AI Chat Paper
Note: Please note that the following content is generated by AMiner AI. SciOpen does not take any responsibility related to this content.
{{lang === 'zh_CN' ? '文章概述' : 'Summary'}}
{{lang === 'en_US' ? '中' : 'Eng'}}
Chat more with AI
PDF (6.1 MB)
Collect
Submit Manuscript AI Chat Paper
Show Outline
Outline
Show full outline
Hide outline
Outline
Show full outline
Hide outline
Original Article | Open Access

Prim-O-glucosylcimifugin ameliorates aging-impaired endogenous tendon regeneration by rejuvenating senescent tendon stem/progenitor cells

Yu Wang1Shanshan Jin1Dan Luo2Danqing He1Min Yu1Lisha Zhu1Zixin Li1Liyuan Chen1Chengye Ding1Xiaolan Wu1Tianhao Wu1Weiran Huang3Xuelin Zhao4Meng Xu4Zhengwei Xie3,( )Yan Liu1, ( )
Laboratory of Biomimetic Nanomaterials, Department of Orthodontics, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials & Translational Research Center for Orocraniofacial Stem Cells and Systemic Health, Beijing 100081, China
CAS Center for Excellence in Nanoscience, Beijing Key Laboratory of Micro-nano Energy and Sensor, Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing 101400, China
Peking University International Cancer Institute, Health Science Center, Peking University, Beijing 100083, China
Department of Orthopedics, the Fourth Medical Center of PLA General Hospital, Beijing 100048, China

These authors jointly supervised this work: Zhengwei Xie, Yan Liu

Show Author Information

Abstract

Adult tendon stem/progenitor cells (TSPCs) are essential for tendon maintenance, regeneration, and repair, yet they become susceptible to senescence with age, impairing the self-healing capacity of tendons. In this study, we employ a recently developed deep-learning-based efficacy prediction system to screen potential stemness-promoting and senescence-inhibiting drugs from natural products using the transcriptional signatures of stemness. The top-ranked candidate, prim-O-glucosylcimifugin (POG), a saposhnikovia root extract, could ameliorate TPSC senescent phenotypes caused by long-term passage and natural aging in rats and humans, as well as restore the self-renewal and proliferative capacities and tenogenic potential of aged TSPCs. In vivo, the systematic administration of POG or the local delivery of POG nanoparticles functionally rescued endogenous tendon regeneration and repair in aged rats to levels similar to those of normal animals. Mechanistically, POG protects TSPCs against functional impairment during both passage-induced and natural aging by simultaneously suppressing nuclear factor-κB and decreasing mTOR signaling with the induction of autophagy. Thus, the strategy of pharmacological intervention with the deep learning-predicted compound POG could rejuvenate aged TSPCs and improve the regenerative capacity of aged tendons.

References

1

Nourissat, G., Berenbaum, F. & Duprez, D. Tendon injury: from biology to tendon repair. Nat. Rev. Rheumatol. 11, 223–233 (2015).

2

LaPrade, C. M. et al. Return-to-play and performance after operative treatment of Achilles tendon rupture in elite male athletes: a scoping review. Br. J. Sports Med. 56, 515–520 (2022).

3

Teunis, T., Lubberts, B., Reilly, B. T. & Ring, D. A systematic review and pooled analysis of the prevalence of rotator cuff disease with increasing age. J. Shoulder Elb. Surg. 23, 1913–1921 (2014).

4

Lui, P. P. Y. & Wong, C. M. Biology of tendon stem cells and tendon in aging. Front. Genet. 10, 1338 (2019).

5

Svensson, R. B., Heinemeier, K. M., Couppé, C., Kjaer, M. & Magnusson, S. P. Effect of aging and exercise on the tendon. J. Appl. Physiol. (1985) 121, 1237–1246 (2016).

6

Marqueti, R. C. et al. Effects of aging and resistance training in rat tendon remodeling. FASEB J. 32, 353–368 (2018).

7

Lee, C. H. et al. Harnessing endogenous stem/progenitor cells for tendon regeneration. J. Clin. Investig. 125, 2690–2701 (2015).

8

Zhou, Z. et al. Tendon-derived stem/progenitor cell aging: defective self-renewal and altered fate. Aging Cell 9, 911–915 (2010).

9

Birch, H. L., Peffers, M. J. & Clegg, P. D. Influence of ageing on tendon homeostasis. Adv. Exp. Med. Biol. 920, 247–260 (2016).

10

Chakkalakal, J. V., Jones, K. M., Basson, M. A. & Brack, A. S. The aged niche disrupts muscle stem cell quiescence. Nature 490, 355–360 (2012).

11

Sousa-Victor, P. et al. Geriatric muscle stem cells switch reversible quiescence into senescence. Nature 506, 316–321 (2014).

12

Sousa-Victor, P., García-Prat, L., Serrano, A. L., Perdiguero, E. & Muñoz-Cánoves, P. Muscle stem cell aging: regulation and rejuvenation. Trends Endocrinol. Metab. 26, 287–296 (2015).

13

Sarkar, T. J. et al. Transient non-integrative expression of nuclear reprogramming factors promotes multifaceted amelioration of aging in human cells. Nat. Commun. 11, 1545 (2020).

14

Ocampo, A. et al. In vivo amelioration of age-associated hallmarks by partial reprogramming. Cell 167, 1719–1733.e1712 (2016).

15

Hu, Y. F. et al. Biomaterial-induced conversion of quiescent cardiomyocytes into pacemaker cells in rats. Nat. Biomed. Eng. 6, 421–434 (2022).

16

Zhang, J., Li, B. & Wang, J. H. The role of engineered tendon matrix in the stemness of tendon stem cells in vitro and the promotion of tendon-like tissue formation in vivo. Biomaterials 32, 6972–6981 (2011).

17

Zhang, C. et al. Histone deacetylase inhibitor treated cell sheet from mouse tendon stem/progenitor cells promotes tendon repair. Biomaterials 172, 66–82 (2018).

18

Li, Z. et al. 3D culture supports long-term expansion of mouse and human nephrogenic progenitors. Cell Stem Cell 19, 516–529 (2016).

19

Dou, X. et al. Short-term rapamycin treatment increases ovarian lifespan in young and middle-aged female mice. Aging Cell 16, 825–836 (2017).

20

Guo, J. et al. Aging and aging-related diseases: from molecular mechanisms to interventions and treatments. Signal Transduct. Target. Ther. 7, 391 (2022).

21

Howell, K. et al. Novel model of tendon regeneration reveals distinct cell mechanisms underlying regenerative and fibrotic tendon healing. Sci. Rep. 7, 45238 (2017).

22

Wang, Y. et al. Functional regeneration and repair of tendons using biomimetic scaffolds loaded with recombinant periostin. Nat. Commun. 12, 1293 (2021).

23

Zhu, J. et al. Prediction of drug efficacy from transcriptional profiles with deep learning. Nat. Biotechnol. 39, 1444–1452 (2021).

24

Zhou, J. et al. Prim-O-glucosylcimifugin attenuates lipopolysaccharideinduced inflammatory response in RAW 264.7 macrophages. Pharmacogn. Mag. 13, 378–384 (2017).

25

Gao, W. et al. Prim-O-glucosylcimifugin enhances the antitumour effect of PD-1 inhibition by targeting myeloid-derived suppressor cells. J. Immunother. Cancer 7, 231 (2019).

26

Wang, W. et al. A genome-wide CRISPR-based screen identifies KAT7 as a driver of cellular senescence. Sci. Transl. Med. 13, eabd2655 (2021).

27

Deng, P. et al. Loss of KDM4B exacerbates bone-fat imbalance and mesenchymal stromal cell exhaustion in skeletal aging. Cell Stem Cell 28, 1057–1073.e1057 (2021).

28

Liao, N. et al. Antioxidants inhibit cell senescence and preserve stemness of adipose tissue-derived stem cells by reducing ROS generation during long-term in vitro expansion. Stem Cell Res. Ther. 10, 306 (2019).

29

Yin, Y., Liu, K. & Li, G. Protective effect of Prim-O-Glucosylcimifugin on ulcerative colitis and its mechanism. Front. Pharmacol. 13, 882924 (2022).

30

Kessler, M. et al. Chronic Chlamydia infection in human organoids increases stemness and promotes age-dependent CpG methylation. Nat. Commun. 10, 1194 (2019).

31

Kessler, M. et al. The Notch and Wnt pathways regulate stemness and differentiation in human fallopian tube organoids. Nat. Commun. 6, 8989 (2015).

32

Castilho, R. M., Squarize, C. H., Chodosh, L. A., Williams, B. O. & Gutkind, J. S. mTOR mediates Wnt-induced epidermal stem cell exhaustion and aging. Cell Stem Cell 5, 279–289 (2009).

33

Chien, Y. et al. Control of the senescence-associated secretory phenotype by NF-κB promotes senescence and enhances chemosensitivity. Genes Dev. 25, 2125–2136 (2011).

34

Chen, Y. et al. Targeted pathological collagen delivery of sustained-release rapamycin to prevent heterotopic ossification. Sci. Adv. 6, eaay9526 (2020).

35

Abraham, A. C. et al. Targeting the NF-κB signaling pathway in chronic tendon disease. Sci. Transl. Med. 11, eaav4319 (2019).

36

Wang, C. et al. Inhibition of IKKβ/NF-κB signaling facilitates tendinopathy healing by rejuvenating inflamm-aging induced tendon-derived stem/progenitor cell senescence. Mol. Ther. Nucleic Acids 27, 562–576 (2022).

37

Chen, S. et al. RelA/p65 inhibition prevents tendon adhesion by modulating inflammation, cell proliferation, and apoptosis. Cell Death Dis. 8, e2710 (2017).

38

Yu, B. et al. Wnt4 signaling prevents skeletal aging and inflammation by inhibiting nuclear factor-κB. Nat. Med. 20, 1009–1017 (2014).

39

Li, W. et al. Subcutaneously engineered autologous extracellular matrix scaffolds with aligned microchannels for enhanced tendon regeneration: aligned microchannel scaffolds for tendon repair. Biomaterials 224, 119488 (2019).

40

Best, K. T. et al. NF-κB activation persists into the remodeling phase of tendon healing and promotes myofibroblast survival. Sci. Signal 13, eabb7209 (2020).

41

Chang, J. et al. NF-κB inhibits osteogenic differentiation of mesenchymal stem cells by promoting β-catenin degradation. Proc. Natl. Acad. Sci. USA 110, 9469–9474 (2013).

42

Jurk, D. et al. Chronic inflammation induces telomere dysfunction and accelerates ageing in mice. Nat. Commun. 2, 4172 (2014).

43

Jeon, O. H. et al. Local clearance of senescent cells attenuates the development of post-traumatic osteoarthritis and creates a pro-regenerative environment. Nat. Med. 23, 775–781 (2017).

44

Hou, Y. et al. NAD(+) supplementation reduces neuroinflammation and cell senescence in a transgenic mouse model of Alzheimer’s disease via cGAS-STING. Proc. Natl. Acad. Sci. USA 118, e2011226118 (2021).

45

Iglesias, M. et al. Downregulation of mTOR signaling increases stem cell population telomere length during starvation of immortal planarians. Stem Cell Rep. 13, 405–418 (2019).

46

Wang, S. et al. Transient activation of autophagy via Sox2-mediated suppression of mTOR is an important early step in reprogramming to pluripotency. Cell Stem Cell 13, 617–625 (2013).

47

García-Prat, L. et al. Autophagy maintains stemness by preventing senescence. Nature 529, 37–42 (2016).

48

Leidal, A. M., Levine, B. & Debnath, J. Autophagy and the cell biology of age-related disease. Nat. Cell Biol. 20, 1338–1348 (2018).

49

Park, J. T., Lee, Y. S., Cho, K. A. & Park, S. C. Adjustment of the lysosomal-mitochondrial axis for control of cellular senescence. Ageing Res. Rev. 47, 176–182 (2018).

50

Young, A. R. et al. Autophagy mediates the mitotic senescence transition. Genes Dev. 23, 798–803 (2009).

51

Leeman, D. S. et al. Lysosome activation clears aggregates and enhances quiescent neural stem cell activation during aging. Science 359, 1277–1283 (2018).

52

Debacq-Chainiaux, F., Erusalimsky, J. D., Campisi, J. & Toussaint, O. Protocols to detect senescence-associated beta-galactosidase (SA-betagal) activity, a biomarker of senescent cells in culture and in vivo. Nat. Protoc. 4, 1798–1806 (2009).

53

Aman, Y. et al. Autophagy in healthy aging and disease. Nat. Aging 1, 634–650 (2021).

54

Carnio, S. et al. Autophagy impairment in muscle induces neuromuscular junction degeneration and precocious aging. Cell Rep. 8, 1509–1521 (2014).

55

Kohler, J. et al. Uncovering the cellular and molecular changes in tendon stem/progenitor cells attributed to tendon aging and degeneration. Aging Cell 12, 988–999 (2013).

56

Wang, Z. et al. Functional regeneration of tendons using scaffolds with physical anisotropy engineered via microarchitectural manipulation. Sci. Adv. 4, eaat4537 (2018).

57

Howell, K. L. et al. Macrophage depletion impairs neonatal tendon regeneration. FASEB J. 35, e21618 (2021).

58

Walia, B., Li, T. M., Crosio, G., Montero, A. M. & Huang, A. H. Axin2-lineage cells contribute to neonatal tendon regeneration. Connect. Tissue Res. 63, 530–543 (2022).

59

Murrell, G. A. et al. The Achilles functional index. J. Orthop. Res. 10, 398–404 (1992).

60

Yang, S. et al. Oriented collagen fiber membranes formed through counter-rotating extrusion and their application in tendon regeneration. Biomaterials 207, 61–75 (2019).

61

Yin, Z. et al. Single-cell analysis reveals a nestin(+) tendon stem/progenitor cell population with strong tenogenic potentiality. Sci. Adv. 2, e1600874 (2016).

62

Saraswat, K. & Rizvi, S. I. Novel strategies for anti-aging drug discovery. Expert Opin. Drug Discov. 12, 955–966 (2017).

63

Conboy, I. M., Conboy, M. J. & Rebo, J. Systemic problems: a perspective on stem cell aging and rejuvenation. Aging (Albany NY) 7, 754–765 (2015).

64

Wang, R., Wang, Y., Zhu, L., Liu, Y. & Li, W. Epigenetic regulation in mesenchymal stem cell aging and differentiation and osteoporosis. Stem Cells Int. 2020, 8836258 (2020).

65

Xie, C. et al. Amelioration of Alzheimer’s disease pathology by mitophagy inducers identified via machine learning and a cross-species workflow. Nat. Biomed. Eng. 6, 76–93 (2022).

66

Stokes, J. M. et al. A deep learning approach to antibiotic discovery. Cell 180, 688–702.e613 (2020).

67

Chou, L. Y., Ho, C. T. & Hung, S. C. Paracrine senescence of mesenchymal stromalcells involves inflammatory cytokines and the NF-κB pathway. Cells 11, 3324 (2022).

68

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. 22, 75–95 (2021).

69

Zhang, X. et al. Characterization of cellular senescence in aging skeletal muscle. Nat. Aging 2, 601–615 (2022).

70

Ji, M. L. et al. Sirt6 attenuates chondrocyte senescence and osteoarthritis progression. Nat. Commun. 13, 7658 (2022).

71

Liu, F., Wu, S., Ren, H. & Gu, J. Klotho suppresses RIG-I-mediated senescence-associated inflammation. Nat. Cell Biol. 13, 254–262 (2011).

72

Li, Y. et al. Melatonin promotes the restoration of bone defects via enhancement of miR-335-5p combined with inhibition of TNFα/NF-κB signaling. FASEB J. 37, e22711 (2023).

73

Wang, N. et al. TNF-α-induced NF-κB activation upregulates microRNA-150-3p and inhibits osteogenesis of mesenchymal stem cells by targeting β-catenin. Open Biol. 6, 150258 (2016).

74

An, S. et al. Inhibition of 3-phosphoinositide-dependent protein kinase 1 (PDK1) can revert cellular senescence in human dermal fibroblasts. Proc. Natl. Acad. Sci. USA 117, 31535–31546 (2020).

75

Graça, A. L., Gomez-Florit, M., Gomes, M. E. & Docheva, D. Tendon aging. Subcell Biochem. 103, 121–147 (2023).

76

Muscat, S., Nichols, A. E. C., Gira, E. & Loiselle, A. E. CCR2 is expressed by tendon resident macrophage and T cells, while CCR2 deficiency impairs tendon healing via blunted involvement of tendon-resident and circulating monocytes/macrophages. FASEB J. 36, e22607 (2022).

77

Xu, H. et al. Bioactive glass-elicited stem cell-derived extracellular vesicles regulate M2 macrophage polarization and angiogenesis to improve tendon regeneration and functional recovery. Biomaterials 294, 121998 (2023).

78

Wang, Y. et al. Aspirin inhibits adipogenesis of tendon stem cells and lipids accumulation in rat injury tendon through regulating PTEN/PI3K/AKT signalling. J. Cell Mol. Med. 23, 7535–7544 (2019).

79

Li, Y. et al. Comparative pharmacokinetics of prim-O-glucosylcimifugin and cimifugin by liquid chromatography-mass spectrometry after oral administration of Radix Saposhnikoviae extract, cimifugin monomer solution and prim-O-glucosylcimifugin monomer solution to rats. Biomed. Chromatogr. 26, 1234–1240 (2012).

80

Gu, Y., Piao, X. & Zhu, D. Simultaneous determination of calycosin, prim-O-glucosylcimifugin, and paeoniflorin in rat plasma by HPLC-MS/MS: application in the pharmacokinetic analysis of HQCF. J. Int. Med. Res. 48, 300060520972902 (2020).

Bone Research
Article number: 54
Cite this article:
Wang Y, Jin S, Luo D, et al. Prim-O-glucosylcimifugin ameliorates aging-impaired endogenous tendon regeneration by rejuvenating senescent tendon stem/progenitor cells. Bone Research, 2023, 11: 54. https://doi.org/10.1038/s41413-023-00288-3

224

Views

1

Downloads

1

Crossref

0

Web of Science

0

Scopus

Altmetrics

Received: 01 July 2023
Revised: 16 August 2023
Accepted: 17 August 2023
Published: 23 October 2023
© The Author(s) 2023

This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons license, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons license and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/.

Return