Next Article in Journal
Signaling Role of Pericytes in Vascular Health and Tissue Homeostasis
Previous Article in Journal
Antibacterial Effect of Spanish Honeys of Different Botanical Origins against Staphylococcus epidermidis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Involvement of Matricellular Proteins in Cellular Senescence: Potential Therapeutic Targets for Age-Related Diseases

1
Department of Molecular Patho-Physiology, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda 278-8510, Chiba, Japan
2
Clinical Research Center in Hiroshima, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-Ku, Hiroshima 734-8551, Japan
3
Department of Pharmacy, Faculty of Pharmaceutical Sciences, Sanyo-Onoda City University, 1-1-1 Daigaku-Doori, Sanyo-Onoda 756-0884, Yamaguchi, Japan
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(12), 6591; https://doi.org/10.3390/ijms25126591
Submission received: 29 May 2024 / Revised: 10 June 2024 / Accepted: 12 June 2024 / Published: 15 June 2024
(This article belongs to the Section Bioactives and Nutraceuticals)

Abstract

:
Senescence is a physiological and pathological cellular program triggered by various types of cellular stress. Senescent cells exhibit multiple characteristic changes. Among them, the characteristic flattened and enlarged morphology exhibited in senescent cells is observed regardless of the stimuli causing the senescence. Several studies have provided important insights into pro-adhesive properties of cellular senescence, suggesting that cell adhesion to the extracellular matrix (ECM), which is involved in characteristic morphological changes, may play pivotal roles in cellular senescence. Matricellular proteins, a group of structurally unrelated ECM molecules that are secreted into the extracellular environment, have the unique ability to control cell adhesion to the ECM by binding to cell adhesion receptors, including integrins. Recent reports have certified that matricellular proteins are closely involved in cellular senescence. Through this biological function, matricellular proteins are thought to play important roles in the pathogenesis of age-related diseases, including fibrosis, osteoarthritis, intervertebral disc degeneration, atherosclerosis, and cancer. This review outlines recent studies on the role of matricellular proteins in inducing cellular senescence. We highlight the role of integrin-mediated signaling in inducing cellular senescence and provide new therapeutic options for age-related diseases targeting matricellular proteins and integrins.

1. Introduction

Cellular senescence is a physiological and pathological cellular process triggered by various cellular stresses, such as telomere shortening, aberrant oncogene activation, and DNA damage accumulation [1]. Senescent cells are characterized by stable proliferation arrest but remain viable and metabolically active. These cells are found not only in aged tissues, but also in embryonic development, wound healing, tissue remodeling, inflammatory lesions, and the tumor microenvironment [1]. Senescent cells can secrete a number of factors, such as chemokines, growth factors, proinflammatory cytokines, proteases, and lipids, collectively known as the senescence-associated secretory phenotype (SASP) [2,3]. The biological functions of the SASP exhibit beneficial and detrimental consequences in a context-dependent manner; transient SASP activity shows beneficial roles in human health, whereas dysregulated and long-lasting SASP activity plays crucial roles in the deleterious effects of age-related diseases [4]. Accordingly, mechanistic analyses of the cellular senescence and SASP development observed in the microenvironments of various diseases have been vigorously conducted in recent years [2]. Recent evidence shows that the onset and progression of age-related diseases, such as intervertebral disc degeneration (IDD) [5,6,7], osteoarthritis (OA) [8,9,10,11], and abdominal aortic aneurysm [12,13,14], are correlated with large numbers of senescent cells within the lesion.
Cellular senescence is induced by the repeated exposure of cells to a variety of stimuli, such as chemotherapeutic drugs, oncogene activation, the exhaustion of cellular replicative potential, and oxidative stress [2]. Regardless of the cause, senescent cells are commonly observed to exhibit a characteristic flattened and enlarged morphology. However, it remains unclear whether this change in cell morphology during the process of cellular senescence is necessary for cellular senescence or merely a consequence. Several studies have provided important insights into pro-adhesive properties of cellular senescence [15,16,17], suggesting that cell adhesion to the extracellular matrix (ECM), which is involved in characteristic morphological changes, may play pivotal roles in cellular senescence. Cell adhesion to the extracellular matrix (ECM) is one of the important determinants of cell morphology, and adhesion receptor integrins are primarily responsible for this binding [18]. Unlike ECM molecules that act as structural components, secreted non-structural ECM components, called matricellular proteins, serve to modulate cell–ECM interactions by interacting with cell adhesion receptors, including integrins [19]. Families of matricellular proteins include centralized or cellular communication network (CCN), periostin, thrombospondin-1 (TSP-1), follistatin-like 1 (FSTL1), osteopontin (OPN), galectin-3 (Gal-3), plasminogen activator inhibitor-1 (PAI-1), pigment epithelium-derived factor (PEDF), and tenascin-C (TNC) [20]. Matricellular proteins exhibit versatile functions, which are governed by alternative splicing to generate multiple isoforms [21] or by proteolytic processing to liberate their bioactive fragments, termed matricryptins [22]. Moreover, matricellular proteins are typically expressed at high levels in developing tissues and show reduced expression in adult tissues [19]. Their expression levels are dysregulated in pathological states, including inflammation and tumorigenesis and in response to different types of injury, inflammation, or tumor development [19]. Therefore, matricellular proteins are considered to play major roles in the pathogenesis of age-related diseases. However, the substantial role of the matricellular proteins responsible for the onset and progression of age-related diseases has not been clarified. Recently, matricellular proteins have been shown to be able to induce cellular senescence [23,24,25], further supporting the belief that matricellular proteins are involved in the onset of age-related diseases. This also raises expectations that matricellular proteins may be a promising target for the treatment of such diseases.
In this review, we describe the recent studies on the role of matricellular proteins in inducing cellular senescence (Table 1). We also highlight the role played by integrin-mediated signaling in inducing cellular senescence and provide new therapeutic options for age-related diseases targeting matricellular proteins and their cell surface receptor integrins. Among matricellular proteins, we pay special attention to TNC and its peptide fragments. As detailed below, a peptide called TNIIIA2, derived from TNC, has been shown to cause cellular transformation and its malignant progression through various effects, including the induction of cellular senescence.

2. Matricellular Proteins in Cellular Senescence

2.1. Centralized or Cellular Communication Network (CCN)

The CCN family comprises six homologous members (CCN1-6) [51]. CCN proteins contain four modules, namely insulin-like growth factor-binding protein (IGFBP) module, von Willebrand factor type C (vWC) module, TSP-1 module, and C-terminal cysteine knot (CT) (with CCN5 lacking the CT module) [52,53,54,55]. The distinct functional modules possessed by each member dictate their interaction with various binding ligands, such as integrins, leading to diverse biological roles of CCN proteins as matricellular proteins.
CCN1, also known as cysteine-rich protein 61 (CYR61) and IGFBP-10, is expressed in multiple tissues [52]. Its expression changes in response to endogenous factors and external stimuli, such as oxidative stress, mechanical stress, and ultraviolet irradiation [56,57,58,59], and aberrant CCN1 expression is associated with aging and age-related diseases, including idiopathic pulmonary fibrosis (IPF) [60] and OA [27,61,62]. Jun and Lau found that human fibroblasts stimulated in the presence of recombinant CCN1 exhibit irreversible growth arrest, an enlarged and flattened cell morphology, the expression of senescence-associated (SA)-β-gal (a lysosomal enzyme used widely as a marker of senescence), p53 and p16 (a commonly used marker of senescence-associated growth arrest), and the production of SASP factors, indicating the induction of cellular senescence [23]. The fibroblasts that adhered to plates coated with CCN1 also exhibited the characteristics of cellular senescence [23]. The authors suggested, using Ccn1dm/dm knockin mice carrying the integrin α6β1 binding-defective mutant, that cellular senescence induced by CCN1–integrin α6β1 interactions might restrict fibrosis during tissue repair [23]. Feng and colleagues found that age is positively correlated with the disease severity of OA and the expression levels of senescence marker p16 and CCN1 in human chondrocytes [27]. CCN1 is also able to induce cellular senescence in human chondrocytes [27], and the suppression of CCN1-induced chondrocyte senescence reduces OA severity in an age-related OA mouse model [27], suggesting that inhibition of cellular senescence caused by CCN1 might be a novel therapeutic strategy for treating OA.
CCN2 is additionally called connective tissue growth factor (CTGF) and IGFBP-8 [63]. CCN2 is expressed in a variety of tissues at the development stages, with particularly high levels in cartilage and vasculature [64], and its expression is maintained in normal adult tissues [59,65]. Furthermore, its expression levels are dysregulated in aging [66,67] and age-related diseases, including IPF [68], and liver fibrosis [69,70]. Thus, CCN2 could play substantial roles in the development and progression of fibrotic diseases [71]. Jun and Lau have found that, similar to CCN1 [23], CCN2 also induces fibroblast senescence by binding to integrin α6β1, revealing that CCN2-induced fibroblast senescence concurrently restrains fibrotic responses [24].
CCN3, also known as nephroblastoma overexpressed (NOV) and IGFBP-9 [63,72], is highly expressed in adult endotheliocytes, smooth muscle cells, fibroblasts, and chondrocytes [73]. Several studies have detected dysregulated expression of CCN3 in aging [32] and a variety of age-related diseases, including OA [74]. The elevated expression of CCN3 in articular cartilage is correlated with OA severity [74]. In OA samples with elevated levels of CCN3, the expression levels of p16 and IL-1β, a proinflammatory SASP, are increased [74]. Kuwahara and colleagues found that the exogenous addition of CCN3 induces increased expression levels of senescence-associated factors p21 and p53 in mouse articular chondrocytes [32]. Moreover, cartilage-specific CCN3-overexpressing mice show degradative changes in knee joints, accompanied by increased expression levels of senescence-associated factor p21 and IL-6, IL-8, and TNF-α, which are not only commonly used SASPs, but also proinflammatory degenerative genes [32]. Collectively, CCN3-induced cellular senescence might be partially involved in the pathology of OA. Further studies are expected regarding the substantial role of CCN3 in the pathology of OA. In addition, trophoblast cells, upon acquisition of senescent traits induced by CCN3, exhibit activation of focal adhesion kinase (FAK) and elevated cell migration, indicating that CCN3-enhanced adhesive activity is involved in the induction of cellular senescence [31].
CCN4, known alternatively as WNT1 inducible signaling pathway protein 1 (WISP1) [55], exhibits elevated expression in the cartilage of OA patients compared with controls, with expression strongly correlated with disease severity [33,75]. Unlike other CCNs, the exogenous addition of recombinant CCN4 suppresses the induction of cellular senescence and chondrocyte apoptosis in human OA [33]. The suppression of cellular senescence and chondrocyte apoptosis by CCN4 is blocked by treatment with anti-integrin αvβ3 antibody [33]. Interestingly, anti-integrin αvβ3 antibody alone is able to induce the cellular senescence of chondrocytes [33]. Furthermore, in the in vivo setting, injection of CCN4 small interfering (si)RNA into the knee joints of a rat model of OA increases disease severity, accompanied by an increased number of SA-β-gal-positive cells and apoptotic cells, compared with non-targeting siRNA [33], suggesting that CCN4 suppression of cellular senescence might be beneficial for OA. However, it should be noted that some reports suggest that CCN4 may demonstrate detrimental effects on the progression of OA [76,77].

2.2. Periostin

Periostin belongs to the fasciclin family [78]. Under physiological conditions, periostin is expressed in limited numbers of tissues, such as periodontal ligament and periosteum [79]. On the other hand, periostin is commonly overexpressed in human tissues during pathological processes, including IDD [80,81,82]. The expression levels of periostin in human IDD tissues are correlated with the severity of the condition [34,80]. Intervertebral discs comprise the highly hydrated and gelatinous nucleus pulposus (NP) core, the peripherally located multilaminar annulus fibrosus and the cartilaginous endplates, and their constituent cells may play a key role in the pathogenesis of IDD [83]. In the context in which abnormal mechano-stress is considered a risk factor for IDD [84], Wu and colleagues found that mechano-stress induces NP cell senescence [34]. Unbiased RNA sequencing and an assay of transposase-accessible chromatin sequencing revealed that mechano-stress induced NP cell senescence and the SASP upregulated periostin transcription [34]. Furthermore, periostin directly accelerated NP cell senescence by binding to integrin αVβ3, forming the self-amplifying loop of cellular senescence [34]. Periostin-induced cellular senescence is also observed in human annulus fibrosus cells [34]. In an in vivo analysis using a rat IDD model, the degenerative phenotype was attenuated by inhibition of periostin [34]. Similar observations were reported by Zhu and colleagues [81]. Collectively, periostin targeting might be beneficial for IDD treatment.

2.3. Thrombospondin-1 (TSP-1)

While the expression levels of TSP-1 are normally low in the healthy state, its expression levels are positively correlated with age [36,85]. Furthermore, its dysregulated expression has been associated with several age-related diseases, such as heart failure [86], glioblastoma [87], age-related macular degeneration [88], and OA [89]. An excellent review has already described the relationship of TSP-1 with aging and age-related diseases (reviewed in [90]). Please refer to the review for information on the alterations in TSP-1 expression and its biochemical functions in aging and age-related diseases.
With regard to the induction of cellular senescence, several research papers suggest that TSP-1 directly induces cellular senescence in endothelial cells (ECs). Bitar reported that wound ECs derived from a diabetic rat model exhibit a characteristic feature of cellular senescence [91]. The cellular senescence induced by TSP-1 is thought to rely on its binding to its receptor CD47 [91]. Supporting this observation, Meijles and colleagues reported that TSP-1-induced cellular senescence in human pulmonary artery ECs occurs at a TSP-1 concentration within the range detected in the plasma of diseased patients [36,92,93]. Moreover, Gao and colleagues found that mouse ECs lacking CD47 have delayed senescence, enhanced proliferation, and increased tube formation in vitro and enhanced angiogenesis in vivo [35]. These results suggest that the TSP-1–CD47 signaling pathway positively regulates cellular senescence in ECs and may be a promising target for senotherapy in vascular diseases [94].
In addition, Baek and colleagues reported that TSP-1 deficiency leads to decreased survival and increased cancer aggression in a mouse model of Kras-induced lung tumorigenesis [37]. Mechanistically, in this mouse model, TSP-1 deficiency attenuated the senescence of lung lesions in lung adenomas [37]. Moreover, TSP-1 reduced the activation of pro-proliferative properties and maintained oncogenic Ras activation-induced senescence by tethering activated phospho-ERK to the cytosol [37], indicating that TSP-1 is involved in oncogenic-induced senescence.

2.4. Follistatin-like 1 (FSTL1)

FSTL1, additionally called follistatin-related protein and transforming growth factor (TGF)-β-stimulated clone 36 (Tsc36), was initially identified as a TGF-β-inducible gene [95], and is a secreted glycoprotein that belongs to the FS-SPARC family [96,97]. FSTL1 is considered to be strongly involved in age-related diseases (reviewed in [98]). Lung tissue and blood samples of IPF exhibit increased expression of FSTL1 compared with healthy controls [38,99,100]. Furthermore, neutralizing antibodies to functionally block FSTL1 and deletion of the FSTL1 gene attenuate bleomycin-induced lung fibrosis in mice [99,100]. However, the mechanisms underlying the detrimental effects of FSTL1 on the progression of pulmonary fibrosis have largely remained elusive.
A growing body of evidence supports the idea that cellular senescence, especially in alveolar type II (ATII) cells, plays key roles in IPF development or progression [101,102,103]. Sun and colleagues revealed that FSTL1 alone promotes cellular senescence in human ATII cells, and FSTL1 administered via intratracheal routes induces lung fibrosis in mice, accompanied by increased cellular senescence [38], indicating that FSTL1-induced ATII senescence might contribute to the pathogenesis of IPF.
In addition, FSTL1 increases with age in the NP of mice, and heterozygous FSTL1 knockout mice exhibit a reduced degree of degeneration compared with wild-type mice in a mouse model of IDD [104]. More recently, Yan and colleagues reported that the expression levels of FSTL1 are positively correlated with the disease severity of human IDD [39]. Mechanistically, recombinant human FSTL1 directly induces cellular senescence, accompanied by upregulated expression of proinflammatory SASP factors in NP cells derived from human IDD tissue [39]. In addition, FSTL1 silencing by siRNA ameliorates IDD progression and decreases the numbers of p16- and IL-1β-positive cells in the rabbit IDD model [39]. Taken together, targeting of FSTL1-induced senescence might be a novel therapeutic strategy for IDD.

2.5. Osteopontin (OPN)

OPN, also known as secreted phosphoprotein 1, bone sialoprotein I, and early T-lymphocyte activation-1, belongs to the small integrin-binding ligand N-linked glycoprotein family [105]. OPN levels increase with aging [106,107,108,109], and high expression levels of OPN are observed in age-related diseases, including OA [110]. The increase in OA disease severity is positively correlated with elevated expression levels of OPN, its receptor integrin αvβ3, and SA-β-gal [111]. In line with these observations, analysis of single-cell RNA sequencing has revealed that OPN is highly expressed in OA cartilage, and these cells possessed more senescent cell characteristics [110]. In contrast, Tian and colleagues have reported decreased expression levels of OPN in human primary chondrocytes of OA cartilage in comparison with normal human primary chondrocytes [40]. Furthermore, the depletion of OPN by siRNA enhances cellular senescence and proinflammatory mediators in human OA chondrocytes [40]. In an in vivo rat model of OA, the depletion of OPN by siRNA results in worse disease severity, accompanied by an increase in senescent cells, compared with non-targeting siRNA [40]. These results suggest that OPN plays a crucial role in modulating human chondrocytes from the inflammatory environment. Due to conflicting research results, further work is required to determine the biological function of OPN and disease development/progression of OA.

2.6. Galectin-3 (Gal-3)

Gal-3 belongs to the galectin family of lectins that binds galactose-containing glycoproteins [112]. Gal-3 is normally localized to the cytoplasm, but it can be secreted to the extracellular space and function as a matricellular protein [113,114]. The elevated expression of Gal-3 has been reported in various age-related diseases [115], including non-alcoholic steatohepatitis [116], OA [117], neurodegenerative diseases [118], heart failure [119], fibrotic diseases [112], and several types of cancer [120], and the expression levels of Gal-3 correlate with disease severity, such as IDD [121], suggesting its role in the onset or progression of age-related diseases.
Vlachou and colleagues have revealed that, although a mouse model of cardiomyopathy exhibits cardiac fibrosis, the Gal-3 deficiency in this model confines the profibrotic responses [122]. Mechanistically, the cardiac fibroblasts with deficient Gal-3 in this model show elevated proliferative potential and a decreased number of SA-β-gal-positive cells [122], indicating that Gal-3 might be involved in cardiac fibrosis and the promoting effect of Gal-3 on cellular senescence.
In contrast, Gal-3 depletion has been associated with decreased proliferative potential and increased numbers of SA-β-gal-positive mouse fibroblasts, human foreskin fibroblasts, and human gastric cancer cells [41]. In in vivo settings using mice bearing human gastric cancer xenografts, the gastric tumor size was reduced in Gal-3-depleted xenografted mice [41], suggesting that Gal-3 suppresses premature senescence and aggravates gastric tumorigenesis. Collectively, Gal-3-induced cellular senescence might be quite diverse and cell type- and context-dependent.

2.7. Plasminogen Activator Inhibitor-1 (PAI-1)

PAI-1 is alternatively called serpin peptidase inhibitor, clade E member 1 (SERPINE1). In fibrinolysis, plasminogen activators (PAs) convert inactive plasminogen into plasmin, which ultimately enables the breakdown of fibrin clots. Conversely, PAI-1 inhibits PAs to reduce the generation of plasmin [123]. In addition, while plasmin has the ability to activate matrix metalloproteinase (MMP) and both plasmin and MMP can degrade ECM, the reduction in plasmin through PA inhibition by PAI-1 induces ECM deposition [123]. Besides its fibrinolytic function, matrix-bound PAI-1 can modulate the activity of cell adhesion via PA receptor [124,125].
An excellent review has already described the relationship between PAI-1 and senescence (reviewed in [126]). Notably, PAI-1 deficiency delays the induction of senescence and protects against organ dysfunction, leading to a prolonged lifespan of Klotho-deficient mice that exhibit a premature aging syndrome [127]. Furthermore, Kortlever and colleagues revealed that MEFs derived from PAI-1 knockout mice proliferate beyond the checkpoint of replicative senescence [43]. The authors suggest that PAI-1 plays a role in inducing replicative senescence as a downstream target of p53 [43]. Moreover, Elzi and colleagues revealed that PAI-1 and IGFBP3 each have the ability to induce cellular senescence [44]. IGFBP3-induced cellular senescence is suppressed by treatment with tissue-type PA (tPA) through the proteolysis of IGFBP3, and PAI-1 stabilizes IGFBP3 by inhibiting tPA-mediated proteolysis of IGFBP3, leading to induction of cellular senescence [44]. In addition, Omer and colleagues reported that the formation of stress granules in the proliferative state impairs the induction of fibroblast senescence by interfering with the pro-senescence function of PAI-1 [45]. Mechanistically, the translocation of PAI-1 to stress granules could inhibit PAI-1-induced cellular senescence by interfering with its secretion [45].
PAI-1-induced cellular senescence may contribute to the progression of age-related diseases, with stimulation with PAI-1 alone inducing cellular senescence in ATII cells [46]. Furthermore, bleomycin-induced ATII cell senescence is associated with an increase in PAI-1 expression and is suppressed by the genetic or pharmacological inhibition of PAI-1 [46]. In vivo studies have revealed that depletion of PAI-1 in ATII cells in mice attenuates bleomycin-induced lung fibrosis [46]. Thus, PAI-1-induced cellular senescence might play a role in the development of lung fibrosis.
Moreover, PAI-1 is highly expressed in human atherosclerotic plaques and positively correlated with the expression of the senescence marker p21 [47]. In the context where senescence of coronary artery smooth muscle cells (SMCs) plays a significant role in atherogenesis [128,129], PAI-1 induces the cellular senescence of human coronary artery SMCs by binding to LRP1, which is a PAI-1 receptor. PAI-1-induced cellular senescence is inhibited by a PAI-1 inhibitor and by an antibody against LRP1 [48]. In in vivo settings, PAI-1 inhibitors attenuate atherosclerosis formation in a mouse model of atherosclerosis [48]. Taken together, the PAI-1–LRP1 axis at least partly promotes the development of atherosclerosis through the induction of cellular senescence.

2.8. Pigment Epithelium-Derived Factor (PEDF)

PEDF, additionally called early population doubling cDNA-1 and SERPINF1, belongs to the SERPIN family. PEDF is a secreted glycoprotein that exhibits anti-angiogenic, anti-oxidant, anti-tumorigenic, and neuroprotective properties [130,131,132,133]. PEDF is associated with aging and age-related diseases (reviewed in [132]), and, additionally, has effects on mesenchymal stem cells (MSCs). MSCs have considerable plasticity and represent a promising candidate for various clinical applications in the field of regenerative medicine [134]. To successfully apply MSCs to clinical applications, MSCs must be expanded. However, the expansion of MSCs is limited by irreversible growth arrest and cellular senescence. PEDF is able to reduce oxidative stress [135] and, based on its ability, PEDF delays cellular senescence in MSCs and allows for greater cell expansion [49], allowing their use as promising tools for regenerative therapy. On the other hand, PEDF expression has been reported to be increased in MSCs derived from old mice, and the expression of PEDF is associated with decreased efficacy of MSCs in treating myocardial infarction in a mouse model [136].

2.9. Tenascin-C (TNC)

TNC is abundantly expressed during embryonic development and around birth before decreasing in adulthood, when expression is at relatively low levels and restricted to sites with high cell turnover [137]. The versatility of TNC function is thought to be based not only on differences in receptors and the presence of multiple alternative splicing variants, but also its context-dependent liberation via proteolytic cleavage by MMP and a disintegrin and metalloproteinase with a TSP motif [138,139]. Aberrant TNC expression is associated with aging [140,141,142] and the pathogenesis of several age-related diseases, including OA [143], heart disease [144], fibrosis [145], and cancer [146]. However, the substantial role of TNC in age-related diseases remains largely unclear.
Proteolytic degradation of TNC has been detected in lung and colon cancer, and early-stage non-small-cell lung cancer patients with TNC degradation show significantly worse prognosis and higher recurrence than those without TNC degradation [147,148,149]. Increased MMP activity has been observed in patients with degraded TNC, indicating that exposure of the TNC functional cryptic site by several inflammatory proteinases may be associated with the malignant progression of cancer [147]. Among the TNC variants, those containing the FNIII A2 repeat are highly expressed in malignancies [150]. The variants containing FNIII A1A2 repeats also inhibit T cell activation in vitro, suggesting that TNC exerts immunosuppressive activity [151]. Additionally, work from our laboratory has shown that TNC harbors a functional cryptic site that comprises the YTITIRGV amino acid sequence located in the FN type III A2 repeat, and fragments containing this sequence, termed TNIIIA2 peptide, can potently enhance cell adhesion by activating β1-integrins; this activation state is sustained for an extended period [152]. Based on these functions, TNIIIA2 contributes to the ability of cancer cells to acquire malignant properties, including hyper-proliferation, disseminative migration, anchorage-independent growth, and anoikis resistance [153,154,155,156,157]. More recently, we found that TNIIIA2 peptide induces cellular senescence in human fibroblasts through β1-integrin activation [25]. In the induction of cellular senescence by TNIIIA2, it was particularly important to maintain the strongly activated state of integrin by TNIIIA2 for several days. This abnormal state of cell adhesion by TNIIIA2 is thought to cause ROS production, resulting in cellular senescence. Moreover, the humoral factors secreted by TNIIIA2-induced senescent fibroblasts cause preneoplastic epithelial cells to acquire malignant properties, perhaps leading to cancer progression [25]. Thus, our study demonstrates that the TNC molecule plays two functional roles that favor the malignant transformation of tumor cells. That is, TNIIIA2, as a TNC-derived matricryptin, not only directly acts on tumor cells to induce cancer aggressiveness, but also indirectly contributes to pro-tumorigenic effects through the induction of cellular senescence in fibroblasts. Functional blocking antibodies against TNIIIA2 [158] are expected to exhibit powerful anticancer effects against malignant tumors based on these two distinct functions.

3. Potential Strategies Targeting Cellular Senescence by Blocking Matricellular Protein Function and Integrin Signaling Pathway

3.1. Blocking Matricellular Protein Function as a Potential Senotherapeutic Strategy

Recently, clinical trials of agents targeting characteristics of cellular senescence, such as senolytic (killing senescent cells), senomorphic/senostatic (modulating phenotypes of senescent cells), or senostatic (modifying or suppressing the SASP) agents, are gradually accumulating insights into the relationship between senescent cells and human diseases [159]. Table 2 summarizes drugs targeting matricellular proteins that are under active clinical development and that are focused on in this review. Although no drugs have yet been approved that target cellular senescence by blocking matricellular proteins, clinical trials are underway for age-related diseases.
In the preceding sections, we summarized the relationship between matricellular protein-induced cellular senescence and the pathogenesis of age-related diseases. While knockout mice of some matricellular proteins, including CCN1, CCN2, and FSTL1, are characterized by embryonic or neonatal lethality, knockout mice of most matricellular proteins exhibit no apparent post-natal phenotypic changes [19]. Considering that the expression of matricellular proteins is context-dependent, as mentioned above, it is conceivable that matricellular protein targeting is unlikely to increase pleiotropic adverse effects. Therefore, the targeting of matricellular proteins may be an attractive strategy. Further research to obtain a deep understanding of both the regulatory mechanisms of matricellular proteins regarding the induction of cellular senescence and their associated pathogenesis in age-related diseases is required for clinical applications.

3.2. Role of Integrin-Mediated Cell Adhesion in Cellular Senescence

As mentioned in the above sections, cellular senescence directly induced by several matricellular proteins is clearly mediated by integrins. Senescent cells generally display a flattened and enlarged cell morphology, regardless of the stimuli inducing the senescence [178]. This suggests that integrin-mediated signaling may play a role in the induction of cellular senescence. Indeed, multiple studies have reported that integrin-mediated signaling is directly associated with cellular senescence induction (Table 3).
Shin and colleagues reported that the downregulation of p21-activated kinase-interacting exchange factor-β (βPIX), a negative regulator of focal adhesion [191], induces fibroblast senescence through enhanced cell adhesive activity; this is accompanied by increases in focal adhesions and actin stress fibers. This cell adhesion-induced cellular senescence is abolished by treatment with arginyl-glycyl-aspartic acid (RGD) peptide or FAK inhibitor [17].
ROS generated within cells are the main cause of stress-induced cellular senescence [192]. Intracellular ROS is produced by FAK–Rac1 activation following integrin engagement through fibronectin–α5-integrin-mediated cell adhesion [193]. Furthermore, it has been reported that cells expressing the auto-clustering β1-integrin mutant have upregulated ROS production [194], indicating that the production of ROS resulting in the induced cellular senescence hinges upon the cell adhesive activity mediated by integrins. Indeed, the accumulation of ROS is required for the binding of CCN1 or CCN2 to β1-integrin in CCN1- or CCN2-induced senescent fibroblasts [23,24]. Our group also found that β1-integrin activation-induced cellular senescence mediated by TNC-derived peptide is caused by ROS production and the resulting DNA damage [25]. Interestingly, our experiments showed that cellular senescence is induced not by the mere transient activation of integrin, but by its continued activation for more than a few days [25]. Cell adhesion via activated integrins is an essential event for cell survival, but maintaining the activated state at a high level for a long period may involve considerable stress for cells. It is possible that other matricellular proteins capable of inducing cellular senescence may similarly cause strong and sustained activation of integrin, thereby inducing ROS production and consequent cellular senescence.
Changes in mechanical properties, the increase in ECM, and tissue stiffness contribute to disease development and progression [195,196,197,198]. Notably, many studies have demonstrated that tissue stiffening enhances integrin signaling [199,200], and increasing evidence on enhanced ECM stiffness-induced cellular senescence has provided important insights into the pathogenesis of age-related diseases, including OA, abdominal aortic aneurysm, and IDD [34,201,202,203,204,205]. Wang and colleagues recently found that the increased stiffness of human NP tissue specimens with IDD is correlated with an increase in the degeneration grade [203]. More recently, a stiff substrate has been reported to induce cellular senescence in NP cells, which is accompanied by upregulation of β1-integrins or activation of the mechanosensitive Piezo1 channel [34,202,203]. Matricellular proteins expressed in age-related diseases also increase ECM stiffness through integrin-mediated signaling pathways. Growing evidence regarding the correlation between ECM stiffness and malignant progression will offer valuable insights into the significant roles of TNC [206,207,208]. Barnes and colleagues have found that glioblastoma exhibits an increased TNC-enriched stiffened ECM and elevated integrin mechanosignaling. Furthermore, glioblastoma xenografts derived from cells expressing auto-clustering active mutants of β1-integrins exhibit enhanced integrin mechanosignaling, increased TNC-enriched stiffened ECM, and, ultimately, worse tumor aggressiveness [207]. Moreover, our group suggested that the TNC-derived matricryptin TNIIIA2 peptide might generate a tumor microenvironment exhibiting pro-adhesive activity via the aberrant activation of β1-integrins [25], possibly resulting in increased stiffness.
Although further studies are needed regarding the matricellular protein–integrin axis in the induction of cellular senescence, the targeting of integrins, which act as receptors for matricellular proteins, may also regulate cellular senescence, offering a promising strategy for the treatment of age-related diseases.

3.3. Integrin Modulators as a Potential Senotherapeutic Strategy

Recent studies have accumulated evidence that several integrin inhibitors have the ability to suppress the induction of cellular senescence and/or the acquisition of the SASP in cell-based assays and preclinical studies (Table 4). These inhibitors could be potential candidates for senotherapy in age-related diseases.
Most common integrin inhibitors are antagonists that competitively inhibit integrin–ligand binding. Unlike these antagonistic peptides, peptide FNIII14 inhibits integrin signaling by an entirely distinct mechanism; peptide FNIII14 induces a conformational shift in β1-integrins from the active to an inactive state, leading to functional inactivation [210,211]. Based on this unique activity, peptide FNIII14 exhibits therapeutic potential in animal models of several conditions, such as metabolic diseases, organ fibrosis, malignant tumors, and atherosclerosis [210,212]. Furthermore, in the previous section (see Section 2.9), we noted that TNIIIA2, a matricryptin of TNC, activates β1-integrins and induces cellular senescence in human fibroblasts [25]. In contrast, peptide FNIII14 inhibits the cellular senescence induced by TNIIIA2 [25]. Peptide FNIII14, due to its novel mechanism targeting integrins, might become a promising senotherapeutic agent. While one intrinsic drawback of peptide drugs is their low bioavailability due to their susceptibility to enzymatic degradation in vivo [213], technology for a drug delivery nanosystem for peptide FNIII14 suitable for in vivo applications has recently been developed [214]. Further progress in consideration of clinical applications is expected.

4. Conclusions and Perspectives

Senotherapeutics have shown promising results in alleviating age-related diseases in animal models [215] and, based on the results, several candidate senotherapeutics have been advanced into clinical trials [216]. However, senotherapeutic intervention strategies, including senolytic drugs, might have some concerning adverse effects, especially with long-term use [217]. Considering that the expression of matricellular proteins is context-dependent and that mice knockout models of most matricellular proteins exhibit no apparent post-natal phenotypic changes [19], the targeting of matricellular proteins for senotherapy is unlikely to result in deleterious pleiotropic adverse effects, making it an attractive strategy. Furthermore, integrin-mediated events, including cell adhesion and sensing of ECM stiffness, contribute to the induction of cellular senescence, suggesting the potential utility of integrin modulators targeting cellular senescence for the treatment of age-related diseases. The clinical development of integrin-targeted therapeutics has vigorously progressed and is marked by the recent approval of orally administered medications [218]. However, the relationship between integrin and the induction of cellular senescence is highly complex [219]; it appears that integrin-regulated cellular senescence is dependent on a multitude of factors, including cell type, stimulus, and integrin subunit. To develop integrin-targeted therapeutics for age-related diseases, more careful consideration of the consequences of integrin blocking, as well as rigorous preclinical applications, are essential.

Author Contributions

Conceptualization, M.F., M.S., T.I. and F.F.; writing—original draft preparation, M.F.; writing—review and editing, M.S., T.I. and F.F.; funding acquisition, T.I. and M.F. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Zhang, L.; Pitcher, L.E.; Yousefzadeh, M.J.; Niedernhofer, L.J.; Robbins, P.D.; Zhu, Y. Cellular Senescence: A Key Therapeutic Target in Aging and Diseases. J. Clin. Investig. 2022, 132, e158450. [Google Scholar] [CrossRef]
  2. Malaquin, N.; Martinez, A.; Rodier, F. Keeping the Senescence Secretome under Control: Molecular Reins on the Senescence-Associated Secretory Phenotype. Exp. Gerontol. 2016, 82, 39–49. [Google Scholar] [CrossRef]
  3. Wiley, C.D.; Sharma, R.; Davis, S.S.; Lopez-Dominguez, J.A.; Mitchell, K.P.; Wiley, S.; Alimirah, F.; Kim, D.E.; Payne, T.; Rosko, A.; et al. Oxylipin Biosynthesis Reinforces Cellular Senescence and Allows Detection of Senolysis. Cell Metab. 2021, 33, 1124–1136.e5. [Google Scholar] [CrossRef]
  4. Paramos-de-Carvalho, D.; Jacinto, A.; Saúde, L. The Right Time for Senescence. eLife 2021, 10, e72449. [Google Scholar] [CrossRef]
  5. Francisco, V.; Pino, J.; González-Gay, M.Á.; Lago, F.; Karppinen, J.; Tervonen, O.; Mobasheri, A.; Gualillo, O. A New Immunometabolic Perspective of Intervertebral Disc Degeneration. Nat. Rev. Rheumatol. 2022, 18, 47–60. [Google Scholar] [CrossRef]
  6. Le Maitre, C.L.; Freemont, A.J.; Hoyland, J.A. Accelerated Cellular Senescence in Degenerate Intervertebral Discs: A Possible Role in the Pathogenesis of Intervertebral Disc Degeneration. Arthritis Res. Ther. 2007, 9, R45. [Google Scholar] [CrossRef]
  7. Liu, Y.; Dou, Y.; Sun, X.; Yang, Q. Mechanisms and Therapeutic Strategies for Senescence-Associated Secretory Phenotype in the Intervertebral Disc Degeneration Microenvironment. J. Orthop. Transl. 2024, 45, 56–65. [Google Scholar] [CrossRef]
  8. Loeser, R.F.; Collins, J.A.; Diekman, B.O. Ageing and the Pathogenesis of Osteoarthritis. Nat. Rev. Rheumatol. 2016, 12, 412–420. [Google Scholar] [CrossRef]
  9. Coryell, P.R.; Diekman, B.O.; Loeser, R.F. Mechanisms and Therapeutic Implications of Cellular Senescence in Osteoarthritis. Nat. Rev. Rheumatol. 2021, 17, 47–57. [Google Scholar] [CrossRef] [PubMed]
  10. Swahn, H.; Li, K.; Duffy, T.; Olmer, M.; D’Lima, D.D.; Mondala, T.S.; Natarajan, P.; Head, S.R.; Lotz, M.K. Senescent Cell Population with ZEB1 Transcription Factor as Its Main Regulator Promotes Osteoarthritis in Cartilage and Meniscus. Ann. Rheum. Dis. 2023, 82, 403–415. [Google Scholar] [CrossRef] [PubMed]
  11. Ansari, M.M.; Ghosh, M.; Lee, D.-S.; Son, Y.-O. Senolytic Therapeutics: An Emerging Treatment Modality for Osteoarthritis. Ageing Res. Rev. 2024, 96, 102275. [Google Scholar] [CrossRef]
  12. Aggarwal, S.; Qamar, A.; Sharma, V.; Sharma, A. Abdominal Aortic Aneurysm: A Comprehensive Review. Exp. Clin. Cardiol. 2011, 16, 11–15. [Google Scholar]
  13. Howard, D.P.J.; Banerjee, A.; Fairhead, J.F.; Handa, A.; Silver, L.E.; Rothwell, P.M. Population-Based Study of Incidence of Acute Abdominal Aortic Aneurysms with Projected Impact of Screening Strategy. J. Am. Heart Assoc. 2015, 4, e001926. [Google Scholar] [CrossRef]
  14. Wang, D.; Hao, X.; Jia, L.; Jing, Y.; Jiang, B.; Xin, S. Cellular Senescence and Abdominal Aortic Aneurysm: From Pathogenesis to Therapeutics. Front. Cardiovasc. Med. 2022, 9, 999465. [Google Scholar] [CrossRef] [PubMed]
  15. Cho, K.A.; Ryu, S.J.; Oh, Y.S.; Park, J.H.; Lee, J.W.; Kim, H.-P.; Kim, K.T.; Jang, I.S.; Park, S.C. Morphological Adjustment of Senescent Cells by Modulating Caveolin-1 Status. J. Biol. Chem. 2004, 279, 42270–42278. [Google Scholar] [CrossRef]
  16. Wolfson, M.; Budovsky, A.; Tacutu, R.; Fraifeld, V. The Signaling Hubs at the Crossroad of Longevity and Age-Related Disease Networks. Int. J. Biochem. Cell Biol. 2009, 41, 516–520. [Google Scholar] [CrossRef] [PubMed]
  17. Shin, E.-Y.; Park, J.-H.; You, S.-T.; Lee, C.-S.; Won, S.-Y.; Park, J.-J.; Kim, H.-B.; Shim, J.; Soung, N.-K.; Lee, O.-J.; et al. Integrin-Mediated Adhesions in Regulation of Cellular Senescence. Sci. Adv. 2020, 6, eaay3909. [Google Scholar] [CrossRef]
  18. Kanchanawong, P.; Calderwood, D.A. Organization, Dynamics and Mechanoregulation of Integrin-Mediated Cell–ECM Adhesions. Nat. Rev. Mol. Cell Biol. 2023, 24, 142–161. [Google Scholar] [CrossRef] [PubMed]
  19. Gerarduzzi, C.; Hartmann, U.; Leask, A.; Drobetsky, E. The Matrix Revolution: Matricellular Proteins and Restructuring of the Cancer Microenvironment. Cancer Res. 2020, 80, 2705–2717. [Google Scholar] [CrossRef]
  20. Cárdenas-León, C.G.; Mäemets-Allas, K.; Klaas, M.; Lagus, H.; Kankuri, E.; Jaks, V. Matricellular Proteins in Cutaneous Wound Healing. Front. Cell Dev. Biol. 2022, 10, 1073320. [Google Scholar] [CrossRef]
  21. Viloria, K.; Hill, N.J. Embracing the Complexity of Matricellular Proteins: The Functional and Clinical Significance of Splice Variation. Biomol. Concepts 2016, 7, 117–132. [Google Scholar] [CrossRef]
  22. Davis, G.E.; Bayless, K.J.; Davis, M.J.; Meininger, G.A. Regulation of Tissue Injury Responses by the Exposure of Matricryptic Sites within Extracellular Matrix Molecules. Am. J. Pathol. 2000, 156, 1489–1498. [Google Scholar] [CrossRef]
  23. Jun, J.-I.; Lau, L.F. The Matricellular Protein CCN1 Induces Fibroblast Senescence and Restricts Fibrosis in Cutaneous Wound Healing. Nat. Cell Biol. 2010, 12, 676–685. [Google Scholar] [CrossRef]
  24. Jun, J.-I.; Lau, L.F. CCN2 Induces Cellular Senescence in Fibroblasts. J. Cell Commun. Signal. 2017, 11, 15–23. [Google Scholar] [CrossRef] [PubMed]
  25. Fujita, M.; Sasada, M.; Eguchi, M.; Iyoda, T.; Okuyama, S.; Osawa, T.; Tsuzuranuki, K.; Sakamoto, M.; Hagihara, Y.; Matsumura, M.; et al. Induction of Cellular Senescence in Fibroblasts through β1-Integrin Activation by Tenascin-C-Derived Peptide and Its Protumor Effect. Am. J. Cancer Res. 2021, 11, 4364–4379. [Google Scholar] [PubMed]
  26. Kim, K.-H.; Chen, C.-C.; Monzon, R.I.; Lau, L.F. Matricellular Protein CCN1 Promotes Regression of Liver Fibrosis through Induction of Cellular Senescence in Hepatic Myofibroblasts. Mol. Cell. Biol. 2013, 33, 2078–2090. [Google Scholar] [CrossRef] [PubMed]
  27. Feng, M.; Peng, H.; Yao, R.; Zhang, Z.; Mao, G.; Yu, H.; Qiu, Y. Inhibition of Cellular Communication Network Factor 1 (CCN1)-Driven Senescence Slows down Cartilage Inflammaging and Osteoarthritis. Bone 2020, 139, 115522. [Google Scholar] [CrossRef]
  28. Jim Leu, S.-J.; Sung, J.-S.; Chen, M.-Y.; Chen, C.-W.; Cheng, J.-Y.; Wang, T.-Y.; Wang, J.-J. The Matricellular Protein CCN1 Suppresses Lung Cancer Cell Growth by Inducing Senescence via the P53/P21 Pathway. J. Cell. Biochem. 2013, 114, 2082–2093. [Google Scholar] [CrossRef]
  29. Du, J.; Klein, J.D.; Hassounah, F.; Zhang, J.; Zhang, C.; Wang, X.H. Aging Increases CCN1 Expression Leading to Muscle Senescence. Am. J. Physiol.-Cell Physiol. 2014, 306, C28–C36. [Google Scholar] [CrossRef]
  30. Noh, B.; Blasco-Conesa, M.P.; Lai, Y.-J.; Ganesh, B.P.; Urayama, A.; Moreno-Gonzalez, I.; Marrelli, S.P.; McCullough, L.D.; Moruno-Manchon, J.F. G-Quadruplexes Stabilization Upregulates CCN1 and Accelerates Aging in Cultured Cerebral Endothelial Cells. Front. Aging 2022, 2, 797562. [Google Scholar] [CrossRef]
  31. Kipkeew, F.; Kirsch, M.; Klein, D.; Wuelling, M.; Winterhager, E.; Gellhaus, A. CCN1 (CYR61) and CCN3 (NOV) Signaling Drives Human Trophoblast Cells into Senescence and Stimulates Migration Properties. Cell Adhes. Migr. 2016, 10, 163–178. [Google Scholar] [CrossRef] [PubMed]
  32. Kuwahara, M.; Kadoya, K.; Kondo, S.; Fu, S.; Miyake, Y.; Ogo, A.; Ono, M.; Furumatsu, T.; Nakata, E.; Sasaki, T.; et al. CCN3 (NOV) Drives Degradative Changes in Aging Articular Cartilage. Int. J. Mol. Sci. 2020, 21, 7556. [Google Scholar] [CrossRef] [PubMed]
  33. Cheng, C.; Tian, J.; Zhang, F.; Deng, Z.; Tu, M.; Li, L.; Yang, H.; Xiao, K.; Guo, W.; Yang, R.; et al. WISP1 Protects against Chondrocyte Senescence and Apoptosis by Regulating αvβ3 and PI3K/Akt Pathway in Osteoarthritis. DNA Cell Biol. 2021, 40, 629–637. [Google Scholar] [CrossRef] [PubMed]
  34. Wu, J.; Chen, Y.; Liao, Z.; Liu, H.; Zhang, S.; Zhong, D.; Qiu, X.; Chen, T.; Su, D.; Ke, X.; et al. Self-Amplifying Loop of NF-κB and Periostin Initiated by PIEZO1 Accelerates Mechano-Induced Senescence of Nucleus Pulposus Cells and Intervertebral Disc Degeneration. Mol. Ther. 2022, 30, 3241–3256. [Google Scholar] [CrossRef]
  35. Gao, Q.; Chen, K.; Gao, L.; Zheng, Y.; Yang, Y.-G. Thrombospondin-1 Signaling through CD47 Inhibits Cell Cycle Progression and Induces Senescence in Endothelial Cells. Cell Death Dis. 2016, 7, e2368. [Google Scholar] [CrossRef] [PubMed]
  36. Meijles, D.N.; Sahoo, S.; Al Ghouleh, I.; Amaral, J.H.; Bienes-Martinez, R.; Knupp, H.E.; Attaran, S.; Sembrat, J.C.; Nouraie, S.M.; Rojas, M.M.; et al. The Matricellular Protein TSP1 Promotes Human and Mouse Endothelial Cell Senescence through CD47 and Nox1. Sci. Signal. 2017, 10, eaaj1784. [Google Scholar] [CrossRef]
  37. Baek, K.-H.; Bhang, D.; Zaslavsky, A.; Wang, L.-C.; Vachani, A.; Kim, C.F.; Albelda, S.M.; Evan, G.I.; Ryeom, S. Thrombospondin-1 Mediates Oncogenic Ras–Induced Senescence in Premalignant Lung Tumors. J. Clin. Invest. 2013, 123, 4375–4389. [Google Scholar] [CrossRef]
  38. Sun, W.; Yang, X.; Chen, L.; Guo, L.; Huang, H.; Liu, X.; Yang, Y.; Xu, Z. FSTL1 Promotes Alveolar Epithelial Cell Aging and Worsens Pulmonary Fibrosis by Affecting SENP1-Mediated DeSUMOylation. Cell Biol. Int. 2023, 47, 1716–1727. [Google Scholar] [CrossRef]
  39. Yan, X.; Ding, J.-Y.; Zhang, R.-J.; Zhang, H.-Q.; Kang, L.; Jia, C.-Y.; Liu, X.-Y.; Shen, C.-L. FSTL1 Accelerates Nucleus Pulposus Cell Senescence and Intervertebral Disc Degeneration through TLR4/NF-κB Pathway. Inflammation 2024, 1–19. [Google Scholar] [CrossRef]
  40. Tian, J.; Cheng, C.; Kuang, S.-D.; Su, C.; Zhao, X.; Xiong, Y.; Li, Y.-S.; Gao, S.-G. OPN Deficiency Increases the Severity of Osteoarthritis Associated with Aberrant Chondrocyte Senescence and Apoptosis and Upregulates the Expression of Osteoarthritis-Associated Genes. Pain Res. Manag. 2020, 2020, e3428587. [Google Scholar] [CrossRef]
  41. Kim, S.-J.; Lee, H.-W.; Gu Kang, H.; La, S.-H.; Choi, I.J.; Ro, J.Y.; Bresalier, R.S.; Song, J.; Chun, K.-H. Ablation of Galectin-3 Induces p27KIP1-Dependent Premature Senescence without Oncogenic Stress. Cell Death Differ. 2014, 21, 1769–1779. [Google Scholar] [CrossRef] [PubMed]
  42. La, S.-H.; Kim, S.-J.; Kang, H.-G.; Lee, H.-W.; Chun, K.-H. Ablation of Human Telomerase Reverse Transcriptase (hTERT) Induces Cellular Senescence in Gastric Cancer through a Galectin-3 Dependent Mechanism. Oncotarget 2016, 7, 57117–57130. [Google Scholar] [CrossRef]
  43. Kortlever, R.M.; Higgins, P.J.; Bernards, R. Plasminogen Activator Inhibitor-1 Is a Critical Downstream Target of P53 in the Induction of Replicative Senescence. Nat. Cell Biol. 2006, 8, 877–884. [Google Scholar] [CrossRef]
  44. Elzi, D.J.; Lai, Y.; Song, M.; Hakala, K.; Weintraub, S.T.; Shiio, Y. Plasminogen Activator Inhibitor 1—Insulin-like Growth Factor Binding Protein 3 Cascade Regulates Stress-Induced Senescence. Proc. Natl. Acad. Sci. USA 2012, 109, 12052–12057. [Google Scholar] [CrossRef] [PubMed]
  45. Omer, A.; Patel, D.; Lian, X.J.; Sadek, J.; Di Marco, S.; Pause, A.; Gorospe, M.; Gallouzi, I.E. Stress Granules Counteract Senescence by Sequestration of PAI-1. EMBO Rep. 2018, 19, e44722. [Google Scholar] [CrossRef] [PubMed]
  46. Jiang, C.; Liu, G.; Luckhardt, T.; Antony, V.; Zhou, Y.; Carter, A.B.; Thannickal, V.J.; Liu, R.-M. Serpine 1 Induces Alveolar Type II Cell Senescence through Activating P53-P21-Rb Pathway in Fibrotic Lung Disease. Aging Cell 2017, 16, 1114–1124. [Google Scholar] [CrossRef]
  47. Wan, Y.-Z.; Gao, P.; Zhou, S.; Zhang, Z.-Q.; Hao, D.-L.; Lian, L.-S.; Li, Y.-J.; Chen, H.-Z.; Liu, D.-P. SIRT1-Mediated Epigenetic Downregulation of Plasminogen Activator Inhibitor-1 Prevents Vascular Endothelial Replicative Senescence. Aging Cell 2014, 13, 890–899. [Google Scholar] [CrossRef]
  48. Khoukaz, H.B.; Ji, Y.; Braet, D.J.; Vadali, M.; Abdelhamid, A.A.; Emal, C.D.; Lawrence, D.A.; Fay, W.P. Drug Targeting of Plasminogen Activator Inhibitor-1 Inhibits Metabolic Dysfunction and Atherosclerosis in a Murine Model of Metabolic Syndrome. Arterioscler. Thromb. Vasc. Biol. 2020, 40, 1479–1490. [Google Scholar] [CrossRef]
  49. Cao, Y.; Yang, T.; Gu, C.; Yi, D. Pigment Epithelium-Derived Factor Delays Cellular Senescence of Human Mesenchymal Stem Cells In Vitro by Reducing Oxidative Stress. Cell Biol. Int. 2013, 37, 305–313. [Google Scholar] [CrossRef]
  50. Rebustini, I.T.; Crawford, S.E.; Becerra, S.P. PEDF Deletion Induces Senescence and Defects in Phagocytosis in the RPE. Int. J. Mol. Sci. 2022, 23, 7745. [Google Scholar] [CrossRef]
  51. Jia, Q.; Xu, B.; Zhang, Y.; Ali, A.; Liao, X. CCN Family Proteins in Cancer: Insight into Their Structures and Coordination Role in Tumor Microenvironment. Front. Genet. 2021, 12, 649387. [Google Scholar] [CrossRef] [PubMed]
  52. Lau, L.F. CCN1/CYR61: The Very Model of a Modern Matricellular Protein. Cell. Mol. Life Sci. 2011, 68, 3149–3163. [Google Scholar] [CrossRef] [PubMed]
  53. Shen, Y.-W.; Zhou, Y.-D.; Chen, H.-Z.; Luan, X.; Zhang, W.-D. Targeting CTGF in Cancer: An Emerging Therapeutic Opportunity. Trends Cancer 2021, 7, 511–524. [Google Scholar] [CrossRef] [PubMed]
  54. Yin, H.; Liu, N.; Zhou, X.; Chen, J.; Duan, L. The Advance of CCN3 in Fibrosis. J. Cell Commun. Signal. 2023, 17, 1219–1227. [Google Scholar] [CrossRef] [PubMed]
  55. Malik, A.R.; Liszewska, E.; Jaworski, J. Matricellular Proteins of the Cyr61/CTGF/NOV (CCN) Family and the Nervous System. Front. Cell. Neurosci. 2015, 9, 237. [Google Scholar] [CrossRef] [PubMed]
  56. Qin, Z.; Robichaud, P.; He, T.; Fisher, G.J.; Voorhees, J.J.; Quan, T. Oxidant Exposure Induces Cysteine-Rich Protein 61 (CCN1) via c-Jun/AP-1 to Reduce Collagen Expression in Human Dermal Fibroblasts. PLoS ONE 2014, 9, e115402. [Google Scholar] [CrossRef] [PubMed]
  57. Grote, K.; Bavendiek, U.; Grothusen, C.; Flach, I.; Hilfiker-Kleiner, D.; Drexler, H.; Schieffer, B. Stretch-Inducible Expression of the Angiogenic Factor CCN1 in Vascular Smooth Muscle Cells Is Mediated by Egr-1. J. Biol. Chem. 2004, 279, 55675–55681. [Google Scholar] [CrossRef] [PubMed]
  58. Qin, Z.; Okubo, T.; Voorhees, J.J.; Fisher, G.J.; Quan, T. Elevated Cysteine-Rich Protein 61 (CCN1) Promotes Skin Aging via Upregulation of IL-1β in Chronically Sun-Exposed Human Skin. Age 2014, 36, 353–364. [Google Scholar] [CrossRef] [PubMed]
  59. Li, J.; Ye, L.; Owen, S.; Weeks, H.P.; Zhang, Z.; Jiang, W.G. Emerging Role of CCN Family Proteins in Tumorigenesis and Cancer Metastasis (Review). Int. J. Mol. Med. 2015, 36, 1451–1463. [Google Scholar] [CrossRef]
  60. Kulkarni, T.; Kurundkar, A.R.; Kim, Y.; de Andrade, J.; Luckhardt, T.; Thannickal, V.J. The Senescence-Associated Matricellular Protein CCN1 in Plasma of Human Subjects with Idiopathic Pulmonary Fibrosis. Respir. Med. 2020, 161, 105821. [Google Scholar] [CrossRef]
  61. Komatsu, M.; Nakamura, Y.; Maruyama, M.; Abe, K.; Watanapokasin, R.; Kato, H. Expression Profiles of Human CCN Genes in Patients with Osteoarthritis or Rheumatoid Arthritis. J. Orthop. Sci. 2015, 20, 708–716. [Google Scholar] [CrossRef]
  62. Chijiiwa, M.; Mochizuki, S.; Kimura, T.; Abe, H.; Tanaka, Y.; Fujii, Y.; Shimizu, H.; Enomoto, H.; Toyama, Y.; Okada, Y. CCN1 (Cyr61) Is Overexpressed in Human Osteoarthritic Cartilage and Inhibits ADAMTS-4 (Aggrecanase 1) Activity. Arthritis Rheumatol. 2015, 67, 1557–1567. [Google Scholar] [CrossRef]
  63. Monsen, V.T.; Attramadal, H. Structural Insights into Regulation of CCN Protein Activities and Functions. J. Cell Commun. Signal. 2023, 17, 371–390. [Google Scholar] [CrossRef]
  64. Leguit, R.J.; Raymakers, R.A.P.; Hebeda, K.M.; Goldschmeding, R. CCN2 (Cellular Communication Network Factor 2) in the Bone Marrow Microenvironment, Normal and Malignant Hematopoiesis. J. Cell Commun. Signal. 2021, 15, 25–56. [Google Scholar] [CrossRef] [PubMed]
  65. Hall-Glenn, F.; Lyons, K.M. Roles for CCN2 in Normal Physiological Processes. Cell. Mol. Life Sci. 2011, 68, 3209–3217. [Google Scholar] [CrossRef] [PubMed]
  66. Qin, Z.; He, T.; Guo, C.; Quan, T. Age-Related Downregulation of CCN2 Is Regulated by Cell Size in a YAP/TAZ-Dependent Manner in Human Dermal Fibroblasts: Impact on Dermal Aging. JID Innov. 2022, 2, 100111. [Google Scholar] [CrossRef] [PubMed]
  67. Xiang, Y.; Qin, Z.; Yang, Y.; Fisher, G.J.; Quan, T. Age-Related Elevation of HGF Is Driven by the Reduction of Fibroblast Size in a YAP/TAZ/CCN2 Axis-Dependent Manner. J. Dermatol. Sci. 2021, 102, 36–46. [Google Scholar] [CrossRef]
  68. Yanagihara, T.; Tsubouchi, K.; Gholiof, M.; Chong, S.G.; Lipson, K.E.; Zhou, Q.; Scallan, C.; Upagupta, C.; Tikkanen, J.; Keshavjee, S.; et al. Connective-Tissue Growth Factor Contributes to TGF-β1-Induced Lung Fibrosis. Am. J. Respir. Cell Mol. Biol. 2022, 66, 260–270. [Google Scholar] [CrossRef]
  69. Tamatani, T.; Kobayashi, H.; Tezuka, K.; Sakamoto, S.; Suzuki, K.; Nakanishi, T.; Takigawa, M.; Miyano, T. Establishment of the Enzyme-Linked Immunosorbent Assay for Connective Tissue Growth Factor (CTGF) and Its Detection in the Sera of Biliary Atresia. Biochem. Biophys. Res. Commun. 1998, 251, 748–752. [Google Scholar] [CrossRef]
  70. Gressner, A.M.; Yagmur, E.; Lahme, B.; Gressner, O.; Stanzel, S. Connective Tissue Growth Factor in Serum as a New Candidate Test for Assessment of Hepatic Fibrosis. Clin. Chem. 2006, 52, 1815–1817. [Google Scholar] [CrossRef]
  71. Fu, M.; Peng, D.; Lan, T.; Wei, Y.; Wei, X. Multifunctional Regulatory Protein Connective Tissue Growth Factor (CTGF): A Potential Therapeutic Target for Diverse Diseases. Acta Pharm. Sin. B 2022, 12, 1740–1760. [Google Scholar] [CrossRef] [PubMed]
  72. Kubota, S.; Kawata, K.; Hattori, T.; Nishida, T. Molecular and Genetic Interactions between CCN2 and CCN3 behind Their Yin-Yang Collaboration. Int. J. Mol. Sci. 2022, 23, 5887. [Google Scholar] [CrossRef]
  73. Peng, L.; Wei, Y.; Shao, Y.; Li, Y.; Liu, N.; Duan, L. The Emerging Roles of CCN3 Protein in Immune-Related Diseases. Mediators Inflamm. 2021, 2021, 5576059. [Google Scholar] [CrossRef] [PubMed]
  74. Hirose, K.; Kuwahara, M.; Nakata, E.; Tetsunaga, T.; Yamada, K.; Saiga, K.; Takigawa, M.; Ozaki, T.; Kubota, S.; Hattori, T. Elevated Expression of CCN3 in Articular Cartilage Induces Osteoarthritis in Hip Joints Irrespective of Age and Weight Bearing. Int. J. Mol. Sci. 2022, 23, 15311. [Google Scholar] [CrossRef] [PubMed]
  75. van den Bosch, M.H.J.; Ramos, Y.F.M.; den Hollander, W.; Bomer, N.; Nelissen, R.G.H.H.; Bovée, J.V.M.G.; van den Berg, W.B.; van Lent, P.L.E.M.; Blom, A.B.; van der Kraan, P.M.; et al. Increased WISP1 Expression in Human Osteoarthritic Articular Cartilage Is Epigenetically Regulated and Decreases Cartilage Matrix Production. Rheumatology 2019, 58, 1065–1074. [Google Scholar] [CrossRef] [PubMed]
  76. van den Bosch, M.H.; Blom, A.B.; Kram, V.; Maeda, A.; Sikka, S.; Gabet, Y.; Kilts, T.M.; van den Berg, W.B.; van Lent, P.L.; van der Kraan, P.M.; et al. WISP1/CCN4 Aggravates Cartilage Degeneration in Experimental Osteoarthritis. Osteoarthr. Cartil. 2017, 25, 1900–1911. [Google Scholar] [CrossRef] [PubMed]
  77. Blom, A.B.; Brockbank, S.M.; van Lent, P.L.; van Beuningen, H.M.; Geurts, J.; Takahashi, N.; van der Kraan, P.M.; van de Loo, F.A.; Schreurs, B.W.; Clements, K.; et al. Involvement of the Wnt Signaling Pathway in Experimental and Human Osteoarthritis: Prominent Role of Wnt-Induced Signaling Protein 1. Arthritis Rheum. 2009, 60, 501–512. [Google Scholar] [CrossRef] [PubMed]
  78. Yang, L.; Guo, T.; Chen, Y.; Bian, K. The Multiple Roles of Periostin in Non-Neoplastic Disease. Cells 2022, 12, 50. [Google Scholar] [CrossRef] [PubMed]
  79. Kii, I. Practical Application of Periostin as a Biomarker for Pathological Conditions. Adv. Exp. Med. Biol. 2019, 1132, 195–204. [Google Scholar] [CrossRef]
  80. Zhu, D.; Wang, Z.; Zhang, G.; Ma, C.; Qiu, X.; Wang, Y.; Liu, M.; Guo, X.; Chen, H.; Deng, Q.; et al. Periostin Promotes Nucleus Pulposus Cells Apoptosis by Activating the Wnt/β-Catenin Signaling Pathway. FASEB J. 2022, 36, e22369. [Google Scholar] [CrossRef]
  81. Tsai, T.-T.; Lai, P.-L.; Liao, J.-C.; Fu, T.-S.; Niu, C.-C.; Chen, L.-H.; Lee, M.S.; Chen, W.-J.; Fang, H.-C.; Ho, N.Y.J.; et al. Increased Periostin Gene Expression in Degenerative Intervertebral Disc Cells. Spine J. 2013, 13, 289–298. [Google Scholar] [CrossRef] [PubMed]
  82. Morimoto, T.; Kobayashi, T.; Ito, H.; Tsukamoto, M.; Yoshihara, T.; Hirata, H.; Otani, K.; Izuhara, K.; Nunomura, S.; Mawatari, M. Serum Periostin Levels Correlate with Severity of Intervertebral Disc Degeneration. Eur. Spine J. 2024, 1–7. [Google Scholar] [CrossRef] [PubMed]
  83. Oichi, T.; Taniguchi, Y.; Oshima, Y.; Tanaka, S.; Saito, T. Pathomechanism of Intervertebral Disc Degeneration. JOR Spine 2020, 3, e1076. [Google Scholar] [CrossRef]
  84. Binch, A.L.A.; Fitzgerald, J.C.; Growney, E.A.; Barry, F. Cell-Based Strategies for IVD Repair: Clinical Progress and Translational Obstacles. Nat. Rev. Rheumatol. 2021, 17, 158–175. [Google Scholar] [CrossRef] [PubMed]
  85. Głuchowska, A.; Cysewski, D.; Baj-Krzyworzeka, M.; Szatanek, R.; Węglarczyk, K.; Podszywałow-Bartnicka, P.; Sunderland, P.; Kozłowska, E.; Śliwińska, M.A.; Dąbrowski, M.; et al. Unbiased Proteomic Analysis of Extracellular Vesicles Secreted by Senescent Human Vascular Smooth Muscle Cells Reveals Their Ability to Modulate Immune Cell Functions. GeroScience 2022, 44, 2863–2884. [Google Scholar] [CrossRef]
  86. van Almen, G.C.; Verhesen, W.; van Leeuwen, R.E.W.; van de Vrie, M.; Eurlings, C.; Schellings, M.W.M.; Swinnen, M.; Cleutjens, J.P.M.; van Zandvoort, M.A.M.J.; Heymans, S.; et al. MicroRNA-18 and microRNA-19 Regulate CTGF and TSP-1 Expression in Age-Related Heart Failure. Aging Cell 2011, 10, 769–779. [Google Scholar] [CrossRef]
  87. Daubon, T.; Léon, C.; Clarke, K.; Andrique, L.; Salabert, L.; Darbo, E.; Pineau, R.; Guérit, S.; Maitre, M.; Dedieu, S.; et al. Deciphering the Complex Role of Thrombospondin-1 in Glioblastoma Development. Nat. Commun. 2019, 10, 1146. [Google Scholar] [CrossRef] [PubMed]
  88. Obasanmi, G.; Zeglinski, M.R.; Hardie, E.; Wilhelm, A.-C.; Turner, C.T.; Hiroyasu, S.; Boivin, W.A.; Tian, Y.; Zhao, H.; To, E.; et al. Granzyme B Contributes to Choroidal Neovascularization and Age-Related Macular Degeneration Through Proteolysis of Thrombospondin-1. Lab. Investig. 2023, 103, 100123. [Google Scholar] [CrossRef] [PubMed]
  89. Li, H.; Huang, L.; Zhao, R.; Wu, G.; Yin, Y.; Zhang, C.; Li, P.; Guo, L.; Wei, X.; Che, X.; et al. TSP-1 Increases Autophagy Level in Cartilage by Upregulating HSP27 Which Delays Progression of Osteoarthritis. Int. Immunopharmacol. 2024, 128, 111475. [Google Scholar] [CrossRef]
  90. Isenberg, J.S.; Roberts, D.D. Thrombospondin-1 in Maladaptive Aging Responses: A Concept Whose Time Has Come. Am. J. Physiol. Cell Physiol. 2020, 319, C45–C63. [Google Scholar] [CrossRef]
  91. Bitar, M.S. Diabetes Impairs Angiogenesis and Induces Endothelial Cell Senescence by Up-Regulating Thrombospondin-CD47-Dependent Signaling. Int. J. Mol. Sci. 2019, 20, 673. [Google Scholar] [CrossRef] [PubMed]
  92. Brostjan, C.; Bayer, A.; Zommer, A.; Gornikiewicz, A.; Roka, S.; Benkö, T.; Yaghubian, R.; Jakesz, R.; Steger, G.; Gnant, M.; et al. Monitoring of Circulating Angiogenic Factors in Dendritic Cell-Based Cancer Immunotherapy. Cancer 2003, 98, 2291–2301. [Google Scholar] [CrossRef] [PubMed]
  93. Novelli, E.M.; Kato, G.J.; Ragni, M.V.; Zhang, Y.; Hildesheim, M.E.; Nouraie, M.; Barge, S.; Meyer, M.P.; Hassett, A.C.; Gordeuk, V.R.; et al. Plasma Thrombospondin-1 Is Increased during Acute Sickle Cell Vaso-Occlusive Events and Associated with Acute Chest Syndrome, Hydroxyurea Therapy, and Lower Hemolytic Rates. Am. J. Hematol. 2012, 87, 326–330. [Google Scholar] [CrossRef] [PubMed]
  94. Han, Y.; Kim, S.Y. Endothelial Senescence in Vascular Diseases: Current Understanding and Future Opportunities in Senotherapeutics. Exp. Mol. Med. 2023, 55, 1–12. [Google Scholar] [CrossRef] [PubMed]
  95. Shibanuma, M.; Mashimo, J.; Mita, A.; Kuroki, T.; Nose, K. Cloning from a Mouse Osteoblastic Cell Line of a Set of Transforming-Growth-Factor-Beta 1-Regulated Genes, One of Which Seems to Encode a Follistatin-Related Polypeptide. Eur. J. Biochem. 1993, 217, 13–19. [Google Scholar] [CrossRef] [PubMed]
  96. Parfenova, O.K.; Kukes, V.G.; Grishin, D.V. Follistatin-like Proteins: Structure, Functions and Biomedical Importance. Biomedicines 2021, 9, 999. [Google Scholar] [CrossRef] [PubMed]
  97. Zhang, Y.; Wang, Y.; Zheng, G.; Liu, Y.; Li, J.; Huang, H.; Xu, C.; Zeng, Y.; Zhang, X.; Qin, J.; et al. Follistatin-like 1 (FSTL1) Interacts with Wnt Ligands and Frizzled Receptors to Enhance Wnt/β-Catenin Signaling in Obstructed Kidneys In Vivo. J. Biol. Chem. 2022, 298, 102010. [Google Scholar] [CrossRef] [PubMed]
  98. Mattiotti, A.; Prakash, S.; Barnett, P.; van den Hoff, M.J.B. Follistatin-like 1 in Development and Human Diseases. Cell. Mol. Life Sci. 2018, 75, 2339–2354. [Google Scholar] [CrossRef]
  99. Dong, Y.; Geng, Y.; Li, L.; Li, X.; Yan, X.; Fang, Y.; Li, X.; Dong, S.; Liu, X.; Li, X.; et al. Blocking Follistatin-like 1 Attenuates Bleomycin-Induced Pulmonary Fibrosis in Mice. J. Exp. Med. 2015, 212, 235–252. [Google Scholar] [CrossRef]
  100. Li, X.; Fang, Y.; Jiang, D.; Dong, Y.; Liu, Y.; Zhang, S.; Guo, J.; Qi, C.; Zhao, C.; Jiang, F.; et al. Targeting FSTL1 for Multiple Fibrotic and Systemic Autoimmune Diseases. Mol. Ther. J. Am. Soc. Gene Ther. 2021, 29, 347–364. [Google Scholar] [CrossRef]
  101. Schneider, J.L.; Rowe, J.H.; Garcia-de-Alba, C.; Kim, C.F.; Sharpe, A.H.; Haigis, M.C. The Aging Lung: Physiology, Disease, and Immunity. Cell 2021, 184, 1990–2019. [Google Scholar] [CrossRef] [PubMed]
  102. Schafer, M.J.; White, T.A.; Iijima, K.; Haak, A.J.; Ligresti, G.; Atkinson, E.J.; Oberg, A.L.; Birch, J.; Salmonowicz, H.; Zhu, Y.; et al. Cellular Senescence Mediates Fibrotic Pulmonary Disease. Nat. Commun. 2017, 8, 14532. [Google Scholar] [CrossRef] [PubMed]
  103. Yao, C.; Guan, X.; Carraro, G.; Parimon, T.; Liu, X.; Huang, G.; Mulay, A.; Soukiasian, H.J.; David, G.; Weigt, S.S.; et al. Senescence of Alveolar Type 2 Cells Drives Progressive Pulmonary Fibrosis. Am. J. Respir. Crit. Care Med. 2021, 203, 707–717. [Google Scholar] [CrossRef] [PubMed]
  104. Wang, S.; Wei, J.; Shi, J.; He, Q.; Zhou, X.; Gao, X.; Cheng, L. Follistatin-like 1 Attenuation Suppresses Intervertebral Disc Degeneration in Mice through Interacting with TNF-α and Smad Signaling Pathway. Oxid. Med. Cell. Longev. 2021, 2021, 6640751. [Google Scholar] [CrossRef] [PubMed]
  105. Du, Y.; Mao, L.; Wang, Z.; Yan, K.; Zhang, L.; Zou, J. Osteopontin—The Stirring Multifunctional Regulatory Factor in Multisystem Aging. Front. Endocrinol. 2022, 13, 1014853. [Google Scholar] [CrossRef] [PubMed]
  106. Sawaki, D.; Czibik, G.; Pini, M.; Ternacle, J.; Suffee, N.; Mercedes, R.; Marcelin, G.; Surenaud, M.; Marcos, E.; Gual, P.; et al. Visceral Adipose Tissue Drives Cardiac Aging through Modulation of Fibroblast Senescence by Osteopontin Production. Circulation 2018, 138, 809–822. [Google Scholar] [CrossRef] [PubMed]
  107. Saker, M.; Lipskaia, L.; Marcos, E.; Abid, S.; Parpaleix, A.; Houssaini, A.; Validire, P.; Girard, P.; Noureddine, H.; Boyer, L.; et al. Osteopontin, a Key Mediator Expressed by Senescent Pulmonary Vascular Cells in Pulmonary Hypertension. Arterioscler. Thromb. Vasc. Biol. 2016, 36, 1879–1890. [Google Scholar] [CrossRef] [PubMed]
  108. Gómez-Santos, B.; Saenz de Urturi, D.; Nuñez-García, M.; Gonzalez-Romero, F.; Buque, X.; Aurrekoetxea, I.; Gutiérrez de Juan, V.; Gonzalez-Rellan, M.J.; García-Monzón, C.; González-Rodríguez, Á.; et al. Liver Osteopontin Is Required to Prevent the Progression of Age-Related Nonalcoholic Fatty Liver Disease. Aging Cell 2020, 19, e13183. [Google Scholar] [CrossRef] [PubMed]
  109. Seiron, P.; Stenwall, A.; Hedin, A.; Granlund, L.; Esguerra, J.L.S.; Volkov, P.; Renström, E.; Korsgren, O.; Lundberg, M.; Skog, O. Transcriptional Analysis of Islets of Langerhans from Organ Donors of Different Ages. PLoS ONE 2021, 16, e0247888. [Google Scholar] [CrossRef]
  110. Qu, Y.; Wang, Y.; Wang, S.; Yu, X.; He, Y.; Lu, R.; Chen, S.; Meng, C.; Xu, H.; Pei, W.; et al. A Comprehensive Analysis of Single-Cell RNA Transcriptome Reveals Unique SPP1+ Chondrocytes in Human Osteoarthritis. Comput. Biol. Med. 2023, 160, 106926. [Google Scholar] [CrossRef]
  111. Cheng, C.; Tian, J.; Gao, S.-G.; Zhou, Z.-H.; Yang, R.-Q.; Xiao, K.; Guo, W.; Liu, L.; Yang, H.; Zhang, F.-J. The Expression of αvβ3 and Osteopontin in Osteoarthritic Knee Cartilage and Their Correlations with Disease Severity and Chondrocyte Senescence. Appl. Immunohistochem. Mol. Morphol. 2023, 31, 57–63. [Google Scholar] [CrossRef] [PubMed]
  112. Bouffette, S.; Botez, I.; De Ceuninck, F. Targeting Galectin-3 in Inflammatory and Fibrotic Diseases. Trends Pharmacol. Sci. 2023, 44, 519–531. [Google Scholar] [CrossRef] [PubMed]
  113. Ochieng, J.; Furtak, V.; Lukyanov, P. Extracellular Functions of Galectin-3. Glycoconj. J. 2002, 19, 527–535. [Google Scholar] [CrossRef] [PubMed]
  114. Dumic, J.; Dabelic, S.; Flögel, M. Galectin-3: An Open-Ended Story. Biochim. Biophys. Acta 2006, 1760, 616–635. [Google Scholar] [CrossRef] [PubMed]
  115. Hara, A.; Niwa, M.; Noguchi, K.; Kanayama, T.; Niwa, A.; Matsuo, M.; Hatano, Y.; Tomita, H. Galectin-3 as a Next-Generation Biomarker for Detecting Early Stage of Various Diseases. Biomolecules 2020, 10, 389. [Google Scholar] [CrossRef] [PubMed]
  116. Cyr, B.; Keane, R.W.; de Rivero Vaccari, J.P. ASC, IL-18 and Galectin-3 as Biomarkers of Non-Alcoholic Steatohepatitis: A Proof of Concept Study. Int. J. Mol. Sci. 2020, 21, 8580. [Google Scholar] [CrossRef] [PubMed]
  117. Toegel, S.; Bieder, D.; André, S.; Kayser, K.; Walzer, S.M.; Hobusch, G.; Windhager, R.; Gabius, H.-J. Human Osteoarthritic Knee Cartilage: Fingerprinting of Adhesion/Growth-Regulatory Galectins In Vitro and In Situ Indicates Differential Upregulation in Severe Degeneration. Histochem. Cell Biol. 2014, 142, 373–388. [Google Scholar] [CrossRef]
  118. Xue, S.; Lozinski, B.M.; Ghorbani, S.; Ta, K.; D’Mello, C.; Yong, V.W.; Dong, Y. Elevated Galectin-3 Is Associated with Aging, Multiple Sclerosis, and Oxidized Phosphatidylcholine-Induced Neurodegeneration. J. Neurosci. 2023, 43, 4725–4737. [Google Scholar] [CrossRef]
  119. Komici, K.; Gnemmi, I.; Bencivenga, L.; Vitale, D.F.; Rengo, G.; Di Stefano, A.; Eleuteri, E. Impact of Galectin-3 Circulating Levels on Frailty in Elderly Patients with Systolic Heart Failure. J. Clin. Med. 2020, 9, 2229. [Google Scholar] [CrossRef]
  120. Dong, R.; Zhang, M.; Hu, Q.; Zheng, S.; Soh, A.; Zheng, Y.; Yuan, H. Galectin-3 as a Novel Biomarker for Disease Diagnosis and a Target for Therapy (Review). Int. J. Mol. Med. 2018, 41, 599–614. [Google Scholar] [CrossRef]
  121. Elshamly, M.; Kinslechner, K.; Grohs, J.G.; Weinmann, D.; Walzer, S.M.; Windhager, R.; Gabius, H.-J.; Toegel, S. Galectins-1 and -3 in Human Intervertebral Disc Degeneration: Non-Uniform Distribution Profiles and Activation of Disease Markers Involving NF-κB by Galectin-1. J. Orthop. Res. 2019, 37, 2204–2216. [Google Scholar] [CrossRef] [PubMed]
  122. Vlachou, F.; Varela, A.; Stathopoulou, K.; Ntatsoulis, K.; Synolaki, E.; Pratsinis, H.; Kletsas, D.; Sideras, P.; Davos, C.H.; Capetanaki, Y.; et al. Galectin-3 Interferes with Tissue Repair and Promotes Cardiac Dysfunction and Comorbidities in a Genetic Heart Failure Model. Cell. Mol. Life Sci. 2022, 79, 250. [Google Scholar] [CrossRef] [PubMed]
  123. Chen, T.-Y.; Zhou, M.; Lin, M.-Q.; Liang, S.-T.; Yan, Y.; Wang, S.-M.; Fang, C.-S.; Li, D.; Ruan, Y. Research Progress on the SERPINE1 Protein and Chronic Inflammatory Diseases of the Upper Respiratory Tract: A Literature Review. Int. Arch. Allergy Immunol. 2021, 182, 1097–1102. [Google Scholar] [CrossRef] [PubMed]
  124. Czekay, R.-P.; Loskutoff, D.J. Plasminogen Activator Inhibitors Regulate Cell Adhesion through a uPAR-Dependent Mechanism. J. Cell. Physiol. 2009, 220, 655–663. [Google Scholar] [CrossRef] [PubMed]
  125. Maquerlot, F.; Galiacy, S.; Malo, M.; Guignabert, C.; Lawrence, D.A.; d’Ortho, M.-P.; Barlovatz-Meimon, G. Dual Role for Plasminogen Activator Inhibitor Type 1 as Soluble and as Matricellular Regulator of Epithelial Alveolar Cell Wound Healing. Am. J. Pathol. 2006, 169, 1624–1632. [Google Scholar] [CrossRef] [PubMed]
  126. Vaughan, D.E.; Rai, R.; Khan, S.S.; Eren, M.; Ghosh, A.K. Plasminogen Activator Inhibitor-1 Is a Marker and a Mediator of Senescence. Arterioscler. Thromb. Vasc. Biol. 2017, 37, 1446–1452. [Google Scholar] [CrossRef] [PubMed]
  127. Eren, M.; Boe, A.E.; Murphy, S.B.; Place, A.T.; Nagpal, V.; Morales-Nebreda, L.; Urich, D.; Quaggin, S.E.; Budinger, G.R.S.; Mutlu, G.M.; et al. PAI-1-Regulated Extracellular Proteolysis Governs Senescence and Survival in Klotho Mice. Proc. Natl. Acad. Sci. USA 2014, 111, 7090–7095. [Google Scholar] [CrossRef]
  128. Childs, B.G.; Baker, D.J.; Wijshake, T.; Conover, C.A.; Campisi, J.; van Deursen, J.M. Senescent Intimal Foam Cells Are Deleterious at All Stages of Atherosclerosis. Science 2016, 354, 472–477. [Google Scholar] [CrossRef]
  129. Wang, J.; Uryga, A.K.; Reinhold, J.; Figg, N.; Baker, L.; Finigan, A.; Gray, K.; Kumar, S.; Clarke, M.; Bennett, M. Vascular Smooth Muscle Cell Senescence Promotes Atherosclerosis and Features of Plaque Vulnerability. Circulation 2015, 132, 1909–1919. [Google Scholar] [CrossRef]
  130. Tombran-Tink, J.; Barnstable, C.J. PEDF: A Multifaceted Neurotrophic Factor. Nat. Rev. Neurosci. 2003, 4, 628–636. [Google Scholar] [CrossRef]
  131. Ek, E.T.H.; Dass, C.R.; Choong, P.F.M. PEDF: A Potential Molecular Therapeutic Target with Multiple Anti-Cancer Activities. Trends Mol. Med. 2006, 12, 497–502. [Google Scholar] [CrossRef]
  132. Abooshahab, R.; Dass, C.R. The Biological Relevance of Pigment Epithelium-Derived Factor on the Path from Aging to Age-Related Disease. Mech. Ageing Dev. 2021, 196, 111478. [Google Scholar] [CrossRef] [PubMed]
  133. Xu, M.; Chen, X.; Yu, Z.; Li, X. Receptors That Bind to PEDF and Their Therapeutic Roles in Retinal Diseases. Front. Endocrinol. 2023, 14, 1116136. [Google Scholar] [CrossRef] [PubMed]
  134. Hoang, D.M.; Pham, P.T.; Bach, T.Q.; Ngo, A.T.L.; Nguyen, Q.T.; Phan, T.T.K.; Nguyen, G.H.; Le, P.T.T.; Hoang, V.T.; Forsyth, N.R.; et al. Stem Cell-Based Therapy for Human Diseases. Signal Transduct. Target. Ther. 2022, 7, 272. [Google Scholar] [CrossRef] [PubMed]
  135. Brook, N.; Brook, E.; Dharmarajan, A.; Chan, A.; Dass, C.R. The Role of Pigment Epithelium-Derived Factor in Protecting against Cellular Stress. Free Radic. Res. 2019, 53, 1166–1180. [Google Scholar] [CrossRef] [PubMed]
  136. Liang, H.; Hou, H.; Yi, W.; Yang, G.; Gu, C.; Lau, W.B.; Gao, E.; Ma, X.; Lu, Z.; Wei, X.; et al. Increased Expression of Pigment Epithelium-Derived Factor in Aged Mesenchymal Stem Cells Impairs Their Therapeutic Efficacy for Attenuating Myocardial Infarction Injury. Eur. Heart J. 2013, 34, 1681–1690. [Google Scholar] [CrossRef] [PubMed]
  137. Giblin, S.P.; Midwood, K.S. Tenascin-C: Form versus Function. Cell Adhes. Migr. 2015, 9, 48–82. [Google Scholar] [CrossRef] [PubMed]
  138. Midwood, K.S.; Chiquet, M.; Tucker, R.P.; Orend, G. Tenascin-C at a Glance. J. Cell Sci. 2016, 129, 4321–4327. [Google Scholar] [CrossRef]
  139. Midwood, K.S.; Hussenet, T.; Langlois, B.; Orend, G. Advances in Tenascin-C Biology. Cell. Mol. Life Sci. 2011, 68, 3175–3199. [Google Scholar] [CrossRef]
  140. Choi, Y.E.; Song, M.J.; Hara, M.; Imanaka-Yoshida, K.; Lee, D.H.; Chung, J.H.; Lee, S.-T. Effects of Tenascin C on the Integrity of Extracellular Matrix and Skin Aging. Int. J. Mol. Sci. 2020, 21, 8693. [Google Scholar] [CrossRef]
  141. Calhoun, C.; Shivshankar, P.; Saker, M.; Sloane, L.B.; Livi, C.B.; Sharp, Z.D.; Orihuela, C.J.; Adnot, S.; White, E.S.; Richardson, A.; et al. Senescent Cells Contribute to the Physiological Remodeling of Aged Lungs. J. Gerontol. A Biol. Sci. Med. Sci. 2016, 71, 153–160. [Google Scholar] [CrossRef] [PubMed]
  142. Gremlich, S.; Cremona, T.P.; Yao, E.; Chabenet, F.; Fytianos, K.; Roth-Kleiner, M.; Schittny, J.C. Tenascin-C: Friend or Foe in Lung Aging? Front. Physiol. 2021, 12, 749776. [Google Scholar] [CrossRef] [PubMed]
  143. Hasegawa, M.; Yoshida, T.; Sudo, A. Tenascin-C in Osteoarthritis and Rheumatoid Arthritis. Front. Immunol. 2020, 11, 577015. [Google Scholar] [CrossRef] [PubMed]
  144. Imanaka-Yoshida, K. Tenascin-C in Heart Diseases-The Role of Inflammation. Int. J. Mol. Sci. 2021, 22, 5828. [Google Scholar] [CrossRef] [PubMed]
  145. Bhattacharyya, S.; Midwood, K.S.; Varga, J. Tenascin-C in Fibrosis in Multiple Organs: Translational Implications. Semin. Cell Dev. Biol. 2022, 128, 130–136. [Google Scholar] [CrossRef] [PubMed]
  146. Yilmaz, A.; Loustau, T.; Salomé, N.; Poilil Surendran, S.; Li, C.; Tucker, R.P.; Izzi, V.; Lamba, R.; Koch, M.; Orend, G. Advances on the Roles of Tenascin-C in Cancer. J. Cell Sci. 2022, 135, jcs260244. [Google Scholar] [CrossRef] [PubMed]
  147. Cai, M.; Onoda, K.; Takao, M.; Kyoko, I.-Y.; Shimpo, H.; Yoshida, T.; Yada, I. Degradation of Tenascin-C and Activity of Matrix Metalloproteinase-2 Are Associated with Tumor Recurrence in Early Stage Non-Small Cell Lung Cancer. Clin. Cancer Res. 2002, 8, 1152–1156. [Google Scholar]
  148. Sakai, T.; Kawakatsu, H.; Hirota, N.; Yokoyama, T.; Sakakura, T.; Saito, M. Specific Expression of Tenascin in Human Colonic Neoplasms. Br. J. Cancer 1993, 67, 1058–1064. [Google Scholar] [CrossRef] [PubMed]
  149. Kusagawa, H.; Onoda, K.; Namikawa, S.; Yada, I.; Okada, A.; Yoshida, T.; Sakakura, T. Expression and Degeneration of Tenascin-C in Human Lung Cancers. Br. J. Cancer 1998, 77, 98–102. [Google Scholar] [CrossRef] [PubMed]
  150. Dueck, M.; Riedl, S.; Hinz, U.; Tandara, A.; Möller, P.; Herfarth, C.; Faissner, A. Detection of Tenascin-C Isoforms in Colorectal Mucosa, Ulcerative Colitis, Carcinomas and Liver Metastases. Int. J. Cancer 1999, 82, 477–483. [Google Scholar] [CrossRef]
  151. Puente Navazo, M.D.; Valmori, D.; Rüegg, C. The Alternatively Spliced Domain TnFnIII A1A2 of the Extracellular Matrix Protein Tenascin-C Suppresses Activation-Induced T Lymphocyte Proliferation and Cytokine Production. J. Immunol. 2001, 167, 6431–6440. [Google Scholar] [CrossRef] [PubMed]
  152. Fujita, M.; Suzuki, H.; Fukai, F. Involvement of Integrin-Activating Peptides Derived from Tenascin-C in Colon Cancer Progression. World J. Gastrointest. Oncol. 2021, 13, 980–994. [Google Scholar] [CrossRef] [PubMed]
  153. Fujita, M.; Sasada, M.; Iyoda, T.; Nagai, R.; Kudo, C.; Yamamoto, T.; Osada, S.; Kodama, H.; Fukai, F. Anoikis Resistance Conferred by Tenascin-C-Derived Peptide TNIIIA2 and Its Disruption by Integrin Inactivation. Biochem. Biophys. Res. Commun. 2021, 536, 14–19. [Google Scholar] [CrossRef] [PubMed]
  154. Iyoda, T.; Fujita, M.; Fukai, F. Biologically Active TNIIIA2 Region in Tenascin-C Molecule: A Major Contributor to Elicit Aggressive Malignant Phenotypes from Tumors/Tumor Stroma. Front. Immunol. 2020, 11, 610096. [Google Scholar] [CrossRef] [PubMed]
  155. Fujita, M.; Yamamoto, T.; Iyoda, T.; Fujisawa, T.; Nagai, R.; Kudo, C.; Sasada, M.; Kodama, H.; Fukai, F. Autocrine Production of PDGF Stimulated by the Tenascin-C-Derived Peptide TNIIIA2 Induces Hyper-Proliferation in Glioblastoma Cells. Int. J. Mol. Sci. 2019, 20, 3183. [Google Scholar] [CrossRef] [PubMed]
  156. Fujita, M.; Yamamoto, T.; Iyoda, T.; Fujisawa, T.; Sasada, M.; Nagai, R.; Kudo, C.; Otsuka, K.; Kamiya, S.; Kodama, H.; et al. Aggressive Progression in Glioblastoma Cells through Potentiated Activation of Integrin α5β1 by the Tenascin-C–Derived Peptide TNIIIA2. Mol. Cancer Ther. 2019, 18, 1649–1658. [Google Scholar] [CrossRef] [PubMed]
  157. Fujita, M.; Ito-Fujita, Y.; Iyoda, T.; Sasada, M.; Okada, Y.; Ishibashi, K.; Osawa, T.; Kodama, H.; Fukai, F.; Suzuki, H. Peptide TNIIIA2 Derived from Tenascin-C Contributes to Malignant Progression in Colitis-Associated Colorectal Cancer via β1-Integrin Activation in Fibroblasts. Int. J. Mol. Sci. 2019, 20, 2752. [Google Scholar] [CrossRef] [PubMed]
  158. Tanaka, R.; Seki, Y.; Saito, Y.; Kamiya, S.; Fujita, M.; Okutsu, H.; Iyoda, T.; Takai, T.; Owaki, T.; Yajima, H.; et al. Tenascin-C-Derived Peptide TNIIIA2 Highly Enhances Cell Survival and Platelet-Derived Growth Factor (PDGF)-Dependent Cell Proliferation through Potentiated and Sustained Activation of Integrin α5β1. J. Biol. Chem. 2014, 289, 17699–17708. [Google Scholar] [CrossRef]
  159. 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, 75–95. [Google Scholar] [CrossRef]
  160. A Phase 3, Randomized, Double-Blind, Placebo-Controlled Efficacy and Safety Study of Pamrevlumab in Subjects with Idiopathic Pulmonary Fibrosis (IPF). Japan Registry of Clinical Trials, jRCT2051210169. Available online: https://jrct.niph.go.jp/latest-detail/jRCT2051210169 (accessed on 10 March 2024).
  161. Evaluation of Efficacy and Safety of Neoadjuvant Treatment with Pamrevlumab in Combination with Chemotherapy (Either Gemcitabine Plus Nab-Paclitaxel or FOLFIRINOX) in Participants with Locally Advanced Pancreatic Cancer (LAPIS). ClinicalTrials.gov, NCT03941093. Available online: https://classic.clinicaltrials.gov/ct2/show/NCT03941093 (accessed on 10 March 2024).
  162. Phase 3 Trial of Pamrevlumab or Placebo with Systemic Corticosteroids in Participants with Non-Ambulatory Duchenne Muscular Dystrophy (DMD) (LELANTOS-1). ClinicalTrials.gov, NCT04371666. Available online: https://classic.clinicaltrials.gov/ct2/show/NCT04371666 (accessed on 10 March 2024).
  163. Study on Intravenous Injection of SHR-1906 in the Treatment of Idiopathic Pulmonary Fibrosis. ClinicalTrials.gov, NCT05722964. Available online: https://classic.clinicaltrials.gov/ct2/show/NCT05722964 (accessed on 10 March 2024).
  164. Study to Evaluate Efficacy of OLX10010 in Reducing Recurrence of Hypertrophic Scarring after Scar Revision Surgery. ClinicalTrials.gov, NCT04877756. Available online: https://classic.clinicaltrials.gov/ct2/show/NCT04877756 (accessed on 10 March 2024).
  165. Study of Single and Multiple Doses of PRS-220 Administered by Oral Inhalation in Healthy Subjects. ClinicalTrials.gov, NCT05473533. Available online: https://classic.clinicaltrials.gov/ct2/show/NCT05473533 (accessed on 10 March 2024).
  166. A Study of Single Ascending Dose of LEM-S401 in Healthy Participants. ClinicalTrials.gov, NCT04707131. Available online: https://classic.clinicaltrials.gov/ct2/show/NCT04707131 (accessed on 10 March 2024).
  167. Phase 1 Study Evaluating VT1021 in Patients with Advanced Solid Tumors. ClinicalTrials.gov, NCT03364400. Available online: https://classic.clinicaltrials.gov/ct2/show/NCT03364400 (accessed on 10 March 2024).
  168. Study Evaluating the Efficacy and Safety of Belapectin for the Prevention of Esophageal Varices in NASH Cirrhosis (NAVIGATE). ClinicalTrials.gov, NCT04365868. Available online: https://classic.clinicaltrials.gov/ct2/show/NCT04365868 (accessed on 10 March 2024).
  169. Prolectin-M in COVID-19 Patients Having Mild to Moderate Symptoms Not Requiring Oxygen Support. (Prolectin-M). ClinicalTrials.gov, NCT04512027. Available online: https://classic.clinicaltrials.gov/ct2/show/NCT04512027 (accessed on 10 March 2024).
  170. A Long Term Extension Study to Assess the Safety of TB006 in Participants with Alzheimer’s Disease. ClinicalTrials.gov, NCT05476783. Available online: https://classic.clinicaltrials.gov/ct2/show/NCT05476783 (accessed on 10 March 2024).
  171. A Single and Repeat Dose Trial in Participants with Hepatic Impairment. ClinicalTrials.gov, NCT05009680. Available online: https://classic.clinicaltrials.gov/ct2/show/NCT05009680 (accessed on 10 March 2024).
  172. GB1211 and Pembrolizumab Versus Pembrolizumab and Placebo in Patients with Metastatic Melanoma and Head and Neck Squamous Cell Carcinoma. ClinicalTrials.gov, NCT05913388. Available online: https://classic.clinicaltrials.gov/ct2/show/NCT05913388 (accessed on 10 March 2024).
  173. Phase 3, Randomized, Double-Blind Study of the Efficacy of TM5614 in Combination with Tyrosine Kinase Inhibitors in Chronic Phase Chronic Myelogenous Leukemia Patients. Japan Registry of Clinical Trials, jRCT2031220084. Available online: https://jrct.niph.go.jp/latest-detail/jRCT2031220084 (accessed on 10 March 2024).
  174. Phase II Study of Combination Treatment of Nivolumab and TM5614, a PAI-1 Inhibitor for Patients with Non-Small Cell Lung Cancer (TM-Uprise Study). Japan Registry of Clinical Trials, jRCT2061230039. Available online: https://jrct.niph.go.jp/latest-detail/jRCT2061230039 (accessed on 10 March 2024).
  175. Phase II Study of the Safety and Efficacy of TM5614 in Combination with Paclitaxel for Cutaneous Angiosarcoma. Japan Registry of Clinical Trials, jRCT2021230016. Available online: https://jrct.niph.go.jp/latest-detail/jRCT2021230016 (accessed on 10 March 2024).
  176. A Phase II Investigator-Initiated Clinical Trial of PAI-1 Inhibitor (TM5614) for Systemic Sclerosis-Associated Interstitial Lung Disease under Immunosuppressive Treatment. Japan Registry of Clinical Trials, jRCT2021230022. Available online: https://jrct.niph.go.jp/latest-detail/jRCT2021230022 (accessed on 10 March 2024).
  177. An Investigator-Initiated Phase I/IIa Trial of Subretinal Injection of DVC1-0401 as Neuroprotective Gene Therapy for Retinitis Pigmentosa. Japan Registry of Clinical Trials, jRCT2073180024. Available online: https://jrct.niph.go.jp/latest-detail/jRCT2073180024 (accessed on 10 March 2024).
  178. Hernandez-Segura, A.; Nehme, J.; Demaria, M. Hallmarks of Cellular Senescence. Trends Cell Biol. 2018, 28, 436–453. [Google Scholar] [CrossRef]
  179. Kren, A.; Baeriswyl, V.; Lehembre, F.; Wunderlin, C.; Strittmatter, K.; Antoniadis, H.; Fässler, R.; Cavallaro, U.; Christofori, G. Increased Tumor Cell Dissemination and Cellular Senescence in the Absence of Beta1-Integrin Function. EMBO J. 2007, 26, 2832–2842. [Google Scholar] [CrossRef]
  180. Bui, T.; Gu, Y.; Ancot, F.; Sanguin-Gendreau, V.; Zuo, D.; Muller, W.J. Emergence of β1 Integrin-Deficient Breast Tumours from Dormancy Involves Both Inactivation of P53 and Generation of a Permissive Tumour Microenvironment. Oncogene 2022, 41, 527–537. [Google Scholar] [CrossRef]
  181. Deng, L.; Jin, K.; Zhou, X.; Zhang, Z.; Ge, L.; Xiong, X.; Su, X.; Jin, D.; Yuan, Q.; Zhang, C.; et al. Blockade of Integrin Signaling Reduces Chemotherapy-Induced Premature Senescence in Collagen Cultured Bladder Cancer Cells. Precis. Clin. Med. 2022, 5, pbac007. [Google Scholar] [CrossRef] [PubMed]
  182. Kozlova, N.I.; Morozevich, G.E.; Berman, A.E. Implication of Integrin α2β1 in Senescence of SK-Mel-147 Human Melanoma Cells. Aging 2021, 13, 18006–18017. [Google Scholar] [CrossRef] [PubMed]
  183. Martinkova, E.; Maglott, A.; Leger, D.Y.; Bonnet, D.; Stiborova, M.; Takeda, K.; Martin, S.; Dontenwill, M. Alpha5beta1 Integrin Antagonists Reduce Chemotherapy-Induced Premature Senescence and Facilitate Apoptosis in Human Glioblastoma Cells. Int. J. Cancer 2010, 127, 1240–1248. [Google Scholar] [CrossRef]
  184. Zhao, J.; Jiang, X.; Yan, L.; Lin, J.; Guo, H.; Yu, S.; Ye, B.; Zhu, J.; Zhang, W. Retinoic Acid Inducible Gene-I Slows down Cellular Senescence through Negatively Regulating the Integrin β3/P38 MAPK Pathway. Cell Cycle 2019, 18, 3378–3392. [Google Scholar] [CrossRef]
  185. Rapisarda, V.; Borghesan, M.; Miguela, V.; Encheva, V.; Snijders, A.P.; Lujambio, A.; O’Loghlen, A. Integrin Beta 3 Regulates Cellular Senescence by Activating the TGF-β Pathway. Cell Rep. 2017, 18, 2480–2493. [Google Scholar] [CrossRef] [PubMed]
  186. Li, S.; Jiang, S.; Zhang, Q.; Jin, B.; Lv, D.; Li, W.; Zhao, M.; Jiang, C.; Dai, C.; Liu, Z. Integrin β3 Induction Promotes Tubular Cell Senescence and Kidney Fibrosis. Front. Cell Dev. Biol. 2021, 9, 733831. [Google Scholar] [CrossRef]
  187. Franovic, A.; Elliott, K.C.; Seguin, L.; Camargo, M.F.; Weis, S.M.; Cheresh, D.A. Glioblastomas Require Integrin αvβ3/PAK4 Signaling to Escape Senescence. Cancer Res. 2015, 75, 4466–4473. [Google Scholar] [CrossRef]
  188. Liu, X.; Yin, D.; Zhang, Y.; Zhao, J.; Zhang, S.; Miao, J. Vascular Endothelial Cell Senescence Mediated by Integrin Beta4 In Vitro. FEBS Lett. 2007, 581, 5337–5342. [Google Scholar] [CrossRef]
  189. Yuan, L.; Du, X.; Tang, S.; Wu, S.; Wang, L.; Xiang, Y.; Qu, X.; Liu, H.; Qin, X.; Liu, C. ITGB4 Deficiency Induces Senescence of Airway Epithelial Cells through P53 Activation. FEBS J. 2019, 286, 1191–1203. [Google Scholar] [CrossRef] [PubMed]
  190. Jung, S.H.; Lee, M.; Park, H.A.; Lee, H.C.; Kang, D.; Hwang, H.J.; Park, C.; Yu, D.-M.; Jung, Y.R.; Hong, M.-N.; et al. Integrin α6β4-Src-AKT Signaling Induces Cellular Senescence by Counteracting Apoptosis in Irradiated Tumor Cells and Tissues. Cell Death Differ. 2019, 26, 245–259. [Google Scholar] [CrossRef]
  191. Kuo, J.-C.; Han, X.; Hsiao, C.-T.; Yates, J.R.; Waterman, C.M. Analysis of the Myosin-II-Responsive Focal Adhesion Proteome Reveals a Role for β-Pix in Negative Regulation of Focal Adhesion Maturation. Nat. Cell Biol. 2011, 13, 383–393. [Google Scholar] [CrossRef]
  192. Lu, T.; Finkel, T. Free Radicals and Senescence. Exp. Cell Res. 2008, 314, 1918–1922. [Google Scholar] [CrossRef]
  193. Chiarugi, P.; Pani, G.; Giannoni, E.; Taddei, L.; Colavitti, R.; Raugei, G.; Symons, M.; Borrello, S.; Galeotti, T.; Ramponi, G. Reactive Oxygen Species as Essential Mediators of Cell Adhesion: The Oxidative Inhibition of a FAK Tyrosine Phosphatase Is Required for Cell Adhesion. J. Cell Biol. 2003, 161, 933–944. [Google Scholar] [CrossRef] [PubMed]
  194. Lee, K.; Chen, Q.K.; Lui, C.; Cichon, M.A.; Radisky, D.C.; Nelson, C.M. Matrix Compliance Regulates Rac1b Localization, NADPH Oxidase Assembly, and Epithelial-Mesenchymal Transition. Mol. Biol. Cell 2012, 23, 4097–4108. [Google Scholar] [CrossRef] [PubMed]
  195. Martinez-Vidal, L.; Murdica, V.; Venegoni, C.; Pederzoli, F.; Bandini, M.; Necchi, A.; Salonia, A.; Alfano, M. Causal Contributors to Tissue Stiffness and Clinical Relevance in Urology. Commun. Biol. 2021, 4, 1011. [Google Scholar] [CrossRef]
  196. Xiong, J.; Xiao, R.; Zhao, J.; Zhao, Q.; Luo, M.; Li, F.; Zhang, W.; Wu, M. Matrix Stiffness Affects Tumor-Associated Macrophage Functional Polarization and Its Potential in Tumor Therapy. J. Transl. Med. 2024, 22, 85. [Google Scholar] [CrossRef]
  197. Kohn, J.C.; Lampi, M.C.; Reinhart-King, C.A. Age-Related Vascular Stiffening: Causes and Consequences. Front. Genet. 2015, 6, 112. [Google Scholar] [CrossRef]
  198. Schnellmann, R.; Ntekoumes, D.; Choudhury, M.I.; Sun, S.; Wei, Z.; Gerecht, S. Stiffening Matrix Induces Age-Mediated Microvascular Phenotype Through Increased Cell Contractility and Destabilization of Adherens Junctions. Adv. Sci. 2022, 9, e2201483. [Google Scholar] [CrossRef]
  199. Levental, K.R.; Yu, H.; Kass, L.; Lakins, J.N.; Egeblad, M.; Erler, J.T.; Fong, S.F.T.; Csiszar, K.; Giaccia, A.; Weninger, W.; et al. Matrix Crosslinking Forces Tumor Progression by Enhancing Integrin Signaling. Cell 2009, 139, 891–906. [Google Scholar] [CrossRef] [PubMed]
  200. Banerjee, S.; Nara, R.; Chakraborty, S.; Chowdhury, D.; Haldar, S. Integrin Regulated Autoimmune Disorders: Understanding the Role of Mechanical Force in Autoimmunity. Front. Cell Dev. Biol. 2022, 10, 852878. [Google Scholar] [CrossRef] [PubMed]
  201. Fu, B.; Shen, J.; Zou, X.; Sun, N.; Zhang, Z.; Liu, Z.; Zeng, C.; Liu, H.; Huang, W. Matrix Stiffening Promotes Chondrocyte Senescence and the Osteoarthritis Development through Downregulating HDAC3. Bone Res. 2024, 12, 32. [Google Scholar] [CrossRef] [PubMed]
  202. Zhao, R.; Yang, L.; He, S.; Xia, T. Nucleus Pulposus Cell Senescence Is Regulated by Substrate Stiffness and Is Alleviated by LOX Possibly through the Integrin β1-p38 MAPK Signaling Pathway. Exp. Cell Res. 2022, 417, 113230. [Google Scholar] [CrossRef] [PubMed]
  203. Wang, B.; Ke, W.; Wang, K.; Li, G.; Ma, L.; Lu, S.; Xiang, Q.; Liao, Z.; Luo, R.; Song, Y.; et al. Mechanosensitive Ion Channel Piezo1 Activated by Matrix Stiffness Regulates Oxidative Stress-Induced Senescence and Apoptosis in Human Intervertebral Disc Degeneration. Oxid. Med. Cell. Longev. 2021, 2021, 8884922. [Google Scholar] [CrossRef] [PubMed]
  204. Liu, Y.; Zhang, Z.; Li, J.; Chang, B.; Lin, Q.; Wang, F.; Wang, W.; Zhang, H. Piezo1 Transforms Mechanical Stress into pro Senescence Signals and Promotes Osteoarthritis Severity. Mech. Ageing Dev. 2023, 216, 111880. [Google Scholar] [CrossRef] [PubMed]
  205. He, H.; Zeng, B.; Wu, X.; Hou, J.; Wang, Y.; Wang, Y.; Lin, Y.; Wu, P.; Zheng, C.; Yin, H.; et al. Higher Matrix Stiffness Promotes VSMC Senescence by Affecting Mitochondria-ER Contact Sites and Mitochondria/ER Dysfunction. FASEB J. 2023, 37, e23318. [Google Scholar] [CrossRef] [PubMed]
  206. Miroshnikova, Y.A.; Mouw, J.K.; Barnes, J.M.; Pickup, M.W.; Lakins, J.N.; Kim, Y.; Lobo, K.; Persson, A.I.; Reis, G.F.; McKnight, T.R.; et al. Tissue Mechanics Promote IDH1-Dependent HIF1α-Tenascin C Feedback to Regulate Glioblastoma Aggression. Nat. Cell Biol. 2016, 18, 1336–1345. [Google Scholar] [CrossRef] [PubMed]
  207. Barnes, J.M.; Kaushik, S.; Bainer, R.O.; Sa, J.K.; Woods, E.C.; Kai, F.; Przybyla, L.; Lee, M.; Lee, H.W.; Tung, J.C.; et al. A Tension-Mediated Glycocalyx-Integrin Feedback Loop Promotes Mesenchymal-like Glioblastoma. Nat. Cell Biol. 2018, 20, 1203–1214. [Google Scholar] [CrossRef]
  208. Bhattacharyya, S.; Wang, W.; Morales-Nebreda, L.; Feng, G.; Wu, M.; Zhou, X.; Lafyatis, R.; Lee, J.; Hinchcliff, M.; Feghali-Bostwick, C.; et al. Tenascin-C Drives Persistence of Organ Fibrosis. Nat. Commun. 2016, 7, 11703. [Google Scholar] [CrossRef]
  209. Suezawa, T.; Kanagaki, S.; Moriguchi, K.; Masui, A.; Nakao, K.; Toyomoto, M.; Tamai, K.; Mikawa, R.; Hirai, T.; Murakami, K.; et al. Disease Modeling of Pulmonary Fibrosis Using Human Pluripotent Stem Cell-Derived Alveolar Organoids. Stem Cell Rep. 2021, 16, 2973–2987. [Google Scholar] [CrossRef] [PubMed]
  210. Fujita, M.; Sasada, M.; Iyoda, T.; Osada, S.; Kodama, H.; Fukai, F. Biofunctional Peptide FNIII14: Therapeutic Potential. Encyclopedia 2021, 1, 350–359. [Google Scholar] [CrossRef]
  211. Fujita, M.; Sasada, M.; Iyoda, T.; Fukai, F. Involvement of Integrin-Activating Peptides Derived from Tenascin-C in Cancer Aggression and New Anticancer Strategy Using the Fibronectin-Derived Integrin-Inactivating Peptide. Mol. Basel Switz. 2020, 25, 3239. [Google Scholar] [CrossRef] [PubMed]
  212. Iyoda, T.; Ohishi, A.; Wang, Y.; Yokoyama, M.-S.; Kazama, M.; Okita, N.; Inouye, S.; Nakagawa, Y.; Shimano, H.; Fukai, F. Bioactive TNIIIA2 Sequence in Tenascin-C Is Responsible for Macrophage Foam Cell Transformation; Potential of FNIII14 Peptide Derived from Fibronectin in Suppression of Atherosclerotic Plaque Formation. Int. J. Mol. Sci. 2024, 25, 1825. [Google Scholar] [CrossRef] [PubMed]
  213. Lee, A.C.-L.; Harris, J.L.; Khanna, K.K.; Hong, J.-H. A Comprehensive Review on Current Advances in Peptide Drug Development and Design. Int. J. Mol. Sci. 2019, 20, 2383. [Google Scholar] [CrossRef] [PubMed]
  214. Liu, Z.; Ji, P.; Liu, H.; Yu, L.; Zhang, S.-M.; Liu, P.; Zhang, X.-Z.; Luo, G.-F.; Shang, Z. FNIII14 Peptide-Enriched Membrane Nanocarrier to Disrupt Stromal Barriers through Reversing CAFs for Augmenting Drug Penetration in Tumors. Nano Lett. 2023, 23, 9963–9971. [Google Scholar] [CrossRef] [PubMed]
  215. Ge, M.; Hu, L.; Ao, H.; Zi, M.; Kong, Q.; He, Y. Senolytic Targets and New Strategies for Clearing Senescent Cells. Mech. Ageing Dev. 2021, 195, 111468. [Google Scholar] [CrossRef] [PubMed]
  216. Zhang, L.; Pitcher, L.E.; Prahalad, V.; Niedernhofer, L.J.; Robbins, P.D. Targeting Cellular Senescence with Senotherapeutics: Senolytics and Senomorphics. FEBS J. 2023, 290, 1362–1383. [Google Scholar] [CrossRef] [PubMed]
  217. Childs, B.G.; Gluscevic, M.; Baker, D.J.; Laberge, R.-M.; Marquess, D.; Dananberg, J.; van Deursen, J.M. Senescent Cells: An Emerging Target for Diseases of Ageing. Nat. Rev. Drug Discov. 2017, 16, 718–735. [Google Scholar] [CrossRef]
  218. Pang, X.; He, X.; Qiu, Z.; Zhang, H.; Xie, R.; Liu, Z.; Gu, Y.; Zhao, N.; Xiang, Q.; Cui, Y. Targeting Integrin Pathways: Mechanisms and Advances in Therapy. Signal Transduct. Target. Ther. 2023, 8, 1. [Google Scholar] [CrossRef]
  219. Kitsugi, K.; Noritake, H.; Matsumoto, M.; Hanaoka, T.; Umemura, M.; Yamashita, M.; Takatori, S.; Ito, J.; Ohta, K.; Chida, T.; et al. Inhibition of Integrin Binding to Ligand Arg-Gly-Asp Motif Induces AKT-Mediated Cellular Senescence in Hepatic Stellate Cells. Mol. Cell. Biochem. 2023, 1–14. [Google Scholar] [CrossRef] [PubMed]
Table 1. Summary of major matricellular proteins in the induction of cellular senescence.
Table 1. Summary of major matricellular proteins in the induction of cellular senescence.
Matricellular
Proteins
Binding
Receptors
Roles in Cellular SenescenceTarget Cell/TissuePossible Relevance to
Disease Onset/Progression
Ref.
CCN1Integrin α6β1InductionWound granulation tissue of mouse skin,
Human fibroblasts
Anti-fibrogenic response[23]
Integrin α6β1InductionMouse model of CCl4 induced liver fibrosis,
Hepatic stellate cells (Mouse, Human),
Portal fibroblasts
Protective role in fibrosis[26]
NDInductionHuman chondrocytesProgression of OA[27]
Integrin α6β1InductionHuman non-small-cell lung carcinoma cellsDecreased cancer
aggressiveness
[28]
NDInductionMouse muscle progenitor cellsProgression of sarcopenia[29]
NDInductionCerebral endothelial cells isolated from aged mice-[30]
NDInductionHuman cytotrophoblast cellModulation of preeclampsia[31]
CCN2Integrin α6β1InductionWound granulation tissue of mouse skin,
Human fibroblasts
Anti-fibrogenic response[24]
CCN3NDInductionMouse articular chondrocytesModulation of OA[32]
NDInductionHuman cytotrophoblast cellModulation of preeclampsia[31]
CCN4Integrin αVβ3SuppressionHuman chondrocytes isolated from knee OAProtective role in OA[33]
PeriostinIntegrin αVβ3InductionHuman nucleus pulposus cells,
Human annulus fibrosus cells
Progression of IDD[34]
TSP-1CD47InductionMouse endothelial cellsModulation of endothelial
dysregulation
[35]
CD47InductionHuman pulmonary artery endothelial cells,
Human aortic endothelial cells, mouse lung
-[36]
NDInductionMouse fibroblasts,
Epithelial cells from LSL-KRASG12D mice,
Mouse model of KrasG12D-mediated lung tumorigenesis
Decreased cancer
aggressiveness
[37]
FSTL1NDInductionHuman alveolar type II cellsProgression of pulmonary
fibrosis
[38]
TLR4InductionHuman nucleus pulposus cells tissues from IDD,
Rabbit IDD model
Progression of IDD[39]
OPNNDSuppressionHuman articular chondrocytes,
Rat knee joint cartilage
Protective role in OA[40]
Gal-3NDSuppressionMouse embryonic fibroblasts,
Human foreskin fibroblasts,
Human gastric carcinoma cell lines
Decreased cancer
aggressiveness
[41,42]
PAI-1NDInductionHuman fibroblasts, Mouse fibroblasts, Human breast cancer cells,
Human retinal pigment epithelial cells
-[43,44,45]
NDInductionAlveolar type II cells (Rat, Mouse)Progression of pulmonary
fibrosis
[46]
NDInductionHuman umbilical vein endothelial cellsProgression of atherosclerosis[47]
LRP1InductionHuman coronary artery smooth muscle cells,
Mouse aorta
Progression of atherosclerosis[48]
PEDFNDSuppressionHuman mesenchymal stem cellsExpansion of MSCs[49]
NDSuppressionMouse retinal pigment epithelium-[50]
TNCβ1-integrinInductionHuman fibroblastsIncreased cancer
aggressiveness
[25]
CCN, centralized or cellular communication network; TSP, thrombospondin; FSTL1, Follistatin-like 1; OPN, osteopontin; Gal, galectin; PAI, plasminogen activator inhibitor; PEDF, pigment epithelium-derived factor; TNC, tenascin-C; TLR4, toll-like receptor 4; LRP, low-density lipoprotein receptor-related protein; CCl4, carbon tetrachloride; OA, osteoarthritis; IDD, intervertebral disc degeneration; MSC, mesenchymal stem cell; ND, not determined.
Table 2. Drugs targeting matricellular proteins under active clinical development and focused on in this review.
Table 2. Drugs targeting matricellular proteins under active clinical development and focused on in this review.
AgentsMode of ActionModalityDiseasesPhaseID Number
Pamrevlumab (FG-3019)Anti-CCN2AntibodyIPFPhase IIIjRCT2051210169 [160]
Pancreatic cancerPhase IIINCT03941093 [161]
Duchenne muscular dystrophyPhase IIINCT04371666 [162]
SHR-1906Anti-CCN2AntibodyIPFPhase IINCT05722964 [163]
OLX-101A (BMT-101)CCN2 expression inhibitorsiRNAHypertrophic scarPhase IINCT04877756 [164]
PRS-220Anti-CCN2Antibody Mimetics IPFPhase INCT05473533 [165]
LEM-S401CCN2 expression inhibitorsiRNAHypertrophic scarPhase INCT04707131 [166]
VT-1021TSP-1 expression inducerPeptidesSolid Tumors, GlioblastomaPhase II/IIINCT03364400 [167]
belapectinGal-3 inhibitorsPolysaccharide polymerNASHPhase II/IIINCT04365868 [168]
proLectin-MGal-3 inhibitorsCarbohydrate polymersCOVID-19Phase IINCT04512027 [169]
TB-006Anti-Gal-3AntibodyAlzheimer’s diseasePhase IINCT05476783 [170]
Selvigaltin (GB1211)Gal-3 inhibitorsSmall Molecules Hepatic impairmentPhase I/IINCT05009680 [171]
Solid TumorsPhase IINCT05913388 [172]
TM5614PAI-1 inhibitorsSmall Molecules CMLPhase IIIjRCT2031220084 [173]
Solid TumorsPhase IIjRCT2061230039 [174]
Cutaneous angiosarcomaPhase IIjRCT2021230016 [175]
SSc-ILDPhase IIjRCT2021230022 [176]
DVC1-0401PEDF expression inducer Virus vectorRetinitis pigmentosaPhase I/IIjRCT2073180024 [177]
CCN, cellular communication network; TSP, thrombospondin; Gal, galectin; PAI, plasminogen activator inhibitor; PEDF, pigment epithelium-derived factor; siRNA, small interfering RNA; IPF, idiopathic pulmonary fibrosis; NASH, nonalcoholic steatohepatitis; COVID-19, coronavirus disease 2019; CML, chronic myelogenous leukemia; SSc-ILD, systemic sclerosis-associated interstitial lung disease. Retrieved from ClinicalTrials.gov and Japan Registry of Clinical Trials, accessed on 10 March 2024.
Table 3. Roles of integrins in the induction of cellular senescence.
Table 3. Roles of integrins in the induction of cellular senescence.
IntegrinsRole in Cellular SenescenceFunctionRef.
β1-integrinInductionβPAK-interacting exchange factor induces cellular senescence in human diploid fibroblasts via persistent activation of β1-integrin[17]
InductionTenascin-C-derived peptides induce cellular senescence in human diploid fibroblasts via activation of β1-integrin[25]
SuppressionSpecific deletion of β1-integrin in a mouse model of spontaneous insulinoma formation resulted in the acquisition of a senescence phenotype in a tumor nest of β tumor cells [179]
SuppressionEpithelial specific depletion of β1-integrin promotes cellular senescence in mammary intra-epithelial neoplasm lesions in a mouse model of luminal B human breast cancer[180]
Integrin α2β1InductionIntegrin α2β1 antagonistic peptide suppresses chemotherapy-induced senescence of human bladder cancer cells[181]
SuppressionIntegrin α2β1 depletion causes cellular senescence in human melanoma cells[182]
Integrin α5β1InductionIntegrin α5β1 antagonist suppresses the ellipticine- or temozolomide-induced senescence of human glioblastoma cells [183]
Integrin α6β1InductionCCN1 induces cellular senescence in human diploid fibroblasts by binding to integrin α6β1[23]
InductionCCN1 induces cellular senescence in hepatic stellate cells and portal fibroblasts by binding to integrin α6β1[26]
InductionCCN1 induces cellular senescence in non-small-cell lung carcinoma cells by binding to integrin α6β1[28]
InductionCCN2 induces cellular senescence in human diploid fibroblasts by binding to integrin α6β1[24]
β3-integrinInductionIncreased expression of β3-integrins render mouse embryonic fibroblasts more susceptible to replicative senescence[184]
InductionIncreased expression of β3-integrins promotes replicative senescence and oncogene-induced senescence in human breast fibroblasts.[185]
Inductionβ3-integrin overexpression induces cellular senescence in human tubular cells[186]
Integrin αvβ3InductionIntegrin αvβ3 antagonist inhibits periostin-induced senescence in human nucleus pulposus cells[34]
SuppressionTreatment with function-blocking integrin αvβ3 antibody induces senescence in human articular chondrocytes[33]
Suppressionβ3-integrin depletion and treatment with the f function-blocking integrin αvβ3 antibody induce senescence in glioblastoma cells[187]
β4-integrinInductionβ4-integrin depletion inhibits senescence of human umbilical vascular endothelial cells[188]
Suppressionβ4-integrin depletion induces senescence of human bronchial epithelial cells[189]
Integrin α6β4InductionIntegrin α6β4 dimerizes, is activated, and induces premature senescence in cancer cells when they are exposed to ionizing radiation[190]
PAK, p21-activated kinase; CCN, cellular communication network.
Table 4. Integrin inhibitors capable of suppressing cellular senescence and/or SASP acquisition.
Table 4. Integrin inhibitors capable of suppressing cellular senescence and/or SASP acquisition.
AgentMode of ActionModalityFunctionRefs.
RGD peptideIntegrin antagonist PeptideSuppression of βPAK-interacting exchange factor-induced cellular senescence in human fibroblasts[17]
TFA peptideIntegrin α2β1 antagonistPeptideSuppression of chemotherapy-induced senescence of primary bladder cancer cells and the human bladder cancer cell line T24 in a 3D collagen I gel model and in T24-bearing mice[181]
CilengitideIntegrin αvβ3 and αvβ5 antagonist PeptideReduction in the SASP secreted from oncogene-induced senescence in human fibroblasts[185]
GSK3008348Integrin αvβ6 antagonist Small moleculeReduction in the SASP secreted from bleomycin-induced senescence in organoids comprising human alveolar epithelial cells and human fibroblasts[209]
T1 peptideIntegrin α6β1-binding peptidePeptideSuppression of CCN1-induced senescence in human fibroblasts[23]
GoH3α6-integrin neutralizing antibodyAntibodySuppression of CCN1- or CCN2-induced senescence in human fibroblasts[23,24]
SJ749Integrin α5β1 antagonist Small moleculeSuppression of ellipticine-induced cellular senescence in human glioblastoma cells[183]
K34cIntegrin α5β1 antagonistSmall moleculeSuppression of ellipticine- or temozolomide-induced cellular senescence in human glioblastoma cells lines or human colon cancer cell lines[183]
Peptide FNIII14β1-integrin inactivator PeptideSuppression of TNC-derived peptide-induced cellular senescence in human fibroblasts[25]
Isoliquiritigenin β3-integrin inactivator Small moleculeInhibition of β3-integrin-mediated cellular senescence in HK-2 cells, and reduction in cellular senescence and interstitial fibrosis in the UUO mouse model[186]
RGD, arginine-glycine-aspartic acid; PAK, p21-activated kinase; SASP, senescence-associated secretory phenotype; CCN, cellular communication network; TNC, tenascin-C; HK-2, human proximal tubule epithelial cell line; UUO, unilateral ureteral obstruction.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Fujita, M.; Sasada, M.; Iyoda, T.; Fukai, F. Involvement of Matricellular Proteins in Cellular Senescence: Potential Therapeutic Targets for Age-Related Diseases. Int. J. Mol. Sci. 2024, 25, 6591. https://doi.org/10.3390/ijms25126591

AMA Style

Fujita M, Sasada M, Iyoda T, Fukai F. Involvement of Matricellular Proteins in Cellular Senescence: Potential Therapeutic Targets for Age-Related Diseases. International Journal of Molecular Sciences. 2024; 25(12):6591. https://doi.org/10.3390/ijms25126591

Chicago/Turabian Style

Fujita, Motomichi, Manabu Sasada, Takuya Iyoda, and Fumio Fukai. 2024. "Involvement of Matricellular Proteins in Cellular Senescence: Potential Therapeutic Targets for Age-Related Diseases" International Journal of Molecular Sciences 25, no. 12: 6591. https://doi.org/10.3390/ijms25126591

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop