You are currently viewing a new version of our website. To view the old version click .
Genes
  • Review
  • Open Access

8 December 2025

Chromosomal Instability and Telomere Attrition in Systemic Sclerosis: A Historical Perspective

Department of Microbiology and Immunology, Drexel University, Philadelphia, PA 19129, USA
This article belongs to the Special Issue The 15th Anniversary of Genes: Feature Papers in the Human Genomics and Genetic Diseases Section

Abstract

Background/Objectives: Systemic sclerosis (SSc) is a rare, complex autoimmune disease characterized by fibrosis of the skin and internal organs. While its pathogenesis is not fully understood, chromosomal instability and telomere attrition have emerged as significant areas of investigation. Methods: This review provides a historical narrative perspective and synthesizes current findings on the role of these genomic anomalies in SSc pathogenesis. We synthesized findings from foundational and recent research articles investigating genotoxic factors, chromosomal aberrations, and telomere biology in SSc. Results: There is a strong historical basis for chromosomal instability in SSc, manifesting as micronuclei, translocations, and breaks. This instability is driven by clastogenic factors and oxidative stress. SSc-specific autoantibodies are implicated; anti-centromere antibodies correlate with aneuploidy and micronuclei, while anti-topoisomerase I may inhibit DNA repair. SSc is also characterized by significant telomere attrition, first reported in 1996 and now confirmed by additional genetic studies. This telomere loss is associated with reduced telomerase activity and the presence of autoantibodies against telomere-associated proteins, including shelterin components. Conclusions: We conclude that inflammation, telomere attrition, and chromosomal instability are linked in a self-perpetuating cycle that drives SSc pathogenesis. We propose that an initial inflammatory stimulus leads to reactive oxygen species production, causing telomere damage and attrition. Critically short telomeres trigger faulty DNA repair mechanisms, such as breakage–fusion–bridge cycles, resulting in chromosomal instability. This genomic damage, in turn, acts as a danger signal, further activating inflammatory pathways and creating a feedback loop that perpetuates fibrosis.

1. Introduction

Systemic sclerosis (SSc) is a rare autoimmune disease that causes fibrosis in the skin and internal organs. Specific autoantibodies develop, which frequently predict the SSc subtype [1]. Raynaud’s phenomenon is one of the earliest symptoms, often occurring before or concurrently with skin thickening [2]. Other features of SSc, such as esophageal dysmotility, kidney failure, and pulmonary and cardiac fibrosis, are associated with this disease and have garnered considerable attention over the decades, likely because they significantly affect the morbidity of the patient [3]. SSc has two major subsets: limited cutaneous (lcSSc) and diffuse cutaneous (dcSSc) disease. In lcSSc, the skin involvement is primarily contained to the face and below the knees and elbows [4]. In this disease subset, organ involvement develops slowly, and the most common autoantibody targets centromeric proteins. In contrast, dcSSc is a rapidly progressing phenotype characterized by widespread skin and internal organ involvement [4]. This subset is primarily associated with autoantibodies that target topoisomerase I and RNA polymerase III.
SSc has been extensively characterized, but more recently, chromosomal instability and telomere attrition have come to the forefront, prompting investigators to examine these anomalies in SSc. This review synthesizes and discusses the findings of these chromosomal aberrations and telomere attrition in SSc, providing a historical narrative of their investigation and current findings.

2. Chromosomal Abnormalities in SSc

The most frequently observed chromosomal rearrangement in the healthy population is balanced translocations and an inversion of chromosome 9. Balanced translocations include the Robertsonian translocation between chromosomes 13 and 14 (rob(13;14)(q10;q10)) [5]. It is found in approximately 1 in 800 individuals [6]. Another translocation occurs between chromosomes 11 and 22, designated as t(11;22)(q23;q11), which is more frequently found in males [7]. The chromosome 9 inversion inv(9)(p11q12) [5] is found in about 1.5% of the general population [8]. However, although these chromosomal rearrangements are present in the general population, the frequency of random chromosomal abnormalities can vary with age. For example, chromosomal aberrations have been reported to be three times higher in older individuals (mean age 75 years) than in younger individuals (mean age 28.5 years [9]. The onset of SSc typically occurs later in life, between the ages of 40 and 60 [10]. When it happens in the elderly (>60 years), it is associated with accelerated disease progression [10,11]. SSc does not have a familial inheritance pattern, and although rare, there are reports of families with more than one affected individual [12,13]. No single, disease-defining chromosomal rearrangement is consistently found in patients.
The Emerit group first studied chromosomal abnormalities in SSc [14,15,16,17]. They identified acentric fragments, dots, dicentric, and ring chromosomes. Other abnormalities seen were the translocation or deletion of chromosomal fragments and intrachromosomal rearrangements [16]. A microchromosome derived from chromosome 11 was reported in a patient [18]. This microchromosome was devoid of telomeric sequences and was somatically stable [18]. The telomeric fusions between sister chromatids in SSc cells have been shown to last for more than two cell generations [19]. Micronuclei are frequently seen in both lcSSc and dcSSc cohorts [20,21,22]. Overall, the chromosomal abnormalities were not confined to lymphocytes but were also found in cultured and uncultured fibroblasts and bone marrow [17]. Micronuclei are found in equal numbers in affected and unaffected fibroblasts [23]. The frequency of abnormalities in various SSc cohorts was significant, and in one instance, it was found to be as high as 95% [24]. First-degree family members also exhibited increased chromosomal aberrations, with 86% found in siblings and 68% in offspring [17], suggesting an environmental or familial abnormality.
Historically (Figure 1), the study of chromosomal instability in SSc started in 1971, when Emerit identified a clastogen in the sera and cell extracts from SSc patients [16]. The clastogenic factor in the sera was later determined to be inosine triphosphate and inosine diphosphate. When added to cell cultures from healthy donors, these compounds induced chromosomal breakage in a dose-dependent manner [25]. Emerit was also the first to propose that reactive oxygen species (ROS) were responsible for sister chromatid exchange [23] and could be a key driver of the genotoxic effects observed in SSc [26]. The discovery that superoxide dismutase could reduce chromosomal aberrations was significant, highlighting ROS as a key driver [27]. The oxidative metabolic activity found in the blood of SSc patients was significantly higher than that of normal individuals [28], and these patients produce more of the superoxide anion (O2 *-) [29] and have more active NADPH oxidase activation [30,31]. Another study found that patients had lower overall levels of superoxide dismutase activity and total antioxidant activity [32], suggesting that ROS overwhelmed the patients’ antioxidant defenses, thereby increasing their risk of additional genotoxic effects. In SSc, urinary levels of 8-hydroxy-2′-deoxyguanosine, a marker of oxidative DNA damage, were significantly higher than those in controls [33].
Figure 1. Historical observations of chromosomal instability in SSc. Chromosomal instability and various genetic alterations have been studied sporadically for the last 50+ years. Created in BioRender. Artlett, C. (2025) https://BioRender.com/lbjflxo. Accessed 19 November 2025.
Several pre-clinical studies have investigated various antioxidants as potential therapeutics for SSc. The antioxidant N-acetylcysteine supported the findings that this molecule reduced fibrosis in both in vitro and in vivo studies [34]. Epigallocatechin-3-gallate, a potent antioxidant and the most abundant polyphenol in green tea, was found to reduce collagen expression, collagen gel contraction, and suppress intracellular ROS, ERK1/2 kinase signaling, and NF-κB activity in SSc cells [35]. One open-labeled study demonstrated an improvement in skin thickness in SSc patients who applied a cream containing 0.6 mg/mL superoxide dismutase [36]. The caveats to this study were that it was open-label and involved a small patient cohort. Although intriguing, to date, no additional studies using this cream have been undertaken to validate this observation. Overall, the studies suggest that redox regulation plays a strong role in the pathogenesis of SSc and may contribute to the development of chromosomal abnormalities.
Using an assay to measure acquired genetic damage, Roberts-Thomson et al. [37] found that SSc patients had a higher proportion of mitotic mutations in the glycophorin A gene. This mutation rate was higher than that of the age-matched controls, and the authors propose an aberrant response to DNA damage and repair in this disease. More recent analyses have shown that SSc has genomic alterations in the variable tandem number repeats [38]. All members in each family analyzed were confirmed using HLA typing. This study was the first to demonstrate that the tandem repeats could be altered in approximately 40% of SSc patients and fibroblast cell lines, with lymphocytes also being affected. Some of the repeats were correctly transmitted through the family, but had an altered size in the proband [38]. In a study using the comet assay, Palomino et al. [39] reported that SSc patients had increased DNA damage, and it was equally prevalent in both lcSSc and dcSSc. The authors proposed that patients with SSc had a less efficient DNA repair process, thus leading to the increased genomic instability seen in lymphocytes [39]. Hypermutation is substantially increased in SSc-lesional skin fibroblasts, and these mutations occur in genes involved in genomic integrity (histone integrity, histone acetylation, helicase, and DNA methylation), comparable to levels observed in some cancers [39].
Loss of centromeric sequences was recently demonstrated in dcSSc patients, with significant deletions on chromosomes 1 and 2, and a specific centromeric expansion on chromosome 19 was also noted [22]. Aneuploidy was reported in 10–50% of chromosome spreads in dcSSc, and there was a loss of centromere identity in the observed micronuclei. The extent of the micronuclei positively correlated with ROS levels [22]. The most recent study in 2024 reported an increase in single-base substitutions (SBS) in SSc patients [40]. The most prevalent signature was the SBS93 mutation signature, which is a prominent feature found in various cancers (Catalog of Somatic Mutations in Cancer, Wellcome Sanger Institute, Saffron Walden, UK). This study also found that doublet base substitutions, insertions, and deletions were elevated in SSc, along with mutation clustering. Loss of heterozygosity was also more often seen in SSc fibroblasts [40]. Overall, like the authors, we also question whether chromosomal instability also places the patient at risk for cancer, as the incidence of cancer is markedly increased in SSc patients [41,42,43,44].
Overall, there is likely an unknown environmental factor, alongside other significant risk factors, such as genetics, which contribute to chronic inflammation in SSc. Chronic inflammation enhances fibroblast proliferation and differentiation and increases the release of ROS and clastogenic factors that damage DNA. The damaged DNA leads to chromosomal abnormalities seen in SSc cells, including acentric fragments, dots, dicentric and ring chromosomes, and increased sister chromatid exchange (Figure 2).
Figure 2. Hypothetical mechanism for the chromosomal instability seen in SSc. Created in BioRender. Artlett, C. (2025) https://BioRender.com/f41g136. Accessed 19 November 2025.

3. Key SSc Autoantibodies and Chromosomal Instability

Two of the most frequently observed autoantibodies in SSc patients are those targeting centromeric proteins and topoisomerase I. These autoantibodies tend to be mutually exclusive; however, when found together, patients exhibit a more severe form of the disease [45]. An additional antibody, less frequently observed, is directed at RNA polymerase III [46].
Autoantibodies were previously considered impervious to the nucleus; however, evidence suggests otherwise. A subset of anti-DNA autoantibodies in systemic lupus erythematosus was found to penetrate the nuclei of live cells, damaging the DNA or inhibiting its repair [47,48,49,50,51,52]. This suggests that the autoantibodies found in SSc may also penetrate the nucleus and affect their target antigens; however, this has not yet been proven for all SSc autoantibodies.

3.1. Centromeric Autoantibodies

The autoantibodies (CENP-A, CENP-B, and CENP-C) against centromeric proteins are primarily found in the lcSSc subset and have been shown to correlate with aneuploidy and chromosomal breaks in SSc patients [53]. Patients who were positive for CENP-A, CENP-B, or CENP-C had significantly more aneuploidy than those who were negative for these autoantibodies (e.g., the dcSSc cohort). However, SSc patients overall (those positive for centromeric autoantibodies and those negative) had substantially greater aneuploidy than the control group. The authors propose that a correlation exists between the presence of autoantibodies to CENP-A, CENP-B, or CENP-C and chromosomal aneuploidy, suggesting that aneuploidy may result from nondisjunction secondary to centromeric dysfunction [53]. In support of this hypothesis, they found that patients with an autoantibody to CENP-C had a higher incidence of chromosomal aneuploidy compared to those without this autoantibody. CENP-A and CENP-B are present on both functional and inactivated centromeres. In contrast, CENP-C localizes to the inner plate of the kinetochore of only functional centromeres, suggesting a causal link to aneuploidy [53]. Further studies have now shown that CENP-A also localizes to the inner plate of the kinetochore at the centromere, which is associated with a satellite DNA complex [54]. In contrast, CENP-B localizes to centromeric heterochromatin beneath the kinetochore [55].
Micronuclei are predominantly found in SSc cells and are thought to reflect the chromosomal damage observed in this disease. Those with lcSSc had significantly higher micronuclei frequencies than those with dcSSc. These results appeared to reflect the autoantibody profile, in which ACA-positive SSc patients had higher micronuclei frequencies than ACA-negative patients [21]. These results support the authors’ proposal that micronuclei correlate with the autoantibody profile and likely cause cytogenetic damage. These observations were confirmed in two additional studies by Fagone [20] and Patterson [56], where both reported a high correlation with micronuclei and the centromeric autoantibody. These key observations are presented in Table 1.

3.2. Topoisomerase I Autoantibodies

Topoisomerase I catalyzes the nicking of DNA, promoting its relaxation and unwinding. This relieves torsional strain in supercoiled DNA during transcription and replication [57]. The autoantibody was initially designated as Scl70, as it was identified in the sera of dcSSc patients and recognized an antigen that had a molecular weight of approximately 70 kDa [58]. This antibody was later identified to be topoisomerase I [59]. Recent studies by May et al. found that Scl70 can penetrate the nucleus of cells, directly inhibiting the activity of topoisomerase I [60]. These elegant studies showed that the transfer of the autoantibody into the nucleus, at least in part, seemed to be dependent on lipid rafts [60]. Currently, there are no direct assessments of chromosomal abnormalities associated with this autoantibody, and, to date, only associations have been reported. In patients with the topoisomerase I autoantibody, there was a prevalence of unstable DNA breaks, overall supporting a clastogenic effect on DNA and the possible interference with protective cellular mechanisms that typically stabilize DNA breaks [20]. Micronuclei have been reported in dcSSc cohorts with the Scl70 autoantibody, but at a lower frequency than in those with the centromeric autoantibody [20,21,56].
However, we speculate that if Scl70 can enter the nucleus and inhibit topoisomerase I activity, it may reflect some of the DNA-damaging effects observed with topoisomerase I inhibitors used in cancer treatments. Topoisomerase I causes single-strand nicks to relax the DNA for replication [61]. Chemical inhibitors of topoisomerase trap the enzyme after it has made the DNA cut, preventing it from resealing the DNA. This creates the Topo I-DNA cleavable complex [62,63]. When the DNA replication fork encounters this complex, it collapses, converting the initial single-strand nick into a double-strand break [62]. The consequences of this process lead to chromosomal fragmentation, translocations, and sister chromatid exchanges [64]. A recent study showed that the topoisomerase autoantibody prevents the formation of the Topo I-DNA cleavable complex [60]. When the replication fork encounters transcription complexes, this causes genomic instability [65]. A summary of these key observations is presented in Table 1.
Table 1. Key observations between chromosomal instability and the SSc autoantibody profile.
Table 1. Key observations between chromosomal instability and the SSc autoantibody profile.
Autoantibody ProfileKey Findings and ImplicationsReferences
Autoantibodies against centromeric proteins CENP-A, CENP-B, and CENP-C autoantibodies can penetrate the nucleus and are associated with increased aneuploidy, chromosomal breaks, and elevated micronuclei [20,21,53,56]
Autoantibody against topoisomerase I (Scl70)Scl70 can penetrate the nucleus. Scl70 can prevent the formation of the Topo I-DNA complex, impede replication, and cause genomic instability. Patients with Scl70 have been associated with unstable DNA breaks and micronuclei.[60]
RNA polymerase IIIRNA polymerase III can repair double-stranded DNA breaks. It is currently unknown whether this autoantibody penetrates the nucleus, but if it does, it could affect RNA polymerase III function, contributing to DNA damage.[66]

3.3. RNA Polymerase III Autoantibodies

The presence of an autoantibody that targets RNA polymerase III is associated with a severe and rapidly progressive form of SSc, characterized by widespread and rapid skin thickening and renal crisis, which is marked by acute kidney failure and malignant hypertension [67,68]. This autoantibody is found in approximately 11% of patients [69], but its prevalence varies by ethnicity and geographic location [70]. RNA polymerase III is a housekeeping gene that specializes in transcribing 5S rRNA, transfer RNAs, and the U6 spliceosomal RNA, all of which play a primary role in translation and related biological processes [71]. Currently, no direct studies have shown that this autoantibody penetrates the nucleus; however, if it did, it would likely cause substantial disruption to DNA repair, as emerging research has demonstrated that RNA polymerase III also plays a role in repairing DNA double-strand breaks [66]. Thus, if the RNA polymerase III autoantibody could cross the nuclear membrane, it could compromise the cell’s ability to repair DNA damage, leading to genomic instability. A summary of these key observations is presented in Table 1.

4. SSc and Telomere Attrition

Telomeres were first discovered in the 1930s by Hermann Müller [72]. They are repetitive hexameric DNA units located at the ends of chromosomes, serving to protect chromosome ends from being recognized and improperly repaired as double-stranded breaks. The most well-known of these sequences is the repeat sequence TTAGGG, which extends between 10 and 15 kilobases in newborns [73]. It is this sequence that is primarily lost during DNA replication. However, the telomere cap also comprises other non-randomly distributed consensus sequences. These are found in the subtelomeric regions and contain TTGGGG and TGAGGG repeat sequences, which play crucial and nuanced roles in regulating the structure and function of the chromosome ends. The telomere is stabilized by shelterin, and the shift from high-affinity (TTAGGG) to low-affinity variants (TTGGGG and TGAGGG) helps mark the transition from the telomere to the rest of the chromosome. Furthermore, reduced shelterin binding in this subtelomeric region can make the chromatin more open and accessible, allowing other regulatory proteins to bind. The G-rich strand overhangs the C-rich strand by 12–16 residues [74] and plays a central role in the function of the telomere [75]. With every round of cell division, 50–200 nucleotides of the TTAGGG sequence are lost. This shortening acts as a biological clock, and once telomeres become critically short, the cell’s DNA damage response is activated, triggering either cellular senescence or apoptosis [76]. Thus, telomere length predicts a cell’s replicative capacity [77].
To shield the telomeres from this response, a protein complex called shelterin binds to the chromosome ends. In some cells, the enzyme telomerase, a reverse transcriptase that uses an RNA template to synthesize new telomere DNA, counteracts this shortening. Telomerase is highly expressed in embryonic stem cells and specific adult cells with high turnover, such as lymphocytes. In most somatic cells with low telomerase activity, the alternative lengthening of telomeres pathway can maintain telomere length via homologous recombination. However, before cell death or senescence occurs, the shortened telomeres can lead to chromosomal aberrations, such as dicentric chromosomes, chromosomal breaks, deletions, and translocations [78,79,80].
Telomerase is an enzyme that elongates telomeres. In adults, it is highly active when cellular longevity is needed, such as during high rates of division, as in stem cells and germ cells. Telomerase contains a telomere-specific reverse transcriptase that adds repeats to the end of the telomeres. Telomerase activity is regulated by the shelterin complex, which comprises six proteins. These proteins are telomeric repeat binding factor-1, telomeric repeat binding factor-2, protection of telomeres-1, adrenocortical dysplasia homolog, TERF1-interacting nuclear factor-2, and repressor/activator protein-1 [81].
SSc was first associated with shortened telomeres in 1996 [82]. At the time, we hypothesized that the telomere attrition we observed could be the source of chromosomal damage and various aberrations resulting from telomeric fusions [82]. We found that the average loss of telomeric sequences was approximately 3 kb greater than in controls, and this was not related to patient age or disease duration [82]. Intriguingly, when analyzing peripheral blood mononuclear cells by Southern blotting, we did not observe a general shift in the telomeric smear, as seen in tumor cell lines [72], but instead we saw a broadening of the smear. We speculated at the time that this could imply that not all chromosomes have shortened telomeres, or not all cells have undergone telomere attrition. We also found that SSc family members, including spouses, had shorter telomeres than controls [82], suggesting an environmental exposure related to the familial chromosomal abnormalities identified by Emerit [17].
One investigation found that a subset of SSc patients had shorter telomeres in lymphocytes but not granulocytes [83]. This observation is interesting because granulocytes, particularly neutrophils, have a significantly faster, more dramatic turnover rate during inflammation than lymphocytes. Neutrophils may play a role in SSc, and they are found to be elevated, correlating with disease severity and tissue damage [84]. As they are elevated and turn over more rapidly, it would be expected that this cell phenotype exhibits telomere attrition.
Two more studies found shorter telomeres in SSc patients with interstitial lung disease and reported them to be substantially shorter in lcSSc than in dcSSc, even when age was taken into consideration [85,86]. Surprisingly, telomere attrition in the Liu study [85] study was greater in patients who did not exhibit any of the common autoantibody profiles associated with SSc. Furthermore, using longitudinal analysis, they found that shorter telomeres were associated with an increased risk for worsening interstitial lung disease [85]. Further support for telomere loss in this study reports substantial loss of telomeric sequences in both lcSSc and dcSSc [87]. Like the first publication investigating telomere loss in SSc [82], they also did not find a correlation between telomere attrition and disease duration [87]. However, contrary to these studies, MacIntyre et al. [88] found increased telomere lengths in an lcSSc cohort, and it is the only research to report this finding. To overcome these discrepancies, a Mendelian randomization study was conducted in 2023 to investigate the relationship between telomere length and SSc [89]. This study reported that shorter telomere lengths were associated with an increased risk of SSc onset, overall supporting the generalized finding that shorter telomeres are found in SSc [82,83,85,87]. However, contrary to the MacIntyre study [88], they observed that the lcSSc patients had shorter telomeres than controls [89]. In a study investigating different connective tissue diseases (rheumatoid arthritis, systemic lupus erythematosus, Sjogren’s syndrome, and SSc), telomerase activity in SSc was found to be significantly lower in lymphocytes than in the other connective tissue diseases and was substantially lower than that of the control cohort [90] and this may be associated with genotype frequencies in the telomerase gene [91].

Autoantibodies Targeting Telomere-Associated Proteins in SSc

Recently, researchers have identified a range of autoantibodies targeting telomerase and its associated proteins, shedding new light on the pathogenesis of SSc and its association with shortened telomeres [92,93,94], and suggesting a potential role for telomere dysfunction in the underlying disease processes. Although rare, these autoantibodies target key components of the cellular machinery that maintain telomeres. About 3% of SSc patients have autoantibodies against the catalytic subunit of telomerase, hTERT [92]. In the shelterin complex (a group of six proteins that bind telomeres), telomeric repeat-binding factor-1 is one of the more frequently identified autoantigens in this category and was found in 9% of patients [92]. The autoantibody against telomeric repeat-binding factor-1 was associated with shorter telomeres in patients with the autoantibody than in those without it. Furthermore, this observation was specific to SSc, as patients with other autoimmune diseases exhibited the same frequency as the controls. However, a similar frequency of the anti-telomere repeat binding factor-1 was observed in patients with idiopathic pulmonary fibrosis, suggesting that telomere-targeting antibodies might trigger fibrotic lung disease [92]. Other, less frequently observed autoantibodies were found to target telomeric repeat binding factor-2, protection of telomeres-1, adrenocortical dysplasia homolog, TERF1-interacting nuclear factor-2, and repressor/activator protein-1 [92]. Beyond the core telomerase and shelterin proteins, studies have also identified autoantibodies against other proteins associated with telomere biology and maintenance. These include THO complex, homeobox-containing-1, RuvBL1, and RuvBL2 [93,94,95]. A timeline of significant findings on telomere attrition and telomere-associated autoantibodies is depicted in Figure 3.
Figure 3. Timeline for the identification of telomere attrition and telomerase-associated autoantigens. Telomere attrition in SSc has received little attention, but in recent years, there has been increased interest in understanding why telomeric sequences are lost at a greater rate in SSc. Created in BioRender. Artlett, C. (2025) https://BioRender.com/il4sfod. Accessed 19 November 2025.

5. Telomere Attrition and Chromosomal Instability in SSc-like Fibrotic Diseases

The tendency for chromosomal breakage and rearrangements, as well as clastogenic factors, has been reported in other diseases and may, in fact, be a feature of inflammation rather than a disease-specific characteristic of SSc. Clastogenic factors have been found in Bloom syndrome [96], lupus [27,97], rheumatoid arthritis [27,98], psoriasis [99], and hepatitis C [100]. They are also found in the autoimmune-prone New Zealand black mouse [27].
SSc and SSc-like diseases have been associated with various occupational exposures, and these chemicals have also been linked to chromosomal abnormalities. Increased chromosomal abnormalities have been associated with cyclophosphamide in SSc and rheumatoid arthritis, suggesting that this chemical increases the risk of chromosomal instability [101]. Various environmental exposures have been linked to SSc and SSc-like illnesses, and these exposures are also associated with chromosomal abnormalities (Table 2). The majority of these associations between SSc and various chemicals, solvents, or environmental toxins are primarily based on case studies or small cohorts, and, to date, it remains unclear whether these observations will hold up to scrutiny in larger patient groups. Studying causative chemical exposures in SSc is difficult, as we are all exposed to various chemicals in daily life. Whether this contributes to disease in certain individuals with specific genetic backgrounds will require intensive study.
Table 2. Chemicals Associated with SSc-like illnesses and Chromosomal Instability *.
The most well-known chemical that induces SSc-like disease in humans is bleomycin. Bleomycin is a glycopeptide antibiotic used as a chemotherapeutic agent. Bleomycin is now more extensively used to induce collagen synthesis in animal models of fibrotic diseases [138]. In humans, the drug-induced pathology is very similar, and they show skin hyperpigmentation, Raynaud’s phenomenon, and interstitial lung disease; however, these patients do not exhibit the classic autoantibodies that are diagnostic of SSc [139,140,141]. Bleomycin enters the cell and generates ROS, which breaks the DNA [142]. Paramagnetic resonance studies showed that bleomycin generates ROS, including superoxide, hydrogen peroxide, and the hydroxyl radical [143,144]. It was previously thought that ROS were primarily responsible for DNA damage; however, it is now accepted that bleomycin reacts with iron and oxygen to damage susceptible DNA sites [145]. The damage caused by bleomycin includes double-strand breaks, translocations, deletions, and the formation of dicentric chromosomes [102,103,104,105,106].
Vinyl chloride was one of the first chemicals associated with features mimicking SSc [107,108,109,110]. It can induce Raynaud’s phenomenon and scleroderma-like skin thickening in some individuals [111]. Vinyl chloride is metabolized in the liver into highly reactive intermediates, primarily chloroethylene oxide and chloroacetaldehyde, which can form DNA adducts that interfere with DNA replication and repair [146]. This leads to genetic mutations and large-scale chromosomal abnormalities in exposed individuals [112]. The types of chromosomal abnormalities observed in SSc, such as aneuploidy, breaks, acentric fragments, dicentric chromosomes, and sister chromatid exchange, are also observed with excessive exposure to vinyl chloride [112].
Other chemicals associated with SSc-like illnesses include trichloroethylene [113,114,115,116] and benzene [119,147,148], among other chemicals (Table 2). These chemicals also cause chromosomal instability upon exposure [122,149]. More recently, toxic oil syndrome [124,150] and nephrogenic fibrosing dermopathy have been associated with SSc-like diseases [151,152]. The cause of toxic oil syndrome in Spain in 1981 was the consumption of adulterated rapeseed oil intended for industrial use that was fraudulently sold as olive oil [153]. The industrial oil was contaminated with aniline, which is metabolized to phenylhydroxylamine and nitrosobenzene. These substances induce the production of ROS, which causes single- and double-strand DNA breaks [125]. Approximately 20,000 were exposed, and 1200 died [154]. Of those who survived, they developed a chronic neuromyopathy and scleroderma-like illness with similar features of collagen vascular disease [153] with overlapping forms of eosinophilic fasciitis [124]. In nephrogenic fibrosing dermopathy, gadolinium is the contrast agent that can bind non-covalently to DNA, causing indirect damage and inducing genotoxic effects, such as the formation of micronuclei and nuclear buds, likely through the generation of oxidative stress [155,156].
Several meta-analyses have reported associations between chemical exposures and the onset of SSc. Regarding occupational exposure to organic solvents, meta-analyses or systematic reviews reported increased relative risk and odds ratios with a greater risk in men [118,135,157,158,159]. SSc has been associated with geographical clustering in some areas, suggesting increased exposures to pollution or environmental toxins [160,161,162,163,164], although this observation did not always hold up to further analysis [165,166].

6. Inflammatory Triggers of Telomere Attrition and Chromosomal Instability in SSc and SSc-like Fibrotic Diseases

Currently, in SSc, and indeed in many other fibrotic diseases, we do not know what the trigger is that initiates fibrosis. However, once inflammation starts, it is a self-perpetuating mechanism. Inflammation and oxidative stress are core features that can cause telomere attrition and chromosomal damage. One fundamental activator of inflammation is the inflammasome [138] and other inflammatory pathways that drive fibrosis play a role [167]. Though not discussed here, the inflammasome is strongly correlated with SSc fibrosis [138], and can increase fibroblast turnover rates [168] and promote fibroblast differentiation [138]. The inflammasomes can be activated by oxidative stress [169] and discussed above SSc has elevated ROS. Furthermore, DNA damage can activate the inflammasome and cGAS-STING [22]. The other inflammatory pathways involved, such as AKT, G-protein-coupled receptors, MAP kinases, and WNT signaling, can promote cell proliferation [167]. To repair the damage, fibroblasts must divide more frequently to replace those lost or injured and this would contribute to telomere attrition. During inflammation, ROS are produced, and telomeres are highly susceptible to oxidative damage because the guanine base is readily oxidized [170]. The damage creates 8-oxo-deoxyguanosine lesions in the telomere [171,172], and if this modification escapes repair, it could lead to the misincorporation of an A within the telomere repeat [173]. Barnes et al. show that 8-oxo-deoxyguanosine disrupts telomere replication, increasing their fragility [174], and Fauquerel showed that this led to telomere loss [175]. NADPH oxidase-mediated superoxide production is strongly associated with shorter telomeres and atherosclerosis [176], implicating a correlation with telomere shortening in SSc [82] with its increased NADPH activity [31]. We believe that if the ROS production exceeds the available antioxidants, as seen in SSc [32], the cell’s ability to repair the sheer number of breaks is overwhelmed. This faulty repair could lead to deletions, translocations, and other chromosomal aberrations seen in SSc. Here, it is likely that oxidative stress from inflammation is the primary culprit. ROS are potent clastogens that cause both single-strand and double-strand breaks. Once telomeres become critically short, the cell’s machinery can no longer distinguish the natural chromosome end from a double-strand break, and the cell tries to fix this by fusing it to another chromosome. This faulty repair can create a dicentric chromosome, which undergoes a random break during cell division. This initiates a catastrophic breakage-fusion-bridge cycle that leads to massive and ongoing chromosomal instability [177]. This abnormality occurs when the telomere breaks or is lost, leading to the fusion of sister chromatids. During cell division, the fused structure forms a bridge between the separating poles of the cell, which then breaks unevenly [178]. This creates daughter cells with unstable, telomere-less chromosomes such as that seen in SSc [18], allowing the cycle to repeat.
We question whether these genomic alterations in SSc are a specific feature of the disease or a generalized phenomenon of fibrosis caused by increased inflammation and oxidative stress. Several other fibrotic disorders also feature chromosomal instability, telomere attrition, and loss of heterozygosity. Werner’s syndrome is caused by a mutation in the Werner gene, which is involved in DNA repair. There is a loss of heterozygosity that leads to genomic instability, accelerated cell senescence, and the characteristic skin-like fibrosis seen in SSc patients [179,180]. Ataxia-telangiectasia is caused by mutations in the ataxia-telangiectasia gene, another master regulator of DNA damage repair. While this is not a classic fibrotic disease, the loss of a functional gene contributes to oxidative stress and chronic inflammation, which can drive fibrosis in some patients [181,182,183]. In idiopathic pulmonary fibrosis, a significant subset of familial and even sporadic cases is caused by mutations in genes essential for telomere maintenance [184,185]. Even without mutations, accelerated telomere shortening is often observed in lung epithelial cells of patients, suggesting it is a key part of the disease process [185,186]. Dyskeratosis congenita is a rare, inherited bone marrow failure syndrome, a classic telomere biology disorder, and is caused by mutations in genes involved in telomere maintenance [187,188]. Patients often develop pulmonary fibrosis and liver cirrhosis as major complications, directly linking telomere dysfunction to fibrosis in these organs [187,188]. Aplastic anemia primarily affects the bone marrow, but some patients also develop telomere attrition, as well as pulmonary fibrosis or liver disease, further highlighting the systemic impact of telomere dysfunction [189]. Telomere shortening is also observed in hepatocytes during chronic liver injury and the progression to cirrhosis, regardless of the initial cause (e.g., viral hepatitis, alcohol, etc.). This shortening contributes to cell senescence and liver fibrosis [190]. Fanconi anemia is caused by mutations in genes involved in repairing DNA crosslinks. Fanconi anemia leads to significant chromosomal instability, and while it is primarily known for bone marrow failure and cancer risk, liver fibrosis can occur [191].

7. Discussion

The reviewed literature established a strong historical basis for the presence of significant genomic anomalies, specifically chromosomal instability and telomere attrition in the pathogenesis of SSc. We propose that these factors are linked in a self-perpetuating cycle that drives the chronic, fibrotic nature of the disease. One key finding on genomic anomalies in SSc includes chromosomal instability. SSc is marked by numerous chromosomal aberrations, including micronuclei, translocations, dicentric chromosomes, acentric fragments, and deletions, which are seen in lymphocytes, fibroblasts, and bone marrow. This instability is driven by clastogenic factors, such as inosine triphosphate and diphosphate, and by oxidative stress from heightened ROS production, which overwhelms the patient’s low antioxidant defenses. The presence of extensive genomic damage, including hypermutation (with a signature common in cancers, SBS93) and loss of heterozygosity, raises the question of whether this instability contributes to the increased incidence of cancer observed in SSc patients [41].
Research investigating clastogenic factors and ROS in SSc requires a modern mechanistic bridge to better understand their roles and define the relationship between the clastogenic factor (e.g., inosine triphosphate/diphosphate or other) and elevated NADPH oxidase activity. Understanding this mechanistic intersection might help to identify the initial biochemical trigger of chromosomal breakage. Inosine triphosphate and inosine diphosphate are central intermediates in the cell’s purine metabolism pathway. They are classified as noncanonical purine nucleotides and are not typically incorporated into DNA or RNA. Under normal conditions, they do not accumulate to high levels because inosine triphosphatase hydrolyzes them to inosine monophosphate and pyrophosphate [192]. This process prevents the incorporation of inosine into nucleic acids, which would cause DNA damage and mutagenesis [193]. The elevated ROS generated by NADPH oxidase causes irreversible damage to DNA, lipids, and enzymes. This extensive cellular damage and high metabolic turnover lead to the production of noncanonical purine nucleotides. The accumulation of inosine triphosphate/diphosphate would further drive genomic instability and inflammation, which, in turn, activates more NADPH oxidase, perpetuating the inflammatory cycle.
A second key finding is telomere attrition. The mechanism of telomere loss is primarily caused by chronic inflammation and oxidative stress, as the guanine-rich telomeres are highly susceptible to damage from ROS [194]. This damage disrupts their replication and increases telomere fragility. The critically short telomeres activate the DNA damage response, leading to faulty DNA repair (e.g., breakage–fusion–bridge cycles) and the chromosomal instability seen in the disease. While autoantibodies targeting shelterin components (e.g., TRF1) and telomerase (hTERT) have been identified in a subset of patients, their functional consequences remain to be investigated. Currently, we do not know whether they directly block telomerase activity or shelterin binding, thereby accelerating telomere loss. Further investigation of telomerase activity in various SSc cell types (lymphocytes, fibroblasts, granulocytes, etc.) is required to determine whether reduced activity is generalized or lineage-specific, and whether this relates to telomerase genotype frequencies [91].
Incorporating recent observations on the cyclic GMP–AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway, damage-associated molecular patterns (inflammasome activation), and the senescence-associated secretory phenotype would provide a clearer understanding of how the core findings of genomic instability and telomere attrition directly lead to the chronic inflammation and tissue fibrosis seen in SSc. This integration could bridge the gap between DNA damage (possibly the cause) and inflammation/fibrosis (the effect) by defining the specific signaling pathways that sense genomic stress. cGAS-STING is one critical molecular sensor that directly links genomic instability to inflammatory and immune responses. When the cell experiences significant genomic stress, fragments of damaged DNA or DNA leakage from damaged mitochondria can accumulate in the cytosol. cGAS senses this aberrant cytosolic DNA. Upon binding the DNA, cGAS produces the secondary messenger cGAMP, which then activates the adaptor protein STING. STING activation triggers the production of type I interferons and other pro-inflammatory cytokines, driving chronic inflammation. This chronic inflammation is a well-established driver of enhanced fibroblast proliferation and differentiation, ultimately leading to fibrosis. Activation of cGAS-STING has been associated with other fibrotic diseases [195,196,197,198,199] and this pathway has garnered much attention in the last 5 years. Indeed, inhibitors of cGAS or STING have been identified that ameliorate fibrosis [200,201].
Damage-associated molecular patterns are endogenous “danger signals” released by damaged or dying cells. Genomic instability directly creates these signals that can activate the inflammasomes, and this would occur in parallel with cGAS-STING. These danger signals are released during inflammation-induced cell death (apoptosis/necrosis) or when telomere attrition leads to micronuclei formation. The damaged DNA fragments function as the “on switch” for inflammation. They are known to activate the inflammasome, which is strongly correlated with fibrosis [138]. This activation releases potent pro-fibrotic cytokines, locking the tissue into an inflammatory state. Critically short telomeres, DNA damage, and chronic stress often force cells, especially fibroblasts, into a state of cellular senescence. Senescent cells, rather than lying dormant, become highly secretory. This potent secretome is characterized by the release of numerous inflammatory and profibrotic molecules, including pro-inflammatory cytokines (e.g., IL-6, IL-1α, and IL-1β), chemokines, and matrix remodeling enzymes. The secretome directly fuels the fibrotic cycle by driving further inflammation, recruiting immune cells, and altering the tissue microenvironment to favor collagen deposition and fibroblast differentiation. This ensures the vicious cycle of inflammation, genomic damage, and fibrosis is perpetuated (Figure 4).
Figure 4. A proposed model for the chronic, self-perpetuating nature of inflammation, telomere loss, and chromosomal instability. The initiating stimulus, whether an environmental toxin or pathogenic, promotes inflammation, leading to the production of ROS. This, in turn, damages DNA, causing chromosomal instability and telomere attrition. The damaged DNA causes parallel activation of cGAS-STING and inflammasomes, leading to fibroblast senescence. Senescent fibroblasts secrete proinflammatory cytokines and fibrotic mediators, thereby increasing extracellular matrix deposition, a feature of fibrosis. The proinflammatory cytokines also exacerbate inflammation, leading to increased ROS production and perpetuating the cycle. The initiating factor’s entry into the cycle is unknown, but it could directly trigger inflammation or ROS. Created in BioRender. Artlett, C. (2025) https://BioRender.com/pswefnm. Accessed 21 November 2025.
Overall, we propose a model of inflammation, ROS production, telomere attrition, and chromosomal instability that initiates a feedback loop, leading to further inflammation and DNA damage, which, in turn, increases telomere attrition and chromosomal instability. Then, the chromosomal instability and damaged DNA act as potent danger signals that further activate inflammatory pathways, such as the inflammasome [138], cGAS-STING, and other inflammatory pathways involved in fibrosis [167]. This can also trigger cellular senescence, leading cells to secrete more pro-inflammatory cytokines, which, in turn, generate even more inflammation and oxidative stress. In essence, these pathways explain that genomic damage is not just an effect but can also be a catalyst, translating the chromosomal damage signal into a chronic, self-perpetuating inflammatory and fibrotic program.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study.

Conflicts of Interest

The author declares no conflicts of interest.

References

  1. Stochmal, A.; Czuwara, J.; Trojanowska, M.; Rudnicka, L. Antinuclear Antibodies in Systemic Sclerosis: An Update. Clin. Rev. Allergy Immunol. 2020, 58, 40–51. [Google Scholar] [CrossRef] [PubMed]
  2. LeRoy, E.C. Raynaud’s phenomenon, scleroderma, overlap syndromes and other fibrosing syndromes. Curr. Opin. Rheumatol. 1993, 4, 821–824. [Google Scholar]
  3. Volkmann, E.R.; Fischer, A. Update on Morbidity and Mortality in Systemic Sclerosis-Related Interstitial Lung Disease. J. Scleroderma Relat. Disord. 2021, 6, 11–20. [Google Scholar] [CrossRef]
  4. van den Hoogen, F.; Khanna, D.; Fransen, J.; Johnson, S.R.; Baron, M.; Tyndall, A.; Matucci-Cerinic, M.; Naden, R.P.; Medsger, T.A., Jr.; Carreira, P.E.; et al. 2013 classification criteria for systemic sclerosis: An American college of rheumatology/European league against rheumatism collaborative initiative. Ann. Rheum. Dis. 2013, 72, 1747–1755. [Google Scholar] [CrossRef] [PubMed]
  5. Ren, J.; Guan, X.; Lv, W.; Yan, Y.; Si, Y.; Yang, S.; Yin, C. A retrospective analysis of 38,652 amniotic fluid karyotype. Front. Genet. 2025, 16, 1655290. [Google Scholar] [CrossRef]
  6. Poot, M.; Hochstenbach, R. Prevalence and Phenotypic Impact of Robertsonian Translocations. Mol. Syndr. 2021, 12, 1–11. [Google Scholar] [CrossRef] [PubMed]
  7. Kurahashi, H.; Inagaki, H.; Ohye, T.; Kogo, H.; Tsutsumi, M.; Kato, T.; Tong, M.; Emanuel, B.S. The constitutional t(11;22): Implications for a novel mechanism responsible for gross chromosomal rearrangements. Clin. Genet. 2010, 78, 299–309. [Google Scholar] [CrossRef]
  8. Teo, S.H.; Tan, M.; Knight, L.; Yeo, S.H.; Ng, I. Pericentric inversion 9--incidence and clinical significance. Ann. Acad. Med. Singap. 1995, 24, 302–304. [Google Scholar]
  9. Aurias, A. (Ed.) Acquired Chromosomal Aberrations in Normal Individuals; RCR Press, Inc.: Fernandina Beach, FL, USA, 1993; pp. 125–139. [Google Scholar]
  10. Manno, R.L.; Wigley, F.M.; Gelber, A.C.; Hummers, L.K. Late-age onset systemic sclerosis. J. Rheumatol. 2011, 38, 1317–1325. [Google Scholar] [CrossRef]
  11. Moinzadeh, P.; Kuhr, K.; Siegert, E.; Mueller-Ladner, U.; Riemekasten, G.; Günther, C.; Kötter, I.; Henes, J.; Blank, N.; Zeidler, G.; et al. Older age onset of systemic sclerosis-accelerated disease progression in all disease subsets. Rheumatology 2020, 59, 3380–3389. [Google Scholar] [CrossRef]
  12. Stephens, C.O.; Briggs, D.C.; Whyte, J.; Artlett, C.M.; Scherbakov, A.B.; Olsen, N.; Gusseva, N.G.; McHugh, N.J.; Maddison, P.J.; Welsh, K.I.; et al. Familial scleroderma--evidence for environmental versus genetic trigger. Br. J. Rheumatol. 1994, 33, 1131–1135. [Google Scholar] [CrossRef]
  13. Englert, H.; Small-McMahon, J.; Chambers, P.; O’Connor, H.; Davis, K.; Manolios, N.; White, R.; Dracos, G.; Brooks, P. Familial risk estimation in systemic sclerosis. Aust.N. Z. J. Med. 1999, 29, 36–41. [Google Scholar] [CrossRef]
  14. Emerit, I.; Housset, E.; de Grouchy, J.; Camus, J.P. Chromosomal breakage in diffuse scleroderma. A study of 27 patients. Rev. Eur. Etud. Clin. Biol. 1971, 16, 684–694. [Google Scholar]
  15. Emerit, I.; Levy, A.; Housset, E. Generalized scleroderma and chromosome breakage. Demonstration of a breaking factor in patients serum. Ann. Genet. 1973, 16, 135–138. [Google Scholar] [PubMed]
  16. Emerit, I.; Levy, A.; Housset, E. Breakage factor in systemic sclerosis and protector effect of L-cysteine. Humangenetik 1974, 25, 221–226. [Google Scholar] [CrossRef] [PubMed]
  17. Emerit, I.; Housset, E.; Feingold, J. Chromosomal breakage and scleroderma: Studies in family members. J. Lab. Clin. Med. 1976, 88, 81–86. [Google Scholar]
  18. Haaf, T.; Sumner, A.T.; Köhler, J.; Willard, H.F.; Schmid, M. A microchromosome derived from chromosome 11 in a patient with the CREST syndrome of scleroderma. Cytogenet. Cell Genet. 1992, 60, 12–17. [Google Scholar] [CrossRef] [PubMed]
  19. Dutrillaux, B.; Aurias, A.; Couturier, J.; Croquette, M.E.; Viegas-Pequignot, E. Multiple telomeric fusions and chain configurations in human somatic chromosomes. Chromosomes Today 1977, 6, 37–44. [Google Scholar]
  20. Majone, F.; Cozzi, F.; Tonello, M.; Olivieri, S.; Montaldi, A.; Favaro, M.; Visentin, S.; Luisetto, R.; Ruffatti, A. Unstabilized DNA breaks in lymphocytes of patients with different subsets of systemic sclerosis. Ann. N. Y. Acad. Sci. 2007, 1108, 240–248. [Google Scholar] [CrossRef]
  21. Porciello, G.; Scarpato, R.; Ferri, C.; Storino, F.; Cagetti, F.; Morozzi, G.; Bellisai, F.; Migliore, L.; Marcolongo, R.; Galeazzi, M. Spontaneous chromosome damage (micronuclei) in systemic sclerosis and Raynaud’s phenomenon. J. Rheumatol. 2003, 30, 1244–1247. [Google Scholar]
  22. Paul, S.; Kaplan, M.H.; Khanna, D.; McCourt, P.M.; Saha, A.K.; Tsou, P.S.; Anand, M.; Radecki, A.; Mourad, M.; Sawalha, A.H.; et al. Centromere defects, chromosome instability, and cGAS-STING activation in systemic sclerosis. Nat. Commun. 2022, 13, 7074. [Google Scholar] [CrossRef]
  23. Martins, E.P.; Fuzzi, H.T.; Kayser, C.; Alarcon, R.T.; Rocha, M.G.; Chauffaille, M.L.; Andrade, L.E. Increased chromosome damage in systemic sclerosis skin fibroblasts. Scand. J. Rheumatol. 2010, 39, 398–401. [Google Scholar] [CrossRef]
  24. Sherer, G.K.; Jackson, B.B.; LeRoy, E.C. Chromosome breakage and sister chromatid exchange frequencies in scleroderma. Arthritis Rheum. 1981, 24, 1409–1413. [Google Scholar] [CrossRef]
  25. Auclair, C.; Gouyette, A.; Levy, A.; Emerit, I. Clastogenic inosine nucleotide as components of the chromosomal breakage factor in scleroderma patients. Arch. Biochem. Biophys. 1990, 278, 238–244. [Google Scholar] [CrossRef]
  26. Emerit, I. Reactive oxygen species, chromosome mutation, and cancer: Possible role of clastogenic factors in carcinogenesis. Free Radic. Biol. Med. 1994, 16, 99–109. [Google Scholar] [CrossRef] [PubMed]
  27. Emerit, I.; Michelson, A.M. Chromosome instability in human and murine autoimmune disease: Anticlastogenic effect of superoxide dismutase. Acta Physiol. Scand. Suppl. 1980, 492, 59–65. [Google Scholar] [PubMed]
  28. Kovacs, I.B.; Meyrick, R.H.; Mackay, T.A.R.; Rustin, M.H.A.; Kirby, J.D.T. Increased chemiluminescence of polymorphonuclear leukocytes from patients with progressive systemic sclerosis. Clin. Sci. 1986, 70, 257–261. [Google Scholar] [CrossRef] [PubMed]
  29. Sambo, P.; Jannino, L.; Candela, M.; Salvi, A.; Donini, M.; Dusi, S.; Luchetti, M.M.; Gabrielli, A. Monocytes of patients wiht systemic sclerosis (scleroderma spontaneously release in vitro increased amounts of superoxide anion. J. Investig. Dermatol. 1999, 112, 78–84. [Google Scholar] [CrossRef]
  30. Sambo, P.; Baroni, S.S.; Luchetti, M.; Paroncini, P.; Dusi, S.; Orlandini, G.; Gabrielli, A. Oxidative stress in scleroderma: Maintenance of scleroderma fibroblast phenotype by the constitutive up-regulation of reactive oxygen species generation through the NADPH oxidase complex pathway. Arthritis Rheum. 2001, 44, 2653–2664. [Google Scholar] [CrossRef]
  31. Yasuoka, H.; Garrett, S.M.; Nguyen, X.X.; Artlett, C.M.; Feghali-Bostwick, C.A. NADPH oxidase-mediated induction of reactive oxygen species and extracellular matrix deposition by insulin-like growth factor binding protein-5. Am. J. Physiol. Lung Cell. Mol. Physiol. 2019, 316, L644–L655. [Google Scholar] [CrossRef]
  32. Sfrent-Cornateanu, R.; Mihai, C.; Stoian, I.; Lixandru, D.; Bara, C.; Moldoveanu, E. Antioxidant defense capacity in scleroderma patients. Clin. Chem. Lab. Med. 2008, 46, 836–841. [Google Scholar] [CrossRef]
  33. Avouac, J.; Borderie, D.; Ekindjian, O.G.; Kahan, A.; Allanore, Y. High DNA oxidative damage in systemic sclerosis. J. Rheumatol. 2010, 37, 2540–2547. [Google Scholar] [CrossRef]
  34. Zhou, C.F.; Yu, J.F.; Zhang, J.X.; Jiang, T.; Xu, S.H.; Yu, Q.Y.; Zhu, Q.X. N-acetylcysteine attenuates subcutaneous administration of bleomycin-induced skin fibrosis and oxidative stress in a mouse model of scleroderma. Clin. Exp. Dermatol. 2013, 38, 403–409. [Google Scholar] [CrossRef]
  35. Dooley, A.; Shi-Wen, X.; Aden, N.; Tranah, T.; Desai, N.; Denton, C.P.; Abraham, D.J.; Bruckdorfer, R. Modulation of collagen type I, fibronectin and dermal fibroblast function and activity, in systemic sclerosis by the antioxidant epigallocatechin-3-gallate. Rheumatology 2010, 49, 2024–2036. [Google Scholar] [CrossRef]
  36. Mizushima, Y.; Hoshi, K.; Yanagawa, A.; Takano, K. Topical application of superoxide dismutase cream. Drugs Exptl. Clin. Res. 1991, XVII, 127–131. [Google Scholar]
  37. Roberts-Thomson, P.J.; Male, D.A.; Walker, J.G.; Cox, S.R.; Shen, X.; Smith, M.D.; Ahern, M.J.; Turner, D.R. Genomic instability in scleroderma. Asian Pac. J. Allergy Immunol. 2004, 22, 153–158. [Google Scholar] [PubMed]
  38. Artlett, C.M.; Black, C.M.; Briggs, D.C.; Stephens, C.; Welsh, K.I. DNA allelic alterations within VNTR loci of scleroderma families. Br. J. Rheumatol. 1996, 35, 1216–1222. [Google Scholar] [CrossRef] [PubMed]
  39. Palomino, G.M.; Bassi, C.L.; Wastowski, I.J.; Xavier, D.J.; Lucisano-Valim, Y.M.; Crispim, J.C.; Rassi, D.M.; Marques-Neto, J.F.; Sakamoto-Hojo, E.T.; Moreau, P.; et al. Patients with systemic sclerosis present increased DNA damage differentially associated with DNA repair gene polymorphisms. J. Rheumatol. 2014, 41, 458–465. [Google Scholar] [CrossRef]
  40. Vijayraghavan, S.; Blouin, T.; McCollum, J.; Porcher, L.; Virard, F.; Zavadil, J.; Feghali-Bostwick, C.; Saini, N. Widespread mutagenesis and chromosomal instability shape somatic genomes in systemic sclerosis. Nat. Commun. 2024, 15, 8889. [Google Scholar] [CrossRef]
  41. Derk, C.T.; Rasheed, M.; Artlett, C.M.; Jimenez, S.A. A cohort study of cancer incidence in systemic sclerosis. J. Rheumatol. 2006, 33, 1113–1116. [Google Scholar]
  42. Derk, C.T.; Sakkas, L.I.; Rasheed, M.; Artlett, C.; Jimenez, S.A. Autoantibodies in patients with systemic sclerosis and cancer: A case-control study. J. Rheumatol. 2003, 30, 1994–1996. [Google Scholar] [PubMed]
  43. Barnes, J.; Mayes, M.D. Epidemiology of systemic sclerosis: Incidence, prevalence, survival, risk factors, malignancy, and environmental triggers. Curr. Opin. Rheumatol. 2012, 24, 165–170. [Google Scholar] [CrossRef] [PubMed]
  44. Mecoli, C.A.; Rosen, A.; Casciola-Rosen, L.; Shah, A.A. Advances at the Interface of Cancer and Systemic Sclerosis. J. Scleroderma Relat. Disord. 2021, 6, 50–57. [Google Scholar] [CrossRef]
  45. Jarzabek-Chorzelska, M.; Blaszczyk, M.; Kolacinska-Strasz, Z.; Chorzelski, T.; Jabłońska, S.; Maul, G.G. Antikinetochore and antitopoisomerase I antibodies in systemic scleroderma: Comparative study using immunoblotted recombinant antigens, immunofluorescence, and double immunodiffusion. Arch. Dermatol. Res. 1990, 282, 76–83. [Google Scholar] [CrossRef]
  46. Fanning, G.C.; Welsh, K.I.; Bunn, C.; Du Bois, R.; Black, C.M. HLA associations in three mutually exclusive autoantibody subgroups in UK systemic sclerosis patients. Br. J. Rheumatol. 1998, 37, 201–207. [Google Scholar] [CrossRef]
  47. Noble, P.W.; Bernatsky, S.; Clarke, A.E.; Isenberg, D.A.; Ramsey-Goldman, R.; Hansen, J.E. DNA-damaging autoantibodies and cancer: The lupus butterfly theory. Nat. Rev. Rheumatol. 2016, 12, 429–434. [Google Scholar] [CrossRef]
  48. Hansen, J.E.; Chan, G.; Liu, Y.; Hegan, D.C.; Dalal, S.; Dray, E.; Kwon, Y.; Xu, Y.; Xu, X.; Peterson-Roth, E.; et al. Targeting cancer with a lupus autoantibody. Sci. Transl. Med. 2012, 4, 157ra142. [Google Scholar] [CrossRef]
  49. Noble, P.W.; Chan, G.; Young, M.R.; Weisbart, R.H.; Hansen, J.E. Optimizing a lupus autoantibody for targeted cancer therapy. Cancer Res. 2015, 75, 2285–2291. [Google Scholar] [CrossRef]
  50. Rattray, Z.; Dubljevic, V.; Rattray, N.J.W.; Greenwood, D.L.; Johnson, C.H.; Campbell, J.A.; Hansen, J.E. Re-engineering and evaluation of anti-DNA autoantibody 3E10 for therapeutic applications. Biochem. Biophys. Res. Commun. 2018, 496, 858–864. [Google Scholar] [CrossRef]
  51. Hansen, J.E.; Tse, C.M.; Chan, G.; Heinze, E.R.; Nishimura, R.N.; Weisbart, R.H. Intranuclear protein transduction through a nucleoside salvage pathway. J. Biol. Chem. 2007, 282, 20790–20793. [Google Scholar] [CrossRef] [PubMed]
  52. Deng, S.X.; Hanson, E.; Sanz, I. In vivo cell penetration and intracellular transport of anti-Sm and anti-La autoantibodies. Int. Immunol. 2000, 12, 415–423. [Google Scholar] [CrossRef]
  53. Jabs, E.W.; Tuck-Muller, C.M.; Anhalt, G.J.; Earnshaw, W.; Wise, R.A.; Wigley, F. Cytogenetic survey in systemic sclerosis: Correlation of aneuploidy with the presence of anticentromere antibodies. Cytogenet. Cell Genet. 1993, 63, 169–175. [Google Scholar] [CrossRef]
  54. Vafa, O.; Shelby, R.D.; Sullivan, K.F. CENP-A associated complex satellite DNA in the kinetochore of the Indian muntjac. Chromosoma 1999, 108, 367–374. [Google Scholar] [CrossRef]
  55. Sugimoto, K.; Fukuda, R.; Himeno, M. Centromere/kinetochore localization of human centromere protein A (CENP-A) exogenously expressed as a fusion to green fluorescent protein. Cell Struct. Funct. 2000, 25, 253–261. [Google Scholar] [CrossRef]
  56. Patterson, K.A.; Walker, J.G.; Roberts-Thomson, P.J.; Bull, C.F.; Fenech, M. Evidence of chromosomal damage in scleroderma. Pathology 2022, 54, 131–133. [Google Scholar] [CrossRef] [PubMed]
  57. Stewart, L.; Redinbo, M.R.; Qiu, X.; Hol, W.G.; Champoux, J.J. A model for the mechanism of human topoisomerase I. Science 1998, 279, 1534–1541. [Google Scholar] [CrossRef]
  58. Douvas, A.S.; Achten, M.; Tan, E.M. Identification of a nuclear protein (Scl-70) as a unique target of human antinuclear antibodies in scleroderma. J. Biol. Chem. 1979, 254, 10514–10522. [Google Scholar] [CrossRef] [PubMed]
  59. Shero, J.H.; Bordwell, B.; Rothfield, N.F.; Earnshaw, W.C. High titres of autoantibodies to topoisomerase 1 (Scl-70) in sera from scleroderma patients. Science 1986, 231, 737–740. [Google Scholar] [CrossRef] [PubMed]
  60. May, C.K.; Noble, P.W.; Herzog, E.L.; Meffre, E.; Hansen, J.E. Nuclear-penetrating scleroderma autoantibody inhibits topoisomerase 1 cleavage complex formation. Biochem. Biophys. Res. Commun. 2024, 720, 150123. [Google Scholar] [CrossRef]
  61. Postow, L.; Crisona, N.J.; Peter, B.J.; Hardy, C.D.; Cozzarelli, N.R. Topological challenges to DNA replication: Conformations at the fork. Proc. Natl. Acad. Sci. USA 2001, 98, 8219–8226. [Google Scholar] [CrossRef]
  62. Cortés, F.; Piñero, J.; Ortiz, T. Importance of replication fork progression for the induction of chromosome damage and SCE by inhibitors of DNA topoisomerases. Mutat. Res. 1993, 303, 71–76. [Google Scholar] [CrossRef] [PubMed]
  63. Ulukan, H.; Muller, M.T.; Swaan, P.W. Downregulation of topoisomerase I in differentiating human intestinal epithelial cells. Int. J. Cancer 2001, 94, 200–207. [Google Scholar] [CrossRef]
  64. Avemann, K.; Knippers, R.; Koller, T.; Sogo, J.M. Camptothecin, a specific inhibitor of type I DNA topoisomerase, induces DNA breakage at replication forks. Mol. Cell Biol. 1988, 8, 3026–3034. [Google Scholar] [CrossRef]
  65. Pomerantz, R.T.; O’Donnell, M. What happens when replication and transcription complexes collide? Cell Cycle 2010, 9, 2537–2543. [Google Scholar] [CrossRef]
  66. Liu, S.; Hua, Y.; Wang, J.; Li, L.; Yuan, J.; Zhang, B.; Wang, Z.; Ji, J.; Kong, D. RNA polymerase III is required for the repair of DNA double-strand breaks by homologous recombination. Cell 2021, 184, 1314–1329.e1310. [Google Scholar] [CrossRef]
  67. Hamaguchi, Y.; Kodera, M.; Matsushita, T.; Hasegawa, M.; Inaba, Y.; Usuda, T.; Kuwana, M.; Takehara, K.; Fujimoto, M. Clinical and immunologic predictors of scleroderma renal crisis in Japanese systemic sclerosis patients with anti-RNA polymerase III autoantibodies. Arthritis Rheumatol. 2015, 67, 1045–1052. [Google Scholar] [CrossRef]
  68. Terras, S.; Hartenstein, H.; Höxtermann, S.; Gambichler, T.; Kreuter, A. RNA polymerase III autoantibodies may indicate renal and more severe skin involvement in systemic sclerosis. Int. J. Dermatol. 2016, 55, 882–885. [Google Scholar] [CrossRef]
  69. Lazzaroni, M.G.; Airò, P. Anti-RNA polymerase III antibodies in patients with suspected and definite systemic sclerosis: Why and how to screen. J. Scleroderma Relat. Disord. 2018, 3, 214–220. [Google Scholar] [CrossRef]
  70. Elhannani, A.; Martel, M.E.; Collet, A.; Chepy, A.; Sanges, S.; Hachulla, É.; Dubucquoi, S.; Launay, D.; Sobanski, V. Anti-RNA polymerase III antibodies in systemic sclerosis: Prevalence and clinical associations from a systematic review and meta-analysis. Rheumatology 2025, 84, 1099–1100. [Google Scholar] [CrossRef]
  71. Schramm, L.; Hernandez, N. Recruitment of RNA polymerase III to its target promoters. Genes Dev. 2002, 16, 2593–2620. [Google Scholar] [CrossRef] [PubMed]
  72. Muller, H.J. The remaking of chromosomes. Collect. Net. 1938, XIII, 181–198. [Google Scholar]
  73. Okuda, K.; Bardeguez, A.; Gardner, J.P.; Rodriguez, P.; Ganesh, V.; Kimura, M.; Skurnick, J.; Awad, G.; Aviv, A. Telomere length in the newborn. Pediatr. Res. 2002, 52, 377–381. [Google Scholar] [CrossRef]
  74. Henderson, E.R.; Blackburn, E.H. An overhanging 3’ terminus is a conserved feature of telomeres. Mol. Cell. Biol. 1989, 9, 345–348. [Google Scholar] [CrossRef] [PubMed]
  75. Weiner, A.M. Eukaryotic nuclear telomeres: Molecular fossils of the RNP world? Cell 1988, 52, 155–158. [Google Scholar] [CrossRef]
  76. Passos, J.F.; Saretzki, G.; von Zglinicki, T. DNA damage in telomeres and mitochondria during cellular senescence: Is there a connection? Nucleic Acids Res. 2007, 35, 7505–7513. [Google Scholar] [CrossRef]
  77. Allsopp, R.C.; Vaziri, H.; Patterson, C.; Goldstein, S.; Younglai, E.V.; Futcher, A.B.; Greider, C.W.; Harley, C.B. Telomere length predicts replicative capacity of human fibroblasts. Proc. Natl. Acad. Sci. USA 1992, 89, 10114–10118. [Google Scholar] [CrossRef]
  78. Bailey, S.M.; Murnane, J.P. Telomeres, chromosome instability and cancer. Nucleic Acids Res. 2006, 34, 2408–2417. [Google Scholar] [CrossRef]
  79. Muraki, K.; Nyhan, K.; Han, L.; Murnane, J.P. Mechanisms of telomere loss and their consequences for chromosome instability. Front. Oncol. 2012, 2, 135. [Google Scholar] [CrossRef]
  80. Gisselsson, D.; Pettersson, L.; Höglund, M.; Heidenblad, M.; Gorunova, L.; Wiegant, J.; Mertens, F.; Dal Cin, P.; Mitelman, F.; Mandahl, N. Chromosomal breakage-fusion-bridge events cause genetic intratumor heterogeneity. Proc. Natl. Acad. Sci. USA 2000, 97, 5357–5362. [Google Scholar] [CrossRef] [PubMed]
  81. Markiewicz-Potoczny, M.; Denchi, E.L. Telomere Protection in Stem Cells. Cold Spring Harb. Perspect. Biol. 2025, 17. [Google Scholar] [CrossRef] [PubMed]
  82. Artlett, C.M.; Black, C.M.; Briggs, D.C.; Stephens, C.O.; Welsh, K.I. Telomere reduction in scleroderma patients: A possible cause for chromosomal instability. Br. J. Rheumatol. 1996, 35, 732–737. [Google Scholar]
  83. Lakota, K.; Hanumanthu, V.S.; Agrawal, R.; Carns, M.; Armanios, M.; Varga, J. Short lymphocyte, but not granulocyte, telomere length in a subset of patients with systemic sclerosis. Ann. Rheum. Dis. 2019, 78, 1142–1144. [Google Scholar] [CrossRef]
  84. Wareing, N.; Mohan, V.; Taherian, R.; Volkmann, E.R.; Lyons, M.A.; Wilhalme, H.; Roth, M.D.; Estrada, Y.M.R.M.; Skaug, B.; Mayes, M.D.; et al. Blood Neutrophil Count and Neutrophil-to-Lymphocyte Ratio for Prediction of Disease Progression and Mortality in Two Independent Systemic Sclerosis Cohorts. Arthritis Care Res. 2023, 75, 648–656. [Google Scholar] [CrossRef]
  85. Liu, S.; Chung, M.P.; Ley, B.; French, S.; Elicker, B.M.; Fiorentino, D.F.; Chung, L.S.; Boin, F.; Wolters, P.J. Peripheral blood leucocyte telomere length is associated with progression of interstitial lung disease in systemic sclerosis. Thorax 2021, 76, 1186–1192. [Google Scholar] [CrossRef]
  86. Yang, M.M.; Lee, S.; Neely, J.; Hinchcliff, M.; Wolters, P.J.; Sirota, M. Gene expression meta-analysis reveals aging and cellular senescence signatures in scleroderma-associated interstitial lung disease. Front. Immunol. 2024, 15, 1326922. [Google Scholar] [CrossRef] [PubMed]
  87. Usategui, A.; Municio, C.; Arias-Salgado, E.G.; Martín, M.; Fernández-Varas, B.; Del Rey, M.J.; Carreira, P.; González, A.; Criado, G.; Perona, R.; et al. Evidence of telomere attrition and a potential role for DNA damage in systemic sclerosis. Immun. Ageing 2022, 19, 7. [Google Scholar] [CrossRef] [PubMed]
  88. MacIntyre, A.; Brouilette, S.W.; Lamb, K.; Radhakrishnan, K.; McGlynn, L.; Chee, M.M.; Parkinson, E.K.; Freeman, D.; Madhok, R.; Shiels, P.G. Association of increased telomere lengths in limited scleroderma, with a lack of age-related telomere erosion. Ann. Rheum. Dis. 2008, 67, 1780–1782. [Google Scholar] [CrossRef]
  89. Rodriguez-Martin, I.; Villanueva-Martin, G.; Guillen-Del-Castillo, A.; Ortego-Centeno, N.; Callejas, J.L.; Simeón-Aznar, C.P.; Martin, J.; Acosta-Herrera, M. Contribution of Telomere Length to Systemic Sclerosis Onset: A Mendelian Randomization Study. Int. J. Mol. Sci. 2023, 24, 15589. [Google Scholar] [CrossRef] [PubMed]
  90. Tarhan, F.; Vural, F.; Kosova, B.; Aksu, K.; Cogulu, O.; Keser, G.; Gündüz, C.; Tombuloglu, M.; Oder, G.; Karaca, E.; et al. Telomerase activity in connective tissue diseases: Elevated in rheumatoid arthritis, but markedly decreased in systemic sclerosis. Rheumatol. Int. 2008, 28, 579–583. [Google Scholar] [CrossRef]
  91. Ohtsuka, T.; Yamakage, A.; Yamazaki, S. The polymorphism of telomerase RNA component gene in patients with systemic sclerosis. Br. J. Dermatol. 2002, 147, 250–254. [Google Scholar] [CrossRef]
  92. Adler, B.L.; Boin, F.; Wolters, P.J.; Bingham, C.O.; Shah, A.A.; Greider, C.; Casciola-Rosen, L.; Rosen, A. Autoantibodies targeting telomere-associated proteins in systemic sclerosis. Ann. Rheum. Dis. 2021, 80, 912–919. [Google Scholar] [CrossRef]
  93. Vulsteke, J.B.; Smith, V.; Bonroy, C.; Derua, R.; Blockmans, D.; De Haes, P.; Vanderschueren, S.; Lenaerts, J.L.; Claeys, K.G.; Wuyts, W.A.; et al. Identification of new telomere- and telomerase-associated autoantigens in systemic sclerosis. J. Autoimmun. 2023, 135, 102988. [Google Scholar] [CrossRef] [PubMed]
  94. Kaji, K.; Fertig, N.; Medsger, T.A., Jr.; Satoh, T.; Hoshino, K.; Hamaguchi, Y.; Hasegawa, M.; Lucas, M.; Schnure, A.; Ogawa, F.; et al. Autoantibodies to RuvBL1 and RuvBL2: A novel systemic sclerosis-related antibody associated with diffuse cutaneous and skeletal muscle involvement. Arthritis Care Res. 2014, 66, 575–584. [Google Scholar] [CrossRef]
  95. Di Pietro, L.; Chiccoli, F.; Salvati, L.; Vivarelli, E.; Vultaggio, A.; Matucci, A.; Bentow, C.; Mahler, M.; Parronchi, P.; Palterer, B. Anti-RuvBL1/2 Autoantibodies Detection in a Patient with Overlap Systemic Sclerosis and Polymyositis. Antibodies 2023, 12, 13. [Google Scholar] [CrossRef]
  96. Emerit, I.; Cerutti, P. Clastogenic activity from Bloom syndrome fibroblast cultures. Proc. Natl. Acad. Sci. USA 1981, 78, 1868–1872. [Google Scholar] [CrossRef] [PubMed]
  97. Emerit, I.; Michelson, A.M. Mechanism of photosensitivity in systemic lupus erythematosus patients. Proc. Natl. Acad. Sci. USA 1981, 78, 2537–2540. [Google Scholar] [CrossRef] [PubMed]
  98. Bhusate, L.L.; Herbert, K.E.; Scott, D.L.; Perrett, D. Increased DNA strand breaks in mononuclear cells from patients with rheumatoid arthritis. Ann. Rheum. Dis. 1992, 51, 8–12. [Google Scholar] [CrossRef]
  99. Emerit, I.; Antunes, J.; Silva, J.M.; Freitas, J.; Pinheiro, T.; Filipe, P. Clastogenic plasma factors in psoriasis--comparison of phototherapy and anti-TNF-α treatments. Photochem. Photobiol. 2011, 87, 1427–1432. [Google Scholar] [CrossRef]
  100. Emerit, I.; Serejo, F.; Filipe, P.; Alaoui Youssefi, A.; Fernandes, A.; Costa, A.; Freitas, J.; Ramalho, F.; Baptista, A.; Carneiro de Moura, M. Clastogenic factors as biomarkers of oxidative stress in chronic hepatitis C. Digestion 2000, 62, 200–207. [Google Scholar] [CrossRef]
  101. Tolchin, S.F.; Winkelstein, A.; Rodnan, G.P.; Pan, S.F.; Nankin, H.R. Chromosomal abnormalities from cyclophosphamide therapy in rheumatoid arthritis and progressive systemic sclerosis (scleroderma). Arthritis Rheum. 1974, 17, 375–382. [Google Scholar] [CrossRef]
  102. Cloos, J.; Reid, C.B.; van der Sterre, M.L.; Tobi, H.; Leemans, C.R.; Snow, G.B.; Braakhuis, B.J. A comparison of bleomycin-induced damage in lymphocytes and primary oral fibroblasts and keratinocytes in 30 subjects. Mutagenesis 1999, 14, 87–93. [Google Scholar] [CrossRef]
  103. Cloos, J.; Temmink, O.; Ceelen, M.; Snel, M.H.; Leemans, C.R.; Braakhuis, B.J. Involvement of cell cycle control in bleomycin-induced mutagen sensitivity. Environ. Mol. Mutagen. 2002, 40, 79–84. [Google Scholar] [CrossRef] [PubMed]
  104. Urbańska, M.; Sofińska, K.; Czaja, M.; Szymoński, K.; Skirlińska-Nosek, K.; Seweryn, S.; Lupa, D.; Szymoński, M.; Lipiec, E. Molecular alterations in metaphase chromosomes induced by bleomycin. Spectrochim. Acta A Mol. Biomol. Spectrosc. 2024, 312, 124026. [Google Scholar] [CrossRef] [PubMed]
  105. Lal, A.S.; Begum, S.K.; Bharadwaj, S.S.; Lalitha, V.; Vijayalakshmi, J.; Paul, S.F.D.; Maddaly, R. Bleomycin-induced genotoxicity in vitro in human peripheral blood lymphocytes evidenced as complex chromosome- and chromatid-type aberrations. Toxicol. Vitr. 2019, 54, 367–374. [Google Scholar] [CrossRef] [PubMed]
  106. Bolzán, A.D.; Bianchi, M.S. DNA and chromosome damage induced by bleomycin in mammalian cells: An update. Mutat. Res. Rev. Mutat. Res. 2018, 775, 51–62. [Google Scholar] [CrossRef] [PubMed]
  107. Bretza, J.; Goldman, J.A. Scleroderma simulating vinyl chloride disease. J. Occup. Med. 1979, 21, 436–438. [Google Scholar]
  108. Black, C.M.; Welsh, K.I.; Walker, A.E.; Bernstein, R.M.; Catoggio, L.J.; McGregor, A.R.; Lloyd Jones, J.K. Genetic susceptibility to scleroderma-like syndrome induced by vinyl chloride. Lancet 1983, 321, 53–55. [Google Scholar] [CrossRef]
  109. Maricq, H.R.; Johnson, M.N.; Whetstone, C.L.; LeRoy, E.C. Capillary abnormalities in polyvinyl chloride production workers. Examination by in vivo microscopy. JAMA 1976, 236, 1368–1371. [Google Scholar] [CrossRef]
  110. Hahn, E.; Aderka, D.; Suprun, H.; Shtamler, B. Occupational acroosteolysis in vinyl chloride workers in Israel. Isr. J. Med. Sci. 1979, 15, 218–222. [Google Scholar]
  111. Nicholson, W.J.; Henneberger, P.K.; Seidman, H. Occupational hazards in the VC-PVC industry. Prog. Clin. Biol. Res. 1984, 141, 155–175. [Google Scholar]
  112. Hansteen, I.L.; Hillestad, L.; Thiis-Evensen, E.; Heldaas, S.S. Effects of vinyl chloride in man: A cytogenetic follow-up study. Mutat. Res. 1978, 51, 271–278. [Google Scholar] [CrossRef] [PubMed]
  113. Flindt-Hansen, H.; Isager, H. Scleroderma after occupational exposure to trichlorethylene and trichlorethane. Acta Derm. Venereol. 1987, 67, 263–264. [Google Scholar] [CrossRef]
  114. Lockey, J.E.; Kelly, C.R.; Cannon, G.W.; Colby, T.V.; Aldrich, V.; Livingston, G.K. Progressive systemic sclerosis associated with exposure to trichloroethylene. J. Occup. Med. 1987, 29, 493–496. [Google Scholar]
  115. Yanez Diaz, S.; Moran, M.; Unamuno, P.; Armijo, M. Silica and trichloroethylene induced progressive systemic sclerosis. Dermatology 1992, 182, 98–102. [Google Scholar] [CrossRef]
  116. Czirjak, L.; Schlammadinger, J.; Szegedi, G. Systemic sclerosis and exposure to trichloroethylene. Dermatology 1993, 186, 236. [Google Scholar] [CrossRef]
  117. Giver, C.R.; Wong, R.; Moore, D.H., 2nd; Pallavicini, M.G. Dermal benzene and trichloroethylene induce aneuploidy in immature hematopoietic subpopulations in vivo. Environ. Mol. Mutagen. 2001, 37, 185–194. [Google Scholar] [CrossRef]
  118. Rubio-Rivas, M.; Moreno, R.; Corbella, X. Occupational and environmental scleroderma. Systematic review and meta-analysis. Clin. Rheumatol. 2017, 36, 569–582. [Google Scholar] [CrossRef]
  119. Diot, E.; Lesire, V.; Guilmot, J.L.; Metzger, M.D.; Pilore, R.; Rogier, S.; Stadler, M.; Diot, P.; Lemarie, E.; Lasfargues, G. Systemic sclerosis and occupational risk factors: A case-control study. Occup. Environ. Med. 2002, 59, 545–549. [Google Scholar] [CrossRef] [PubMed]
  120. Zhou, Y.; Wang, K.; Wang, B.; Pu, Y.; Zhang, J. Occupational benzene exposure and the risk of genetic damage: A systematic review and meta-analysis. BMC Public Health 2020, 20, 1113. [Google Scholar] [CrossRef] [PubMed]
  121. Silvestre, R.T.; Bravo, M.; Santiago, F.; Delmonico, L.; Scherrer, L.; Otero, U.B.; Liehr, T.; Alves, G.; Chantre-Justino, M.; Ornellas, M.H. Hypermethylation in Gene Promoters Are Induced by Chronic Exposure to Benzene, Toluene, Ethylbenzene and Xylenes. Pak. J. Biol. Sci. 2020, 23, 518–525. [Google Scholar] [CrossRef]
  122. Villalba-Campos, M.; Chuaire-Noack, L.; Sánchez-Corredor, M.C.; Rondón-Lagos, M. High chromosomal instability in workers occupationally exposed to solvents and paint removers. Mol. Cytogenet. 2016, 9, 46. [Google Scholar] [CrossRef]
  123. Brasington, R.D., Jr.; Thorpe-Swenson, A.J. Systemic sclerosis associated with cutaneous exposure to solvent: Case report and review of the literature. Arthritis Rheum. 1991, 34, 631–633. [Google Scholar] [CrossRef] [PubMed]
  124. Alonso-Ruiz, A.; Zea-Mendoza, A.C.; Salazar-Vallinas, J.M.; Rocamora-Ripoll, A.; Beltrán-Gutiérrez, J. Toxic oil syndrome: A syndrome with features overlapping those of various forms of scleroderma. Semin. Arthritis Rheum. 1986, 15, 200–212. [Google Scholar] [CrossRef]
  125. Phillips, D.H.; Hewer, A.; Arlt, V.M. 32P-postlabeling analysis of DNA adducts. Methods Mol. Biol. 2005, 291, 3–12. [Google Scholar] [CrossRef]
  126. Kanaya, N. Activation of aniline by extracts from plants and induction of chromosomal damages in Chinese hamster ovary cells. Genes Genet. Syst. 1996, 71, 319–322. [Google Scholar] [CrossRef]
  127. Stefanidou, M.E.; Hatzi, V.I.; Terzoudi, G.I.; Loutsidou, A.C.; Maravelias, C.P. Effect of cocaine and crack on the ploidy status of Tetrahymena pyriformis: A study using DNA image analysis. Cytotechnology 2011, 63, 35–40. [Google Scholar] [CrossRef] [PubMed]
  128. Attoussi, S.; Faulkner, M.L.; Oso, A.; Umoru, B. Cocaine-induced scleroderma and scleroderma renal crisis. South. Med. J. 1998, 91, 961–963. [Google Scholar] [CrossRef]
  129. Andreussi, R.; Silva, L.M.B.; da Silva, H.C.; Luppino-Assad, A.P.; Andrade, D.C.O.; Sampaio-Barros, P.D. Systemic sclerosis induced by the use of cocaine: Is there an association? Rheumatol. Int. 2019, 39, 387–393. [Google Scholar] [CrossRef] [PubMed]
  130. Landers, D.; Hehir, D.; Murphy, G. Three distinct presentations of systemic sclerosis in patients with previous silica dust exposure. BMJ Case Rep. 2025, 18, e264237. [Google Scholar] [CrossRef]
  131. Freire, M.; Sopeña, B.; Bravo, S.; Spuch, C.; Argibay, A.; Estévez, M.; Pena, C.; Naya, M.; Lama, A.; González-Quintela, A. Serum Proteomic Markers in Patients with Systemic Sclerosis in Relation to Silica Exposure. J. Clin. Med. 2025, 14, 2019. [Google Scholar] [CrossRef]
  132. Wultsch, G.; Setayesh, T.; Kundi, M.; Kment, M.; Nersesyan, A.; Fenech, M.; Knasmüller, S. Induction of DNA damage as a consequence of occupational exposure to crystalline silica: A review and meta-analysis. Mutat. Res. Rev. Mutat. Res. 2021, 787, 108349. [Google Scholar] [CrossRef] [PubMed]
  133. Yan, K.; Ma, Y.; Shi, X.; Liang, C.; Ding, R.; Sun, Z.; Duan, J. Long-term SiNPs exposure induced genetic instability and malignant transformation via SQSTM1/p62-mediated autophagy dysfunction in lungs. Mater. Today Bio 2025, 33, 101972. [Google Scholar] [CrossRef]
  134. Freire, M.; Sopeña, B.; González-Quintela, A.; Guillén Del Castillo, A.; Moraga, E.C.; Lledó-Ibañez, G.M.; Rubio-Rivas, M.; Trapiella, L.; Argibay, A.; Tolosa, C.; et al. Exposure to different occupational chemicals and clinical phenotype of a cohort of patients with systemic sclerosis. Autoimmun. Rev. 2024, 23, 103542. [Google Scholar] [CrossRef]
  135. Abbot, S.; Bossingham, D.; Proudman, S.; de Costa, C.; Ho-Huynh, A. Risk factors for the development of systemic sclerosis: A systematic review of the literature. Rheumatol. Adv. Pract. 2018, 2, rky041. [Google Scholar] [CrossRef]
  136. Varga, J.; Peltonen, J.; Uitto, J.; Jimenez, S. Development of diffuse fasciitis with eosinophilia during L-tryptophan treatment: Demonstration of elevated type I collagen gene expression in affected tissues: A Clinicopathologic Study of Four Patients. Ann. Intern. Med. 1990, 112, 344–351. [Google Scholar] [CrossRef] [PubMed]
  137. Silver, R.M.; Heyes, M.P.; Maize, J.C.; Quearry, B.; Vionnet-Fuasset, M.; Sternberg, E.M. Scleroderma, fasciitis, and eosinophilia associated with the ingestion of tryptophan. N. Engl. J. Med. 1990, 322, 874–881. [Google Scholar] [CrossRef]
  138. Artlett, C.M.; Sassi-Gaha, S.; Rieger, J.L.; Boesteanu, A.C.; Feghali-Bostwick, C.A.; Katsikis, P.D. The inflammasome activating caspase-1 mediates fibrosis and myofibroblast differentiation in systemic sclerosis. Arthritis Rheum. 2011, 63, 3563–3574. [Google Scholar] [CrossRef] [PubMed]
  139. Kerr, L.D.; Spiera, H. Scleroderma in association with the use of bleomycin: A report of 3 cases. J. Rheumatol. 1992, 19, 294–296. [Google Scholar]
  140. Kim, K.H.; Yoon, T.J.; Oh, C.W.; Ko, G.H.; Kim, T.H. A case of bleomycin-induced scleroderma. J. Korean Med. Sci. 1996, 11, 454–456. [Google Scholar] [CrossRef]
  141. Passiu, G.; Cauli, A.; Atzeni, F.; Aledda, M.; Dessole, G.; Sanna, G.; Nurchis, P.; Vacca, A.; Garau, P.; Laudadio, M.; et al. Bleomycin-induced scleroderma: Report of a case with a chronic course rather than the typical acute/subacute self-limiting form. Clin. Rheumatol. 1999, 18, 422–424. [Google Scholar] [CrossRef]
  142. Limoli, C.L.; Kaplan, M.I.; Phillips, J.W.; Adair, G.M.; Morgan, W.F. Differential induction of chromosomal instability by DNA strand-breaking agents. Cancer Res. 1997, 57, 4048–4056. [Google Scholar] [PubMed]
  143. Oberley, L.W.; Buettner, G.R. Role of superoxide dismutase in cancer: A review. Cancer Res. 1979, 39, 1141–1149. [Google Scholar]
  144. Sausville, E.A.; Stein, R.W.; Peisach, J.; Horwitz, S.B. Properties and products of the degradation of DNA by bleomycin and iron(II). Biochemistry 1978, 17, 2746–2754. [Google Scholar] [CrossRef] [PubMed]
  145. Rodriguez, L.O.; Hecht, S.M. Iron(II)-bleomycin. Biochemical and spectral properties in the presence of radical scavengers. Biochem. Biophys. Res. Commun. 1982, 104, 1470–1476. [Google Scholar] [CrossRef]
  146. Bartsch, H.; Malaveille, C.; Barbin, A.; Bresil, H.; Tomatis, L.; Montesano, R. Mutagenicity and metabolism of vinyl chloride and related compounds. Environ. Health Perspect. 1976, 17, 193–198. [Google Scholar] [CrossRef]
  147. Czirják, L.; Szegedi, G. Benzene exposure and systemic sclerosis. Ann. Intern. Med. 1987, 107, 118. [Google Scholar] [CrossRef] [PubMed]
  148. Borghini, A.; Poscia, A.; Bosello, S.; Teleman, A.A.; Bocci, M.; Iodice, L.; Ferraccioli, G.; La Milìa, D.I.; Moscato, U. Environmental Pollution by Benzene and PM(10) and Clinical Manifestations of Systemic Sclerosis: A Correlation Study. Int. J. Environ. Res. Public Health 2017, 14, 1297. [Google Scholar] [CrossRef]
  149. Rithidech, K.; Dunn, J.J.; Bond, V.P.; Gordon, C.R.; Cronkite, E.P. Characterization of genetic instability in radiation- and benzene-induced murine acute leukemia. Mutat. Res. 1999, 428, 33–39. [Google Scholar] [CrossRef]
  150. Rush, P.J.; Bell, M.J.; Fam, A.G. Toxic oil syndrome (Spanish oil disease) and chemically induced scleroderma-like conditions. J. Rheumatol. 1984, 11, 262–264. [Google Scholar]
  151. Dupont, A.; Majithia, V.; Ahmad, S.; McMurray, R. Nephrogenic fibrosing dermopathy, a new mimicker of systemic sclerosis. Am. J. Med. Sci. 2005, 330, 192–194. [Google Scholar] [CrossRef] [PubMed]
  152. Jimenez, S.A.; Artlett, C.M.; Sandorfi, N.; Derk, C.; Latinis, K.; Sawaya, H.; Haddad, R.; Shanahan, J.C. Dialysis-associated systemic fibrosis (nephrogenic fibrosing dermopathy): Study of inflammatory cells and transforming growth factor beta1 expression in affected skin. Arthritis Rheum. 2004, 50, 2660–2666. [Google Scholar] [CrossRef] [PubMed]
  153. Phelps, R.G.; Fleischmajer, R. Clinical, pathologic, and immunopathologic manifestations of the toxic oil syndrome. Analysis of fourteen cases. J. Am. Acad. Dermatol. 1988, 18, 313–324. [Google Scholar] [CrossRef]
  154. Posada de la Paz, M.; Philen, R.M.; Borda, A.I. Toxic oil syndrome: The perspective after 20 years. Epidemiol. Rev. 2001, 23, 231–247. [Google Scholar] [CrossRef]
  155. Russell, E.; McMahon, S.J.; Russell, B.; Mohamud, H.; McGarry, C.K.; Schettino, G.; Prise, K.M. Effects of Gadolinium MRI Contrast Agents on DNA Damage and Cell Survival when Used in Combination with Radiation. Radiat. Res. 2020, 194, 298–309. [Google Scholar] [CrossRef] [PubMed]
  156. Pinto, T.G.; Dedivits, R.A.; Ribeiro, D.A. Do Gadolinium-Based Contrasts Represent a High Risk for Genotoxicity in Mammalian Cells? A Systematic Review. J. Appl. Toxicol. 2025, 45, 1935–1946. [Google Scholar] [CrossRef]
  157. Aryal, B.K.; Khuder, S.A.; Schaub, E.A. Meta-analysis of systemic sclerosis and exposure to solvents. Am. J. Ind. Med. 2001, 40, 271–274. [Google Scholar] [CrossRef]
  158. Kettaneh, A.; Al Moufti, O.; Tiev, K.P.; Chayet, C.; Tolédano, C.; Fabre, B.; Fardet, L.; Cabane, J. Occupational exposure to solvents and gender-related risk of systemic sclerosis: A metaanalysis of case-control studies. J. Rheumatol. 2007, 34, 97–103. [Google Scholar] [PubMed]
  159. Barragán-Martínez, C.; Speck-Hernández, C.A.; Montoya-Ortiz, G.; Mantilla, R.D.; Anaya, J.M.; Rojas-Villarraga, A. Organic solvents as risk factor for autoimmune diseases: A systematic review and meta-analysis. PLoS ONE 2012, 7, e51506. [Google Scholar] [CrossRef]
  160. Kosarek, N.N.; Romano, M.E.; Moen, E.L.; Simms, R.W.; Erickson, A.; Khanna, D.; Pioli, P.A.; Whitfield, M.L. Geographic Clustering of Systemic Sclerosis in Areas of Environmental Pollution. Arthritis Care Res. 2025, 77, 855–866. [Google Scholar] [CrossRef]
  161. Cayuela, L.; Pereyra-Rodríguez, J.J.; Ramos, P.C.; Grande, N.G.; Cayuela, A. Unveiling spatial clusters of systemic sclerosis mortality in Spain: A comprehensive geographical analysis. Med. Clin. 2025, 164, 403–409. [Google Scholar] [CrossRef]
  162. Kassamali, B.; Kassamali, A.A.; Muntyanu, A.; Netchiporouk, E.; Vleugels, R.A.; LaChance, A. Geographic distribution and environmental triggers of systemic sclerosis cases from 2 large academic tertiary centers in Massachusetts. J. Am. Acad. Dermatol. 2022, 86, 925–927. [Google Scholar] [CrossRef] [PubMed]
  163. Radić, M.; Martinović Kaliterna, D.; Fabijanić, D.; Radić, J. Prevalence of systemic sclerosis in Split-Dalmatia county in Southern Croatia. Clin. Rheumatol. 2010, 29, 419–421. [Google Scholar] [CrossRef]
  164. Silman, A.J.; Howard, Y.; Hicklin, A.J.; Black, C.M. Geographical clustering of scleroderma in south and west London. Br. J. Rheumatol. 1990, 29, 92–96. [Google Scholar] [CrossRef]
  165. Walker, U.A.; Tyndall, A.; Czirják, L.; Denton, C.P.; Farge-Bancel, D.; Kowal-Bielecka, O.; Müller-Ladner, U.; Matucci-Cerinic, M. Geographical variation of disease manifestations in systemic sclerosis: A report from the EULAR Scleroderma Trials and Research (EUSTAR) group database. Ann. Rheum. Dis. 2009, 68, 856–862. [Google Scholar] [CrossRef]
  166. Roberts-Thomson, P.J.; Jones, M.; Hakendorf, P.; Kencana Dharmapatni, A.A.S.S.; MacFarlane, J.G.; Smith, M.D.; Ahern, M.J. Scleroderma in South Australia: Epidemiological observations of possible pathogenic significance. Int. Med. J. 2001, 31, 220–221. [Google Scholar] [CrossRef]
  167. Sadatpour, O.; Azizan, A.; Kavosi, H.; Vodjgani, M.; Farhadi, E.; Mahmoudi, M. Systemic sclerosis, main culprits and involved signaling pathways. Inflamm. Res. Off. J. Eur. Histamine Res. Soc. 2025, 74, 158. [Google Scholar] [CrossRef] [PubMed]
  168. Nguyen, T.H.; Nguyen, H.H.; Nguyen, T.D.; Tran, V.T.; Nguyen, H.A.; Pham, D.V. NLRP3 inflammasome activation contributes to the development of the pro-fibrotic phenotype of lung fibroblasts. Biochem. Pharmacol. 2024, 229, 116496. [Google Scholar] [CrossRef] [PubMed]
  169. Feng, J.; Liu, H.; Jiang, K.; Gong, X.; Huang, R.; Zhou, C.; Mao, J.; Chen, Y.; Xu, H.; Zhang, X.; et al. Enhanced oxidative stress aggravates BLM-induced pulmonary fibrosis by promoting cellular senescence through enhancing NLRP3 activation. Life Sci. 2024, 358, 123128. [Google Scholar] [CrossRef]
  170. Rivadeneira, D.B.; Thosar, S.; Quann, K.; Gunn, W.G.; Dean, V.G.; Xie, B.; Parise, A.; McGovern, A.C.; Spahr, K.; Lontos, K.; et al. Oxidative-stress-induced telomere instability drives T cell dysfunction in cancer. Immunity 2025, 58, 2524–2540.e2525. [Google Scholar] [CrossRef]
  171. Oikawa, S.; Tada-Oikawa, S.; Kawanishi, S. Site-specific DNA damage at the GGG sequence by UVA involves acceleration of telomere shortening. Biochemistry 2001, 40, 4763–4768. [Google Scholar] [CrossRef]
  172. Sekoguchi, S.; Nakajima, T.; Moriguchi, M.; Jo, M.; Nishikawa, T.; Katagishi, T.; Kimura, H.; Minami, M.; Itoh, Y.; Kagawa, K.; et al. Role of cell-cycle turnover and oxidative stress in telomere shortening and cellular senescence in patients with chronic hepatitis C. J. Gastroenterol. Hepatol. 2007, 22, 182–190. [Google Scholar] [CrossRef] [PubMed]
  173. Kuchino, Y.; Mori, F.; Kasai, H.; Inoue, H.; Iwai, S.; Miura, K.; Ohtsuka, E.; Nishimura, S. Misreading of DNA templates containing 8-hydroxydeoxyguanosine at the modified base and at adjacent residues. Nature 1987, 327, 77–79. [Google Scholar] [CrossRef]
  174. Barnes, R.P.; de Rosa, M.; Thosar, S.A.; Detwiler, A.C.; Roginskaya, V.; Van Houten, B.; Bruchez, M.P.; Stewart-Ornstein, J.; Opresko, P.L. Telomeric 8-oxo-guanine drives rapid premature senescence in the absence of telomere shortening. Nat. Struct. Mol. Biol. 2022, 29, 639–652. [Google Scholar] [CrossRef] [PubMed]
  175. Fouquerel, E.; Barnes, R.P.; Uttam, S.; Watkins, S.C.; Bruchez, M.P.; Opresko, P.L. Targeted and Persistent 8-Oxoguanine Base Damage at Telomeres Promotes Telomere Loss and Crisis. Mol. Cell 2019, 75, 117–130.e116. [Google Scholar] [CrossRef]
  176. Pejenaute, Á.; Cortés, A.; Marqués, J.; Montero, L.; Beloqui, Ó.; Fortuño, A.; Martí, A.; Orbe, J.; Zalba, G. NADPH Oxidase Overactivity Underlies Telomere Shortening in Human Atherosclerosis. Int. J. Mol. Sci. 2020, 21, 1434. [Google Scholar] [CrossRef]
  177. Bhargava, R.; Fischer, M.; O’Sullivan, R.J. Genome rearrangements associated with aberrant telomere maintenance. Curr. Opin. Genet. Dev. 2020, 60, 31–40. [Google Scholar] [CrossRef]
  178. Cleal, K.; Baird, D.M. Catastrophic Endgames: Emerging Mechanisms of Telomere-Driven Genomic Instability. Trends Genet. 2020, 36, 347–359. [Google Scholar] [CrossRef]
  179. Goto, M. Werner’s syndrome: From clinics to genetics. Clin. Exp. Rheumatol. 2000, 18, 760–766. [Google Scholar]
  180. Foti, R.; Leonardi, R.; Rondinone, R.; Di Gangi, M.; Leonetti, C.; Canova, M.; Doria, A. Scleroderma-like disorders. Autoimmun. Rev. 2008, 7, 331–339. [Google Scholar] [CrossRef]
  181. Dunn, H.G.; Meuwissen, H.; Livingstone, C.S.; Pump, K.K. Ataxia-Telangiectasia. Can. Med. Assoc. J. 1964, 91, 1106–1118. [Google Scholar]
  182. Duecker, R.; Baer, P.; Eickmeier, O.; Strecker, M.; Kurz, J.; Schaible, A.; Henrich, D.; Zielen, S.; Schubert, R. Oxidative stress-driven pulmonary inflammation and fibrosis in a mouse model of human ataxia-telangiectasia. Redox Biol. 2018, 14, 645–655. [Google Scholar] [CrossRef]
  183. Barreto, T.L.N.; de Carvalho Filho, R.J.; Shigueoka, D.C.; Fonseca, F.L.A.; Ferreira, A.C.; Kochi, C.; Aranda, C.S.; Sarni, R.O.S. Hepatic fibrosis: A manifestation of the liver disease evolution in patients with Ataxia-telangiectasia. Orphanet J. Rare Dis. 2023, 18, 105. [Google Scholar] [CrossRef]
  184. Alonso-González, A.; Véliz-Flores, I.; Tosco-Herrera, E.; González-Barbuzano, S.; Mendoza-Alvarez, A.; Galván-Fernández, H.; Sastre, L.; Fernández-Varas, B.; Corrales, A.; Rubio-Rodríguez, L.A.; et al. A tiered strategy to identify relevant genetic variants in familial pulmonary fibrosis: A proof of concept for the clinical practice. Eur. J. Hum. Genet. 2025, 33, 1509–1519. [Google Scholar] [CrossRef]
  185. Alder, J.K.; Armanios, M. Telomere-mediated lung disease. Physiol. Rev. 2022, 102, 1703–1720. [Google Scholar] [CrossRef]
  186. Dahlqvist, C.; Planté-Bordeneuve, T.; Muca, T.; de Leener, A.; Ghaye, B.; Coche, E.; Decottignies, A.; van Dievoet, M.A.; Froidure, A. Use of Telomere Length as a Biomarker in Idiopathic Pulmonary Fibrosis. Lung 2025, 203, 78. [Google Scholar] [CrossRef]
  187. Cortesão, C.; Balanco, L.; Ferreira, P.G. Familial pulmonary fibrosis with dyskeratosis congenita associated with a rare RTEL1 gene mutation. BMJ Case Rep. 2025, 18, e265092. [Google Scholar] [CrossRef]
  188. Roka, K.; Solomou, E.; Kattamis, A.; Stiakaki, E. Telomere biology disorders: From dyskeratosis congenita and beyond. Postgrad. Med. J. 2024, 100, 879–889. [Google Scholar] [CrossRef]
  189. Batista, L.F.Z.; Dokal, I.; Parker, R. Telomere biology disorders: Time for moving towards the clinic? Trends Mol. Med. 2022, 28, 882–891. [Google Scholar] [CrossRef] [PubMed]
  190. Thakur, W.; Anwar, N.; Samad, S.; Fatima, N.; Ahmed, R.; Tariq, F.; Ashfaq, J.; Sharif, S.; Borhany, M. Assessment of Hepatic Profile in Acquired Aplastic Anemia: An Experience from Pakistan. Cureus 2022, 14, e29079. [Google Scholar] [CrossRef] [PubMed]
  191. Snyder, A.J.; Campbell, K.M.; Lane, A.; Mehta, P.A.; Myers, K.; Davies, S.M.; Koo, J. Liver abnormalities are frequent and persistent in patients with Fanconi anemia. Blood Adv. 2024, 8, 1427–1438. [Google Scholar] [CrossRef] [PubMed]
  192. Schroader, J.H.; Handley, M.T.; Reddy, K. Inosine triphosphate pyrophosphatase: A guardian of the cellular nucleotide pool and potential mediator of RNA function. Wiley Interdiscip. Rev. RNA 2023, 14, e1790. [Google Scholar] [CrossRef]
  193. Ji, D.; Stepchenkova, E.I.; Cui, J.; Menezes, M.R.; Pavlov, Y.I.; Kool, E.T. Measuring deaminated nucleotide surveillance enzyme ITPA activity with an ATP-releasing nucleotide chimera. Nucleic Acids Res. 2017, 45, 11515–11524. [Google Scholar] [CrossRef]
  194. Wu, S.; Jiang, L.; Lei, L.; Fu, C.; Huang, J.; Hu, Y.; Dong, Y.; Chen, J.; Zeng, Q. Crosstalk between G-quadruplex and ROS. Cell Death Dis. 2023, 14, 37. [Google Scholar] [CrossRef]
  195. Rech, L.; Abdellatif, M.; Pöttler, M.; Stangl, V.; Mabotuwana, N.; Hardy, S.; Rainer, P.P. Small molecule STING inhibition improves myocardial infarction remodeling. Life Sci. 2022, 291, 120263. [Google Scholar] [CrossRef]
  196. Shen, R.; Yang, K.; Cheng, X.; Guo, C.; Xing, X.; Sun, H.; Liu, D.; Liu, X.; Wang, D. Accumulation of polystyrene microplastics induces liver fibrosis by activating cGAS/STING pathway. Environ. Pollut. 2022, 300, 118986. [Google Scholar] [CrossRef]
  197. Zhang, D.; Liu, Y.; Zhu, Y.; Zhang, Q.; Guan, H.; Liu, S.; Chen, S.; Mei, C.; Chen, C.; Liao, Z.; et al. A non-canonical cGAS-STING-PERK pathway facilitates the translational program critical for senescence and organ fibrosis. Nat. Cell Biol. 2022, 24, 766–782. [Google Scholar] [CrossRef]
  198. Wang, L.; Zhang, Y.; Ren, Y.; Yang, X.; Ben, H.; Zhao, F.; Yang, S.; Wang, L.; Qing, J. Pharmacological targeting of cGAS/STING-YAP axis suppresses pathological angiogenesis and ameliorates organ fibrosis. Eur. J. Pharmacol. 2022, 932, 175241. [Google Scholar] [CrossRef] [PubMed]
  199. Jiang, A.; Liu, J.; Wang, Y.; Zhang, C. cGAS-STING signaling pathway promotes hypoxia-induced renal fibrosis by regulating PFKFB3-mediated glycolysis. Free Radic. Biol. Med. 2023, 208, 516–529. [Google Scholar] [CrossRef]
  200. Gairola, S.; Kaundal, R.K. Amlexanox alleviates renal inflammation and fibrosis by inhibiting cGAS/STING/TBK1 and TGF-β1/Smad signaling. Eur. J. Pharmacol. 2025, 1007, 178266. [Google Scholar] [CrossRef] [PubMed]
  201. Luo, W.; Xu, G.; Song, Z.; Mu, W.; Wen, J.; Hui, S.; Zhao, J.; Zhan, X.; Bai, Z.; Xiao, X. Licorice extract inhibits the cGAS-STING pathway and protects against non-alcoholic steatohepatitis. Front. Pharmacol. 2023, 14, 1160445. [Google Scholar] [CrossRef] [PubMed]
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.

Article Metrics

Citations

Article Access Statistics

Multiple requests from the same IP address are counted as one view.