Next Article in Journal
Have New Plate Designs Reduced the Rate of Hardware Removal Following Midshaft Clavicle Fracture Fixation?
Previous Article in Journal
The Role of Minimally Invasive Surgery in Mitral Valve Repair: Through the Female Gaze
Previous Article in Special Issue
The Role of Neutrophil to Lymphocytes Ratio (NLR) as a Predictor of Disease Activity in Behcet’s Syndrome—A Comprehensive Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Uncovering the New Biology of Giant Cell Arteritis to Guide Therapeutic Strategies

1
Department of Rheumatology, Hyogo Prefectural Amagasaki General Medical Center, Amagasaki 660-8550, Japan
2
Department of Clinical Immunology, Graduate School of Medicine, Osaka Metropolitan University, 1-4-3, Asahi-Machi, Abeno-Ku, Osaka 545-8585, Japan
3
Department of Legal Medicine, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2025, 14(18), 6350; https://doi.org/10.3390/jcm14186350
Submission received: 29 July 2025 / Revised: 30 August 2025 / Accepted: 6 September 2025 / Published: 9 September 2025

Abstract

Giant cell arteritis (GCA) is a form of large vessel vasculitis that primarily affects older adults and forms granulomatous inflammation in the aorta and its major branches. Recent advances in immunology and multi-omics technologies have elucidated several key mechanisms involved in the pathogenesis of GCA, including immune checkpoint dysregulation, clonal hematopoiesis, and age-associated immune dysfunction. From the perspective of immune cell subsets, a diverse range of immune cells—including tissue-resident memory T cells, stem-like T cells, macrophage subsets, B cells, and myofibroblasts—play distinct roles in sustaining vascular inflammation and tissue remodeling. This review summarizes the latest immunopathological and omics-based insights into GCA, proposes potential therapeutic targets, and discusses future directions for precision medicine aimed at achieving sustained remission.

1. Introduction

Giant cell arteritis (GCA) predominantly affects individuals over 50 years of age and is the most common form of vasculitis in the elderly. It primarily involves the aorta and its major branches [1]. Epidemiologically, GCA has the highest prevalence in North America and Northern Europe and is relatively rare among East Asian populations [2].
Clinical manifestations vary depending on the arteries involved. For example, disease affecting the external and internal carotid arteries may present with visual loss, headache, or jaw claudication, whereas vertebral artery involvement can result in stroke. When the subclavian or axillary arteries are affected, patients may develop arm claudication and diminished pulses. While stenosis and occlusion are characteristic of medium-sized arteries, thoracic aortic disease can lead to severe complications, including aneurysm formation, dissection, and even rupture [1]. Early-onset GCA is often characterized by pronounced systemic symptoms and frequent relapses, whereas late-onset GCA is more commonly associated with intracranial involvement and comorbidities [3,4].
In recent years, advances in immunological omics-based analytical techniques have significantly enhanced our understanding of the pathophysiology of GCA. Emerging insights have revealed novel perspectives, including the interplay between aging and inflammation, epigenetic modifications, and the intricate networks of various immune cell subsets and cytokines. In this review, we summarize the latest findings on the emerging pathogenic mechanisms underlying GCA and discuss potential future therapeutic strategies informed by these insights.

2. Histopathology

The histopathological examination of temporal artery biopsy (TAB) remains the gold standard for the definitive diagnosis of GCA. The characteristic finding is concentric thickening of the vascular wall with marked intimal hyperplasia, which contributes to vascular occlusion and subsequent ischemic complications. Granulomatous inflammation is typically observed at the media–intima junction, characterized by prominent infiltration of CD4+ T cells and macrophages, often accompanied by multinucleated giant cells. Another key diagnostic feature is the destruction or fragmentation of the internal elastic lamina, which can be clearly demonstrated using elastin stains such as EVG. The inflammatory infiltrate consists predominantly of CD4+ T cells and macrophages, with only a minor contribution from CD8+ T cells. Additional features include neovascularization and adventitial inflammation, both reflecting ongoing vascular injury and reparative processes. Ultimately, persistent vascular inflammation results in intimal hyperplasia with vascular occlusion, or in arterial wall destruction leading to aneurysm formation (Figure 1) [1].

3. Pathophysiology

The pathophysiology of GCA involves a complex interplay of genetic predisposition, age-related immune dysfunction, environmental triggers, and aberrant immune signaling. Vascular dendritic cells (vas-DCs) residing at the media–adventitia border are activated upon stimulation through Toll-like receptors (TLRs), leading to the release of IL-12 and IL-23. These cytokines promote the differentiation of recruited CD4+ T cells into Th1 and Th17 subsets. Simultaneously, monocytes differentiate into macrophages that produce interleukin-6 (IL-6), matrix metalloproteinases (MMPs), tumor necrosis factor-α (TNF-α), and vascular endothelial growth factor (VEGF), thereby contributing to vascular wall destruction and remodeling. VEGF not only promotes neovascularization but also contributes to fibroblast proliferation and adventitial fibrosis. Myofibroblasts proliferate within the intima, resulting in intimal hyperplasia [5,6]. A schematic overview of these inflammatory signaling pathways is provided in Figure 2. It is important to further dissect these classical inflammatory cascades not only from the perspective of genetic susceptibility but also in the context of various immune dysregulations and immunosenescence.

3.1. Genetic Factors

Genetic susceptibility is an essential factor in understanding the pathophysiology of GCA, and it has long been associated with the HLA-DRB1*04 allele [7,8].
The largest genome-wide association study (GWAS) to date, involving GCA patients from Europe and North America, confirmed previously established associations with HLA class II alleles and the plasminogen gene, and additionally identified three novel genetic loci. These include MFGE8 and VTN, both implicated in angiogenesis, as well as CCDC25, a gene associated with neutrophil extracellular traps (NETs) [9]. Although the HLA-DRB1*15 haplotype has previously been considered protective against GCA, recent evidence suggests its involvement in classical cranial GCA, with the HLA-B15:01 allele frequently observed in patients with large-vessel involvement [10]. Most recently, integrative analyses combining single-cell RNA sequencing with Mendelian randomization have suggested that three genes—RCAN3, RPS6, and HLA-DQB1—may confer a protective effect against the development of GCA [11].
Our recent work identified additional genetic risk factors. The GWAS data from the UK Biobank and FinnGen cohorts revealed that the T allele of the SNP rs7044155, located within the very low-density lipoprotein receptor gene region, is associated with an increased risk of developing GCA. Furthermore, the same study employed Mendelian randomization analysis, suggesting that elevated levels of low-density lipoprotein cholesterol may contribute to the risk of GCA onset [12].
These advances in genetic research have substantially expanded our understanding of the heritable components of GCA, and ongoing investigation is expected to further elucidate the genetic architecture underlying disease susceptibility.

3.2. Environmental Factors

Environmental factors, such as bacterial or viral infections (e.g., Mycoplasma pneumoniae, Chlamydia pneumoniae, Parvovirus B19, Herpes Zoster Virus), have been proposed as triggers for GCA. Although direct invasion of the vascular wall by infectious pathogens and deposition of immune complexes have been proposed as potential mechanisms of vascular injury, no studies have definitively demonstrated these processes, and the supporting evidence remains limited [13,14]. Reductions in beneficial microbes such as Lachnospiraceae and Bacteroidales may disrupt regulatory T-cell (Treg) homeostasis and promote Th17 polarization, thereby contributing to the development of vascular inflammation [15,16].

3.3. Immune Checkpoint Dysregulation

While genetic studies have shed light on the heritable predisposition to GCA, accumulating evidence highlights the critical role of immune checkpoint dysregulation in its pathophysiology [5]. In vascular lesions of patients with GCA, vascular dendritic cells exhibit reduced expression of the immunosuppressive ligand programmed death-ligand 1 (PD-L1), whereas infiltrating CD4+ T cells display elevated expression of programmed death-1 (PD-1) [17,18]. Supporting these findings, a study using C57BL/6 mice demonstrated that, under non-inflammatory conditions, vascular-resident dendritic cells express higher levels of PD-L1 compared to splenic dendritic cells, while PD-L1 expression is significantly downregulated following inflammation induced by lipopolysaccharide [19]. These observations collectively support the concept that local immune checkpoint dysfunction plays a crucial role in GCA.
While immune checkpoint signaling is locally suppressed in vascular tissues, patients with GCA and polymyalgia rheumatica (PMR) exhibit elevated systemic levels of soluble immune checkpoint molecules (sICMs), including soluble cytotoxic T-lymphocyte antigen 4 (sCTLA-4), sPD-1, sPD-L1, and sPD-L2, compared to healthy individuals. These molecules have shown strong discriminatory ability in distinguishing PMR and GCA from healthy controls [20]. Additionally, patients with GCA exhibit significantly increased frequencies of PD-1-positive circulating peripheral helper T cells and T follicular helper cells compared to healthy controls [21]. These findings suggest that systemic immunoregulatory mechanisms may be activated in these conditions. However, the functional relevance of these elevated sICM levels remains unclear, and it is not yet known whether these immunosuppressive pathways are effectively functioning.
The possible association between immune checkpoint inhibitors and large-vessel vasculitis has also been reported. In a human artery-engrafted Severe Combined Immunodeficiency (SCID) mouse model, injection of peripheral blood mononuclear cells (PBMCs) from GCA patients followed by administration of an anti-PD-1 antibody resulted in marked T-cell infiltration into the vessel wall, accompanied by excessive production of proinflammatory cytokines such as IFN-γ, IL-17, and IL-21. Histologically, this was associated with pronounced intimal hyperplasia, neoangiogenesis, and vascular remodeling [17].
Furthermore, clinical studies have documented a significantly increased incidence of GCA in cancer patients treated with the anti-CTLA-4 antibody ipilimumab. In patients with GCA, dysfunctional Treg overexpress CTLA-4, which may increase their sensitivity to CTLA-4 blockade by ipilimumab [22]. These findings suggest that the CTLA-4 pathway plays a critical role in the immunopathogenesis of GCA.
Recent studies have also identified functional impairments in two major inhibitory checkpoint pathways in GCA, the PD-1/PD-L1 axis and the CD96/CD155 axis, leading to the concept of “checkpoint dysfunction” [23]. This weakening of inhibitory signaling is believed to promote persistent T-cell activation and sustained generation of proinflammatory effector T cells, including Th1, Th17, and Th9 subsets [23]. Th1 cells enhance inflammation via IFN-γ production, while Th17 cells stimulate vascular endothelial cells to secrete IL-6 and granulocyte–macrophage colony-stimulating factor (GM-CSF), which in turn activate macrophages and drive vascular remodeling. Additionally, reduced expression of CD155 on antigen-presenting cells (APCs) may impair CD96-mediated T-cell suppression, thereby perpetuating inflammation and aggravating vasculitis through expansion of Th9 cells [23].
These findings underscore the need for further research to clarify the regulatory mechanisms governing immune checkpoint expression in vascular inflammation.

4. Aging and Inflammation

GCA predominantly affects individuals over the age of 50 years, suggesting that aging is a significant risk factor for disease onset. Recent advances in aging research have increasingly highlighted physiological changes, as well as the roles of immunosenescence and senescence-associated immune dysregulation, in the pathogenesis of GCA. The concept of inflammaging has recently emerged to describe the phenomenon of chronic, low-grade systemic inflammation that develops with age in the absence of overt infection and has been reported to be associated with persistent elevations of proinflammatory cytokines, including IL-6 and TNF-α [24,25,26,27,28,29]. As central mechanisms of inflammaging, age-related mitochondrial dysfunction and epigenetic alterations are believed to drive cellular senescence, with senescent cells acquiring a senescence-associated secretory phenotype (SASP) that further amplifies chronic inflammation [30].
In hematopoietic stem cells (HSCs), aging is associated with chromatin remodeling that facilitates the transcription of genes involved in inflammation and oxidative stress. This shift promotes the preferential generation of myeloid-biased HSC subsets over lymphoid-biased subsets. Notably, while myeloid-biased HSCs retain their self-renewal capacity, the self-renewal ability of lymphoid-biased HSCs declines with age. These findings may partially explain the diminished immune competence observed in elderly individuals [31]. Recent evidence indicates that clonal hematopoiesis of indeterminate potential (CHIP), an age-associated somatic mutation in hematopoietic stem cells, contributes to sustained inflammation through the emergence of immune cells with heightened pro-inflammatory cytokine production [32].
This persistent inflammatory state is believed to contribute to the development of age-related diseases, including atherosclerosis, heart failure, stroke, and vascular stenosis [32,33]. Recent evidence further suggests that inflammaging may contribute to the pathophysiology of GCA [33,34]. In this section, we summarize the key features of these mechanisms and examine the potential impact of age-related immunological changes on GCA (Figure 3).

4.1. Epigenetic Alterations

Among the mechanisms linking aging to chronic inflammation in GCA, epigenetic alterations have emerged as key contributors. Genomic instability plays a critical role in the aging process and is accompanied by diverse epigenetic changes, typically involving transcriptional regulation through DNA methylation, histone modifications, and transposable elements. Post-transcriptional gene regulation is further mediated by non-coding RNAs such as microRNAs (miRNAs) [35,36]. Notably, the epigenetic alterations observed in patients with GCA appear to overlap with those found in senescent T cells [37,38].
Epigenetic profiling using genome-wide DNA methylation arrays on TAB specimens from GCA patients revealed hypomethylation events associated with increased activity of the calcineurin/nuclear factor of activated T cells (NFAT) pathway compared to healthy controls [37].
NFATC1, a member of the NFAT transcription factor family, was localized to CD3+, CD4+, and CD8+ T lymphocytes, as well as CD68+ macrophages infiltrating the vascular wall in TAB specimens. Within these tissues, several pro-inflammatory miRNAs associated with T-cell activation, including those regulated by NFATC1, were found to be upregulated. In contrast, multiple regulatory immune-related miRNAs were downregulated. These findings suggest that dysregulation of the calcineurin/NFAT signaling pathway may impair the proper execution of T cell-mediated immune responses [39].
One such miRNA, miR-21, is known to increase with age and promote the differentiation of activated CD4+ T cells from memory to inflammatory effector phenotypes, thereby contributing to vascular remodeling [40,41]. Importantly, elevated levels of miR-21 have been demonstrated in inflamed temporal arteries from GCA patients [38]. Although a causal relationship between these epigenetic changes and the development of GCA has not yet been established, these aberrantly expressed miRNAs in TAB specimens from GCA patients may serve as potential diagnostic biomarkers or therapeutic targets.

4.2. Clonal Hematopoiesis of Indeterminate Potential (CHIP)

In addition to epigenetic modifications, clonal alterations in HSCs have also been recognized as important age-associated contributors to chronic inflammation. CHIP refers to the age-related acquisition of somatic mutations in HSCs—most commonly loss-of-function mutations in DNMT3A and TET2—in the absence of overt hematologic malignancy or other known clonal disorders. CHIP is defined by the presence of such somatic mutations in peripheral blood with a variant allele frequency of ≥2% [42,43].
Recent murine studies have demonstrated that aged mice exhibit increased intestinal permeability and microbial dysbiosis, both of which correlate with the expansion of mutant HSCs. Notably, treatment with broad-spectrum antibiotics resulted in a reduction in these mutant HSCs. Conversely, fecal microbiota transplantation from colitic or aged mice into healthy recipients promoted the expansion of DNMT3A-deficient HSCs. These findings suggest a potential role for the gut microbiome in driving the clonal expansion of mutant HSCs [44]. In particular, recent work has identified ADP-D-glycero-β-D-manno-heptose (ADP-heptose)—a metabolic byproduct specific to Gram-negative bacteria—as a key microbial-derived ligand. ADP-heptose activates transcriptional reprogramming and NF-κB signaling via the host receptor ALPK1, thereby promoting CHIP development [45,46].
Once somatic mutations arise, monocytes derived from these mutant HSCs can be recruited from the bloodstream into atherosclerotic plaques, where they produce excessive amounts of IL-6, IL-1β, and chemokines, contributing to the progression of atherosclerosis [47,48,49,50]. CHIP has also been associated with hematologic malignancies and may influence the efficacy and toxicity of therapies such as hematopoietic stem cell transplantation and CAR-T cell therapy [51]. In solid tumors, TET2 mutations in CHIP have been linked to tumor-infiltrating clonal hematopoiesis, which in turn has been associated with an increased risk of recurrence and mortality in patients with solid cancers [52,53]. Moreover, CHIP—particularly DNMT3A mutations—has been implicated in osteoporosis through IL-20-mediated osteoclast activation [54], as well as in the exacerbation of periodontitis via enhanced osteoclast activity [55].
More recently, CHIP has been reported to be associated not only with autoinflammatory syndromes such as VEXAS syndrome, but also with autoimmune diseases, including rheumatoid arthritis, systemic lupus erythematosus, and vasculitis [32,56,57,58,59]. A study investigating the prevalence of CHIP in patients with GCA, Takayasu arteritis (TAK), and ANCA-associated vasculitis (AAV) found that CHIP was detected in 60% of GCA patients, including in temporal artery tissue specimens. The frequency was higher than that in age-matched healthy individuals and greater than that in patients with TAK or AAV. However, other studies have shown that the difference between GCA patients and healthy controls becomes statistically insignificant after adjusting for age; thus, the true prevalence of CHIP in GCA remains uncertain [58,60,61].
In ex vivo stimulation assays using macrophages derived from GCA patients, those harboring CHIP produced significantly higher levels of inflammatory chemokines, including MCP-1, MIP-1α, and MIP-1β. Moreover, a higher variant allele frequency of mutated clones was associated with an increased risk of relapse [61], particularly among patients carrying TET2 or JAK2 mutations [60]. Notably, TET2 mutations have been linked to an elevated risk of ischemic vision loss in GCA [62,63]. Additionally, male GCA patients exhibited a 4.8-fold increased risk of developing myeloid malignancies such as myeloproliferative neoplasms compared to the general population, with a high prevalence of JAK2 V617F mutations among affected individuals [64]. GCA cases harboring JAK2 mutations were characterized by fewer cranial symptoms, such as headaches, and elevated platelet counts [65].
Interestingly, while CHIP has traditionally been attributed solely to somatic genetic mutations, recent evidence suggests a possible contribution of DNA methylation instability (DMI). DMI refers to increased variability in methylation levels at CpG sites that are normally stable. This phenomenon has been associated with disease progression and relapse in hematologic malignancies, cardiovascular disease, and aging. Although a direct causal link to CHIP has not yet been established, DMI may represent a novel, mutation-independent mechanism contributing to CHIP development [66].
In summary, mutant clones arising from CHIP may contribute to the chronic inflammatory response and risk of relapse in GCA through the excessive production of proinflammatory cytokines and chemokines. However, whether CHIP directly contributes to the onset of GCA remains uncertain, and definitive evidence is currently lacking.

4.3. Immunosenescence

Immunosenescence encompasses both the age-related functional decline of immune cells in the innate and adaptive immune systems and cellular senescence, characterized by cell-cycle arrest, evasion of apoptosis, and acquisition of a SASP. This process is recognized as a major driver of inflammaging. For instance, DCs exhibit age-associated impairments in TLR function, increased production of proinflammatory cytokines, and heightened reactivity to self-antigens [67]. In T cells, age-related changes are linked to thymic involution, leading to alterations in both composition and function. Specifically, aging is associated with a reduction in naïve T cells and Tregs, along with a relative expansion of memory and effector T-cell populations [68,69,70,71,72]. In patients with GCA, a decreased number of CD4+ Tregs has been reported compared to healthy controls, suggesting their involvement in disease pathogenesis [73]. More recently, attention has shifted toward functional defects in CD8+ Tregs and their association with autoimmune diseases [68]. In GCA, CD8+ Treg deficiency has been clearly identified. Aberrant signaling via NOTCH4 receptors expressed on CD8+ Tregs disrupts the secretion of exosomes containing NOX2, resulting in a failure to suppress CD4+ T-cell activation and thereby contributing to persistent and exacerbated inflammation [74]. Moreover, dysregulation of glycolytic enzymes and reduced T-cell receptor (TCR)-induced calcium influx have also been implicated in Treg dysfunction in GCA [75].
Interestingly, aging markers such as p21 and p16 are highly expressed not only in macrophages and T cells infiltrating the temporal arteries of GCA patients, but also in vascular smooth muscle cells and fibroblasts [76]. The senescence of vascular endothelial and smooth muscle cells induces a so-called SASP, characterized by the production of inflammatory cytokines (e.g., IL-6, IL-1β), growth factors (e.g., VEGF), and MMPs [77]. This SASP contributes to structural fragility of the vessel wall and breakdown of tissue tolerance, creating a microenvironment conducive to the onset of GCA [78]. This tissue damage is mediated via IL-6-dependent pathways, and tocilizumab has been suggested to suppress this inflammation-promoting mechanism [79].
Taken together, immunosenescence, characterized by the reduction in and functional impairment of Tregs, may permit excessive activation of pro-inflammatory T cells and thereby contribute to vascular wall inflammation. In addition, cellular senescence, through the SASP, may help sustain and amplify the inflammatory milieu. These processes may act synergistically in the pathogenesis and progression of GCA.

4.4. The Dual Roles of Senescence

A fundamental question in aging research is whether aging is entirely detrimental. Surprisingly, individuals with CHIP have been reported to have a lower risk of developing Alzheimer’s disease, a prototypical age-related disorder. Furthermore, among CHIP carriers who have not developed dementia, neuropathological changes—such as amyloid plaques and tau pathology—have been observed to be relatively mild [80,81]. One hypothesis proposes that myeloid cells bearing CHIP-associated mutations may infiltrate and engraft in the brain, potentially compensating for the age-related functional decline of resident microglia by acting as microglia-like cells [82].
In a murine model of liver fibrosis, the elimination of p16 Ink4a-positive macrophages (senescent macrophages) resulted in an improvement in fibrosis. In contrast, depletion of p16 Ink4a-positive endothelial cells (senescent endothelial cells) led to a worsening of fibrosis. These findings suggest that, while senescent macrophages exacerbate fibrosis, senescent endothelial cells may exert protective effects by expressing anti-fibrotic and reparative factors such as VEGF, thereby contributing to tissue repair and angiogenesis [83]. Thus, senescent cells are not uniformly detrimental, and future senolytic therapies will likely require cell-type-specific targeting [84].

5. Omics-Based Pathophysiological Insights

Recent advances in omics technologies have significantly enhanced our understanding of the molecular mechanisms underlying GCA. Through transcriptomic, proteomic, and spatial transcriptomic analyses, diverse immune pathways, cell types, and potential therapeutic targets have been identified. This section provides an overview of the key findings from recent omics-based studies, as summarized in Table 1, with particular emphasis on their implications for therapeutic intervention.
The role of CD4+ T cells has garnered significant attention as a key immune component in the pathophysiology of GCA. Single-cell RNA sequencing (scRNA-seq) analysis has revealed a marked expansion of cytotoxic CD4+ T cells (CTLs) expressing identical TCR sequences during active disease. These CTLs exhibit high expression levels of granzyme genes (GZMB, GZMK) and chemokine genes (CCL4, CCL5), suggesting their involvement in apoptosis induction, pro-inflammatory responses, and the recruitment of other immune cells. Notably, Maraviroc, a CCR5 antagonist that targets the receptor for CCL5, has been proposed as a potential therapeutic agent [85].
In relation to CD8+ T cells, an increased frequency of Ki-67-positive cells has been observed in the peripheral blood of patients with GCA, indicating a proliferative phenotype. These cells display a reduced activation threshold for TCR stimulation and can be activated even in the absence of co-stimulatory signals. Indeed, the scRNA-seq analysis of PBMCs demonstrated upregulation of genes such as KLRD1 and IFITM1, which are associated with antiviral responses and immune activation, and downregulation of GNLY and ZFP36L2, which are involved in cytotoxic function and proliferative control. These gene expression profiles are consistent with previous findings. Furthermore, although CD8+ T cells in affected vascular lesions expressed IFN-γ, they lacked Ki-67 expression, suggesting that these cells were not undergoing local clonal expansion but were instead recruited from the peripheral circulation following prior activation and proliferation [86].
In studies investigating the histological heterogeneity of GCA, gene expression profiles have been compared between two subtypes of TAB tissue: the transmural inflammation (TMI) type and the inflammation-limited-to-the-adventitia (ILA) type. In the TMI subtype, a broad range of immune-activating molecules—including members of the TNF superfamily, immune checkpoints, chemokines and their receptors, TLRs, complement components, IgG Fc receptors, signaling lymphocytic activation molecule (SLAM) family members, JAK3, STAT1, and STAT4—were significantly upregulated. In contrast, the gene expression profile of the ILA subtype resembled that of normal tissue, indicating that TMI-type GCA exhibits a transcriptional profile reflective of marked inflammation. These findings highlight distinct molecular signatures that correspond to different histopathological classifications of GCA [87].
A spatial transcriptomics study further demonstrated upregulated gene expression across all arterial layers in TAB specimens from patients with GCA, with the most pronounced changes observed in the intima and media. In addition to increased expression of macrophage-related genes such as MMP2 and MMP9, CD74 was identified as the most highly expressed gene across all layers. CD74, a transmembrane receptor for macrophage migration inhibitory factor (MIF), is expressed on antigen-presenting cells and is known to be involved in apoptosis and tissue repair. These findings support the notion that angiogenic activity is primarily localized to the intima and media and suggest that several druggable molecular targets—including CD74, MMPs, and CXCR4—may represent promising candidates for future therapeutic development [88].
Furthermore, spatial transcriptomics combined with pseudotime analysis demonstrated a sustained increase in MMP12 expression throughout the disease course, reaching up to a 50-fold elevation. These findings suggest that MMP12 may serve as a potential biomarker reflecting disease activity in GCA [89]. In addition, trans-omics analysis of systemic vasculitis revealed that MMP12 is specifically expressed by CD206+ macrophages and multinucleated giant cells. Notably, MMP12 expression remained indicative of disease activity even under IL-6 blockade, suggesting its potential utility as a predictor of disease relapse [90].

6. Current Therapeutic Strategies and Their Limitations

In this section, we provide an overview of the current therapeutic challenges and the therapeutic targets that have been proposed to date. The pathophysiology of GCA involves diverse immune cells and cytokine pathways, and understanding these mechanisms is essential for the development of novel therapeutic strategies. A conceptual diagram summarizing these pathogenic mechanisms is presented in Figure 4.

6.1. Glucocorticoid

Glucocorticoid (GC) remain the cornerstone of remission induction in GCA, exerting their anti-inflammatory effects through downregulation of pro-inflammatory genes, upregulation of GC receptor targets (e.g., FKBP5), and increased expression of anti-inflammatory markers such as CD163 [91,92].
However, in GCA patients treated with GC, a second TAB performed on the contralateral side revealed persistent vascular inflammation in approximately half of the patients, even one year after treatment initiation [93]. Moreover, prolonged GC use is associated with considerable toxicity. A recent study comparing patients receiving methylprednisolone pulse therapy followed by oral GC with those receiving GC alone demonstrated no significant difference in visual recovery, while the risk of developing diabetes within one year was significantly higher in the pulse group [94]. Furthermore, reports have indicated that even a 26-week rapid GC tapering regimen does not adequately prevent GC-related adverse events [95]. These adverse effects warrant particular attention in elderly patients with GCA.

6.2. Tocilizumab

The addition of tocilizumab (TCZ), an IL-6 inhibitor, significantly reduces relapse rates and cumulative GC exposure in GCA, establishing it as a major therapeutic option [96]. In real-world clinical practice in Japan, 85% of patients were able to taper GC without relapse at 52 weeks [97]. Furthermore, TCZ has been reported to prevent new visual complications and may reduce the risk of blindness [98].
However, tapering or discontinuation of TCZ is associated with relapse risk [99], and approximately 47% of patients with GCA relapse within five years [100]. In a study of patients with GCA treated with GC and TCZ who underwent re-biopsy at a median of 2.4 years after treatment initiation, persistent vasculitis was observed in about 40% of cases, despite well-controlled clinical disease activity [101]. These challenges highlight the need for more fundamental therapeutic strategies.

6.3. JAK Inhibitor

JAK inhibitors target intracellular signaling downstream of multiple pro-inflammatory cytokines (IL-6, IFN-γ, GM-CSF). Baricitinib, tofacitinib, and upadacitinib have been demonstrated to suppress vascular inflammation in murine models and to achieve clinical remission in approximately 60% of patients with relapsing GCA in real-world settings, including those refractory to IL-6 blockade [102]. In a Phase 3 trial of upadacitinib (a JAK1-selective inhibitor) in GCA, sustained remission rates at 52 weeks were significantly higher (15 mg (46%) vs. placebo (29%)), with additional steroid-sparing effects [103]. Based on these results, upadacitinib has been approved for the treatment of GCA. However, the potential cardiovascular and malignancy risks associated with JAK inhibitors require careful consideration [104].

6.4. GM-CSFR Inhibitor

GM-CSF and its receptor (GM-CSFRα) are upregulated in GCA lesions, particularly in macrophages, T cells, and vascular endothelial cells, where they activate JAK2/STAT5 signaling. Mavrilimumab, a monoclonal antibody targeting GM-CSFRα, has been shown in ex vivo cultures of GCA arteries to reduce inflammatory infiltrates, pro-inflammatory cytokines (IL-6, TNFα, IL-1β), oxidative stress, and angiogenesis [105]. In a Phase 2 trial, mavrilimumab demonstrated superior efficacy to placebo in achieving sustained remission, yielding promising results; however, a Phase 3 trial is not currently underway [106].

6.5. CTLA-4 Ig

As noted above, dysregulation of immune checkpoint pathways contributes to the pathophysiology of GCA. Abatacept (CTLA-4 Ig), which modulates T-cell co-stimulation via CD80/86, demonstrated superiority over standard therapy in achieving relapse-free survival in a Phase 2 trial of GCA [107]. A Phase 3 trial is currently underway (NCT04474847).

6.6. IL-17 Inhibitor

IL-17A is highly expressed in GCA arteries and correlates with the intensity of inflammation. Secukinumab (a monoclonal antibody against IL-17A) showed promising results in a Phase 2 trial (70% remission maintenance vs. 20% with placebo at 28 weeks). Phase 3 trials are currently ongoing (NCT04930094, NCT05380453) [108].

6.7. IL-12/IL-23 Inhibitor

IL-23 is a key cytokine for the proliferation and maintenance of Th17 cells. Inflammatory aortic aneurysms exhibit upregulated expression of the IL-23 receptor gene, suggesting an association with GCA [109]. However, studies of Ustekinumab (an IL-12/23 inhibitor) in GCA have not demonstrated consistent efficacy. Furthermore, a clinical trial of Guselkumab (an anti–IL-23 antibody) was terminated (NCT04633447) [108]. Thus, although targeting the IL-12/23 pathway remains theoretically promising, its clinical efficacy in GCA has yet to be established.

6.8. IL-21 Inhibitor

IL-21 is primarily produced by Tfh cells and has been reported to promote B-cell activation and support the maintenance of effector CD8+ T-cell responses [110]. Its expression is epigenetically suppressed by the transcription factor Blimp-1, whose downregulation is frequently observed in autoimmune diseases. Inhibition of IL-21 has been shown to decrease Th1 and Th17 cells, while potentially increasing Th2 cells with protective functions [111]. However, evidence for its clinical application in GCA remains limited.

7. Emerging Immune Cells and Potential Therapeutic Targets

In this section, we provide an overview of the major immune cell populations that have recently gained attention in the pathogenesis of GCA and examine their potential as therapeutic targets.

7.1. Macrophage

Macrophages are the primary cellular source of IL-6, MMP-9, and VEGF, and they play a pivotal role in mediating vascular wall destruction and remodeling in GCA [112,113]. The ontogeny of macrophages varies depending on the inflammatory state and their localization within the arterial wall [114].
Fate-mapping analyses combined with single-cell RNA sequencing have revealed that the developmental origin of arterial macrophages differs between homeostatic and inflammatory conditions. Under steady-state conditions, arterial macrophages are derived equally from yolk sac erythro-myeloid progenitors and the bone marrow, whereas during vascular inflammation, the majority originate from bone marrow-derived circulating monocytes that differentiate into adventitial macrophages. Notably, a subset of adventitial macrophages, characterized by LYVE1 expression and derived from the bone marrow under homeostatic conditions, may play a reparative role that appears to decline with age [115].
In a study investigating the pathogenesis of intimal inflammation using coronary artery specimens from patients with Kawasaki disease, intimal macrophages were found to express the chemokine receptor CCR2, suggesting their derivation from circulating monocytes. These macrophages are believed to contribute to vascular occlusion, remodeling, and thrombus formation [116].
Moreover, macrophages in GCA exhibit phenotypic diversity and functional heterogeneity, with distinct spatial distribution patterns depending on the disease stage. CD206+/MMP-9+ macrophages, which are involved in tissue destruction and angiogenesis, are induced in the early phase by GM-CSF and are localized to areas of tissue damage. In contrast, FRβ+ macrophages are induced in the later stages by macrophage colony-stimulating factor and are hypothesized to activate myofibroblasts and promote intimal hyperplasia, thereby contributing to luminal stenosis [114,117].
Recent transcriptomic analyses of treatment-naïve GCA lesions have identified macrophages and multinucleated giant cells expressing a wide array of molecules implicated in GCA pathogenesis, including MMP12 (critical for elastin degradation), ACP5 and ATP6V0D2 (osteoclast-associated bone resorption molecules), VDR and TREM2 (involved in osteoclastogenesis and phagocytosis), MRC1 (a marker of fibrosis-associated macrophages), and HLA-DRA (involved in antigen presentation to CD4+ T cells) [118]. These molecules are not unique to GCA but are also expressed in other granulomatous diseases such as sarcoidosis, tuberculosis, and TAK, suggesting the potential for shared therapeutic targets across these conditions.

7.2. Tissue-Resident Memory T Cell (Trm)

In patients with GCA definitively diagnosed by TAB, a study that performed a second TAB on the contralateral side revealed that approximately half of the patients exhibited persistent vascular inflammation even one year after the initiation of treatment [93]. Histopathological analysis demonstrated that T cells were the predominant residual cell population, drawing increasing attention to tissue-resident memory T cells (Trm) [68]. Trm are a subset of memory T cells that persist long term within tissues without recirculating. Upon reactivation, they exert local effector functions, including Th1-, Th2-, and Th17-type responses.
In a murine model of GCA, CD4+ CD103+ Trm were found to reside within the arterial wall [119]. In this study, arteries containing Trm were transplanted into SCID mice without the addition of PBMCs, yet the Trm remained viable over the long term and continued to produce cytokines such as IFN-γ, IL-17, and IL-21. The survival, maintenance, and cytokine production of Trm were shown to depend on CD28-mediated co-stimulation, which enhances glucose uptake in T cells, and on JAK1/3-STAT signaling for sustained activity [120]. Indeed, it has been suggested that tofacitinib may have therapeutic efficacy against Trm [119,121].
Recent omics analyses have identified DDIT4 and ARHGAP15 as susceptibility genes associated with GCA. In particular, DDIT4 promotes mitochondrial fission and contributes to the maintenance of chronic inflammation in Trm by regulating lipid metabolism and reactive oxygen species (ROS). Notably, deletion of DDIT4 resulted in a significant reduction in chronic inflammation, highlighting its potential as a novel therapeutic target [122].

7.3. Stem-like T Cell

Recent studies have reported the formation of lymph node-like tertiary lymphoid structures within the aortic wall of patients with GCA, where clusters of CD4+ T cells and B cells are present. Among these, TCF1 hi IL-7R+ stem-like CD4+ T cells express the IL-7R and exhibit stem cell-like properties, including self-renewal and the ability to differentiate into Eomesodermin (EOMES)+ cytotoxic T cells and B-cell lymphoma 6+ follicular helper-like T cells. These stem-like CD4+ T cells play a central role in sustaining chronic inflammation. Notably, in mouse models, the depletion of these cells resulted in the prevention of vasculitis relapse [123].
EOMES is a transcription factor essential for the differentiation of T cells—particularly CD8+ T cells—into memory cells and for their cytotoxic function. T cells harboring TET2 mutations exhibit increased EOMES expression and display a bias toward differentiation into memory CD8+ T cells. As a result, although primary immune responses may remain intact, secondary responses—such as those elicited upon reinfection—may become exaggerated [32]. Eliminating IL-7R+ TCF1hi CD4+ stem-like T cells in TLS or inhibiting TLS formation represents a novel strategy to prevent relapse [123].

7.4. B Cell

Although the pathogenesis of GCA has traditionally been investigated primarily through the lens of cellular immunity, recent studies have increasingly emphasized the role of humoral immunity. In GCA, B cells that infiltrate the vessel wall from the adventitial vasculature form tertiary lymphoid organs and secrete a range of inflammatory cytokines—including IL-6, GM-CSF, TNF-α, and lymphotoxin-β (LTβ)—as well as the anti-inflammatory cytokine IL-10. These B cells may promote the differentiation of macrophages migrating from the vasculature toward the media into a proinflammatory phenotype [124].
Moreover, a recent study reported that 57% of GCA patients tested positive for anti-VSIG10L antibodies and 43% for anti-DCBLD1 antibodies. In contrast, these antibodies were not detected in patients with TAK or in healthy controls, suggesting that antibody-mediated humoral immunity may also play a role in the pathogenesis of GCA [125]. These findings highlight the potential of B cell-targeted therapies.

7.5. Vascular Smooth Muscle Cell

Vascular smooth muscle cells (VSMCs) play a pivotal role in vascular inflammation and remodeling. In vitro, they respond to stimulation with IFN-γ or PDGF by producing chemokines such as CXCL9, CXCL10, CXCL11, and CCL2, thereby promoting the chemotaxis of monocytes and Th1 cells and recruiting CD8+ T cells into the vessel wall. In addition, VSMCs release matrix metalloproteinases (MMP-2 and MMP-9) and interact with fibroblasts and myofibroblasts to drive intimal hyperplasia and vascular occlusion [6]. Experimental studies have further demonstrated that inhibition of PDGF signaling with imatinib reduces VSMC-derived CCL2 production, thereby attenuating T-cell chemotaxis [126].

7.6. Myofibroblast

Myofibroblasts contribute to intimal hyperplasia and vascular occlusion. Their origins vary depending on the organ and tissue context, arising from multiple cell types [127]. In GCA, myofibroblasts are believed to differentiate from various sources, including vascular smooth muscle cells [128,129], endothelial cells [17], and fibroblasts [130]. Morphologically, myofibroblasts are irregular, spindle-shaped cells with high migratory and proliferative capacities, and they exhibit approximately twice the contractile strength of quiescent fibroblasts [127]. Myofibroblasts produce profibrotic mediators such as TGF-β1, angiotensin II, and IL-1, thereby contributing to both fibrosis and tissue repair [131].
Recent studies have suggested that, in temporal artery specimens from patients with GCA, adventitial CD90+ fibroblasts differentiate into activated fibroblasts that migrate from the adventitia toward the intima, contributing to neointimal formation [132]. Signals that induce fibroblast-to-myofibroblast differentiation include TGF-β signaling and IL-6 [131].
Neointimal myofibroblasts (characterized by α-SMA+ CD90+ Desmin+ MYH11+ expression) in GCA lesions are not merely structural cells but are immunologically active. These cells exhibit activated IFN-γ signaling, as evidenced by phosphorylated STAT1, and promote Th1/Tc1 and Th17/Tc17 polarization by producing IL-12 and IL-23, thereby creating a pro-inflammatory microenvironment. Accordingly, JAK inhibitors represent a potential therapeutic strategy [6,133]. Furthermore, recent genome-wide gene expression profiling has revealed that myofibroblasts involved in intimal hyperplasia express LRRC15, suggesting that these cells contribute to the formation of an immunosuppressive microenvironment by inhibiting cytotoxic CD8+ T cells and highlighting LRRC15 as a potential therapeutic target [118].

7.7. Neutrophil

In chronic inflammatory states, immature neutrophils are released into peripheral blood, and patients with GCA exhibit significantly higher numbers of circulating immature neutrophils compared to healthy individuals. These immature neutrophils have a longer lifespan than their mature counterparts and demonstrate resistance to apoptosis. They produce large quantities of ROS, disrupt the vascular endothelial barrier, and increase vascular permeability [134,135].
Temporal artery biopsies from GCA patients have revealed the presence of NETs, primarily localized near the vasa vasorum in the adventitia. These NETs contain proinflammatory cytokines such as IL-6 and IL-17A [136]. Mitochondrial-derived N-formyl methionine peptides (fMET), released during NET formation, act as potent neutrophil chemoattractants and correlate strongly with inflammatory markers such as C-reactive protein (CRP) and erythrocyte sedimentation rate. fMET induces ROS production via the G-protein-coupled receptor formyl peptide receptor 1 (FPR1), and inhibition of FPR1 suppresses ROS generation [137]. Moreover, ROS production is elevated in GCA leukocytes, contributing to oxidative modifications of fibrinogen and potentially promoting thrombosis. This effect is attenuated by treatment with tocilizumab [138].
These potential therapeutic targets remain under investigation, and further studies are needed to evaluate their efficacy and facilitate their translation into clinical practice.

8. Conclusions and Future Perspectives

Recent studies have revealed that age-related mechanisms, including immunosenescence and CHIP, contribute to the pathogenesis of giant cell arteritis. Moreover, emerging evidence has highlighted the roles of macrophages, Trm, and stromal cells as novel immune players in disease progression. Integrating these insights with multi-omics approaches holds promise for developing precision medicine strategies aimed at achieving sustained remission and preventing irreversible vascular damage.

Author Contributions

M.S., writing—original draft preparation; R.W., writing—review and editing; S.T. provided pathological images and offered expert pathological insights; R.I. and G.K. provided general support for the study; M.H., supervision. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We sincerely thank the faculty members of the Department of Clinical Immunology at Osaka Metropolitan University for their invaluable advice and guidance throughout this study. We are also deeply grateful to the staff of the Department of Rheumatology at Hyogo Prefectural Amagasaki General Medical Center for their generous support and collaboration, which greatly contributed to the successful completion of this research. We also acknowledge BioRender.com (https://biorender.com, accessed on 1 July 2022) for providing the schematic illustration.

Conflicts of Interest

R.W. received a research grant from AbbVie and speaker’s fee from AbbVie, Asahi Kasei, Chugai, Eli Lilly, GSK, and Taisho. M.H. received research grants and/or speaker’s fee from AbbVie, Asahi Kasei, Astellas, Bristol Meyers, Chugai, EA Pharma, Eisai, Daiichi Sankyo, Eli Lilly, Novartis Pharma, Taisho Toyama, Tanabe Mitsubishi, Towa Pharma, and UCB Japan. These pharmaceutical companies had no role in the writing of the manuscript.

References

  1. Weyand, C.M.; Goronzy, J.J. Medium- and large-vessel vasculitis. N. Engl. J. Med. 2003, 349, 160–169. [Google Scholar] [CrossRef] [PubMed]
  2. Fernández-Lozano, D.; Hernández-Rodríguez, I.; Narvaez, J.; Domínguez-Álvaro, M.; De Miguel, E.; Silva-Díaz, M.; Belzunegui, J.M.; Moriano Morales, C.; Sánchez, J.; Galíndez-Agirregoikoa, E.; et al. Incidence and clinical manifestations of giant cell arteritis in Spain: Results of the ARTESER register. RMD Open 2024, 10, e003824. [Google Scholar] [CrossRef]
  3. Monti, S.; Milanesi, A.; Klersy, C.; Tomelleri, A.; Dagna, L.; Campochiaro, C.; Farina, N.; Muratore, F.; Galli, E.; Marvisi, C.; et al. Age at diagnosis influences the clinical phenotype, treatment strategies and outcomes in patients with giant cell arteritis: Results from the observational GCAGE study on a large cohort of 1004 patients. Ann. Rheum. Dis. 2023, 82, 1098–1106. [Google Scholar] [CrossRef]
  4. Hysa, E.; Mantello, E.; Gotelli, E.; Campitiello, R.; Cutolo, C.A.; Matteson, E.L. Environmental factors in polymyalgia rheumatica and giant cell arteritis. J. Environ. Rheumatol. 2025, 2. [Google Scholar] [CrossRef]
  5. Pugh, D.; Karabayas, M.; Basu, N.; Cid, M.C.; Goel, R.; Goodyear, C.S.; Grayson, P.C.; McAdoo, S.P.; Mason, J.C.; Owen, C.; et al. Large-vessel vasculitis. Nat. Rev. Dis. Primers 2022, 7, 93. [Google Scholar] [CrossRef]
  6. Karabayas, M.; Ibrahim, H.E.; Roelofs, A.J.; Reynolds, G.; Kidder, D.; De Bari, C. Vascular disease persistence in giant cell arteritis: Are stromal cells neglected? Ann. Rheum. Dis. 2024, 83, 1100–1109. [Google Scholar] [CrossRef]
  7. Carmona, F.D.; Vaglio, A.; Mackie, S.L.; Hernández-Rodríguez, J.; Monach, P.A.; Castañeda, S.; Solans, R.; Morado, I.C.; Narváez, J.; Ramentol-Sintas, M.; et al. A Genome-wide Association Study Identifies Risk Alleles in Plasminogen and P4HA2 Associated with Giant Cell Arteritis. Am. J. Hum. Genet. 2017, 100, 64–74. [Google Scholar] [CrossRef] [PubMed]
  8. Carmona, F.D.; Coit, P.; Saruhan-Direskeneli, G.; Hernández-Rodríguez, J.; Cid, M.C.; Solans, R.; Castañeda, S.; Vaglio, A.; Direskeneli, H.; Merkel, P.A.; et al. Analysis of the common genetic component of large-vessel vasculitides through a meta-Immunochip strategy. Sci. Rep. 2017, 7, 43953. [Google Scholar] [CrossRef]
  9. Borrego-Yaniz, G.; Ortiz-Fernández, L.; Madrid-Paredes, A.; Kerick, M.; Hernández-Rodríguez, J.; Mackie, S.L.; Vaglio, A.; Castañeda, S.; Solans, R.; Mestre-Torres, J.; et al. Risk loci involved in giant cell arteritis susceptibility: A genome-wide association study. Lancet Rheumatol. 2024, 6, e374–e383. [Google Scholar] [CrossRef]
  10. Prieto-Peña, D.; Remuzgo-Martínez, S.; Ocejo-Vinyals, J.G.; Atienza-Mateo, B.; Genre, F.; Muñoz-Jimenez, A.; Ortiz-Sanjuán, F.; Romero-Yuste, S.; Moriano, C.; Galindez-Agirregoikoa, E.; et al. The presence of both HLA-DRB1*04:01 and HLA-B*15:01 increases the susceptibility to cranial and extracranial giant cell arteritis. Clin. Exp. Rheumatol. 2021, 39 (Suppl. S129), 21–26. [Google Scholar] [CrossRef]
  11. Cheng, T.; Chen, J.; Shan, X.; Jia, B.; Sang, S.; Wang, Y.; Wei, Y.; Hu, Y. Potential Key Genes for Giant Cell Arteritis Revealed Based on Single-Cell Sequencing and Mendelian Randomization Analysis. Int Arch Allergy Immunol 2025, 1–13. [Google Scholar] [CrossRef] [PubMed]
  12. Iwasaki, T.; Watanabe, R.; Zhang, H.; Hashimoto, M.; Morinobu, A.; Matsuda, F. Identification of the VLDLR locus associated with giant cell arteritis and the possible causal role of low-density lipoprotein cholesterol in its pathogenesis. Rheumatology 2024, 63, 2754–2762. [Google Scholar] [CrossRef] [PubMed]
  13. Lidar, M.; Lipschitz, N.; Langevitz, P.; Shoenfeld, Y. The infectious etiology of vasculitis. Autoimmunity 2009, 42, 432–438. [Google Scholar] [CrossRef]
  14. Miyabe, C.; Miyabe, Y.; Miyata, R.; Ishiguro, N. Pathogens in Vasculitis: Is It Really Idiopathic? Jma J. 2021, 4, 216–224. [Google Scholar] [CrossRef]
  15. Omenetti, S.; Pizarro, T.T. The Treg/Th17 Axis: A Dynamic Balance Regulated by the Gut Microbiome. Front. Immunol. 2015, 6, 639. [Google Scholar] [CrossRef]
  16. Wu, M.; Liao, Z.; Zeng, K.; Jiang, Q. Exploring the causal role of gut microbiota in giant cell arteritis: A Mendelian randomization analysis with mediator insights. Front. Immunol. 2023, 14, 1280249. [Google Scholar] [CrossRef] [PubMed]
  17. Zhang, H.; Watanabe, R.; Berry, G.J.; Vaglio, A.; Liao, Y.J.; Warrington, K.J.; Goronzy, J.J.; Weyand, C.M. Immunoinhibitory checkpoint deficiency in medium and large vessel vasculitis. Proc. Natl. Acad. Sci. USA 2017, 114, E970–E979. [Google Scholar] [CrossRef]
  18. Watanabe, R.; Zhang, H.; Berry, G.; Goronzy, J.J.; Weyand, C.M. Immune checkpoint dysfunction in large and medium vessel vasculitis. Am. J. Physiol. Heart Circ. Physiol. 2017, 312, H1052–H1059. [Google Scholar] [CrossRef]
  19. Sun, L.; Zhang, W.; Zhao, L.; Zhao, Y.; Wang, F.; Lew, A.M.; Xu, Y. Self-Tolerance of Vascular Tissues Is Broken Down by Vascular Dendritic Cells in Response to Systemic Inflammation to Initiate Regional Autoinflammation. Front. Immunol. 2022, 13, 823853. [Google Scholar] [CrossRef]
  20. Hysa, E.; Camellino, D.; Dejaco, C.; Bauckneht, M.; Pesce, G.; Morbelli, S.; Bagnasco, M.; Cutolo, M.; Matteson, E.L.; Cimmino, M.A.; et al. Soluble Co-Inhibitory Immune Checkpoint Molecules Are Increased in Patients With Polymyalgia Rheumatica Without Significant Correlations With Clinical Status: A Case-Control Study. ACR Open Rheumatol. 2025, 7, e70045. [Google Scholar] [CrossRef]
  21. Monjo-Henry, I.; Nieto-Carvalhal, B.; Uyaguari, M.; García-Carazo, S.; Balsa, A.; de Miguel, E.; Miranda-Carús, M.E. Circulating PD-1hi CXCR5- and CXCR5+ CD4 T cells are elevated in patients with newly diagnosed giant cell arteritis, and predict relapse. Rheumatology 2025, 64, 3996–4004. [Google Scholar] [CrossRef]
  22. Régnier, P.; Le Joncour, A.; Maciejewski-Duval, A.; Darrasse-Jèze, G.; Dolladille, C.; Meijers, W.C.; Bastarache, L.; Fouret, P.; Bruneval, P.; Arbaretaz, F.; et al. CTLA-4 Pathway Is Instrumental in Giant Cell Arteritis. Circ. Res. 2023, 133, 298–312. [Google Scholar] [CrossRef]
  23. Sato, Y.; Tada, M.; Goronzy, J.J.; Weyand, C.M. Immune checkpoints in autoimmune vasculitis. Best. Pract. Res. Clin. Rheumatol. 2024, 38, 101943. [Google Scholar] [CrossRef] [PubMed]
  24. Ferrucci, L.; Harris, T.B.; Guralnik, J.M.; Tracy, R.P.; Corti, M.C.; Cohen, H.J.; Penninx, B.; Pahor, M.; Wallace, R.; Havlik, R.J. Serum IL-6 level and the development of disability in older persons. J. Am. Geriatr. Soc. 1999, 47, 639–646. [Google Scholar] [CrossRef] [PubMed]
  25. Harris, T.B.; Ferrucci, L.; Tracy, R.P.; Corti, M.C.; Wacholder, S.; Ettinger, W.H., Jr.; Heimovitz, H.; Cohen, H.J.; Wallace, R. Associations of elevated interleukin-6 and C-reactive protein levels with mortality in the elderly. Am. J. Med. 1999, 106, 506–512. [Google Scholar] [CrossRef] [PubMed]
  26. Jylhä, M.; Paavilainen, P.; Lehtimäki, T.; Goebeler, S.; Karhunen, P.J.; Hervonen, A.; Hurme, M. Interleukin-1 receptor antagonist, interleukin-6, and C-reactive protein as predictors of mortality in nonagenarians: The vitality 90+ study. J. Gerontol. A Biol. Sci. Med. Sci. 2007, 62, 1016–1021. [Google Scholar] [CrossRef] [PubMed]
  27. Bruunsgaard, H.; Ladelund, S.; Pedersen, A.N.; Schroll, M.; Jørgensen, T.; Pedersen, B.K. Predicting death from tumour necrosis factor-alpha and interleukin-6 in 80-year-old people. Clin. Exp. Immunol. 2003, 132, 24–31. [Google Scholar] [CrossRef]
  28. Bruunsgaard, H.; Andersen-Ranberg, K.; Hjelmborg, J.; Pedersen, B.K.; Jeune, B. Elevated levels of tumor necrosis factor alpha and mortality in centenarians. Am. J. Med. 2003, 115, 278–283. [Google Scholar] [CrossRef]
  29. Grants, J.M.; Wegrzyn, J.; Hui, T.; O’Neill, K.; Shadbolt, M.; Knapp, D.; Parker, J.; Deng, Y.; Gopal, A.; Docking, T.R.; et al. Altered microRNA expression links IL6 and TNF-induced inflammaging with myeloid malignancy in humans and mice. Blood 2020, 135, 2235–2251. [Google Scholar] [CrossRef]
  30. Campisi, J.; d’Adda di Fagagna, F. Cellular senescence: When bad things happen to good cells. Nat. Rev. Mol. Cell Biol. 2007, 8, 729–740. [Google Scholar] [CrossRef]
  31. Su, T.Y.; Hauenstein, J.; Somuncular, E.; Dumral, Ö.; Leonard, E.; Gustafsson, C.; Tzortzis, E.; Forlani, A.; Johansson, A.S.; Qian, H.; et al. Aging is associated with functional and molecular changes in distinct hematopoietic stem cell subsets. Nat. Commun. 2024, 15, 7966. [Google Scholar] [CrossRef] [PubMed]
  32. Belizaire, R.; Wong, W.J.; Robinette, M.L.; Ebert, B.L. Clonal haematopoiesis and dysregulation of the immune system. Nat. Rev. Immunol. 2023, 23, 595–610. [Google Scholar] [CrossRef]
  33. Liberale, L.; Badimon, L.; Montecucco, F.; Lüscher, T.F.; Libby, P.; Camici, G.G. Inflammation, Aging, and Cardiovascular Disease: JACC Review Topic of the Week. J. Am. Coll. Cardiol. 2022, 79, 837–847. [Google Scholar] [CrossRef]
  34. Patricia, H.; Sharon, C.; Robert, H.; David, K.; Richard, C. The role of immune aging in Giant Cell Arteritis. Aging Pathobiology and Therapeutics 2023, 5, 92–100. [Google Scholar] [CrossRef]
  35. Zhang, H.; Weyand, C.M.; Goronzy, J.J. Hallmarks of the aging T-cell system. Febs J. 2021, 288, 7123–7142. [Google Scholar] [CrossRef]
  36. Berger, S.L.; Kouzarides, T.; Shiekhattar, R.; Shilatifard, A. An operational definition of epigenetics. Genes. Dev. 2009, 23, 781–783. [Google Scholar] [CrossRef] [PubMed]
  37. Coit, P.; De Lott, L.B.; Nan, B.; Elner, V.M.; Sawalha, A.H. DNA methylation analysis of the temporal artery microenvironment in giant cell arteritis. Ann. Rheum. Dis. 2016, 75, 1196–1202. [Google Scholar] [CrossRef]
  38. Croci, S.; Zerbini, A.; Boiardi, L.; Muratore, F.; Bisagni, A.; Nicoli, D.; Farnetti, E.; Pazzola, G.; Cimino, L.; Moramarco, A.; et al. MicroRNA markers of inflammation and remodelling in temporal arteries from patients with giant cell arteritis. Ann. Rheum. Dis. 2016, 75, 1527–1533. [Google Scholar] [CrossRef]
  39. Bolha, L.; Hočevar, A.; Suljič, A.; Jurčić, V. Inflammatory Cell Composition and Immune-Related microRNA Signature of Temporal Artery Biopsies From Patients With Giant Cell Arteritis. Front. Immunol. 2021, 12, 791099. [Google Scholar] [CrossRef] [PubMed]
  40. Legido, A.; Sarría, A.; Bueno, M.; Garagorri, J.; Fleta, J.; Abós, M.D.; Pérez-González, J. Body fat distribution and hyperinsulinemia in childhood. Am. J. Clin. Nutr. 1988, 48, 686–687. [Google Scholar] [CrossRef]
  41. Bolha, L.; Hočevar, A.; Jurčić, V. Current state of epigenetics in giant cell arteritis: Focus on microRNA dysregulation. Autoimmun. Rev. 2025, 24, 103739. [Google Scholar] [CrossRef]
  42. Steensma, D.P.; Bejar, R.; Jaiswal, S.; Lindsley, R.C.; Sekeres, M.A.; Hasserjian, R.P.; Ebert, B.L. Clonal hematopoiesis of indeterminate potential and its distinction from myelodysplastic syndromes. Blood 2015, 126, 9–16. [Google Scholar] [CrossRef] [PubMed]
  43. Cook, E.K.; Luo, M.; Rauh, M.J. Clonal hematopoiesis and inflammation: Partners in leukemogenesis and comorbidity. Exp. Hematol. 2020, 83, 85–94. [Google Scholar] [CrossRef]
  44. Aman, Y. Clonal hematopoiesis driven by microbial metabolite. Nat. Aging 2025, 5, 957. [Google Scholar] [CrossRef]
  45. Agarwal, P.; Sampson, A.; Hueneman, K.; Choi, K.; Jakobsen, N.A.; Uible, E.; Ishikawa, C.; Yeung, J.; Bolanos, L.; Zhao, X.; et al. Microbial metabolite drives ageing-related clonal haematopoiesis via ALPK1. Nature 2025, 642, 201–211. [Google Scholar] [CrossRef]
  46. Zeng, S.; Mu, D.; Wang, S. Fighting aging-associated clonal hematopoiesis with microbial metabolite. Trends Mol. Med. 2025, 31, 685–687. [Google Scholar] [CrossRef] [PubMed]
  47. Qu, F.; Park, S.; Martinez, K.; Gray, J.L.; Thowfeik, F.S.; Lundeen, J.A.; Kuhn, A.E.; Charboneau, D.J.; Gerlach, D.L.; Lockart, M.M.; et al. Ruthenium Complexes are pH-Activated Metallo Prodrugs (pHAMPs) with Light-Triggered Selective Toxicity Toward Cancer Cells. Inorg. Chem. 2017, 56, 7519–7532. [Google Scholar] [CrossRef] [PubMed]
  48. Tyrrell, D.J.; Goldstein, D.R. Ageing and atherosclerosis: Vascular intrinsic and extrinsic factors and potential role of IL-6. Nat. Rev. Cardiol. 2021, 18, 58–68. [Google Scholar] [CrossRef]
  49. Marnell, C.S.; Bick, A.; Natarajan, P. Clonal hematopoiesis of indeterminate potential (CHIP): Linking somatic mutations, hematopoiesis, chronic inflammation and cardiovascular disease. J. Mol. Cell Cardiol. 2021, 161, 98–105. [Google Scholar] [CrossRef] [PubMed]
  50. Jaiswal, S.; Natarajan, P.; Silver, A.J.; Gibson, C.J.; Bick, A.G.; Shvartz, E.; McConkey, M.; Gupta, N.; Gabriel, S.; Ardissino, D.; et al. Clonal Hematopoiesis and Risk of Atherosclerotic Cardiovascular Disease. N. Engl. J. Med. 2017, 377, 111–121. [Google Scholar] [CrossRef]
  51. von Bonin, M.; Jambor, H.K.; Teipel, R.; Stölzel, F.; Thiede, C.; Damm, F.; Kroschinsky, F.; Schetelig, J.; Chavakis, T.; Bornhäuser, M. Clonal hematopoiesis and its emerging effects on cellular therapies. Leukemia 2021, 35, 2752–2758. [Google Scholar] [CrossRef]
  52. Pich, O.; Bernard, E.; Zagorulya, M.; Rowan, A.; Pospori, C.; Slama, R.; Huerga Encabo, H.; O’Sullivan, J.; Papazoglou, D.; Anastasiou, P.; et al. Tumor-Infiltrating Clonal Hematopoiesis. N. Engl. J. Med. 2025, 392, 1594–1608. [Google Scholar] [CrossRef]
  53. Weeks, L.D.; Ebert, B.L. Clonal Hematopoiesis as a Driver of Solid Tumors. N. Engl. J. Med. 2025, 392, 1654–1656. [Google Scholar] [CrossRef]
  54. Kim, P.G.; Niroula, A.; Shkolnik, V.; McConkey, M.; Lin, A.E.; Słabicki, M.; Kemp, J.P.; Bick, A.; Gibson, C.J.; Griffin, G.; et al. Dnmt3a-mutated clonal hematopoiesis promotes osteoporosis. J. Exp. Med. 2021, 218, e20211872. [Google Scholar] [CrossRef] [PubMed]
  55. Wang, H.; Divaris, K.; Pan, B.; Li, X.; Lim, J.H.; Saha, G.; Barovic, M.; Giannakou, D.; Korostoff, J.M.; Bing, Y.; et al. Clonal hematopoiesis driven by mutated DNMT3A promotes inflammatory bone loss. Cell 2024, 187, 3690–3711.e3619. [Google Scholar] [CrossRef]
  56. Torreggiani, S.; Castellan, F.S.; Aksentijevich, I.; Beck, D.B. Somatic mutations in autoinflammatory and autoimmune disease. Nat. Rev. Rheumatol. 2024, 20, 683–698. [Google Scholar] [CrossRef]
  57. Gutierrez-Rodrigues, F.; Kusne, Y.; Fernandez, J.; Lasho, T.; Shalhoub, R.; Ma, X.; Alessi, H.; Finke, C.; Koster, M.J.; Mangaonkar, A.; et al. Spectrum of clonal hematopoiesis in VEXAS syndrome. Blood 2023, 142, 244–259. [Google Scholar] [CrossRef]
  58. Salzbrunn, J.B.; van Zeventer, I.A.; de Graaf, A.O.; Kamphuis, P.; van Bergen, M.; van Sleen, Y.; van der Reijden, B.A.; Schuringa, J.J.; Brouwer, E.; Diepstra, A.; et al. Clonal haematopoiesis and UBA1 mutations in individuals with biopsy-proven giant cell arteritis and population-based controls. Rheumatology 2024, 63, e45–e47. [Google Scholar] [CrossRef]
  59. Watanabe, R.; Kiji, M.; Hashimoto, M. Vasculitis associated with VEXAS syndrome: A literature review. Front. Med. 2022, 9, 983939. [Google Scholar] [CrossRef] [PubMed]
  60. Guedon, A.F.; Ouafdi, A.; Belfeki, N.; Dellal, A.; Ghriss, N.; Scheen, M.; Haidar, F.; Espitia, O.; Scoazec, J.Y.; Fain, O.; et al. Higher risk profile among patients with TET2-mutated giant cell arteritis: A cluster analysis. RMD Open 2024, 10, e004694. [Google Scholar] [CrossRef] [PubMed]
  61. Gutierrez-Rodrigues, F.; Wells, K.V.; Jones, A.I.; Hironaka, D.; Rankin, C.; Gadina, M.; Sikora, K.A.; Alemu, L.; Calado, R.T.; Quinn, K.A.; et al. Clonal haematopoiesis across the age spectrum of vasculitis patients with Takayasu’s arteritis, ANCA-associated vasculitis and giant cell arteritis. Ann. Rheum. Dis. 2024, 83, 508–517. [Google Scholar] [CrossRef]
  62. Bucala, R.; Tsao, B.P. The Emerging Spectrum of Somatic Mutation in Rheumatic Disease: Clonal Hematopoiesis Connects Aging With Giant Cell Arteritis. Arthritis Rheumatol. 2024, 76, 351–353. [Google Scholar] [CrossRef]
  63. Robinette, M.L.; Weeks, L.D.; Kramer, R.J.; Agrawal, M.; Gibson, C.J.; Yu, Z.; Sekar, A.; Mehta, A.; Niroula, A.; Brown, J.T.; et al. Association of Somatic TET2 Mutations With Giant Cell Arteritis. Arthritis Rheumatol. 2024, 76, 438–443. [Google Scholar] [CrossRef]
  64. Greigert, H.; Mounier, M.; Arnould, L.; Creuzot-Garcher, C.; Ramon, A.; Martin, L.; Tarris, G.; Ponnelle, T.; Audia, S.; Bonnotte, B.; et al. Heamatological malignancies in giant cell arteritis: A French population-based study. Rheumatology 2021, 60, 5408–5412. [Google Scholar] [CrossRef] [PubMed]
  65. Papo, M.; Friedrich, C.; Delaval, L.; Boysson, H.; Viallard, J.F.; Bachmeyer, C.; Sené, T.; Humbert, S.; Duffau, P.; Contis, A.; et al. Myeloproliferative neoplasms and clonal haematopoiesis in patients with giant cell arteritis: A case-control and exploratory study. Rheumatology 2022, 61, 775–780. [Google Scholar] [CrossRef] [PubMed]
  66. Salman, B.; Ido, N.-M.; Rajesh, D.; Fernando, L.S.; Mehran, B.; Andrea, A.; Tracy, M.; Scott, V.B.; Steven, M.C.; Mark, D.M.; et al. Blood-Based Epigenetic Instability Linked to Human Aging and Disease. bioRxiv 2025. [Google Scholar] [CrossRef]
  67. Panda, A.; Qian, F.; Mohanty, S.; van Duin, D.; Newman, F.K.; Zhang, L.; Chen, S.; Towle, V.; Belshe, R.B.; Fikrig, E.; et al. Age-associated decrease in TLR function in primary human dendritic cells predicts influenza vaccine response. J. Immunol. 2010, 184, 2518–2527. [Google Scholar] [CrossRef] [PubMed]
  68. Watanabe, R.; Hashimoto, M. Vasculitogenic T Cells in Large Vessel Vasculitis. Front. Immunol. 2022, 13, 923582. [Google Scholar] [CrossRef]
  69. Carrasco, E.; Gómez de Las Heras, M.M.; Gabandé-Rodríguez, E.; Desdín-Micó, G.; Aranda, J.F.; Mittelbrunn, M. The role of T cells in age-related diseases. Nat. Rev. Immunol. 2022, 22, 97–111. [Google Scholar] [CrossRef]
  70. Goronzy, J.J.; Weyand, C.M. Mechanisms underlying T cell ageing. Nat. Rev. Immunol. 2019, 19, 573–583. [Google Scholar] [CrossRef]
  71. Goronzy, J.J.; Weyand, C.M. Successful and Maladaptive T Cell Aging. Immunity 2017, 46, 364–378. [Google Scholar] [CrossRef]
  72. Mittelbrunn, M.; Kroemer, G. Hallmarks of T cell aging. Nat. Immunol. 2021, 22, 687–698. [Google Scholar] [CrossRef]
  73. Terrier, B.; Geri, G.; Chaara, W.; Allenbach, Y.; Rosenzwajg, M.; Costedoat-Chalumeau, N.; Fouret, P.; Musset, L.; Benveniste, O.; Six, A.; et al. Interleukin-21 modulates Th1 and Th17 responses in giant cell arteritis. Arthritis Rheum. 2012, 64, 2001–2011. [Google Scholar] [CrossRef] [PubMed]
  74. Jin, K.; Parreau, S.; Warrington, K.J.; Koster, M.J.; Berry, G.J.; Goronzy, J.J.; Weyand, C.M. Regulatory T Cells in Autoimmune Vasculitis. Front. Immunol. 2022, 13, 844300. [Google Scholar] [CrossRef] [PubMed]
  75. Adriawan, I.R.; Atschekzei, F.; Dittrich-Breiholz, O.; Garantziotis, P.; Hirsch, S.; Risser, L.M.; Kosanke, M.; Schmidt, R.E.; Witte, T.; Sogkas, G. Novel aspects of regulatory T cell dysfunction as a therapeutic target in giant cell arteritis. Ann. Rheum. Dis. 2022, 81, 124–131. [Google Scholar] [CrossRef]
  76. Jiemy, W.F.; van Sleen, Y.; Graver, J.C.; Pringle, S.; Brouwer, E.; van der Geest, K.S.M.; Cornec, D.; Boots, A.M.H.; Sandovici, M. Indication of Activated Senescence Pathways in the Temporal Arteries of Patients With Giant Cell Arteritis. Arthritis Rheumatol. 2023, 75, 1812–1818. [Google Scholar] [CrossRef] [PubMed]
  77. Chaib, S.; Tchkonia, T.; Kirkland, J.L. Cellular senescence and senolytics: The path to the clinic. Nat. Med. 2022, 28, 1556–1568. [Google Scholar] [CrossRef]
  78. Watanabe, R.; Hashimoto, M. Aging-Related Vascular Inflammation: Giant Cell Arteritis and Neurological Disorders. Front. Aging Neurosci. 2022, 14, 843305. [Google Scholar] [CrossRef]
  79. Veroutis, D.; Argyropoulou, O.D.; Goules, A.V.; Kambas, K.; Palamidas, D.A.; Evangelou, K.; Havaki, S.; Polyzou, A.; Valakos, D.; Xingi, E.; et al. Senescent cells in giant cell arteritis display an inflammatory phenotype participating in tissue injury via IL-6-dependent pathways. Ann. Rheum. Dis. 2024, 83, 342–350. [Google Scholar] [CrossRef]
  80. Bouzid, H.; Belk, J.A.; Jan, M.; Qi, Y.; Sarnowski, C.; Wirth, S.; Ma, L.; Chrostek, M.R.; Ahmad, H.; Nachun, D.; et al. Clonal hematopoiesis is associated with protection from Alzheimer’s disease. Nat. Med. 2023, 29, 1662–1670. [Google Scholar] [CrossRef]
  81. Sánchez Vela, P.; Trowbridge, J.J.; Levine, R.L. Clonal hematopoiesis, aging and Alzheimer’s disease. Nat. Med. 2023, 29, 1605–1606. [Google Scholar] [CrossRef]
  82. López-de-Mesa-Aragón, J.; Silva-Buriticá, C.; Salazar-Londoño, S. Clonal hematopoiesis as the intersection between genetics and resilience in Alzheimer’s disease. Npj Dementia 2025, 1, 11. [Google Scholar] [CrossRef]
  83. Zhao, H.; Liu, Z.; Chen, H.; Han, M.; Zhang, M.; Liu, K.; Jin, H.; Liu, X.; Shi, M.; Pu, W.; et al. Identifying specific functional roles for senescence across cell types. Cell 2024, 187, 7314–7334.e7321. [Google Scholar] [CrossRef] [PubMed]
  84. de Magalhães, J.P. Cellular senescence in normal physiology. Science 2024, 384, 1300–1301. [Google Scholar] [CrossRef] [PubMed]
  85. Carmona, E.G.; Callejas-Rubio, J.L.; Raya, E.; Ríos-Fernández, R.; Villanueva-Martín, G.; Cid, M.C.; Hernández-Rodríguez, J.; Ballestar, E.; Timmermann, B.; Ortego-Centeno, N.; et al. Single-cell transcriptomic profiling reveals a pathogenic role of cytotoxic CD4(+) T cells in giant cell arteritis. J. Autoimmun. 2024, 142, 103124. [Google Scholar] [CrossRef] [PubMed]
  86. Reitsema, R.D.; van der Geest, K.S.M.; Sandovici, M.; Jiemy, W.F.; Graver, J.C.; Abdulahad, W.H.; Boots, A.M.H.; Heeringa, P.; Brouwer, E. Phenotypic, transcriptomic and functional profiling reveal reduced activation thresholds of CD8+ T cells in giant cell arteritis. Rheumatology 2022, 62, 417–427. [Google Scholar] [CrossRef]
  87. Ferrigno, I.; Bonacini, M.; Rossi, A.; Nicastro, M.; Muratore, F.; Boiardi, L.; Cavazza, A.; Bisagni, A.; Cimino, L.; Ghidini, A.; et al. Genes deregulated in giant cell arteritis by Nanostring nCounter gene expression profiling in temporal artery biopsies. RMD Open 2024, 10, e004600. [Google Scholar] [CrossRef]
  88. Parreau, S.; Molina, E.; Dumonteil, S.; Goulabchand, R.; Naves, T.; Bois, M.C.; Akil, H.; Terro, F.; Fauchais, A.L.; Liozon, E.; et al. Use of high-plex data provides novel insights into the temporal artery processes of giant cell arteritis. Front. Immunol. 2023, 14, 1237986. [Google Scholar] [CrossRef]
  89. Su, Y.; Jia, R.L.; Han, L.; Li, Z.G. Role of anti-nucleosome antibody in the diagnosis of systemic lupus erythematosus. Clin. Immunol. 2007, 122, 115–120. [Google Scholar] [CrossRef]
  90. Matsumoto, K.; Suzuki, K.; Magi, M.; Onishi, S.; Yoshida, H.; Takeshita, M.; Kuramoto, J.; Yazawa, M.; Kato, T.; Shimizu, H.; et al. Trans-omics landscape of systemic vasculitis identified matrix metalloproteinase 12 as a novel signature molecule. Rheumatology 2025, 64, 4766–4775. [Google Scholar] [CrossRef]
  91. Estupiñán-Moreno, E.; Ortiz-Fernández, L.; Li, T.; Hernández-Rodríguez, J.; Ciudad, L.; Andrés-León, E.; Terron-Camero, L.C.; Prieto-González, S.; Espígol-Frigolé, G.; Cid, M.C.; et al. Methylome and transcriptome profiling of giant cell arteritis monocytes reveals novel pathways involved in disease pathogenesis and molecular response to glucocorticoids. Ann. Rheum. Dis. 2022, 81, 1290–1300. [Google Scholar] [CrossRef]
  92. Wagner, A.D.; Wittkop, U.; Thalmann, J.; Willmen, T.; Gödecke, V.; Hodam, J.; Ronicke, S.; Zenke, M. Glucocorticoid Effects on Tissue Residing Immune Cells in Giant Cell Arteritis: Importance of GM-CSF. Front. Med. 2021, 8, 709404. [Google Scholar] [CrossRef]
  93. Maleszewski, J.J.; Younge, B.R.; Fritzlen, J.T.; Hunder, G.G.; Goronzy, J.J.; Warrington, K.J.; Weyand, C.M. Clinical and pathological evolution of giant cell arteritis: A prospective study of follow-up temporal artery biopsies in 40 treated patients. Mod. Pathol. 2017, 30, 788–796. [Google Scholar] [CrossRef] [PubMed]
  94. Henningson, H.; Hammar, B.; Mohammad, A.J. The use of intravenous methylprednisolone in giant cell arteritis: A population-based study. Rheumatology 2025, 64, 2083–2090. [Google Scholar] [CrossRef] [PubMed]
  95. Muratore, F.; Marvisi, C.; Castrignanò, P.; Croci, S.; Bonacini, M.; Boiardi, L.; Ricordi, C.; Galli, E.; Besutti, G.; Spaggiari, L.; et al. Effectiveness and safety of a 26-week taper regimen of glucocorticoid in GCA patients: Results from a prospective cohort study. Semin. Arthritis Rheum. 2024, 64, 152351. [Google Scholar] [CrossRef] [PubMed]
  96. Ford, J.A.; Gewurz, D.; Gewurz-Singer, O. Tocilizumab in giant cell arteritis: An update for the clinician. Curr. Opin. Rheumatol. 2023, 35, 135–140. [Google Scholar] [CrossRef]
  97. Harigai, M.; Miyamae, T.; Hashimoto, H.; Umetsu, K.; Yamashita, K.; Nakaoka, Y. A multicentre, large-scale, observational study of tocilizumab in patients with giant cell arteritis in Japan. Mod. Rheumatol. 2024, 34, 775–783. [Google Scholar] [CrossRef]
  98. Unizony, S.; McCulley, T.J.; Spiera, R.; Pei, J.; Sidiropoulos, P.N.; Best, J.H.; Birchwood, C.; Pavlov, A.; Stone, J.H. Clinical outcomes of patients with giant cell arteritis treated with tocilizumab in real-world clinical practice: Decreased incidence of new visual manifestations. Arthritis Res. Ther. 2021, 23, 8. [Google Scholar] [CrossRef]
  99. Ito, T.; Fukui, S.; Nagase, F.N.; Yamaguchi, T.; Oda, N.; Inokuchi, H.; Suda, M.; Takizawa, N.; Suyama, Y.; Rokutanda, R.; et al. A feasible treatment strategy for tapering subcutaneous tocilizumab in giant cell arteritis: A 24-month multi-center retrospective study. Rheumatol. Int. 2025, 45, 45. [Google Scholar] [CrossRef]
  100. Moreel, L.; Betrains, A.; Molenberghs, G.; Vanderschueren, S.; Blockmans, D. Epidemiology and predictors of relapse in giant cell arteritis: A systematic review and meta-analysis. Jt. Bone Spine 2023, 90, 105494. [Google Scholar] [CrossRef]
  101. Ricordi, C.; Marvisi, C.; Macchioni, P.; Boiardi, L.; Cavazza, A.; Croci, S.; Bonacini, M.; Malchiodi, G.; Durmo, R.; Versari, A.; et al. Does tocilizumab eliminate inflammation in GCA? A cohort study on repeated temporal artery biopsies. RMD Open 2024, 10, e005132. [Google Scholar] [CrossRef]
  102. Loricera, J.; Tofade, T.; Prieto-Peña, D.; Romero-Yuste, S.; de Miguel, E.; Riveros-Frutos, A.; Ferraz-Amaro, I.; Labrador, E.; Maiz, O.; Becerra, E.; et al. Effectiveness of janus kinase inhibitors in relapsing giant cell arteritis in real-world clinical practice and review of the literature. Arthritis Res. Ther. 2024, 26, 116. [Google Scholar] [CrossRef]
  103. Blockmans, D.; Penn, S.K.; Setty, A.R.; Schmidt, W.A.; Rubbert-Roth, A.; Hauge, E.M.; Keen, H.I.; Ishii, T.; Khalidi, N.; Dejaco, C.; et al. A Phase 3 Trial of Upadacitinib for Giant-Cell Arteritis. N. Engl. J. Med. 2025, 392, 2013–2024. [Google Scholar] [CrossRef]
  104. Nash, P.; Kerschbaumer, A.; Konzett, V.; Aletaha, D.; Dörner, T.; Fleischmann, R.; McInnes, I.; Primdahl, J.; Sattar, N.; Tanaka, Y.; et al. Expert consensus statement on the treatment of immune-mediated inflammatory diseases with Janus kinase inhibitors: 2024 update. Ann. Rheum. Dis. 2025, 84, 664–679. [Google Scholar] [CrossRef] [PubMed]
  105. Corbera-Bellalta, M.; Alba-Rovira, R.; Muralidharan, S.; Espígol-Frigolé, G.; Ríos-Garcés, R.; Marco-Hernández, J.; Denuc, A.; Kamberovic, F.; Pérez-Galán, P.; Joseph, A.; et al. Blocking GM-CSF receptor α with mavrilimumab reduces infiltrating cells, pro-inflammatory markers and neoangiogenesis in ex vivo cultured arteries from patients with giant cell arteritis. Ann. Rheum. Dis. 2022, 81, 524–536. [Google Scholar] [CrossRef]
  106. Cid, M.C.; Unizony, S.H.; Blockmans, D.; Brouwer, E.; Dagna, L.; Dasgupta, B.; Hellmich, B.; Molloy, E.; Salvarani, C.; Trapnell, B.C.; et al. Efficacy and safety of mavrilimumab in giant cell arteritis: A phase 2, randomised, double-blind, placebo-controlled trial. Ann. Rheum. Dis. 2022, 81, 653–661. [Google Scholar] [CrossRef] [PubMed]
  107. Langford, C.A.; Cuthbertson, D.; Ytterberg, S.R.; Khalidi, N.; Monach, P.A.; Carette, S.; Seo, P.; Moreland, L.W.; Weisman, M.; Koening, C.L.; et al. A Randomized, Double-Blind Trial of Abatacept (CTLA-4Ig) for the Treatment of Giant Cell Arteritis. Arthritis Rheumatol. 2017, 69, 837–845. [Google Scholar] [CrossRef] [PubMed]
  108. Thiel, J. Giant cell arteritis-New treatment targets at the horizon. Semin. Arthritis Rheum. 2025, 72s, 152686. [Google Scholar] [CrossRef]
  109. Hur, B.; Koster, M.J.; Jang, J.S.; Weyand, C.M.; Warrington, K.J.; Sung, J. Global Transcriptomic Profiling Identifies Differential Gene Expression Signatures Between Inflammatory and Noninflammatory Aortic Aneurysms. Arthritis Rheumatol. 2022, 74, 1376–1386. [Google Scholar] [CrossRef]
  110. Zander, R.; Kasmani, M.Y.; Chen, Y.; Topchyan, P.; Shen, J.; Zheng, S.; Burns, R.; Ingram, J.; Cui, C.; Joshi, N.; et al. Tfh-cell-derived interleukin 21 sustains effector CD8(+) T cell responses during chronic viral infection. Immunity 2022, 55, 475–493.e475. [Google Scholar] [CrossRef]
  111. Liu, Y.W.; Fu, S.H.; Chien, M.W.; Hsu, C.Y.; Lin, M.H.; Dong, J.L.; Lu, R.J.; Lee, Y.J.; Chen, P.Y.; Wang, C.H.; et al. Blimp-1 molds the epigenetic architecture of IL-21-mediated autoimmune diseases through an autoregulatory circuit. JCI Insight 2022, 7, e151614. [Google Scholar] [CrossRef] [PubMed]
  112. Hernández-Rodríguez, J.; Segarra, M.; Vilardell, C.; Sánchez, M.; García-Martínez, A.; Esteban, M.J.; Queralt, C.; Grau, J.M.; Urbano-Márquez, A.; Palacín, A.; et al. Tissue production of pro-inflammatory cytokines (IL-1beta, TNFalpha and IL-6) correlates with the intensity of the systemic inflammatory response and with corticosteroid requirements in giant-cell arteritis. Rheumatology 2004, 43, 294–301. [Google Scholar] [CrossRef] [PubMed]
  113. Watanabe, R.; Maeda, T.; Zhang, H.; Berry, G.J.; Zeisbrich, M.; Brockett, R.; Greenstein, A.E.; Tian, L.; Goronzy, J.J.; Weyand, C.M. MMP (Matrix Metalloprotease)-9-Producing Monocytes Enable T Cells to Invade the Vessel Wall and Cause Vasculitis. Circ. Res. 2018, 123, 700–715. [Google Scholar] [CrossRef]
  114. Watanabe, R.; Hashimoto, M. Pathogenic role of monocytes/macrophages in large vessel vasculitis. Front. Immunol. 2022, 13, 859502. [Google Scholar] [CrossRef]
  115. Weinberger, T.; Esfandyari, D.; Messerer, D.; Percin, G.; Schleifer, C.; Thaler, R.; Liu, L.; Stremmel, C.; Schneider, V.; Vagnozzi, R.J.; et al. Ontogeny of arterial macrophages defines their functions in homeostasis and inflammation. Nat. Commun. 2020, 11, 4549. [Google Scholar] [CrossRef]
  116. Stock, A.T.; Parsons, S.; Sharma, V.J.; James, F.; Starkey, G.; D’Costa, R.; Gordon, C.L.; Wicks, I.P. Intimal macrophages develop from circulating monocytes during vasculitis. Clin. Transl. Immunol. 2022, 11, e1412. [Google Scholar] [CrossRef]
  117. Jiemy, W.F.; van Sleen, Y.; van der Geest, K.S.; Ten Berge, H.A.; Abdulahad, W.H.; Sandovici, M.; Boots, A.M.; Heeringa, P.; Brouwer, E. Distinct macrophage phenotypes skewed by local granulocyte macrophage colony-stimulating factor (GM-CSF) and macrophage colony-stimulating factor (M-CSF) are associated with tissue destruction and intimal hyperplasia in giant cell arteritis. Clin. Transl. Immunol. 2020, 9, e1164. [Google Scholar] [CrossRef]
  118. Watanabe, N.; Hara, Y.; Nishito, Y.; Kounoe, M.; Sekiyama, K.; Takamasu, E.; Kise, T.; Chinen, N.; Shimada, K.; Sugihara, M.; et al. Tissue degrading and remodelling molecules in giant cell arteritis. Rheumatology 2025, 64, 3095–3103. [Google Scholar] [CrossRef]
  119. Zhang, H.; Watanabe, R.; Berry, G.J.; Tian, L.; Goronzy, J.J.; Weyand, C.M. Inhibition of JAK-STAT Signaling Suppresses Pathogenic Immune Responses in Medium and Large Vessel Vasculitis. Circulation 2018, 137, 1934–1948. [Google Scholar] [CrossRef]
  120. Zhang, H.; Watanabe, R.; Berry, G.J.; Nadler, S.G.; Goronzy, J.J.; Weyand, C.M. CD28 Signaling Controls Metabolic Fitness of Pathogenic T Cells in Medium and Large Vessel Vasculitis. J. Am. Coll. Cardiol. 2019, 73, 1811–1823. [Google Scholar] [CrossRef] [PubMed]
  121. Watanabe, R.; Hashimoto, M. Perspectives of JAK Inhibitors for Large Vessel Vasculitis. Front. Immunol. 2022, 13, 881705. [Google Scholar] [CrossRef]
  122. Yu, Q.; Wu, Y.; Ma, X.; Zhang, Y. Causal genes identification of giant cell arteritis in CD4+ Memory t cells: An integration of multi-omics and expression quantitative trait locus analysis. Inflamm. Res. 2025, 74, 3. [Google Scholar] [CrossRef] [PubMed]
  123. Sato, Y.; Jain, A.; Ohtsuki, S.; Okuyama, H.; Sturmlechner, I.; Takashima, Y.; Le, K.C.; Bois, M.C.; Berry, G.J.; Warrington, K.J.; et al. Stem-like CD4(+) T cells in perivascular tertiary lymphoid structures sustain autoimmune vasculitis. Sci. Transl. Med. 2023, 15, eadh0380. [Google Scholar] [CrossRef] [PubMed]
  124. Graver, J.C.; Jiemy, W.F.; Altulea, D.H.A.; van Sleen, Y.; Xu, S.; van der Geest, K.S.M.; Verstappen, G.; Heeringa, P.; Abdulahad, W.H.; Brouwer, E.; et al. Cytokine producing B-cells and their capability to polarize macrophages in giant cell arteritis. J. Autoimmun. 2023, 140, 103111. [Google Scholar] [CrossRef] [PubMed]
  125. Pesce, E.; Bombaci, M.; Croci, S.; Bonacini, M.; Marvisi, C.; Ricordi, C.; Monti, S.; Muratore, F.; Abrignani, S.; Caporali, R.; et al. Identification of two autoantigens recognised by circulating autoantibodies as potential biomarkers for diagnosing giant cell arteritis. Clin. Exp. Rheumatol. 2024, 42, 1317–1320. [Google Scholar] [CrossRef]
  126. Lozano, E.; Segarra, M.; García-Martínez, A.; Hernández-Rodríguez, J.; Cid, M.C. Imatinib mesylate inhibits in vitro and ex vivo biological responses related to vascular occlusion in giant cell arteritis. Ann. Rheum. Dis. 2008, 67, 1581–1588. [Google Scholar] [CrossRef]
  127. Tai, Y.; Woods, E.L.; Dally, J.; Kong, D.; Steadman, R.; Moseley, R.; Midgley, A.C. Myofibroblasts: Function, Formation, and Scope of Molecular Therapies for Skin Fibrosis. Biomolecules 2021, 11, 1095. [Google Scholar] [CrossRef]
  128. Greigert, H.; Genet, C.; Ramon, A.; Bonnotte, B.; Samson, M. New Insights into the Pathogenesis of Giant Cell Arteritis: Mechanisms Involved in Maintaining Vascular Inflammation. J. Clin. Med. 2022, 11, 2905. [Google Scholar] [CrossRef]
  129. Weyand, C.M.; Goronzy, J.J. Immune mechanisms in medium and large-vessel vasculitis. Nat. Rev. Rheumatol. 2013, 9, 731–740. [Google Scholar] [CrossRef]
  130. Gibb, A.A.; Lazaropoulos, M.P.; Elrod, J.W. Myofibroblasts and Fibrosis: Mitochondrial and Metabolic Control of Cellular Differentiation. Circ. Res. 2020, 127, 427–447. [Google Scholar] [CrossRef]
  131. Xu, S.; Jiemy, W.F.; Brouwer, E.; Burgess, J.K.; Heeringa, P.; van der Geest, K.S.M.; Alba-Rovira, R.; Corbera-Bellalta, M.; Boots, A.H.; Cid, M.C.; et al. Current evidence on the role of fibroblasts in large-vessel vasculitides: From pathogenesis to therapeutics. Autoimmun. Rev. 2024, 23, 103574. [Google Scholar] [CrossRef]
  132. Parreau, S.; Vedrenne, N.; Regent, A.; Richard, L.; Sindou, P.; Mouthon, L.; Fauchais, A.L.; Jauberteau, M.O.; Ly, K.H. An immunohistochemical analysis of fibroblasts in giant cell arteritis. Ann. Diagn. Pathol. 2021, 52, 151728. [Google Scholar] [CrossRef] [PubMed]
  133. Greigert, H.; Ramon, A.; Genet, C.; Cladière, C.; Gerard, C.; Cuidad, M.; Corbera-Bellalta, M.; Alba-Rovira, R.; Arnould, L.; Creuzot-Garcher, C.; et al. Neointimal myofibroblasts contribute to maintaining Th1/Tc1 and Th17/Tc17 inflammation in giant cell arteritis. J. Autoimmun. 2024, 142, 103151. [Google Scholar] [CrossRef]
  134. Wang, L.; Luqmani, R.; Udalova, I.A. The role of neutrophils in rheumatic disease-associated vascular inflammation. Nat. Rev. Rheumatol. 2022, 18, 158–170. [Google Scholar] [CrossRef]
  135. Wang, L.; Ai, Z.; Khoyratty, T.; Zec, K.; Eames, H.L.; van Grinsven, E.; Hudak, A.; Morris, S.; Ahern, D.; Monaco, C.; et al. ROS-producing immature neutrophils in giant cell arteritis are linked to vascular pathologies. JCI Insight 2020, 5, e139163. [Google Scholar] [CrossRef]
  136. Palamidas, D.A.; Argyropoulou, O.D.; Georgantzoglou, N.; Karatza, E.; Xingi, E.; Kapsogeorgou, E.K.; Anagnostopoulos, C.D.; Lazaris, A.C.; Ritis, K.; Goules, A.V.; et al. Neutrophil extracellular traps in giant cell arteritis biopsies: Presentation, localization and co-expression with inflammatory cytokines. Rheumatology 2022, 61, 1639–1644. [Google Scholar] [CrossRef] [PubMed]
  137. Michailidou, D.; Duvvuri, B.; Kuley, R.; Cuthbertson, D.; Grayson, P.C.; Khalidi, N.A.; Koening, C.L.; Langford, C.A.; McAlear, C.A.; Moreland, L.W.; et al. Neutrophil activation in patients with anti-neutrophil cytoplasmic autoantibody-associated vasculitis and large-vessel vasculitis. Arthritis Res. Ther. 2022, 24, 160. [Google Scholar] [CrossRef] [PubMed]
  138. Bettiol, A.; Argento, F.R.; Fini, E.; Bello, F.; Di Scala, G.; Taddei, N.; Emmi, G.; Prisco, D.; Becatti, M.; Fiorillo, C. ROS-driven structural and functional fibrinogen modifications are reverted by interleukin-6 inhibition in Giant Cell Arteritis. Thromb. Res. 2023, 230, 1–10. [Google Scholar] [CrossRef]
Figure 1. Representative histopathological features of temporal artery specimens from patients with giant cell arteritis. (A) Typical giant cell arteritis. Inflammation mainly in the adventitia, extending to the media and the intima. Concentric intimal hyperplasia with luminal occlusion. Hematoxylin and eosin stain (H&E), ×40. (B) Higher-power view of A. Prominent inflammatory cell infiltrates in the adventitia, extending into the media, with relatively sparse infiltration in the hyperplastic intima. H&E, ×100. (C) Granulomatous inflammation. Macrophages (blue arrowheads) aligned at the media–intima junction with multinucleated giant cell (red arrow). Lymphocyte-predominant inflammatory infiltrates in the adventitia (red arrowhead). H&E, ×200. (D) Elastica van Gieson stain (EVG), low magnification, ×40. (E) Higher-power view of D. EVG, ×100. (F) Infiltration of multinucleated giant cells (red arrows) with phagocytosing elastic fibers (blue arrows). EVG, ×400. (G) Disruption of the internal elastic lamina (green arrows). EVG, ×200. (H) CD4+ T-cell infiltration from the adventitia into the media–intima junction. CD4 stain, ×40. (I) Minimal infiltration of CD8+ T cells. ×40.
Figure 1. Representative histopathological features of temporal artery specimens from patients with giant cell arteritis. (A) Typical giant cell arteritis. Inflammation mainly in the adventitia, extending to the media and the intima. Concentric intimal hyperplasia with luminal occlusion. Hematoxylin and eosin stain (H&E), ×40. (B) Higher-power view of A. Prominent inflammatory cell infiltrates in the adventitia, extending into the media, with relatively sparse infiltration in the hyperplastic intima. H&E, ×100. (C) Granulomatous inflammation. Macrophages (blue arrowheads) aligned at the media–intima junction with multinucleated giant cell (red arrow). Lymphocyte-predominant inflammatory infiltrates in the adventitia (red arrowhead). H&E, ×200. (D) Elastica van Gieson stain (EVG), low magnification, ×40. (E) Higher-power view of D. EVG, ×100. (F) Infiltration of multinucleated giant cells (red arrows) with phagocytosing elastic fibers (blue arrows). EVG, ×400. (G) Disruption of the internal elastic lamina (green arrows). EVG, ×200. (H) CD4+ T-cell infiltration from the adventitia into the media–intima junction. CD4 stain, ×40. (I) Minimal infiltration of CD8+ T cells. ×40.
Jcm 14 06350 g001
Figure 2. Inflammatory signaling pathways and vessel wall remodeling. PAMPs and DAMPs activate vasDCs in the adventitia through TLRs, leading to NF-κB activation and production of cytokines such as IL-12 and IL-23, as well as chemokines (CCL19, CCL21) that recruit monocytes and CD4+ T cells. Monocytes differentiate into macrophages, which contribute to granuloma formation and tissue damage. Activated vasDCs promote differentiation of naïve CD4+ T cells into Th1 and Th17 subsets via IL-12 and IL-23, respectively. Th1 cells secrete IFNγ and Th17 cells produce IL-17, while IL-6 further amplifies T-cell activation. IL-6, IL-12, IL-23, and IFNγ signal through the JAK–STAT pathway to sustain T-cell activation and chronic inflammation. Macrophages release VEGF, inducing neoangiogenesis, and MMPs, which disrupt the internal elastic lamina. These processes lead to VSMC migration and proliferation, endothelial cell dysfunction, intimal hyperplasia, and vascular remodeling. Abbreviations: CCL, chemokine-C motif ligand; DAMPs, damage-associated molecular patterns; IFN, interferon; IL, interleukin; MMP, matrix metalloproteinase; PAMPs, pathogen-associated molecular patterns; TLR, Toll-like receptor; vasDC, vascular dendritic cells; VEGF, vascular endothelial growth factor; VSMC, vascular smooth muscle cell.
Figure 2. Inflammatory signaling pathways and vessel wall remodeling. PAMPs and DAMPs activate vasDCs in the adventitia through TLRs, leading to NF-κB activation and production of cytokines such as IL-12 and IL-23, as well as chemokines (CCL19, CCL21) that recruit monocytes and CD4+ T cells. Monocytes differentiate into macrophages, which contribute to granuloma formation and tissue damage. Activated vasDCs promote differentiation of naïve CD4+ T cells into Th1 and Th17 subsets via IL-12 and IL-23, respectively. Th1 cells secrete IFNγ and Th17 cells produce IL-17, while IL-6 further amplifies T-cell activation. IL-6, IL-12, IL-23, and IFNγ signal through the JAK–STAT pathway to sustain T-cell activation and chronic inflammation. Macrophages release VEGF, inducing neoangiogenesis, and MMPs, which disrupt the internal elastic lamina. These processes lead to VSMC migration and proliferation, endothelial cell dysfunction, intimal hyperplasia, and vascular remodeling. Abbreviations: CCL, chemokine-C motif ligand; DAMPs, damage-associated molecular patterns; IFN, interferon; IL, interleukin; MMP, matrix metalloproteinase; PAMPs, pathogen-associated molecular patterns; TLR, Toll-like receptor; vasDC, vascular dendritic cells; VEGF, vascular endothelial growth factor; VSMC, vascular smooth muscle cell.
Jcm 14 06350 g002
Figure 3. Proposed mechanisms underlying the onset of giant cell arteritis through age-related immune dysregulation and inflammaging. The onset of GCA is driven by a complex interplay of systemic, genetic, epigenetic, and cellular factors, all of which collectively contribute to vascular tissue dysfunction. Systemic aging and gut dysbiosis increase intestinal permeability and disrupt microbial homeostasis, thereby promoting systemic inflammation. Genetic and epigenetic alterations—such as TET2 and DNMT3A mutations, DNA methylation instability, and microRNA dysregulation—further enhance immune activation. CHIP leads to the emergence of proinflammatory macrophages, while immunosenescence impairs T-cell function, Treg populations, and promotes effector T-cell responses. Senescent cells in vascular and stromal compartments—including endothelial cells, VSMC, and fibroblasts—are characterized by elevated expression of p21 and p16. These cells acquire SASP, producing proinflammatory cytokines, VEGF, and MMPs, which compromise tissue tolerance and vascular integrity. The convergence of these pathological processes may ultimately result in the clinical onset of GCA. Abbreviations: CHIP, Clonal hematopoiesis of indeterminate potential; MMPs, matrix metalloproteinases; SASP, Senescence-associated secretory phenotype; Treg, Regulatory T cell; VSMC, vascular smooth muscle cell.
Figure 3. Proposed mechanisms underlying the onset of giant cell arteritis through age-related immune dysregulation and inflammaging. The onset of GCA is driven by a complex interplay of systemic, genetic, epigenetic, and cellular factors, all of which collectively contribute to vascular tissue dysfunction. Systemic aging and gut dysbiosis increase intestinal permeability and disrupt microbial homeostasis, thereby promoting systemic inflammation. Genetic and epigenetic alterations—such as TET2 and DNMT3A mutations, DNA methylation instability, and microRNA dysregulation—further enhance immune activation. CHIP leads to the emergence of proinflammatory macrophages, while immunosenescence impairs T-cell function, Treg populations, and promotes effector T-cell responses. Senescent cells in vascular and stromal compartments—including endothelial cells, VSMC, and fibroblasts—are characterized by elevated expression of p21 and p16. These cells acquire SASP, producing proinflammatory cytokines, VEGF, and MMPs, which compromise tissue tolerance and vascular integrity. The convergence of these pathological processes may ultimately result in the clinical onset of GCA. Abbreviations: CHIP, Clonal hematopoiesis of indeterminate potential; MMPs, matrix metalloproteinases; SASP, Senescence-associated secretory phenotype; Treg, Regulatory T cell; VSMC, vascular smooth muscle cell.
Jcm 14 06350 g003
Figure 4. Cellular and molecular mechanisms underlying vascular inflammation in giant cell arteritis and potential therapeutic targets. This schematic illustrates the immunopathogenesis of GCA within the temporal artery, highlighting the spatial organization of immune cells and their interactions with vascular stromal components. CD4+ T cells become activated via antigen presentation by APCs in the adventitial vasa vasorum and differentiate into effector subsets, including Th1, Th17, Treg, and Trm. Key inflammatory pathways include the IL-12/23–Th1 axis, IL-6/IL-21–Th17 axis, and GM-CSF–macrophage axis, all of which contribute to vascular injury and intimal hyperplasia. Monocyte-derived macrophages express MMP-9 and interact with VSMCs and myofibroblasts, promoting structural remodeling and the production of ROS. The NOTCH1–JAGGED1–mTOR signaling cascade, as well as the JAK–STAT pathway, supports CD4+ T-cell survival and activation. Infiltrating neutrophils form NETs, further amplifying inflammation. Tertiary lymphoid structures containing TCF1+ CD4+ T cells, BCL6+ T follicular helper-like cells, and EOMES+ subsets are also observed. Multiple therapeutic targets are highlighted, including inhibitors of the mTOR, IL-12/23, IL-17, IL-21, GM-CSFR, CTLA-4, and JAK pathways. Abbreviations: APCs, antigen-presenting cells; BCL6, B-cell lymphoma6+ T follicular helper-like T cell; CTLA-4, cytotoxic T-lymphocyte antigen 4; EOMES, Eomesodermin+ cytotoxic T cell; GM-CSF, granulocyte–macrophage colony-stimulating factor; MMP, matrix metalloproteinase; NET, neutrophil extracellular trap; ROS, reactive oxygen species; Treg, Regulatory T cell; Trm, tissue-resident memory T cell; VSMC, vascular smooth muscle cell.
Figure 4. Cellular and molecular mechanisms underlying vascular inflammation in giant cell arteritis and potential therapeutic targets. This schematic illustrates the immunopathogenesis of GCA within the temporal artery, highlighting the spatial organization of immune cells and their interactions with vascular stromal components. CD4+ T cells become activated via antigen presentation by APCs in the adventitial vasa vasorum and differentiate into effector subsets, including Th1, Th17, Treg, and Trm. Key inflammatory pathways include the IL-12/23–Th1 axis, IL-6/IL-21–Th17 axis, and GM-CSF–macrophage axis, all of which contribute to vascular injury and intimal hyperplasia. Monocyte-derived macrophages express MMP-9 and interact with VSMCs and myofibroblasts, promoting structural remodeling and the production of ROS. The NOTCH1–JAGGED1–mTOR signaling cascade, as well as the JAK–STAT pathway, supports CD4+ T-cell survival and activation. Infiltrating neutrophils form NETs, further amplifying inflammation. Tertiary lymphoid structures containing TCF1+ CD4+ T cells, BCL6+ T follicular helper-like cells, and EOMES+ subsets are also observed. Multiple therapeutic targets are highlighted, including inhibitors of the mTOR, IL-12/23, IL-17, IL-21, GM-CSFR, CTLA-4, and JAK pathways. Abbreviations: APCs, antigen-presenting cells; BCL6, B-cell lymphoma6+ T follicular helper-like T cell; CTLA-4, cytotoxic T-lymphocyte antigen 4; EOMES, Eomesodermin+ cytotoxic T cell; GM-CSF, granulocyte–macrophage colony-stimulating factor; MMP, matrix metalloproteinase; NET, neutrophil extracellular trap; ROS, reactive oxygen species; Treg, Regulatory T cell; Trm, tissue-resident memory T cell; VSMC, vascular smooth muscle cell.
Jcm 14 06350 g004
Table 1. Summary of recent omics-based studies investigating the pathogenesis of giant cell arteritis. This table provides a summary of recent studies employing transcriptomic, proteomic, and spatial omics approaches to investigate the immunopathology of GCA. The columns present the year of publication, type and source of specimens (e.g., peripheral blood mononuclear cells [PBMCs], temporal artery biopsy [TAB], arterial tissue), sample sizes (N = number of cases; C = number of controls), analytical platforms used, key findings, and potential therapeutic targets. The final column provides PubMed IDs (PMIDs) for reference. * Publicly available datasets were used exclusively. Analysis was performed by integrating data from GSE157007 (PMID: 37117750) and GSE198891 (PMID: 35460236). Abbreviations: scRNA-seq, single-cell RNA sequencing; SV, systemic vasculitis; TCR, T-cell receptor; TLS, tertiary lymphoid structure; TMI, transmural inflammation; IL, interleukin; ROS, reactive oxygen species.
Table 1. Summary of recent omics-based studies investigating the pathogenesis of giant cell arteritis. This table provides a summary of recent studies employing transcriptomic, proteomic, and spatial omics approaches to investigate the immunopathology of GCA. The columns present the year of publication, type and source of specimens (e.g., peripheral blood mononuclear cells [PBMCs], temporal artery biopsy [TAB], arterial tissue), sample sizes (N = number of cases; C = number of controls), analytical platforms used, key findings, and potential therapeutic targets. The final column provides PubMed IDs (PMIDs) for reference. * Publicly available datasets were used exclusively. Analysis was performed by integrating data from GSE157007 (PMID: 37117750) and GSE198891 (PMID: 35460236). Abbreviations: scRNA-seq, single-cell RNA sequencing; SV, systemic vasculitis; TCR, T-cell receptor; TLS, tertiary lymphoid structure; TMI, transmural inflammation; IL, interleukin; ROS, reactive oxygen species.
YearSpecimenNCModality/PlatformKey Findings/Potential Therapeutic TargetPMID
2022GCA
PBMC
33scRNA-seq
/10x Chromium
Genes associated with antiviral defense and immune activation (KLRD1, IFITM1) were upregulated, whereas those involved in cytotoxicity and proliferation control (GNLY, ZFP36L2) were downregulated.35460236
2022GCA
Arterial tissue
3NonescRNA-seq, TCR repertoire
/10x Chromium
TCF1+IL7R+ stem-like CD4+ T cells expand clonally in TLSs, suggesting IL-7R+ CD4+ T cells as pathogenic and therapeutic targets.37672564
2023GCA
TAB
97Spatial transcriptomics
/NanoString GeoMx DSP
CD74 and macrophage-associated pathways—including MMP2, MMP9, and CXCR4—were identified as key upregulated targets in GCA, particularly within the intima and media layers.37744332
2024GCA
PBMC
88scRNA-seq, TCR repertoire
/10x Genomics Chromium
Clonally expanded cytotoxic CD4+ T cells in active GCA highly expressed GZMB and CCL5, suggesting Maraviroc as a potential therapy.37952293
2024GCA
TAB
497Gene expression profiling
/NanoString nCounter
In TMI lesions, genes encoding TNF superfamily members, immune checkpoints, chemokines and their receptors, toll-like receptors, complement components, Fc receptors for IgG, signaling lymphocytic activation molecules, as well as JAK3, STAT1, and STAT4 were upregulated.39317454
2024GCA
PBMC *
(a) 40
(b) 14
(a) 31
(b) 6
(a) Bulk RNA-seq
(b) scRNA-seq
/Illumina, 10x Chromium
Targeting DDIT4, a causal gene for GCA, may suppress persistent inflammation in CD4+ memory T cells.39762453
2024GCA
TAB
106Gene expression profiling
/Agilent microarray
MMP12, ACP5, and TREM2 are potential therapeutic targets in GCA, associated with macrophage-driven tissue destruction, while LRRC15 marks myofibroblasts contributing to intimal hyperplasia in granulomatous inflammation.39837478
2025SV
(c) Serum, whole blood,
tissue
332
(c) GCA2,
GPA1
30Proteome analysis,
RNA sequencing,
Spatial transcriptomics
/Olink Proteomics,
Illumina.
10x Genomics
MMP12 is selectively elevated in vasculitis and specifically expressed in CD206-positive macrophages and multinucleated giant cells within lesions. It reflects disease activity even under IL-6 blockade and helps predict relapse.40139687
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Shiomi, M.; Watanabe, R.; Ishihara, R.; Tanaka, S.; Kageyama, G.; Hashimoto, M. Uncovering the New Biology of Giant Cell Arteritis to Guide Therapeutic Strategies. J. Clin. Med. 2025, 14, 6350. https://doi.org/10.3390/jcm14186350

AMA Style

Shiomi M, Watanabe R, Ishihara R, Tanaka S, Kageyama G, Hashimoto M. Uncovering the New Biology of Giant Cell Arteritis to Guide Therapeutic Strategies. Journal of Clinical Medicine. 2025; 14(18):6350. https://doi.org/10.3390/jcm14186350

Chicago/Turabian Style

Shiomi, Mayu, Ryu Watanabe, Ryuhei Ishihara, Sayaka Tanaka, Goichi Kageyama, and Motomu Hashimoto. 2025. "Uncovering the New Biology of Giant Cell Arteritis to Guide Therapeutic Strategies" Journal of Clinical Medicine 14, no. 18: 6350. https://doi.org/10.3390/jcm14186350

APA Style

Shiomi, M., Watanabe, R., Ishihara, R., Tanaka, S., Kageyama, G., & Hashimoto, M. (2025). Uncovering the New Biology of Giant Cell Arteritis to Guide Therapeutic Strategies. Journal of Clinical Medicine, 14(18), 6350. https://doi.org/10.3390/jcm14186350

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

Article Metrics

Back to TopTop