Increase of cell surface vimentin is associated with vimentin network disruption and subsequent stress-induced premature senescence in human chondrocytes

  1. Jana Riegger  Is a corresponding author
  2. Rolf E Brenner
  1. University of Ulm, Germany

Abstract

Accumulation of dysfunctional chondrocytes has detrimental consequences on the cartilage homeostasis and is thus thought to play a crucial role during the pathogenesis of osteoarthritis (OA). However, the underlying mechanisms of phenotypical alteration in chondrocytes are incompletely understood. Here, we provide evidence that disruption of the intracellular vimentin network and consequent phenotypical alteration in human chondrocytes results in an externalization of the intermediate filament. The presence of so-called cell surface vimentin (CSV) on chondrocytes was associated with the severity of tissue degeneration in clinical OA samples and was enhanced after mechanical injury of cartilage tissue. By means of a doxorubicine-based in vitro model of stress-induced premature senescence (SIPS), we could confirm the connection between cellular senescence and amount of CSV. Although siRNA-mediated silencing of CDKN2A clearly reduced the senescent phenotype as well as CSV levels of human chondrocytes, cellular senescence could not be completely reversed. Interestingly, knockdown of vimentin resulted in a SIPS-like phenotype and consequently increased CSV. Therefore, we concluded that the integrity of the intracellular vimentin network is crucial to maintain cellular function in chondrocytes. This assumption could be confirmed by chemically-induced collapse of the vimentin network, which resulted in cellular stress and enhanced CSV expression. Regarding its biological function, CSV was found to be associated with enhanced chondrocyte adhesion and plasticity. While osteogenic capacities seemed to be enhanced in chondrocytes expressing high levels of CSV, the chondrogenic potential was clearly compromised. Overall, our study reinforces the importance of the vimentin network in maintenance of the chondrogenic phenotype and introduces CSV as a novel membrane-bound marker of dysfunctional chondrocytes.

Data availability

All data generated or analysed during this study are included in the manuscript and supporting file.

Article and author information

Author details

  1. Jana Riegger

    Department of Orthopedics, University of Ulm, Ulm, Germany
    For correspondence
    jana.riegger@uni-ulm.de
    Competing interests
    The authors declare that no competing interests exist.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0003-0048-5047
  2. Rolf E Brenner

    Department of Orthopedics, University of Ulm, Ulm, Germany
    Competing interests
    The authors declare that no competing interests exist.

Funding

European Social Fund

  • Jana Riegger

Ministry of Science, Research, and Arts Baden-Württemberg

  • Jana Riegger

University of Ulm

  • Jana Riegger

The funders had no role in study design, data collection and interpretation, or the decision to submit the work for publication.

Reviewing Editor

  1. Di Chen, Chinese Academy of Sciences, China

Version history

  1. Preprint posted: May 9, 2023 (view preprint)
  2. Received: July 30, 2023
  3. Accepted: October 4, 2023
  4. Accepted Manuscript published: October 19, 2023 (version 1)
  5. Version of Record published: November 2, 2023 (version 2)

Copyright

© 2023, Riegger & Brenner

This article is distributed under the terms of the Creative Commons Attribution License permitting unrestricted use and redistribution provided that the original author and source are credited.

Metrics

  • 639
    views
  • 144
    downloads
  • 3
    citations

Views, downloads and citations are aggregated across all versions of this paper published by eLife.

Download links

A two-part list of links to download the article, or parts of the article, in various formats.

Downloads (link to download the article as PDF)

Open citations (links to open the citations from this article in various online reference manager services)

Cite this article (links to download the citations from this article in formats compatible with various reference manager tools)

  1. Jana Riegger
  2. Rolf E Brenner
(2023)
Increase of cell surface vimentin is associated with vimentin network disruption and subsequent stress-induced premature senescence in human chondrocytes
eLife 12:e91453.
https://doi.org/10.7554/eLife.91453

Share this article

https://doi.org/10.7554/eLife.91453

Further reading

    1. Cell Biology
    2. Developmental Biology
    Corey D Holman, Alexander P Sakers ... Patrick Seale
    Research Article

    The energy-burning capability of beige adipose tissue is a potential therapeutic tool for reducing obesity and metabolic disease, but this capacity is decreased by aging. Here, we evaluate the impact of aging on the profile and activity of adipocyte stem and progenitor cells (ASPCs) and adipocytes during the beiging process in mice. We found that aging increases the expression of Cd9 and other fibro-inflammatory genes in fibroblastic ASPCs and blocks their differentiation into beige adipocytes. Fibroblastic ASPC populations from young and aged mice were equally competent for beige differentiation in vitro, suggesting that environmental factors suppress adipogenesis in vivo. Examination of adipocytes by single nucleus RNA-sequencing identified compositional and transcriptional differences in adipocyte populations with aging and cold exposure. Notably, cold exposure induced an adipocyte population expressing high levels of de novo lipogenesis (DNL) genes, and this response was severely blunted in aged animals. We further identified Npr3, which encodes the natriuretic peptide clearance receptor, as a marker gene for a subset of white adipocytes and an aging-upregulated gene in adipocytes. In summary, this study indicates that aging blocks beige adipogenesis and dysregulates adipocyte responses to cold exposure and provides a resource for identifying cold and aging-regulated pathways in adipose tissue.

    1. Cell Biology
    Tongtong Ma, Ruimin Ren ... Heng Wang
    Research Article

    Current studies on cultured meat mainly focus on the muscle tissue reconstruction in vitro, but lack the formation of intramuscular fat, which is a crucial factor in determining taste, texture, and nutritional contents. Therefore, incorporating fat into cultured meat is of superior value. In this study, we employed the myogenic/lipogenic transdifferentiation of chicken fibroblasts in 3D to produce muscle mass and deposit fat into the same cells without the co-culture or mixture of different cells or fat substances. The immortalized chicken embryonic fibroblasts were implanted into the hydrogel scaffold, and the cell proliferation and myogenic transdifferentiation were conducted in 3D to produce the whole-cut meat mimics. Compared to 2D, cells grown in 3D matrix showed elevated myogenesis and collagen production. We further induced fat deposition in the transdifferentiated muscle cells and the triglyceride content could be manipulated to match and exceed the levels of chicken meat. The gene expression analysis indicated that both lineage-specific and multifunctional signalings could contribute to the generation of muscle/fat matrix. Overall, we were able to precisely modulate muscle, fat, and extracellular matrix contents according to balanced or specialized meat preferences. These findings provide new avenues for customized cultured meat production with desired intramuscular fat contents that can be tailored to meet the diverse demands of consumers.