Full Text:   <2098>

CLC number: Q28

On-line Access: 2019-05-15

Received: 2019-04-13

Revision Accepted: 2019-04-24

Crosschecked: 2019-05-09

Cited: 0

Clicked: 3406

Citations:  Bibtex RefMan EndNote GB/T7714

-   Go to

Article info.
Open peer comments

Journal of Zhejiang University SCIENCE B 2019 Vol.20 No.6 P.467-475

http://doi.org/10.1631/jzus.B1900197


Novel insights into cell cycle regulation of cell fate determination


Author(s):  Su-Wei Gao, Feng Liu

Affiliation(s):  State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; more

Corresponding email(s):   liuf@ioz.ac.cn

Key Words:  Cell cycle, Cell fate, Heterogeneity, Fate conversion, Stem/progenitor cell


Su-Wei Gao, Feng Liu. Novel insights into cell cycle regulation of cell fate determination[J]. Journal of Zhejiang University Science B, 2019, 20(6): 467-475.

@article{title="Novel insights into cell cycle regulation of cell fate determination",
author="Su-Wei Gao, Feng Liu",
journal="Journal of Zhejiang University Science B",
volume="20",
number="6",
pages="467-475",
year="2019",
publisher="Zhejiang University Press & Springer",
doi="10.1631/jzus.B1900197"
}

%0 Journal Article
%T Novel insights into cell cycle regulation of cell fate determination
%A Su-Wei Gao
%A Feng Liu
%J Journal of Zhejiang University SCIENCE B
%V 20
%N 6
%P 467-475
%@ 1673-1581
%D 2019
%I Zhejiang University Press & Springer
%DOI 10.1631/jzus.B1900197

TY - JOUR
T1 - Novel insights into cell cycle regulation of cell fate determination
A1 - Su-Wei Gao
A1 - Feng Liu
J0 - Journal of Zhejiang University Science B
VL - 20
IS - 6
SP - 467
EP - 475
%@ 1673-1581
Y1 - 2019
PB - Zhejiang University Press & Springer
ER -
DOI - 10.1631/jzus.B1900197


Abstract: 
The stem/progenitor cell has long been regarded as a central cell type in development, homeostasis, and regeneration, largely owing to its robust self-renewal and multilineage differentiation abilities. The balance between self-renewal and stem/progenitor cell differentiation requires the coordinated regulation of cell cycle progression and cell fate determination. Extensive studies have demonstrated that cell cycle states determine cell fates, because cells in different cell cycle states are characterized by distinct molecular features and functional outputs. Recent advances in high-resolution epigenome profiling, single-cell transcriptomics, and cell cycle reporter systems have provided novel insights into the cell cycle regulation of cell fate determination. Here, we review recent advances in cell cycle-dependent cell fate determination and functional heterogeneity, and the application of cell cycle manipulation for cell fate conversion. These findings will provide insight into our understanding of cell cycle regulation of cell fate determination in this field, and may facilitate its potential application in translational medicine.

细胞周期调控细胞命运决定的新见解

概要:长期以来,干/祖细胞由于其具备的自我更新和多谱系分化能力,因而被视为生物体发育、稳态和再生过程中的一类重要细胞类型.干/祖细胞自我更新和多谱系分化之间的平衡是由细胞周期进程和细胞命运决定之间的协同调控来完成的.大量研究表明细胞周期状态可以决定细胞的命运,体现在处于不同细胞周期状态的细胞具有不同的分子特征和功能.目前,随着高分辨率的表观基因组学、单细胞转录组学和细胞周期实时标记系统的开发,我们对细胞周期如何调控细胞命运有了新的认识.本文总结了细胞周期调控细胞命运决定和功能异质性的分子机制,以及通过操纵细胞周期进而影响细胞命运转变的研究进展.这些发现将加深我们对细胞周期调控细胞命运决定机制的理解,同时也能促进其在转化医学中的潜在应用.
关键词:细胞周期;细胞命运;异质性;命运转变;干/祖细胞

Darkslateblue:Affiliate; Royal Blue:Author; Turquoise:Article

Reference

[1]Akdemir KC, Jain AK, Allton K, et al., 2014. Genome-wide profiling reveals stimulus-specific functions of p53 during differentiation and DNA damage of human embryonic stem cells. Nucleic Acids Res, 42(1):205-223.

[2]Barnum KJ, O'Connell MJ, 2014. Cell cycle regulation by checkpoints. In: Noguchi E, Gadaleta MC (Eds.), Cell Cycle Control: Methods in Molecular Biology (Methods and Protocols), Vol. 1170. Humana Press, New York, p.29-40.

[3]Besson A, Dowdy SF, Roberts JM, 2008. CDK inhibitors: cell cycle regulators and beyond. Dev Cell, 14(2):159-169.

[4]Betschinger J, Nichols J, Dietmann S, et al., 2013. Exit from pluripotency is gated by intracellular redistribution of the bHLH transcription factor Tfe3. Cell, 153(2):335-347.

[5]Biswas SC, Sanphui P, Chatterjee N, et al., 2017. Cdc25A phosphatase: a key cell cycle protein that regulates neuron death in disease and development. Cell Death Dis, 8(3):e2692.

[6]Buettner F, Natarajan KN, Casale FP, et al., 2015. Computational analysis of cell-to-cell heterogeneity in single-cell RNA-sequencing data reveals hidden subpopulations of cells. Nat Biotechnol, 33(2):155-160.

[7]Calder A, Roth-Albin I, Bhatia S, et al., 2013. Lengthened G1 phase indicates differentiation status in human embryonic stem cells. Stem Cells Dev, 22(2):279-295.

[8]Chatterjee N, Sanphui P, Kemeny S, et al., 2016. Role and regulation of Cdc25A phosphatase in neuron death induced by NGF deprivation or β-amyloid. Cell Death Discov, 2:16083.

[9]Cheung TH, Rando TA, 2013. Molecular regulation of stem cell quiescence. Nat Rev Mol Cell Biol, 14(6):329-340.

[10]Chia NY, Chan YS, Feng B, et al., 2010. A genome-wide RNAi screen reveals determinants of human embryonic stem cell identity. Nature, 468(7321):316-320.

[11]Coronado D, Godet M, Bourillot PY, et al., 2013. A short G1 phase is an intrinsic determinant of naïve embryonic stem cell pluripotency. Stem Cell Res, 10(1):118-131.

[12]Dalton S, 2013. G1 compartmentalization and cell fate coordination. Cell, 155(1):13-14.

[13]Dalton S, 2015. Linking the cell cycle to cell fate decisions. Trends Cell Biol, 25(10):592-600.

[14]Fang JS, Coon BG, Gillis N, et al., 2017. Shear-induced Notch-Cx37-p27 axis arrests endothelial cell cycle to enable arterial specification. Nat Commun, 8:2149.

[15]Gonzales KA, Liang HQ, Lim YS, et al., 2015. Deterministic restriction on pluripotent state dissolution by cell-cycle pathways. Cell, 162(3):564-579.

[16]Gruenheit N, Parkinson K, Brimson CA, et al., 2018. Cell cycle heterogeneity can generate robust cell type proportioning. Dev Cell, 47(4):494-508.e4.

[17]Gurdon JB, 2016. Cell fate determination by transcription factors. Curr Top Dev Biol, 116:445-454.

[18]Haas S, Trumpp A, Milsom MD, 2018. Causes and consequences of hematopoietic stem cell heterogeneity. Cell Stem Cell, 22(5):627-638.

[19]Harper JW, Adami GR, Wei N, et al., 1993. The p21 Cdk-interacting protein Cip1 is a potent inhibitor of G1 cyclin-dependent kinases. Cell, 75(4):805-816.

[20]Hartwell LH, Weinert TA, 1989. Checkpoints: controls that ensure the order of cell cycle events. Science, 246(4930):629-634.

[21]Haug JS, He XC, Grindley JC, et al., 2008. N-cadherin expression level distinguishes reserved versus primed states of hematopoietic stem cells. Cell Stem Cell, 2(4):367-379.

[22]Ito K, Suda T, 2014. Metabolic requirements for the maintenance of self-renewing stem cells. Nat Rev Mol Cell Biol, 15(4):243-256.

[23]Jiang HB, Xu ZM, Zhong P, et al., 2015. Cell cycle and p53 gate the direct conversion of human fibroblasts to dopaminergic neurons. Nat Commun, 6:10100.

[24]Johnson A, Skotheim JM, 2013. Start and the restriction point. Curr Opin Cell Biol, 25(6):717-723.

[25]Kar S, Wang MF, Yao W, et al., 2006. PM-20, a novel inhibitor of Cdc25A, induces extracellular signal-regulated kinase 1/2 phosphorylation and inhibits hepatocellular carcinoma growth in vitro and in vivo. Mol Cancer Ther, 5(6):1511-1519.

[26]Koledova Z, Kafkova LR, Calabkova L, et al., 2010. Cdk2 inhibition prolongs G1 phase progression in mouse embryonic stem cells. Stem Cells Dev, 19(2):181-194.

[27]Lange C, Huttner WB, Calegari F, 2009. Cdk4/cyclinD1 overexpression in neural stem cells shortens G1, delays neurogenesis, and promotes the generation and expansion of basal progenitors. Cell Stem Cell, 5(3):320-331.

[28]Lauridsen FKB, Jensen TL, Rapin N, et al., 2018. Differences in cell cycle status underlie transcriptional heterogeneity in the HSC compartment. Cell Rep, 24(3):766-780.

[29]Lee MH, Reynisdóttir I, Massagué J, 1995. Cloning of p57KIP2, a cyclin-dependent kinase inhibitor with unique domain structure and tissue distribution. Genes Dev, 9(6):639-649.

[30]Lein E, Borm LE, Linnarsson S, 2017. The promise of spatial transcriptomics for neuroscience in the era of molecular cell typing. Science, 358(6359):64-69.

[31]Li LH, Clevers H, 2010. Coexistence of quiescent and active adult stem cells in mammals. Science, 327(5965):542-545.

[32]Lim S, Kaldis P, 2013. Cdks, cyclins and CKIs: roles beyond cell cycle regulation. Development, 140(15):3079-3093.

[33]Lis R, Karrasch CC, Poulos MG, et al., 2017. Conversion of adult endothelium to immunocompetent haematopoietic stem cells. Nature, 545(7655):439-445.

[34]Liu ZQ, Wang L, Welch JD, et al., 2017. Single-cell transcriptomics reconstructs fate conversion from fibroblast to cardiomyocyte. Nature, 551(7678):100-104.

[35]Lu CJ, Fan XY, Guo YF, et al., 2019. Single-cell analyses identify distinct and intermediate states of zebrafish pancreatic islet development. J Mol Cell Biol, mjy064.

[36]Lu YC, Sanada C, Xavier-Ferrucio J, et al., 2018. The molecular signature of megakaryocyte-erythroid progenitors reveals a role for the cell cycle in fate specification. Cell Rep, 25(8):2083-2093.e4.

[37]Lugert S, Basak O, Knuckles P, et al., 2010. Quiescent and active hippocampal neural stem cells with distinct morphologies respond selectively to physiological and pathological stimuli and aging. Cell Stem Cell, 6(5):445-456.

[38]Ma YQ, Kanakousaki K, Buttitta L, 2015. How the cell cycle impacts chromatin architecture and influences cell fate. Front Genet, 6:19.

[39]Maimets T, Neganova I, Armstrong L, et al., 2008. Activation of p53 by nutlin leads to rapid differentiation of human embryonic stem cells. Oncogene, 27(40):5277-5287.

[40]Matson JP, Cook JG, 2017. Cell cycle proliferation decisions: the impact of single cell analyses. FEBS J, 284(3):362-375.

[41]Matsuoka S, Edwards MC, Bai C, et al., 1995. p57KIP2, a structurally distinct member of the p21CIP1 Cdk inhibitor family, is a candidate tumor suppressor gene. Genes Dev, 9(6):650-662.

[42]McDavid A, Finak G, Gottardo R, 2016. The contribution of cell cycle to heterogeneity in single-cell RNA-seq data. Nat Biotechnol, 34(6):591-593.

[43]Mende N, Kuchen EE, Lesche M, et al., 2015. CCND1-CDK4-mediated cell cycle progression provides a competitive advantage for human hematopoietic stem cells in vivo. J Exp Med, 212(8):1171-1183.

[44]Nagano T, Lubling Y, Stevens TJ, et al., 2013. Single-cell Hi-C reveals cell-to-cell variability in chromosome structure. Nature, 502(7469):59-64.

[45]Nagano T, Lubling Y, Várnai C, et al., 2017. Cell-cycle dynamics of chromosomal organization at single-cell resolution. Nature, 547(7661):61-67.

[46]Ohnuma S, Harris WA, 2003. Neurogenesis and the cell cycle. Neuron, 40(2):199-208.

[47]Orford KW, Scadden DT, 2008. Deconstructing stem cell self-renewal: genetic insights into cell-cycle regulation. Nat Rev Genet, 9(2):115-128.

[48]Otsuki L, Brand AH, 2018. Cell cycle heterogeneity directs the timing of neural stem cell activation from quiescence. Science, 360(6384):99-102.

[49]Pauklin S, Vallier L, 2013. The cell-cycle state of stem cells determines cell fate propensity. Cell, 155(1):135-147.

[50]Pietenpol JA, Stewart ZA, 2002. Cell cycle checkpoint signaling: cell cycle arrest versus apoptosis. Toxicology, 181-182:475-481.

[51]Polyak K, Lee MH, Erdjument-Bromage H, et al., 1994. Cloning of p27kip1, a cyclin-dependent kinase inhibitor and a potential mediator of extracellular antimitogenic signals. Cell, 78(1):59-66.

[52]Qian PX, He XC, Paulson A, et al., 2016. The Dlk1-Gtl2 locus preserves LT-HSC function by inhibiting the PI3K-mTOR pathway to restrict mitochondrial metabolism. Cell Stem Cell, 18(2):214-228.

[53]Roccio M, Schmitter D, Knobloch M, et al., 2013. Predicting stem cell fate changes by differential cell cycle progression patterns. Development, 140(2):459-470.

[54]Sakaue-Sawano A, Kurokawa H, Morimura T, et al., 2008. Visualizing spatiotemporal dynamics of multicellular cell-cycle progression. Cell, 132(3):487-498.

[55]Salazar-Roa M, Malumbres M, 2017. Fueling the cell division cycle. Trends Cell Biol, 27(1):69-81.

[56]Salomoni P, Calegari F, 2010. Cell cycle control of mammalian neural stem cells: putting a speed limit on G1. Trends Cell Biol, 20(5):233-243.

[57]Sela Y, Molotski N, Golan S, et al., 2012. Human embryonic stem cells exhibit increased propensity to differentiate during the G1 phase prior to phosphorylation of retinoblastoma protein. Stem Cells, 30(6):1097-1108.

[58]Sherr CJ, Roberts JM, 1999. CDK inhibitors: positive and negative regulators of G1-phase progression. Genes Dev, 13(12):1501-1512.

[59]Singh AM, Chappell J, Trost R, et al., 2013. Cell-cycle control of developmentally regulated transcription factors accounts for heterogeneity in human pluripotent cells. Stem Cell Rep, 1(6):532-544.

[60]Singh AM, Sun YH, Li L, et al., 2015. Cell-cycle control of bivalent epigenetic domains regulates the exit from pluripotency. Stem Cell Rep, 5(3):323-336.

[61]Skinner SO, Xu H, Nagarkar-Jaiswal S, et al., 2016. Single-cell analysis of transcription kinetics across the cell cycle. eLife, 5:e12175.

[62]Su TY, Stanley G, Sinha R, et al., 2018. Single-cell analysis of early progenitor cells that build coronary arteries. Nature, 559(7714):356-362.

[63]Sun N, Yu XM, Li F, et al., 2017. Inference of differentiation time for single cell transcriptomes using cell population reference data. Nat Commun, 8(1):1856.

[64]Thomson I, Gilchrist S, Bickmore WA, et al., 2004. The radial positioning of chromatin is not inherited through mitosis but is established de novo in early G1. Curr Biol, 14(2):166-172.

[65]Tomás-Loba A, Manieri E, González-Terán B, et al., 2019. p38γ is essential for cell cycle progression and liver tumorigenesis. Nature, 568(7753):557-560.

[66]Toyoshima H, Hunter T, 1994. p27, a novel inhibitor of G1 cyclin-Cdk protein kinase activity, is related to p21. Cell, 78(1):67-74.

[67]Upadhaya S, Sawai CM, Papalexi E, et al., 2018. Kinetics of adult hematopoietic stem cell differentiation in vivo. J Exp Med, 215(11):2815-2832.

[68]Vallier L, 2015. Cell cycle rules pluripotency. Cell Stem Cell, 17(2):131-132.

[69]Vierbuchen T, Ostermeier A, Pang ZP, et al., 2010. Direct conversion of fibroblasts to functional neurons by defined factors. Nature, 463(7284):1035-1041.

[70]Walter D, Lier A, Geiselhart A, et al., 2015. Exit from dormancy provokes DNA-damage-induced attrition in haematopoietic stem cells. Nature, 520(7548):549-552.

[71]Xie HF, Ye M, Feng R, et al., 2004. Stepwise reprogramming of B cells into macrophages. Cell, 117(5):663-676.

[72]Zerjatke T, Gak IA, Kirova D, et al., 2017. Quantitative cell cycle analysis based on an endogenous all-in-one reporter for cell tracking and classification. Cell Rep, 19(9):1953-1966.

[73]Zetterberg A, Larsson O, Wiman KG, 1995. What is the restriction point? Curr Opin Cell Biol, 7(6):835-842.

[74]Zhang MY, Dong Y, Hu FX, et al., 2018. Transcription factor Hoxb5 reprograms B cells into functional T lymphocytes. Nat Immunol, 19(3):279-290.

Open peer comments: Debate/Discuss/Question/Opinion

<1>

Please provide your name, email address and a comment





Journal of Zhejiang University-SCIENCE, 38 Zheda Road, Hangzhou 310027, China
Tel: +86-571-87952783; E-mail: cjzhang@zju.edu.cn
Copyright © 2000 - 2024 Journal of Zhejiang University-SCIENCE