Single-nucleus profiling unveils a geroprotective role of the FOXO3 in primate skeletal muscle aging

Ying Jing, Yuesheng Zuo, Yang Yu, Liang Sun, Zhengrong Yu, Shuai Ma, Qian Zhao, Guoqiang Sun, Huifang Hu, Jingyi Li, Daoyuan Huang, Lixiao Liu, Jiaming Li, Zijuan Xin, Haoyan Huang, Juan Carlos Izpisua Belmonte, Weiqi Zhang, Si Wang, Jing Qu, Guang-Hui Liu

PDF(8762 KB)
PDF(8762 KB)
Protein Cell ›› 2023, Vol. 14 ›› Issue (7) : 497-512. DOI: 10.1093/procel/pwac061
RESEARCH ARTICLE
RESEARCH ARTICLE

Single-nucleus profiling unveils a geroprotective role of the FOXO3 in primate skeletal muscle aging

Author information +
History +

Abstract

Age-dependent loss of skeletal muscle mass and function is a feature of sarcopenia, and increases the risk of many aging-related metabolic diseases. Here, we report phenotypic and single-nucleus transcriptomic analyses of non-human primate skeletal muscle aging. A higher transcriptional fluctuation was observed in myonuclei relative to other interstitial cell types, indicating a higher susceptibility of skeletal muscle fiber to aging. We found a downregulation of FOXO3 in aged primate skeletal muscle, and identified FOXO3 as a hub transcription factor maintaining skeletal muscle homeostasis. Through the establishment of a complementary experimental pipeline based on a human pluripotent stem cell-derived myotube model, we revealed that silence of FOXO3 accelerates human myotube senescence, whereas genetic activation of endogenous FOXO3 alleviates human myotube aging. Altogether, based on a combination of monkey skeletal muscle and human myotube aging research models, we unraveled the pivotal role of the FOXO3 in safeguarding primate skeletal muscle from aging, providing a comprehensive resource for the development of clinical diagnosis and targeted therapeutic interventions against human skeletal muscle aging and the onset of sarcopenia along with aging-related disorders.

Keywords

single-nucleus RNA sequencing / primate / aging / skeletal muscle / FOXO3

Cite this article

Download citation ▾
Ying Jing, Yuesheng Zuo, Yang Yu, Liang Sun, Zhengrong Yu, Shuai Ma, Qian Zhao, Guoqiang Sun, Huifang Hu, Jingyi Li, Daoyuan Huang, Lixiao Liu, Jiaming Li, Zijuan Xin, Haoyan Huang, Juan Carlos Izpisua Belmonte, Weiqi Zhang, Si Wang, Jing Qu, Guang-Hui Liu. Single-nucleus profiling unveils a geroprotective role of the FOXO3 in primate skeletal muscle aging. Protein Cell, 2023, 14(7): 497‒512 https://doi.org/10.1093/procel/pwac061

References

[1]
Aibar S, González-Blas CB, Moerman T et al. SCENIC: single-cell regulatory network inference and clustering. Nat Methods 2017;14:1083–1086.
CrossRef Google scholar
[2]
Anders S, Pyl PT, Huber W. HTSeq—a Python framework to work with high-throughput sequencing data. Bioinformatics 2015;31:166–169.
CrossRef Google scholar
[3]
Anderson RM, Colman RJ. Prospects and perspectives in primate aging research. Antioxid Redox Signal 2011;14:203–205.
CrossRef Google scholar
[4]
Angelidis I, Simon LM, Fernandez IE et al. An atlas of the aging lung mapped by single cell transcriptomics and deep tissue proteomics. Nat Commun 2019;10:963.
CrossRef Google scholar
[5]
Askanas V, Engel WK. Inclusion-body myositis, a multifactorial muscle disease associated with aging: current concepts of pathogenesis. Curr Opin Rheumatol 2007;19:550–559.
CrossRef Google scholar
[6]
Askanas V, Engel WK, Nogalska A. Pathogenic considerations in sporadic inclusion-body myositis, a degenerative muscle disease associated with aging and abnormalities of myoproteostasis. J Neuropathol Exp Neurol 2012;71:680–693.
CrossRef Google scholar
[7]
Baskin KK, Winders BR, Olson EN. Muscle as a “mediator” of systemic metabolism. Cell Metab 2015;21:237–248.
CrossRef Google scholar
[8]
Butler A, Hoffman P, Smibert P et al. Integrating single-cell transcriptomic data across different conditions, technologies, and species. Nat Biotechnol 2018;36:411–420.
CrossRef Google scholar
[9]
Cadot B, Gache V, Gomes ER. Moving and positioning the nucleus in skeletal muscle – one step at a time. Nucleus 2015;6:373–381.
CrossRef Google scholar
[10]
Cai Y, Song W, Li J et al. The landscape of aging. Sci China Life Sci 2022; 65(12), 2354–2454.
CrossRef Google scholar
[11]
Calissi G, Lam EW, Link W. Therapeutic strategies targeting FOXO transcription factors. Nat Rev Drug Discov 2021;20:21–38.
CrossRef Google scholar
[12]
Dalle S, Koppo K. Cannabinoid receptor 1 expression is higher in muscle of old vs. young males, and increases upon resistance exercise in older adults. Sci Rep 2021;11:18349.
CrossRef Google scholar
[13]
De Micheli AJ, Spector JA, Elemento O et al. A reference single-cell transcriptomic atlas of human skeletal muscle tissue reveals bifurcated muscle stem cell populations. Skelet Muscle 2020;10:19.
CrossRef Google scholar
[14]
Dell’Orso S, Juan AH, Ko KD et al. Single cell analysis of adult mouse skeletal muscle stem cells in homeostatic and regenerative conditions. Development 2019;146:dev174177.
CrossRef Google scholar
[15]
Demontis F, Piccirillo R, Goldberg AL et al. Mechanisms of skeletal muscle aging: insights from Drosophila and mammalian models. Dis Model Mech 2013;6:1339–1352.
CrossRef Google scholar
[16]
Dirks AJ, Hofer T, Marzetti E et al. Mitochondrial DNA mutations, energy metabolism and apoptosis in aging muscle. Ageing Res Rev 2006;5:179–195.
CrossRef Google scholar
[17]
Domingues-Faria C, Vasson MP, Goncalves-Mendes N et al. Skeletal muscle regeneration and impact of aging and nutrition. Ageing Res Rev 2016;26:22–36.
CrossRef Google scholar
[18]
Dos Santos M, Backer S, Saintpierre B et al. Single-nucleus RNA-seq and FISH identify coordinated transcriptional activity in mammalian myofibers. Nat Commun 2020;11:5102.
CrossRef Google scholar
[19]
Fleming SJ, Marioni JC, Babadi M. CellBender remove-background: a deep generative model for unsupervised removal of background noise from scRNA-seq datasets. bioRxiv 2019.
CrossRef Google scholar
[20]
Frontera WR, Ochala J. Skeletal muscle: a brief review of structure and function. Calcif Tissue Int 2015;96:183–195.
CrossRef Google scholar
[21]
Fuchs E, Blau HM. Tissue stem cells: architects of their niches. Cell Stem Cell 2020;27:532–556.
CrossRef Google scholar
[22]
Giordani L, He GJ, Negroni E et al. High-dimensional single-cell cartography reveals novel skeletal muscle-resident cell populations. Mol Cell 2019;74:609–621.e6.
CrossRef Google scholar
[23]
Gomarasca M, Banfi G, Lombardi G. Myokines: the endocrine coupling of skeletal muscle and bone. Adv Clin Chem 2020;94:155–218.
CrossRef Google scholar
[24]
González JP, Queralt-Rosinach N, Bravo A et al. DisGeNET: a discovery platform for the dynamical exploration of human diseases and their genes. Database 2015;2015:bav028.
CrossRef Google scholar
[25]
Guang-Hui Liu YB, Qu J, Zhang W et al. Aging Atlas: a multi-omics database for aging biology. Nucleic Acids Res 2021;49:D825–d830.
CrossRef Google scholar
[26]
He X, Memczak S, Qu J et al. Single-cell omics in ageing: a young and growing field. Nat Metab 2020;2:293–302.
CrossRef Google scholar
[27]
Hershberg EA, Camplisson CK, Close JL et al. PaintSHOP enables the interactive design of transcriptome- and genome-scale oligonucleotide FISH experiments. Nat Methods 2021;18:937–944.
CrossRef Google scholar
[28]
Hu H, Ji Q, Song M et al. ZKSCAN3 counteracts cellular senescence by stabilizing heterochromatin. Nucleic Acids Res 2020;48:6001–6018.
CrossRef Google scholar
[29]
Iizuka K, Machida T, Hirafuji M. Skeletal muscle is an endocrine organ. J Pharmacol Sci 2014;125:125–131.
CrossRef Google scholar
[30]
Irrthum A, Wehenkel L, Geurts P. Inferring regulatory networks from expression data using tree-based methods. PLoS One 2010;5:e12776.
CrossRef Google scholar
[31]
Jiang C, Wen Y, Kuroda K et al. Notch signaling deficiency underlies age-dependent depletion of satellite cells in muscular dystrophy. Dis Model Mech 2014;7:997–1004.
CrossRef Google scholar
[32]
Jones RA, Harrison C, Eaton SL et al. Cellular and molecular anatomy of the human neuromuscular junction. Cell Rep 2017;21:2348–2356.
CrossRef Google scholar
[33]
Kalinkovich A, Livshits G. Sarcopenic obesity or obese sarcopenia: a cross talk between age-associated adipose tissue and skeletal muscle inflammation as a main mechanism of the pathogenesis. Ageing Res Rev 2017;35:200–221.
CrossRef Google scholar
[34]
Kanehisa M, Furumichi M, Sato Y et al. KEGG: integrating viruses and cellular organisms. Nucleic Acids Res 2021;49:D545–D551.
CrossRef Google scholar
[35]
Kang W, Jin T, Zhang T et al. Regeneration Roadmap: data-base resources for regenerative biology. Nucleic Acids Res 2022;50:D1085–D1090.
[36]
Kim D, Langmead B, Salzberg SL. HISAT: a fast spliced aligner with low memory requirements. Nat Methods 2015;12:357–360.
CrossRef Google scholar
[37]
Kim M, Franke V, Brandt B et al. Single-nucleus transcriptomics reveals functional compartmentalization in syncytial skeletal muscle cells. Nat Commun 2020;11:6375.
CrossRef Google scholar
[38]
Kishi JY, Lapan SW, Beliveau BJ et al. SABER amplifies FISH: enhanced multiplexed imaging of RNA and DNA in cells and tissues. Nat Methods 2019;16:533–544.
CrossRef Google scholar
[39]
Krishnaswami SR, Grindberg RV, Novotny M et al. Using single nuclei for RNA-seq to capture the transcriptome of postmortem neurons. Nat Protoc 2016;11:499–524.
CrossRef Google scholar
[40]
Larsson L, Degens H, Li M et al. Sarcopenia: aging-related loss of muscle mass and function. Physiol Rev 2019;99:427–511.
CrossRef Google scholar
[41]
Leger B, Derave W, De Bock K et al. Human sarcopenia reveals an increase in SOCS-3 and myostatin and a reduced efficiency of Akt phosphorylation. Rejuvenation Res 2008;11:163–175B.
CrossRef Google scholar
[42]
Lei J, Wang S, Kang W et al. FOXO3-engineered human mesenchymal progenitor cells efficiently promote cardiac repair after myocardial infarction. Protein Cell 2021;12:145–151.
CrossRef Google scholar
[43]
Leng SX, Pawelec G. Single-cell immune atlas for human aging and frailty. Life Med 2022:lnac013.
CrossRef Google scholar
[44]
Li J, Zheng Y, Yan P et al. A single-cell transcriptomic atlas of primate pancreatic islet aging. Natl Sci Rev 2021;8:nwaa127.
CrossRef Google scholar
[45]
Liberzon A, Birger C, Thorvaldsdóttir H et al. The Molecular Signatures Database (MSigDB) hallmark gene set collection. Cell Syst 2015;1:417–425.
CrossRef Google scholar
[46]
Limbad C, Doi R, McGirr J et al. Senolysis induced by 25-hydroxycholesterol targets CRYAB in multiple cell types. iScience 2022;25:103848.
CrossRef Google scholar
[47]
Lipina C, Hundal HS. Lipid modulation of skeletal muscle mass and function. J Cachexia Sarcopenia Muscle 2017;8:190–201.
CrossRef Google scholar
[48]
Liu L, Liu X, Bai Y et al. Neuregulin-1β modulates myogenesis in septic mouse serum-treated C2C12 myotubes in vitro through PPARγ/NF-κB signaling. Mol Biol Rep 2018;45:1611–1619.
CrossRef Google scholar
[49]
Liu Z, Li W, Geng L et al. Cross-species metabolomic analysis identifies uridine as a potent regeneration promoting factor. Cell Discov 2022;8:6.
CrossRef Google scholar
[50]
Livshits G, Kalinkovich A. Inflammaging as a common ground for the development and maintenance of sarcopenia, obesity, cardiomyopathy and dysbiosis. Ageing Res Rev 2019;56:100980.
CrossRef Google scholar
[51]
Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol 2014;15:550.
CrossRef Google scholar
[52]
Ma S, Sun S, Geng L et al. Caloric restriction reprograms the single-cell transcriptional landscape of Rattus Norvegicus aging. Cell 2020;180:1001.e22.
CrossRef Google scholar
[53]
Ma S, Sun S, Li J et al. Single-cell transcriptomic atlas of primate cardiopulmonary aging. Cell Res 2021;31:415–432.
CrossRef Google scholar
[54]
Ma S, Wang S, Ye Y et al. Heterochronic parabiosis induces stem cell revitalization and systemic rejuvenation across aged tissues. Cell Stem Cell 2022;29:1005.e10.
CrossRef Google scholar
[55]
Maeda S, Miyagawa S, Kawamura T et al. Notch signaling-modified mesenchymal stem cells improve tissue perfusion by induction of arteriogenesis in a rat hindlimb ischemia model. Sci Rep 2021;11:2543.
CrossRef Google scholar
[56]
Maffioletti SM, Gerli MF, Ragazzi M et al. Efficient derivation and inducible differentiation of expandable skeletal myogenic cells from human ES and patient-specific iPS cells. Nat Protoc 2015;10:941–958.
CrossRef Google scholar
[57]
Mammucari C, Milan G, Romanello V et al. FoxO3 controls autophagy in skeletal muscle in vivo. Cell Metab 2007;6:458–471.
CrossRef Google scholar
[58]
McGinnis CS, Murrow LM, Gartner ZJ. DoubletFinder: doublet detection in single-cell RNA sequencing data using artificial nearest neighbors. Cell Syst 2019;8:329–337.e4.
CrossRef Google scholar
[59]
McKellar DW, Walter LD, Song LT et al. Large-scale integration of single-cell transcriptomic data captures transitional progenitor states in mouse skeletal muscle regeneration. Commun Biol 2021;4:1280.
CrossRef Google scholar
[60]
Mercken EM, Capri M, Carboneau BA et al. Conserved and species-specific molecular denominators in mammalian skeletal muscle aging. NPJ Aging Mech Dis 2017;3:8.
CrossRef Google scholar
[61]
Morris BJ, Willcox DC, Donlon TA et al. FOXO3: A major gene for human longevity — a mini-review. Gerontology 2015;61:515–525.
CrossRef Google scholar
[62]
Murgia M, Toniolo L, Nagaraj N et al. Single muscle fiber proteomics reveals fiber-type-specific features of human muscle aging. Cell Rep 2017;19:2396–2409.
CrossRef Google scholar
[63]
Navarro G. A guided tour to approximate string matching. ACM Comput Surv 2001;33:31–88.
CrossRef Google scholar
[64]
Ohsawa N, Koebis M, Suo S et al. Alternative splicing of PDLIM3/ALP, for α-actinin-associated LIM protein 3, is aberrant in persons with myotonic dystrophy. Biochem Biophys Res Commun 2011;409:64–69.
CrossRef Google scholar
[65]
Orchard P, Manickam N, Ventresca C et al. Human and rat skeletal muscle single-nuclei multi-omic integrative analyses nominate causal cell types, regulatory elements, and SNPs for complex traits. Genome Res 2021;31:2258–2275.
CrossRef Google scholar
[66]
Podnar J, Deiderick H, Hunicke-Smith S. Next-generation sequencing fragment library construction. Curr Protoc Mol Biol 2014;107:7.17.11–7.17.16.
CrossRef Google scholar
[67]
Rubenstein AB, Smith GR, Raue U et al. Single-cell transcriptional profiles in human skeletal muscle. Sci Rep 2020;10:229.
CrossRef Google scholar
[68]
Salih DA, Brunet A. FoxO transcription factors in the maintenance of cellular homeostasis during aging. Curr Opin Cell Biol 2008;20:126–136.
CrossRef Google scholar
[69]
Sandri M, Sandri C, Gilbert A et al. Foxo transcription factors induce the atrophy-related ubiquitin ligase atrogin-1 and cause skeletal muscle atrophy. Cell 2004;117:399–412.
CrossRef Google scholar
[70]
Shannon P, Markiel A, Ozier O et al. Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res 2003;13:2498–2504.
CrossRef Google scholar
[71]
Simon M, Van Meter M, Ablaeva J et al. LINE1 derepression in aged wild-type and SIRT6-deficient mice drives inflammation. Cell Metab 2019;29:871–885.e5 e875.
CrossRef Google scholar
[72]
Stokes T, Hector AJ, Morton RW et al. Recent perspectives regarding the role of dietary protein for the promotion of muscle hypertrophy with resistance exercise training. Nutrients 2018;10:180.
CrossRef Google scholar
[73]
Tieland M, Trouwborst I, Clark BC. Skeletal muscle performance and ageing. J Cachexia Sarcopenia Muscle 2018;9:3–19.
CrossRef Google scholar
[74]
Verma M, Asakura Y, Murakonda BSR et al. Muscle satellite cell cross-talk with a vascular niche maintains quiescence via VEGF and notch signaling. Cell Stem Cell 2018;23:530–543.e9.
CrossRef Google scholar
[75]
Wagers AJ, Conboy IM. Cellular and molecular signatures of muscle regeneration: current concepts and controversies in adult myogenesis. Cell 2005;122:659–667.
CrossRef Google scholar
[76]
Wang S, Cheng F, Ji Q et al. Hyperthermia differentially affects specific human stem cells and their differentiated derivatives. Protein Cell 2022;13:615–622.
CrossRef Google scholar
[77]
Wang S, Hu B, Ding Z et al. ATF6 safeguards organelle homeostasis and cellular aging in human mesenchymal stem cells. Cell Discov 2018;4:2.
CrossRef Google scholar
[78]
Wang S, Yao X, Ma S et al. A single-cell transcriptomic landscape of the lungs of patients with COVID-19. Nat Cell Biol 2021a;23:1314–1328.
CrossRef Google scholar
[79]
Wang S, Zheng Y, Li J et al. Single-cell transcriptomic atlas of primate ovarian aging. Cell 2020;180:585–600.e19.
CrossRef Google scholar
[80]
Wang S, Zheng Y, Li Q et al. Deciphering primate retinal aging at single-cell resolution. Protein Cell 2021b;12:889–898.
CrossRef Google scholar
[81]
Wang Y, Ng S-C. Sphingolipids mediate lipotoxicity in muscular dystrophies. Life Med 2022:lnac015.
CrossRef Google scholar
[82]
Wickham, H. 2016. ggplot2: Elegant Graphics for Data Analysis. Springer
CrossRef Google scholar
[83]
Wilkinson DJ, Piasecki M, Atherton PJ. The age-related loss of skeletal muscle mass and function: measurement and physiology of muscle fibre atrophy and muscle fibre loss in humans. Ageing Res Rev 2018;47:123–132.
CrossRef Google scholar
[84]
Yan P, Li Q, Wang L et al. FOXO3-engineered human ESC-derived vascular cells promote vascular protection and regeneration. Cell Stem Cell 2019;24:447–461.e8.
CrossRef Google scholar
[85]
Yates AD, Achuthan P, Akanni W et al. Ensembl 2020. Nucleic Acids Res 2020;48:D682–D688.
[86]
Yu W, Clyne M, Khoury MJ et al. Phenopedia and genopedia: disease- centered and gene-centered views of the evolving knowledge of human genetic associations. Bioinformatics 2010;26:145–146.
CrossRef Google scholar
[87]
Zhang W, Li J, Suzuki K, et al. Aging stem cells. A Werner syndrome stem cell model unveils heterochromatin alterations as a driver of human aging. Sci 2015;348:1160–1163.
CrossRef Google scholar
[88]
Zhang H, Li J, Ren J et al. Single-nucleus transcriptomic landscape of primate hippocampal aging. Protein Cell 2021;12:695–716.
CrossRef Google scholar
[89]
Zhang W, Wan H, Feng G et al. SIRT6 deficiency results in developmental retardation in cynomolgus monkeys. Nature 2018;560:661–665.
CrossRef Google scholar
[90]
Zhang W, Zhang S, Yan P et al. A single-cell transcriptomic landscape of primate arterial aging. Nat Commun 2020a;11:2202.
CrossRef Google scholar
[91]
Zhang W, Qu J, Liu G-H, Belmonte JCI. The ageing epigenome and its rejuvenation. Nat Rev Mol Cell Biol 2020b;21:137–150.
CrossRef Google scholar
[92]
Zhang Y, Zheng Y, Wang S et al. Single-nucleus transcriptomics reveals a gatekeeper role for FOXP1 in primate cardiac aging. Protein & Cell 2022:pwac038.
CrossRef Google scholar
[93]
Zhao D, Chen S. Failures at every level: breakdown of the epigenetic machinery of aging. Life Med 2022:lnac016.
CrossRef Google scholar
[94]
Zhao J, Brault JJ, Schild A et al. FoxO3 coordinately activates protein degradation by the autophagic/lysosomal and proteasomal pathways in atrophying muscle cells. Cell Metab 2007;6:472–483.
CrossRef Google scholar
[95]
Zhou Y, Zhou B, Pache L et al. Metascape provides a biologist-oriented resource for the analysis of systems-level datasets. Nat Commun 2019;10:1523.
CrossRef Google scholar
[96]
Zou, X., Dai, X., Mentis, A.F.A., Esteban, M.A., Liu, L., and Han, L. From monkey single-cell atlases into a broader biomedical perspective. Life Med. 2022:lnac028.
CrossRef Google scholar
[97]
Zou Z, Long X, Zhao Q et al. A single-cell transcriptomic atlas of human skin aging. Dev Cell 2021;56:383–397.e8.
CrossRef Google scholar

RIGHTS & PERMISSIONS

2022 The Author(s) 2022. Published by Oxford University Press on behalf of Higher Education Press.
AI Summary AI Mindmap
PDF(8762 KB)

Accesses

Citations

Detail

Sections
Recommended

/