Next Article in Journal
The Role of β-Dystroglycan in Nuclear Dynamics
Previous Article in Journal
Advanced Glycation End Products Upregulate CD40 in Human Retinal Endothelial and Müller Cells: Relevance to Diabetic Retinopathy
Previous Article in Special Issue
The Contribution of Innate Immunity in Large-Vessel Vasculitis: Detangling New Pathomechanisms beyond the Onset of Vascular Inflammation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Current Insights into Tissue Injury of Giant Cell Arteritis: From Acute Inflammatory Responses towards Inappropriate Tissue Remodeling

by
Dimitris Anastasios Palamidas
1,
Loukas Chatzis
1,2,
Maria Papadaki
2,
Ilias Gissis
3,
Konstantinos Kambas
4,
Evangelos Andreakos
2,
Andreas V. Goules
1,2 and
Athanasios G. Tzioufas
1,2,5,*
1
Department of Pathophysiology, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
2
Laboratory of Immunobiology, Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece
3
Department of Thoracic and Cardiovascular Surgery, Evangelismos General Hospital, 11473 Athens, Greece
4
Laboratory of Molecular Genetics, Department of Immunology, Hellenic Pasteur Institute, 11521 Athens, Greece
5
Research Institute for Systemic Autoimmune Diseases, 11527 Athens, Greece
*
Author to whom correspondence should be addressed.
Cells 2024, 13(5), 430; https://doi.org/10.3390/cells13050430
Submission received: 31 January 2024 / Revised: 25 February 2024 / Accepted: 27 February 2024 / Published: 29 February 2024
(This article belongs to the Special Issue Cellular and Molecular Mechanisms Underlying Large-Vessel Vasculitis)

Abstract

:
Giant cell arteritis (GCA) is an autoimmune disease affecting large vessels in patients over 50 years old. It is an exemplary model of a classic inflammatory disorder with IL-6 playing the leading role. The main comorbidities that may appear acutely or chronically are vascular occlusion leading to blindness and thoracic aorta aneurysm formation, respectively. The tissue inflammatory bulk is expressed as acute or chronic delayed-type hypersensitivity reactions, the latter being apparent by giant cell formation. The activated monocytes/macrophages are associated with pronounced Th1 and Th17 responses. B-cells and neutrophils also participate in the inflammatory lesion. However, the exact order of appearance and mechanistic interactions between cells are hindered by the lack of cellular and molecular information from early disease stages and accurate experimental models. Recently, senescent cells and neutrophil extracellular traps have been described in tissue lesions. These structures can remain in tissues for a prolonged period, potentially favoring inflammatory responses and tissue remodeling. In this review, current advances in GCA pathogenesis are discussed in different inflammatory phases. Through the description of these—often overlapping—phases, cells, molecules, and small lipid mediators with pathogenetic potential are described.

Graphical Abstract

1. Introduction

Giant cell arteritis (GCA) stands out as the most prevalent form of primary systemic vasculitis in the elderly [1]. It is an autoimmune granulomatous disease that can affect large arteries with a notable predilection of the aortic arch and its branches. The superficial cranial branches of the external carotids, along with the ophthalmic branch of the internal carotid artery, are typically involved. Despite historical recognition of the systemic repercussions, manifested through constitutional symptoms and elevated inflammatory marker levels, GCA has traditionally been considered a predominantly cranial disease [2]. However, applying advanced imaging techniques has unveiled a broader spectrum, extending beyond cranial vasculature to involve other extracranial large and medium-sized vessels, underscoring the inherently systemic nature of GCA at the tissue level [3] (Figure 1). GCA is often associated with polymyalgia rheumatica (PMR), characterized by abrupt-onset pain, aching, and morning stiffness in the shoulder and hip girdle muscles. PMR is also an independent disease, suggesting that these two disorders could represent distinct expressions of a common underlying pathological process [4].

1.1. Epidemiology

Despite its global prevalence, GCA exhibits geographic variability in its incidence and prevalence. Regardless of the diagnostic criteria used, several studies consistently identify people of northern European ancestry with the highest reported incidence (44 cases per 100,000 persons aged more than 50 years) and Asians, particularly in southeast Asia, as the area with the lowest (0.4 per 100,000 persons aged 50 years and above) [5]. Notably, there is a discernible north-to-south decreasing gradient even across Europe, highlighting the potential influence of ethnic origins in disease development. Females exhibit an almost threefold increased likelihood of developing GCA compared to males, with a propensity for more extracranial artery involvement and a higher prevalence of PMR [6]. Seasonal variation adds another layer to the epidemiological profile, with a notable preponderance of diagnoses during the spring and summer [7,8].

1.2. Risk Factors

Among the various risk factors associated with the development of GCA, age emerges as the single most important determinant. GCA abstains from affecting individuals under the age of 50, and after that, its incidence steadily ascents, with more than 80 percent of diagnosed patients surpassing the age of 70 [9]. This age-dependent pattern highlights the intricate interplay between aging and the pathogenesis of the disease. The etiology hangs between the age-related restructuring of the immune system and the age-related vascular remodeling and dysfunction. Immunosenescence, characterized by the reduction of the naïve T-cell pool, contraction of T-cell diversity, and accumulation of memory T-cells, is accompanied by low-grade inflammation, commonly referred to as inflammaging [10]. Vascular aging introduces pivotal changes in the structural properties of the vascular wall, leading to increased arterial stiffness and reduced compliance [11]. The interplay between these two aging-related phenomena forms a complex network that contributes to the initiation of GCA. Unraveling the most prominent driving forces in the aging process remains a critical challenge for a better understanding of the pathophysiology of GCA.

1.3. Clinical Picture

Regardless of the underlying pathogenetic origins, GCA presents with a variety of clinical features extending from constitutional symptoms to headaches, scalp tenderness, jaw claudication, and visual manifestations [12]. The onset of the disease typically follows a subacute trajectory over several weeks, although abrupt presentations are not uncommon. If left untreated, GCA poses the ominous risk of its most dreaded complication, permanent visual loss and, less frequently, stroke. Notably, recent reports indicate a decline in the incidence of visual loss, dwindling from 20% of patients to less than 10%, attributed to the advancements in awareness, timely diagnosis facilitated by rapid temporal artery color Doppler ultrasonography assessment [13], and swift medical intervention [14]. Although visual loss can occur abruptly and without warning, single or multiple episodes of transient visual loss (amaurosis fungax) may precede [15]. These brief episodes, though temporary, serve as critical warning signs, offering a window of opportunity for prompt recognition and initiation of treatment. According to the latest recommendations from EULAR in 2023, ultrasound, including assessment of the axillary arteries, is now advocated as the first-line imaging diagnostic test for GCA. If there is a high clinical suspicion coupled with a positive imaging test, a biopsy can be omitted from the diagnostic procedure [16]. However, clinical suspicion should always be heightened for GCA mimickers, with the most prominent being ANCA vasculitis, amyloidosis, and atherosclerosis [17]. Lately, COVID-19 has also been added to the list of conditions with similar presenting symptoms, such as headaches and acute phase responses. Thrombocytosis may lean more towards GCA, while lymphopenia may suggest COVID-19, aiding in the correct diagnosis [18].

1.4. Treatment

The current management of GCA faces challenges typified by suboptimal rates of maintaining remission. Glucocorticoids (GCs) serve as the cornerstone of GCA treatment employed for both initiation and maintenance therapy in the majority of cases. While GCs effectively inhibit the Th17-dependent pathway, their impact on the Th-1-dependent pathway—crucial for ischemic complications and long-term arterial sequelae—is limited [19]. Notably, when the GC dose is tapered below 15–20 mg/day, relapses occur in approximately 50% of patients. To address this, the incorporation of conventional synthetic disease-modifying anti-rheumatic drugs (csDMARDs) becomes necessary. However, the recent approval of tocilizumab, an anti-interleukin 6 (IL-6) receptor inhibitor, provided new treatment options, especially for relapsing and refractory disease [20]. Several other agents, including sarilumab (a monoclonal antibody against IL-6Rα), ustekinumab (a monoclonal antibody targeting both IL-12 and IL-23), mavrilimumab (a human monoclonal antibody inhibiting the GM-CSF receptor), and baricitinib (inhibitor of JAK1 and JAK2 enzymes), are currently undergoing clinical investigation [21].

1.5. Histopathology

Similar to the diversity of clinical phenotypes, a remarkable heterogeneity also exists in the histopathologic pattern of the disease, delineated into four distinct categories: (i) transmural inflammatory pattern, (ii) inflammation confined to the adventitia, (iii) small vessel vasculitis distal to the adventitia, and (iv) vasa vasoritis [22] (Figure 2).
GCA is considered the prototype autoimmune disease where both acute and chronic delayed-type hypersensitivity reactions (DTH) may unfold. Beyond the chronic granuloma lesion, which stands as the hallmark of the disease, various subsets of immune cells, cytokines, and other molecules priming the autoimmune response have been described. However, the wealth of knowledge is vastly limited at single time points, and these are often compartmentalized, posing challenges in the synthesis and interpretation of such data. In this review, we shall discuss recent advances in understanding the pathogenetic mechanisms of GCA. The findings will be structured according to the disease’s different stages, including the initiation, perpetuation, and sustainability of the inflammatory response and the operative mechanisms also involving stromal cells and, eventually, leading to tissue injury. While acknowledging the overlap inherent to many of these phases, this organized presentation seeks to provide a comprehensive and accessible framework to navigate the intricate landscape of GCA pathogenesis.

2. Pathogenetic Mechanism in GCA

2.1. Initiation Phase

Similar to almost all systemic autoimmune and autoinflammatory disorders, the triggering factor(s) of GCA remains unknown. Several reports in the past have tried to elucidate the role of infections. Furthermore, tissue alterations via intrinsic mechanisms such as cell stress and accelerated atherosclerosis have also been proposed as initiating disease events. The lack of spontaneous experimental models, along with the poor understanding of tissue injury in GCA at the very early stages of the disease, are serious obstacles to understanding the initial mechanistic events. The initiating elements, when present, act synergistically upon a particular gene profile of patients with GCA that extends from antigen presentation (MHCs), adaptive and innate immunity genes (e.g., IL-2, IFN-γ), and several regulatory genes (e.g., TLRs).

2.1.1. Genetics

The genetic component has been considered an essential factor in GCA pathogenesis, based mainly on two observations: (i) reports on familial clustering of the disease [23,24,25] and (ii) an increased prevalence in Europeans of northern and Scandinavian ancestry [26]. With the utilization of molecular techniques from different scientific groups, they sought to decipher the genetic landscape of the disease. In the past two decades, genetic studies have been categorized into two types, according to the candidate-tested genes: (i) studies of targeted gene polymorphisms associated with disease susceptibility (Table 1) and (ii) genome-wide association studies.
In the first category, the main focus of research has included genes encoding the human leukocyte antigen (HLA) classes I and II and different molecules participating in the inflammatory response of GCA (IL-1β, IL-6, IL-10, IL-17A, IL-21, IL-23R, IL-33, TNF-α, VEGF, MMP-9, MPO, and others). Several studies in small patient cohorts interrogated the role of the HLA region in the disease (Table 1). They revealed an association of GCA with the expression of HLA-DRB1*04 alleles (HLA-DRB1*0401, HLADRB1*0404, and HLADRB1*0408 haplotypes) [27,28,29,30,31,32,33,34,35,36]. Studies of genes encoding inflammatory molecules yielded various results. The rs2250889 polymorphism of the MMP-9 gene was associated with GCA susceptibility, but the study included only 30 patients [37]. Salvarani et al. demonstrated an association of the G/R 241 SNP in the ICAM-1 gene with the disease [38], but Amoli et al. reported a lack of association [39]. These studies included several weaknesses in sample size, lack of successful replicability, absence of appropriate geographical sample representation, and the use of low throughput technologies that did not allow for a large number of tested SNPs (Table 1) [40].
The research in this field entered the high-throughput screening era, producing more interesting results for the community. A large-scale study revealed that the HLA class II region is strongly associated with GCA [41], differentiating GCA from other systemic vasculitides that are genetically linked to HLA class I molecules (Takayasu arteritis, Behçet’s disease) [42] and supporting the implication of antigenic presentation processes in the initiation phase of the disease. A genome-wide association study (GWAS) confirmed this association, in addition to the identification of the plasminogen (PLG) gene polymorphism rs4252134 and the Prolyl 4-hydroxylase subunit alpha-2 (P4HA2) gene polymorphism rs128738 as genetic risk factors in GCA [43]. Furthermore, a meta-analysis highlighted the IL12B gene polymorphism rs755374 as a new risk SNP for GCA and a re-verification of the strongly associated HLA class II genes [44].
Overall, different studies exploring the contribution of SNPs in genes encoding proteins associated with the proinflammatory and tissue remodeling phases of GCA suggested the relevance of a plethora of innate and adaptive immunity molecules to disease pathogenesis. Additionally, GWAS introduced new genes as risk factors in GCA and reinforced the hypothesis of an unknown antigen-driven immune response initiating the pathogenesis of the disease.
Table 1. Genetic studies in GCA focused on genes encoding MHC class II and various inflammatory molecules.
Table 1. Genetic studies in GCA focused on genes encoding MHC class II and various inflammatory molecules.
Genes and No. of SNPs
Tested
GCA Patients/
Controls
Associated SNP/Haplotype/
Variant with GCA *
MethodRef, Year
HLA DRB1 alleles20/242-MLCA[33], 1991
HLA DRB1 alleles41/384 HLA DRB1*04 (<0.001)PCR[31], 1998
HLA DRB1 alleles42/63 HLA DRB1*0401 or B1*0404/8 (<0.03)PCR[34], 1992
HLA DRB1 alleles65/200HLA DR4 (<0.05)MLCA[27], 1988
HLA DRB1 alleles42/1609 HLA DRB1*04 (0.0005)PCR[28], 1998
HLA DRB1 alleles52/72 HLA DRB1*04 (0.0001)PCR[35], 1994
MBL, HLA DRB1 alleles65/193 HLA DRB1*04 (0.01)PCR[29], 2002
HLA DRB1 alleles53/145 HLA DRB1* 0401 (<0.05)PCR[36], 1998
HLA DRB1 alleles44/99 HLA-DRB1* 0401 HLA (0.02) and -DRB1* 0404 (0.04) PCR[30], 2004
CCR5: 1 SNP176/180-PCR-RFLP[45], 2013
CD24: 1 SNP120/195 rs3838646 (0.01) and rs8743 (0.001)Real-Time PCR[46], 2008
CRH: 2 SNPs62/147 -PCR-RFLP[47], 2002
CRP: 4 SNPs125/234 -Real-Time PCR[48], 2009
eNOS: 3 SNPs57/117 C/1/T (0.04) and C/1/G (0.02) haplotypes PCR[49], 2003
eNOS gene: 2 SNPs91/133 894 G/T (0.003)PCR-RFLP[50], 2003
FCGR2A, FCGR3A, FCGR3B and FCGR2B:
biallelic polymorphisms
85/132 FCGR2A-FCGR3A 131R-158F haplotype (0.01)SSCP Assay and Sanger Sequencing[51], 2006
ICAM-1: 2 SNPs121/228G/R 241 (0.00005)PCR-RFLP[38], 2000
ICAM-1: 2 SNPs58/129-PCR-RFLP[39], 2001
IFN-γ microsatellite 50/129 -PCR[52], 2004
IFN-γ: 4 SNPs and IL-4: 5 SNPs IFN-γ: SNP2 rs2227284 (0.0001) and IL-4 haplotype: T-T-C-A-C (0.02)Real-Time PCR[53], 2004
IL-6: 1 SNP in the promoter62/124−174 G/C (0.03)PCR[54], 2002
IL-6: 1 SNP in the promoter126/112 -PCR[55], 2005
IL-10: 2 SNPs in the promoter140/200 IL-10 promoter: −592 C/A (0.004)PCR-RFLP[56], 2006
IL-10: 2 SNPs in the promoter103/226 IL-10 promoter: −592 C/A (0.004)Real-Time PCR[57], 2007
IL17A: 5 SNPs1266/3779rs2275913 (0.01) and rs7747909 (0.04)Real-Time PCR[58], 2014
IL-18: 3 SNPs212/405rs360719 (0.003) and rs1946518 (0.02)Real-Time PCR[59], 2010
IL2/21: 1 SNP272/791-Real-Time PCR[60], 2011
IL23R: 1 SNP and IL12RB2: 1 SNP357/574IL12RB2: rs3790567 (0.039)Real-Time PCR[61], 2011
IL33: 6 SNPs and IL1RL1: 3 SNPs1363/3908IL33 gene: rs7025417 (0.041)Real-Time PCR[62], 2014
IRAK1: 1 SNP and MECP2: 1 SNP627/1520
(only females)
-Real-Time PCR[63], 2014
IRF-5: 1 SNP and 1 indel220/520 -Real-Time PCR and Sanger Sequencing[64], 2010
MCP-1: 1 SNP in the promoter79/99 haplotypes C-C (0.03) and T-T (0.005)Real-Time PCR[65], 2005
MICA-TM microsatellite polymorphism and HLA-B genotyping98/225 MICA A5 (0.0005) and HLA-B*15 alleles (0.04)PCR[66], 2007
MIF: 1 SNP in the promoter83/122 -Sanger Sequencing[67], 2005
MMP-9: 7 SNPs30/23 SNP rs2250889 (0.0009)PCR-RFLP[37], 2008
MPO: 1 SNP in the promoter156/235 −463 G/A (0.0002)PCR-RFLP[68], 2008
NFKB1: 1 SNP in the promoter96/204 -Real-Time PCR[69], 2006
NLRP1: 1 SNP685/2898rs8182352 (0.0026)Real-Time PCR[70], 2013
PTPN22: 1 SNP99/229 -Real-Time PCR[71], 2005
PTPN22: 2 SNPs and CSK: 2 Variants911/8136PTPN22: rs2476601/R620W (0.00002)Real-Time PCR[72], 2013
STAT4: 1 SNP212/678 -Real-Time PCR[73], 2009
TLR4: 1 SNP210/678 SNP rs4986790 (0.01)Real-Time PCR[74], 2009
TNF microsatellite markers a-d62/147 TNFa2 allele (0.003) and TNFa10 allele (0.02)Sanger Sequencing[75], 2000
TNF-α, IL-1A and IL-1B genes: 5 SNPs57/128-PCR[76], 2002
TRAF1: 1 SNP and C5: 1 SNP220/410 -Real-Time PCR[77], 2010
VEGF: 3 SNPS in the promoter92/200 I (0.025) and C634 (0.015)PCR-RFLP[78], 2003
VEGF: 2 SNPs 103/226 -Real-Time PCR[79], 2005
CCL2, CCR7, IL10, IL12A, IL1A, IL1B, IL1RN, IL6, IL8, IFNG, LTA, eNOS, TNF and VEGF: 130 SNPs82/166eNOS (rs2779251), VEGF (rs1885657, rs2010963, rs699946 and rs699947), IL1RN (rs17207494), IL-6 (rs7805828 and rs1546766) and CCL2 (rs1860190) SNPs (<0.05)Illumina Bead Array System[80], 2012
CCL2: C motif chemokine ligand 2; CCR5: C-C chemokine receptor 5 gene; CCR7: C-C chemokine receptor type 7; CD24: cluster of differentiation 24; CRH: corticotropin-releasing hormone; CRP: C-reactive protein; CSK: C-terminal Src kinase; eNOS: endothelial nitric oxide synthase; FCGR2A: Fc gamma receptor 2A; ICAM-1: intercellular adhesion molecule 1; IFN-γ: interferon gamma; IL: interleukin; IL1RL1: interleukin 1 receptor-like 1; IL12RB2: interleukin 12 receptor subunit beta 2; IL23R: interleukin 23 receptor; indel: insertion/deletion; IRAK1: interleukin-1 receptor-associated kinase 1; IRF-5: interferon regulatory factor 5; LTA: lymphotoxin-α; MBL gene: mannose-binding lectin gene; MCP-1: monocyte chemoattractant protein 1; MECP2: methyl CpG binding protein 2; MLCA: microlympocytotoxicity assay; MICA-TM: major histocompatibility complex class I chain-related gene A transmembrane; MIF: macrophage migration inhibitory factor; MMP-9: matrix metalloproteinase 9; MPO: myeloperoxidase; NLRP1: NACHT; LRR; FIIND; CARD domain and PYD domains-containing protein 1; PCR: polymerase chain reaction; PTPN22: protein tyrosine phosphatase non-receptor type 22; RFLP: restriction fragment length polymorphism; SNP: single-nucleotide polymorphism; SSCP: single-stranded conformational polymorphism; STAT4: signal transducer and activator of transcription 4; TLR4: toll-like receptor 4; TNF: tumor necrosis factor; TRAF1: tumor necrosis factor receptor-associated factor 1; VEGF: vascular endothelial growth factor. * p-values are in parentheses.

2.1.2. Infections and Microbiome

The antigen-driven hypothesis of triggering the initiation of vascular inflammation observed in GCA has been a matter of debate for many years in the literature. In this direction, several studies have investigated the implication of different infectious agents in vascular dendritic cell activation and subsequent disease onset [81]. Of the infectious agents interrogated, human herpes virus (HHV)-6, HHV-7, varicella-zoster virus, and Epstein–Barr virus have not been associated with GCA. Although cytomegalovirus, parvovirus B19, herpes simplex virus, human parainfluenza 1, and chlamydia pneumonia were initially correlated with the disease, none of these results were confirmed by subsequent studies in larger cohorts [81]. A previous study reported the presence of varicella-zoster virus (VZV) antigen in 64–73% of temporal artery biopsies (TABs) of large vessel vasculitis (LVV) patients, with or without temporal artery involvement, as opposed to 22% of normal arteries tested [82]. Although this finding implied a possible disease-causing mechanism of VZV infection, the study lost its validity after Pisapia et al. reported an increased false-positive rate due to non-specific staining of the antibody used in the immunohistochemistry assay for VZV detection [83].
State-of-the-art approaches such as whole genome sequencing and 16S Ribosomal RNA gene sequencing have also been utilized in search of infectious agents implicated in GCA pathogenesis. In a shotgun sequencing-based study, none of the previously reported pathogens were detected in GCA TABs. Moreover, the microbiome did not differ between GCA cases and controls since only members of the normal skin flora were observed [84]. Two studies of another group that followed aseptic techniques during tissue collection found that the microbiome of thoracic aortic aneurysms from isolated aortitis and GCA differ substantially from the microbiome of non-inflammatory aortic aneurysms [85] and that from TABs of GCA patients [86]. These studies provided novel insights into the candidate role of the microbiome of different vascular components in health and disease. However, the translation of these findings in a comprehensive manner that could enrich our knowledge of GCA pathogenesis still has a long way to go.

2.1.3. Vascular Aging/Inflammaging

Undoubtedly, increasing age is a major etiological factor in GCA’s initiation, with different compartments of immunity being affected [11]. T-cells, in particular, exhibit a senescent phenotype due to the loss of the CD28 cell surface marker [87]. CD4+ CD28− and CD8+ CD28− cells are reported to be enriched in T-cell subsets of aged individuals [88]. There is also a marked decrease in the numbers of naïve T-cells and the T-Cell Receptor (TCR) repertoire diversity as opposed to an increase in T effector and memory cells [89]. Dendritic cells (DCs) can also be impacted by aging, displaying impaired activation and migration capacity [90]. Senescent macrophages display an altered, more inflammatory phenotype characterized by the expression of proinflammatory mediators (IL-1β, IL-6, TNF-α) [91]. It is reported that epigenetics may play a vital role during this so-called inflammaging process. Indeed, DNA methyltransferase 1 (DNMT1) levels in T-cells are decreased during the aging process [92]. DNMT1 is an enzyme that facilitates the maintenance of DNA methylation levels between cell divisions, a disturbed process during aging that leads to altered methylation levels in T-cells, a phenomenon associated with increased risk for autoimmunity and cancer. On the other hand, structural arterial changes are observed that correlate with increasing age, including calcium deposition, increased artery stiffness, wall thickening, and alterations in the extracellular matrix of the arteries [93]. The effect of inflammaging on the immune system, which is disturbed in GCA, and the vasculature, the target organ of the disease, supports the central role of senescence in GCA pathogenesis.

2.2. Perpetuation and Sustainability of the Inflammatory Response

Different studies of tissue biopsies of GCA patients reveal many types of immunocytes involved in innate and adaptive immune responses that are usually activated as attested by cellular markers and cytokines. Essentially, the perpetuation of tissue inflammatory response is performed through acute DTH mechanisms in which activated monocytes play a central role. At the same time, sustainability is implemented through chronic DTH responses with granuloma formation, which is the histological hallmark of the disease. In some instances, the local autoimmune injury is probably continuously fueled by the formation of ectopic germinal centers [94]. Disturbances in immune cell subpopulation frequencies in peripheral blood mononuclear cells (PBMCs) are also observed during active and inactive disease states [95]. An unmet need of GCA is the exact definition of cells participating in the acute inflammatory response in correlation with the cells in remission. Indeed, the type of cells and the inflammatory mediators differ in the acute and remission phases since the metabolic landscape is also different [96]. To this end, other groups and ours have suggested the investigation of pairs of biological samples in both activity and remission states in an attempt to gain clinically relevant biomarkers. Hereafter, the types and functions of major immune cell types are described.

2.2.1. Dendritic Cells and TLRs

The activation of resident DCs and subsequent activation of T-cells in the affected GCA arteries is thought to be the initial step of the intense inflammatory response. Following sensing intrinsic or extrinsic antigens, these cells induce strong innate and adaptive immune responses via TLR ligation and cell activation, which leads to chemokine and cytokine production, orchestrating tissue infiltration from T-cells. Thus, TLR differential expression and activation in the context of this pathology is of great importance [97].
Resident DCs observed in the adventitia of normal arteries display an immature phenotype [98,99] and contribute to immune surveillance. Detection of activated DCs in TABs is not exclusive to GCA but has also been described in TABs of patients with PMR. Activated DCs are located in the adventitia of PMR patient TABs as opposed to GCA patient TABs, in which activated DCs are extended in all three layers of the vascular wall [99]. Moreover, DCs are restricted to the artery and do not migrate to lymph nodes [98], a phenomenon facilitated by a positive-feedback loop of specific chemokine production (CCL19 and CCL21) and their receptor, CCR7 [99], leading to the migration of more DCs at the site of inflammation and restricting their escape to the lymph nodes. Additionally, DCs express CD83, an activation marker, and CD86, a co-stimulatory molecule that renders them capable of activating naïve T-cells. A study in a human artery–mouse chimera model of GCA showed that DC depletion with an antibody against CD83 was able to effectively reduce the inflammatory response, T-cell infiltration, and IFN-γ production [99], recapitulating the significance of DCs in the initial inflammatory cascade of GCA. It has also been demonstrated that the immunoprotective PD-1/PD-L1 immune checkpoint is defective in GCA-affected tissue DCs, thus rendering them more susceptible to activation [100]. Interestingly, a study investigating the expression profile of TLR genes of 6 GCA-affected arteries obtained during autopsy showed a distinct TLR profile for each affected vessel studied [101]. Until recently, there were no reports on DC subpopulations in the peripheral blood of GCA patients. Reitsema et al. investigated the frequencies of DC subsets in the peripheral blood of GCA and PMR patients compared to healthy individuals for the first time [102]. The authors noted that plasmacytoid DCs (pDCs) and type-2 conventional dendritic cells (cDC2) frequencies did not differ between patients and controls, whereas cDC1 frequencies were substantially reduced in patients. Additionally, in GCA/PMR patients, pDCs exhibited higher expression of the immune checkpoint CD86 and reduced expression of CD40, whereas cDC2 subsets exhibited lower activation status as shown by lower HLA-DR activation marker expression [102]. Overall, the interpretation of these results is challenging in understanding GCA pathogenesis since there are no surrogate data of DC subsets on the inflamed tissue level.
Examining the mechanism of DC activation in the context of the disease is pivotal in understanding the clinical heterogeneity in GCA, with some patients exhibiting disease restricted to temporal arteries while others display a more pronounced inflammatory response in different large vessels. A multidimensional study correlating TLR repertoire, pathogen expression (bacteria and viruses), and the activation status of different DC subpopulations in different anatomically affected arteries has never been conducted and may provide insights into understanding the etiology and initiation phase of GCA.

2.2.2. CD4 T-Cells

In GCA, a pronounced T-cell compartment participates in the underlying inflammation [103]. Indeed, activated DCs recruit CD4+ T-cells at the site of inflammation through CCL18, CCL19, CCL20, and CCL21 chemokine expression [104]. Upon arrival, activated CD4+ cells differentiate into Th1 and Th17 T-cell subsets through proinflammatory signals of the inflamed artery (IL-1β, IL-6, IL-12, IL-18, and IL23) [105,106]. In Th1 responses, CD4+ cells, under the influence of IL-12 and IL-18, differentiate into Th1 cells that secrete IFN-γ [107], a critical regulatory cytokine in the disease’s pathogenesis [108]. On the other hand, Th17 responses include the differentiation of CD4+ cells into Th17 cells under the effect of IL-1β, IL-6, and IL23 [107]. It is well established that these two T-cell axes operate in the vicious, inflammatory cycle observed in the disease. However, the order of appearance of Th1 and Th17 responses is unknown. GC treatment is highly effective in restricting Th17 responses, as proved by the reduction of both the number of Th17 cells in the peripheral blood and IL-17 serum levels [105,109]. However, IFN-γ producing Th1 responses show resistance after GC treatment in GCA patients [105,110].
Another dysregulated CD4+ subset implicated in GCA pathogenesis is T regulatory cells (Tregs). Tregs are reduced in both peripheral blood and at the tissue level in GCA [105,106,111]. Moreover, the high levels of IL-6, IL-21, and IL-23 in the inflamed tissue microenvironment lead to the restricted expression of FOXP3 transcriptional factor, which is crucial for the differentiation of Tregs, favoring the upregulation of RORγt transcriptional factor that mediates Th17 differentiation [112]. An imbalance of the Th17/Treg axis participating in the GCA pathogenetic mechanism has been observed and is restored by IL-6R blockade as shown by normalization in Treg peripheral blood frequencies and a decrease in activation status [111,113]. Miyabe et al. also highlighted that Tregs in GCA present a reduced suppressor capacity, possessing a proinflammatory phenotype with increased production of IL-17 [114].
Recent evidence has emerged for the role of Th9 cells in GCA. Th9 cells constitute a distinct lineage of T-cells differentiated from CD4+ naïve cells under the influence of IL-4, TGF-β, and thymic stromal lymphopoietin (TSLP) and are effective producers of IL-9. In GCA, Th9 cells are mainly observed in artery tissues with transmural inflammation and small vessel vasculitis, as opposed to their absence in vasa vasorum vasculitis [115]. Furthermore, patients with GCA exhibit a defective, macrophage-induced CD155-CD96 immune checkpoint that leads to Th9 expansion [116]. The same study showed increased vessel wall destruction due to increased IL-9 production in a humanized mouse model of GCA [116]. Although all groups have not demonstrated the functions and disease relevance of the Th9 subset in GCA, their implication in the pathogenic phenomena appears important and requires further research.
GCA-affected tissue may possess ectopic germinal center (EGC)-like structures that are important for perpetuating local inflammatory responses. In the formation of such structures, Follicular helper T (Tfh) cells play a central role. Tfh cells have been detected in the inflamed artery and the peripheral blood of GCA patients [117]. Tfh cells are capable of producing IL-21, which, among other functions, increases B-cell differentiation into plasmablasts and triggers germinal centers to produce immunoglobulins [118]. However, autoantibodies characterizing the disease have not been detected yet.

2.2.3. CD8 T-Cells

For years, the role of CD8+ T-cells in GCA has not attracted much attention due to their low number in both the periphery and tissue [119,120,121]. Nonetheless, CD8+ cells present oligoclonality in GCA tissue. They may infiltrate temporal arteries, producing proinflammatory cytokines such as IL-17A and IFN-γ as well as cytotoxic substances such as granzymes A and B [119]. These observations give rise, once again, to an antigen-driven theory of GCA initiation. Furthermore, a decreased number of immunosuppressive CD8+ CCR7+ FoxP3+ Tregs has been observed in the peripheral blood of GCA patients, potentially leading to a failure in the control of CD4+ T-cell proliferation and activation through a NADPH oxidase-2 dependent mechanism [122]. In addition, a decrease in CD8+ T-cells was associated with aging and GCA pathogenesis [122]. The fact that this defect was not abrogated by GC treatment in patients raises new questions about the pathogenetic role of CD8+ cells.

2.2.4. B-Cells

The role of B-cells in GCA had been overlooked for many years due to the initial reports of a low number of B-cells infiltrating the temporal artery [123,124], suggesting that humoral immunity plays a marginal role in the inflammatory process. Data on serum autoantibodies in GCA patients support a more active role for B-cells in the inflammatory process. Autoantibodies that were identified include (i) low titers of anti-cardiolipin of the IgG isotype that disappeared following GCs treatment [125,126,127,128]; (ii) anti-endothelial cell antibodies which were not specific for GCA and were also detected in other systemic vasculitides [129,130]; and (iii) anti-smooth muscle cell antibodies, but the study included a low number of patients [130]; additionally, (iv) Baerlecken et al. reported antibodies against the human ferritin heavy chain in a high percentage of GCA patients (92%) [131] while Régent et al. reported lower titers, but yet with high prevalence (71.9%) in GCA patients and 34% in patients with a diagnosis other than GCA diagnosis [132]. These results collectively support the idea that these autoantibodies are not disease-specific but an epiphenomenon of the inflammatory bulk.
In recent years, the formation of tertiary lymphoid organs (ATLOs) has been identified in the aortas of GCA patients but not in the affected temporal arteries [94,133]. Moreover, those ATLOs were found in the adventitia of inflamed aortas and were absent from GCA-positive temporal arteries, suggesting different organization according to the size of the affected artery. ATLOs consist of follicular dendritic cells located near T-cells and endothelial cells in the media layer, supporting the organization of lymphoid tissue against arterial wall-derived antigens. In the peripheral blood of treatment-naïve GCA and PMR patients, B effector cells are decreased compared to healthy controls and reach normal levels after GC treatment, exhibiting an enhanced IL-6 production capacity [134]. In contrast, serum levels of CXCL9 and CXCL13, major chemokines for the organization of ATLOs, are elevated [135,136]. The same study highlighted that B-cells were detected in the inflamed arterial tissue, and their migration pattern followed the CXCR3–CXCL9 and CXCR5–CXCL13 chemokine axes [136]. The above data indicate that B-cells in active disease are reduced in the peripheral blood due to their migration to the affected temporal arteries. In cases of large vessel involvement, ectopic lymphoid tissue is generated and is possibly connected to chronic inflammatory processes.

2.2.5. Monocytes

The abundant expression of IFN-γ in the initial steps of inflammation induces CCL2 expression by vascular smooth muscle cells (VSMCs), leading to tissue infiltration by monocytes of the classical subset, expressing the CCR2 receptor [137]. In addition, non-classical monocytes also participate in the inflammatory process and reach out to the tissue lesion under stimuli of the CX3CR1–CX3CL1 axis [138]. The predominant type of monocytes in the GCA tissue lesions is still a matter of debate, with different groups reporting either the classical monocyte subset [137] or non-classical subsets as the major monocytic type [138]. In the peripheral blood of GCA patients during diagnosis, elevated classical monocyte numbers [95,138] persisted after three months of GC treatment. Interestingly, disease remission correlated with a decrease in the number of intermediate and non-classical monocytic lineages, indicating that GCs act partly towards normalizing monocyte numbers [138]. Following integrative analysis of the methylome and transcriptome of CD14+ cells in GCA active and inactive disease, a recent study identified IL-11 as a novel cytokine pathway associated with the disease [139]. However, functional studies on the relevance of their findings to disease pathogenesis are needed.

2.2.6. Cytokines with a Significant Contribution to the Inflammatory Response of GCA

IL-6: IL-6 is considered to be the leading inflammatory cytokine in GCA’s pathogenetic mechanism. Serum levels of IL-6 appear to be elevated early during the disease course [105] and decrease rapidly after a few hours of GC initiation [140], without, however, reaching the levels of healthy individuals, even after chronic administration of GCs [141]. In addition, after GC tapering, IL-6 is among the first cytokines that arise in GCA patients’ serum [140]. IL-6 also correlates highly with acute phase reactants (erythrocyte sedimentation rate and C-reactive protein) and may serve as a biomarker of disease activity [142]. The above clinical observations made IL-6 a therapeutic target, and tocilizumab, a humanized monoclonal antibody to the IL-6 receptor (IL-6R), is now consistently used for patients with relapsing or refractory disease [143].
Although targeting of IL-6 is successful in therapy, the exact mechanism of action in GCA pathogenesis has not been elucidated, probably due to its pleiotropic effects. IL-6 is expressed by various immune and stromal cells, including activated monocytes, macrophages, B-cells, T-cells, fibroblasts, and endothelial cells [144]. IL-6 signal transduction is mediated by the complex IL-6/IL-6R/gp-130, with the latter being ubiquitously expressed, while hepatocytes, activated monocytes, macrophages, B-cells, and endothelial cells mainly express IL-6R. An established function of IL-6 is to control the balance between Th17 cells and Tregs [145], which is also evident in GCA [111,114]. The reduced frequency and the proinflammatory phenotype of Th17 expressing Tregs are reversed in GCA patients after tocilizumab treatment. There are conflicting reports on the effect of IL-6 on tissue remodeling. In a study by O’Neill et al., serum amyloid A (SAA) protein, which is triggered by IL-6 hepatocyte signaling and is increased in GCA patients’ serum, induced the protein expression of vascular endothelial growth factor (VEGF) and MMP-9 in an ex vivo culture model of temporal arteries. However, a more recent study of the same group argues a role of IL-6 in tissue remodeling since there was no effect of IL-6 treatment on myofibroblast proliferation and migration in a myofibroblast outgrowth culture model of GCA [146].
GM-CSF: Granulocyte-macrophage colony-stimulating factor (GM-CSF) has been recently suggested as a highly influential cytokine in GCA [147]. This proinflammatory cytokine is expressed by fibroblasts, endothelial, epithelial, myeloid, and T-cells upon appropriate stimulatory cues [148]. The GM-CSF heterodimeric receptor is composed of an α chain specific to GM-CSF and a signal transduction β chain, which is also found in the IL-3 and IL-5 receptors and, upon phosphorylation, triggers the activation of the JAK2/STAT5 signaling pathway [148]. Patient serum levels of GM-CSF are extremely low and comparable with healthy individuals [147]. On the contrary, GM-CSF protein levels are increased in GCA TABs [147] and PBMCs upon stimulation in vitro [106], suggesting a paracrine function of this cytokine on the inflamed tissue. The only functional study about GM-CSF in GCA was by Corbera-Bellalta et al. and highlighted the role of this cytokine in GCA. GM-CSF blockade by mavrilimumab, a fully human IgG4 monoclonal antibody to GM-CSFRα, abolished immune cell infiltration, inflammatory markers, and tissue remodeling factors, suggesting a role of GM-CSF in both the perpetuation of the inflammatory response and tissue injury phases [147]. A phase-2 clinical trial on mavrilimumab showed promising results on sustaining disease remission in week 26. However, further clinical trials are needed to determine whether this treatment modality is superior to current treatment options [149].

2.3. Tissue Injury

GCA is characterized by vital organ and life-threatening comorbidities as a result of ongoing tissue injury. The most severe and common are blindness and aneurysm formation. To this end, the mechanisms involved in tissue remodeling following acute inflammatory responses are of paramount importance. This section will discuss cells and molecules mediating tissue injury and their relation to acute inflammatory responses (MMPs, cytokines, and immune-mediated thrombosis).

2.3.1. Neutrophils

Until recently, there was scarce information in the literature about the presence and involvement of neutrophils in GCA. A few reports suggested a low prevalence of neutrophil infiltration in GCA biopsies [22,150]. Our group has reported the presence of neutrophils in GCA TABs and identified their localization in the adventitia and media of inflamed arteries [151]. Interestingly, we identified the release of neutrophil extracellular traps (NETs) decorated with proinflammatory mediators (IL-6 and IL-17A) in the adventitia adjacent to the vasa vasorum. Another group identified a subset of immature neutrophils with a phenotype of CD66b+CD15+CD10lo/–CD64– in peripheral blood of GCA patients and granulomatosis with polyangiitis (GPA) patients, but not in healthy individuals [152]. This immature cell subset was resistant to apoptosis, exhibited high potential for generating reactive oxygen species (ROS), and infiltrated the inflamed tissue. Neutrophilia is observed in the peripheral blood of GCA treatment-naïve patients during active disease, [153]. Neutrophils possess three distinct phenotypes with GC treatment over 24 weeks [153]. Neutrophils in weeks 1 and 24 exhibit high capabilities for endothelial adhesion, and neutrophils in week one can suppress T-cell proliferation as opposed to week 24. Based on the loss of neutrophil subsets with a capacity to suppress T-cell responses during GC tapering, the authors suggested a possible role of neutrophils in disease relapse, but further research is needed to support this hypothesis. Serum biomarkers related to neutrophils have also been detected in GCA patients. These include calprotectin and N-formyl methionine (fMET), markers of neutrophil activation, which are increased in the serum of GCA [154], as well as higher levels of circulating NETs in LVV patients when compared to healthy controls [155]. However, the circulating levels of NET components are non-indicative of NET release [156] and these findings have not been validated in larger cohorts. Another unexplored role of NETs is their implication in immunothrombosis, which may also play a role in tissue injury observed in GCA since intralaminal thrombosis is observed in 10–20% of positive GCA biopsies. There have been reports of decorated NETs with tissue factor (TF) in active lupus kidney lesions. Moreover, in a mouse model of sepsis-induced acute lung injury, NET generation mediated by stimulator of interferon genes (STING) activation resulted in endothelial damage via enhanced production of TF. Taken together, these findings suggest further research on the role of neutrophils in promoting immunothrombosis in GCA.
Overall, neutrophil biology in GCA requires further exploration to understand their role in tissue remodeling and/or disease relapse.

2.3.2. Macrophages

Macrophages are critical players in the inflammatory bulk identified in TABs of GCA patients. Therefore, they are thought to be of major importance both during the perpetuation of the inflammatory response and the subsequent vascular remodeling phase. Monocytes that reach the inflamed artery differentiate into macrophages and form multinucleated giant cells, a phenomenon that serves as the histological hallmark of the disease since giant cells are detected in 57–74.8% of GCA tissue biopsies [22,157]. The critical role of macrophages is manifested by producing several inflammatory mediators, including matrix metalloproteinases (MMPs) and growth factors, leading to tissue remodeling and vascular occlusion [158,159]. Macrophage subsets are heterogeneous, exhibiting high plasticity and various functions [160]; in GCA, research has focused on M1 and M2 macrophage subsets [161,162]. M1-like macrophages are located in the adventitia and media layers of the artery [163], secreting the proinflammatory cytokines IL-1β and IL-6, leading to the perpetuation of the inflammatory response [161]. MMPs that act on matrix degradation [117], reactive oxygen species (ROS) that disrupt the homeostasis of VSMCs, and endothelial cells (ECs) [163] have also been described. M2-like macrophages are distributed in the intima-media border of the artery and promote wound healing via the production of platelet-derived growth factor (PDGF) [164] and VEGF [165], forcing extensive tissue remodeling and vascular occlusion. Macrophage-mediated tissue destruction has been associated with CD206+/MMP-9+ macrophages, while FRβ+ macrophages are linked to intima proliferation, and their tissue distribution is regulated by local expression of GM-CSF and M-CSF, respectively [162]. The same group recently reported that GM-CSF skewed CD206+ macrophage production of the angiogenic factor YLK-40, which is also elevated in the serum of GC-treated patients. Moreover, the knockdown of YLK-40 resulted in a vast decrease in MMP-9 production from macrophages [166]. To this end, the authors propose that targeting the blockade of YLK-40 or GM-CSF could offer a therapeutic option in controlling inappropriate tissue remodeling, a still unmet need in the management of the disease.
Given the central role of macrophages in GCA pathogenesis and the high disease relapse rate after GC withdrawal, it is of great importance to study inflammasome activation pathways in the context of this disease. NLRP3 inflammasome hyperactivation has been associated with multiple autoimmune and autoinflammatory diseases, including Sjögren’s syndrome, SLE, ankylosing spondylitis, and RA [167], and has been proposed as a therapeutic target in managing chronic inflammation in these diseases. However, there are no studies interrogating the implication of inflammasome activation during perpetuation and sustainability of the inflammatory response in GCA.
With the advancements in the field of novel PET/CT radiotracers and in an effort to implement precision medicine approaches in disease diagnosis and monitoring, several candidate radiotracers have been proposed for GCA imaging, including those targeting macrophages [168]. Although macrophages are highly relevant to disease pathogenesis during initiation and relapse, there is a limited number of studies investigating the role of this immune cell subset in GCA. We propose the implementation of a structured research strategy that involves the integrative analysis of different omics technologies in two-time points of the disease, activity and inactivity, to identify the most sensitive-to-change parameters that could serve as next-generation biomarkers to be utilized as radiotracers for non-invasive disease diagnosis and monitoring (Figure 3).

2.3.3. Matrix Metalloproteinases (MMPs)

Macrophages, along with VSMCs, are the primary cellular source of MMP-2 and MMP-9 production, two enzymes thought to be involved in GCA tissue destruction via internal elastic lamina disruption [169]. There are several reports on the strong expression of MMP-9 in GCA lesions [162,170,171] as well as an increased ratio of MMP-9/TIMP1 [172], with the latter being a natural inhibitor of this gelatinase type. As mentioned before, MMP-9-mediated matrix degradation facilitates T-cell infiltration in the affected GCA arteries as described in a study utilizing a human artery-SCID-mouse model [117]. On the other hand, MMP-2 expression in GCA lesions is controversial since it is constitutively expressed by VSMCs and ECs, and its expression is comparable both in GCA tissue and non-GCA control arteries [169,173]. MMP-12 gene expression is highly upregulated in positive TABs and its expression is even more pronounced compared to MMP-9. However, its protein levels and enzymatic activity have not been evaluated in the affected tissue [174].

2.3.4. Vascular Smooth Muscle Cells and Myofibroblasts

Previous studies have shown that VSMCs possess a critical role in the pathogenetic mechanism of GCA and, more specifically, in the perpetuation of the inflammatory response and the tissue remodeling stages [175]. Upon IFN-γ stimulation, VSMCs produce the chemokines CXCL9, CXCL10, CXCL11, and CCL2 that facilitate the recruitment of T-cells and monocytes at the site of inflammation. In an in vitro model of GCA, the blockade of IFN-γ resulted in the reduction of macrophage infiltration in cultured arteries due to reduced VSMC chemokine production [176]. In large arteries, VSMCs reside in the media layer and, upon stimulation, are capable of migrating into the intima, where they differentiate into myofibroblasts that proliferate, producing extracellular matrix proteins (collagen I, collagen III, and fibronectin) [177], leading to lumen stenosis and vessel occlusion [178]. The molecules that mediate the signaling cascade of VSMCs regarding wound healing are thought to be PDGF [164], TGF-β, and endothelin 1 (ET-1) [179,180]. ET-1 receptors A and B are expressed on the surface of VSMCs and their blockade reduces the proliferation and migration of VSMCs across the inflamed tissue [181]. Furthermore, imatinib mesylate treatment, which inhibits the PDGF receptor and is currently used in the treatment of lung fibrosis, reduced VSMC migration and proliferation in vitro, highlighting the central role of PDGF in mediating the activation of signaling pathways regulating the VSMC profile and possibly vascular occlusion in GCA [177]. Another element determining the fate of VSMCs during GCA is neurotrophin expression. In a study investigating their role in GCA, Ly KH et al. reported an enhanced expression of nerve growth factor (NGF), brain-derived neurotrophic factor (BDNF), tropomyosin receptor kinase B (TrkB), and sortilin in GCA temporal arteries compared to control arteries that also express these molecules to a lesser extent [182]. The authors also reported that NGF and BDNF facilitated VSMC migration and proliferation in vitro. However, these findings need further confirmation to elucidate whether neurotrophins have a primary or secondary role in vascular occlusion in GCA.

2.3.5. Endothelial Cells

Endothelial cells (ECs) are observed as a thin layer surrounding the lumen of large vessels, but can also be found in the vasa vasorum of large arteries. Their primary function is to protect the inner walls of the artery from immune cell infiltration. However, during the inflammatory response in GCA, ECs are activated and express adhesion molecules (ICAM-1, ICAM-2, P-selectin, E-selectin, and VCAM-1) [183,184], thus facilitating T-cell adhesion and tissue infiltration. Some authors, among them our group, consider activation of the endothelium in vasa vasorum to be an initial finding resulting in the breach of tolerance in this immune-privileged region and spreading the inflammatory response in all layers of the artery, thus creating transmural inflammation, the most common histologic finding of GCA-positive biopsies. Vasa vasorum ECs, under the influence of serum VEGF, produce an enhanced expression of Jagged1, which is a Notch ligand [185]. The activation and upregulation of the Notch-mTORC1 pathway are currently considered the central module for CD4+ T-cell recruitment and polarization into Th1 and Th17 cells [186]. This mechanistic study is coupled to the already observed NOTCH1 upregulated expression in CD4+ cells of GCA patients [187].

3. Chronic Inflammatory Components on the Tissue Level

3.1. NETs and Senescent Cells

In recent studies, the properties of two cellular structures in tissue injury of GCA have emerged. These are neutrophil extracellular traps (NETs) and senescent cells, which we have recently detected in GCA TABs and have been associated with IL-6 expression [151,188] (Figure 4).
Netosis is a lytic type of cell death that is activated upon neutrophils’ encounter with pathogens as seen during infection, but also under sterile inflammation [189]. A characteristic disassembly of neutrophil cytoskeleton, histone citrullination, chromatin decondensation, and assembly of toxic granules containing myeloperoxidase (MPO) and neutrophil elastase (NE) are the hallmarks of this mechanism [190]. There is increased scientific interest in the organ-specific effects of NETs in autoimmune diseases such as systemic lupus erythematosus (SLE), ANCA vasculitis, and psoriasis [191,192,193]. The protein decoration of NETs (MPO, proteinase 3) causes the maintenance of the inflammatory response via autoantibody generation in an ANCA-associated vasculitis mouse model [194]. NETs also serve as a scaffold for thrombus build-up and platelet aggregation in patients with antiphospholipid syndrome (APS) [195]. In this direction, our group has reported the presence of NETs decorated with IL-6 and IL-17A in TABs from GCA patients [151]. However, the role of these toxic structures requires further clarification in the disease’s pathogenesis.
Cellular senescence is a phenomenon in which normal cells, upon intrinsic and extrinsic stimuli, are led to cell cycle arrest followed by metabolism dysregulation, macromolecular damage, and a specific senescence-associated secretory phenotype (SASP) [196]. The SASP secretome includes a variety of proinflammatory mediators, chemokines, growth factors, angiogenic factors, bioactive lipids, and MMPs that may act in a paracrine or autocrine manner to induce senescence in vivo, triggering a positive-feedback loop that leads to the perpetuation of the inflammatory response [197]. Senescent cells have been identified in the context of human diseases, including COVID-19 [198], where senescent cells were detected in SARS-CoV-2-infected lung tissue, and Hodgkin lymphoma, where a high percentage of senescent cells were detected in the affected lymph nodes [199,200]. Our group has reported the presence of senescent cells in biopsy-proven GCA specimens [188]. Senescent cells in GCA tissue mainly originate from fibroblasts, endothelial cells, and macrophages, while the detected SASP involves IL-6 (fibroblasts and macrophages) and MMP-9, with the latter restricted to senescent fibroblasts. CGA artery culture-conditioned media induced IL-6-associated senescence in primary fibroblasts in vitro, an effect that was reduced upon successful IL-6 signaling inhibition by tocilizumab. Another recent study has also reported the implication of activated senescent pathways in the pathogenesis of GCA [201]. However, the presence of senescent cells was not illustrated since their approach did not involve the consensus guideline multimarker algorithm adopted by the International Cell Senescence Association [196,202].
Interestingly, these two structures are potentially linked. A recent study addressing tissue remodeling mechanisms in the context of retinopathies demonstrated that senescent cells, via their SASP components, attract neutrophils, which release NETs and ultimately clear senescent cells, contributing to tissue remodeling [203].
NETs and senescent cells share common properties that may affect the sustainability of autoimmune response to tissue injury. Indeed, they may stay for long periods in inflamed or (probably most importantly) in post-inflamed tissue. They are detected in GCA-affected tissue decorated with inflammatory cytokines and/or MMPs, thus affecting tissue remodeling. Further studies are required to address the function of these inflammatory structures in detail and to decipher whether they contribute to the perpetuation of the inflammatory response or chronic tissue injury detected in GCA (Figure 5).

3.2. Lipid Mediators in Vascular Inflammation

Unresolved inflammation is crucial for the pathophysiology of commonly occurring vascular diseases such as atherosclerosis and aneurysm formation [204]. Despite being an immune-driven vascular disease, research on GCA pathogenesis has consistently neglected and never investigated mechanisms of the resolution of inflammation, mediated by ω6 and ω3 polyunsaturated fatty acids (PUFAs), critical regulators of inflammation [205]. It is well established that within self-limited acute inflammatory exudates, pro-resolving families of lipid mediators are identified, termed resolvins, protectins, and maresins, which actively stimulate cardinal signals of resolution, including cessation of leukocytic infiltration, counter-regulation of proinflammatory mediators, and uptake of apoptotic neutrophils and cellular debris [205]. The biosynthesis of these resolution-phase mediators in sensu stricto is initiated during lipid-mediator class switching, which is the biochemical transition from inflammation to resolution, wherein early proinflammatory lipid mediators (LMs) such as prostaglandins and leukotrienes are replaced by specialized pro-resolving lipid mediators (SPMs) in exudates. The mechanisms of this shift in the LMs profile involve temporal changes in the expression, localization, and activity of critical cellular enzymes such as 5- and 15-lipoxygenases (LOXs) [206]. Once available, SPMs coordinate crosstalk between leukocytes and local cell populations, promoting an M1–M2 phenotypic transition in macrophages that is central to tissue repair [207]. SPMs further promote resolution by positive-feedback effects on LOX activity and SPM receptor expression in leukocytes [208]. Translation of these concepts into therapies for complex human diseases is the current challenge faced by investigators in the field of inflammation.
Evidence suggests that the resolution of vascular inflammation is an essential driver of vessel wall remodeling and functional recovery [204]. According to recent studies on vascular inflammation, stromal cells and their interactions with leukocytes are directly impacted by SPMs, which are locally synthesized in vascular tissues and have protective effects on the injury response [204]. Moreover, studies imply that the ratio of proinflammatory LMs to SPMs may reflect a systemic indicator of poor prognosis, greater tissue damage, and high mortality [209,210]. For this reason, measuring SPMs and proinflammatory LMs in both TABs and the periphery of GCA patients is of the greatest importance.
Studies of lipid mediators in GCA are lacking, and this is a significant unmet need for the disease. A speculation that urgently needs to be proven is that GCA patients might have an imbalance of proinflammatory LM/SPMs with a predominance of a “hazardous” LM signature that exacerbates hyperinflammation and tissue damage. Research is required to identify the potential for “resolution therapeutics” in GCA, enhance clinical measurement tools, and comprehend the molecular and cellular mechanisms of resolution in the vasculature.

4. Experimental Models in GCA

4.1. Mouse Models in GCA

While no specific mouse model perfectly mimics all aspects of GCA, researchers have developed a few experimental models to study different aspects of vasculitis [211]. Of the proposed animal models in GCA, the most widely used is the human artery xenograft mouse model, in which human artery implantation is established in immunodeficient mice [185,212]. It is the only model that has been useful to test various therapeutics for GCA (Table 2). Although the use of immunodeficient mice provides a simulation of the human environment in vivo, several challenges persist, including, among others, the increase in the survival rate of mice to extend the observation duration and the efficient reconstruction of the human B-cell immune response [211].
As TLRs have been implicated in the pathogenesis of LVV [213], activating adventitial DCs either in the vasa vasorum area or in perivascular adipose tissue is another method to induce arteritis in mice. However, this method is technically challenging, particularly in rodents, due to the thin artery width and the questionable presence of vasa vasorum in the mouse aorta [214,215] (Table 2). Another effort for the development of vasculitis in mice is the induction of arterial injury through arterial cuffing in prone models for arteritis or through an intraluminal balloon (coupled with TLR ligands). More modified arteritis-prone models have shown encouraging data for establishing an LVV model [216,217,218].
It is important to note that while these models exhibit certain features of vasculitis, none fully replicate the complexity and heterogeneity of human LVV, while others have not been investigated in the context of GCA pathogenesis yet (Table 2). Researchers often use a combination of these models and study various aspects to gain insights into the pathogenesis, underlying mechanisms, and potential therapeutic interventions for large vessel vasculitis in humans. Additionally, advances in modeling techniques continue to refine these models for better representation and understanding of systemic autoimmune vasculitis.
Table 2. Mouse models in the research of GCA pathogenesis.
Table 2. Mouse models in the research of GCA pathogenesis.
Animal ModelMechanism of ActionLimitationsRef, Year
Human artery engraftin NOD-SCID miceleakage, spontaneous thymomas, very short lifespans[212], 1997
[185], 2017
Induction of arteritis through administration of TLRs and activation of adventitial DCsat the vasa vasorumtechnically challenging for rodents[214], 2022
[215], 2003
at perivascular adipose tissue (PVAT)involvement in atherosclerosis, speculation for GCA pathogenesis[219], 2022
[220], 2000
[221], 2005
Induction of arterial injury through arterial cuffingin genetically modified atherosclerosis-prone micespeculation for GCA pathogenesis[216], 2002
in IBP-deficient or IL-1Ra deficient micenot sufficient to initiate pan-arteritis[217], 2003
[218], 2014
Induction of arterial injury through intraluminal balloonin atherosclerotic micetested in mouse and rat models of atherosclerosis, speculation for GCA pathogenesis[222], 2007
Modified arteritis-prone modelsIL1-A deficiency mice upon artery injury arteritis development is dependent on the genetic background[223], 2005
or IL25 administrationspeculation for GCA pathogenesis[224], 2019
IBP-deficient mice with TCR specific to OVA peptidesspeculation for GCA pathogenesis[225], 2008
Infection-related chronic vasculitisIFNg-receptor-deficient mice infected with γ-herpes virus 68IFNg signaling deficiency not investigated in LVV patients[226], 2001
[227], 1998

4.2. Current and Future In Vitro Models of LVV

Artery culture models: For the last decade, most groups have depended on using in vitro models of whole temporal artery culture to address questions requiring functional studies. In this context, two models have been proposed in the literature. The first and the most widely used model involves small temporal artery sections cultured in a tri-dimensional matrix for five days that facilitates the study of protein and RNA kinetics after specific drug treatments [109]. This model has been successfully used to investigate the effect of significant cytokines in disease pathogenesis [147,176]. However, it involves some limitations, including a bias in the levels of different molecules originating from the use of Matrigel and the ex vivo manipulations of the model. The second proposed in vitro model involves the culture of temporal artery sections for a shorter period (24 h) without using any supporting matrix [228]. This model is implemented to collect the secreted inflammatory microenvironment of a GCA-affected artery and use it as a conditioned medium for further applications (e.g., in treating PBMCs, endothelial cells, and myofibroblasts) [229].
Even though these models have expanded our understanding of GCA pathogenesis, they do not facilitate the investigation of the early phases of disease initiation or chronic tissue remodeling. With the advancements in the diagnostic approaches in GCA and when temporal artery biopsy for diagnostic purposes will be obsolete, establishing such models will not be feasible. Thus, new in vitro models are required for functional studies in this field.
Bioengineered arteries: Bioengineered blood vessels have attracted the scientific community’s interest in the last decade in an attempt to establish in vitro models for studying cardiovascular diseases and reducing the use of laboratory animals [230]. Along this line, several vasculature models have been suggested for constructing vessels of different sizes. A bioartificial artery model for large arteries has been described, implementing a collagen type I matrix that served as a scaffold for VSMC and EC lining [231]. The constructed bioartificial arteries were cultured for two days and were populated with DCs, monocytes, and macrophages to mimic the human vasculature in the initiating events of vasculitis. Even though the construct closely resembled human medium-sized arteries, the model exhibited several limitations, including the short period of viability, lack of patient-derived vascular cells, and the lack of following culture protocols underflow to mimic human contractile vasculature [231]. Another proposed model for the construction of human arteries incorporating 3D bioprinting approaches was described by Xu et al. This novel approach successfully produced vessels that closely mimic human medium-sized vessels but did not involve using immune cells to test this methodology in the setting of autoimmune disease research [232]. Moreover, patient-derived induced pluripotent stem cells are strongly suggested in constructing 3D in vitro models to retain the epigenetic, transcriptomic, and metabolic profile of the patient of origin [233,234]. The abovementioned efforts to construct bioengineered human arteries indicate that the ever-expanding tools and novel methodologies hold the potential for the generation of more physiologically relevant in vitro models of large vessel vasculitis in the foreseeable future.

5. Conclusions

GCA constitutes an ideal model to study acute and chronic DTH responses. In this review, we described, for academic purposes, the continuous inflammatory process as four distinct phases: (i) the initiation of the disease; (ii) the perpetuation and sustainability of the inflammatory response; (iii) the impaired tissue remodeling; and (iv) the chronic tissue injury phase (Figure 5). Despite the enormous progress over the past few years in understanding disease mechanisms, diagnosis, and application of targeted therapies, many unanswered questions still need to be answered. New queries have been generated, leading eventually to new unmet needs for the disease and, therefore, a revised research agenda. Among them, the generation of accurate animal models of the disease or the use of bioengineered artery models for obtaining more insights into pathogenetic mechanisms, the application of different omics technologies, and the integration of generated data in artificial intelligence-empowered platforms in pairs of samples during activity and inactivity of the disease, will undoubtedly provide new biomarkers with direct mechanistic involvement and composite characteristics. New treatment targets will be identified, and revised treatment selection tools will be developed, taking into consideration patient characteristics and future comorbidities.

Funding

This manuscript has partly been supported by the George and Victoria Karelias Foundation through donations to the Research Institute for Autoimmune Diseases (RISAD) and the Immunome project consortium for AutoInflammatory Disorders (ImmunAID) project, which has received funding from the European Union’s Horizon 2020 research and innovation program under Grant Agreement No. 779295.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. 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]
  2. Dejaco, C.; Brouwer, E.; Mason, J.C.; Buttgereit, F.; Matteson, E.L.; Dasgupta, B. Giant cell arteritis and polymyalgia rheumatica: Current challenges and opportunities. Nat. Rev. Rheumatol. 2017, 13, 578–592. [Google Scholar] [CrossRef]
  3. van der Geest, K.S.M.; Sandovici, M.; van Sleen, Y.; Sanders, J.S.; Bos, N.A.; Abdulahad, W.H.; Stegeman, C.A.; Heeringa, P.; Rutgers, A.; Kallenberg, C.G.M.; et al. Review: What Is the Current Evidence for Disease Subsets in Giant Cell Arteritis? Arthritis Rheumatol. 2018, 70, 1366–1376. [Google Scholar] [CrossRef]
  4. Buttgereit, F.; Matteson, E.L.; Dejaco, C. Polymyalgia Rheumatica and Giant Cell Arteritis. JAMA 2020, 324, 993–994. [Google Scholar] [CrossRef]
  5. Li, K.J.; Semenov, D.; Turk, M.; Pope, J. A meta-analysis of the epidemiology of giant cell arteritis across time and space. Arthritis Res. Ther. 2021, 23, 82. [Google Scholar] [CrossRef] [PubMed]
  6. Salvarani, C.; Crowson, C.S.; O’Fallon, W.M.; Hunder, G.G.; Gabriel, S.E. Reappraisal of the epidemiology of giant cell arteritis in Olmsted County, Minnesota, over a fifty-year period. Arthritis Rheum. 2004, 51, 264–268. [Google Scholar] [CrossRef] [PubMed]
  7. Kønig, E.B.; Stormly Hansen, M.; Foldager, J.; Siersma, V.; Loft, A.; Terslev, L.; Møller Døhn, U.; Radmer Jensen, M.; Wiencke, A.K.; Faber, C.; et al. Seasonal variation in biopsy-proven giant cell arteritis in Eastern Denmark from 1990–2018. Acta Ophthalmol. 2021, 99, 527–532. [Google Scholar] [CrossRef] [PubMed]
  8. Gokoffski, K.K.; Chatterjee, A.; Khaderi, S.K. Seasonal incidence of biopsy-proven giant cell arteritis: A 20-year retrospective study of the University of California Davis Medical System. Clin. Exp. Rheumatol. 2019, 37 (Suppl. 117), 90–97. [Google Scholar] [PubMed]
  9. Hoffman, G.S. Giant Cell Arteritis. Ann. Intern. Med. 2016, 165, Itc65–Itc80. [Google Scholar] [CrossRef] [PubMed]
  10. Tu, W.; Rao, S. Mechanisms Underlying T Cell Immunosenescence: Aging and Cytomegalovirus Infection. Front. Microbiol. 2016, 7, 2111. [Google Scholar] [CrossRef] [PubMed]
  11. Mohan, S.V.; Liao, Y.J.; Kim, J.W.; Goronzy, J.J.; Weyand, C.M. Giant cell arteritis: Immune and vascular aging as disease risk factors. Arthritis Res. Ther. 2011, 13, 231. [Google Scholar] [CrossRef]
  12. Calamia, K.T.; Hunder, G.G. Giant cell arteritis (temporal arteritis) presenting as fever of undetermined origin. Arthritis Rheum. 1981, 24, 1414–1418. [Google Scholar] [CrossRef]
  13. Diamantopoulos, A.P.; Haugeberg, G.; Lindland, A.; Myklebust, G. The fast-track ultrasound clinic for early diagnosis of giant cell arteritis significantly reduces permanent visual impairment: Towards a more effective strategy to improve clinical outcome in giant cell arteritis? Rheumatology 2016, 55, 66–70. [Google Scholar] [CrossRef]
  14. Soriano, A.; Muratore, F.; Pipitone, N.; Boiardi, L.; Cimino, L.; Salvarani, C. Visual loss and other cranial ischaemic complications in giant cell arteritis. Nat. Rev. Rheumatol. 2017, 13, 476–484. [Google Scholar] [CrossRef]
  15. Hayreh, S.S.; Podhajsky, P.A.; Zimmerman, B. Ocular manifestations of giant cell arteritis. Am. J. Ophthalmol. 1998, 125, 509–520. [Google Scholar] [CrossRef]
  16. Dejaco, C.; Ramiro, S.; Bond, M.; Bosch, P.; Ponte, C.; Mackie, S.L.; Bley, T.A.; Blockmans, D.; Brolin, S.; Bolek, E.C.; et al. EULAR recommendations for the use of imaging in large vessel vasculitis in clinical practice: 2023 update. Ann. Rheum. Dis. 2023; Online ahead of print. [Google Scholar] [CrossRef]
  17. Evangelatos, G.; Grivas, A.; Pappa, M.; Kouna, K.; Iliopoulos, A.; Fragoulis, G.E. Cranial giant cell arteritis mimickers: A masquerade to unveil. Autoimmun. Rev. 2022, 21, 103083. [Google Scholar] [CrossRef]
  18. Mehta, P.; Sattui, S.E.; van der Geest, K.S.M.; Brouwer, E.; Conway, R.; Putman, M.S.; Robinson, P.C.; Mackie, S.L. Giant Cell Arteritis and COVID-19: Similarities and Discriminators. A Systematic Literature Review. J. Rheumatol. 2021, 48, 1053–1059. [Google Scholar] [CrossRef]
  19. Castañeda, S.; Prieto-Peña, D.; Vicente-Rabaneda, E.F.; Triguero-Martínez, A.; Roy-Vallejo, E.; Atienza-Mateo, B.; Blanco, R.; González-Gay, M.A. Advances in the Treatment of Giant Cell Arteritis. J. Clin. Med. 2022, 11, 1588. [Google Scholar] [CrossRef] [PubMed]
  20. Maz, M.; Chung, S.A.; Abril, A.; Langford, C.A.; Gorelik, M.; Guyatt, G.; Archer, A.M.; Conn, D.L.; Full, K.A.; Grayson, P.C.; et al. 2021 American College of Rheumatology/Vasculitis Foundation Guideline for the Management of Giant Cell Arteritis and Takayasu Arteritis. Arthritis Rheumatol. 2021, 73, 1349–1365. [Google Scholar] [CrossRef] [PubMed]
  21. Koster, M.J.; Warrington, K.J. Giant cell arteritis: Pathogenic mechanisms and new potential therapeutic targets. BMC Rheumatol. 2017, 1, 2. [Google Scholar] [CrossRef] [PubMed]
  22. Cavazza, A.; Muratore, F.; Boiardi, L.; Restuccia, G.; Pipitone, N.; Pazzola, G.; Tagliavini, E.; Ragazzi, M.; Rossi, G.; Salvarani, C. Inflamed temporal artery: Histologic findings in 354 biopsies, with clinical correlations. Am. J. Surg. Pathol. 2014, 38, 1360–1370. [Google Scholar] [CrossRef]
  23. Kemp, A. Monozygotic twins with temporal arteritis and ophthalmic arteritis. Acta Ophthalmol. 1977, 55, 183–190. [Google Scholar] [CrossRef] [PubMed]
  24. Fietta, P.; Manganelli, P.; Zanetti, A.; Neri, T.M. Familial giant cell arteritis and polymyalgia rheumatica: Aggregation in 2 families. J. Rheumatol. 2002, 29, 1551–1555. [Google Scholar]
  25. Liozon, E.; Ouattara, B.; Rhaiem, K.; Ly, K.; Bezanahary, H.; Loustaud, V.; Letellier, P.; Drouet, M.; Vidal, E. Familial aggregation in giant cell arteritis and polymyalgia rheumatica: A comprehensive literature review including 4 new families. Clin. Exp. Rheumatol. 2009, 27, S89–S94. [Google Scholar]
  26. Lee, J.L.; Naguwa, S.M.; Cheema, G.S.; Gershwin, M.E. The geo-epidemiology of temporal (giant cell) arteritis. Clin. Rev. Allergy Immunol. 2008, 35, 88–95. [Google Scholar] [CrossRef] [PubMed]
  27. Cid, M.C.; Ercilla, G.; Vilaseca, J.; Sanmarti, R.; Villalta, J.; Ingelmo, M.; Urbano-Marquez, A. Polymyalgia rheumatica: A syndrome associated with HLA-DR4 antigen. Arthritis Rheum. 1988, 31, 678–682. [Google Scholar] [CrossRef] [PubMed]
  28. Combe, B.; Sany, J.; Le Quellec, A.; Clot, J.; Eliaou, J.F. Distribution of HLA-DRB1 alleles of patients with polymyalgia rheumatica and giant cell arteritis in a Mediterranean population. J. Rheumatol. 1998, 25, 94–98. [Google Scholar] [PubMed]
  29. Jacobsen, S.; Baslund, B.; Madsen, H.O.; Tvede, N.; Svejgaard, A.; Garred, P. Mannose-binding lectin variant alleles and HLA-DR4 alleles are associated with giant cell arteritis. J. Rheumatol. 2002, 29, 2148–2153. [Google Scholar] [PubMed]
  30. Martínez-Taboda, V.M.; Bartolome, M.J.; Lopez-Hoyos, M.; Blanco, R.; Mata, C.; Calvo, J.; Corrales, A.; Rodriguez-Valverde, V. HLA-DRB1 allele distribution in polymyalgia rheumatica and giant cell arteritis: Influence on clinical subgroups and prognosis. Semin. Arthritis Rheum. 2004, 34, 454–464. [Google Scholar] [CrossRef]
  31. Rauzy, O.; Fort, M.; Nourhashemi, F.; Alric, L.; Juchet, H.; Ecoiffier, M.; Abbal, M.; Adoue, D. Relation between HLA DRB1 alleles and corticosteroid resistance in giant cell arteritis. Ann. Rheum. Dis. 1998, 57, 380–382. [Google Scholar] [CrossRef]
  32. Richardson, J.E.; Gladman, D.D.; Fam, A.; Keystone, E.C. HLA-DR4 in giant cell arteritis: Association with polymyalgia rheumatica syndrome. Arthritis Rheum. 1987, 30, 1293–1297. [Google Scholar] [CrossRef]
  33. Salvarani, C.; Macchioni, P.; Zizzi, F.; Mantovani, W.; Rossi, F.; Castri, C.; Capozzoli, N.; Baricchi, R.; Boiardi, L.; Chiaravalloti, F.; et al. Epidemiologic and immunogenetic aspects of polymyalgia rheumatica and giant cell arteritis in northern Italy. Arthritis Rheum. 1991, 34, 351–356. [Google Scholar] [CrossRef]
  34. Weyand, C.M.; Hicok, K.C.; Hunder, G.G.; Goronzy, J.J. The HLA-DRB1 locus as a genetic component in giant cell arteritis. Mapping of a disease-linked sequence motif to the antigen binding site of the HLA-DR molecule. J. Clin. Investig. 1992, 90, 2355–2361. [Google Scholar] [CrossRef]
  35. Weyand, C.M.; Hunder, N.N.; Hicok, K.C.; Hunder, G.G.; Goronzy, J.J. HLA-DRB1 alleles in polymyalgia rheumatica, giant cell arteritis, and rheumatoid arthritis. Arthritis Rheum. 1994, 37, 514–520. [Google Scholar] [CrossRef] [PubMed]
  36. Dababneh, A.; Gonzalez-Gay, M.A.; Garcia-Porrua, C.; Hajeer, A.; Thomson, W.; Ollier, W. Giant cell arteritis and polymyalgia rheumatica can be differentiated by distinct patterns of HLA class II association. J. Rheumatol. 1998, 25, 2140–2145. [Google Scholar] [PubMed]
  37. Rodríguez-Pla, A.; Beaty, T.H.; Savino, P.J.; Eagle, R.C., Jr.; Seo, P.; Soloski, M.J. Association of a nonsynonymous single-nucleotide polymorphism of matrix metalloproteinase 9 with giant cell arteritis. Arthritis Rheum. 2008, 58, 1849–1853. [Google Scholar] [CrossRef] [PubMed]
  38. Salvarani, C.; Casali, B.; Boiardi, L.; Ranzi, A.; Macchioni, P.; Nicoli, D.; Farnetti, E.; Brini, M.; Portioli, I. Intercellular adhesion molecule 1 gene polymorphisms in polymyalgia rheumatica/giant cell arteritis: Association with disease risk and severity. J. Rheumatol. 2000, 27, 1215–1221. [Google Scholar] [PubMed]
  39. Amoli, M.M.; Shelley, E.; Mattey, D.L.; Garcia-Porrua, C.; Thomson, W.; Hajeer, A.H.; Ollier, W.E.; Gonzalez-Gay, M.A. Lack of association between intercellular adhesion molecule-1 gene polymorphisms and giant cell arteritis. J. Rheumatol. 2001, 28, 1600–1604. [Google Scholar] [PubMed]
  40. Carmona, F.D.; González-Gay, M.A.; Martín, J. Genetic component of giant cell arteritis. Rheumatology 2014, 53, 6–18. [Google Scholar] [CrossRef]
  41. Carmona, F.D.; Mackie, S.L.; Martín, J.E.; Taylor, J.C.; Vaglio, A.; Eyre, S.; Bossini-Castillo, L.; Castañeda, S.; Cid, M.C.; Hernández-Rodríguez, J.; et al. A large-scale genetic analysis reveals a strong contribution of the HLA class II region to giant cell arteritis susceptibility. Am. J. Hum. Genet. 2015, 96, 565–580. [Google Scholar] [CrossRef] [PubMed]
  42. Carmona, F.D.; Martín, J.; González-Gay, M.A. Genetics of vasculitis. Curr. Opin. Rheumatol. 2015, 27, 10–17. [Google Scholar] [CrossRef]
  43. 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]
  44. 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] [PubMed]
  45. Pazzola, G.; Boiardi, L.; Casali, B.; Farnetti, E.; Nicoli, D.; Pipitone, N.; Padovano, I.; Caruso, A.; Catanoso, M.; Salvarani, C. CC chemokine receptor 5 polymorphism in Italian patients with giant cell arteritis. Mod. Rheumatol. 2013, 23, 851–855. [Google Scholar] [CrossRef] [PubMed]
  46. Rueda, B.; Miranda-Filloy, J.A.; Martin, J.; Gonzalez-Gay, M.A. Association of CD24 gene polymorphisms with susceptibility to biopsy-proven giant cell arteritis. J. Rheumatol. 2008, 35, 850–854. [Google Scholar] [PubMed]
  47. Gonzalez-Gay, M.A.; Hajeer, A.H.; Dababneh, A.; Garcia-Porrua, C.; Amoli, M.M.; Thomson, W.; Ollier, W.E. Corticotropin releasing hormone promoter polymorphisms in giant cell arteritis and polymyalgia rheumatica. Clin. Exp. Rheumatol. 2002, 20, 133–138. [Google Scholar] [PubMed]
  48. Palomino-Morales, R.; Vazquez-Rodriguez, T.R.; Miranda-Filloy, J.A.; Martin, J.; Gonzalez-Gay, M.A. C-reactive protein gene polymorphisms in biopsy-proven giant cell arteritis from Northwestern Spain. J. Rheumatol. 2009, 36, 341–346. [Google Scholar] [CrossRef] [PubMed]
  49. Amoli, M.M.; Garcia-Porrua, C.; Llorca, J.; Ollier, W.E.; Gonzalez-Gay, M.A. Endothelial nitric oxide synthase haplotype associations in biopsy-proven giant cell arteritis. J. Rheumatol. 2003, 30, 2019–2022. [Google Scholar]
  50. Salvarani, C.; Casali, B.; Nicoli, D.; Farnetti, E.; Macchioni, P.; Catanoso, M.G.; Chen, Q.; Bajocchi, G.; Boiardi, L. Endothelial nitric oxide synthase gene polymorphisms in giant cell arteritis. Arthritis Rheum. 2003, 48, 3219–3223. [Google Scholar] [CrossRef] [PubMed]
  51. Morgan, A.W.; Robinson, J.I.; Barrett, J.H.; Martin, J.; Walker, A.; Babbage, S.J.; Ollier, W.E.; Gonzalez-Gay, M.A.; Isaacs, J.D. Association of FCGR2A and FCGR2A-FCGR3A haplotypes with susceptibility to giant cell arteritis. Arthritis Res. Ther. 2006, 8, R109. [Google Scholar] [CrossRef]
  52. Gonzalez-Gay, M.A.; Hajeer, A.H.; Dababneh, A.; Garcia-Porrua, C.; Amoli, M.M.; Llorca, J.; Ollier, W.E. Interferon-gamma gene microsatellite polymorphisms in patients with biopsy-proven giant cell arteritis and isolated polymyalgia rheumatica. Clin. Exp. Rheumatol. 2004, 22, S18–S20. [Google Scholar] [PubMed]
  53. Amoli, M.M.; Gonzalez-Gay, M.A.; Zeggini, E.; Salway, F.; Garcia-Porrua, C.; Ollier, W.E. Epistatic interactions between HLA-DRB1 and interleukin 4, but not interferon-gamma, increase susceptibility to giant cell arteritis. J. Rheumatol. 2004, 31, 2413–2417. [Google Scholar] [PubMed]
  54. Gonzalez-Gay, M.A.; Hajeer, A.H.; Dababneh, A.; Garcia-Porrua, C.; Mattey, D.L.; Amoli, M.M.; Thomson, W.; Ollier, W.E. IL-6 promoter polymorphism at position -174 modulates the phenotypic expression of polymyalgia rheumatica in biopsy-proven giant cell arteritis. Clin. Exp. Rheumatol. 2002, 20, 179–184. [Google Scholar] [PubMed]
  55. Salvarani, C.; Casali, B.; Farnetti, E.; Pipitone, N.; Nicoli, D.; Macchioni, P.; Cimino, L.; Bajocchi, G.; Catanoso, M.G.; Boiardi, L. Interleukin-6 promoter polymorphism at position -174 in giant cell arteritis. J. Rheumatol. 2005, 32, 2173–2177. [Google Scholar]
  56. Boiardi, L.; Casali, B.; Farnetti, E.; Pipitone, N.; Nicoli, D.; Macchioni, P.; Cimino, L.; Bajocchi, G.; Catanoso, M.G.; Pattacini, L.; et al. Interleukin-10 promoter polymorphisms in giant cell arteritis. Arthritis Rheum. 2006, 54, 4011–4017. [Google Scholar] [CrossRef] [PubMed]
  57. Rueda, B.; Roibas, B.; Martin, J.; Gonzalez-Gay, M.A. Influence of interleukin 10 promoter polymorphisms in susceptibility to giant cell arteritis in Northwestern Spain. J. Rheumatol. 2007, 34, 1535–1539. [Google Scholar] [PubMed]
  58. Márquez, A.; Hernández-Rodríguez, J.; Cid, M.C.; Solans, R.; Castañeda, S.; Fernández-Contreras, M.E.; Ramentol, M.; Morado, I.C.; Narváez, J.; Gómez-Vaquero, C.; et al. Influence of the IL17A locus in giant cell arteritis susceptibility. Ann. Rheum. Dis. 2014, 73, 1742–1745. [Google Scholar] [CrossRef] [PubMed]
  59. Palomino-Morales, R.J.; Vazquez-Rodriguez, T.R.; Torres, O.; Morado, I.C.; Castañeda, S.; Miranda-Filloy, J.A.; Callejas-Rubio, J.L.; Fernandez-Gutierrez, B.; Gonzalez-Gay, M.A.; Martin, J. Association between IL-18 gene polymorphisms and biopsy-proven giant cell arteritis. Arthritis Res. Ther. 2010, 12, R51. [Google Scholar] [CrossRef]
  60. Rodríguez-Rodríguez, L.; Castañeda, S.; Vázquez-Rodríguez, T.R.; Morado, I.C.; Gómez-Vaquero, C.; Marí-Alfonso, B.; Miranda-Filloy, J.A.; Narvaez, J.; Ortego-Centeno, N.; Vicente, E.F.; et al. Role of the rs6822844 gene polymorphism at the IL2-IL21 region in biopsy-proven giant cell arteritis. Clin. Exp. Rheumatol. 2011, 29, S12–S16. [Google Scholar]
  61. Rodríguez-Rodríguez, L.; Carmona, F.D.; Castañeda, S.; Miranda-Filloy, J.A.; Morado, I.C.; Narváez, J.; Marí-Alfonso, B.; Gómez-Vaquero, C.; Amigo-Díaz, E.; Ríos-Fernández, R.; et al. Role of rs1343151 IL23R and rs3790567 IL12RB2 polymorphisms in biopsy-proven giant cell arteritis. J. Rheumatol. 2011, 38, 889–892. [Google Scholar] [CrossRef]
  62. Márquez, A.; Solans, R.; Hernández-Rodríguez, J.; Cid, M.C.; Castañeda, S.; Ramentol, M.; Rodriguez-Rodriguez, L.; Narváez, J.; Blanco, R.; Ortego-Centeno, N.; et al. A candidate gene approach identifies an IL33 genetic variant as a novel genetic risk factor for GCA. PLoS ONE 2014, 9, e113476. [Google Scholar] [CrossRef] [PubMed]
  63. Márquez, A.; Solans, R.; Hernández-Rodríguez, J.; Cid, M.C.; Castañeda, S.; Ramentol, M.; Morado, I.C.; Rodriguez-Rodriguez, L.; Narváez, J.; Gómez-Vaquero, C.; et al. Analysis of two autoimmunity genes, IRAK1 and MECP2, in giant cell arteritis. Clin. Exp. Rheumatol. 2014, 32, S30–S33. [Google Scholar] [PubMed]
  64. Torres, O.; Palomino-Morales, R.; Vazquez-Rodriguez, T.R.; Castañeda, S.; Morado, I.C.; Miranda-Filloy, J.A.; Valero, F.; Callejas-Rubio, J.L.; Fernandez-Gutierrez, B.; Martin, J.; et al. Lack of association between IRF5 gene polymorphisms and biopsy-proven giant cell arteritis. J. Rheumatol. 2010, 37, 136–140. [Google Scholar] [CrossRef] [PubMed]
  65. Amoli, M.M.; Salway, F.; Zeggini, E.; Ollier, W.E.; Gonzalez-Gay, M.A. MCP-1 gene haplotype association in biopsy proven giant cell arteritis. J. Rheumatol. 2005, 32, 507–510. [Google Scholar] [PubMed]
  66. Gonzalez-Gay, M.A.; Rueda, B.; Vilchez, J.R.; Lopez-Nevot, M.A.; Robledo, G.; Ruiz, M.P.; Fernández, O.; Garcia-Porrua, C.; Gonzalez-Escribano, M.F.; Martín, J. Contribution of MHC class I region to genetic susceptibility for giant cell arteritis. Rheumatology 2007, 46, 431–434. [Google Scholar] [CrossRef] [PubMed]
  67. Amoli, M.M.; Garcia-Porrua, C.; Ollier, W.E.; Gonzalez-Gay, M.A. Lack of association between macrophage migration inhibitory factor gene polymorphism and giant cell arteritis. J. Rheumatol. 2005, 32, 74–76. [Google Scholar]
  68. Salvarani, C.; Casali, B.; Farnetti, E.; Pipitone, N.; Nicoli, D.; Macchioni, P.L.; Cimino, L.; Bajocchi, G.L.; Catanoso, M.G.; Pattacini, L.; et al. −463 G/A myeloperoxidase promoter polymorphism in giant cell arteritis. Ann. Rheum. Dis. 2008, 67, 485–488. [Google Scholar] [CrossRef]
  69. Martin, J.; Perez-Armengol, C.; Miranda-Filloy, J.A.; Vilchez, J.R.; Lopez-Nevot, M.A.; Garcia-Porrua, C.; Gonzalez-Gay, M.A. Lack of association of a functional -94ins/delATTG NFKB1 promoter polymorphism with susceptibility and clinical expression of biopsy-proven giant cell arteritis in northwest Spain. J. Rheumatol. 2006, 33, 285–288. [Google Scholar] [PubMed]
  70. Serrano, A.; Carmona, F.D.; Castañeda, S.; Solans, R.; Hernández-Rodríguez, J.; Cid, M.C.; Prieto-González, S.; Miranda-Filloy, J.A.; Rodríguez-Rodríguez, L.; Morado, I.C.; et al. Evidence of association of the NLRP1 gene with giant cell arteritis. Ann. Rheum. Dis. 2013, 72, 628–630. [Google Scholar] [CrossRef]
  71. Gonzalez-Gay, M.A.; Oliver, J.; Orozco, G.; Garcia-Porrua, C.; Lopez-Nevot, M.A.; Martin, J. Lack of association of a functional single nucleotide polymorphism of PTPN22, encoding lymphoid protein phosphatase, with susceptibility to biopsy-proven giant cell arteritis. J. Rheumatol. 2005, 32, 1510–1512. [Google Scholar]
  72. Serrano, A.; Márquez, A.; Mackie, S.L.; Carmona, F.D.; Solans, R.; Miranda-Filloy, J.A.; Hernández-Rodríguez, J.; Cid, M.C.; Castañeda, S.; Morado, I.C.; et al. Identification of the PTPN22 functional variant R620W as susceptibility genetic factor for giant cell arteritis. Ann. Rheum. Dis. 2013, 72, 1882–1886. [Google Scholar] [CrossRef]
  73. Palomino-Morales, R.; Vazquez-Rodriguez, T.R.; Morado, I.C.; Castañeda, S.; Ortego-Centeno, N.; Miranda-Filloy, J.A.; Lamas, J.R.; Martin, J.; Gonzalez-Gay, M.A. Lack of association between STAT4 gene polymorphism and biopsy-proven giant cell arteritis. J. Rheumatol. 2009, 36, 1021–1025. [Google Scholar] [CrossRef]
  74. Palomino-Morales, R.; Torres, O.; Vazquez-Rodriguez, T.R.; Morado, I.C.; Castañeda, S.; Callejas-Rubio, J.L.; Miranda-Filloy, J.A.; Fernandez-Gutierrez, B.; Martin, J.; Gonzalez-Gay, M.A. Association between toll-like receptor 4 gene polymorphism and biopsy-proven giant cell arteritis. J. Rheumatol. 2009, 36, 1501–1506. [Google Scholar] [CrossRef]
  75. Mattey, D.L.; Hajeer, A.H.; Dababneh, A.; Thomson, W.; González-Gay, M.A.; García-Porrúa, C.; Ollier, W.E. Association of giant cell arteritis and polymyalgia rheumatica with different tumor necrosis factor microsatellite polymorphisms. Arthritis Rheum. 2000, 43, 1749–1755. [Google Scholar] [CrossRef] [PubMed]
  76. Gonzalez-Gay, M.A.; Di Giovine, F.S.; Silvestri, T.; Amoli, M.M.; Garcia-Porrua, C.; Thomson, W.; Ollier, W.E.; Hajeer, A.H. Lack of association between IL-1 cluster and TNF-alpha gene polymorphisms and giant cell arteritis. Clin. Exp. Rheumatol. 2002, 20, 431. [Google Scholar] [PubMed]
  77. Torres, O.; Palomino-Morales, R.; Vazquez-Rodriguez, T.R.; Castañeda, S.; Morado, I.C.; Miranda-Filloy, J.A.; Ortego-Centeno, N.; Gonzalez-Alvaro, I.; Fernandez-Gutierrez, B.; Martin, J.; et al. Lack of association between TRAF1/C5 gene polymorphisms and biopsy-proven giant cell arteritis. J. Rheumatol. 2010, 37, 131–135. [Google Scholar] [CrossRef] [PubMed]
  78. Boiardi, L.; Casali, B.; Nicoli, D.; Farnetti, E.; Chen, Q.; Macchioni, P.; Catanoso, M.G.; Pulsatelli, L.; Meliconi, R.; Salvarani, C. Vascular endothelial growth factor gene polymorphisms in giant cell arteritis. J. Rheumatol. 2003, 30, 2160–2164. [Google Scholar] [PubMed]
  79. Rueda, B.; Lopez-Nevot, M.A.; Lopez-Diaz, M.J.; Garcia-Porrua, C.; Martín, J.; Gonzalez-Gay, M.A. A functional variant of vascular endothelial growth factor is associated with severe ischemic complications in giant cell arteritis. J. Rheumatol. 2005, 32, 1737–1741. [Google Scholar] [PubMed]
  80. Enjuanes, A.; Benavente, Y.; Hernández-Rodríguez, J.; Queralt, C.; Yagüe, J.; Jares, P.; de Sanjosé, S.; Campo, E.; Cid, M.C. Association of NOS2 and potential effect of VEGF, IL6, CCL2 and IL1RN polymorphisms and haplotypes on susceptibility to GCA—A simultaneous study of 130 potentially functional SNPs in 14 candidate genes. Rheumatology 2012, 51, 841–851. [Google Scholar] [CrossRef] [PubMed]
  81. Ly, K.H.; Régent, A.; Tamby, M.C.; Mouthon, L. Pathogenesis of giant cell arteritis: More than just an inflammatory condition? Autoimmun. Rev. 2010, 9, 635–645. [Google Scholar] [CrossRef]
  82. Nagel, M.A.; White, T.; Khmeleva, N.; Rempel, A.; Boyer, P.J.; Bennett, J.L.; Haller, A.; Lear-Kaul, K.; Kandasmy, B.; Amato, M.; et al. Analysis of Varicella-Zoster Virus in Temporal Arteries Biopsy Positive and Negative for Giant Cell Arteritis. JAMA Neurol. 2015, 72, 1281–1287. [Google Scholar] [CrossRef]
  83. Pisapia, D.J.; Lavi, E. VZV, temporal arteritis, and clinical practice: False positive immunohistochemical detection due to antibody cross-reactivity. Exp. Mol. Pathol. 2016, 100, 114–115. [Google Scholar] [CrossRef] [PubMed]
  84. Bhatt, A.S.; Manzo, V.E.; Pedamallu, C.S.; Duke, F.; Cai, D.; Bienfang, D.C.; Padera, R.F.; Meyerson, M.; Docken, W.P. In search of a candidate pathogen for giant cell arteritis: Sequencing-based characterization of the giant cell arteritis microbiome. Arthritis Rheumatol. 2014, 66, 1939–1944. [Google Scholar] [CrossRef] [PubMed]
  85. Getz, T.M.; Hoffman, G.S.; Padmanabhan, R.; Villa-Forte, A.; Roselli, E.E.; Blackstone, E.; Johnston, D.; Pettersson, G.; Soltesz, E.; Svensson, L.G.; et al. Microbiomes of Inflammatory Thoracic Aortic Aneurysms Due to Giant Cell Arteritis and Clinically Isolated Aortitis Differ From Those of Non-Inflammatory Aneurysms. Pathog. Immun. 2019, 4, 105–123. [Google Scholar] [CrossRef]
  86. Hoffman, G.S.; Getz, T.M.; Padmanabhan, R.; Villa-Forte, A.; Clifford, A.H.; Funchain, P.; Sankunny, M.; Perry, J.D.; Blandford, A.; Kosmorsky, G.; et al. The Microbiome of Temporal Arteries. Pathog. Immun. 2019, 4, 21–38. [Google Scholar] [CrossRef] [PubMed]
  87. Czesnikiewicz-Guzik, M.; Lee, W.W.; Cui, D.; Hiruma, Y.; Lamar, D.L.; Yang, Z.Z.; Ouslander, J.G.; Weyand, C.M.; Goronzy, J.J. T cell subset-specific susceptibility to aging. Clin. Immunol. 2008, 127, 107–118. [Google Scholar] [CrossRef]
  88. Vallejo, A.N.; Weyand, C.M.; Goronzy, J.J. T-cell senescence: A culprit of immune abnormalities in chronic inflammation and persistent infection. Trends Mol. Med. 2004, 10, 119–124. [Google Scholar] [CrossRef]
  89. Sun, X.; Nguyen, T.; Achour, A.; Ko, A.; Cifello, J.; Ling, C.; Sharma, J.; Hiroi, T.; Zhang, Y.; Chia, C.W.; et al. Longitudinal analysis reveals age-related changes in the T cell receptor repertoire of human T cell subsets. J. Clin. Investig. 2022, 132, e158122. [Google Scholar] [CrossRef]
  90. Grolleau-Julius, A.; Harning, E.K.; Abernathy, L.M.; Yung, R.L. Impaired dendritic cell function in aging leads to defective antitumor immunity. Cancer Res. 2008, 68, 6341–6349. [Google Scholar] [CrossRef]
  91. Behmoaras, J.; Gil, J. Similarities and interplay between senescent cells and macrophages. J. Cell Biol. 2021, 220, e202010162. [Google Scholar] [CrossRef]
  92. Li, Y.; Liu, Y.; Strickland, F.M.; Richardson, B. Age-dependent decreases in DNA methyltransferase levels and low transmethylation micronutrient levels synergize to promote overexpression of genes implicated in autoimmunity and acute coronary syndromes. Exp. Gerontol. 2010, 45, 312–322. [Google Scholar] [CrossRef]
  93. Lee, H.Y.; Oh, B.H. Aging and arterial stiffness. Circ. J. 2010, 74, 2257–2262. [Google Scholar] [CrossRef]
  94. Graver, J.C.; Boots, A.M.H.; Haacke, E.A.; Diepstra, A.; Brouwer, E.; Sandovici, M. Massive B-Cell Infiltration and Organization Into Artery Tertiary Lymphoid Organs in the Aorta of Large Vessel Giant Cell Arteritis. Front. Immunol. 2019, 10, 83. [Google Scholar] [CrossRef] [PubMed]
  95. van Sleen, Y.; Graver, J.C.; Abdulahad, W.H.; van der Geest, K.S.M.; Boots, A.M.H.; Sandovici, M.; Brouwer, E. Leukocyte Dynamics Reveal a Persistent Myeloid Dominance in Giant Cell Arteritis and Polymyalgia Rheumatica. Front. Immunol. 2019, 10, 1981. [Google Scholar] [CrossRef] [PubMed]
  96. Iliou, A.; Argyropoulou, O.D.; Palamidas, D.A.; Karagiannakou, M.; Benaki, D.; Tsezou, K.; Vlachoyiannopoulos, P.G.; Mikros, E.; Tzioufas, A.G. NMR-based metabolomics in giant cell arteritis and polymyalgia rheumatica sequential sera differentiates active and inactive disease. Rheumatology 2023, kead590. [Google Scholar] [CrossRef] [PubMed]
  97. Deng, J.; Ma-Krupa, W.; Gewirtz, A.T.; Younge, B.R.; Goronzy, J.J.; Weyand, C.M. Toll-like receptors 4 and 5 induce distinct types of vasculitis. Circ. Res. 2009, 104, 488–495. [Google Scholar] [CrossRef] [PubMed]
  98. Krupa, W.M.; Dewan, M.; Jeon, M.S.; Kurtin, P.J.; Younge, B.R.; Goronzy, J.J.; Weyand, C.M. Trapping of misdirected dendritic cells in the granulomatous lesions of giant cell arteritis. Am. J. Pathol. 2002, 161, 1815–1823. [Google Scholar] [CrossRef] [PubMed]
  99. Ma-Krupa, W.; Jeon, M.S.; Spoerl, S.; Tedder, T.F.; Goronzy, J.J.; Weyand, C.M. Activation of arterial wall dendritic cells and breakdown of self-tolerance in giant cell arteritis. J. Exp. Med. 2004, 199, 173–183. [Google Scholar] [CrossRef]
  100. 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]
  101. Pryshchep, O.; Ma-Krupa, W.; Younge, B.R.; Goronzy, J.J.; Weyand, C.M. Vessel-specific Toll-like receptor profiles in human medium and large arteries. Circulation 2008, 118, 1276–1284. [Google Scholar] [CrossRef]
  102. Reitsema, R.D.; Hesselink, B.C.; Abdulahad, W.H.; van der Geest, K.S.M.; Brouwer, E.; Heeringa, P.; van Sleen, Y. Aberrant phenotype of circulating antigen presenting cells in giant cell arteritis and polymyalgia rheumatica. Front. Immunol. 2023, 14, 1201575. [Google Scholar] [CrossRef]
  103. Brack, A.; Geisler, A.; Martinez-Taboada, V.M.; Younge, B.R.; Goronzy, J.J.; Weyand, C.M. Giant cell vasculitis is a T cell-dependent disease. Mol. Med. 1997, 3, 530–543. [Google Scholar] [CrossRef]
  104. Weyand, C.M.; Goronzy, J.J. Medium- and large-vessel vasculitis. N. Engl. J. Med. 2003, 349, 160–169. [Google Scholar] [CrossRef]
  105. Samson, M.; Audia, S.; Fraszczak, J.; Trad, M.; Ornetti, P.; Lakomy, D.; Ciudad, M.; Leguy, V.; Berthier, S.; Vinit, J.; et al. Th1 and Th17 lymphocytes expressing CD161 are implicated in giant cell arteritis and polymyalgia rheumatica pathogenesis. Arthritis Rheum. 2012, 64, 3788–3798. [Google Scholar] [CrossRef] [PubMed]
  106. 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]
  107. Kaiko, G.E.; Horvat, J.C.; Beagley, K.W.; Hansbro, P.M. Immunological decision-making: How does the immune system decide to mount a helper T-cell response? Immunology 2008, 123, 326–338. [Google Scholar] [CrossRef]
  108. Weyand, C.M.; Younge, B.R.; Goronzy, J.J. IFN-γ and IL-17: The two faces of T-cell pathology in giant cell arteritis. Curr. Opin. Rheumatol. 2011, 23, 43–49. [Google Scholar] [CrossRef] [PubMed]
  109. Corbera-Bellalta, M.; García-Martínez, A.; Lozano, E.; Planas-Rigol, E.; Tavera-Bahillo, I.; Alba, M.A.; Prieto-González, S.; Butjosa, M.; Espígol-Frigolé, G.; Hernández-Rodríguez, J.; et al. Changes in biomarkers after therapeutic intervention in temporal arteries cultured in Matrigel: A new model for preclinical studies in giant-cell arteritis. Ann. Rheum. Dis. 2014, 73, 616–623. [Google Scholar] [CrossRef]
  110. Deng, J.; Younge, B.R.; Olshen, R.A.; Goronzy, J.J.; Weyand, C.M. Th17 and Th1 T-cell responses in giant cell arteritis. Circulation 2010, 121, 906–915. [Google Scholar] [CrossRef]
  111. Samson, M.; Greigert, H.; Ciudad, M.; Gerard, C.; Ghesquière, T.; Trad, M.; Corbera-Bellalta, M.; Genet, C.; Ouandji, S.; Cladière, C.; et al. Improvement of Treg immune response after treatment with tocilizumab in giant cell arteritis. Clin. Transl. Immunol. 2021, 10, e1332. [Google Scholar] [CrossRef]
  112. Kimura, A.; Kishimoto, T. IL-6: Regulator of Treg/Th17 balance. Eur. J. Immunol. 2010, 40, 1830–1835. [Google Scholar] [CrossRef]
  113. Samson, M.; Audia, S.; Janikashvili, N.; Ciudad, M.; Trad, M.; Fraszczak, J.; Ornetti, P.; Maillefert, J.F.; Miossec, P.; Bonnotte, B. Brief report: Inhibition of interleukin-6 function corrects Th17/Treg cell imbalance in patients with rheumatoid arthritis. Arthritis Rheum. 2012, 64, 2499–2503. [Google Scholar] [CrossRef]
  114. Miyabe, C.; Miyabe, Y.; Strle, K.; Kim, N.D.; Stone, J.H.; Luster, A.D.; Unizony, S. An expanded population of pathogenic regulatory T cells in giant cell arteritis is abrogated by IL-6 blockade therapy. Ann. Rheum. Dis. 2017, 76, 898–905. [Google Scholar] [CrossRef] [PubMed]
  115. Ciccia, F.; Rizzo, A.; Guggino, G.; Cavazza, A.; Alessandro, R.; Maugeri, R.; Cannizzaro, A.; Boiardi, L.; Iacopino, D.G.; Salvarani, C.; et al. Difference in the expression of IL-9 and IL-17 correlates with different histological pattern of vascular wall injury in giant cell arteritis. Rheumatology 2015, 54, 1596–1604. [Google Scholar] [CrossRef]
  116. Ohtsuki, S.; Wang, C.; Watanabe, R.; Zhang, H.; Akiyama, M.; Bois, M.C.; Maleszewski, J.J.; Warrington, K.J.; Berry, G.J.; Goronzy, J.J.; et al. Deficiency of the CD155-CD96 immune checkpoint controls IL-9 production in giant cell arteritis. Cell Rep. Med. 2023, 4, 101012. [Google Scholar] [CrossRef] [PubMed]
  117. 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] [PubMed]
  118. Nurieva, R.I.; Chung, Y. Understanding the development and function of T follicular helper cells. Cell. Mol. Immunol. 2010, 7, 190–197. [Google Scholar] [CrossRef] [PubMed]
  119. Samson, M.; Ly, K.H.; Tournier, B.; Janikashvili, N.; Trad, M.; Ciudad, M.; Gautheron, A.; Devilliers, H.; Quipourt, V.; Maurier, F.; et al. Involvement and prognosis value of CD8(+) T cells in giant cell arteritis. J. Autoimmun. 2016, 72, 73–83. [Google Scholar] [CrossRef] [PubMed]
  120. Banks, P.M.; Cohen, M.D.; Ginsburg, W.W.; Hunder, G.G. Immunohistologic and cytochemical studies of temporal arteritis. Arthritis Rheum. 1983, 26, 1201–1207. [Google Scholar] [CrossRef] [PubMed]
  121. Schaufelberger, C.; Andersson, R.; Nordborg, E.; Hansson, G.K.; Nordborg, C.; Wahlström, J. An uneven expression of T cell receptor V genes in the arterial wall and peripheral blood in giant cell arteritis. Inflammation 2008, 31, 372–383. [Google Scholar] [CrossRef]
  122. Wen, Z.; Shimojima, Y.; Shirai, T.; Li, Y.; Ju, J.; Yang, Z.; Tian, L.; Goronzy, J.J.; Weyand, C.M. NADPH oxidase deficiency underlies dysfunction of aged CD8+ Tregs. J. Clin. Investig. 2016, 126, 1953–1967. [Google Scholar] [CrossRef]
  123. Cid, M.C.; Campo, E.; Ercilla, G.; Palacin, A.; Vilaseca, J.; Villalta, J.; Ingelmo, M. Immunohistochemical analysis of lymphoid and macrophage cell subsets and their immunologic activation markers in temporal arteritis. Influence of corticosteroid treatment. Arthritis Rheum. 1989, 32, 884–893. [Google Scholar] [CrossRef]
  124. Lavignac, C.; Jauberteau-Marchan, M.O.; Liozon, E.; Vidal, E.; Catanzano, G.; Liozon, F. [Immunohistochemical study of lesions in Horton’s temporal arteritis before and during corticotherapy]. Rev. Med. Interne 1996, 17, 814–820. [Google Scholar] [CrossRef] [PubMed]
  125. Cid, M.C.; Cervera, R.; Font, J.; Lopez-Soto, A.; Pallarés, L.; Navarro, M.; Ingelmo, M. Late thrombotic events in patients with temporal arteritis and anticardiolipin antibodies. Clin. Exp. Rheumatol. 1990, 8, 359–363. [Google Scholar] [PubMed]
  126. Duhaut, P.; Berruyer, M.; Pinede, L.; Demolombe-Rague, S.; Loire, R.; Seydoux, D.; Dechavanne, M.; Ninet, J.; Pasquier, J. Anticardiolipin antibodies and giant cell arteritis: A prospective, multicenter case-control study. Groupe de Recherche sur l’Artérite à Cellules Géantes. Arthritis Rheum. 1998, 41, 701–709. [Google Scholar] [CrossRef]
  127. Liozon, E.; Roblot, P.; Paire, D.; Loustaud, V.; Liozon, F.; Vidal, E.; Jauberteau, M.O. Anticardiolipin antibody levels predict flares and relapses in patients with giant-cell (temporal) arteritis. A longitudinal study of 58 biopsy-proven cases. Rheumatology 2000, 39, 1089–1094. [Google Scholar] [CrossRef] [PubMed]
  128. Manna, R.; Latteri, M.; Cristiano, G.; Todaro, L.; Scuderi, F.; Gasbarrini, G. Anticardiolipin antibodies in giant cell arteritis and polymyalgia rheumatica: A study of 40 cases. Br. J. Rheumatol. 1998, 37, 208–210. [Google Scholar] [CrossRef]
  129. Alard, J.E.; Hillion, S.; Guillevin, L.; Saraux, A.; Pers, J.O.; Youinou, P.; Jamin, C. Autoantibodies to endothelial cell surface ATP synthase, the endogenous receptor for hsp60, might play a pathogenic role in vasculatides. PLoS ONE 2011, 6, e14654. [Google Scholar] [CrossRef]
  130. Régent, A.; Dib, H.; Ly, K.H.; Agard, C.; Tamby, M.C.; Tamas, N.; Weksler, B.; Federici, C.; Broussard, C.; Guillevin, L.; et al. Identification of target antigens of anti-endothelial cell and anti-vascular smooth muscle cell antibodies in patients with giant cell arteritis: A proteomic approach. Arthritis Res. Ther. 2011, 13, R107. [Google Scholar] [CrossRef]
  131. Baerlecken, N.T.; Linnemann, A.; Gross, W.L.; Moosig, F.; Vazquez-Rodriguez, T.R.; Gonzalez-Gay, M.A.; Martin, J.; Kötter, I.; Henes, J.C.; Melchers, I.; et al. Association of ferritin autoantibodies with giant cell arteritis/polymyalgia rheumatica. Ann. Rheum. Dis. 2012, 71, 943–947. [Google Scholar] [CrossRef]
  132. Régent, A.; Ly, K.H.; Blet, A.; Agard, C.; Puéchal, X.; Tamas, N.; Le-Jeunne, C.; Vidal, E.; Guillevin, L.; Mouthon, L. Contribution of antiferritin antibodies to diagnosis of giant cell arteritis. Ann. Rheum. Dis. 2013, 72, 1269–1270. [Google Scholar] [CrossRef]
  133. Ciccia, F.; Rizzo, A.; Maugeri, R.; Alessandro, R.; Croci, S.; Guggino, G.; Cavazza, A.; Raimondo, S.; Cannizzaro, A.; Iacopino, D.G.; et al. Ectopic expression of CXCL13, BAFF, APRIL and LT-β is associated with artery tertiary lymphoid organs in giant cell arteritis. Ann. Rheum. Dis. 2017, 76, 235–243. [Google Scholar] [CrossRef] [PubMed]
  134. van der Geest, K.S.; Abdulahad, W.H.; Chalan, P.; Rutgers, A.; Horst, G.; Huitema, M.G.; Roffel, M.P.; Roozendaal, C.; Kluin, P.M.; Bos, N.A.; et al. Disturbed B cell homeostasis in newly diagnosed giant cell arteritis and polymyalgia rheumatica. Arthritis Rheumatol. 2014, 66, 1927–1938. [Google Scholar] [CrossRef] [PubMed]
  135. van der Geest, K.S.; Abdulahad, W.H.; Rutgers, A.; Horst, G.; Bijzet, J.; Arends, S.; Roffel, M.P.; Boots, A.M.; Brouwer, E. Serum markers associated with disease activity in giant cell arteritis and polymyalgia rheumatica. Rheumatology 2015, 54, 1397–1402. [Google Scholar] [CrossRef] [PubMed]
  136. Graver, J.C.; Abdulahad, W.; van der Geest, K.S.M.; Heeringa, P.; Boots, A.M.H.; Brouwer, E.; Sandovici, M. Association of the CXCL9-CXCR3 and CXCL13-CXCR5 axes with B-cell trafficking in giant cell arteritis and polymyalgia rheumatica. J. Autoimmun. 2021, 123, 102684. [Google Scholar] [CrossRef] [PubMed]
  137. Cid, M.C.; Hoffman, M.P.; Hernández-Rodríguez, J.; Segarra, M.; Elkin, M.; Sánchez, M.; Vilardell, C.; García-Martínez, A.; Pla-Campo, M.; Grau, J.M.; et al. Association between increased CCL2 (MCP-1) expression in lesions and persistence of disease activity in giant-cell arteritis. Rheumatology 2006, 45, 1356–1363. [Google Scholar] [CrossRef] [PubMed]
  138. van Sleen, Y.; Wang, Q.; van der Geest, K.S.M.; Westra, J.; Abdulahad, W.H.; Heeringa, P.; Boots, A.M.H.; Brouwer, E. Involvement of Monocyte Subsets in the Immunopathology of Giant Cell Arteritis. Sci. Rep. 2017, 7, 6553. [Google Scholar] [CrossRef] [PubMed]
  139. 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]
  140. Roche, N.E.; Fulbright, J.W.; Wagner, A.D.; Hunder, G.G.; Goronzy, J.J.; Weyand, C.M. Correlation of interleukin-6 production and disease activity in polymyalgia rheumatica and giant cell arteritis. Arthritis. Rheum. 1993, 36, 1286–1294. [Google Scholar] [CrossRef]
  141. García-Martínez, A.; Hernández-Rodríguez, J.; Espígol-Frigolé, G.; Prieto-González, S.; Butjosa, M.; Segarra, M.; Lozano, E.; Cid, M.C. Clinical relevance of persistently elevated circulating cytokines (tumor necrosis factor alpha and interleukin-6) in the long-term followup of patients with giant cell arteritis. Arthritis Care Res. 2010, 62, 835–841. [Google Scholar] [CrossRef]
  142. Weyand, C.M.; Fulbright, J.W.; Hunder, G.G.; Evans, J.M.; Goronzy, J.J. Treatment of giant cell arteritis: Interleukin-6 as a biologic marker of disease activity. Arthritis Rheum. 2000, 43, 1041–1048. [Google Scholar] [CrossRef]
  143. Villiger, P.M.; Adler, S.; Kuchen, S.; Wermelinger, F.; Dan, D.; Fiege, V.; Bütikofer, L.; Seitz, M.; Reichenbach, S. Tocilizumab for induction and maintenance of remission in giant cell arteritis: A phase 2, randomised, double-blind, placebo-controlled trial. Lancet 2016, 387, 1921–1927. [Google Scholar] [CrossRef]
  144. Tanaka, T.; Narazaki, M.; Kishimoto, T. IL-6 in inflammation, immunity, and disease. Cold Spring Harb. Perspect. Biol. 2014, 6, a016295. [Google Scholar] [CrossRef] [PubMed]
  145. Bettelli, E.; Carrier, Y.; Gao, W.; Korn, T.; Strom, T.B.; Oukka, M.; Weiner, H.L.; Kuchroo, V.K. Reciprocal developmental pathways for the generation of pathogenic effector TH17 and regulatory T cells. Nature 2006, 441, 235–238. [Google Scholar] [CrossRef] [PubMed]
  146. O’Neill, L.; McCormick, J.; Gao, W.; Veale, D.J.; McCarthy, G.M.; Murphy, C.C.; Fearon, U.; Molloy, E.S. Interleukin-6 does not upregulate pro-inflammatory cytokine expression in an ex vivo model of giant cell arteritis. Rheumatol. Adv. Pract. 2019, 3, rkz011. [Google Scholar] [CrossRef] [PubMed]
  147. 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] [PubMed]
  148. Becher, B.; Tugues, S.; Greter, M. GM-CSF: From Growth Factor to Central Mediator of Tissue Inflammation. Immunity 2016, 45, 963–973. [Google Scholar] [CrossRef] [PubMed]
  149. 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]
  150. Foell, D.; Hernández-Rodríguez, J.; Sánchez, M.; Vogl, T.; Cid, M.C.; Roth, J. Early recruitment of phagocytes contributes to the vascular inflammation of giant cell arteritis. J. Pathol. 2004, 204, 311–316. [Google Scholar] [CrossRef]
  151. 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]
  152. 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]
  153. Nadkarni, S.; Dalli, J.; Hollywood, J.; Mason, J.C.; Dasgupta, B.; Perretti, M. Investigational analysis reveals a potential role for neutrophils in giant-cell arteritis disease progression. Circ. Res. 2014, 114, 242–248. [Google Scholar] [CrossRef] [PubMed]
  154. 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]
  155. Hanata, N.; Shoda, H.; Tsuchida, Y.; Nagafuchi, Y.; Fujio, K. Comment on: Neutrophil extracellular traps in giant cell arteritis biopsies: Presentation, localization and co-expression with inflammatory cytokines. Rheumatology 2022, 61, e154–e155. [Google Scholar] [CrossRef] [PubMed]
  156. Wang, H.; Sha, L.L.; Ma, T.T.; Zhang, L.X.; Chen, M.; Zhao, M.H. Circulating Level of Neutrophil Extracellular Traps Is Not a Useful Biomarker for Assessing Disease Activity in Antineutrophil Cytoplasmic Antibody-Associated Vasculitis. PLoS ONE 2016, 11, e0148197. [Google Scholar] [CrossRef]
  157. Chatelain, D.; Duhaut, P.; Schmidt, J.; Loire, R.; Bosshard, S.; Guernou, M.; Pellet, H.; Piette, J.C.; Sevestre, H.; Ducroix, J.P. Pathological features of temporal arteries in patients with giant cell arteritis presenting with permanent visual loss. Ann. Rheum. Dis. 2009, 68, 84–88. [Google Scholar] [CrossRef]
  158. Newby, A.C. Dual role of matrix metalloproteinases (matrixins) in intimal thickening and atherosclerotic plaque rupture. Physiol. Rev. 2005, 85, 1–31. [Google Scholar] [CrossRef]
  159. Cho, A.; Reidy, M.A. Matrix metalloproteinase-9 is necessary for the regulation of smooth muscle cell replication and migration after arterial injury. Circ. Res. 2002, 91, 845–851. [Google Scholar] [CrossRef]
  160. Locati, M.; Curtale, G.; Mantovani, A. Diversity, Mechanisms, and Significance of Macrophage Plasticity. Annu. Rev. Pathol. 2020, 15, 123–147. [Google Scholar] [CrossRef]
  161. Wagner, A.D.; Goronzy, J.J.; Weyand, C.M. Functional profile of tissue-infiltrating and circulating CD68+ cells in giant cell arteritis. Evidence for two components of the disease. J. Clin. Investig. 1994, 94, 1134–1140. [Google Scholar] [CrossRef]
  162. 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]
  163. Rittner, H.L.; Kaiser, M.; Brack, A.; Szweda, L.I.; Goronzy, J.J.; Weyand, C.M. Tissue-destructive macrophages in giant cell arteritis. Circ. Res. 1999, 84, 1050–1058. [Google Scholar] [CrossRef]
  164. Kaiser, M.; Weyand, C.M.; Björnsson, J.; Goronzy, J.J. Platelet-derived growth factor, intimal hyperplasia, and ischemic complications in giant cell arteritis. Arthritis Rheum. 1998, 41, 623–633. [Google Scholar] [CrossRef] [PubMed]
  165. Kaiser, M.; Younge, B.; Björnsson, J.; Goronzy, J.J.; Weyand, C.M. Formation of new vasa vasorum in vasculitis. Production of angiogenic cytokines by multinucleated giant cells. Am. J. Pathol. 1999, 155, 765–774. [Google Scholar] [CrossRef] [PubMed]
  166. van Sleen, Y.; Jiemy, W.F.; Pringle, S.; van der Geest, K.S.M.; Abdulahad, W.H.; Sandovici, M.; Brouwer, E.; Heeringa, P.; Boots, A.M.H. A Distinct Macrophage Subset Mediating Tissue Destruction and Neovascularization in Giant Cell Arteritis: Implication of the YKL-40/Interleukin-13 Receptor α2 Axis. Arthritis Rheumatol. 2021, 73, 2327–2337. [Google Scholar] [CrossRef] [PubMed]
  167. Li, Z.; Guo, J.; Bi, L. Role of the NLRP3 inflammasome in autoimmune diseases. Biomed. Pharmacother. 2020, 130, 110542. [Google Scholar] [CrossRef] [PubMed]
  168. van der Geest, K.S.M.; Sandovici, M.; Nienhuis, P.H.; Slart, R.; Heeringa, P.; Brouwer, E.; Jiemy, W.F. Novel PET Imaging of Inflammatory Targets and Cells for the Diagnosis and Monitoring of Giant Cell Arteritis and Polymyalgia Rheumatica. Front. Med. 2022, 9, 902155. [Google Scholar] [CrossRef]
  169. Rodríguez-Pla, A.; Bosch-Gil, J.A.; Rosselló-Urgell, J.; Huguet-Redecilla, P.; Stone, J.H.; Vilardell-Tarres, M. Metalloproteinase-2 and -9 in giant cell arteritis: Involvement in vascular remodeling. Circulation 2005, 112, 264–269. [Google Scholar] [CrossRef]
  170. Weyand, C.M.; Wagner, A.D.; Björnsson, J.; Goronzy, J.J. Correlation of the topographical arrangement and the functional pattern of tissue-infiltrating macrophages in giant cell arteritis. J. Clin. Investig. 1996, 98, 1642–1649. [Google Scholar] [CrossRef]
  171. Nikkari, S.T.; Höyhtyä, M.; Isola, J.; Nikkari, T. Macrophages contain 92-kd gelatinase (MMP-9) at the site of degenerated internal elastic lamina in temporal arteritis. Am. J. Pathol. 1996, 149, 1427–1433. [Google Scholar]
  172. Segarra, M.; García-Martínez, A.; Sánchez, M.; Hernández-Rodríguez, J.; Lozano, E.; Grau, J.M.; Cid, M.C. Gelatinase expression and proteolytic activity in giant-cell arteritis. Ann. Rheum. Dis. 2007, 66, 1429–1435. [Google Scholar] [CrossRef] [PubMed]
  173. Galis, Z.S.; Khatri, J.J. Matrix metalloproteinases in vascular remodeling and atherogenesis: The good, the bad, and the ugly. Circ. Res. 2002, 90, 251–262. [Google Scholar] [CrossRef]
  174. Rodríguez-Pla, A.; Martínez-Murillo, F.; Savino, P.J.; Eagle, R.C., Jr.; Seo, P.; Soloski, M.J. MMP-12, a novel matrix metalloproteinase associated with giant cell arteritis. Rheumatology 2009, 48, 1460–1461. [Google Scholar] [CrossRef] [PubMed]
  175. Piggott, K.; Biousse, V.; Newman, N.J.; Goronzy, J.J.; Weyand, C.M. Vascular damage in giant cell arteritis. Autoimmunity 2009, 42, 596–604. [Google Scholar] [CrossRef]
  176. Corbera-Bellalta, M.; Planas-Rigol, E.; Lozano, E.; Terrades-García, N.; Alba, M.A.; Prieto-González, S.; García-Martínez, A.; Albero, R.; Enjuanes, A.; Espígol-Frigolé, G.; et al. Blocking interferon γ reduces expression of chemokines CXCL9, CXCL10 and CXCL11 and decreases macrophage infiltration in ex vivo cultured arteries from patients with giant cell arteritis. Ann. Rheum. Dis. 2016, 75, 1177–1186. [Google Scholar] [CrossRef]
  177. 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]
  178. Salvarani, C.; Cantini, F.; Hunder, G.G. Polymyalgia rheumatica and giant-cell arteritis. Lancet 2008, 372, 234–245. [Google Scholar] [CrossRef]
  179. Lozano, E.; Segarra, M.; Corbera-Bellalta, M.; García-Martínez, A.; Espígol-Frigolé, G.; Plà-Campo, A.; Hernández-Rodríguez, J.; Cid, M.C. Increased expression of the endothelin system in arterial lesions from patients with giant-cell arteritis: Association between elevated plasma endothelin levels and the development of ischaemic events. Ann. Rheum. Dis. 2010, 69, 434–442. [Google Scholar] [CrossRef] [PubMed]
  180. Planas-Rigol, E.; Terrades-Garcia, N.; Corbera-Bellalta, M.; Lozano, E.; Alba, M.A.; Segarra, M.; Espígol-Frigolé, G.; Prieto-González, S.; Hernández-Rodríguez, J.; Preciado, S.; et al. Endothelin-1 promotes vascular smooth muscle cell migration across the artery wall: A mechanism contributing to vascular remodelling and intimal hyperplasia in giant-cell arteritis. Ann. Rheum. Dis. 2017, 76, 1624–1634. [Google Scholar] [CrossRef]
  181. Régent, A.; Ly, K.H.; Groh, M.; Khifer, C.; Lofek, S.; Clary, G.; Chafey, P.; Baud, V.; Broussard, C.; Federici, C.; et al. Molecular analysis of vascular smooth muscle cells from patients with giant cell arteritis: Targeting endothelin-1 receptor to control proliferation. Autoimmun. Rev. 2017, 16, 398–406. [Google Scholar] [CrossRef]
  182. Ly, K.H.; Régent, A.; Molina, E.; Saada, S.; Sindou, P.; Le-Jeunne, C.; Brézin, A.; Witko-Sarsat, V.; Labrousse, F.; Robert, P.Y.; et al. Neurotrophins are expressed in giant cell arteritis lesions and may contribute to vascular remodeling. Arthritis Res. Ther. 2014, 16, 487. [Google Scholar] [CrossRef] [PubMed]
  183. Coll-Vinent, B.; Vilardell, C.; Font, C.; Oristrell, J.; Hernández-Rodríguez, J.; Yagüe, J.; Urbano-Márquez, A.; Grau, J.M.; Cid, M.C. Circulating soluble adhesion molecules in patients with giant cell arteritis. Correlation between soluble intercellular adhesion molecule-1 (sICAM-1) concentrations and disease activity. Ann. Rheum. Dis. 1999, 58, 189–192. [Google Scholar] [CrossRef] [PubMed]
  184. Cid, M.C.; Hernández-Rodríguez, J.; Esteban, M.J.; Cebrián, M.; Gho, Y.S.; Font, C.; Urbano-Márquez, A.; Grau, J.M.; Kleinman, H.K. Tissue and serum angiogenic activity is associated with low prevalence of ischemic complications in patients with giant-cell arteritis. Circulation 2002, 106, 1664–1671. [Google Scholar] [CrossRef] [PubMed]
  185. Wen, Z.; Shen, Y.; Berry, G.; Shahram, F.; Li, Y.; Watanabe, R.; Liao, Y.J.; Goronzy, J.J.; Weyand, C.M. The microvascular niche instructs T cells in large vessel vasculitis via the VEGF-Jagged1-Notch pathway. Sci. Transl. Med. 2017, 9, eaal3322. [Google Scholar] [CrossRef] [PubMed]
  186. Amsen, D.; Helbig, C.; Backer, R.A. Notch in T Cell Differentiation: All Things Considered. Trends Immunol. 2015, 36, 802–814. [Google Scholar] [CrossRef] [PubMed]
  187. Piggott, K.; Deng, J.; Warrington, K.; Younge, B.; Kubo, J.T.; Desai, M.; Goronzy, J.J.; Weyand, C.M. Blocking the NOTCH pathway inhibits vascular inflammation in large-vessel vasculitis. Circulation 2011, 123, 309–318. [Google Scholar] [CrossRef]
  188. 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. 2023, 83, 342–350. [Google Scholar] [CrossRef] [PubMed]
  189. Wigerblad, G.; Kaplan, M.J. Neutrophil extracellular traps in systemic autoimmune and autoinflammatory diseases. Nat. Rev. Immunol. 2023, 23, 274–288. [Google Scholar] [CrossRef]
  190. Papayannopoulos, V.; Metzler, K.D.; Hakkim, A.; Zychlinsky, A. Neutrophil elastase and myeloperoxidase regulate the formation of neutrophil extracellular traps. J. Cell Biol. 2010, 191, 677–691. [Google Scholar] [CrossRef]
  191. Mistry, P.; Nakabo, S.; O’Neil, L.; Goel, R.R.; Jiang, K.; Carmona-Rivera, C.; Gupta, S.; Chan, D.W.; Carlucci, P.M.; Wang, X.; et al. Transcriptomic, epigenetic, and functional analyses implicate neutrophil diversity in the pathogenesis of systemic lupus erythematosus. Proc. Natl. Acad. Sci. USA 2019, 116, 25222–25228. [Google Scholar] [CrossRef]
  192. Nakazawa, D.; Tomaru, U.; Yamamoto, C.; Jodo, S.; Ishizu, A. Abundant neutrophil extracellular traps in thrombus of patient with microscopic polyangiitis. Front. Immunol. 2012, 3, 333. [Google Scholar] [CrossRef]
  193. Hu, S.C.; Yu, H.S.; Yen, F.L.; Lin, C.L.; Chen, G.S.; Lan, C.C. Neutrophil extracellular trap formation is increased in psoriasis and induces human β-defensin-2 production in epidermal keratinocytes. Sci. Rep. 2016, 6, 31119. [Google Scholar] [CrossRef] [PubMed]
  194. Sangaletti, S.; Tripodo, C.; Chiodoni, C.; Guarnotta, C.; Cappetti, B.; Casalini, P.; Piconese, S.; Parenza, M.; Guiducci, C.; Vitali, C.; et al. Neutrophil extracellular traps mediate transfer of cytoplasmic neutrophil antigens to myeloid dendritic cells toward ANCA induction and associated autoimmunity. Blood 2012, 120, 3007–3018. [Google Scholar] [CrossRef] [PubMed]
  195. Sule, G.; Kelley, W.J.; Gockman, K.; Yalavarthi, S.; Vreede, A.P.; Banka, A.L.; Bockenstedt, P.L.; Eniola-Adefeso, O.; Knight, J.S. Increased Adhesive Potential of Antiphospholipid Syndrome Neutrophils Mediated by β2 Integrin Mac-1. Arthritis Rheumatol. 2020, 72, 114–124. [Google Scholar] [CrossRef] [PubMed]
  196. Gorgoulis, V.; Adams, P.D.; Alimonti, A.; Bennett, D.C.; Bischof, O.; Bishop, C.; Campisi, J.; Collado, M.; Evangelou, K.; Ferbeyre, G.; et al. Cellular Senescence: Defining a Path Forward. Cell 2019, 179, 813–827. [Google Scholar] [CrossRef]
  197. Kumari, R.; Jat, P. Mechanisms of Cellular Senescence: Cell Cycle Arrest and Senescence Associated Secretory Phenotype. Front. Cell Dev. Biol. 2021, 9, 645593. [Google Scholar] [CrossRef]
  198. Evangelou, K.; Veroutis, D.; Paschalaki, K.; Foukas, P.G.; Lagopati, N.; Dimitriou, M.; Papaspyropoulos, A.; Konda, B.; Hazapis, O.; Polyzou, A.; et al. Pulmonary infection by SARS-CoV-2 induces senescence accompanied by an inflammatory phenotype in severe COVID-19: Possible implications for viral mutagenesis. Eur. Respir. J. 2022, 60, 2102951. [Google Scholar] [CrossRef]
  199. Evangelou, K.; Belogiannis, K.; Papaspyropoulos, A.; Petty, R.; Gorgoulis, V.G. Escape from senescence: Molecular basis and therapeutic ramifications. J. Pathol. 2023, 260, 649–665. [Google Scholar] [CrossRef]
  200. Kyriazopoulou, L.; Karpathiou, G.; Hatzimichael, E.; Peoc’h, M.; Papoudou-Bai, A.; Kanavaros, P. Autophagy and cellular senescence in classical Hodgkin lymphoma. Pathol. Res. Pract. 2022, 236, 153964. [Google Scholar] [CrossRef] [PubMed]
  201. 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]
  202. Evangelou, K.; Lougiakis, N.; Rizou, S.V.; Kotsinas, A.; Kletsas, D.; Muñoz-Espín, D.; Kastrinakis, N.G.; Pouli, N.; Marakos, P.; Townsend, P.; et al. Robust, universal biomarker assay to detect senescent cells in biological specimens. Aging Cell 2017, 16, 192–197. [Google Scholar] [CrossRef]
  203. Binet, F.; Cagnone, G.; Crespo-Garcia, S.; Hata, M.; Neault, M.; Dejda, A.; Wilson, A.M.; Buscarlet, M.; Mawambo, G.T.; Howard, J.P.; et al. Neutrophil extracellular traps target senescent vasculature for tissue remodeling in retinopathy. Science 2020, 369, 934. [Google Scholar] [CrossRef] [PubMed]
  204. Conte, M.S.; Desai, T.A.; Wu, B.; Schaller, M.; Werlin, E. Pro-resolving lipid mediators in vascular disease. J. Clin. Investig. 2018, 128, 3727–3735. [Google Scholar] [CrossRef] [PubMed]
  205. Serhan, C.N. Pro-resolving lipid mediators are leads for resolution physiology. Nature 2014, 510, 92–101. [Google Scholar] [CrossRef] [PubMed]
  206. Reátegui, E.; Jalali, F.; Khankhel, A.H.; Wong, E.; Cho, H.; Lee, J.; Serhan, C.N.; Dalli, J.; Elliott, H.; Irimia, D. Microscale arrays for the profiling of start and stop signals coordinating human-neutrophil swarming. Nat. Biomed. Eng. 2017, 1, 0094. [Google Scholar] [CrossRef]
  207. Dalli, J.; Colas, R.A.; Serhan, C.N. Novel n-3 immunoresolvents: Structures and actions. Sci. Rep. 2013, 3, 1940. [Google Scholar] [CrossRef]
  208. Motwani, M.P.; Colas, R.A.; George, M.J.; Flint, J.D.; Dalli, J.; Richard-Loendt, A.; De Maeyer, R.P.; Serhan, C.N.; Gilroy, D.W. Pro-resolving mediators promote resolution in a human skin model of UV-killed Escherichia coli-driven acute inflammation. JCI Insight 2018, 3, e94463. [Google Scholar] [CrossRef]
  209. Khan, B.; O’Laughlin, A.; Perelas, A.; Thatcher, T.H.; Jackson, P.D. Polyunsaturated Fatty Acid-Derived Lipid Mediators as Biomarkers in Critical Care. In Biomarkers in Trauma, Injury and Critical Care; Rajendram, R., Preedy, V.R., Patel, V.B., Eds.; Springer International Publishing: Cham, Switzerland, 2023; pp. 941–969. [Google Scholar]
  210. Sertoglu, E.; Yucel, C.; Balık, A.R.; Taşçı, C.; Bilge, S.; Ertuğrul, M.S.; Nazaroğlu, N.K.; Ozgurtas, T. Evaluation of erythrocyte membrane fatty-acid compositions in association with interleukin-6 levels in patients with COVID-19. Nutrition 2022, 96, 111581. [Google Scholar] [CrossRef]
  211. Chu, C.Q. Animal models for large vessel vasculitis—The unmet need. Rheumatol. Immunol. Res. 2023, 4, 4–10. [Google Scholar] [CrossRef]
  212. Brack, A.; Rittner, H.L.; Younge, B.R.; Kaltschmidt, C.; Weyand, C.M.; Goronzy, J.J. Glucocorticoid-mediated repression of cytokine gene transcription in human arteritis-SCID chimeras. J. Clin. Investig. 1997, 99, 2842–2850. [Google Scholar] [CrossRef]
  213. Kabeerdoss, J.; Thomas, M.; Goel, R.; Mohan, H.; Danda, S.; L, J.; Danda, D. High expression of S100 calgranulin genes in peripheral blood mononuclear cells from patients with Takayasu arteritis. Cytokine 2019, 114, 61–66. [Google Scholar] [CrossRef] [PubMed]
  214. Phillippi, J.A. On vasa vasorum: A history of advances in understanding the vessels of vessels. Sci. Adv. 2022, 8, eabl6364. [Google Scholar] [CrossRef] [PubMed]
  215. Gössl, M.; Rosol, M.; Malyar, N.M.; Fitzpatrick, L.A.; Beighley, P.E.; Zamir, M.; Ritman, E.L. Functional anatomy and hemodynamic characteristics of vasa vasorum in the walls of porcine coronary arteries. Anat. Rec. A Discov. Mol. Cell. Evol. Biol. 2003, 272, 526–537. [Google Scholar] [CrossRef]
  216. Egashira, K.; Zhao, Q.; Kataoka, C.; Ohtani, K.; Usui, M.; Charo, I.F.; Nishida, K.; Inoue, S.; Katoh, M.; Ichiki, T.; et al. Importance of monocyte chemoattractant protein-1 pathway in neointimal hyperplasia after periarterial injury in mice and monkeys. Circ. Res. 2002, 90, 1167–1172. [Google Scholar] [CrossRef] [PubMed]
  217. Isoda, K.; Shiigai, M.; Ishigami, N.; Matsuki, T.; Horai, R.; Nishikawa, K.; Kusuhara, M.; Nishida, Y.; Iwakura, Y.; Ohsuzu, F. Deficiency of interleukin-1 receptor antagonist promotes neointimal formation after injury. Circulation 2003, 108, 516–518. [Google Scholar] [CrossRef] [PubMed]
  218. Isoda, K.; Akita, K.; Isobe, S.; Niida, T.; Adachi, T.; Iwakura, Y.; Daida, H. Interleukin-1 receptor antagonist originating from bone marrowderived cells and non-bone marrow-derived cells helps to suppress arterial inflammation and reduce neointimal formation after injury. J. Atheroscler. Thromb. 2014, 21, 1208–1218. [Google Scholar] [CrossRef]
  219. Hillock-Watling, C.; Gotlieb, A.I. The pathobiology of perivascular adipose tissue (PVAT), the fourth layer of the blood vessel wall. Cardiovasc. Pathol. 2022, 61, 107459. [Google Scholar] [CrossRef]
  220. Nicklin, M.J.; Hughes, D.E.; Barton, J.L.; Ure, J.M.; Duff, G.W. Arterial inflammation in mice lacking the interleukin 1 receptor antagonist gene. J. Exp. Med. 2000, 191, 303–312. [Google Scholar] [CrossRef]
  221. Shepherd, J.; Nicklin, M.J. Elastic-vessel arteritis in interleukin-1 receptor antagonist-deficient mice involves effector Th1 cells and requires interleukin-1 receptor. Circulation 2005, 111, 3135–3140. [Google Scholar] [CrossRef]
  222. Ali, Z.A.; Alp, N.J.; Lupton, H.; Arnold, N.; Bannister, T.; Hu, Y.; Mussa, S.; Wheatcroft, M.; Greaves, D.R.; Gunn, J.; et al. Increased in-stent stenosis in ApoE knockout mice: Insights from a novel mouse model of balloon angioplasty and stenting. Arterioscler. Thromb. Vasc. Biol. 2007, 27, 833–840. [Google Scholar] [CrossRef]
  223. Matsuki, T.; Isoda, K.; Horai, R.; Nakajima, A.; Aizawa, Y.; Suzuki, K.; Ohsuzu, F.; Iwakura, Y. Involvement of tumor necrosis factor-alpha in the development of T cell-dependent aortitis in interleukin-1 receptor antagonist-deficient mice. Circulation 2005, 112, 1323–1331. [Google Scholar] [CrossRef] [PubMed]
  224. Yoshizaki, T.; Itoh, S.; Yamaguchi, S.; Numata, T.; Nambu, A.; Kimura, N.; Suto, H.; Okumura, K.; Sudo, K.; Yamaguchi, A.; et al. IL-25 exacerbates autoimmune aortitis in IL-1 receptor antagonist-deficient mice. Sci. Rep. 2019, 9, 17067. [Google Scholar] [CrossRef] [PubMed]
  225. Chen, Q.; Yang, W.; Gupta, S.; Biswas, P.; Smith, P.; Bhagat, G.; Pernis, A.B. IRF-4-binding protein inhibits interleukin-17 and interleukin-21 production by controlling the activity of IRF-4 transcription factor. Immunity 2008, 29, 899–911. [Google Scholar] [CrossRef] [PubMed]
  226. Dal Canto, A.J.; Swanson, P.E.; O’Guin, A.K.; Speck, S.H.; Virgin, H.W. IFN-gamma action in the media of the great elastic arteries, a novel immunoprivileged site. J. Clin. Investig. 2001, 107, R15–R22. [Google Scholar] [CrossRef] [PubMed]
  227. Presti, R.M.; Pollock, J.L.; Dal Canto, A.J.; O’Guin, A.K.; Virgin, H.W.t. Interferon gamma regulates acute and latent murine cytomegalovirus infection and chronic disease of the great vessels. J. Exp. Med. 1998, 188, 577–588. [Google Scholar] [CrossRef] [PubMed]
  228. O’Neill, L.; Rooney, P.; Molloy, D.; Connolly, M.; McCormick, J.; McCarthy, G.; Veale, D.J.; Murphy, C.C.; Fearon, U.; Molloy, E. Regulation of Inflammation and Angiogenesis in Giant Cell Arteritis by Acute-Phase Serum Amyloid A. Arthritis Rheumatol. 2015, 67, 2447–2456. [Google Scholar] [CrossRef] [PubMed]
  229. Conway, R.; O’Neill, L.; McCarthy, G.M.; Murphy, C.C.; Fabre, A.; Kennedy, S.; Veale, D.J.; Wade, S.M.; Fearon, U.; Molloy, E.S. Interleukin 12 and interleukin 23 play key pathogenic roles in inflammatory and proliferative pathways in giant cell arteritis. Ann. Rheum. Dis. 2018, 77, 1815–1824. [Google Scholar] [CrossRef] [PubMed]
  230. Pan, C.; Gao, Q.; Kim, B.S.; Han, Y.; Gao, G. The Biofabrication of Diseased Artery In Vitro Models. Micromachines 2022, 13, 326. [Google Scholar] [CrossRef]
  231. Han, J.W.; Shimada, K.; Ma-Krupa, W.; Johnson, T.L.; Nerem, R.M.; Goronzy, J.J.; Weyand, C.M. Vessel wall-embedded dendritic cells induce T-cell autoreactivity and initiate vascular inflammation. Circ. Res. 2008, 102, 546–553. [Google Scholar] [CrossRef]
  232. Xu, L.; Varkey, M.; Jorgensen, A.; Ju, J.; Jin, Q.; Park, J.H.; Fu, Y.; Zhang, G.; Ke, D.; Zhao, W.; et al. Bioprinting small diameter blood vessel constructs with an endothelial and smooth muscle cell bilayer in a single step. Biofabrication 2020, 12, 045012. [Google Scholar] [CrossRef]
  233. Naderi-Meshkin, H.; Cornelius, V.A.; Eleftheriadou, M.; Potel, K.N.; Setyaningsih, W.A.W.; Margariti, A. Vascular organoids: Unveiling advantages, applications, challenges, and disease modelling strategies. Stem Cell Res. Ther. 2023, 14, 292. [Google Scholar] [CrossRef] [PubMed]
  234. Markou, M.; Kouroupis, D.; Badounas, F.; Katsouras, A.; Kyrkou, A.; Fotsis, T.; Murphy, C.; Bagli, E. Tissue Engineering Using Vascular Organoids From Human Pluripotent Stem Cell Derived Mural Cell Phenotypes. Front. Bioeng. Biotechnol. 2020, 8, 278. [Google Scholar] [CrossRef] [PubMed]
Figure 1. A representative image of an 18F-fluorodeoxyglucose positron emission tomography/computed tomography (PET/CT) of a patient with giant cell arteritis and extracranial large vessel involvement. The image displays coronal (Panel (A)) and sagittal views (Panel (B)), highlighting robust tracer uptake within the arch of the aorta, which is shown by arrows (kindly provided by our collaborator Dr CD Anagnostopoulos (BRFAA)).
Figure 1. A representative image of an 18F-fluorodeoxyglucose positron emission tomography/computed tomography (PET/CT) of a patient with giant cell arteritis and extracranial large vessel involvement. The image displays coronal (Panel (A)) and sagittal views (Panel (B)), highlighting robust tracer uptake within the arch of the aorta, which is shown by arrows (kindly provided by our collaborator Dr CD Anagnostopoulos (BRFAA)).
Cells 13 00430 g001
Figure 2. A visual representation showing the four different histological patterns observed in giant cell arteritis. Upper left: transmural involvement; upper right: Inflammation limited to adventitia; lower left: small vessel vasculitis; and lower right: inflammation limited to vasa vasorum [22]. Created with BioRender.com (accessed on 24 December 2023).
Figure 2. A visual representation showing the four different histological patterns observed in giant cell arteritis. Upper left: transmural involvement; upper right: Inflammation limited to adventitia; lower left: small vessel vasculitis; and lower right: inflammation limited to vasa vasorum [22]. Created with BioRender.com (accessed on 24 December 2023).
Cells 13 00430 g002
Figure 3. Proposal for the identification of novel next-generation biomarkers in GCA that possess two characteristics: (i) they are tightly related to the underlying pathogenetic mechanism; and (ii) they are the outcome of integrative analysis of high-throughput assays during disease activity and inactivity. They may also be potentially used as activity indices, organ damage markers, and treatment selection tools.
Figure 3. Proposal for the identification of novel next-generation biomarkers in GCA that possess two characteristics: (i) they are tightly related to the underlying pathogenetic mechanism; and (ii) they are the outcome of integrative analysis of high-throughput assays during disease activity and inactivity. They may also be potentially used as activity indices, organ damage markers, and treatment selection tools.
Cells 13 00430 g003
Figure 4. (A) Representative picture of IL-6+ NETs in the TABs from GCA patients [151]. White arrows indicate the co-staining of MPO/citH3/IL-6. (B) Representative picture of senescent cells positive for IL-6 in the TABs from GCA. White arrows indicate the co-staining of GL13/IL-6. (C) Representative picture of senescent cells positive for MMP-9 in the TABs from GCA [188]. White arrows indicate co-staining of MMP-9/GL13. Blue: DAPI; green: (A) + (B) IL-6, (C) MMP9; red: (A) citrullinated H3, (C) vimentin; magenta: (A) MPO, (B) + (C) GL13/lipofuscin. Objective (A) + (B) 20×, (C) 40×. Scale bar: 25 μm.
Figure 4. (A) Representative picture of IL-6+ NETs in the TABs from GCA patients [151]. White arrows indicate the co-staining of MPO/citH3/IL-6. (B) Representative picture of senescent cells positive for IL-6 in the TABs from GCA. White arrows indicate the co-staining of GL13/IL-6. (C) Representative picture of senescent cells positive for MMP-9 in the TABs from GCA [188]. White arrows indicate co-staining of MMP-9/GL13. Blue: DAPI; green: (A) + (B) IL-6, (C) MMP9; red: (A) citrullinated H3, (C) vimentin; magenta: (A) MPO, (B) + (C) GL13/lipofuscin. Objective (A) + (B) 20×, (C) 40×. Scale bar: 25 μm.
Cells 13 00430 g004
Figure 5. Phases of inflammation and tissue injury in giant cell arteritis presented as sequential events. In most cases, these phases overlap.
Figure 5. Phases of inflammation and tissue injury in giant cell arteritis presented as sequential events. In most cases, these phases overlap.
Cells 13 00430 g005
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

Palamidas, D.A.; Chatzis, L.; Papadaki, M.; Gissis, I.; Kambas, K.; Andreakos, E.; Goules, A.V.; Tzioufas, A.G. Current Insights into Tissue Injury of Giant Cell Arteritis: From Acute Inflammatory Responses towards Inappropriate Tissue Remodeling. Cells 2024, 13, 430. https://doi.org/10.3390/cells13050430

AMA Style

Palamidas DA, Chatzis L, Papadaki M, Gissis I, Kambas K, Andreakos E, Goules AV, Tzioufas AG. Current Insights into Tissue Injury of Giant Cell Arteritis: From Acute Inflammatory Responses towards Inappropriate Tissue Remodeling. Cells. 2024; 13(5):430. https://doi.org/10.3390/cells13050430

Chicago/Turabian Style

Palamidas, Dimitris Anastasios, Loukas Chatzis, Maria Papadaki, Ilias Gissis, Konstantinos Kambas, Evangelos Andreakos, Andreas V. Goules, and Athanasios G. Tzioufas. 2024. "Current Insights into Tissue Injury of Giant Cell Arteritis: From Acute Inflammatory Responses towards Inappropriate Tissue Remodeling" Cells 13, no. 5: 430. https://doi.org/10.3390/cells13050430

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