Cảnh quan biểu hiện và sự đồng điều chỉnh của các gen lão hóa đặc hiệu theo mô và loại tế bào ở người
Tóm tắt
Từ khóa
#lão hóa tế bào #gen lão hóa #phân tích mạng lưới gen #RNA-seq #mô người #tính đặc hiệu theo loại tế bàoTài liệu tham khảo
Gorgoulis V, Adams PD, Alimonti A, Bennett DC, Bischof O, Bishop C, et al. Cellular senescence: defining a path forward. Cell. 2019;179(4):813–27. https://doi.org/10.1016/j.cell.2019.10.005.
Hayflick L, Moorhead PS. The serial cultivation of human diploid cell strains. Exp Cell Res. 1961;25(3):585–621. https://doi.org/10.1016/0014-4827(61)90192-6.
de Magalhães JP, Passos JF. Stress, cell senescence and organismal ageing. Mech Ageing Dev. 2018;170:2–9. https://doi.org/10.1016/j.mad.2017.07.001.
Rhinn M, Ritschka B, Keyes WM. Cellular senescence in development, regeneration and disease. Development. 2019;146(20). https://doi.org/10.1242/dev.151837.
Demaria M, Ohtani N, Youssef SA, Rodier F, Toussaint W, Mitchell JR, et al. An essential role for senescent cells in optimal wound healing through secretion of PDGF-AA. Dev Cell. 2014;31(6):722–33. https://doi.org/10.1016/j.devcel.2014.11.012.
Lee S, Schmitt CA. The dynamic nature of senescence in cancer. Nat Cell Biol. 2019;21(1):94–101. https://doi.org/10.1038/s41556-018-0249-2.
Ogrodnik M, Miwa S, Tchkonia T, Tiniakos D, Wilson CL, Lahat A, et al. Cellular senescence drives age-dependent hepatic steatosis. Nat Commun. 2017;8(1):15691. https://doi.org/10.1038/ncomms15691.
Schafer MJ, White TA, Iijima K, Haak AJ, Ligresti G, Atkinson EJ, et al. Cellular senescence mediates fibrotic pulmonary disease. Nat Commun. 2017;8(1):14532. https://doi.org/10.1038/ncomms14532.
Aguayo-Mazzucato C, Andle J, Lee TB Jr, Midha A, Talemal L, Chipashvili V, et al. Acceleration of beta cell aging determines diabetes and Senolysis improves disease outcomes. Cell Metab. 2019;30(1):129–42 e124. https://doi.org/10.1016/j.cmet.2019.05.006.
Baker DJ, Petersen RC. Cellular senescence in brain aging and neurodegenerative diseases: evidence and perspectives. J Clin Invest. 2018;128(4):1208–16. https://doi.org/10.1172/JCI95145.
Johmura Y, Yamanaka T, Omori S, Wang T-W, Sugiura Y, Matsumoto M, et al. Senolysis by glutaminolysis inhibition ameliorates various age-associated disorders. Science. 2021;371(6526):265–70. https://doi.org/10.1126/science.abb5916.
Zhang P, Kishimoto Y, Grammatikakis I, Gottimukkala K, Cutler RG, Zhang S, et al. Senolytic therapy alleviates Abeta-associated oligodendrocyte progenitor cell senescence and cognitive deficits in an Alzheimer’s disease model. Nat Neurosci. 2019;22(5):719–28. https://doi.org/10.1038/s41593-019-0372-9.
van Deursen JM. The role of senescent cells in ageing. Nature. 2014;509(7501):439–46. https://doi.org/10.1038/nature13193.
Noren Hooten N, Evans MK. Techniques to induce and quantify cellular senescence. JoVE. 2017:e55533.
He S, Sharpless NE. Senescence in health and disease. Cell. 2017;169(6):1000–11. https://doi.org/10.1016/j.cell.2017.05.015.
Coppe JP, Patil CK, Rodier F, Sun Y, Munoz DP, Goldstein J, et al. Senescence-associated secretory phenotypes reveal cell-nonautonomous functions of oncogenic RAS and the p53 tumor suppressor. PLoS Biol. 2008;6(12):2853–68. https://doi.org/10.1371/journal.pbio.0060301.
Kuilman T, Michaloglou C, Vredeveld LC, Douma S, van Doorn R, Desmet CJ, et al. Oncogene-induced senescence relayed by an interleukin-dependent inflammatory network. Cell. 2008;133(6):1019–31. https://doi.org/10.1016/j.cell.2008.03.039.
Chien Y, Scuoppo C, Wang X, Fang X, Balgley B, Bolden JE, et al. Control of the senescence-associated secretory phenotype by NF-kappaB promotes senescence and enhances chemosensitivity. Genes Dev. 2011;25(20):2125–36. https://doi.org/10.1101/gad.17276711.
Freund A, Patil CK, Campisi J. p38MAPK is a novel DNA damage response-independent regulator of the senescence-associated secretory phenotype. EMBO J. 2011;30(8):1536–48. https://doi.org/10.1038/emboj.2011.69.
Herranz N, Gallage S, Mellone M, Wuestefeld T, Klotz S, Hanley CJ, et al. mTOR regulates MAPKAPK2 translation to control the senescence-associated secretory phenotype. Nat Cell Biol. 2015;17(9):1205–17. https://doi.org/10.1038/ncb3225.
Laberge RM, Sun Y, Orjalo AV, Patil CK, Freund A, Zhou L, et al. MTOR regulates the pro-tumorigenic senescence-associated secretory phenotype by promoting IL1A translation. Nat Cell Biol. 2015;17(8):1049–61. https://doi.org/10.1038/ncb3195.
Sharpless NE, Sherr CJ. Forging a signature of in vivo senescence. Nat Rev Cancer. 2015;15(7):397–408. https://doi.org/10.1038/nrc3960.
Hernandez-Segura A, de Jong TV, Melov S, Guryev V, Campisi J, Demaria M. Unmasking transcriptional heterogeneity in senescent cells. Curr Biol. 2017;27(17):2652–60 e2654. https://doi.org/10.1016/j.cub.2017.07.033.
Casella G, Munk R, Kim KM, Piao Y, De S, Abdelmohsen K, et al. Transcriptome signature of cellular senescence. Nucleic Acids Res. 2019;47(14):7294–305. https://doi.org/10.1093/nar/gkz555.
Basisty N, Kale A, Jeon OH, Kuehnemann C, Payne T, Rao C, et al. A proteomic atlas of senescence-associated secretomes for aging biomarker development. PLoS Biol. 2020;18(1):e3000599. https://doi.org/10.1371/journal.pbio.3000599.
Avelar RA, Ortega JG, Tacutu R, Tyler EJ, Bennett D, Binetti P, et al. A multidimensional systems biology analysis of cellular senescence in aging and disease. Genome Biol. 2020;21(1):91. https://doi.org/10.1186/s13059-020-01990-9.
Song WM, Agrawal P, Von Itter R, Fontanals-Cirera B, Wang M, Zhou X, et al. Network models of primary melanoma microenvironments identify key melanoma regulators underlying prognosis. Nat Commun. 2021;12(1):1214. https://doi.org/10.1038/s41467-021-21457-0.
Wang Q, Zhang Y, Wang M, Song WM, Shen Q, McKenzie A, et al. The landscape of multiscale transcriptomic networks and key regulators in Parkinson’s disease. Nat Commun. 2019;10(1):5234. https://doi.org/10.1038/s41467-019-13144-y.
Wang M, Li A, Sekiya M, Beckmann ND, Quan X, Schrode N, et al. Transformative network modeling of multi-omics data reveals detailed circuits, key regulators, and potential therapeutics for Alzheimer’s disease. Neuron. 2020;109(2):257-272.e14. https://doi.org/10.1016/j.neuron.2020.11.002.
Song WM, Zhang B. Multiscale embedded gene co-expression network analysis. PLoS Comput Biol. 2015;11(11):e1004574. https://doi.org/10.1371/journal.pcbi.1004574.
Robinson MD, McCarthy DJ, Smyth GK. edgeR: a bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics. 2010;26(1):139–40. https://doi.org/10.1093/bioinformatics/btp616.
Tumminello M, Aste T, Di Matteo T, Mantegna RN. A tool for filtering information in complex systems. Proc Natl Acad Sci U S A. 2005;102(30):10421–6. https://doi.org/10.1073/pnas.0500298102.
Yu G, Wang LG, Han Y, He QY. clusterProfiler: an R package for comparing biological themes among gene clusters. Omics. 2012;16(5):284–7. https://doi.org/10.1089/omi.2011.0118.
Franzen O, Gan LM, Bjorkegren JLM. PanglaoDB: a web server for exploration of mouse and human single-cell RNA sequencing data. Database. 2019;2019. https://doi.org/10.1093/database/baz046.
Gu Z, Eils R, Schlesner M. Complex heatmaps reveal patterns and correlations in multidimensional genomic data. Bioinformatics. 2016;32(18):2847–9. https://doi.org/10.1093/bioinformatics/btw313.
Li M, Santpere G, Imamura Kawasawa Y, Evgrafov OV, Gulden FO, Pochareddy S, et al. Integrative functional genomic analysis of human brain development and neuropsychiatric risks. Science. 2018;362(6420). https://doi.org/10.1126/science.aat7615.
Stuart T, Butler A, Hoffman P, Hafemeister C, Papalexi E, Mauck WM 3rd, et al. Comprehensive integration of single-cell data. Cell. 2019;177(7):1888–902 e1821. https://doi.org/10.1016/j.cell.2019.05.031.
Guo J, Grow EJ, Mlcochova H, Maher GJ, Lindskog C, Nie X, et al. The adult human testis transcriptional cell atlas. Cell Res. 2018;28(12):1141–57. https://doi.org/10.1038/s41422-018-0099-2.
Baron M, Veres A, Wolock SL, Faust AL, Gaujoux R, Vetere A, et al. A single-cell transcriptomic map of the human and mouse pancreas reveals inter- and intra-cell population structure. Cell Syst. 2016;3(4):346–60 e344. https://doi.org/10.1016/j.cels.2016.08.011.
Madissoon E, Wilbrey-Clark A, Miragaia RJ, Saeb-Parsy K, Mahbubani KT, Georgakopoulos N, et al. scRNA-seq assessment of the human lung, spleen, and esophagus tissue stability after cold preservation. Genome Biol. 2019;21(1):1. https://doi.org/10.1186/s13059-019-1906-x.
Jin S, Guerrero-Juarez CF, Zhang L, Chang I, Ramos R, Kuan CH, et al. Inference and analysis of cell-cell communication using CellChat. Nat Commun. 2021;12(1):1088. https://doi.org/10.1038/s41467-021-21246-9.
Maynard KR, Collado-Torres L, Weber LM, Uytingco C, Barry BK, Williams SR, et al. Transcriptome-scale spatial gene expression in the human dorsolateral prefrontal cortex. Nat Neurosci. 2021;24(3):425–36. https://doi.org/10.1038/s41593-020-00787-0.
Hafemeister C, Satija R. Normalization and variance stabilization of single-cell RNA-seq data using regularized negative binomial regression. Genome Biol. 2019;20(1):296. https://doi.org/10.1186/s13059-019-1874-1.
Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci. 2005;102(43):15545–50. https://doi.org/10.1073/pnas.0506580102.
Kiss T, Nyul-Toth A, Balasubramanian P, Tarantini S, Ahire C, DelFavero J, et al. Single-cell RNA sequencing identifies senescent cerebromicrovascular endothelial cells in the aged mouse brain. Geroscience. 2020;42(2):429–44. https://doi.org/10.1007/s11357-020-00177-1.
Korotkevich G, Sukhov V, Budin N, Shpak B, Artyomov MN, Sergushichev A. Fast gene set enrichment analysis. bioRxiv. 2021:060012.
Consortium GT. The genotype-tissue expression (GTEx) project. Nat Genet. 2013;45:580–5.
Consortium GT. Human genomics. The genotype-tissue expression (GTEx) pilot analysis: multitissue gene regulation in humans. Science. 2015;348(6235):648–60. https://doi.org/10.1126/science.1262110.
Keen JC, Moore HM. The genotype-tissue expression (GTEx) project: linking clinical data with molecular analysis to advance personalized medicine. Journal of Personalized Medicine. 2015;5(1):22–9. https://doi.org/10.3390/jpm5010022.
Brown JP, Wei W, Sedivy JM. Bypass of senescence after disruption of p21CIP1/WAF1 gene in normal diploid human fibroblasts. Science. 1997;277(5327):831–4. https://doi.org/10.1126/science.277.5327.831.
Sebastian T, Malik R, Thomas S, Sage J, Johnson PF. C/EBPbeta cooperates with RB:E2F to implement Ras(V12)-induced cellular senescence. EMBO J. 2005;24(18):3301–12. https://doi.org/10.1038/sj.emboj.7600789.
Kortlever RM, Higgins PJ, Bernards R. Plasminogen activator inhibitor-1 is a critical downstream target of p53 in the induction of replicative senescence. Nat Cell Biol. 2006;8(8):877–84. https://doi.org/10.1038/ncb1448.
Ohtani N, Zebedee Z, Huot TJ, Stinson JA, Sugimoto M, Ohashi Y, et al. Opposing effects of Ets and id proteins on p16INK4a expression during cellular senescence. Nature. 2001;409(6823):1067–70. https://doi.org/10.1038/35059131.
Jun JI, Lau LF. The matricellular protein CCN1 induces fibroblast senescence and restricts fibrosis in cutaneous wound healing. Nat Cell Biol. 2010;12(7):676–85. https://doi.org/10.1038/ncb2070.
Martinez-Zamudio RI, Roux PF, de Freitas J, Robinson L, Dore G, Sun B, et al. AP-1 imprints a reversible transcriptional programme of senescent cells. Nat Cell Biol. 2020;22(7):842–55. https://doi.org/10.1038/s41556-020-0529-5.
Saini Y, Chen J, Patial S. The Tristetraprolin family of RNA-binding proteins in cancer: progress and future prospects. Cancers. 2020;12(6). https://doi.org/10.3390/cancers12061539.
Soliman MA, Berardi P, Pastyryeva S, Bonnefin P, Feng X, Colina A, et al. ING1a expression increases during replicative senescence and induces a senescent phenotype. Aging Cell. 2008;7(6):783–94. https://doi.org/10.1111/j.1474-9726.2008.00427.x.
Di Micco R, Krizhanovsky V, Baker D, d'Adda di Fagagna F. Cellular senescence in ageing: from mechanisms to therapeutic opportunities. Nat Rev Mol Cell Biol. 2021;22(2):75–95. https://doi.org/10.1038/s41580-020-00314-w.
Craig RW. MCL1 provides a window on the role of the BCL2 family in cell proliferation, differentiation and tumorigenesis. Leukemia. 2002;16(4):444–54. https://doi.org/10.1038/sj.leu.2402416.
Tonnessen-Murray CA, Lozano G, Jackson JG. The regulation of cellular functions by the p53 protein: cellular senescence. Cold Spring Harb Perspect Med. 2017;7(2). https://doi.org/10.1101/cshperspect.a026112.
Li Q, Tang L, Roberts PC, Kraniak JM, Fridman AL, Kulaeva OI, et al. Interferon regulatory factors IRF5 and IRF7 inhibit growth and induce senescence in immortal Li-Fraumeni fibroblasts. Mol Cancer Res. 2008;6(5):770–84. https://doi.org/10.1158/1541-7786.MCR-07-0114.
Wysk M, Yang DD, Lu H-T, Flavell RA, Davis RJ. Requirement of mitogen-activated protein kinase kinase 3 (MKK3) for tumor necrosis factor-induced cytokine expression. Proc Natl Acad Sci. 1999;96(7):3763–8. https://doi.org/10.1073/pnas.96.7.3763.
Wotton SF, Blyth K, Kilbey A, Jenkins A, Terry A, Bernardin-Fried F, et al. RUNX1 transformation of primary embryonic fibroblasts is revealed in the absence of p53. Oncogene. 2004;23(32):5476–86. https://doi.org/10.1038/sj.onc.1207729.
Cui H, Kong Y, Xu M, Zhang H. Notch3 functions as a tumor suppressor by controlling cellular senescence. Cancer Res. 2013;73(11):3451–9. https://doi.org/10.1158/0008-5472.CAN-12-3902.
Chatsirisupachai K, Palmer D, Ferreira S, de Magalhaes JP. A human tissue-specific transcriptomic analysis reveals a complex relationship between aging, cancer, and cellular senescence. Aging Cell. 2019;18:e13041.
Sturmlechner I, Zhang C, Sine CC, van Deursen EJ, Jeganathan KB, Hamada N, et al. p21 produces a bioactive secretome that places stressed cells under immunosurveillance. Science. 2021;374:eabb3420.
Lee BY, Han JA, Im JS, Morrone A, Johung K, Goodwin EC, et al. Senescence-associated beta-galactosidase is lysosomal beta-galactosidase. Aging Cell. 2006;5(2):187–95. https://doi.org/10.1111/j.1474-9726.2006.00199.x.
Cho SJ, Rossi A, Jung YS, Yan W, Liu G, Zhang J, et al. Ninjurin1, a target of p53, regulates p53 expression and p53-dependent cell survival, senescence, and radiation-induced mortality. Proc Natl Acad Sci U S A. 2013;110(23):9362–7. https://doi.org/10.1073/pnas.1221242110.
Zemskova M, Lilly MB, Lin YW, Song JH, Kraft AS. p53-dependent induction of prostate cancer cell senescence by the PIM1 protein kinase. Mol Cancer Res. 2010;8(8):1126–41. https://doi.org/10.1158/1541-7786.MCR-10-0174.
Musi N, Valentine JM, Sickora KR, Baeuerle E, Thompson CS, Shen Q, et al. Tau protein aggregation is associated with cellular senescence in the brain. Aging Cell. 2018;17(6):e12840. https://doi.org/10.1111/acel.12840.
Burd CE, Sorrentino JA, Clark KS, Darr DB, Krishnamurthy J, Deal AM, et al. Monitoring tumorigenesis and senescence in vivo with a p16(INK4a)-luciferase model. Cell. 2013;152(1-2):340–51. https://doi.org/10.1016/j.cell.2012.12.010.
Zezula J, Casaccia-Bonnefil P, Ezhevsky SA, Osterhout DJ, Levine JM, Dowdy SF, et al. p21cip1 is required for the differentiation of oligodendrocytes independently of cell cycle withdrawal. EMBO Rep. 2001;2(1):27–34. https://doi.org/10.1093/embo-reports/kve008.
Munoz-Espin D, Canamero M, Maraver A, Gomez-Lopez G, Contreras J, Murillo-Cuesta S, et al. Programmed cell senescence during mammalian embryonic development. Cell. 2013;155(5):1104–18. https://doi.org/10.1016/j.cell.2013.10.019.
Storer M, Mas A, Robert-Moreno A, Pecoraro M, Ortells MC, Di Giacomo V, et al. Senescence is a developmental mechanism that contributes to embryonic growth and patterning. Cell. 2013;155(5):1119–30. https://doi.org/10.1016/j.cell.2013.10.041.
Minamino T, Miyauchi H, Yoshida T, Ishida Y, Yoshida H, Komuro I. Endothelial cell senescence in human atherosclerosis: role of telomere in endothelial dysfunction. Circulation. 2002;105(13):1541–4. https://doi.org/10.1161/01.CIR.0000013836.85741.17.
Schmid N, Flenkenthaler F, Stockl JB, Dietrich KG, Kohn FM, Schwarzer JU, et al. Insights into replicative senescence of human testicular peritubular cells. Sci Rep. 2019;9(1):15052. https://doi.org/10.1038/s41598-019-51380-w.
Lee KE, Bar-Sagi D. Oncogenic KRas suppresses inflammation-associated senescence of pancreatic ductal cells. Cancer Cell. 2010;18(5):448–58. https://doi.org/10.1016/j.ccr.2010.10.020.
Bryant AG, Hu M, Carlyle BC, Arnold SE, Frosch MP, Das S, et al. Cerebrovascular senescence is associated with tau pathology in Alzheimer’s disease. Front Neurol. 2020;11:575953. https://doi.org/10.3389/fneur.2020.575953.
Bussian TJ, Aziz A, Meyer CF, Swenson BL, van Deursen JM, Baker DJ. Clearance of senescent glial cells prevents tau-dependent pathology and cognitive decline. Nature. 2018;562(7728):578–82. https://doi.org/10.1038/s41586-018-0543-y.
Bhat R, Crowe EP, Bitto A, Moh M, Katsetos CD, Garcia FU, et al. Astrocyte senescence as a component of Alzheimer's disease. PLoS One. 2012;7(9):e45069. https://doi.org/10.1371/journal.pone.0045069.
Nguyen QH, Pervolarakis N, Nee K, Kessenbrock K. Experimental considerations for single-cell RNA sequencing approaches. Front Cell Dev Biol. 2018;6:108. https://doi.org/10.3389/fcell.2018.00108.