Migrasomes trigger innate immune activation and mediate transmission of senescence signals across human cells
Xiaoqian Liu, Haifeng Jiao, Baohu Zhang, Sheng Zhang, Kaowen Yan, Jing Qu, Weiqi Zhang, Li Yu, Guang-Hui Liu
Migrasomes trigger innate immune activation and mediate transmission of senescence signals across human cells
Aging is a complex and heterogeneous process, raising important questions about how aging is differently impacted by underlying genetics and external factors. Recently, migrasomes, newly discovered organelles, have been identified to play important roles in various physiological and pathological processes by facilitating cell-to-cell communication. Thus far, their involvement in cellular senescence and aging remains largely unexplored. In this study, we aimed to investigate how migrasomes impact on cellular aging by leveraging multiple cellular senescence models, including replicatively senescent (RS), pathologically senescent and stress-induced senescent human mesenchymal stem cells (hMSCs), as well as RS human primary fibroblasts. In all cellular aging models, we detected an enhanced formation of migrasomes. Notably, migrasomes in senescent cells exhibited an accumulation of numerous aging hallmarks, such as dysfunctional mitochondria, endogenous retroviruses, and senescence-associated pro-inflammatory cytokines. Furthermore, we discovered that migrasomes derived from senescent cells can be taken up by young cells, thereby transferring aging signals and subsequently causing premature senescence phenotypes in recipient cells. Mechanistically, we found that treatment with migrasomes derived from senescent cells activated the innate immune response. Thus, our study sheds light on a pivotal role of migrasomes in mediating the contagiousness of aging.
migrasomes / cellular senescence / endogenous retrovirus / senescence-associated secretory phenotype / aging
[1] |
Aging Biomarker Consortium; Bao H, Cao J, Chen M, et al. Biomarkers of aging. Sci China Life Sci 2023;66:893–1066.
CrossRef
Google scholar
|
[2] |
Guo J, Huang X, Dou L, et al. Aging and aging-related diseases: from molecular mechanisms to interventions and treatments. Signal Transduct Target Ther 2022;7:391.
CrossRef
Google scholar
|
[3] |
Cai Y, Song W, Li J, et al. The landscape of aging. Sci China Life Sci 2022;65:2354–454.
CrossRef
Google scholar
|
[4] |
Aging Biomarker Consortium, Jia Y-J, Yu-Juan Jia, et al. A framework of biomarkers for brain aging: a consensus statement by the Aging Biomarker Consortium. Life Med 2023;2:lnad017.
CrossRef
Google scholar
|
[5] |
Cai Y, Ji Z, Wang S, et al. Genetic enhancement: an avenue to combat aging-related diseases. Life Med 2022;1:307–18.
CrossRef
Google scholar
|
[6] |
Lopez-Otin C, Blasco MA, Partridge L, et al. Hallmarks of aging: An expanding universe. Cell 2023;186:243–78.
CrossRef
Google scholar
|
[7] |
Vu R, Jin S, Sun P, et al. Wound healing in aged skin exhibits systems-level alterations in cellular composition and cell-cell communication. Cell Rep 2022;40:111155.
CrossRef
Google scholar
|
[8] |
Ma S, Wang S, Ye Y, et al. Heterochronic parabiosis induces stem cell revitalization and systemic rejuvenation across aged tissues. Cell Stem Cell 2022;29:990–1005.e10.
CrossRef
Google scholar
|
[9] |
Liu X, Liu Z, Wu Z, et al. Resurrection of endogenous retroviruses during aging reinforces senescence. Cell 2023;186:287–304.e26.
CrossRef
Google scholar
|
[10] |
Zhang H, Li J, Yu Y, et al. Nuclear lamina erosion-induced resurrection of endogenous retroviruses underlies neuronal aging. Cell Rep 2023;42:112593.
CrossRef
Google scholar
|
[11] |
Rando TA, Wyss-Coray T. Asynchronous, contagious and digital aging. Nat Aging 2021;1:29–35.
CrossRef
Google scholar
|
[12] |
Ma L, Li Y, Peng J, et al. Discovery of the migrasome, an organelle mediating release of cytoplasmic contents during cell migration. Cell Res 2015;25:24–38.
CrossRef
Google scholar
|
[13] |
Yu S, Yu L. Migrasome biogenesis and functions. FEBS J 2022;289:7246–54.
CrossRef
Google scholar
|
[14] |
Jiang D, Li Y, Yu L. Detection, purification, characterization, and manipulation of migrasomes. Curr Protoc 2023;3:e856.
CrossRef
Google scholar
|
[15] |
Dharan R, Huang Y, Cheppali SK, et al. Tetraspanin 4 stabilizes membrane swellings and facilitates their maturation into migrasomes. Nat Commun 2023;14:1037.
CrossRef
Google scholar
|
[16] |
Dharan R, Goren S, Cheppali SK, et al. Transmembrane proteins tetraspanin 4 and CD9 sense membrane curvature. Proc Natl Acad Sci USA 2022;119:e2208993119.
CrossRef
Google scholar
|
[17] |
Fan C, Shi X, Zhao K, et al. Cell migration orchestrates migrasome formation by shaping retraction fibers. J Cell Biol 2022;221:e202109168.
CrossRef
Google scholar
|
[18] |
Zhu M, Zou Q, Huang R, et al. Lateral transfer of mRNA and protein by migrasomes modifies the recipient cells. Cell Res 2021;31:237–40.
CrossRef
Google scholar
|
[19] |
Zhao X, Lei Y, Zheng J, et al. Identification of markers for migrasome detection. Cell Discov 2019;5:27.
CrossRef
Google scholar
|
[20] |
Zhang C, Li T, Yin S, et al. Monocytes deposit migrasomes to promote embryonic angiogenesis. Nat Cell Biol 2022;24:1726–38.
CrossRef
Google scholar
|
[21] |
Jiang D, Jiang Z, Lu D, et al. Migrasomes provide regional cues for organ morphogenesis during zebrafish gastrulation. Nat Cell Biol 2019;21:966–77.
CrossRef
Google scholar
|
[22] |
Qin Y, Yang J, Liang C, et al. Pan-cancer analysis identifies migrasome-related genes as a potential immunotherapeutic target: A bulk omics research and single cell sequencing validation. Front Immunol 2022;13:994828.
CrossRef
Google scholar
|
[23] |
Hu M, Li T, Ma X, et al. Macrophage lineage cells-derived migrasomes activate complement-dependent blood-brain barrier damage in cerebral amyloid angiopathy mouse model. Nat Commun 2023;14:3945.
CrossRef
Google scholar
|
[24] |
Zheng Y, Lang Y, Qi B, et al. TSPAN4 and migrasomes in atherosclerosis regression correlated to myocardial infarction and pan-cancer progression. Cell Adh Migr 2023;17:14–9.
CrossRef
Google scholar
|
[25] |
Wu L, Yang S, Li H, et al. TSPAN4-positive migrasome derived from retinal pigmented epithelium cells contributes to the development of proliferative vitreoretinopathy. J Nanobiotechnol 2022;20:519.
CrossRef
Google scholar
|
[26] |
Wu Z, Zhang W, Song M, et al. Differential stem cell aging kinetics in Hutchinson-Gilford progeria syndrome and Werner syndrome. Protein Cell 2018;9:333–50.
CrossRef
Google scholar
|
[27] |
Chen Y, Li Y, Ma L, et al. Detection of migrasomes. Methods Mol Biol 2018;1749:43–9.
CrossRef
Google scholar
|
[28] |
Deniz IA, Karbanová J, Wobus M, et al. Mesenchymal stromal cell-associated migrasomes: a new source of chemoattractant for cells of hematopoietic origin. Cell Commun Signal 2023;21:36.
CrossRef
Google scholar
|
[29] |
Chen L, Ma L, Yu L. WGA is a probe for migrasomes. Cell Discov 2019;5:13.
CrossRef
Google scholar
|
[30] |
Kudlow BA, Kennedy BK, Monnat R Jr. Werner and Hutchinson-Gilford progeria syndromes: mechanistic basis of human progeroid diseases. Nat Rev Mol Cell Biol 2007;8:394–404.
CrossRef
Google scholar
|
[31] |
Liu GH, Barkho BZ, Ruiz S, et al. Recapitulation of premature ageing with iPSCs from Hutchinson-Gilford progeria syndrome. Nature 2011;472:221–5.
CrossRef
Google scholar
|
[32] |
Gonzalo S, Kreienkamp R, Askjaer P. Hutchinson-Gilford Progeria Syndrome: A premature aging disease caused by LMNA gene mutations. Ageing Res Rev 2017;33:18–29.
CrossRef
Google scholar
|
[33] |
Zhang B, Yan H, Liu X, et al. SenoIndex: S100A8/S100A9 as a novel aging biomarker. Life Med 2023;2:lnad022.
CrossRef
Google scholar
|
[34] |
Duan J, Duan J, Zhang Z, et al. Irreversible cellular senescence induced by prolonged exposure to H2O2 involves DNA-damage-and-repair genes and telomere shortening. Int J Biochem Cell Biol 2005;37:1407–20.
CrossRef
Google scholar
|
[35] |
Cheng F, Ji Q, Wang L, et al. Reducing oxidative protein folding alleviates senescence by minimizing ER-to-nucleus H(2) O(2) release. EMBO Rep 2023;24:e56439.
CrossRef
Google scholar
|
[36] |
Jiao H, Jiang D, Hu X, et al. Mitocytosis, a migrasome-mediated mito-chondrial quality-control process. Cell 2021;184:2896–2910.e13.
CrossRef
Google scholar
|
[37] |
Wang C, Yang K, Liu X, et al. MAVS antagonizes human stem cell senescence as a mitochondrial stabilizer. Research 2023;6:0192.
CrossRef
Google scholar
|
[38] |
He Y, Ji Q, Wu Z, et al. 4E-BP1 counteracts human mesenchymal stem cell senescence via maintaining mitochondrial homeostasis. Protein Cell 2023;14:202–16.
CrossRef
Google scholar
|
[39] |
Amorim JA, Coppotelli G, Rolo AP, et al. Mitochondrial and metabolic dysfunction in ageing and age-related diseases. Nat Rev Endocrinol 2022;18:243–58.
CrossRef
Google scholar
|
[40] |
Shpilka T, Haynes CM. The mitochondrial UPR: mechanisms, physiological functions and implications in ageing. Nat Rev Mol Cell Biol 2018;19:109–20.
CrossRef
Google scholar
|
[41] |
Schleiff E, Turnbull JL. Functional and structural properties of the mitochondrial outer membrane receptor Tom20. Biochemistry 1998;37:13043–51.
CrossRef
Google scholar
|
[42] |
Garcia-Montojo M, Doucet-O’Hare T, Henderson L, et al. Human endogenous retrovirus-K (HML-2): a comprehensive review. Crit Rev Microbiol 2018;44:715–38.
CrossRef
Google scholar
|
[43] |
Subramanian RP, Wildschutte JH, Russo C, et al. Identification, characterization, and comparative genomic distribution of the HERV-K (HML-2) group of human endogenous retroviruses. Retrovirology 2011;8:90.
CrossRef
Google scholar
|
[44] |
De Cecco M, Criscione SW, Peckham EJ, et al. Genomes of replicatively senescent cells undergo global epigenetic changes leading to gene silencing and activation of transposable elements. Aging Cell 2013;12:247–56.
CrossRef
Google scholar
|
[45] |
Chen YG, Hur S. Cellular origins of dsRNA, their recognition and consequences. Nat Rev Mol Cell Biol 2022;23:286–301.
CrossRef
Google scholar
|
[46] |
Chiappinelli KB, Strissel PL, Desrichard A, et al. Inhibiting DNA methylation causes an interferon response in cancer via dsRNA including endogenous retroviruses. Cell 2015;162:974–86.
CrossRef
Google scholar
|
[47] |
Lima-Junior DS, Krishnamurthy SR, Bouladoux N, et al. Endogenous retroviruses promote homeostatic and inflammatory responses to the microbiota. Cell 2021;184:3794–3811.e19.
CrossRef
Google scholar
|
[48] |
Canadas I, Thummalapalli R, Kim JW, et al. Tumor innate immunity primed by specific interferon-stimulated endogenous retroviruses. Nat Med 2018;24:1143–50.
CrossRef
Google scholar
|
[49] |
Birch J, Gil J. Senescence and the SASP: many therapeutic avenues. Genes Dev 2020;34:1565–76.
CrossRef
Google scholar
|
[50] |
Lopes-Paciencia S, Saint-Germain E, Rowell M-C, et al. The senescence- associated secretory phenotype and its regulation. Cytokine 2019;117:15–22.
CrossRef
Google scholar
|
[51] |
Li X, Li C, Zhang W, et al. Inflammation and aging: signaling pathways and intervention therapies. Signal Transduct Target Ther 2023;8:239.
CrossRef
Google scholar
|
[52] |
Seth RB, Sun L, Ea C-K, et al. Identification and characterization of MAVS, a mitochondrial antiviral signaling protein that activates NF-kappaB and IRF 3. Cell 2005;122:669–82.
CrossRef
Google scholar
|
[53] |
Schoggins JW, MacDuff DA, Imanaka N, et al. Pan-viral specificity of IFN-induced genes reveals new roles for cGAS in innate immunity. Nature 2014;505:691–5.
CrossRef
Google scholar
|
[54] |
Aging Atlas C. Aging Atlas: a multi-omics database for aging biology. Nucleic Acids Res 2021;49:D825–30.
CrossRef
Google scholar
|
[55] |
Ju Z, Choudhury AR, Rudolph KL. A dual role of p21 in stem cell aging. Ann N Y Acad Sci 2007;1100:333–44.
CrossRef
Google scholar
|
[56] |
Robin JD, Magdinier F. Physiological and pathological aging affects chromatin dynamics, structure and function at the nuclear edge. Front Genet 2016;7:153.
CrossRef
Google scholar
|
[57] |
Pittenger MF, Discher DE, Péault BM, et al. Mesenchymal stem cell perspective: cell biology to clinical progress. NPJ Regen Med 2019;4:22.
CrossRef
Google scholar
|
[58] |
Yan H, Wang R, Ma S, et al. Lineage Landscape: a comprehensive database that records lineage commitment across species. Nucleic Acids Res 2023;51:D1061–6.
CrossRef
Google scholar
|
[59] |
Giroud J, Bouriez I, Paulus H, et al. Exploring the communication of the SASP: dynamic, interactive, and adaptive effects on the microenvironment. Int J Mol Sci 2023;24:10788.
CrossRef
Google scholar
|
[60] |
Wu D, Xu Y, Ding T, et al. Pairing of integrins with ECM proteins determines migrasome formation. Cell Res 2017;27:1397–400.
CrossRef
Google scholar
|
[61] |
Huang Y, Zucker B, Zhang S, et al. Migrasome formation is mediated by assembly of micron-scale tetraspanin macrodomains. Nat Cell Biol 2019;21:991–1002.
CrossRef
Google scholar
|
[62] |
Liang H, Ma X, Zhang Y, et al. The formation of migrasomes is initiated by the assembly of sphingomyelin synthase 2 foci at the leading edge of migrating cells. Nat Cell Biol 2023;25:1173–84.
CrossRef
Google scholar
|
[63] |
Ding T, Ji J, Zhang W, et al. The phosphatidylinositol (4,5)-bisphosphate-Rab35 axis regulates migrasome formation. Cell Res 2023;33:617–27.
CrossRef
Google scholar
|
[64] |
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
|
/
〈 | 〉 |