Abstract
Objective.
The correlation between age and incidence of osteoarthritis (OA) is well known but the causal mechanisms involved are not completely understood. This narrative review summarizes selected key findings from the past 30 years that have elucidated key aspects of the relationship between aging and OA.
Methods.
The peer-reviewed English language literature was searched on PubMed using key words including senescence, aging, cartilage, and osteoarthritis, for original studies and reviews published from 1993-2023 with a major focus on more recent studies. Manuscripts most relevant to aging and OA that examined one or more of the hallmarks of aging were selected for further review.
Results.
All proposed hallmarks of aging have been observed in articular cartilage and some have also been described in other joint tissues. Hallmarks include genomic instability, telomere attrition, epigenetic alterations, loss of proteostasis, deregulated nutrient sensing, mitochondrial dysfunction, cellular senescence, stem cell exhaustion, altered intercellular communication, disabled macroautophagy, chronic inflammation, and dysbiosis. There is evidence that these age-related changes contribute to the development of OA in part by promoting cellular senescence. Senescence may therefore serve as a downstream mediator that connects numerous aging hallmarks to OA, likely through the senescence-associated secretory phenotype (SASP) that is characterized by increased production of proinflammatory cytokines and matrix metalloproteinases.
Conclusions.
Progress over the past 30 years has provided the foundation for emerging therapies, such as senolytics and senomorphics, that hold promise for OA disease modification. Mechanistic studies utilizing physiologically-aged animals and cadaveric human joint tissues will be important for continued progress.
Keywords: Aging, senescence, cartilage, Chondrocyte
Introduction
The increasing incidence of osteoarthritis (OA) with age, coupled with the lack of effective non-surgical treatments to mitigate disease progression, make OA one of the most common causes of chronic pain and disability in older adults1, 2. Similar to other chronic age-related conditions, OA is not an inevitable aspect of “normal” aging. For example, a study of 90-year-old participants in the Leiden 85-plus Study found 16% of the 82 individuals examined did not exhibit radiographic OA in their hands, hips, or knees3. Aging itself does not cause OA, but instead the changes that occur with age in cells, tissues, and organ systems contribute to the development of OA in conjunction with other risk factors such as prior joint injury, obesity, biological sex, and genetics. As an example of these complex interactions, Magnusson and colleagues used a large twin study to illustrate that the genetic contribution to knee replacement risk is consistent from age 50 to death in women, whereas heritability increased in importance for men starting at age 684.
Over the past 30 years since Osteoarthritis and Cartilage was first published, the knowledge gained from studies in the basic science of aging, as well as studies specific to joint tissues, have significantly advanced our understanding of how aging contributes to the development and progression of OA. Thirty years ago, in a review of basic mechanisms of aging and OA written by David Hamerman5, much of the focus was on age-related changes in the extracellular matrix of articular cartilage. As an example, a study published in the first volume of Osteoarthritis and Cartilage in 1993 reported on age-related decreases in the content of aggrecan molecules containing the epidermal growth factor-like (EGF-like) domain6. Amongst the numerous changes to the cartilage extra-cellular matrix, the accumulation of advanced glycation end-products (AGEs) has been one of the most consistent age-related changes noted in cartilage. AGE accumulation alters the biomechanical properties of the tissue7 and is associated with reduced chondrocyte anabolic activity8.
More recent studies have broadened the scope of investigation into age-related changes present in other tissues and cell types of the joint and have incorporated a wide range of tools to investigate causal mechanisms. Over this time frame the terminology describing OA has changed to reflect a more nuanced understanding of the disease and these shifts in language closely parallel the emergence of similar concepts in aging research. For example, “degenerative joint disease” has fallen out of favor because it evokes a primarily passive mechanical concept of “wear and tear” without sufficient recognition of the biological processes that actively drive OA. In the aging field, the “geroscience hypothesis” proposes that there are a common set of active processes underlying most (if not all) age-related diseases that are consistent enough across tissue types and organ systems to be modifiable at an organism-wide scale for improved healthspan9. Similarly, the more consistent use of “osteoarthritis” in place of “osteoarthrosis” emphasizes that inflammation plays an important role in the disease. Heightened baseline inflammation has been recognized as one of the most consistent features of aging and the tight linkage of these processes has been termed “inflammaging”10,11.
In this narrative review, we will examine the intersection of the aging “hallmarks” with features that emerge in joint tissues during OA. Of note, in part due to the widespread use of surgical waste tissue from end-stage OA, it is often challenging to parse whether these hallmarks emerge before initiation of OA or at some point during OA progression. We will then use cellular senescence, a key hallmark of aging that is driven by many of the other hallmarks, as a lens to examine how a better understanding of aging may translate into improved therapies for OA. Senescent cells exhibit the senescence-associated secretory phenotype (SASP). SASP factors include a host of proinflammatory cytokines and matrix degrading enzymes that are found in OA joints and directly contribute to OA pathogenesis. Finally, we will highlight several topics and experimental approaches that are likely to enhance rigor and accelerate research in this field.
Hallmarks of aging and the relation to the development of OA
The original nine hallmarks of aging described in 2013 included genomic instability, telomere attrition, epigenetic alterations, loss of proteostasis, deregulated nutrient sensing, mitochondrial dysfunction, cellular senescence, stem cell exhaustion, and altered intercellular communication12. These were revised in 2022 to add disabled macroautophagy, chronic inflammation, and dysbiosis13. There have been numerous studies published over the past 30 years that support the presence of these hallmarks in aging and/or OA joints. As consistent with the geroscience framework14, we propose that these hallmarks are more than just correlative but instead represent fundamental changes that enable a higher likelihood of OA onset and progression. The various hallmarks do not exist independently but rather are quite interconnected, with senescence likely positioned as a central mediator between processes that lead to initial cellular dysfunction and those that directly lead to OA pathology (Figure 1).
Figure 1.
The contribution of the hallmarks of aging to cellular senescence and the development of osteoarthritis (OA). Changes that occur within joint tissues during aging include the hallmarks of aging that lead to cellular senescence and the senescence-associated secretory phenotype (SASP). SASP factors promote chronic inflammation and degradation of joint tissues resulting in OA. Graphic created with Biorender.com.
Telomere attrition is a classic cause of in vitro senescence. One of the early studies on senescence in human chondrocytes showed that mean telomere length shortens with age and that shortened telomeres correlate positively to senescence-associated beta galactosidase15. A follow-up study demonstrated that telomerase overexpression can help avoid chondrocyte senescence, but only if cell cycle inhibitors are also inactivated16. Genomic instability in terms of mutational burden has not been extensively investigated in joint tissues, but chondrocytes did show higher chromosomal abnormalities with age and in OA tissue as compared to normal, with more abnormalities than peripheral blood leukocytes from the same subjects17. DNA damage burden is related to genomic instability, and both aged cadaveric donors and end-stage OA samples harbor DNA damage at levels high enough to induce senescence in chondrocytes18–20.
Mitochondrial function is critical for healthy aging of joint tissues. Variations in mitochondrial DNA that can affect mitochondrial function have been associated with increased risk of OA. In order to determine a causal link between mitochondrial DNA haplogroups and the development of OA, conplastic mice were generated that had high risk mitochondrial DNA variants swapped with the low risk mitochondrial DNA variants and vice versa21. The mice generated with the low mitochondrial DNA risk variants developed less severe OA using a surgical model (destabilized medial meniscus, DMM) than the mice with the high-risk variants, providing strong evidence that differences in mitochondrial DNA that had been associated with changes in mitochondrial function can affect OA development. A consequence of mitochondrial dysfunction, which occurs in OA chondrocytes22, is increased levels of mitochondrial reactive oxygen species that can contribute to chondrocyte senescence as can other causes of redox imbalance23. Selenoproteins are important intracellular antioxidants and a decrease in their levels seen in mice with genetic deficiency of selenophosphate synthetase 1 resulted in more severe age-related and surgically-induced OA that was associated with chondrocyte DNA damage, senescence, and increased levels of SASP factors24.
Various epigenetic alterations, including those associated with aging, have been observed in OA25. Sirt6 is a nuclear histone deacetylase that also functions in DNA repair. Loss of Sirt6 in mice results in a premature aging phenotype26 while transgenic overexpression of Sirt6 promotes longevity27. Recent studies have shown that Sirt6 loss in chondrocytes results in cell senescence and premature OA in mice28,29. This was associated with increased STAT5 activity, which promotes proinflammatory signaling28, and with decreased activity of the IGF-1-Akt pathway, which reduces anabolic activity29. Age-related epigenetic changes may also interact with other OA risk factors such as genetics, for example by methylating promoters or increasing the accessibility of regulatory regions30.
There is extensive evidence that a decline in autophagy occurs in aged and OA cartilage31. A recent study linked loss of the chaperone protein HSP90A to disrupted proteostasis and disabled macroautophagy32. FOXO proteins are important regulators of autophagy and their deletion in mice results in more severe age-related OA33 while promotion of FOXO activity by TGF-β improves autophagy and reduces the susceptibility of chondrocytes to oxidative stress34. There is also evidence for deregulated nutrient sensing in aging and OA including reduced activity of the nutrient sensor AMPK35. Treatment of mice with metformin activates AMPK and is being studied for its ability to protect from certain age-related diseases in humans36. Metformin reduced the severity of surgically-induced OA in mice37 and could potentially serve as a disease-modifying drug in humans with OA38.
Stem cell exhaustion is another hallmark of aging that may contribute to OA development. There is evidence to suggest that mesenchymal stromal cells can become senescent in OA and contribute to the pro-inflammatory and matrix destructive environment through production of SASP factors39, 40. SASP factors promote chronic inflammation which is another hallmark of aging clearly associated with OA11. Altered intercellular communication is hallmark of aging that has been studied at the systemic level through parabiosis experiments where pairs of young and old mice share circulatory factors through parabiosis surgery. OA was less severe in old mice paired with young mice compared to old mice paired with old mice and the reduction in OA severity was attributed to GDF-11 present in the circulation of the younger mice41. Another example of intercellular communication related to aging and OA is the promotion of cell senescence by extracellular vesicles produced by senescent chondrocytes42. Finally, dysbiosis is one of the new proposed hallmarks of aging based on a growing body of literature demonstrating that alterations in the gut microbiota contribute to aging and age-related conditions13. A role for gut microbial dysbiosis in the development of OA is not clear43 although there is some evidence that increased intestinal permeability resulting in higher systemic endotoxin (lipopolysaccharide) may be involved44.
Cellular senescence and the SASP
A primary beneficial function of senescent cells in tumor prevention is to elicit a robust inflammatory response by the release of SASP factors to clear persistently damaged cells. However, with increased rates of senescence induction and reduced clearance efficiency during aging, the continuous secretion of these same SASP factors becomes detrimental45. There is a very strong correlation between the pro-inflammatory cytokines, chemokines, and matrix degrading enzymes found in the OA joint and those that constitute the SASP23 (Table 1). In addition to cartilage, joint tissues such as synovium46, bone47, and meniscus48 also harbor senescent cells. Approaches such as single-cell RNA-seq have started to unravel the cell types that are responsible for secreting factors into the OA joint49. Overlapping this approach with rigorous definitions of senescence will likely enable a better understanding of the various sources of the SASP in joint tissues.
Table 1.
Selected SASP Factors Relevant to OA*
Group | SASP Factors | Potential Role in OA |
---|---|---|
Pro-inflammatory cytokines | IL-1(α,β), IL-6, IL-7, IL-15, IL-17 | Promote inflammation and expression of matrix degrading enzymes |
Anti-inflammatory cytokines | IL-13 | Reduce inflammation and are protective in OA |
Chemokines | IL-8, GRO(α,β,δ), MIF, MIP(1α,3α), MCP(1,2,3,4), ENA-78, CCL5, SDF1 | Promote OA by attracting inflammatory cells into the joint or through direct action on joint tissue cells |
Growth factors and growth factor regulators | EGF, bFGF, HGF, TGF-β, IGF-2, IGFBP(2,3,4,6), LTBP-1, NGF, VEGF, CTGF, GDF15 | Can promote OA or be protective depending on the cellular context including receptor expression |
Matrix degrading enzymes and inhibitors | MMP(1,3,10,13,14), TIMP(1,2), PAI-1, tPA, uPA, cathepsin(B,D) | Degradative enzymes promote OA while TIMPs and PAI-1 may be protective. |
Matrix proteins | Fibronectin, collagen (VI), tenascin, periostin, biglycan | Degradation and loss of matrix proteins occurs in OA. Periostin and certain matrix fragments, such as fibronectin fragments, can promote OA. |
Other soluble factors | GM-CSF, HMGB-1, PGE2, HSP-90, 14-3-3 protein epsilon | Can promote OA (GM-CSF, HMGB-1, HSP-90, PGE2) or be protective (14-3-3 protein epsilon) |
bFGF, basic fibroblast growth factor; CCL, CC chemokine ligand; CTGF, connective tissue growth factor; EGF, endothelial growth factor; ENA, epithelial neutrophil-activating protein, GDF, growth and differentiation factor; GM-CSF, granulocyte-macrophage colony-stimulating factor; GRO, growth-related oncogene; HGF, hepatocyte growth factor; HMGB, high mobility group box; HSP, heat shock protein; IGF, insulin-like growth factor; IGFBP, IGF binding protein; IL, interleukin; LTBP, latent transforming growth factor binding protein; MCP, membrane cofactor protein; MIF, macrophage inhibitory factor; MIP, macrophage inflammatory protein, MMP, matrix metalloproteinase; NGF, nerve growth factor; PAI, plasminogen activator inhibitor; PGE2, prostaglandin E2; SDF, stromal cell derived factor; t-PA, tissue-type plasminogen activator; TGF, transforming growth factor; TIMP, tissue inhibitor of metalloproteinases; u-PA, urokinase-type plasminogen activator; uPAR, u-PA receptor; VEGF, vascular endothelial growth factor.
Senescent cells are challenging to define, especially in vivo, due to the insufficient specificity of any particular senescence marker50. The overuse of senescence-associated β-galactosidase (SA-β-gal) as a single marker has been a particular problem due to its lack of specificity, particularly in cultured cells51. One approach is to simultaneously analyze a large number of putative senescence markers and identify the sub-populations that are enriched with aging and depleted with the targeted removal of senescent cells; this paradigm was applied to bone and led to the prioritization of p16-positive; Ki-67 negative; and Bcl2-positive cells52. There is evidence that chondrocytes expressing senescence markers likely represent a heterogeneous population in that 5 of the 25 clusters identified by single-cell mass cytometry in OA samples contain p16-positive cells53. A cell surface marker that holds promise for the identification of senescent chondrocytes is dipeptidylpeptidase-4 (CD26) which was associated with increased p16 and production of SASP factors54. The recent “algorithmic assessment” paradigm posits that after an initial characterization, analyzing the SASP is likely to be the most informative for identifying the specific type of senescence present in a given biological sample55.
Another challenge in identifying senescent cells is their dynamic nature and therefore the specific markers that are positive above background levels may change depending on the “stage” or “depth” of senescence. Using both physiological aging and “aging” through repeated passaging or extended time after a senescence-inducing stimulus, it was shown that some features of senescence such as activation of interferon signaling occur relatively late as compared to more commonly used markers such as p2156. The widespread use of acute damage models to induce senescence and short time points (less than a week) to monitor cellular changes may provide a bias towards markers and states of senescence that are somewhat transient. While these model systems are valuable, care must be taken to distinguish an acute-phase response from a phenotype (senescence) that is defined at least in part by its persistence. For example, IL-1 has been used in many studies to stimulate a senescent-like phenotype in chondrocytes but this was found to be less potent as compared to direct DNA damage in causing sustained growth arrest and increased p2157. Given that characterizing senescence under controlled in vitro conditions has limitations, it is no surprise that the complexity of physiological aging is even more challenging. A goal of the Cellular Senescence Network (SenNet) program funded by the US National Institutes of Health is to develop atlases of senescent cells from various tissues and disease states.
Targeting aging as therapeutic strategy for OA
All twelve of the aging hallmarks could be viewed as potential targets for the development of OA therapies, but over the last 10-15 years much of the translational focus has been on senescence. The burgeoning interest in cellular senescence as a phenotype can be traced to the intriguing finding that elimination of these cells in genetically-engineered mouse models can increase longevity and healthspan58. This also supports the notion that the hallmarks of aging drive age-related conditions by promoting cellular senescence. Fortunately, the idea of targeting senescent cells for therapy has spurred more detailed and mechanistic investigations about the phenotype itself as a way to improve the likelihood of clinical success. Identifying small molecule interventions to mimic the results of the engineered mice became a major focus for aging researchers, as well as those seeking novel treatments for age-related diseases such as OA. Proposed senescence-targeting drugs and their molecular mechanisms have recently been reviewed in the context of OA therapeutics59 and so will only be summarized here.
Senolytics are designed to cause selective cell death and remove the cells from tissues where they have accumulated. Senolytics typically inhibit specific cell survival proteins that are upregulated in senescent cells. By killing senescent cells, SASP factor production is reduced which is a major contributor to tissue dysfunction. In pre-clinical studies using the mouse ACLT model, intra-articular (IA) delivery of the senolytic UBX0101, which blocks the interaction of p53 and MDM2 allowing p53 to induce apoptosis, significantly reduced histologic features of OA, as well as pain behavior, in young 10-week-old animals60. This intervention was less effective in 72-week old mice, but significant mitigation of post-traumatic OA was achieved using a combination of IA UBX0101 plus systemic delivery of Navitoclax as a second senolytic61. This latter finding may explain, at least in part, why IA UBX0101 given to older adult humans with symptomatic knee OA in a Phase 2 clinical trial was not effective in reducing pain or improving function after 12 weeks62.
Fisetin is a flavonoid that has antioxidant and anti-inflammatory properties that also appears to have senolytic activity through interference with cell survival pathways63. Fisetin has been examined in preclinical models of osteoporosis and OA64 and was found to reduce the severity of DMM-induced OA in mice65. Fisetin, given as an oral supplement, was recently tested in humans in a phase 1/2 clinical trial for symptomatic knee OA but the results have not yet been reported (ClinicalTrials.Gov NCT04210986).
Rather than killing senescent cells, senomorphics target SASP factors such as cytokines or the pathways that promote SASP factor production such as JAK/STAT signaling or mTOR59. In the mouse anterior ligament transection model, IL-17 was found to contribute to the development of senescence and the SASP61. In that study, intra-articular injection of an IL-17 antibody reduced senescence accompanied by a reduction in the severity of OA, suggesting that IL-17 inhibition could be considered as a senomorphic. Other senomorphics include the repurposed drugs metformin, which activates AMPK, and rapamycin, which targets mTOR. Metformin has been shown in pre-clinical models to reduce OA severity and is beginning to be examined as an OA therapeutic in humans38. A retrospective study of individuals with type II diabetes found that those treated with metformin had a lower risk of incident OA, but not of joint replacement, compared to those treated with a sulfonylurea66.
Because the toxicity of rapamycin limits its usefulness as a senomorphic and could make OA worse67, rapamycin analogs (“rapalogs”) with less toxicity are being developed68. Modulating levels of NAD+ is an intriguing approach for a senomorphic that would inhibit several aging pathways that promote OA (oxidative stress, DNA damage, cell senescence) and would promote activity of the sirtuins, including Sirt669. In an OA pre-clinical model, NAD+ levels were increased by inhibition of CD38, which mediates NAD degradation, resulting in a reduction in OA severity70. As we learn more details of the mechanisms by which aging contributes to OA, additional therapies will be developed that hold promise for reducing OA progression in humans.
Challenges and promising areas for future research
In the 30 years since Osteoarthritis & Cartilage was first published, the global prevalence of osteoarthritis has more than doubled71. This has coincided with an increase in individuals within older age brackets: in 1993 only 3.9% of the population was ≥ 70 years old whereas this increased to 6.4% for 2023 and is estimated to jump to 12.3% in 205372. With these demographic trends, it is imperative that age-related changes are considered across all aspects of OA research. One area that needs particular attention is the more consistent use of aged mice in analysis of genetically-engineered models or therapeutic interventions. Despite the many benefits of joint injury models such as the DMM model, there are numerous examples of divergent findings compared to OA that develops with physiological aging73. One challenge is that the variability of spontaneous OA in aged mice requires large cohorts, which may be accentuated in genetically-mixed backgrounds such as those used to test treatments for healthspan effects in the National Institute on Aging Interventions Testing Program74.
For studies with human joint tissue, increased use of cadaveric samples from wide age ranges are a valuable complement to the more widely accessible waste tissue from joint replacement surgeries. This facilitates the investigation of the potential initiating events that lead to OA by identifying changes that occur with age, such as increased DNA damage20, before any macroscopic tissue damage is present. A challenge is the extensive overlap between SASP factors and the pro-inflammatory mediators and matrix degrading enzymes produced by cells within the OA joint that are not senescent. It will be important to make a distinction between senescent and non-senescent joint tissue cells in order to study the causal mechanisms distinct to the different ways that cells become dysfunctional and contribute to OA.
The role of senescence and SASP factors in mediating OA pain is another promising area for future studies. There is recent evidence that senolytics can reduce pain associated with OA in aged mice75. SASP factors include the chemokine MCP-1 which is a mediator of OA pain in mice through binding to its receptor CCR276,77. It will also be important to determine if targeting senescence locally within the joint will be sufficient for treatment of OA in humans or if systemic targeting will be required. The answer may depend on the OA phenotype. A young to middle-aged adult with post-traumatic OA may only have senescent cells producing SASP factors in the affected joint while older adults with OA may have a greater contribution from senescence in multiple tissues outside of the joint.
While not a major focus of current translational research, the next 30 years could welcome more sophisticated genetic approaches to ameliorate or reverse age-related cellular changes. One example is the burgeoning field of partial reprogramming, which seeks to use temporary expression of rejuvenating factors to jump start the typically dormant regenerative potential of differentiated cells. Overexpression of six transcription factors for three days helped chondrocytes from older end-stage OA patients achieve a more “youthful” phenotype78. Similarly, SOX5 was identified as a factor that overcomes senescence through the use of a CRISPR activation screen, and further experiments demonstrated that lentiviral overexpression of SOX5 in the joints of aged mice increased cartilage volume and decreased the level of catabolic features such as MMP-1379.
With the progress made to date and the promising research focused on causal mechanisms underlying the link between aging and OA, it should not take another 30 years before we see the development of successful therapeutics that will help patients live more active and pain free lives.
Supplementary Material
Role of the funding source
This work was supported by grants from the National Institute on Aging (R01 AG081734 to BD and RO1 AG044034 to RL). The study sponsors had no role in the study design, collection, analysis, and interpretation of data; the writing of the manuscript; or the decision to submit the manuscript for publication.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Competing Interest Statement
The authors declare that they have no competing interests.
References
- 1.Prieto-Alhambra D, Judge A, Javaid MK, Cooper C, Diez-Perez A, Arden NK. Incidence and risk factors for clinically diagnosed knee, hip and hand osteoarthritis: influences of age, gender and osteoarthritis affecting other joints. Ann Rheum Dis 2014; 73: 1659–1664. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Safiri S, Kolahi AA, Cross M, Hill C, Smith E, Carson-Chahhoud K, et al. Prevalence, deaths, and disability-sdjusted life years due to musculoskeletal disorders for 195 countries and territories 1990–2017. Arthritis Rheumatol 2021; 73: 702–714. [DOI] [PubMed] [Google Scholar]
- 3.Goekoop RJ, Kloppenburg M, Kroon HM, Frolich M, Huizinga TW, Westendorp RG, et al. Low innate production of interleukin-1beta and interleukin-6 is associated with the absence of osteoarthritis in old age. Osteoarthritis Cartilage 2010; 18: 942–947. [DOI] [PubMed] [Google Scholar]
- 4.Magnusson K, Turkiewicz A, Englund M. Nature vs nurture in knee osteoarthritis – the importance of age, sex and body mass index. Osteoarthritis and Cartilage 2019; 27: 586–592. [DOI] [PubMed] [Google Scholar]
- 5.Hamerman D Aging and osteoarthritis: basic mechanisms. J Am Geriatr Soc 1993; 41: 760–770. [DOI] [PubMed] [Google Scholar]
- 6.Stanescu V, Chaminade F, Stanescu R. The epidermal growth factor-like domain of the large proteoglycans from articular cartilage (aggrecans). Estimate of content at different ages and in osteoarthritis. Osteoarthritis Cartilage 1993; 1: 179–184. [DOI] [PubMed] [Google Scholar]
- 7.Bank RA, Bayliss MT, Lafeber FP, Maroudas A, Tekoppele JM. Ageing and zonal variation in post-translational modification of collagen in normal human articular cartilage. The age-related increase in non-enzymatic glycation affects biomechanical properties of cartilage. Biochem J 1998; 330 (Pt 1): 345–351. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.DeGroot J, Verzijl N, Bank RA, Lafeber FP, Bijlsma JW, TeKoppele JM. Age-related decrease in proteoglycan synthesis of human articular chondrocytes: the role of nonenzymatic glycation. Arthritis Rheum 1999; 42: 1003–1009. [DOI] [PubMed] [Google Scholar]
- 9.Kennedy BK, Berger SL, Brunet A, Campisi J, Cuervo AM, Epel ES, et al. Geroscience: linking aging to chronic disease. Cell 2014; 159: 709–713. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Franceschi C, Campisi J. Chronic inflammation (inflammaging) and its potential contribution to age-associated diseases. J Gerontol A Biol Sci Med Sci 2014; 69 Suppl 1: S4–9. [DOI] [PubMed] [Google Scholar]
- 11.Greene MA, Loeser RF. Aging-related inflammation in osteoarthritis. Osteoarthritis and Cartilage 2015; 23: 1966–1971. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Lopez-Otin C, Blasco MA, Partridge L, Serrano M, Kroemer G. The hallmarks of aging. Cell 2013; 153: 1194–1217. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.López-Otín C, Blasco MA, Partridge L, Serrano M, Kroemer G. Hallmarks of aging: An expanding universe. Cell 2022; 186: 243–278. [DOI] [PubMed] [Google Scholar]
- 14.Sierra F. The Emergence of Geroscience as an Interdisciplinary Approach to the Enhancement of Health Span and Life Span. Cold Spring Harbor Perspectives in Medicine 2016; 6: a025163. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Martin JA, Buckwalter JA. Telomere erosion and senescence in human articular cartilage chondrocytes. J Gerontol A Biol Sci Med Sci 2001; 56: B172–179. [DOI] [PubMed] [Google Scholar]
- 16.Martin JA, Mitchell CJ, Klingelhutz AJ, Buckwalter JA. Effects of telomerase and viral oncogene expression on the in vitro growth of human chondrocytes. J Gerontol A Biol Sci Med Sci 2002; 57: B48–53. [DOI] [PubMed] [Google Scholar]
- 17.Tamayo M, Mosquera A, Rego I, Blanco FJ, Gosálvez J, Fernández JL. Decreased length of telomeric DNA sequences and increased numerical chromosome aberrations in human osteoarthritic chondrocytes. Mutat Res 2011; 708: 50–58. [DOI] [PubMed] [Google Scholar]
- 18.Rose J, Soder S, Skhirtladze C, Schmitz N, Gebhard PM, Sesselmann S, et al. DNA damage, discoordinated gene expression and cellular senescence in osteoarthritic chondrocytes. Osteoarthritis Cartilage 2012; 20: 1020–1028. [DOI] [PubMed] [Google Scholar]
- 19.Copp ME, Flanders MC, Gagliardi R, Gilbertie JM, Sessions GA, Chubinskaya S, et al. The combination of mitogenic stimulation and DNA damage induces chondrocyte senescence. Osteoarthritis Cartilage 2021; 29: 402–412. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Copp ME, Chubinskaya S, Bracey DN, Shine J, Sessions G, Loeser RF, et al. Comet assay for quantification of the increased DNA damage burden in primary human chondrocytes with aging and osteoarthritis. Aging Cell 2022; 21: e13698. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Scotece M, Vaamonde-Garcia C, Lechuga-Vieco AV, Cortes AC, Gomez MCJ, Filgueira-Fernandez P, et al. mtDNA variability determines spontaneous joint aging damage in a conplastic mouse model. Aging 2022; 14: 5966–5983. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Fernández-Moreno M, Rego-Pãrez I, Blanco FJ. Is osteoarthritis a mitochondrial disease? What is the evidence. Curr Opin Rheumatol 2021; 34: 46–63. [DOI] [PubMed] [Google Scholar]
- 23.Coryell PR, Diekman BO, Loeser RF. Mechanisms and therapeutic implications of cellular senescence in osteoarthritis. Nat Rev Rheumatol 2021; 17: 47–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Kang D, Lee J, Jung J, Carlson BA, Chang MJ, Chang CB, et al. Selenophosphate synthetase 1 deficiency exacerbates osteoarthritis by dysregulating redox homeostasis. Nat Comm 2022; 13779. 10.1038/s41467-022-28385-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.McCulloch K, Litherland GJ, Rai TS. Cellular senescence in osteoarthritis pathology. Aging Cell 2017; 16: 210–218. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Mostoslavsky R, Chua KF, Lombard DB, Pang WW, Fischer MR, Gellon L, et al. Genomic instability and aging-like phenotype in the absence of mammalian SIRT6. Cell 2006; 124: 315–329. [DOI] [PubMed] [Google Scholar]
- 27.Kanfi Y, Naiman S, Amir G, Peshti V, Zinman G, Nahum L, et al. The sirtuin SIRT6 regulates lifespan in male mice. Nature 2012; 483: 218–221. [DOI] [PubMed] [Google Scholar]
- 28.Ji M-L, Jiang H, Li Z, Geng R, Hu JZ, Lin YC, et al. Sirt6 attenuates chondrocyte senescence and osteoarthritis progression. Nat Comm 2022; 13: 7658. 10.1038/s41467-022-35424-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Collins JA, Kim C, Coleman A, Little A, Perez MM, Clarke EJ, et al. Cartilage-specific Sirt6 deficiency represses IGF-1 and enhances osteoarthritis severity in mice. Ann Rheum Dis 2023. (in press). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Richard D, Capellini TD, Diekman BO. Epigenetics as a mediator of genetic risk in osteoarthritis: role during development, homeostasis, aging, and disease progression. Am J Physiol Cell Physiol 2023; 324: C1078–C1088. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Lotz MK, Carames B. Autophagy and cartilage homeostasis mechanisms in joint health, aging and OA. Nat Rev Rheumatol 2011; 7: 579–587. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Lorenzo-Gómez I, Nogueira-Recalde U, García-Domínguez C, Oreiro N, Lotz M, Pinto-Tasende JA, et al. Defective chaperone-mediated autophagy is a hallmark of joint disease in patients with knee osteoarthritis. Osteoarthritis Cartilage 2023; 31: 919–933. [DOI] [PubMed] [Google Scholar]
- 33.Matsuzaki T, Alvarez-Garcia O, Mokuda S, Nagira K, Olmer M, Gamini R, et al. FoxO transcription factors modulate autophagy and proteoglycan 4 in cartilage homeostasis and osteoarthritis. Sci Transl Med 2018; 10: 428. doi: 10.1126/scitranslmed.aan0746. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Kurakazu I, Akasaki Y, Tsushima H, Sueishi T, Toya M, Kuwahara M, et al. TGFβ1 signaling protects chondrocytes against oxidative stress via FOXO1-autophagy axis. Osteoarthritis Cartilage 2021; 29: 1600–1613. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Liu-Bryan R. Inflammation and intracellular metabolism: new targets in OA. Osteoarthritis Cartilage 2015; 23: 1835–1842. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Espinoza SE, Khosla S, Baur JA, de Cabo R, Musi N. Drugs targeting mechanisms of aging to delay age-related disease and promote healthspan: Proceedings of a National Institute on Aging Workshop. J Gerontol A Biol Sci Med Sci 2023; 78: 53–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Li J, Zhang B, Liu WX, Lu K, Pan H, Wang T, et al. Metformin limits osteoarthritis development and progression through activation of AMPK signalling. Ann Rheum Dis 2020; 79: 635–645. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Lim YZ, Wang Y, Estee M, Abidi J, Kumar MU, Hussain SM, et al. Metformin as a potential disease-modifying drug in osteoarthritis: a systematic review of pre-clinical and human studies. Osteoarthritis Cartilage 2022; 30: 1434–1442. [DOI] [PubMed] [Google Scholar]
- 39.Malaise O, Tachikart Y, Constantinides M, Mumme M, Ferreira-Lopez R, Noack S, et al. Mesenchymal stem cell senescence alleviates their intrinsic and seno-suppressive paracrine properties contributing to osteoarthritis development. Aging 2019; 11: 9128–9146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Liu W, Brodsky AS, Feng M, Liu Y, Ding J, Jayasuriya CT, et al. Senescent tissue-resident mesenchymal stromal cells are an internal source of inflammation in human osteoarthritic cartilage. Front Cell Dev Biol 2021; 9: 725071. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Li L, Wei X, Wang D, Lv Z, Geng X, Li P, et al. Positive effects of a young systemic environment and high growth differentiation factor 11 levels on chondrocyte proliferation and cartilage matrix synthesis in old mice. Arthritis Rheumatol 2020; 72: 1123–1133. [DOI] [PubMed] [Google Scholar]
- 42.Jeon OH, Wilson DR, Clement CC, Rathod S, Cherry C, Powell B, et al. Senescence cell-associated extracellular vesicles serve as osteoarthritis disease and therapeutic markers. JCI Insight 2019; 4 DOI 10.1172/jci.insight.125019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Liu S, Li G, Xu H, Wang Q, Wei Y, Yang Q, et al. “Cross-talk” between gut microbiome dysbiosis and osteoarthritis progression: a systematic review. Front Immunol 2023; 14: 1150572. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Loeser RF, Arbeeva L, Kelley K, Fodor AA, Sun S, Ulici V, et al. Association of increased serum lipopolysaccharide, but not microbial dysbiosis, with obesity-related osteoarthritis. Arthritis Rheumatol 2022; 74: 227–236. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Muñoz-Espín D, Serrano M. Cellular senescence: from physiology to pathology. Nature Reviews Molecular Cell Biology 2014; 15: 482–496. [DOI] [PubMed] [Google Scholar]
- 46.Chen X, Gong W, Shao X, Shi T, Zhang L, Dong J, et al. METTL3-mediated m(6)A modification of ATG7 regulates autophagy-GATA4 axis to promote cellular senescence and osteoarthritis progression. Ann Rheum Dis 2021; 81: 85–97. [DOI] [PubMed] [Google Scholar]
- 47.Farr JN, Fraser DG, Wang H, Jaehn K, Ogrodnik MB, Weivoda MM, et al. Identification of senescent cells in the bone microenvironment. J Bone Miner Res 2016; 31: 1920–1929. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Zhang J, Zhu J, Zhao B, Nie D, Wang W, Qi Y, et al. LTF induces senescence and degeneration in the meniscus via the NF-kappaB signaling pathway: A study based on integrated bioinformatics analysis and experimental validation. Front Mol Biosci 2023; 10: 1134253. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Chou CH, Jain V, Gibson J, Attarian DE, Haraden CA, Yohn CB, et al. Synovial cell cross-talk with cartilage plays a major role in the pathogenesis of osteoarthritis. Sci Rep 2020; 10: 10868. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Sharpless NE, Sherr CJ. Forging a signature of in vivo senescence. Nat Rev Cancer 2015; 15: 397–408. [DOI] [PubMed] [Google Scholar]
- 51.Gorgoulis V, Adams PD, Alimonti A, Bennett DC, Bischof O, Bishop C, et al. Cellular senescence: defining a path forward. Cell 2019; 179: 813–827. [DOI] [PubMed] [Google Scholar]
- 52.Doolittle ML, Saul D, Kaur J, Rowsey JL, Vos SJ, Pavelko KD, et al. Multiparametric senescent cell phenotyping reveals targets of senolytic therapy in the aged murine skeleton. Nature Communications 2023; 14: 4587. 10.1038/s41467-023-40393-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Sahu N, Grandi FC, Bhutani N. A single-cell mass cytometry platform to map the effects of preclinical drugs on cartilage homeostasis. JCI Insight 2022; 7: e160702. doi: 10.1172/jci.insight.160702. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Chen YH, Zhang X, Chou CH, Hsueh MF, Attarian D, Li YJ, et al. Dipeptidyl-peptidase 4 (CD26) is associated with chondrocyte senescence and radiographic progression in knee osteoarthritis. Arthritis Rheumatol 2023; 75: 1120–1131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Kohli J, Wang B, Brandenburg SM, Basisty N, Evangelou K, Varela-Eirin M, et al. Algorithmic assessment of cellular senescence in experimental and clinical specimens. Nat Protoc 2021; 16: 2471–2498. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.De Cecco M, Ito T, Petrashen AP, Elias AE, Skvir NJ, Criscione SW, et al. L1 drives IFN in senescent cells and promotes age-associated inflammation. Nature 2019; 566: 73–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Georget M, Defois A, Guiho R, Bon N, Allain S, Boyer C, et al. Development of a DNA damage-induced senescence model in osteoarthritic chondrocytes. Aging 2023; 15: 8576–8593. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Baker DJ, Childs BG, Durik M, Wijers ME, Sieben CJ, Zhong J, et al. Naturally occurring p16-positive cells shorten healthy lifespan. Nature 2016; 530: 184–189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Astrike-Davis EM, Coryell P, Loeser RF. Targeting cellular senescence as a novel treatment for osteoarthritis. Curr Opin Pharmacol 2022; 64: 102213. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Jeon OH, Kim C, Laberge RM, Demaria M, Rathod S, Vasserot AP, et al. Local clearance of senescent cells attenuates the development of post-traumatic osteoarthritis and creates a pro-regenerative environment. Nat Med 2017; 23: 775–781. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Faust HJ, Zhang H, Han J, Wolf MT, Jeon OH, Sadtler K, et al. IL-17 and immunologically induced senescence regulate response to injury in osteoarthritis. J Clin Invest 2020; 130: 5453–5507. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Lane NHB, Visich J, Xie B, Khan A, Dananberg J. A phase 2 rendomized, double-blind, placebo-controlled study of senolytic molecule UBX0101 in the treatment of painful knee osteoarthritis. Osteoarthritis Cartilage 2021; 29: S52. [Google Scholar]
- 63.Kirkland JL, Tchkonia T. Senolytic drugs: from discovery to translation. J Intern Med 2020; 288: 518–536. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Yamaura K, Nelson AL, Nishimura H, Rutledge JC, Ravuri SK, Bahney C, et al. The effects of fisetin on bone and cartilage: A systematic review. Pharmacol Res 2022; 185: 106504. [DOI] [PubMed] [Google Scholar]
- 65.Zheng W, Feng Z, You S, Zhang H, Tao Z, Wang Q, et al. Fisetin inhibits IL-1beta-induced inflammatory response in human osteoarthritis chondrocytes through activating SIRT1 and attenuates the progression of osteoarthritis in mice. Int Immunopharmacol 2017; 45: 135–147. [DOI] [PubMed] [Google Scholar]
- 66.Baker MC, Sheth K, Liu Y, Lu D, Lu R, Robinson WH. Development of osteoarthritis in adults with Type 2 diabetes treated with Metformin vs a Sulfonylurea. JAMA Netw Open 2023; 6: e233646. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Minton DM, Elliehausen CJ, Javors MA, Santangello KS, Konopka AR. Rapamycin-induced hyperglycemia is associated with exacerbated age-related osteoarthritis. Arthritis Research & Therapy 2021; 23: 253. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Lamming DW, Ye L, Sabatini DM, Baur JA. Rapalogs and mTOR inhibitors as anti-aging therapeutics. Journal of Clinical Investigation 2013; 123: 980–989. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Covarrubias AJ, Perrone R, Grozio A, Verdin E. NAD+ metabolism and its roles in cellular processes during ageing. Nature Reviews Molecular Cell Biology 2021; 22: 119–141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Gil Alabarse P, Chen LY, Oliveira P, Qin H, Liu-Bryan R. Targeting CD38 to suppress osteoarthritis development and associated pain after joint injury in mice. Arthritis Rheumatol 2023; 75: 364–374. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Long H, Liu Q, Yin H, Wang K, Diao N, Zhang Y, et al. Prevalence trends of site-specific osteoarthritis from 1990 to 2019: findings from the global burden of disease study 2019. Arthritis Rheumatol 2022; 74: 1172–1183. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.United Nations Department of Economic and Social Affairs, Population Division. World population prospects 2022.
- 73.Loeser RF. Using mice to model human osteoarthritis: age, biologic sex, and pain matter. Arthritis Rheumatol 2023; 75: 1733–1735. [DOI] [PubMed] [Google Scholar]
- 74.Ewart D, Harper L, Gravely A, Miller RA, Carlson CS, Loeser RF. Naturally occurring osteoarthritis in male mice with an extended lifespan. Connect Tissue Res 2020; 61: 95–103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Gil TH, Zheng H, Lee HG, Shin JW, Hwang SW, Jang KM, et al. Senolytic drugs relieve pain by reducing peripheral nociceptive signaling without modifying joint tissue damage in spontaneous osteoarthritis. Aging 2022; 14: 6006–6027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Miller RE, Tran PB, Das R, Ghoreishi-Haack N, Ren D, Miller RJ, et al. CCR2 chemokine receptor signaling mediates pain in experimental osteoarthritis. Proc Natl Acad Sci U S A 2012; 109: 20602–20607. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Longobardi L, Temple JD, Tagliafierro L, Willcockson H, Esposito A, D’Onofrio N, et al. Role of the C-C chemokine receptor-2 in a murine model of injury-induced osteoarthritis. Osteoarthritis Cartilage 2017; 25: 914–925. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Sarkar TJ, Quarta M, Mukherjee S, Colville A, Paine P, Doan L, et al. Transient non-integrative expression of nuclear reprogramming factors promotes multifaceted amelioration of aging in human cells. Nature Communications 2020; 11. doi 10.1038/s41467-020-15174-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Jing Y, Jiang X, Ji Q, Wu Z, Wang W, Liu Z, et al. Genome-wide CRISPR activation screening in senescent cells reveals SOX5 as a driver and therapeutic target of rejuvenation. Cell Stem Cell 2023; 30: 1452–1471.e1410. [DOI] [PubMed] [Google Scholar]
- 80.Coppé J-P, Desprez P-Y, Krtolica A, Campisi J. The Senescence-associated secretory phenotype: The dark side of tumor suppression. Annual Review of Pathology: Mechanisms of Disease 2010; 5: 99–118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.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: e3000599. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.