zhao
- Center for Personal Dynamic Regulomes, Stanford, CA, USA
| | - Alina Garbuzov
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
| | - M Ryan Corces
- Center for Personal Dynamic Regulomes, Stanford, CA, USA
| | - Patrick Neuhöfer
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
| | - Victoria M Gillespie
- Nuclear Dynamics and Cancer Program, Cancer Epigenetics Institute, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Peggie Cheung
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
| | - Julia A Belk
- Department of Computer Science, Stanford University, Stanford, CA, USA
| | | | - Yuning Wei
- Center for Personal Dynamic Regulomes, Stanford, CA, USA
| | - Lu Chen
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
- Nuclear Dynamics and Cancer Program, Cancer Epigenetics Institute, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Howard Y Chang
- Center for Personal Dynamic Regulomes, Stanford, CA, USA
- Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA
| | - Steven E Artandi
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
2
|
Rehman A, Fatima I, Noor F, Qasim M, Wang P, Jia J, Alshabrmi FM, Liao M. Role of small molecules as drug candidates for reprogramming somatic cells into induced pluripotent stem cells: A comprehensive review. Comput Biol Med 2024; 177:108661. [PMID: 38810477 DOI: 10.1016/j.compbiomed.2024.108661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 04/08/2024] [Accepted: 05/26/2024] [Indexed: 05/31/2024]
Abstract
With the use of specific genetic factors and recent developments in cellular reprogramming, it is now possible to generate lineage-committed cells or induced pluripotent stem cells (iPSCs) from readily available and common somatic cell types. However, there are still significant doubts regarding the safety and effectiveness of the current genetic methods for reprogramming cells, as well as the conventional culture methods for maintaining stem cells. Small molecules that target specific epigenetic processes, signaling pathways, and other cellular processes can be used as a complementary approach to manipulate cell fate to achieve a desired objective. It has been discovered that a growing number of small molecules can support lineage differentiation, maintain stem cell self-renewal potential, and facilitate reprogramming by either increasing the efficiency of reprogramming or acting as a genetic reprogramming factor substitute. However, ongoing challenges include improving reprogramming efficiency, ensuring the safety of small molecules, and addressing issues with incomplete epigenetic resetting. Small molecule iPSCs have significant clinical applications in regenerative medicine and personalized therapies. This review emphasizes the versatility and potential safety benefits of small molecules in overcoming challenges associated with the iPSCs reprogramming process.
Collapse
Affiliation(s)
- Abdur Rehman
- Center of Bioinformatics, College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, 712100, PR China
| | - Israr Fatima
- Center of Bioinformatics, College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, 712100, PR China
| | - Fatima Noor
- Institute of Molecular Biology and Biotechnology, The University of Lahore, Lahore, Pakistan; Department of Bioinformatics and Biotechnology, Government College University of Faisalabad, 38000, Pakistan
| | - Muhammad Qasim
- Department of Bioinformatics and Biotechnology, Government College University of Faisalabad, 38000, Pakistan
| | - Peng Wang
- Center of Bioinformatics, College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, 712100, PR China
| | - Jinrui Jia
- Laboratory of Animal Fat Deposition and Muscle Development, Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, PR China
| | - Fahad M Alshabrmi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, 51452, Saudi Arabia
| | - Mingzhi Liao
- Center of Bioinformatics, College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, 712100, PR China.
| |
Collapse
|
3
|
Mukhamadiarov RI, Ciarchi M, Olmeda F, Rulands S. Clonal dynamics of surface-driven growing tissues. Phys Rev E 2024; 109:064407. [PMID: 39021023 DOI: 10.1103/physreve.109.064407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 05/17/2024] [Indexed: 07/20/2024]
Abstract
The self-organization of cells into complex tissues relies on a tight coordination of cell behavior. Identifying the cellular processes driving tissue growth is key to understanding the emergence of tissue forms and devising targeted therapies for aberrant growth, such as in cancer. Inferring the mode of tissue growth, whether it is driven by cells on the surface or by cells in the bulk, is possible in cell culture experiments but difficult in most tissues in living organisms (in vivo). Genetic tracing experiments, where a subset of cells is labeled with inheritable markers, have become important experimental tools to study cell fate in vivo. Here we show that the mode of tissue growth is reflected in the size distribution of the progeny of marked cells. To this end, we derive the clone size distributions using analytical calculations in the limit of negligible cell migration and cell death, and we test our predictions with an agent-based stochastic sampling technique. We show that for surface-driven growth the clone size distribution takes a characteristic power-law form with an exponent determined by fluctuations of the tissue surface. Our results propose a possible way of determining the mode of tissue growth from genetic tracing experiments.
Collapse
|
4
|
Kuo YP, Nombela-Arrieta C, Carja O. A theory of evolutionary dynamics on any complex population structure reveals stem cell niche architecture as a spatial suppressor of selection. Nat Commun 2024; 15:4666. [PMID: 38821923 PMCID: PMC11143212 DOI: 10.1038/s41467-024-48617-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 05/02/2024] [Indexed: 06/02/2024] Open
Abstract
How the spatial arrangement of a population shapes its evolutionary dynamics has been of long-standing interest in population genetics. Most previous studies assume a small number of demes or symmetrical structures that, most often, act as well-mixed populations. Other studies use network theory to study more heterogeneous spatial structures, however they usually assume small, regular networks, or strong constraints on the strength of selection considered. Here we build network generation algorithms, conduct evolutionary simulations and derive general analytic approximations for probabilities of fixation in populations with complex spatial structure. We build a unifying evolutionary theory across network families and derive the relevant selective parameter, which is a combination of network statistics, predictive of evolutionary dynamics. We also illustrate how to link this theory with novel datasets of spatial organization and use recent imaging data to build the cellular spatial networks of the stem cell niches of the bone marrow. Across a wide variety of parameters, we find these networks to be strong suppressors of selection, delaying mutation accumulation in this tissue. We also find that decreases in stem cell population size also decrease the suppression strength of the tissue spatial structure.
Collapse
Affiliation(s)
- Yang Ping Kuo
- Computational Biology Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA, USA
- Joint Carnegie Mellon University-University of Pittsburgh Ph.D. Program in Computational Biology, Carnegie Mellon University, Pittsburgh, PA, USA
| | - César Nombela-Arrieta
- Department of Medical Oncology and Hematology, University and University Hospital Zurich, Zurich, Switzerland
| | - Oana Carja
- Computational Biology Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA, USA.
| |
Collapse
|
5
|
Bener MB, Slepchenko BM, Inaba M. Asymmetric stem cell division maintains the genetic heterogeneity of tissue cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.16.594576. [PMID: 38798517 PMCID: PMC11118488 DOI: 10.1101/2024.05.16.594576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Within a given tissue, the stem cell niche provides the microenvironment for stem cells suitable for their self-renewal. Conceptually, the niche space constrains the size of a stem-cell pool, as the cells sharing the niche compete for its space. It has been suggested that either neutral- or non-neutral-competition of stem cells changes the clone dynamics of stem cells. Theoretically, if the rate of asymmetric division is high, the stem cell competition is limited, thus suppressing clonal expansion. However, the effects of asymmetric division on clone dynamics have never been experimentally tested. Here, using the Drosophila germline stem cell (GSC) system, as a simple model of the in-vivo niche, we examine the effect of division modes (asymmetric or symmetric) on clonal dynamics by combining experimental approaches with mathematical modeling. Our experimental data and computational model both suggest that the rate of asymmetric division is proportional to the time a stem cell clone takes to expand. Taken together, our data suggests that asymmetric division is essential for maintaining the genetic variation of stem cells and thus serves as a critical mechanism for safeguarding fertility over the animal age or preventing multiple disorders caused by the clonal expansion of stem cells.
Collapse
Affiliation(s)
- Muhammed Burak Bener
- Department of Cell Biology, University of Connecticut School of Medicine, Farmington, CT 06030
| | - Boris M. Slepchenko
- Department of Cell Biology, University of Connecticut School of Medicine, Farmington, CT 06030
- Richard D. Berlin Center for Cell Analysis and Modeling, University of Connecticut School of Medicine, Farmington, CT 06030
| | - Mayu Inaba
- Department of Cell Biology, University of Connecticut School of Medicine, Farmington, CT 06030
| |
Collapse
|
6
|
Yang L, Liao J, Huang H, Lee TL, Qi H. Stage-specific regulation of undifferentiated spermatogonia by AKT1S1-mediated AKT-mTORC1 signaling during mouse spermatogenesis. Dev Biol 2024; 509:11-27. [PMID: 38311163 DOI: 10.1016/j.ydbio.2024.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 11/03/2023] [Accepted: 02/01/2024] [Indexed: 02/10/2024]
Abstract
Undifferentiated spermatogonia are composed of a heterogeneous cell population including spermatogonial stem cells (SSCs). Molecular mechanisms underlying the regulation of various spermatogonial cohorts during their self-renewal and differentiation are largely unclear. Here we show that AKT1S1, an AKT substrate and inhibitor of mTORC1, regulates the homeostasis of undifferentiated spermatogonia. Although deletion of Akt1s1 in mouse appears not grossly affecting steady-state spermatogenesis and male mice are fertile, the subset of differentiation-primed OCT4+ spermatogonia decreased significantly, whereas self-renewing GFRα1+ and proliferating PLZF+ spermatogonia were sustained. Both neonatal prospermatogonia and the first wave spermatogenesis were greatly reduced in Akt1s1-/- mice. Further analyses suggest that OCT4+ spermatogonia in Akt1s1-/- mice possess altered PI3K/AKT-mTORC1 signaling, gene expression and carbohydrate metabolism, leading to their functionally compromised developmental potential. Collectively, these results revealed an important role of AKT1S1 in mediating the stage-specific signals that regulate the self-renewal and differentiation of spermatogonia during mouse spermatogenesis.
Collapse
Affiliation(s)
- Lele Yang
- Center for Cell Lineage and Development, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China; CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China; Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China; GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Center, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China; GIBH-CUHK Joint Research Laboratory on Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Jinyue Liao
- GIBH-CUHK Joint Research Laboratory on Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China; School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong
| | - Hongying Huang
- The Experimental Animal Center, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Tin Lap Lee
- GIBH-CUHK Joint Research Laboratory on Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China; School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong
| | - Huayu Qi
- Center for Cell Lineage and Development, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China; CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China; Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China; GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Center, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China; GIBH-CUHK Joint Research Laboratory on Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China.
| |
Collapse
|
7
|
Murugesh V, Ritting M, Salem S, Aalam SMM, Garcia J, Chattha AJ, Zhao Y, Knapp DJHF, Kalthur G, Granberg CF, Kannan N. Puberty Blocker and Aging Impact on Testicular Cell States and Function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.23.586441. [PMID: 38585884 PMCID: PMC10996503 DOI: 10.1101/2024.03.23.586441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Spermatogonial stem cell (SSC) acquisition of meiotogenetic state during puberty to produce genetically diverse gametes is blocked by drugs collectively referred as 'puberty blocker' (PB). Investigating the impact of PB on juvenile SSC state and function is challenging due to limited tissue access and clinical data. Herein, we report largest clinically annotated juvenile testicular biorepository with all children with gender dysphoria on chronic PB treatment highlighting shift in pediatric patient demography in US. At the tissue level, we report mild-to-severe sex gland atrophy in PB treated children. We developed most extensive integrated single-cell RNA dataset to date (>100K single cells; 25 patients), merging both public and novel (52 month PB-treated) datasets, alongside innovative computational approach tailed for germ cells and evaluated the impact of PB and aging on SSC. We report novel constitutional ranges for each testicular cell type across the entire age spectrum, distinct effects of treatments on prepubertal vs adult SSC, presence of spermatogenic epithelial cells exhibiting post-meiotic-state, irrespective of age, puberty status, or PB treatment. Further, we defined distinct effects of PB and aging on testicular cell lineage composition, and SSC meiotogenetic state and function. Using single cell data from prepubertal and young adult, we were able to accurately predict sexual maturity based both on overall cell type proportions, as well as on gene expression patterns within each major cell type. Applying these models to a PB-treated patient that they appeared pre-pubertal across the entire tissue. This combined with the noted gland atrophy and abnormalities from the histology data raise a potential concern regarding the complete 'reversibility' and reproductive fitness of SSC. The biorepository, data, and research approach presented in this study provide unique opportunity to explore the impact of PB on testicular reproductive health.
Collapse
Affiliation(s)
- Varshini Murugesh
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Megan Ritting
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Salem Salem
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | | | - Joaquin Garcia
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Asma J Chattha
- Department of Pediatrics, Mayo Clinic, Rochester, MN, USA
| | - Yulian Zhao
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Mayo Clinic, Rochester, MN, USA
| | - David JHF Knapp
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
- Institut de Recherche en Immunologie et Cancérologie, and Département de Pathologie et Biologie Cellulaire, Université de Montréal, Montreal, QC, Canada
- Senior authors
| | - Guruprasad Kalthur
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
- Division of Reproductive Biology, Department of Reproductive Science, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, India
- Senior authors
| | | | - Nagarajan Kannan
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
- Mayo Clinic Comprehensive Cancer Center, Mayo Clinic, Rochester, MN, USA
- Center for Regenerative Biotherapeutics, Mayo Clinic, Rochester, MN, USA
- Senior authors
- Lead contact
| |
Collapse
|
8
|
Beumer J, Clevers H. Hallmarks of stemness in mammalian tissues. Cell Stem Cell 2024; 31:7-24. [PMID: 38181752 PMCID: PMC10769195 DOI: 10.1016/j.stem.2023.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 12/03/2023] [Accepted: 12/08/2023] [Indexed: 01/07/2024]
Abstract
All adult tissues experience wear and tear. Most tissues can compensate for cell loss through the activity of resident stem cells. Although the cellular maintenance strategies vary greatly between different adult (read: postnatal) tissues, the function of stem cells is best defined by their capacity to replace lost tissue through division. We discuss a set of six complementary hallmarks that are key enabling features of this basic function. These include longevity and self-renewal, multipotency, transplantability, plasticity, dependence on niche signals, and maintenance of genome integrity. We discuss these hallmarks in the context of some of the best-understood adult stem cell niches.
Collapse
Affiliation(s)
- Joep Beumer
- Institute of Human Biology (IHB), Roche Pharma Research and Early Development, Basel, Switzerland.
| | - Hans Clevers
- Institute of Human Biology (IHB), Roche Pharma Research and Early Development, Basel, Switzerland.
| |
Collapse
|
9
|
Nelson JO, Kumon T, Yamashita YM. rDNA magnification is a unique feature of germline stem cells. Proc Natl Acad Sci U S A 2023; 120:e2314440120. [PMID: 37967216 PMCID: PMC10666004 DOI: 10.1073/pnas.2314440120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 10/16/2023] [Indexed: 11/17/2023] Open
Abstract
Ribosomal DNA (rDNA) encodes ribosomal RNA and exists as tandem repeats of hundreds of copies in the eukaryotic genome to meet the high demand of ribosome biogenesis. Tandemly repeated DNA elements are inherently unstable; thus, mechanisms must exist to maintain rDNA copy number (CN), in particular in the germline that continues through generations. A phenomenon called rDNA magnification was discovered over 50 y ago in Drosophila as a process that recovers the rDNA CN on chromosomes that harbor minimal CN. Our recent studies indicated that rDNA magnification is the mechanism to maintain rDNA CN under physiological conditions to counteract spontaneous CN loss that occurs during aging. Our previous studies that explored the mechanism of rDNA magnification implied that asymmetric division of germline stem cells (GSCs) may be particularly suited to achieve rDNA magnification. However, it remains elusive whether GSCs are the unique cell type that undergoes rDNA magnification or differentiating germ cells are also capable of magnification. In this study, we provide empirical evidence that suggests that rDNA magnification operates uniquely in GSCs, but not in differentiating germ cells. We further provide computer simulation that suggests that rDNA magnification is only achievable through asymmetric GSC divisions. We propose that despite known plasticity and transcriptomic similarity between GSCs and differentiating germ cells, GSCs' unique ability to divide asymmetrically serves a critical role of maintaining rDNA CN through generations, supporting germline immortality.
Collapse
Affiliation(s)
- Jonathan O Nelson
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142
- HHMI, Chevy Chase, MD 20815
| | - Tomohiro Kumon
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142
- HHMI, Chevy Chase, MD 20815
| | - Yukiko M Yamashita
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142
- HHMI, Chevy Chase, MD 20815
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142
| |
Collapse
|
10
|
Reifarth L, Körber H, Packeiser EM, Goericke-Pesch S. Detection of spermatogonial stem cells in testicular tissue of dogs with chronic asymptomatic orchitis. Front Vet Sci 2023; 10:1205064. [PMID: 37396999 PMCID: PMC10311113 DOI: 10.3389/fvets.2023.1205064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 05/25/2023] [Indexed: 07/04/2023] Open
Abstract
Chronic asymptomatic idiopathic orchitis (CAO) is an important but neglected cause of acquired infertility due to non-obstructive azoospermia (NOA) in male dogs. The similarity of the pathophysiology in infertile dogs and men supports the dog's suitability as a possible animal model for studying human diseases causing disruption of spermatogenesis and evaluating the role of spermatogonial stem cells (SSCs) as a new therapeutic approach to restore or recover fertility in cases of CAO. To investigate the survival of resilient stem cells, the expression of the protein gene product (PGP9.5), deleted in azoospermia like (DAZL), foxo transcription factor 1 (FOXO1) and tyrosine-kinase receptor (C-Kit) were evaluated in healthy and CAO-affected canine testes. Our data confirmed the presence of all investigated germ cell markers at mRNA and protein levels. In addition, we postulate a specific expression pattern of FOXO1 and C-Kit in undifferentiated and differentiating spermatogonia, respectively, whereas DAZL and PGP9.5 expressions were confirmed in the entire spermatogonial population. Furthermore, this is the first study revealing a significant reduction of PGP9.5, DAZL, and FOXO1 in CAO at protein and/or gene expression level indicating a severe disruption of spermatogenesis. This means that chronic asymptomatic inflammatory changes in CAO testis are accompanied by a significant loss of SSCs. Notwithstanding, our data confirm the survival of putative stem cells with the potential of self-renewal and differentiation and lay the groundwork for further research into stem cell-based therapeutic options to reinitialize spermatogenesis in canine CAO-affected patients.
Collapse
Affiliation(s)
| | | | | | - Sandra Goericke-Pesch
- Reproductive Unit – Clinic for Small Animals, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| |
Collapse
|
11
|
Shamhari A‘A, Jefferi NES, Abd Hamid Z, Budin SB, Idris MHM, Taib IS. The Role of Promyelocytic Leukemia Zinc Finger (PLZF) and Glial-Derived Neurotrophic Factor Family Receptor Alpha 1 (GFRα1) in the Cryopreservation of Spermatogonia Stem Cells. Int J Mol Sci 2023; 24:ijms24031945. [PMID: 36768269 PMCID: PMC9915902 DOI: 10.3390/ijms24031945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 12/08/2022] [Accepted: 12/14/2022] [Indexed: 01/20/2023] Open
Abstract
The cryopreservation of spermatogonia stem cells (SSCs) has been widely used as an alternative treatment for infertility. However, cryopreservation itself induces cryoinjury due to oxidative and osmotic stress, leading to reduction in the survival rate and functionality of SSCs. Glial-derived neurotrophic factor family receptor alpha 1 (GFRα1) and promyelocytic leukemia zinc finger (PLZF) are expressed during the self-renewal and differentiation of SSCs, making them key tools for identifying the functionality of SSCs. To the best of our knowledge, the involvement of GFRα1 and PLZF in determining the functionality of SSCs after cryopreservation with therapeutic intervention is limited. Therefore, the purpose of this review is to determine the role of GFRα1 and PLZF as biomarkers for evaluating the functionality of SSCs in cryopreservation with therapeutic intervention. Therapeutic intervention, such as the use of antioxidants, and enhancement in cryopreservation protocols, such as cell encapsulation, cryoprotectant agents (CPA), and equilibrium of time and temperature increase the expression of GFRα1 and PLZF, resulting in maintaining the functionality of SSCs. In conclusion, GFRα1 and PLZF have the potential as biomarkers in cryopreservation with therapeutic intervention of SSCs to ensure the functionality of the stem cells.
Collapse
Affiliation(s)
- Asma’ ‘Afifah Shamhari
- Center of Diagnostics, Therapeutics, and Investigative Studies (CODTIS), Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Wilayah Persekutuan, Malaysia
| | - Nur Erysha Sabrina Jefferi
- Center of Diagnostics, Therapeutics, and Investigative Studies (CODTIS), Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Wilayah Persekutuan, Malaysia
| | - Zariyantey Abd Hamid
- Center of Diagnostics, Therapeutics, and Investigative Studies (CODTIS), Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Wilayah Persekutuan, Malaysia
| | - Siti Balkis Budin
- Center of Diagnostics, Therapeutics, and Investigative Studies (CODTIS), Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Wilayah Persekutuan, Malaysia
| | - Muhd Hanis Md Idris
- Integrative Pharmacogenomics Institute (iPROMISE), Universiti Teknologi MARA (UiTM), Puncak Alam Campus, Bandar Puncak Alam 42300, Selangor, Malaysia
| | - Izatus Shima Taib
- Center of Diagnostics, Therapeutics, and Investigative Studies (CODTIS), Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Wilayah Persekutuan, Malaysia
- Correspondence: ; Tel.: +603-928-97608
| |
Collapse
|
12
|
Zhang P, Wang M, Chen X, Jing K, Li Y, Liu X, Ran H, Qin J, Zhong J, Cai X. Dysregulated genes in undifferentiated spermatogonia and Sertoli cells are associated with the spermatogenic arrest in cattleyak. Mol Reprod Dev 2022; 89:632-645. [PMID: 36409004 DOI: 10.1002/mrd.23653] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 11/04/2022] [Accepted: 11/07/2022] [Indexed: 11/23/2022]
Abstract
Hybrid male sterility (HMS) is a reproductive isolation mechanism limiting the formation of fertile offspring through interspecific fertilization. Cattleyak is the interspecific hybrid presenting significant heterosis in several economic traits, but HMS restricted its wide reproduction in cettleyak breeding. In this study, we detected the specifically expressed genes of a variety of cells (undifferentiated spermatogonia, primary spermatocytes, secondary spermatocytes, haploid spermatids, sperm, Sertoli cells, Leydig cells, and macrophages) in the testis of yak and cattleyak, and found that the spermatogenesis of cattleyak might be blocked at meiosis I, and the expression of niche factors (NR5A1, GATA4, STAR, CYP11A1, CD68, TNF, and CX3CR1) in undifferentiated spermatogonia niche was abnormal. Then we isolated the undifferentiated spermatogonia and Sertoli cells from yak and cattleyak by enzyme digestion, and detected the specific genes in the two bovid testicular cells as well as the proliferation capacity of the undifferentiated spermatogonia. These results indicated that weak proliferation ability and scarce number of undifferentiated spermatogonia and abnormal gene expressions in Sertoli cells may contribute to male sterility of cattleyak.
Collapse
Affiliation(s)
- Peng Zhang
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization of Sichuan Province and Ministry of Education, Institute of Qinghai-Tibetan Plateau, Southwest Minzu University, Chengdu, Sichuan, China
| | - Mingxiu Wang
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization of Sichuan Province and Ministry of Education, Institute of Qinghai-Tibetan Plateau, Southwest Minzu University, Chengdu, Sichuan, China
| | - Xuemei Chen
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization of Sichuan Province and Ministry of Education, Institute of Qinghai-Tibetan Plateau, Southwest Minzu University, Chengdu, Sichuan, China
| | - Kemin Jing
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization of Sichuan Province and Ministry of Education, Institute of Qinghai-Tibetan Plateau, Southwest Minzu University, Chengdu, Sichuan, China
| | - Yuqian Li
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization of Sichuan Province and Ministry of Education, Institute of Qinghai-Tibetan Plateau, Southwest Minzu University, Chengdu, Sichuan, China
| | - Xinrui Liu
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization of Sichuan Province and Ministry of Education, Institute of Qinghai-Tibetan Plateau, Southwest Minzu University, Chengdu, Sichuan, China
| | - Hongbiao Ran
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization of Sichuan Province and Ministry of Education, Institute of Qinghai-Tibetan Plateau, Southwest Minzu University, Chengdu, Sichuan, China
| | - Jie Qin
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization of Sichuan Province and Ministry of Education, Institute of Qinghai-Tibetan Plateau, Southwest Minzu University, Chengdu, Sichuan, China
| | - Jincheng Zhong
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization of Sichuan Province and Ministry of Education, Institute of Qinghai-Tibetan Plateau, Southwest Minzu University, Chengdu, Sichuan, China
| | - Xin Cai
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization of Sichuan Province and Ministry of Education, Institute of Qinghai-Tibetan Plateau, Southwest Minzu University, Chengdu, Sichuan, China
| |
Collapse
|
13
|
Huelsz-Prince G, Kok RNU, Goos Y, Bruens L, Zheng X, Ellenbroek S, Van Rheenen J, Tans S, van Zon JS. Mother cells control daughter cell proliferation in intestinal organoids to minimize proliferation fluctuations. eLife 2022; 11:e80682. [PMID: 36445322 PMCID: PMC9708068 DOI: 10.7554/elife.80682] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 10/20/2022] [Indexed: 11/30/2022] Open
Abstract
During renewal of the intestine, cells are continuously generated by proliferation. Proliferation and differentiation must be tightly balanced, as any bias toward proliferation results in uncontrolled exponential growth. Yet, the inherently stochastic nature of cells raises the question how such fluctuations are limited. We used time-lapse microscopy to track all cells in crypts of growing mouse intestinal organoids for multiple generations, allowing full reconstruction of the underlying lineage dynamics in space and time. Proliferative behavior was highly symmetric between sister cells, with both sisters either jointly ceasing or continuing proliferation. Simulations revealed that such symmetric proliferative behavior minimizes cell number fluctuations, explaining our observation that proliferating cell number remained constant even as crypts increased in size considerably. Proliferative symmetry did not reflect positional symmetry but rather lineage control through the mother cell. Our results indicate a concrete mechanism to balance proliferation and differentiation with minimal fluctuations that may be broadly relevant for other tissues.
Collapse
Affiliation(s)
| | | | | | - Lotte Bruens
- Department of Molecular Pathology, Oncode Institute, Netherlands Cancer InstituteAmterdamNetherlands
| | | | - Saskia Ellenbroek
- Department of Molecular Pathology, Oncode Institute, Netherlands Cancer InstituteAmterdamNetherlands
| | - Jacco Van Rheenen
- Department of Molecular Pathology, Oncode Institute, Netherlands Cancer InstituteAmterdamNetherlands
| | | | | |
Collapse
|
14
|
Nakamuta A, Yoshido K, Naoki H. Stem cell homeostasis regulated by hierarchy and neutral competition. Commun Biol 2022; 5:1268. [PMID: 36400843 PMCID: PMC9674595 DOI: 10.1038/s42003-022-04218-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 11/03/2022] [Indexed: 11/19/2022] Open
Abstract
Tissue stem cells maintain themselves through self-renewal while constantly supplying differentiating cells. Two distinct models have been proposed as mechanisms of stem cell homeostasis. According to the classical model, there is hierarchy among stem cells, and master stem cells produce stem cells by asymmetric division; whereas, according to the recent model, stem cells are equipotent and neutrally compete. However, the mechanism remains controversial in several tissues and species. Here, we developed a mathematical model linking the two models, named the hierarchical neutral competition (hNC) model. Our theoretical analysis showed that the combination of the hierarchy and neutral competition exhibited bursts in clonal expansion, which was consistent with experimental data of rhesus macaque hematopoiesis. Furthermore, the scaling law in clone size distribution, considered a unique characteristic of the recent model, was satisfied even in the hNC model. Based on the findings above, we proposed the criterion for distinguishing the three models based on experiments.
Collapse
Affiliation(s)
- Asahi Nakamuta
- grid.258799.80000 0004 0372 2033Laboratory of Theoretical Biology, Graduate School of Biostudies, Kyoto University, Yoshidakonoecho, Sakyo, Kyoto, 606-8315 Japan ,grid.258799.80000 0004 0372 2033Faculty of Science, Kyoto University, Yoshidakonoecho, Sakyo, Kyoto, 606-8315 Japan
| | - Kana Yoshido
- grid.258799.80000 0004 0372 2033Laboratory of Theoretical Biology, Graduate School of Biostudies, Kyoto University, Yoshidakonoecho, Sakyo, Kyoto, 606-8315 Japan
| | - Honda Naoki
- grid.258799.80000 0004 0372 2033Laboratory of Theoretical Biology, Graduate School of Biostudies, Kyoto University, Yoshidakonoecho, Sakyo, Kyoto, 606-8315 Japan ,grid.257022.00000 0000 8711 3200Laboratory of Data-driven Biology, Graduate School of Integrated Sciences for Life, Hiroshima University, Kagamiyama, Higashi-hiroshima, Hiroshima, 739-8526 Japan ,grid.257022.00000 0000 8711 3200Kansei-Brain Informatics Group, Center for Brain, Mind and Kansei Sciences Research (BMK Center), Hiroshima University, Kasumi, Minami-ku, Hiroshima, 734-8551 Japan ,grid.250358.90000 0000 9137 6732Theoretical Biology Research Group, Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Okazaki, Aichi 444-8787 Japan
| |
Collapse
|
15
|
Chen C, Liao Y, Peng G. Connecting past and present: single-cell lineage tracing. Protein Cell 2022; 13:790-807. [PMID: 35441356 PMCID: PMC9237189 DOI: 10.1007/s13238-022-00913-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 03/06/2022] [Indexed: 01/16/2023] Open
Abstract
Central to the core principle of cell theory, depicting cells' history, state and fate is a fundamental goal in modern biology. By leveraging clonal analysis and single-cell RNA-seq technologies, single-cell lineage tracing provides new opportunities to interrogate both cell states and lineage histories. During the past few years, many strategies to achieve lineage tracing at single-cell resolution have been developed, and three of them (integration barcodes, polylox barcodes, and CRISPR barcodes) are noteworthy as they are amenable in experimentally tractable systems. Although the above strategies have been demonstrated in animal development and stem cell research, much care and effort are still required to implement these methods. Here we review the development of single-cell lineage tracing, major characteristics of the cell barcoding strategies, applications, as well as technical considerations and limitations, providing a guide to choose or improve the single-cell barcoding lineage tracing.
Collapse
Affiliation(s)
- Cheng Chen
- Center for Cell Lineage and Development, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Yuanxin Liao
- Center for Cell Lineage and Development, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Guangdun Peng
- Center for Cell Lineage and Development, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China.
- Center for Cell Lineage and Atlas, Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, 510005, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
| |
Collapse
|
16
|
Altshuler A, Wickström SA, Shalom-Feuerstein R. Spotlighting adult stem cells: advances, pitfalls, and challenges. Trends Cell Biol 2022; 33:477-494. [PMID: 36270939 DOI: 10.1016/j.tcb.2022.09.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 09/09/2022] [Accepted: 09/16/2022] [Indexed: 11/06/2022]
Abstract
The existence of stem cells (SCs) at the tip of the cellular differentiation hierarchy has fascinated the scientific community ever since their discovery in the early 1950s to 1960s. Despite the remarkable success of the SC theory and the development of SC-based treatments, fundamental features of SCs remain enigmatic. Recent advances in single-cell lineage tracing, live imaging, and genomic technologies have allowed capture of life histories and transcriptional signatures of individual cells, leaving SCs much less space to 'hide'. Focusing on epithelial SCs and comparing them to other SCs, we discuss new paradigms of the SC niche, dynamics, and pathology, highlighting key open questions in SC biology that need to be resolved for harnessing SC potential in regenerative medicine.
Collapse
|
17
|
Chechekhin VI, Kulebyakin KY, Tyurin-Kuzmin PA. Specific Features of Regulation of Hormonal Sensitivity in Stem Cells. Russ J Dev Biol 2022. [DOI: 10.1134/s106236042203002x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
18
|
Song H, Park HJ, Lee WY, Lee KH. Models and Molecular Markers of Spermatogonial Stem Cells in Vertebrates: To Find Models in Nonmammals. Stem Cells Int 2022; 2022:4755514. [PMID: 35685306 PMCID: PMC9174007 DOI: 10.1155/2022/4755514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 03/21/2022] [Accepted: 04/17/2022] [Indexed: 11/24/2022] Open
Abstract
Spermatogonial stem cells (SSCs) are the germline stem cells that are essential for the maintenance of spermatogenesis in the testis. However, it has not been sufficiently understood in amphibians, reptiles, and fish because numerous studies have been focused mainly on mammals. The aim of this review is to discuss scientific ways to elucidate SSC models of nonmammals in the context of the evolution of testicular organization since rodent SSC models. To further understand the SSC models in nonmammals, we point out common markers of an SSC pool (undifferentiated spermatogonia) in various types of testes where the kinetics of the SSC pool appears. This review includes the knowledge of (1) common molecular markers of vertebrate type A spermatogonia including putative SSC markers, (2) localization of the markers on the spermatogonia that have been reported in previous studies, (3) highlighting the most common markers in vertebrates, and (4) suggesting ways of finding SSC models in nonmammals.
Collapse
Affiliation(s)
- Hyuk Song
- Department of Stem Cell and Regenerative Technology, KIT, Konkuk University, Seoul 05029, Republic of Korea
| | - Hyun-Jung Park
- Department of Animal Biotechnology, College of Life Science and Natural Resources, Sangji University, Wonju-si 26339, Republic of Korea
| | - Won-Young Lee
- Department of Animal Science, Korea National College of Agriculture and Fisheries, Jeonju-si 54874, Republic of Korea
| | - Kyung Hoon Lee
- Department of Stem Cell and Regenerative Technology, KIT, Konkuk University, Seoul 05029, Republic of Korea
| |
Collapse
|
19
|
Amar A, Hubbard EJA, Kugler H. Modeling the C. elegans germline stem cell genetic network using automated reasoning. Biosystems 2022; 217:104672. [PMID: 35469833 PMCID: PMC9142837 DOI: 10.1016/j.biosystems.2022.104672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Revised: 03/31/2022] [Accepted: 04/04/2022] [Indexed: 11/18/2022]
Abstract
Computational methods and tools are a powerful complementary approach to experimental work for studying regulatory interactions in living cells and systems. We demonstrate the use of formal reasoning methods as applied to the Caenorhabditis elegans germ line, which is an accessible system for stem cell research. The dynamics of the underlying genetic networks and their potential regulatory interactions are key for understanding mechanisms that control cellular decision-making between stem cells and differentiation. We model the “stem cell fate” versus entry into the “meiotic development” pathway decision circuit in the young adult germ line based on an extensive study of published experimental data and known/hypothesized genetic interactions. We apply a formal reasoning framework to derive predictive networks for control of differentiation. Using this approach we simultaneously specify many possible scenarios and experiments together with potential genetic interactions, and synthesize genetic networks consistent with all encoded experimental observations. In silico analysis of knock-down and overexpression experiments within our model recapitulate published phenotypes of mutant animals and can be applied to make predictions on cellular decision-making. A methodological contribution of this work is demonstrating how to effectively model within a formal reasoning framework a complex genetic network with a wealth of known experimental data and constraints. We provide a summary of the steps we have found useful for the development and analysis of this model and can potentially be applicable to other genetic networks. This work also lays a foundation for developing realistic whole tissue models of the C. elegans germ line where each cell in the model will execute a synthesized genetic network.
Collapse
Affiliation(s)
- Ani Amar
- The Faculty of Engineering, Bar-Ilan University, Ramat Gan 5290002, Israel.
| | - E Jane Albert Hubbard
- Skirball Institute of Biomolecular Medicine, Department of Cell Biology, Department of Pathology, NYU Grossman School of Medicine, 540 First Avenue, New York, NY 10016, United States of America.
| | - Hillel Kugler
- The Faculty of Engineering, Bar-Ilan University, Ramat Gan 5290002, Israel.
| |
Collapse
|
20
|
The netrin-1 receptor UNC5C contributes to the homeostasis of undifferentiated spermatogonia in adult mice. Stem Cell Res 2022; 60:102723. [PMID: 35247845 DOI: 10.1016/j.scr.2022.102723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 02/21/2022] [Accepted: 02/23/2022] [Indexed: 11/24/2022] Open
Abstract
In adult testis, the cell mobility is essential for spermatogonia differentiation and is suspected to regulate spermatogonial stem cell fate. Netrin-1 controls cell migration and/or survival according to the cellular context. Its involvement in some self-renewing lineages raises the possibility that Netrin-1 could have a role in spermatogenesis. We show that in addition to Sertoli cells, a fraction of murine undifferentiated spermatogonia express the Netrin-1 receptor UNC5c and that UNC5c contributes to spermatogonia differentiation. Receptor loss in Unc5crcm males leads to the concomitant accumulation of transit-amplifying progenitors and short syncytia of spermatogonia. Without altering cell death rates, the consequences of Unc5c loss worsen with age: the increase in quiescent undifferentiated progenitors associated with a higher spermatogonial stem cell enriched subset leads to the spermatocyte I decline. We demonstrate in vitro that Netrin-1 promotes a guidance effect as it repulses both undifferentiated and differentiating spermatogonia. Finally, we propose that UNC5c triggers undifferentiated spermatogonia adhesion/ migration and that the repulsive activity of Netrin-1 receptors could regulate spermatogonia differentiation, and maintain germ cell homeostasis.
Collapse
|
21
|
Kitadate Y, Yoshida S. Regulation of spermatogenic stem cell homeostasis by mitogen competition in an open niche microenvironment. Gene 2022; 97:15-25. [DOI: 10.1266/ggs.21-00062] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Affiliation(s)
- Yu Kitadate
- Division of Germ Cell Biology, National Institute for Basic Biology, National Institutes of Natural Sciences
| | - Shosei Yoshida
- Division of Germ Cell Biology, National Institute for Basic Biology, National Institutes of Natural Sciences
| |
Collapse
|
22
|
Nakagawa T, Jörg DJ, Watanabe H, Mizuno S, Han S, Ikeda T, Omatsu Y, Nishimura K, Fujita M, Takahashi S, Kondoh G, Simons BD, Yoshida S, Nagasawa T. A multistate stem cell dynamics maintains homeostasis in mouse spermatogenesis. Cell Rep 2021; 37:109875. [PMID: 34686326 DOI: 10.1016/j.celrep.2021.109875] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 08/17/2021] [Accepted: 09/29/2021] [Indexed: 01/15/2023] Open
Abstract
In mouse testis, a heterogeneous population of undifferentiated spermatogonia (Aundiff) harbors spermatogenic stem cell (SSC) potential. Although GFRα1+ Aundiff maintains the self-renewing pool in homeostasis, the functional basis of heterogeneity and the implications for their dynamics remain unresolved. Here, through quantitative lineage tracing of SSC subpopulations, we show that an ensemble of heterogeneous states of SSCs supports homeostatic, persistent spermatogenesis. Such heterogeneity is maintained robustly through stochastic interconversion of SSCs between a renewal-biased Plvap+/GFRα1+ state and a differentiation-primed Sox3+/GFRα1+ state. In this framework, stem cell commitment occurs not directly but gradually through entry into licensed but uncommitted states. Further, Plvap+/GFRα1+ cells divide slowly, in synchrony with the seminiferous epithelial cycle, while Sox3+/GFRα1+ cells divide much faster. Such differential cell-cycle dynamics reduces mitotic load, and thereby the potential to acquire harmful de novo mutations of the self-renewing pool, while keeping the SSC density high over the testicular open niche.
Collapse
Affiliation(s)
- Toshinori Nakagawa
- Division of Germ Cell Biology, National Institute for Basic Biology, National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji, Okazaki 444-8787, Japan; Department of Basic Biology, School of Life Science, Graduate University for Advanced Studies (Sokendai), 5-1 Higashiyama, Myodaiji, Okazaki 444-8787, Japan; Department of Immunobiology and Hematology, Institute for Frontier Medical Sciences, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan.
| | - David J Jörg
- The Wellcome Trust/Cancer Research UK Gurdon Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, UK; Cavendish Laboratory, Department of Physics, J. J. Thomson Avenue, University of Cambridge, Cambridge CB3 0HE, UK
| | - Hitomi Watanabe
- Laboratory of Integrative Biological Science, Institute for Frontier Life and Medical Sciences, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Seiya Mizuno
- Laboratory Animal Resource Center and Trans-border Medical Research Center, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Seungmin Han
- The Wellcome Trust/Cancer Research UK Gurdon Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, UK; Wellcome Trust-Medical Research Council Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge CB2 A0W, UK
| | - Tatsuro Ikeda
- Division of Germ Cell Biology, National Institute for Basic Biology, National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji, Okazaki 444-8787, Japan
| | - Yoshiki Omatsu
- Department of Immunobiology and Hematology, Institute for Frontier Medical Sciences, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan; Laboratory of Stem Cell Biology and Developmental Immunology, Graduate School of Frontier Biosciences and Graduate School of Medicine, World Premier International Immunology Frontier Research Center, Osaka University, 1-3 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Keiko Nishimura
- Division of Germ Cell Biology, National Institute for Basic Biology, National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji, Okazaki 444-8787, Japan
| | - Miyako Fujita
- Division of Germ Cell Biology, National Institute for Basic Biology, National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji, Okazaki 444-8787, Japan
| | - Satoru Takahashi
- Laboratory Animal Resource Center and Trans-border Medical Research Center, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan; Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Gen Kondoh
- Laboratory of Integrative Biological Science, Institute for Frontier Life and Medical Sciences, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Benjamin D Simons
- The Wellcome Trust/Cancer Research UK Gurdon Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, UK; Wellcome Trust-Medical Research Council Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge CB2 A0W, UK; Department of Applied Mathematics and Theoretical Physics, Centre for Mathematical Sciences, Wilberforce Road, Cambridge CB3 0WA, UK
| | - Shosei Yoshida
- Division of Germ Cell Biology, National Institute for Basic Biology, National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji, Okazaki 444-8787, Japan; Department of Basic Biology, School of Life Science, Graduate University for Advanced Studies (Sokendai), 5-1 Higashiyama, Myodaiji, Okazaki 444-8787, Japan.
| | - Takashi Nagasawa
- Department of Immunobiology and Hematology, Institute for Frontier Medical Sciences, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan; Laboratory of Stem Cell Biology and Developmental Immunology, Graduate School of Frontier Biosciences and Graduate School of Medicine, World Premier International Immunology Frontier Research Center, Osaka University, 1-3 Yamadaoka, Suita, Osaka 565-0871, Japan.
| |
Collapse
|
23
|
Martin-Inaraja M, Ferreira M, Taelman J, Eguizabal C, Chuva De Sousa Lopes SM. Improving In Vitro Culture of Human Male Fetal Germ Cells. Cells 2021; 10:cells10082033. [PMID: 34440801 PMCID: PMC8393746 DOI: 10.3390/cells10082033] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 07/29/2021] [Accepted: 08/05/2021] [Indexed: 12/14/2022] Open
Abstract
Male human fetal germ cells (hFGCs) give rise to spermatogonial stem cells (SSCs), which are the adult precursors of the male gametes. Human SSCs are a promising (autologous) source of cells for male fertility preservation; however, in contrast to mouse SSCs, we are still unable to culture them in the long term. Here, we investigated the effect of two different culture media and four substrates (laminin, gelatin, vitronectin and matrigel) in the culture of dissociated second trimester testes, enriched for hFGCs. After 6 days in culture, we quantified the presence of POU5F1 and DDX4 expressing hFGCs. We observed a pronounced difference in hFGC number in different substrates. The combination of gelatin-coated substrate and medium containing GDNF, LIF, FGF2 and EGF resulted in the highest percentage of hFGCs (10% of the total gonadal cells) after 6 days of culture. However, the vitronectin-coated substrate resulted in a comparable percentage of hFGCs regardless of the media used (3.3% of total cells in Zhou-medium and 4.8% of total cells in Shinohara-medium). We provide evidence that not only the choices of culture medium but also choices of the adequate substrate are crucial for optimizing culture protocols for male hFGCs. Optimizing culture conditions in order to improve the expansion of hFGCs will benefit the development of gametogenesis assays in vitro.
Collapse
Affiliation(s)
- Myriam Martin-Inaraja
- Cell Therapy, Stem Cells and Tissues Group, Basque Centre for Blood Transfusion and Human Tissues, 48960 Galdakao, Spain; (M.M.-I.); (C.E.)
- Biocruces Bizkaia Health Research Institute, Cell Therapy, Stem Cells and Tissues Group, 48903 Barakaldo, Spain
| | - Monica Ferreira
- Department of Anatomy and Embryology, Leiden University Medical Centre, Einthovenweg 20, 2333 ZC Leiden, The Netherlands; (M.F.); (J.T.)
| | - Jasin Taelman
- Department of Anatomy and Embryology, Leiden University Medical Centre, Einthovenweg 20, 2333 ZC Leiden, The Netherlands; (M.F.); (J.T.)
| | - Cristina Eguizabal
- Cell Therapy, Stem Cells and Tissues Group, Basque Centre for Blood Transfusion and Human Tissues, 48960 Galdakao, Spain; (M.M.-I.); (C.E.)
- Biocruces Bizkaia Health Research Institute, Cell Therapy, Stem Cells and Tissues Group, 48903 Barakaldo, Spain
| | - Susana M. Chuva De Sousa Lopes
- Department of Anatomy and Embryology, Leiden University Medical Centre, Einthovenweg 20, 2333 ZC Leiden, The Netherlands; (M.F.); (J.T.)
- Ghent-Fertility and Stem Cell Team (G-FaST), Department for Reproductive Medicine, Ghent University Hospital, Corneel Heymanslaan 10, 9000 Ghent, Belgium
- Correspondence: ; Tel.: +31-71-526-9350
| |
Collapse
|
24
|
Tigyi G, Lin KH, Jang IH, Lee SC. Revisiting the role of lysophosphatidic acid in stem cell biology. Exp Biol Med (Maywood) 2021; 246:1802-1809. [PMID: 34038224 DOI: 10.1177/15353702211019283] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Stem cells possess unique biological characteristics such as the ability to self-renew and to undergo multilineage differentiation into specialized cells. Whereas embryonic stem cells (ESC) can differentiate into all cell types of the body, somatic stem cells (SSC) are a population of stem cells located in distinct niches throughout the body that differentiate into the specific cell types of the tissue in which they reside in. SSC function mainly to restore cells as part of normal tissue homeostasis or to replenish cells that are damaged due to injury. Cancer stem-like cells (CSC) are said to be analogous to SSC in this manner where tumor growth and progression as well as metastasis are fueled by a small population of CSC that reside within the corresponding tumor. Moreover, emerging evidence indicates that CSC are inherently resistant to chemo- and radiotherapy that are often the cause of cancer relapse. Hence, major research efforts have been directed at identifying CSC populations in different cancer types and understanding their biology. Many factors are thought to regulate and maintain cell stemness, including bioactive lysophospholipids such as lysophosphatidic acid (LPA). In this review, we discuss some of the newly discovered functions of LPA not only in the regulation of CSC but also normal SSC, the similarities in these regulatory functions, and how these discoveries can pave way to the development of novel therapies in cancer and regenerative medicine.
Collapse
Affiliation(s)
- Gábor Tigyi
- Department of Physiology, University of Tennessee Health Science Center Memphis, Memphis, TN 38163, USA
| | - Kuan-Hung Lin
- Department of Physiology, University of Tennessee Health Science Center Memphis, Memphis, TN 38163, USA
| | - Il Ho Jang
- Department of Oral Biochemistry, Pusan National University School of Dentistry, Yangsan 50612, Republic of Korea.,Dental and Life Science Institute, Pusan National University School of Dentistry, Yangsan 50612, Republic of Korea
| | - Sue Chin Lee
- Department of Physiology, University of Tennessee Health Science Center Memphis, Memphis, TN 38163, USA
| |
Collapse
|
25
|
Marca JEL, Somers WG. The Drosophila gonads: models for stem cell proliferation, self-renewal, and differentiation. AIMS GENETICS 2021. [DOI: 10.3934/genet.2014.1.55] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
AbstractThe male and female gonads of Drosophila melanogaster have developed into powerful model systems for both the study of stem cell behaviours, and for understanding how stem cell misregulation can lead to cancers. Using these systems, one is able to observe and manipulate the resident stem cell populations in vivo with a great deal of licence. The tractability of the testis and ovary also allow researchers to explore a range of cellular mechanisms, such as proliferation and polarity, as well as the influence exerted by the local environment through a host of highly-conserved signalling pathways. Importantly, many of the cellular behaviours and processes studied in the Drosophila testis and ovary are known to be disrupted, or otherwise misregulated, in human tumourigenic cells. Here, we review the mechanisms relating to stem cell behaviour, though we acknowledge there are many other fascinating aspects of gametogenesis, including the invasive behaviour of migratory border cells in the Drosophila ovary that, though relevant to the study of tumourigenesis, will unfortunately not be covered.
Collapse
Affiliation(s)
- John E. La Marca
- Department of Genetics, La Trobe University, Melbourne, VIC 3086, Australia
| | | |
Collapse
|
26
|
Nakamura Y, Jörg DJ, Kon Y, Simons BD, Yoshida S. Transient suppression of transplanted spermatogonial stem cell differentiation restores fertility in mice. Cell Stem Cell 2021; 28:1443-1456.e7. [PMID: 33848470 PMCID: PMC8351876 DOI: 10.1016/j.stem.2021.03.016] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 12/16/2020] [Accepted: 03/19/2021] [Indexed: 01/11/2023]
Abstract
A remarkable feature of tissue stem cells is their ability to regenerate the structure and function of host tissue following transplantation. However, the dynamics of donor stem cells during regeneration remains largely unknown. Here we conducted quantitative clonal fate studies of transplanted mouse spermatogonial stem cells in host seminiferous tubules. We found that, after a large population of donor spermatogonia settle in host testes, through stochastic fate choice, only a small fraction persist and regenerate over the long term, and the rest are lost through differentiation and cell death. Further, based on these insights, we showed how repopulation efficiency can be increased to a level where the fertility of infertile hosts is restored by transiently suppressing differentiation using a chemical inhibitor of retinoic acid synthesis. These findings unlock a range of potential applications of spermatogonial transplantation, from fertility restoration in individuals with cancer to conservation of biological diversity.
Collapse
Affiliation(s)
- Yoshiaki Nakamura
- Division of Germ Cell Biology, National Institute for Basic Biology, National Institutes of Natural Sciences, Higashiyama 5-1, Myodaiji, Okazaki 444-8787, Japan; Laboratory of Animal Breeding and Genetics, Graduate School of Integrated Sciences for Life, Hiroshima University, 1-4-4 Kagamiyama, Higashi-Hiroshima, Hiroshima 739-8528, Japan; Japan Society for the Promotion of Science, 5-3-1 Kojimachi, Chiyoda-ku, Tokyo 102-0083, Japan
| | - David J Jörg
- Wellcome Trust-Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge CB2 1QN, UK; Cavendish Laboratory, Department of Physics, University of Cambridge, Cambridge CB3 0HE, UK
| | - Yayoi Kon
- Division of Germ Cell Biology, National Institute for Basic Biology, National Institutes of Natural Sciences, Higashiyama 5-1, Myodaiji, Okazaki 444-8787, Japan
| | - Benjamin D Simons
- Wellcome Trust-Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge CB2 1QN, UK; Department of Applied Mathematics and Theoretical Physics, Centre for Mathematical Sciences, University of Cambridge, Wilberforce Road, Cambridge CB3 0WA, UK; Wellcome Trust-Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge CB2 1QR, UK.
| | - Shosei Yoshida
- Division of Germ Cell Biology, National Institute for Basic Biology, National Institutes of Natural Sciences, Higashiyama 5-1, Myodaiji, Okazaki 444-8787, Japan; Department of Basic Biology, School of Life Science, Graduate University for Advanced Studies (Sokendai), Okazaki 444-8787, Japan.
| |
Collapse
|
27
|
Rocchi C, Barazzuol L, Coppes RP. The evolving definition of salivary gland stem cells. NPJ Regen Med 2021; 6:4. [PMID: 33526786 PMCID: PMC7851389 DOI: 10.1038/s41536-020-00115-x] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 12/03/2020] [Indexed: 02/07/2023] Open
Abstract
Dysfunction of the salivary gland and irreversible hyposalivation are the main side effects of radiotherapy treatment for head and neck cancer leading to a drastic decrease of the quality of life of the patients. Approaches aimed at regenerating damaged salivary glands have been proposed as means to provide long-term restoration of tissue function in the affected patients. In studies to elucidate salivary gland regenerative mechanisms, more and more evidence suggests that salivary gland stem/progenitor cell behavior, like many other adult tissues, does not follow that of the hard-wired professional stem cells of the hematopoietic system. In this review, we provide evidence showing that several cell types within the salivary gland epithelium can serve as stem/progenitor-like cells. While these cell populations seem to function mostly as lineage-restricted progenitors during homeostasis, we indicate that upon damage specific plasticity mechanisms might be activated to take part in regeneration of the tissue. In light of these insights, we provide an overview of how recent developments in the adult stem cell research field are changing our thinking of the definition of salivary gland stem cells and their potential plasticity upon damage. These new perspectives may have important implications on the development of new therapeutic approaches to rescue radiation-induced hyposalivation.
Collapse
Affiliation(s)
- Cecilia Rocchi
- Department of Biomedical Sciences of Cells and Systems, University Medical Center Groningen, University of Groningen, 9713 AV, Groningen, The Netherlands. .,Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, 9700 RB, Groningen, The Netherlands. .,Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh BioQuarter, 5 Little France Drive, Edinburgh, EH16 4UU, UK.
| | - Lara Barazzuol
- Department of Biomedical Sciences of Cells and Systems, University Medical Center Groningen, University of Groningen, 9713 AV, Groningen, The Netherlands.,Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, 9700 RB, Groningen, The Netherlands
| | - Rob P Coppes
- Department of Biomedical Sciences of Cells and Systems, University Medical Center Groningen, University of Groningen, 9713 AV, Groningen, The Netherlands.,Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, 9700 RB, Groningen, The Netherlands
| |
Collapse
|
28
|
Strinkovsky L, Havkin E, Shalom-Feuerstein R, Savir Y. Spatial correlations constrain cellular lifespan and pattern formation in corneal epithelium homeostasis. eLife 2021; 10:56404. [PMID: 33433326 PMCID: PMC7803374 DOI: 10.7554/elife.56404] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 12/08/2020] [Indexed: 02/06/2023] Open
Abstract
Homeostasis in adult tissues relies on the replication dynamics of stem cells, their progenitors and the spatial balance between them. This spatial and kinetic coordination is crucial to the successful maintenance of tissue size and its replenishment with new cells. However, our understanding of the role of cellular replicative lifespan and spatial correlation between cells in shaping tissue integrity is still lacking. We developed a mathematical model for the stochastic spatial dynamics that underlie the rejuvenation of corneal epithelium. Our model takes into account different spatial correlations between cell replication and cell removal. We derive the tradeoffs between replicative lifespan, spatial correlation length, and tissue rejuvenation dynamics. We determine the conditions that allow homeostasis and are consistent with biological timescales, pattern formation, and mutants phenotypes. Our results can be extended to any cellular system in which spatial homeostasis is maintained through cell replication.
Collapse
Affiliation(s)
- Lior Strinkovsky
- Department of Physiology, Biophysics and System Biology, Faculty of Medicine, Technion, Haifa, Israel
| | - Evgeny Havkin
- Department of Physiology, Biophysics and System Biology, Faculty of Medicine, Technion, Haifa, Israel
| | - Ruby Shalom-Feuerstein
- Department of Genetics & Developmental Biology, Faculty of Medicine, Technion, Haifa, Israel.,The Rappaport Family Institute for Research in the Medical Sciences, Technion, Haifa, Israel
| | - Yonatan Savir
- Department of Physiology, Biophysics and System Biology, Faculty of Medicine, Technion, Haifa, Israel.,The Rappaport Family Institute for Research in the Medical Sciences, Technion, Haifa, Israel
| |
Collapse
|
29
|
Clonal Analysis of Gliogenesis in the Cerebral Cortex Reveals Stochastic Expansion of Glia and Cell Autonomous Responses to Egfr Dosage. Cells 2020; 9:cells9122662. [PMID: 33322301 PMCID: PMC7764668 DOI: 10.3390/cells9122662] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 12/02/2020] [Accepted: 12/07/2020] [Indexed: 02/07/2023] Open
Abstract
Development of the nervous system undergoes important transitions, including one from neurogenesis to gliogenesis which occurs late during embryonic gestation. Here we report on clonal analysis of gliogenesis in mice using Mosaic Analysis with Double Markers (MADM) with quantitative and computational methods. Results reveal that developmental gliogenesis in the cerebral cortex occurs in a fraction of earlier neurogenic clones, accelerating around E16.5, and giving rise to both astrocytes and oligodendrocytes. Moreover, MADM-based genetic deletion of the epidermal growth factor receptor (Egfr) in gliogenic clones revealed that Egfr is cell autonomously required for gliogenesis in the mouse dorsolateral cortices. A broad range in the proliferation capacity, symmetry of clones, and competitive advantage of MADM cells was evident in clones that contained one cellular lineage with double dosage of Egfr relative to their environment, while their sibling Egfr-null cells failed to generate glia. Remarkably, the total numbers of glia in MADM clones balance out regardless of significant alterations in clonal symmetries. The variability in glial clones shows stochastic patterns that we define mathematically, which are different from the deterministic patterns in neuronal clones. This study sets a foundation for studying the biological significance of stochastic and deterministic clonal principles underlying tissue development, and identifying mechanisms that differentiate between neurogenesis and gliogenesis.
Collapse
|
30
|
Yoshida S. Mouse Spermatogenesis Reflects the Unity and Diversity of Tissue Stem Cell Niche Systems. Cold Spring Harb Perspect Biol 2020; 12:cshperspect.a036186. [PMID: 32152184 DOI: 10.1101/cshperspect.a036186] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Mouse spermatogenesis is supported by spermatogenic stem cells (SSCs). SSCs maintain their pool while migrating over an open (or facultative) niche microenvironment of testicular seminiferous tubules, where ligands that support self-renewal are likely distributed widely. This contrasts with the classic picture of closed (or definitive) niches in which stem cells are gathered and the ligands are highly localized. Some of the key properties observed in the dynamics of SSCs in the testicular niche in vivo, which show the flexible and stochastic (probabilistic) fate behaviors, are found to be generic for a wide range of, if not all, tissue stem cells. SSCs also show properties characteristic of an open niche-supported system, such as high motility. Motivated by the properties of SSCs, in this review, I will reconsider the potential unity and diversity of tissue stem cell systems, with an emphasis on the varying degrees of ligand distribution and stem cell motility.
Collapse
Affiliation(s)
- Shosei Yoshida
- Division of Germ Cell Biology, National Institute for Basic Biology, National Institutes of Natural Sciences; and Department of Basic Biology, School of Life Science, SOKENDAI (Graduate University for Advanced Studies), Okazaki, Aichi 444-8787, Japan
| |
Collapse
|
31
|
Abstract
Modern single cell experiments have revealed unexpected heterogeneity in apparently functionally 'pure' cell populations. However, we are still lacking a conceptual framework to understand this heterogeneity. Here, we propose that cellular memories-changes in the molecular status of a cell in response to a stimulus, that modify the ability of the cell to respond to future stimuli-are an essential ingredient in any such theory. We illustrate this idea by considering a simple age-structured model of stem cell proliferation that takes account of mitotic memories. Using this model we argue that asynchronous mitosis generates heterogeneity that is central to stem cell population function. This model naturally explains why stem cell numbers increase through life, yet regenerative potency simultaneously declines.
Collapse
Affiliation(s)
- Patrick S Stumpf
- Joint Research Center for Computational Biomedicine, RWTH Aachen University, Aachen, 52074, Germany
| | | | | |
Collapse
|
32
|
A Hierarchy of Proliferative and Migratory Keratinocytes Maintains the Tympanic Membrane. Cell Stem Cell 2020; 28:315-330.e5. [PMID: 33181078 DOI: 10.1016/j.stem.2020.10.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 08/30/2020] [Accepted: 10/14/2020] [Indexed: 12/29/2022]
Abstract
The tympanic membrane (TM) is critical for hearing and requires continuous clearing of cellular debris, but little is known about homeostatic mechanisms in the TM epidermis. Using single-cell RNA sequencing, lineage tracing, whole-organ explant, and live-cell imaging, we show that homeostatic TM epidermis is distinct from other epidermal sites and has discrete proliferative zones with a three-dimensional hierarchy of multiple keratinocyte populations. TM stem cells reside in a discrete location of the superior TM and generate long-lived clones and committed progenitors (CPs). CP clones exhibit lateral migration, and their proliferative capacity is supported by Pdgfra+ fibroblasts, generating migratory but non-proliferative progeny. Single-cell sequencing of the human TM revealed similar cell types and transcriptional programming. Thus, during homeostasis, TM keratinocytes transit through a proliferative CP state and exhibit directional lateral migration. This work forms a foundation for understanding TM disorders and modeling keratinocyte biology.
Collapse
|
33
|
Azizi H, Niazi Tabar A, Skutella T, Govahi M. In Vitro and In Vivo Determinations of The Anti-GDNF Family Receptor Alpha 1 Antibody in Mice by Immunochemistry and RT-PCR. INTERNATIONAL JOURNAL OF FERTILITY & STERILITY 2020; 14:228-233. [PMID: 33098391 PMCID: PMC7604702 DOI: 10.22074/ijfs.2020.6051] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 01/11/2020] [Indexed: 12/31/2022]
Abstract
Background The glial cell-derived neurotrophic factor (GDNF) family plays essential roles in the maintenance, growth, regulatory and signalling pathways of spermatogonial stem cells (SSCs). In this study, we analysed the expression of anti-GDNF family receptor alpha 1 antibody (GFRa1) by immunohistochemistry (IHC), immunocytochemistry (ICC), Fluidigm real-time polymerase chain reaction (RT-PCR) and flow cytometry analyses. Materials and Methods In this experiment study, ICC, IHC, Fluidigm RT-PCR and flow cytometry were used to analyse the expression of the germ cell marker GFRa1 in testis tissue and SSC culture. Results IHC analysis showed that there were two groups of GFRa1 positive cells in the seminiferous tubules based on their location and expression shape - a small round punctuated shape on the basal compartment donut shape and a C-shaped expression located between the basal and the luminal compartments of the seminiferous tubules. OCT4 and PLZF positive cells may have similar patterns of expression as the first group. Assessment of the seminiferous tubule sections demonstrated that about 27% of the SSCs were positive for GFRa1. Fluidigm RT-PCR confirmed the significant expression (P<0.001) of GFRa1 in the SSCs compared to testicular stromal cells (TSCs). Flow cytometry analysis demonstrated that about 75% of the isolated SSCs colonies were positive for GFRa1. Conclusion The results indicated that GFRa1 had a specific expression pattern both in vivo and in vitro. This finding could be helpful for understanding the proliferation, maintenance and signalling pathways of SSCs, and differentiation of meiotic and haploid germ cells.
Collapse
Affiliation(s)
- Hossein Azizi
- Faculty of Biotechnology, Amol University of Special Modern Technologies, Amol, Iran. Electronic Address:
| | - Amirreza Niazi Tabar
- Faculty of Biotechnology, Amol University of Special Modern Technologies, Amol, Iran
| | - Thomas Skutella
- Institute for Anatomy and Cell Biology, Medical Faculty, University of Heidelberg, Heidelberg, Germany
| | - Mostafa Govahi
- Faculty of Biotechnology, Amol University of Special Modern Technologies, Amol, Iran
| |
Collapse
|
34
|
Zhou S, Feng S, Qin W, Wang X, Tang Y, Yuan S. Epigenetic Regulation of Spermatogonial Stem Cell Homeostasis: From DNA Methylation to Histone Modification. Stem Cell Rev Rep 2020; 17:562-580. [PMID: 32939648 DOI: 10.1007/s12015-020-10044-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/11/2020] [Indexed: 12/27/2022]
Abstract
Spermatogonial stem cells(SSCs)are the ultimate germline stem cells with the potential of self-renewal and differentiation, and a dynamic balance of SSCs play an essential role in spermatogenesis. During the gene expression process, genomic DNA and nuclear protein, working together, contribute to SSC homeostasis. Recently, emerging studies have shown that epigenome-related molecules such as chromatin modifiers play an important role in SSC homeostasis through regulating target gene expression. Here, we focus on two types of epigenetic events, including DNA methylation and histone modification, and summarize their function in SSC homeostasis. Understanding the molecular mechanism during SSC homeostasis will promote the recognition of epigenetic biomarkers in male infertility, and bring light into therapies of infertile patients.Graphical Abstract.
Collapse
Affiliation(s)
- Shumin Zhou
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, China
| | - Shenglei Feng
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, China
| | - Weibing Qin
- NHC Key Laboratory of Male Reproduction and Genetics, Family Planning Research Institute of Guangdong Province, 510500, Guangzhou, China
| | - Xiaoli Wang
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, China
| | - Yunge Tang
- NHC Key Laboratory of Male Reproduction and Genetics, Family Planning Research Institute of Guangdong Province, 510500, Guangzhou, China.
| | - Shuiqiao Yuan
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, China. .,Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen, 518057, China.
| |
Collapse
|
35
|
Parekh PA, Garcia TX, Hofmann MC. Regulation of GDNF expression in Sertoli cells. Reproduction 2020; 157:R95-R107. [PMID: 30620720 DOI: 10.1530/rep-18-0239] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 01/08/2019] [Indexed: 12/15/2022]
Abstract
Sertoli cells regulate male germ cell proliferation and differentiation and are a critical component of the spermatogonial stem cell (SSC) niche, where homeostasis is maintained by the interplay of several signaling pathways and growth factors. These factors are secreted by Sertoli cells located within the seminiferous epithelium, and by interstitial cells residing between the seminiferous tubules. Sertoli cells and peritubular myoid cells produce glial cell line-derived neurotrophic factor (GDNF), which binds to the RET/GFRA1 receptor complex at the surface of undifferentiated spermatogonia. GDNF is known for its ability to drive SSC self-renewal and proliferation of their direct cell progeny. Even though the effects of GDNF are well studied, our understanding of the regulation its expression is still limited. The purpose of this review is to discuss how GDNF expression in Sertoli cells is modulated within the niche, and how these mechanisms impact germ cell homeostasis.
Collapse
Affiliation(s)
- Parag A Parekh
- Department of Endocrine Neoplasia, UT MD Anderson Cancer Center, Houston, Texas, USA
| | - Thomas X Garcia
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas, USA.,Department of Biological and Environmental Sciences, University of Houston-Clear Lake, Houston, Texas, USA
| | - Marie-Claude Hofmann
- Department of Endocrine Neoplasia, UT MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
36
|
Defining Adult Stem Cell Function at Its Simplest: The Ability to Replace Lost Cells through Mitosis. Cell Stem Cell 2020; 25:174-183. [PMID: 31374197 DOI: 10.1016/j.stem.2019.07.002] [Citation(s) in RCA: 105] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Classic studies on hematopoiesis indicate that blood cell numbers are maintained by rare, hard-wired, transplantable stem cells (SCs). Subsequent studies in other organs have implicitly assumed that all SC hierarchies follow the design of the hematopoietic system. Lineage tracing techniques have revolutionized the study of solid tissue SCs. It thus appears that key characteristics of the hematopoietic SC hierarchy (rarity of SCs, specific marker expression, quiescence, asymmetric division, and unidirectional differentiation) are not generalizable to other tissues. In light of these insights, we offer a revised, generalizable definition of SC function: the ability to replace lost tissue through cell division.
Collapse
|
37
|
Ledesma-Terrón M, Peralta-Cañadas N, Míguez DG. FGF2 modulates simultaneously the mode, the rate of division and the growth fraction in cultures of radial glia. Development 2020; 147:147/14/dev189712. [PMID: 32709691 PMCID: PMC7390635 DOI: 10.1242/dev.189712] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 06/18/2020] [Indexed: 01/16/2023]
Abstract
Radial glial progenitors in the mammalian developing neocortex have been shown to follow a deterministic differentiation program restricted to an asymmetric-only mode of division. This feature seems incompatible with their well-known ability to increase in number when cultured in vitro, driven by fibroblast growth factor 2 and other mitogenic signals. The changes in their differentiation dynamics that allow this transition from in vivo asymmetric-only division mode to an in vitro self-renewing culture have not been fully characterized. Here, we combine experiments of radial glia cultures with numerical models and a branching process theoretical formalism to show that fibroblast growth factor 2 has a triple effect by simultaneously increasing the growth fraction, promoting symmetric divisions and shortening the length of the cell cycle. These combined effects partner to establish and sustain a pool of rapidly proliferating radial glial progenitors in vitro. We also show that, in conditions of variable proliferation dynamics, the branching process tool outperforms other commonly used methods based on thymidine analogs, such as BrdU and EdU, in terms of accuracy and reliability. Highlighted Article: When mode and/or rate of division are changing, a branching process, rather than a thymidine analog method, provides temporal resolution, it is more robust and does not interfere with cell homeostasis.
Collapse
Affiliation(s)
- Mario Ledesma-Terrón
- Departamento de Física de la Materia Condensada, Instituto de Física de la Materia Condensada, IFIMAC, Instituto Nicolas Cabrera, INC, Centro de Biología Molecular Severo Ochoa, CBMSO, Universidad Autónoma de Madrid, Madrid 28012, Spain
| | - Nuria Peralta-Cañadas
- Departamento de Física de la Materia Condensada, Instituto de Física de la Materia Condensada, IFIMAC, Instituto Nicolas Cabrera, INC, Centro de Biología Molecular Severo Ochoa, CBMSO, Universidad Autónoma de Madrid, Madrid 28012, Spain
| | - David G Míguez
- Departamento de Física de la Materia Condensada, Instituto de Física de la Materia Condensada, IFIMAC, Instituto Nicolas Cabrera, INC, Centro de Biología Molecular Severo Ochoa, CBMSO, Universidad Autónoma de Madrid, Madrid 28012, Spain
| |
Collapse
|
38
|
Parigini C, Greulich P. Universality of clonal dynamics poses fundamental limits to identify stem cell self-renewal strategies. eLife 2020; 9:56532. [PMID: 32687057 PMCID: PMC7444910 DOI: 10.7554/elife.56532] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 07/03/2020] [Indexed: 12/21/2022] Open
Abstract
How adult stem cells maintain self-renewing tissues is commonly assessed by analysing clonal data from in vivo cell lineage-tracing assays. To identify strategies of stem cell self-renewal requires that different models of stem cell fate choice predict sufficiently different clonal statistics. Here, we show that models of cell fate choice can, in homeostatic tissues, be categorized by exactly two 'universality classes', whereby models of the same class predict, under asymptotic conditions, the same clonal statistics. Those classes relate to generalizations of the canonical asymmetric vs. symmetric stem cell self-renewal strategies and are distinguished by a conservation law. This poses both challenges and opportunities to identify stem cell self-renewal strategies: while under asymptotic conditions, self-renewal models of the same universality class cannot be distinguished by clonal data only, models of different classes can be distinguished by simple means.
Collapse
Affiliation(s)
- Cristina Parigini
- School of Mathematical Science, University of Southampton, Southampton, United Kingdom.,Institute for Life Sciences, University of Southampton, Southampton, United Kingdom
| | - Philip Greulich
- School of Mathematical Science, University of Southampton, Southampton, United Kingdom.,Institute for Life Sciences, University of Southampton, Southampton, United Kingdom
| |
Collapse
|
39
|
Abstract
The mechanisms that regulate the balance between stem cell duplication and differentiation in adult tissues remain in debate. Using a combination of genetic lineage tracing and marker-based assays, the quantitative statistical analysis of clone size and cell composition has provided insights into the patterns of stem cell fate across a variety of tissue types and organisms. These studies have emphasized the role of niche factors and environmental cues in promoting stem cell competence, fate priming, and stochastic renewal programs. At the same time, evidence for injury-induced "cellular reprogramming" has revealed the remarkable flexibility of cell states, allowing progenitors to reacquire self-renewal potential during regeneration. Together, these findings have questioned the nature of stem cell identity and function. Here, focusing on a range of canonical tissue types, we review how quantitative modeling-based approaches have uncovered conserved patterns of stem cell fate and provided new insights into the mechanisms that regulate self-renewal.
Collapse
Affiliation(s)
- Lemonia Chatzeli
- Wellcome Trust/CRUK Gurdon Institute, University of Cambridge, Cambridge CB2 1QN, United Kingdom
- Wellcome Trust/MRC Stem Cell Institute, University of Cambridge, Cambridge CB2 1QR, United Kingdom
| | - Benjamin D Simons
- Wellcome Trust/CRUK Gurdon Institute, University of Cambridge, Cambridge CB2 1QN, United Kingdom
- Wellcome Trust/MRC Stem Cell Institute, University of Cambridge, Cambridge CB2 1QR, United Kingdom
- Department of Applied Mathematics and Theoretical Physics, Centre for Mathematical Sciences, University of Cambridge, Cambridge CB3 0WA, United Kingdom
| |
Collapse
|
40
|
McKinley KL, Castillo-Azofeifa D, Klein OD. Tools and Concepts for Interrogating and Defining Cellular Identity. Cell Stem Cell 2020; 26:632-656. [PMID: 32386555 PMCID: PMC7250495 DOI: 10.1016/j.stem.2020.03.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Defining the mechanisms that generate specialized cell types and coordinate their functions is critical for understanding organ development and renewal. New tools and discoveries are challenging and refining our definitions of a cell type. A rapidly growing toolkit for single-cell analyses has expanded the number of markers that can be assigned to a cell simultaneously, revealing heterogeneity within cell types that were previously regarded as homogeneous populations. Additionally, cell types defined by specific molecular markers can exhibit distinct, context-dependent functions; for example, between tissues in homeostasis and those responding to damage. Here we review the current technologies used to identify and characterize cells, and we discuss how experimental and pathological perturbations are adding increasing complexity to our definitions of cell identity.
Collapse
Affiliation(s)
- Kara L McKinley
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA
| | - David Castillo-Azofeifa
- Department of Orofacial Sciences, University of California, San Francisco, San Francisco, CA, USA; Program in Craniofacial Biology, University of California, San Francisco, San Francisco, CA, USA
| | - Ophir D Klein
- Department of Orofacial Sciences, University of California, San Francisco, San Francisco, CA, USA; Program in Craniofacial Biology, University of California, San Francisco, San Francisco, CA, USA; Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA; Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
41
|
Wang Z, Xu X, Li JL, Palmer C, Maric D, Dean J. Sertoli cell-only phenotype and scRNA-seq define PRAMEF12 as a factor essential for spermatogenesis in mice. Nat Commun 2019; 10:5196. [PMID: 31729367 PMCID: PMC6858368 DOI: 10.1038/s41467-019-13193-3] [Citation(s) in RCA: 35] [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: 08/27/2018] [Accepted: 10/22/2019] [Indexed: 01/14/2023] Open
Abstract
Spermatogonial stem cells (SSCs) have the dual capacity to self-renew and differentiate into progenitor spermatogonia that develop into mature spermatozoa. Here, we document that preferentially expressed antigen of melanoma family member 12 (PRAMEF12) plays a key role in maintenance of the spermatogenic lineage. In male mice, genetic ablation of Pramef12 arrests spermatogenesis and results in sterility which can be rescued by transgenic expression of Pramef12. Pramef12 deficiency globally decreases expression of spermatogenic-related genes, and single-cell transcriptional analysis of post-natal male germline cells identifies four spermatogonial states. In the absence of Pramef12 expression, there are fewer spermatogonial stem cells which exhibit lower expression of SSC maintenance-related genes and are defective in their ability to differentiate. The disruption of the first wave of spermatogenesis in juvenile mice results in agametic seminiferous tubules. These observations mimic a Sertoli cell-only syndrome in humans and may have translational implications for reproductive medicine.
Collapse
Affiliation(s)
- Zhengpin Wang
- Laboratory of Cellular and Developmental Biology, NIDDK, National Institutes of Health, Bethesda, MD, 20892, USA.
| | - Xiaojiang Xu
- Integrative Bioinformatics, NIEHS, National Institutes of Health, Research Triangle Park, NC, 27709, USA
| | - Jian-Liang Li
- Integrative Bioinformatics, NIEHS, National Institutes of Health, Research Triangle Park, NC, 27709, USA
| | - Cameron Palmer
- Laboratory of Cellular and Developmental Biology, NIDDK, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Dragan Maric
- NINDS Flow Cytometry Core Facility, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Jurrien Dean
- Laboratory of Cellular and Developmental Biology, NIDDK, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
42
|
Shi Z, Enayatullah H, Lv Z, Dai H, Wei Q, Shen L, Karwand B, Shi F. Freeze-Dried Royal Jelly Proteins Enhanced the Testicular Development and Spermatogenesis in Pubescent Male Mice. Animals (Basel) 2019; 9:ani9110977. [PMID: 31731648 PMCID: PMC6912309 DOI: 10.3390/ani9110977] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Revised: 11/07/2019] [Accepted: 11/07/2019] [Indexed: 12/20/2022] Open
Abstract
Simple Summary Spermatogenesis and hormones secretions are serious life-threating and complicated process, which can be improve through science-based approaches. Royal jelly is a thick white milky fluid secreted by the hypopharyngeal and mandibular glands of young nurse worker bees (Apis mellifera) and used to feed their queen to expand their life. The results of the study revealed that, the growth performance of testis in exposed mice to freeze-dried Royal Jelly for 35 consecutive days were significantly enhanced in moderate dose among other treated doses. However, at Post Natal Days (PNDs 14 and PNDs 21), obviously changes were observed in histological examination of the testis while at PNDs-07 no major changes were observed. The Tunnel assay showed that, less apoptotic cells were detected in the testis of mice in high dose of freeze-dried RJ and oral administration of freeze-dried royal jelly can aggravate adverse effects via tempestuous on sexual hormone secretion at both PNDs 21 and PNDs 35 respectively. Abstract Spermatogenesis and hormones secretions are crucial endocrine and physiological process for maintaining the life. Royal Jelly (RJ) bioactive components are Major Royal Jelly Proteins (MRJPs), owing exceptional biological properties. However, the effects of RJ on pup’s testicular development during neonatal and pubertal period exposure hasn’t been adequately studied. The aim of the study was to detect neonatal sexual hormones concentration and histopathological changes on testicular development of the male progeny after oral exposure to freeze-dried RJ for 35 consecutive days. After mice give birth, male pups were collected together, separated by sex, and randomly standardized to seven (7) male pups per dam. Male pups were assigned to control diet (CON group), low dose RJ (L-RJ group) diet (control diet + 125 mg/kg/day RJ), moderate dose RJ (M-RJ group) diet (control diet + 250 mg/kg/day RJ) and high dose of RJ (H-RJ group) diet (control diet + 500 mg/kg/day RJ). After weaning, male pups were continuously fed with freeze-dried RJ until the age of PNDs 35. The results revealed that, oral M-RJ (250 mg/kg/day) administration significantly (p < 0.05) increased the testis weight, the diameter of seminiferous tubule and the height of seminiferous epithelium of offspring mice at PNDs 14. However, high-dose RJ (500 mg/kg/day) decreased the diameter of seminiferous tubule but increased the height of seminiferous epithelium of male offspring (p < 0.05) at the same time point. Furthermore, oral M-RJ treatment significantly (p < 0.05) increased the testis weight and spermatogenesis at PNDs 21. Whereas, oral H-RJ treatment significantly (p < 0.05) reduced the diameter of seminiferous tubule and the height of seminiferous epithelium at PNDs 21. At PNDs 35, oral M-RJ treatment increased the testis weight, the diameter of seminiferous tubule and the level of FSH. While, high-dose of RJ reduced testis weight and size (diameter of seminiferous tubule and height of seminiferous epithelium), ratio of apoptotic germ cells and incomplete spermatogenesis collectively. In addition, sexual hormone secretions (FSH, LH, E2) were decreased after RJs treatment (L-RJ, M-RJ, H-RJ) at PNDs 21 respectively. In conclusion, the results concluded that oral administration of low and moderate doses of RJ could enhance the development of testis at neonate period until pubescent, whereas unfavorable adverse effects induced by high dose of RJ might remain.
Collapse
Affiliation(s)
- Zhicheng Shi
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (Z.S.); (H.E.); (Z.L.); (H.D.); (Q.W.)
| | - Hamdard Enayatullah
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (Z.S.); (H.E.); (Z.L.); (H.D.); (Q.W.)
| | - Zengpeng Lv
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (Z.S.); (H.E.); (Z.L.); (H.D.); (Q.W.)
| | - Hongjian Dai
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (Z.S.); (H.E.); (Z.L.); (H.D.); (Q.W.)
| | - Quanwei Wei
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (Z.S.); (H.E.); (Z.L.); (H.D.); (Q.W.)
| | - Lirong Shen
- College of Food Science, Zhejiang University, Hangzhou 310058, China;
| | - Babrak Karwand
- Faculty of Veterinary Science, Kunduz University, Kunduz City 0093, Afghanistan;
| | - Fangxiong Shi
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (Z.S.); (H.E.); (Z.L.); (H.D.); (Q.W.)
- Correspondence: ; Tel./Fax: +86-25-84399112
| |
Collapse
|
43
|
Singh P, Patel RK, Palmer N, Grenier JK, Paduch D, Kaldis P, Grimson A, Schimenti JC. CDK2 kinase activity is a regulator of male germ cell fate. Development 2019; 146:dev180273. [PMID: 31582414 PMCID: PMC6857589 DOI: 10.1242/dev.180273] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 09/21/2019] [Indexed: 12/27/2022]
Abstract
The ability of men to remain fertile throughout their lives depends upon establishment of a spermatogonial stem cell (SSC) pool from gonocyte progenitors, and thereafter balancing SSC renewal versus terminal differentiation. Here, we report that precise regulation of the cell cycle is crucial for this balance. Whereas cyclin-dependent kinase 2 (Cdk2) is not necessary for mouse viability or gametogenesis stages prior to meiotic prophase I, mice bearing a deregulated allele (Cdk2Y15S ) are severely deficient in spermatogonial differentiation. This allele disrupts an inhibitory phosphorylation site (Tyr15) for the kinase WEE1. Remarkably, Cdk2Y15S/Y15S mice possess abnormal clusters of mitotically active SSC-like cells, but these are eventually removed by apoptosis after failing to differentiate properly. Analyses of lineage markers, germ cell proliferation over time, and single cell RNA-seq data revealed delayed and defective differentiation of gonocytes into SSCs. Biochemical and genetic data demonstrated that Cdk2Y15S is a gain-of-function allele causing elevated kinase activity, which underlies these differentiation defects. Our results demonstrate that precise regulation of CDK2 kinase activity in male germ cell development is crucial for the gonocyte-to-spermatogonia transition and long-term spermatogenic homeostasis.
Collapse
Affiliation(s)
- Priti Singh
- Cornell University, College of Veterinary Medicine, Department of Biomedical Sciences, Ithaca, NY 14853, USA
| | - Ravi K Patel
- Cornell University, Department of Molecular Biology and Genetics, Ithaca, NY 14853, USA
| | - Nathan Palmer
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology, and Research (A*STAR), Singapore 138673
- Department of Biochemistry, National University of Singapore, Singapore 117599, Republic of Singapore
| | - Jennifer K Grenier
- Cornell University, College of Veterinary Medicine, Department of Biomedical Sciences, Ithaca, NY 14853, USA
| | - Darius Paduch
- Cornell University, Weill Cornell Medicine, Department of Urology, New York, NY 10065, USA
| | - Philipp Kaldis
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology, and Research (A*STAR), Singapore 138673
- Department of Biochemistry, National University of Singapore, Singapore 117599, Republic of Singapore
| | - Andrew Grimson
- Cornell University, Department of Molecular Biology and Genetics, Ithaca, NY 14853, USA
| | - John C Schimenti
- Cornell University, College of Veterinary Medicine, Department of Biomedical Sciences, Ithaca, NY 14853, USA
| |
Collapse
|
44
|
La HM, Hobbs RM. Mechanisms regulating mammalian spermatogenesis and fertility recovery following germ cell depletion. Cell Mol Life Sci 2019; 76:4071-4102. [PMID: 31254043 PMCID: PMC11105665 DOI: 10.1007/s00018-019-03201-6] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 06/07/2019] [Accepted: 06/19/2019] [Indexed: 12/19/2022]
Abstract
Mammalian spermatogenesis is a highly complex multi-step process sustained by a population of mitotic germ cells with self-renewal potential known as spermatogonial stem cells (SSCs). The maintenance and regulation of SSC function are strictly dependent on a supportive niche that is composed of multiple cell types. A detailed appreciation of the molecular mechanisms underpinning SSC activity and fate is of fundamental importance for spermatogenesis and male fertility. However, different models of SSC identity and spermatogonial hierarchy have been proposed and recent studies indicate that cell populations supporting steady-state germline maintenance and regeneration following damage are distinct. Importantly, dynamic changes in niche properties may underlie the fate plasticity of spermatogonia evident during testis regeneration. While formation of spermatogenic colonies in germ-cell-depleted testis upon transplantation is a standard assay for SSCs, differentiation-primed spermatogonial fractions have transplantation potential and this assay provides readout of regenerative rather than steady-state stem cell capacity. The characterisation of spermatogonial populations with regenerative capacity is essential for the development of clinical applications aimed at restoring fertility in individuals following germline depletion by genotoxic treatments. This review will discuss regulatory mechanisms of SSCs in homeostatic and regenerative testis and the conservation of these mechanisms between rodent models and man.
Collapse
Affiliation(s)
- Hue M La
- Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, 3800, Australia
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Melbourne, VIC, 3800, Australia
| | - Robin M Hobbs
- Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, 3800, Australia.
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Melbourne, VIC, 3800, Australia.
| |
Collapse
|
45
|
Ageing affects DNA methylation drift and transcriptional cell-to-cell variability in mouse muscle stem cells. Nat Commun 2019; 10:4361. [PMID: 31554804 PMCID: PMC6761124 DOI: 10.1038/s41467-019-12293-4] [Citation(s) in RCA: 135] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 09/03/2019] [Indexed: 12/29/2022] Open
Abstract
Age-related tissue alterations have been associated with a decline in stem cell number and function. Although increased cell-to-cell variability in transcription or epigenetic marks has been proposed to be a major hallmark of ageing, little is known about the molecular diversity of stem cells during ageing. Here we present a single cell multi-omics study of mouse muscle stem cells, combining single-cell transcriptome and DNA methylome profiling. Aged cells show a global increase of uncoordinated transcriptional heterogeneity biased towards genes regulating cell-niche interactions. We find context-dependent alterations of DNA methylation in aged stem cells. Importantly, promoters with increased methylation heterogeneity are associated with increased transcriptional heterogeneity of the genes they drive. These results indicate that epigenetic drift, by accumulation of stochastic DNA methylation changes in promoters, is associated with the degradation of coherent transcriptional networks during stem cell ageing. Furthermore, our observations also shed light on the mechanisms underlying the DNA methylation clock. Age-related tissue alterations have been associated with a decline in stem cell number and function. Here the authors report a single cell multi-omics study of mouse muscle stem cells, combining single cell transcriptome and DNA methylome profiling and find that aged cells have a global increase of uncoordinated transcriptional heterogeneity biased towards genes regulating cell-niche interactions.
Collapse
|
46
|
Sharir A, Marangoni P, Zilionis R, Wan M, Wald T, Hu JK, Kawaguchi K, Castillo-Azofeifa D, Epstein L, Harrington K, Pagella P, Mitsiadis T, Siebel CW, Klein AM, Klein OD. A large pool of actively cycling progenitors orchestrates self-renewal and injury repair of an ectodermal appendage. Nat Cell Biol 2019; 21:1102-1112. [PMID: 31481792 PMCID: PMC6935352 DOI: 10.1038/s41556-019-0378-2] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 07/23/2019] [Indexed: 02/06/2023]
Abstract
The classical model of tissue renewal posits that small numbers of quiescent stem cells (SCs) give rise to proliferating transit-amplifying cells before terminal differentiation. However, many organs house pools of SCs with proliferative and differentiation potentials that diverge from this template. Resolving SC identity and organization is therefore central to understanding tissue renewal. Here, using a combination of single-cell RNA sequencing (scRNA-seq), mouse genetics and tissue injury approaches, we uncover cellular hierarchies and mechanisms that underlie the maintenance and repair of the continuously growing mouse incisor. Our results reveal that, during homeostasis, a group of actively cycling epithelial progenitors generates enamel-producing ameloblasts and adjacent layers of non-ameloblast cells. After injury, tissue repair was achieved through transient increases in progenitor-cell proliferation and through direct conversion of Notch1-expressing cells to ameloblasts. We elucidate epithelial SC identity, position and function, providing a mechanistic basis for the homeostasis and repair of a fast-turnover ectodermal appendage.
Collapse
Affiliation(s)
- Amnon Sharir
- Program in Craniofacial Biology and Department of Orofacial Sciences, University of California, San Francisco, CA, USA
| | - Pauline Marangoni
- Program in Craniofacial Biology and Department of Orofacial Sciences, University of California, San Francisco, CA, USA
| | - Rapolas Zilionis
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
- Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Mian Wan
- Program in Craniofacial Biology and Department of Orofacial Sciences, University of California, San Francisco, CA, USA
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases and Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Tomas Wald
- Program in Craniofacial Biology and Department of Orofacial Sciences, University of California, San Francisco, CA, USA
| | - Jimmy K Hu
- Program in Craniofacial Biology and Department of Orofacial Sciences, University of California, San Francisco, CA, USA
| | - Kyogo Kawaguchi
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
- Universal Biology Institute, The University of Tokyo, Tokyo, Japan
| | - David Castillo-Azofeifa
- Program in Craniofacial Biology and Department of Orofacial Sciences, University of California, San Francisco, CA, USA
| | - Leo Epstein
- Bioinformatics and Computational Biology Program, University of Idaho, Moscow, ID, USA
| | - Kyle Harrington
- Bioinformatics and Computational Biology Program, University of Idaho, Moscow, ID, USA
- Virtual Technology and Design, University of Idaho, Moscow, ID, USA
| | - Pierfrancesco Pagella
- Orofacial Development and Regeneration, Institute of Oral Biology, Centre of Dental Medicine, University of Zurich, Zurich, Switzerland
| | - Thimios Mitsiadis
- Orofacial Development and Regeneration, Institute of Oral Biology, Centre of Dental Medicine, University of Zurich, Zurich, Switzerland
| | - Christian W Siebel
- Department of Discovery Oncology, Genentech, South San Francisco, CA, USA
| | - Allon M Klein
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA.
| | - Ophir D Klein
- Program in Craniofacial Biology and Department of Orofacial Sciences, University of California, San Francisco, CA, USA.
- Department of Pediatrics and Institute for Human Genetics, University of California, San Francisco, CA, USA.
| |
Collapse
|
47
|
Jilkine A. Mathematical Models of Stem Cell Differentiation and Dedifferentiation. CURRENT STEM CELL REPORTS 2019. [DOI: 10.1007/s40778-019-00156-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
48
|
Sharma S, Wistuba J, Pock T, Schlatt S, Neuhaus N. Spermatogonial stem cells: updates from specification to clinical relevance. Hum Reprod Update 2019; 25:275-297. [DOI: 10.1093/humupd/dmz006] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 11/23/2018] [Accepted: 02/22/2019] [Indexed: 12/20/2022] Open
Affiliation(s)
- Swati Sharma
- Centre of Reproductive Medicine and Andrology, Institute of Reproductive and Regenerative Biology, Albert-Schweitzer Campus 1, Building D11, Münster, Germany
| | - Joachim Wistuba
- Centre of Reproductive Medicine and Andrology, Institute of Reproductive and Regenerative Biology, Albert-Schweitzer Campus 1, Building D11, Münster, Germany
| | - Tim Pock
- Centre of Reproductive Medicine and Andrology, Institute of Reproductive and Regenerative Biology, Albert-Schweitzer Campus 1, Building D11, Münster, Germany
| | - Stefan Schlatt
- Centre of Reproductive Medicine and Andrology, Institute of Reproductive and Regenerative Biology, Albert-Schweitzer Campus 1, Building D11, Münster, Germany
| | - Nina Neuhaus
- Centre of Reproductive Medicine and Andrology, Institute of Reproductive and Regenerative Biology, Albert-Schweitzer Campus 1, Building D11, Münster, Germany
| |
Collapse
|
49
|
Abstract
Studying cell fate dynamics is complicated by the fact that direct in vivo observation of individual cell fate outcomes is usually not possible and only multicellular data of cell clones can be obtained. In this situation, experimental data alone is not sufficient to validate biological models because the hypotheses and the data cannot be directly compared and thus standard statistical tests cannot be leveraged. On the other hand, mathematical modelling can bridge the scales between a hypothesis and measured data via quantitative predictions from a mathematical model. Here, we describe how to implement the rules behind a hypothesis (cell fate outcomes) one-to-one as a stochastic model, how to evaluate such a rule-based model mathematically via analytical calculation or stochastic simulations of the model's Master equation, and to predict the outcomes of clonal statistics for respective hypotheses. We also illustrate two approaches to compare these predictions directly with the clonal data to assess the models.
Collapse
Affiliation(s)
- Philip Greulich
- Mathematical Sciences, University of Southampton, Southampton, UK.
- Institute for Life Sciences, University of Southampton, Southampton, UK.
| |
Collapse
|
50
|
|