Next Article in Journal
Quantification of Liver Fat Content with the iATT Algorithm: Correlation with Controlled Attenuation Parameter
Next Article in Special Issue
Use of Anti-Thrombotic Drugs and In-Hospital Mortality in Acute Aortic Dissection Patients
Previous Article in Journal
Temporal Trends in the Prevalence, Treatment and Outcomes of Patients with Acute Coronary Syndrome at High Bleeding Risk
Previous Article in Special Issue
Bicuspid Aortic Valve in Children and Adolescents: A Comprehensive Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Tracking an Elusive Killer: State of the Art of Molecular-Genetic Knowledge and Laboratory Role in Diagnosis and Risk Stratification of Thoracic Aortic Aneurysm and Dissection

1
Department of Experimental and Clinical Medicine, University of Florence—Atherothrombotic Diseases Unit, Careggi University Hospital, 50134 Florence, Italy
2
Department of Cardiology, CMSR, 36077 Altavilla Vicentina, Italy
3
Reference Regional (Tuscany) Center for Marfan Syndrome and Related Disorders, Largo Brambilla 3, 50134 Florence, Italy
*
Author to whom correspondence should be addressed.
Diagnostics 2022, 12(8), 1785; https://doi.org/10.3390/diagnostics12081785
Submission received: 9 June 2022 / Revised: 12 July 2022 / Accepted: 19 July 2022 / Published: 22 July 2022
(This article belongs to the Special Issue Thoracic Aortic Disease: From Bench to Bedside)

Abstract

:
The main challenge in diagnosing and managing thoracic aortic aneurysm and dissection (TAA/D) is represented by the early detection of a disease that is both deadly and “elusive”, as it generally grows asymptomatically prior to rupture, leading to death in the majority of cases. Gender differences exist in aortic dissection in terms of incidence and treatment options. Efforts have been made to identify biomarkers that may help in early diagnosis and in detecting those patients at a higher risk of developing life-threatening complications. As soon as the hereditability of the TAA/D was demonstrated, several genetic factors were found to be associated with both the syndromic and non-syndromic forms of the disease, and they currently play a role in patient diagnosis/prognosis and management-guidance purposes. Likewise, circulating biomarker could represent a valuable resource in assisting the diagnosis, and several studies have attempted to identify specific molecules that may help with risk stratification outside the emergency department. Even if promising, those data lack specificity/sensitivity, and, in most cases, they need more testing before entering the “clinical arena”. This review summarizes the state of the art of the laboratory in TAA/D diagnostics, with particular reference to the current and future role of molecular-genetic testing.

1. Introduction

Despite the considerable advancements pursued over the past decades in pathophysiology knowledge and diagnostic imaging techniques for thoracic aortic aneurysm (TAA), plus the more consistent surveillance programs after diagnosis, it remains a life-threatening and still “subtle” disease for which the true epidemiology is hard to be determined. Ninety-five percent of TAA patients do not show any symptoms unless an acute aortic event occurs [1]. Thus, the estimated prevalence remains at 6–10 cases/100,000 patients/year, with ascending TAAs being more common compared to descending TAAs (60% vs. 35%) [2]. The most feared consequences of TAA are represented by aortic dissections/ruptures, the most common of the catastrophic events affecting the aorta [3]. In this respect too, the true incidence of aortic dissection is not easy to assess, as hospital-based reports clearly do not account for pre-admission deaths, which are known to involve a substantial proportion of TAA individuals [4]. Gender differences in aortic dissection were observed in a recent Swedish population-based study, estimating a decreasing incidence in men during 15 years. Differently, in women, the incidence did not show a significant variation. Moreover, fewer women were treated with thoracic endovascular aortic repair (TEVAR), and they showed a higher postoperative mortality when compared to men [5].
TAA has traditionally been divided based on the presence of extra-aortic clinical manifestations—syndromic TAA—or their absence—non-syndromic TAA. Syndromic TAA patients present with systemic features reflecting the presence of diseases involving connective tissue disorders such as Marfan syndrome (MFS), Loeys-Dietz syndrome (LDS), vascular Ehlers-Danlos syndrome (vEDS), and arterial tortuosity syndrome [6]. Bicuspid aortic valve (BAV) represents another condition often found in association with TAA, which is currently considered an independent risk factor for its development, with the aorta being dilated in some cases, even in the presence of a normally functioning BAV and/or regardless of physical activity, as has been recently assessed in pediatric patients [7,8]. Non-syndromic TAAs account for 95% of all TAA cases [6] and are further classified as sporadic or familial, in the case of at least one of the first-degree family members being affected [9]. The different presentations of TAA share similarities with regards to the molecular pathophysiology underlying dilatation development (impaired extracellular matrix (ECM)), collagen homeostasis, alteration of the TGF-β signaling pathways, disruption of smooth muscle and cytoskeletal apparatus, and, even if with different penetrance, a significant heritability [10].
According to the ESC (European Society of Cardiology) guidelines, TAA presentation has an impact on management, measures of intervention, and therapeutic strategies, as aortic growth speed and progression vary between syndromic and non-syndromic cases and also between sporadic and familial ones [11]. Thus, early identification of asymptomatic patients is crucial to allow timely monitoring and management strategies that could potentially prevent the progression of TAA and TAD [6]. This is especially important in non-syndromic cases in which the lack of distinct clinical manifestations (involving musculoskeletal or ocular signs) may delay detection. From the laboratory point of view, given that syndromic/non-syndromic TAA are most often inherited in an autosomal dominant pattern, molecular-genetic analysis represents a most valuable tool allowing early identification of affected individual as well as those subjects at higher risk of developing adverse outcomes in terms of aneurysm-growth speed and dissection [12]. This is particularly relevant for TAAs involving the ascending aorta, whose etiology is predominantly linked to a crucial genetic component. In addition, easily detectable and specific circulating biomarkers might further improve diagnosis and overall management of the disease. This review will address the state of the art of TAA’s pathophysiological drivers and related current laboratory tests, as well as genetic approaches and their major implications (most particularly regarding novel technologies/strategies) in supporting the detection and management of a condition with features (slow, gradual, and painless aneurysm formation, usually “accidentally” diagnosed via imaging study carried out for another purpose), that have made it earn, over the years, the sinister reputation of a “silent killer” [13].

2. Drivers of TAA Formation: A Constant Journey through Gene Discovery

The complexity and heterogeneity of TAA characteristics, syndromic presentation, and/or progression is the consequence of multiple but unique cellular and molecular-genetic mechanisms underlying its development, which often result in similar clinical presentation [14]. As familial-aggregation studies have suggested, more than 20% of patients have at least one first-degree family member with an arterial aneurysm, basically defining an increased risk for relatives of the affected individuals [15]. The first clue about the heritability of the trait is derived from case-control studies comparing the prevalence of thoracic aortic aneurysms, thoracic aortic dissections, and sudden death in first-degree relatives of patients referred for thoracic aortic surgery [16], identifying a higher risk for developing those diseases in the proband first-degree relatives with respect to the control groups (with relative risks of 1.8, 10.9, and 1.8 in proband fathers, brothers, and sisters, respectively). More evidence on genetic factors contributing to TAA development was provided by an analysis of a database comprising 598 patients evaluated for TAA in the United States [17], which showed a faster growth rate of aortic aneurysm in patients with familial cases with respect to the sporadic ones, with a younger age of presentation. In addition, pedigrees also showed different patterns of inheritance (autosomal dominant, X-linked, autosomal recessive). The role of genetic factors in causing TAA was further confirmed by more recent studies as well, analyzing different type of aneurysms [18]. It was mainly through genetic and animal models’ studies that the combination of disrupted/altered cellular processes driving the TAA formation were elucidated as well as the specific associated genes (Table 1). In this regard, it has to be noticed, which some causative genes exert an overlapping effect on both syndromic and non-syndromic TAAs, even if these two conditions have traditionally been considered as distinct entities (e.g., ACTA2, SMAD3) (Table 1) [6].

2.1. Extracellular Matrix Components

Among the genes causing and/or influencing TAA development, those codifying the ECM components are always mentioned first due to the amount of data that was collected over the years through animal studies, providing evidence of their impact on maintaining the structural integrity of the aortic wall. Those components are in close relationship and represent key factors, upstream/downstream, and intermediate elements of cellular pathways with impairment that has been demonstrated to have consequences in aneurysm development/predisposition in different ways, namely depletion of the elastic lamina in the aortic wall, lengthening of the ascending aorta, impaired assembly of collagen and elastic fibers, and altered TGF-β signaling (Figure 1).
The FBN1 gene (15q21.1) encodes the key component of extracellular microfibrils fibrillin-1, with a major role in elastin (encoded by ELN gene, 7q11.23) assembly and support by promoting adhesion to vascular smooth muscle cells (VSMCs) through interaction with lysyl oxidase (encoded by LOX gene, 5q23.1) and fibronectin [45]. Robust data are available on TAA-causing variants, which were found to be responsible of protein synthesis/secretion’s impairment or incorporation of mutant fibrillin in the microfibrillar architecture [46]. FBN1 represents the causative gene of Marfan syndrome [47] in syndromic TAA (Table 1), but mutations involving this gene were found in sporadic, non-syndromic TAA as well [25]; besides, a large Whole Exome Sequencing (WES) performed on syndromic and non-syndromic TAAD reported FBN1 as the most mutated gene of the cohort [26].
As mentioned, fibrillin 1 regulates the assembly of elastin, a protein that, when dysfunctional and/or depleted, has been found in association with TAA, even if additional mechanisms are required to initiate the dilatation development (altered integrins signaling, focal adhesion) [46]. Mutations involving ELN gene are causative of cutis laxa, which has an association with aortic dilatation that was found in 30–50% of patients [22]. Regarding the non-syndromic presentation of TAA, a triplication around the elastin gene was found to segregate in a family with supravalvular non-syndromic aortic aneurysm and in which the diagnosis of cutis laxa was excluded [23].
Stabilization and assembly of elastin is also regulated by the enzymatic activity of the lysyl oxidase protein, encoded by the LOX gene (5q23.1), catalyzing the lysin residues’ oxidation and those crosslinking reactions required for the stability of the elastin molecules [48]. Inactivating mutations involving the LOX gene were found to be causative of TAA development in patients with MFS, familial TAA and dissection (FTAAD), and BAV, through mechanisms that remain to be entirely clarified [28,49].
Fibulin-4 (EFEMP2 11q13.1) and type III collagen (encoded by COL3A1 gene 2q32.2) represent substrates of the enzyme encoded by the LOX gene that were shown to promote TAA development as well, when impaired in response to mutations involving the two codifying genes. The matrix glycoprotein fibulin-4 functions as an enhancer of lysyl oxidase activity and as a recruiter of immature elastin molecules: when mutated, the disruption of the elastic fibers and collagens on the aortic wall drives the aneurysm formation, as shown in both human and mouse models [50]. A similar mechanism described for fibulin-4 results from mutations in COL3A1, the causative gene of vEDS, with a main function to protect against the catastrophic disruption of the aortic wall that may result from an impaired deposition and maturation of collagen [51], mostly in syndromic forms of TAA [20].
Other ECM key components with dysregulation that has been found to drive TAA formation are the microfibril-associated glycoprotein 2 (MFAP5 gene, 12p13.3) and biglycan (BGN, Xq28). Loss-of-function mutations in the first gene were shown to impair TGF-β and notch-1 signaling, as a consequence of the interaction’s loss with fibrillin-1 [52]. A likely pathogenic variant was identified in a patient with mild TAA restricted to the ascending aorta, BAV, and subtle craniofacial features consistent with a connective-tissue disorder [31]. BGN mutations cause syndromic forms of TAA [19]. Its function is once again related to an alteration of the TGF-β signaling pathways, in which it acts as negative regulator, increasing TGF-β bioavailability [53].

2.2. SMCs (Smooth Muscle Cells) Compartment

The aorta is, for the most part, constituted by populations of SMCs, and the maintenance of the contractile properties of their cellular components is highly controlled and regulated. Molecular studies have, in fact, demonstrated that the impairment at different levels of this strictly regulated system predispose individuals to TAA development. Among proteins participating in this cellular machinery, α-smooth muscle aorta encoded by ACTA2 gene (10q23.31) exerts an important function in maintaining contractility, and its depletion is, in turn, associated with a decrease ability of the monomer to assemble into polymeric filaments, eventually impairing the actin-myosin contractile unit [54]. Decades of study has proven mutations in this gene represent an important cause of familial and nonfamilial non-syndromic TAAD [55,56,57], accounting for 12–21% of TAAD.
Filamin A (FLNA gene, Xq28) is an anchoring cytoskeletal protein that, among its diverse functions, acts as a linker between actin filaments [58]. An 18.4% TAA frequency was found in a large systematic analysis of both pediatric and adult patients with periventricular nodular heterotopia carrying loss-of-function FLNA mutations [33] and, very recently, in a patient developing TAA with an history of systemic lupus erythematosus [59].
Myosin heavy chain 11 (MYH11 gene, 16p13.11) interacts with α-actin, controlling its state change and enabling the actin filaments binding. MYH11 is also traditionally classified as a mostly non-syndromic gene for TAA and was demonstrated to be associated with other manifestations such as aortic stiffness, an early hallmark of the disease [34,60]. However, it has to be noticed that MYH11 defects account for <1% of all non-syndromic TAAD, with an aorta diameter prior to dissection usually >5 cm [35]. Robust data support the idea of shared defects in FLNA, MYH11, and ACTA2 machinery, determining mechanical-strength disruption of the vascular walls and contractility-maintenance failure as significant causes of TAA progression, especially in non-syndromic cases [32].
The MYLK gene (3q21.1) encodes the myosin light chain kinase, regulating the actin-myosin interaction and phosphorylating myosin light-chains, and its most destructive mutations were once again mostly found in non-syndromic TAAD [9] (Figure 2).

2.3. TGF-β Signaling

A mutational repertoire in the genes coding for positive and negative regulators of TGF-β signaling has been reported in patients developing TAA in association with MFS and LDS, with those large volumes of animal studies research representing the first, and one of the major, contributions to our understanding of aneurysmal onset and growth [61]. TGF-β signaling, in fact, plays a critical role in a series of vascular cellular processes such as blood-vessel development and maintenance, positive regulation of contractile proteins’ expression, cell differentiation, proliferation, and homeostasis, and those mechanisms have been demonstrated to be dysregulated in all types of LDSs [61]. Different models have been proposed as the basis of aortic-wall dilatation, one of those suggesting a reduction in TGF-β signaling that would cause an impaired expression of contractile proteins, resembling the effect of mutations in ACTA2 and MYH11 genes [62]. Alternatively, LDS-causing mutations increase the VSMCs’ signaling capacity, resulting in a defective responsiveness to the TGF-β of cardiac neural crest-derived VSMCs, which are highly abundant in the proximal thoracic aorta and in excessive activation of TGF-β signaling [63,64]. TGF-β is secreted by many types of cells, including macrophages, as part of a large latent complex that consists of the mature TGF-β cytokine, a dimer of its processed latency associated peptide (LAP), and one of three latent TGF-β binding protein-isoforms (LTBP1, 3, or 4). The latter binds to ECM components such as fibronectin or microfibrils composed of fibrillin-1. Among the LTBP protein family, a deletion involving the LTBP1 gene (2p22.3) has been found to segregate in a three-generation family presenting with TAA [38], while a variant involving LTBP3 gene (11q13.1) was suggested to be predisposing to TAA/D development [65]. Upon release, TGF-β binds to its heterodimeric receptor, activating the phosphorylation of the SMAD2 and SMAD3 proteins, which transmit the signal to the nucleus via the association with SMAD4 and, in turn, activate gene transcription [66]. Upon LDS-causing mutations, those affecting elements of the TGF-β signaling pathways involve the receptor heterodimer, made of the two components codified by the TGFBR1 (9q22.33) and TGFBR2 (3p24.1) genes; …these mutations result in decreased kinase activity with a reduction in the transduction molecules levels codified by the SMAD2 (18q21.1) and SMAD3 (15q22.33) genes [46]. The SMAD2 and SMAD3 proteins belong, in fact, to the receptor-activated (R)-SMAD family, intracellular effectors of the canonical TGF-β signaling pathway, with activated ligands that include the TGF-β2 and TGF-β3 encoded by TGFB2 (1q41) and TGFB3 (14q24.3). Mutations involving those genes have been associated with different subtypes of LDS, sharing aneurysm formation as a common clinical manifestation and being characterized by the presence/absence of other systemic features such as aortic or arterial tortuosity, cleft palate, bifid uvula, mitral valve disease, skeletal overgrowth, and so on [39] as well as a different tendency to early aortic dissection. TGFBR1, TGFBR2 and SMAD3 mutations also account for up to 3%, 5% and 2%, respectively, of non-syndromic FTAAD [32] (Figure 3).

2.4. TAA in the Context of Bicuspid Aortic Valve and the Role of Proteases

Since approximately 40% of BAV patients are prone to develop ascending aortic dilatation, this congenital heart defect is currently considered as an independent risk factor for TAA [7,67]. As the two phenotypes frequently occur together, and given the autosomal pattern of inheritance with incomplete penetrance that has been proposed for BAV, hypotheses have been made about the pathophysiological mechanisms driving the valve anomaly, along with its more frequent complication that, among others, involves the potential role of genetic syndromic TAA-associated variants such as NOTCH1, ROBO4, SMAD6, ELN, FBN1, ACTA2, and LOX [24,68]. Metalloproteases (MMPs) have been hypothesized to participate in TAA development in the context of BAV, following the observation of a significant increase in MMP-2 levels associated with a reduction in TIMP-1 in BAV/TAA compared with TAA subjects, in the context of a normal tricuspid valve; a greater activity of MMP-2 and MMP-9 in aneurysms was associated with BAV, which could explain the higher prevalence of TAA in these patients [69,70]. Apart from BAV, the combination of altered levels of MMPs, ADAMTS, and TIMPs (the main proteases and inhibitors within the media controlling the ECM environment’s integrity and maintenance) are proven to actively contribute to the medial degeneration triggering TAA [71]. Studies on human and animal models led to the observation of increased expression of MMP-2 and MMP-9 in TAA intima and media [72] and higher levels of ADAMTS-1 and ADAMTS-4 in sporadic, ascending TAA tissues from human patients [73]. Despite the indisputable value of these observation in understanding TAA pathogenesis and their potential implications in diagnosis, it has to be noticed that those markers are not specific for the phenotype, and their levels are altered in several other processes such as AAA (MMP-2, MMP-9), cancer (MMP-9, TIMP-1, TIMP-2), renal disease (TIMP-2), and so on [74] and are currently not included in the recommended work-up for TAA diagnosis and management nor in the genetic screening.

3. Mechanisms of TAA Progression: The Dissection Menace

As the pathophysiological mechanisms underlying TAA onset in both its syndromic and non-syndromic presentation have been illustrated, and their specific molecular features continue to be unraveled thanks to the advancements in wide genetic-screening technologies, detection of the disease remains a clinical challenge. This is especially relevant with respect to its most catastrophic complications, rupture and dissection, leading to death in the great majority of patients without timely treatment [4,75]. In fact, once aortic dissection has occurred, mortality is 1–2% for each hour afterwards, resulting in a 48-h mortality of approximately 50% [76]. However, in the case of survival, serious complications may follow such as lethal malperfusion syndrome, aortic regurgitation, cardiac failure, and stroke [77]. Dissection represents a considerable diagnostic challenge for physicians due to the rarity of the condition and the characteristic symptomatology often mimicking other, more common diseases, determining a delay in diagnosis in >30% of cases [4]. Thus, the understanding of the pathophysiology, the key features, and the potential biochemical/molecular markers of TAA progression into aortic dissection has been crucial during the last decades to improve outcomes, for long-term prognosis, and eventually for patients’ risk-stratification purposes.

3.1. Pathophysiology and Risk Factors

The instability and the deteriorating integrity of the aortic wall may be due to being predisposed to inherited conditions (such as inherited connective tissue disorders) or can be acquired, as happens with atherosclerotic degeneration due to ageing. Two mechanisms have been proposed to initiate the dissection cascade: (1) in most cases, a tear in the intima exposes the medial layer to the pulsatile blood flow; and (2) in fewer cases, the rupture of the vasa vasorum leads to the weakening of the inner aortic wall [78]. In the first scenario, a false lumen derives from the progressive separation of the aortic wall layers, and its propagation leads to aortic rupture where the adventitia is disrupted: rupture quickly leads to exsanguination and death [79]. In the latter case, the bleeding results in intramural hematoma that may progress in aortic dissection. It has actually been hypothesized that a co-existence of these two conditions may, in turn, constitute a spectrum [80]. From a molecular point of view, dissection occurs as a consequence of the aortic-wall structure’s remodeling, due to inflammation and ECM-degradation processes. Once again, proteases exert an important role, since the infiltration of the activated macrophages and pro-inflammatory cytokines in the tunica media leads to an excessive production of MMP-1, MMP-9, and MMP-12 and to an imbalance between them and their inhibitors (TIMPs), which, in turn, results in the degradation of collagen and elastin fibers [81,82]. Wall remodeling is also maintained by VEGF-mediated neo-angiogenesis, as the production of VEGF (also functioning as a pro-inflammatory molecule) is increased in degraded medial layers [83].
Among classical risk factors associated with aortic dissection, namely older age, dyslipidemia, and increased levels of apolipoprotein A1, 80% of patients developing dissection have hypertension [84], which has a direct effect on the pathogenic mechanisms described above. Specifically, hypertension is demonstrated to promote a pro-inflammatory environment mainly by inducing macrophage recruitment and activation [85]; hypertensive patients, in fact, show high concentrations of VEGF, IL-6, MMP-2, and MMP-9 [86,87]. Other risk factors are recognized such as the male sex, a smoking habit, and the concurrence of connective tissue disorders such as MFS, LDS, vEDS, and BAV. Aortic dilatation is known to increase the risk of dissection, with the incidence complications reaching 30% at diameters > 60 mm [88]. Still, dilatation is proven not to be essential for developing a dissection, as ~60% of non-syndromic type A aortic dissection have diameters < 55 mm, while, in the absence of hypertension, MFS or BAV patients show a tendency to dissect at larger diameters [89,90]. Rare risk factors for aortic dissection are further represented by vascular inflammation due to autoimmune disorders such as Giant-cell arteritis, Takayasu Arteritis, Behçet disease, and systemic lupus erythematosus, while 1% to 5% of aortic dissections are secondary to aortitis [91].

3.2. Genetic Profiles of Dissection

To date, more than 30 TAAD-causative genes have been discovered (Table 1) and in the context of risk-prediction of TAA progression into potentially fatal dissection, it appears of primary interest to identify those genes or the specific type of variants/genetic profiles that, among others, are more prone to trigger and drive those processes eventually leading to sudden aortic-wall rupture. Marked in bold in Table 1, those genes were demonstrated to increase the risk of dissection at certain aortic sizes. Some of them represent causative genes of peculiar connective-tissue disorders described in association with TAA in its syndromic presentation. Pathogenic variants in FBN1, the MFS-causative gene (see Section 2.1), were found to increase the risk for Stanford type A and B dissection, even in the context of a normal or minimally dilated ascending aorta [92,93,94]. Indeed, a diagnosis of MFS is established in ~5% of patients with aortic dissection [95]. Haploinsufficiency, mainly resulting from truncating or splicing FBN1 mutations, is described as the leading mechanism behind the increased rate of aortic events [27]. Some overlapping cardiovascular clinical manifestations characterize MFS and LDS as basically reflecting a predisposition of patients affected by one or the other connective disorder to develop aneurysm and dissections of aorta and other arteries [92]. Implicated genes are TGFBR1, TGFBR2, and SMAD3, causing LDS type I and III, which have been associated with increased aortic risk of dissection at diameters < 50 mm [9]. TGFBR1 and TGFBR2 mutations’ carriers are often reported as a comparable clinical picture regarding presentation and natural history, even though clinical differences have actually been observed between the two populations of mutated subjects [96]. Regarding TGFBR1 families, a gender-based difference in survival has been observed, resulting in significantly better outcomes in women than in men. Differences were also observed in terms of aortic diameter, with TGBFR2 carriers dissecting at minimal aortic dilatation with respect to TGFBR1 carriers in which the ascending aorta diameter at the time of type A dissections was 50 mm. A more recent and large multicenter retrospective registry of patients with genetically triggered thoracic aortic disease reported the data of 441 subjects harboring mutations on the TGF-β receptor genes, somewhat confirming the previous observations in TGFBR2 mutations’ female carriers, that type A dissections of moderately dilated ascending aorta appeared more frequently than in males, which was not the case with TGFBR1, suggesting a more aggressive aortic disease in TGFBR2 patients, especially in women [97]. Together with a TGFBR2 mutation and the female sex, other features such as aortic tortuosity, hypertelorism, and translucent skin were found to be associated with an increased aortic dissection risk and may be taken into consideration in determining the optimal surgical timing (45 mm in the general population, lowered toward 40 in females with low body surface area, harboring a TGFBR2 mutation, and presenting extra-aortic features). The literature data support the role of SMAD3 as a dissection-predisposing gene [66,98,99,100], with mutations’ carriers having a cumulative risk of dissection or prophylactic surgical repair of 50% by age 50 and 85% by age 80. Interestingly, these subjects are characterized by a later onset of aortic events, possibly leading to a delayed diagnosis if compared, for instance, to MFS patients presenting with wider systemic features [101]. The early recognition of the disease, in this subset of patients, consequently lies in the family history of thoracic aortic disease (as a key element), so this observation remarks on the need to identify those subjects before dissection occurs. The majority (63%) of SMAD3 mutations are missense and reside in the MH2 domain, which regulates the oligomerization with SMAD2 or SMAD4 and the subsequent activation of transcription, with those variants being associated with earlier aortic events compared to truncating, non-sense, gene-disrupting ones [102]. Even if generally associated with a specific connective-tissue disorder, it must be reasserted that FBN1, TGFBR1, TGFBR2, SMAD3, and TGFB2 mutations also account for an additional 14% of non-syndromic familial TAA [40], in which the diagnosis of aortic disease may be complicated by the absence of peculiar systemic clinical features.
A vEDS-causing gene, COL3A1, is also among those referenced as dissection associated ones, due to the supporting literature data on population studies and case reports. The dissection was found to develop in different locations of the arterial tree, such as the abdominal aorta, as well as the iliac, coronary, and cervical arteries [103,104,105,106]. Concerning syndromic TAA in patients diagnosed as vEDS, few post-mortem cases were reported in 2010 [107], and 33 unrelated individuals or families were found to carry COL3A1 splicing mutations or small deletions partially removing splice-junctions sequences [108], and patients developing post-surgical or sudden aortic events are reported [109,110,111]. COL3A1 variants were additionally found to be associated with sporadic forms of TAAD in recent WES and case-control studies [112,113]. The type of variant involving the COL3A1 gene was also been suggested to correlate with the phenotype severity of vEDS; specifically, a subgroup of patients in a large European cohort bearing non-glycine missense and/or genetic variations at the C- and N-termini of type III procollagens was found to develop a later-onset and a milder phenotype with higher rates of aortic complications [114], while mutations at splice-donor sites were associated with higher mortality rates with respect to those involving the splice-acceptor sequences [115]. The literature data on animal models/human cohorts have identified a number of other genes, with mutations that were suggested to increase the risk of aortic dissection, such as EFEMP2 at the level of the ascending aorta [21], MYH11, ACTA2, and MAT2A in the thoracic aorta [43,116], and SLC2A10 at the aorta, as well as the arteries [44] LOX and PRKG1, with more limited data [28,117], and FOXE3 and MFAP5 as identified through WES studies [30,31] (see also Section 5).

4. Recommended Laboratory Workup for TAA/D Diagnosis and Risk Prediction

4.1. Medical History and Physical Examination

The 2014 ESC guidelines [11] stress the need to report any reference to a family history in the medical record in the initial evaluation along with the physical examination [118]. Silaschi and co-workers characterized some features that might be considered during the medical history’s collection as “red flags”, raising suspicion of acute aortic dissection, specifically: (a) known MFS or other connective tissue diseases; (b) family history; (c) known aortic valve disease; (d) known aortic aneurysm; and (e) previous aortic manipulation/surgery [76]. However, given that TAA patients are asymptomatic in the majority of cases, those few who present with premonitory symptoms have chest pain that should be ascribable to the presence of TAA where no other causes are found [76,119]. In the case of an aortic dissection occurring, however, the chest pain is characterized by a sudden onset and a more severe nature (sharp or “knife-like” pain, tearing, localized between the shoulder blades) often, but not invariably, manifesting after a strong physical or emotional strain [76,119]. The risk-assessment tool proposed by the 2010 North American guidelines is based on clinical data extracted from three groups of information, which are designated as the presence of (1) a “high-risk condition” (as MFS, family history of aortic disease); (2) “high-risk pain features” (severe chest pain); and (3) “high-risk examination features” (pulse deficit, aortic diastolic murmur). The resulting scoring system considers the mere number of those groups that are involved, ranging from 0 to 3, with the score being associated to an increased/decreased pre-test probability that influences, in turn, the diagnostic approach. Still, validation of this scoring system is still needed [120]. The diagnostic flow chart then combines the pre-test probabilities according to clinical data and the laboratory and imaging tests, as should be done in the clinical practice in emergency or chest-pain units [11].

4.2. “Traditional” Circulating Biomarkers for TAA/D

Biochemical markers currently do not play a major role in the overall diagnostic TAA flowchart and their support to diagnostic screening and decision-making, especially concerning time of intervention, is still limited. However, decades of studies have led to the identification and validation of a number of molecules that can assist physicians in: (a) identifying high-risk patients among those presenting with thoracic aortic aneurysm; (b) establishing prognostic stratification of individuals; and (c) evaluating aortic disease during the follow-up [121]. In addition, regarding the diagnostic workup described in the previous section, it has to be noticed that 5–13% of patients are classified at low risk for dissection and an even greater number of those who are classified at intermediate risk eventually turn out not to develop the complication [122]. Consequently, circulating biomarkers represent, in all respects, an appealing tool in assisting physicians, especially during the diagnostic algorithm proposed for aortic dissection. Among the potentially useful biomarkers, a number of studies have focused on D-Dimer (DD) evaluation, which became widely used in the clinical workup of suspected deep-vein thrombosis and pulmonary embolism due to its negative predictive value [123]. Its predictive power in identifying TAA patients at risk for developing dissection has been tested by several studies [124,125,126,127], showing DD to represent a highly sensitive yet largely unspecific biomarker, since DD levels increase in cancer infections, trauma, and surgery apart from deep-vein thrombosis, pulmonary embolism, and disseminated intravascular coagulopathy. However, negative DD could safely rule out aortic dissection, when associated with a sufficiently low pre-test probability of the disease [128]. DD might be used in prognosis prediction and in monitoring adverse events in patients in which the diagnosis of aortic dissection has been established [121].
Altered levels of metalloproteases and their inhibitors have been extensively studied in TAA samples (see Section 3.1), and their expression was also found to be different depending on the TAA etiology (atherosclerotic or non-atherosclerotic), size, growth rate, and location of the aneurysm (ascending or descending portion of the aorta) [129,130]. Concerning the potential role of these molecules as outcome predictors, preliminary data are available for MMP isoforms 1, 2, 3, 8, 9, 12, and 13. Among these, a low cutoff for plasma MMP8 has been correlated with ideal sensitivity and a negative predictive value for aortic dissection, suggesting a potential role in ruling out the occurrence of the disease. In addition, a combined use of MMP8 and DD has been proposed in ruling out aortic dissection in the diagnostic workup: as a matter of fact, the application of low cutoffs for both biomarkers was proven to be highly sensitive, and it allowed safe dissection’s being ruled out in 20% of patients [131]. A 2018 case-control study reported a close association between serum MMP9 and the existence of aortic aneurysms, suggesting a role for this isoform as a valuable marker for the discrimination of aortic aneurysm, especially for TAA [132]. Although suggestive, these data and those derived from MMP12, with levels that were found to be significantly increased in patients with Stanford type A aortic dissection with respect to coronary artery disease and control groups [133], are not yet sufficient to implement MMPs in clinical practice, so additional insights potentially coming from gene-expression studies are warranted [134].
Elastolytic process is augmented in TAA in which the elastin’s structure is modified, leading to increased levels of products of elastin degradation (soluble elastin fragments, sELAFs) and serum propeptide of type III procollagen, which were suggested as promising biomarkers for TAA [135]. Those were also found in patients developing aortic dissection, but their potential role as biomarkers is currently precluded due to the poorly measured sensitivity and specificity [136].
Increased C-reactive protein (CRP) levels have been observed in TAA, with respect to coronary artery disease patients and healthy controls [137], which may reflect the extensive inflammatory reaction and severe coagulopathies in individuals with acute type A aortic dissection and thoracic aortic aneurysm. In addition, CRP levels positively correlate with DD levels in aortic dissection [138], suggesting the presence of a strict relationship between these two factors in the pathophysiology of the complication and with plasma MMP8 and MMP9 in patients suspected of dissection [131]. These observations confirm the idea of a model in which inflammation, thrombosis, and ECM remodeling act synergically in driving the aortic rupture. However, the use of CRP does not apply in clinical practice as a specific diagnostic biomarker for aortic dissection, since increased levels can be observed in a number of other conditions (e.g., acute abdominal disease, pleuritis, pericarditis). A role for CRP may instead be considered for follow-up, as the stratification of patients with aortic dissection and studies have described an association between this acute-phase protein and the period of hospitalization [139]. CRP has been reported as an independent risk factor for in-hospital mortality [138]. Recently, Erdolu and as suggested the use of CRP and neutrophil to lymphocyte ratio values to predict mortality in patients with aortic dissections [140].
Sbarouni and co-workers analyzed 31 consecutive patients presenting acute aortic dissection showing higher homocysteine (Hcy) and lower folate compared to both chronic aneurysms and controls [141]. These data are not consistent with those resulting from studies on animal models with thoracic aortic dilatation, dissection, and rupture, in which hyperhomocysteinemia in the physiologic range did not induce/accelerate abnormal aortic growth in wild-type mice nor adverse pathologic progression in mice with an underlying predisposition for aortic dilatation [142]. An association has been found between elevated total Hcy (tHcy) levels and development of aortic dissection in a cohort of MFS patients [143] carrying the C677T polymorphism at the homozygous state. Although suggestive, especially for the marked remodeling of the extracellular matrix of the arterial wall induced by elevated Hcy through the activation of metalloproteases, further studies are needed to investigate the actual role of Hcy levels as a predictor of aneurysm progression [144].
Evidence was reported once again by a number of studies on MFS patients, underlying the increased levels of TGF-β as promoters of a significant weakening of the aortic wall resulting in TAA [145]. The usefulness of measuring TGF-β levels in such a specific population of patients as those affected by MFS, characterized by high variability in clinical manifestations, age of onset, and rate of aortic involvement, has been argued since the discovery of the alteration of this pathway in driving aortic complications observed in the disease [146]. This is of particular interest considering the potential advantages the evaluation of this plasma biomarker would represent in predicting disease severity and progression of artic dilatation. At the moment, the transition of this biomarker into the routine clinical evaluation of MFS and risk-stratification is not considered to be applicable, due to the inconsistency of results of different studies in retrieving elevated TGF-β levels in MFS patients; this is possibly due to the different population size, type of FBN1 mutation underlying the clinical picture and impacting the TGF-β pathway impairment, disease severity shown by patients, and use of medication [147]. Nonetheless, TGF-β concentration correlates with the level of aortic growth, faster aortic growth rate, previous aortic surgery, and acute aortic events during follow-up [148,149,150], suggesting its role as useful clinical marker in humans [151]. Acute elevations of TGF-β1 were found to be potentially predictive of aortic dissection in non-MFS patients [152], while the development of the complication has been associated with a significantly enhanced function of TGF-β1/Smad-signaling transduction, as a result of aortic remodeling incorporating both vascular injury and repair [153]; further evidence is emerging in this respect [154,155,156], suggesting an increased/unbalanced in plasma TGF-β in promoting aortic disease in different populations affected by genetic syndromes sharing aortic dilation as clinical complication, but this still has inconsistent results [157]. For those reasons, evaluation of this cytokine in the clinical practice of TAAD is not included in the 2014 ESC guidelines, unlike the molecular screening of those genes implicated in the TGF-β signaling.
Plasma levels of SMC protein have been evaluated in the context of aortic dissection. Among these, smooth muscle myosin heavy chain (smMHC) was first reported to acutely peak after aortic dissection [158] and later to be elevated in dissected patients compared with myocardial infarction subjects within 12 h from presentation or 3 h after onset [159,160].
Isozyme MM of creatine kinase (CK-MM, in which the protein is composed of two type M subunits) was observed to be selectively increased in aortic dissection patients 6 h after onset [161], while isozyme BB (2 type B subunits) was reported to peak at 12 h, in dissected patients with respect to controls, and return to normal within 24–36 h [162].
Calponin, a protein regulating myosin-actin interaction and SMCs contractility, was raised in serum of dissected patients in a study that enrolled multiple clinical centers [163], but its role in the diagnosis of dissection was eventually disregarded as the best results were obtained in early presenters and in Stanford Type A individuals.
More recently, smMHC, sELAF, D-dimer, or Polycystin 1 (PC1) alone were suggested as biomarkers for early diagnosis of acute aortic dissection, but the combination of these markers has been pointed out as having a significantly higher diagnostic value [164]. The application of these biomarkers in the current diagnostic flowchart has, however, not been considered.
Platelet activation and coagulopathy are also associated with aortic dissection and dissection extent [165]. A higher mean platelet volume/platelet (MPV/PLT) ratio was observed in aortic dissection patients along with a lower platelet count, which was also associated with an increased risk of in-hospital death before and after intervention; MPV/PLT, negatively associated with survival and platelet distribution, was established as a negative independent predictor of mortality in cohorts of dissected individuals [166,167]. Taken together, these data support the hypothesis of suppression of the PLT activation as future targets of therapy in acute aortic dissection, with particular reference to the prevention of systemic inflammation [165].
Data continue to emerge on potential biomarkers, e.g., aggrecan plasma levels were proposed as reliable biomarker to detect the presence of an acute type A aortic dissection in a very sensitive manner [168]; high-sensitivity cardiac troponin T concentration was proposed as an early biomarker for the risk stratification of patients with the same disease in the emergency department [169]; angiopoietin-like protein 8 (ANGPTL8), a hormone involved in the regulation of lipid metabolism and inflammation, combined with D-dimer and CRP was proposed as useful clinical predictor of TAAD [170]; and so on. Although promising, none of the abovementioned biomarkers showed a satisfactory profile for initial patient screening on large populations and case-control studies unavoidably provide limited information and need to be sustained by prospective enrolment of patients, such that further investigations are still necessary for implementing the guidelines, and novel, specific, and more readable biomarkers need to be unraveled [121]. To date, in fact, specific and promptly assessable biomarkers in the emergency departments worldwide are not available: the only validated biomarker is represented by DD, whose only validated cutoff for acute aortic syndromes is a 500 ng/mL fibrinogen equivalent unit (FEU) [121], the ESC guidelines indicating its measurement to rule out aortic dissection [11]. Still, as already mentioned, even in this case, the plasma biomarker lacks definitive specificity, as it may increase in several diseases including pericarditis, sepsis, and pulmonary embolism, and it may result falsely negative in conditions such as small intramural aortic hematomas or in those patients showing very early or very late symptoms of aortic dissection [121]. In this context, the broad-spectrum omic profiling (genomics, transcriptomics, metabolomics) may in the near future provide the necessary support (see Section 5) (Table 2).

4.3. Genetic Testing in Supporting TAA/D Diagnostics and in Risk Prediction: Where Do We Stand?

Although primarily considered as surgical disease, TAA’s optimal management greatly relies on an appropriate workup with the major purpose of identifying those features suggestive of a rapid progression of the aortic anomaly, thus predicting potentially life-threatening consequences of the disease. In this context, an accurate genetic evaluation/diagnosis serves different purposes: (a) guidance for overall medical management and surgical options; (b) timely evaluation of other organs that could be affected essentially in syndromic forms of TAA; (c) better definition of the prognosis; (d) identification of high-risk first-degree family members; (e) estimation of recurrence risk for future pregnancies in the prenatal diagnosis’ framework; and (f) support for imaging techniques in capturing nonsyndromic TAA patients who may be missed while developing dissection or rupture before reaching the guidelines-defined aortic diameter thresholds for aortic intervention [171]. As previously mentioned, syndromic and non-syndromic heritable thoracic aortic disease are, in most cases, inherited in an autosomal dominant manner except for rare X-linked and recessive conditions [172]. The accurate clinical evaluation of at-risk relatives is critical in this context, and ordinary and reproductive pre- and post-test genetic counseling allow for the early identification of an undiagnosed aortic disease in the first case and provide awareness about the risk of transmission to the offspring in the latter. Mutations are described to have variable penetrance depending on the TAA presentation, from almost 100% in MFS and 90% in LDS, to 50% in FTAAD and BAV in the presence of ascending aortic aneurysm. In fact, in the case of FTAAD, the causal mutation is found in much fewer cases (<10%) than in MFS or LDS, this discrepancy also being evident at the phenotypic level, presenting with a different severity of clinical manifestations along with age of presentation or diagnosis. When features of a connective-tissue disorder are present, patients should undergo genetic counselling and testing where appropriate [10]. The current ESC guidelines recommend genetic screening in first-degree relatives of TAA or aortic dissection and a diagnosis of familial aortic disease. In absence of a genetic diagnosis, at-risk relatives should undergo examination every 5 years. Screening should cover the entire arterial tree (including cerebral arteries) in families with nonsyndromic familial aortic disease [173]. According to the North American guidelines and related Class I recommendations, in case of identification of a mutation in one of the following genes, FBN1, TGFBR1, TGFBR2, COL3A1, ACTA2, and MYH11, which are associated with aortic aneurysm and/or dissection, first-degree relatives should undergo counseling and testing. Then, only the relatives with the genetic mutation should undergo aortic imaging. The guidelines provide some more recommendations (Class IIa and IIb): (a) ACTA2 sequencing should be considered in case of family history of thoracic aortic aneurysm and/or dissection; (b) TGFBR1, TGFBR2, and MYH11 sequencing may be considered in patients with a family history and clinical features associated with mutations in these genes; and (c) if one or more first-degree relatives of a patient with known thoracic aortic aneurysm and/or dissection are found to have thoracic aortic dilatation, aneurysm, or dissection, then referral to a geneticist may be considered [174]. Following the exclusion of a syndromic condition, nonsyndromic TAA, in which mutations in genes known to be involved in syndromic forms of TAAD are rarely found, may present suggestive features of a genetic etiology, which might include young age at presentation (<50 years old), multiple aneurysms or dissections, and aortic root aneurysm [175,176]. In this scenario, genetic counseling should begin with the collection of the most detailed information of a three-generation family history, for the presence of aneurysm, dissection, sudden deaths, and syndromic features that would help in determining the inheritance pattern, identifying at-risk relatives, and recognizing syndromic signs [172]. In 2009, Ripperger and co-workers reported three cases of sudden, unexpected death due to thoracic aortic dissection, pointing out the great benefit that could be derived from alerting the at-risk relatives of the deceased about a potential heritable etiology of the disease [177]. The authors propose the development of a standard procedure which includes genetic counseling for at-risk relatives and storage of DNA or unfixed tissue for molecular investigations that would eventually allow differential diagnostic reappraisal from a genetic point of view. In any case, during genetic consultation, patients should become aware of the limitations, benefits, and personal and familial implications of genetic testing. Besides, awareness should be raised on the possibility of a negative genetic test that would not necessarily exclude a genetic etiology, thus indicating the imaging to be performed anyways in the first-degree family members in the search for aortic disease [174]. In fact, some types of genetic variants may be undetectable by standard assays and, similarly, the causative mutation may involve a gene that has not yet been associated to TAAD, due to absence of data supporting the actual pathological effect of that variant [174]. As a matter of fact, regarding the most appropriate genetic test selection, no specific indications are provided by the European guidelines. Genetic-testing panels vary significantly among laboratories and despite the enthusiasm for the so-called “exome-first” approach in diagnosing such a complex disease as TAA, its actual benefit and routine application in the diagnostic workup currently represent a matter of debate within the international scientific community.

5. Latest Findings on TAA/D Genetic and Non-Genetic Biomarkers

5.1. RNA Signatures: A Novel, Noninvasive, and Promising Screening Option?

As discussed in the previous section, the identification of noninvasive approaches, which could support and extend the diagnostic/risk-prediction capabilities workup for TAAD, has been the subject of numerous studies that reported a number of molecules potentially driving the aneurysm formation and/or dissection. Yet, their real value lies in preclinical verification (in terms of sensitivity and specificity) and validation on large cohorts of patients vs. controls and in comparing subsets of patients that are affected by the disease but in which the presentation is highly variable. Recently, evidence is accumulating about the role of micro-RNAs (miRNAs), non-coding RNAs (ncRNAs), and circular RNAs in the development of many cardiovascular diseases, including aortic dissection [170]. Together, these molecules contribute to determining RNA-expression patterns and, ultimately, in defining the so-called “RNA signatures”, with the potential ability to accurately differentiate between different pathologic phenotypes (including the variable clinical manifestations of the same disease, as in the case of TAA), which has drawn the attention of numerous research groups during the last decades. This was also facilitated by the advancements in microarray and high-throughput technologies allowing the interrogation of different RNA populations in a single assay [178] (Table 3).
A 2007 study analyzed whole-genome gene-expression profiles from 94 peripheral blood samples (58 TAA subjects and 36 controls), identifying, with high accuracy (80% overall) a signature set of 41 biomarker genes specific of asymptomatic TAA patients [179]. This signature RNA test had also the ability to differentiate between ascending vs. descending aneurysms and between familial and sporadic TAA, also highlighting potential targets for intervention. Suggestions are derived from studies on miRNA profiles. The miR-29 family comprises three members, miR-29a/-29b/-29c, with expression that has been tested in a number of studies involving animal models and human normal/aneurysmatic/dissected aorta tissues [178]. Among these, miRNA-29b was found to play pivotal role in the formation of aneurysms [180,181], post-transcriptionally regulating the expression levels of multiple targets with a function in the ECM collagens, elastin, and fibrillin and modulating the aortic SMCs’ synthetic phenotype switch [182]. A therapeutic potential has been consequently proposed for miRNA-29b, with inhibition that could prevent TAA expansion. A cross-talk between miRNA-29b and TGF-β was observed, thus confirming its potential role in TAA, aortic dissection, and other diseases in which the disruption of the TGF-β signaling represents an underlying pathophysiological mechanism [178]. Two more miRNAs have been proposed as therapeutic targets for TAA, specifically miRNA-143 and miRNA-145, which appear to have a role in VSMCs’ phenotype switch [183] and, when up-regulated, an increased expression of VSMCs’ differentiation markers was observed [184].
Table 3. Novel potential circulating biomarkers for TAAD.
Table 3. Novel potential circulating biomarkers for TAAD.
MarkerAnimal ModelsHuman CohortTAATAAD
miR-1-aortic tissue specimens from ascending TAA patients (30)/3 tissues of patients with AAA, 11 tissues of patients with TAA and 8 controls [185,186]+
miR-21+
miR-29a++
miR-133a++
miR-15a-10 patients with TAA/3 tissue specimens from AAA patients, 11 from TAA patients and 8 controls/aortic tissue specimens from AAA patients (10) [186,187,188]++
miR-22++
miR-25+
miR-29b+
miR-125a-3p+
miR-126-3p+
miR-128+
miR-133b++
miR-138-1++
miR-142–5p+
miR-145++
miR-146b-5p+
miR-183++
miR-422a+
miR-433++
miR-486–5p+
miR-487b+
miR-491–3p++
miR-553++
miR-638+
miR-940++
miR-193a-3p++
miR-768–5p++
miR-886–5p ++
miR-195++
miR-140–5p++
miR-30e++
miR-101++
miR-744++
miR-193a-5p++
miR-30c-3 tissues specimens from AAA patients, 11 from TAA patients and 8 controls [186]+
miR-155+
miR-204+
miR-143mouse models [183]-++
AAA: abdominal aortic aneurysm; miR: microRNA; TAA: aortic aneurysm, thoracic.
Both miRNAs were down-regulated in aorta from acute aortic dissection patients [189], while their expression levels appeared increased or decreased in TAA depending on the study [183,188]. Other mi-RNAs were suggested to modulate aortic SMC towards the differentiation process (miR-1 [190], miR-133 [191], miR-663 [192], miR-424 [193], miR-195 [194], miR-138 [192]), de-differentiation process (miR-221/-222 [195], miR26a [196], miR-146a [197], miR-155 [198], miR-31 [199], miR-181b [200]), and phenotype switch mechanisms (miR-21 [201], miR-24 [202]) (Table 3). The involvement of all these molecules in aortic SMCs biology and ECM integrity maintenance and composition, as well as their differential expression in aneurysmatic tissues with respect to controls, supports their potential role in TAAD pathogenesis. Still, miRNAs actively promoting those processes leading to aortic dilatation and from aortic dilatation to aortic dissection need to be clearly differentiated from simple bystanders, in order to identify precise therapeutic targets [178]. A validation study was carried out by Moushi and co-workers to verify the literature data on the association between a large number of miRNAs using plasma from TAA patients collected before and after surgery [203]. In this study, where 24 papers and 11 miRNAs were selected for validation, miR-193a-5p and miR-30-b-5p were found to be down-regulated in plasma samples collected before the aneurysm removal with respect to post-surgical ones, making these molecules the most promising among those tested. A recent study analyzed 19 TAA patients and 19 controls allowing the identification of 232 differentially expressed miRNAs, among which miR-574-5p was proposed as a potential therapeutic target [178]. A cohort of 40 MFS patients undergoing elective ascending aorta surgery were enrolled in a study conducted by D’Amico and co-workers, which aimed to compare TAA histomorphological features, a miRNA profile and related target genes, and to find specific alterations that may explain the earlier and more severe clinical outcomes in MFS patients [204]. Twenty-five miRNAs, including miR-26a, miR-29, miR-143, and miR-145, were found to be downregulated, while miR-632 was upregulated in MFS/TAA in vivo; in addition, 28 upregulated and 7 downregulated genes were identified, some of them belonging to the CDH1/APC and CCNA2/TP53 signaling pathways, which the authors propose to be further tested as potential therapeutic targets to counteract the rapid progression of MFS aortopathy.
Studies have also addressed a potential role of long ncRNAs (lncRNAs) in modulating TAA development and progression acting via several molecular pathways. With regards to known pathophysiological mechanisms underlying TAA onset, including aortic-wall expansion and VSMCs’ dedifferentiation from the contractile to the synthetic phenotype, which is regulated by lncRNAs’ CARMN, LUCAT1, SMILR, and MALAT1, Patamsytė and co-workers tested these molecules in clinical aortic tissue and blood-plasma samples from TAA and non-TAA patients using the qRT-PCR method [205], attributing to LUCAT1 alone the ability to discriminate aneurysmal disease in patients’ blood plasma and a diagnostic potential for TAA. Subsequently, high-throughput sequencing was used to analyze lncRNAs’ expression profile in human thoracic aortic dissection, revealing a set of dysregulated lncRNAs and predicting their multiple potential functions in the disease (lnc_1421, ENSG00000269936, lncRNA XIST, NSG00000248508, ENSG00000226530, EG00000259719 [206]). Further evidence continues to emerge, as LncRNA Sox2ot was shown to modulate TAA progression by regulating miR-330-5p/Myh11, which was suggested as new potential mode to treat TAA [207], as well as lncRNA CDKN2B-AS1, which was found to aggravate the pathogenesis of human thoracic aortic dissection [208], or LncRNA Xist’s contributing to arterial smooth muscle cell apoptosis through the miR-29b-3p/Eln pathway [209]. Additional data have been reported on lncRNA H19, which has the ability to regulate smooth muscle cell functions participating in the development of aortic dissection through sponging miR-193b-3p [210], and on lncRNA OIP5-AS1, which exacerbates aorta intima, media, and adventitia injury in the development of aortic dissection, through the upregulation of TUB via sponging miR-143-3p [211].
Circular RNAs (CircRNAs) represent endogenous lncRNAs with regulatory roles at both the transcriptional and post-transcriptional levels, by binding and interacting with miRNAs. Among them, circRNA-101238 was found to be highly expressed in human thoracic aortic dissection specimens, leading to a lower expression of the downstream target miR-320a, and, in turn, to increased MMP9 expression [212], while circMARK3-miR-1273-Fgr interaction was suggested to have a certain clinical significance in human acute Stanford type A aortic dissection (AAAD) tested by RNA-seq [213].
Although promising, miRNA research needs to be extended to other cellular components besides VSMCs and endothelial cells, such as macrophages and fibroblasts; other ncRNAs, such as repeat-associated small interfering RNAs (rasiRNAs) and Piwi-interacting RNAs (piRNAs), may also be involved in the disease and should undergo further investigation; strategies for local and safe ncRNA or miRNA delivery in patients are required as well as the development of in vivo imaging techniques mimicking those effects that were observed in animal models [214]. In this respect, it seems that there is still a long way to go and a lot to be done in order to test these hypotheses and to include RNA markers in the routinely diagnostic flowchart for TAA.

5.2. Novel Genes and the WES Outbreak: Pros and Cons in the Clinical Practice and Applicability

As is well known, the high-throughput molecular technologies’ outbreak in the mid-2000s, represented an actual “revolution” in the genetic research and diagnostic fields for Mendelian disorders as well as those pathologies recognizing, among other factors, a genetic etiology. Still, the opportunity to interrogate the entire codifying component of the human genome (WES) or the entire human genome itself (Whole Genome Sequencing (WGS)) in a single assay, at a decreasing cost and in a multiplex mode, even if unarguably beneficial for gene-discovery purposes, led to a debate over its actual usefulness and applicability in the routinely diagnostic flowchart for a series of pathologies in the place of targeted gene/gene panels approaches. Regarding TAA, disease-specific gene panels are expanding in terms of the number of included genes. That is especially due to the phenotypic heterogeneity and allelic overlap between syndromic and non-syndromic cases [31]. Positive genetic testing for TAA has important implications for disease management and for at-risk family members screening, so that guidance in decision-making about when to re-test, as panels continue to expand, is necessary.
Back in 2014, WES analysis identified a new mutation in TGFB2 involved in a familial case of non-syndromic aortic disease [215]. As discussed in Section 3.1, TGFB2, together with FBN1, TGFBR1, TGFBR2, and SMAD3 mutations, accounts for 14% of non-syndromic familial TAA [40], but current American Guidelines do not include this gene among those that should be tested in patients with a TAA family history [174]. WES identified recurrent gain-of-function mutations in PRKG1 as causative for thoracic aortic aneurysms and acute aortic dissections [117], meanwhile MAT2A, LOX, and FOXE3 have been suggested as predisposing genes [28,30,43]. Milewicz and co-workers applied WES to identify causative mutations in novel genes for TAAD [216]. Their strategy resided in sequencing distant relatives with TAAD, in order to reduce the large number of rare variants identified using WES, and filtering for heterozygous rare variants that are shared between relatives and that are predicted to disrupt protein function and segregate with the TAAD phenotype in other family members. This strategy led to the successful identification of novel genes for FTAAD, which were validated through minimal additional molecular, cellular, or animal studies. Despite the advantages of this approach in gene discovery, the authors also stressed the importance of the molecular, cellular, and animal studies required to link the gene variant to the disease phenotype that also needs the development of novel techniques and the establishment of new collaborations, with this last issue to be especially encouraged. WES was also applied in an attempt to identify the causative mutation in two young patients with acute type B aortic dissection without syndromic features, who were afterwards diagnosed with MFS as they were found to carry two pathogenic FBN1 mutations [217]. In this work, the authors emphasized the necessity of genetic testing for young patients with type B aortic dissection through WES, which is a timely, robust, and inexpensive technique for genetic sequencing, particularly for TAAD that is caused by numerous genes, in order to identify syndromic conditions, such as MFS, which has a diagnosis that may facilitate periodic surveillance, prophylactic surgical measures, and genetic counseling. Another study performed WES on 183 FTAAD families without significant systemic features of MFS identifying FBN1 mutations in 11 families, suggesting the screening of this gene in this category of FTAAD subjects [218]. WES was used as routine genetic test in 102 TAAD patients [219], allowing for the opportunity for the personalized management of patients tailored to the specifics of the genetic mutation (e.g., prophylactic surgical interventions earlier and at smaller aortic for malignant ones). Candidate genetic modifiers of syndromic and familial TAA severity were identified through WES in a cohort of 27 subjects with syndromic or familial TAA and presenting extreme phenotypes (ADCK4 and COL15A1 genes [220]). Authors speculate that these hypotheses-generating findings initiate a path toward risk stratification through genetic testing at an early stage of disease and identify novel therapeutic targets, pinpointing WES as the most suitable technology for those purposes. However, they also point out the need to develop novel statistical and experimental platforms to define how specific variants interact to actually influence phenotype. New genes (MLX, DAB2IP, EP300, ZFYVE9, PML, PRKCD) were suggested as candidate aortic dissection-associated genes, through correlation analyses performed on the results of a WES study on a cohort of 99 Chinese cases [112]. Variants of TES and other focal adhesion scaffold genes were shown to predispose individuals to isolated TAA, by affecting vascular smooth-muscle-cell phenotypic modulation and vasoconstrictive function [221]. These results were obtained from a WES study on a cohort of 551 sporadic isolated TAA cases and 1071 controls, which expanded the genetic landscape across this disease, showing focal adhesion scaffold genes as a novel category of TAA causal genes. The application of different methodologies (other than WES, such as Next Generation Sequencing (NGS) panels and mouse models), led to the identification of a number of other suggestive genes for TAA/D (Table 4).
A flowchart for a dedicated screening program for relatives of patients affected by nonsyndromic TAA was proposed by Faggion Vinholo and co-workers, in which genetic screening is strongly supported by the literature data [6]. In all these studies, authors list the advantages of applying WES in a TAA clinical setting: (1) it is comprehensive, including (but not limited to) testing all the 30-plus TAAD genes that have been identified to date; (2) the WES of the proband permits straightforward, comprehensive screening of the family members by less complex and less-costly single-site (Sanger) sequencing and non-mutation-carrying family members can be spared repeated imaging studies; and (3) patients with no mutation of a known TAAD gene, especially those with affected family members, are likely to provide a clinical “gold mine” for mining WES data for totally novel TAAD genes, especially in light of the genes identified to date that only account for approximately 30% of aortic diseases in nonsyndromic TAAD patients [219].
On the other hand, Renard and co-workers reported the results of a study aimed to accurately identify TAAD-predisposing genes using the Clinical Genome Resource (ClinGen) framework [36]. Fifty-three candidate genes were analyzed, nine of which were categorized as “TAAD genes with definitive or strong gene disease-association” (ACTA2, COL3A1, FBN1, MYH11, MYLK, SMAD3, TGFB2, TGFBR1, TGFBR2), while eight were defined as “potentially diagnostic genes, with moderate or limited gene-disease association”, as they could allow diagnosis of thoracic aortic enlargement but are primarily associated with other clinical features and do not carry a significant risk for dissection (EFEMP2, ELN, FBN2, FLNA, NOTCH1, SLC2A10, SMAD4, SKI). The other genes had limited or no clinical evidence for TAA/D. These findings led the authors to question the usefulness of very large panels (and, as a consequence, WES) that instead increase the risk for ambiguous and even false-positive reports and raise the probability of identifying variants of uncertain significance (VUS), which may cause unnecessary distress for patients and family members and create diagnostic confusion, misallocation of resources for follow-up, and even unjustified genetic discrimination [228]. A gene panel containing the most established genes associated with the highest risk of early fatal complications in patients with hereditary TAAD (ACTA1, COL3A1, FBN1, MYH11, SMAD3, TGFB2, TGFBR1, TGFBR2, MYLK) was evaluated as sufficient to identify the prevailing majority of variants most likely to be causative of the disease, with respect to the larger panel including 174 genes [37]. The authors suggest that a curated list of genes associated with heritable TAAD has the ability to identify patients and families at risk and reduce inconclusive diagnostic testing by limiting the scope of screened genes. The quality of a diagnostic panel should be judged on the clinical validity of genes included in that panel rather than on the number. In this respect, efforts need to be made in the direction of more complete and uniform genetic profiles with greater power to diagnose TAAD and predict its fatal consequences [36]. However, data analysis of a WES experiment could definitely focus on the most suspected genes and/or on those with a role in causing/modulating TAA and its complications, which is supported by the most robust data, and then broaden the evaluation in case of negative results. The probability of incidental findings and VUSs remains high and this issue should probably be addressed in determining the most appropriate way of discussing it during genetic counseling.

6. Conclusions

TAA’s bad reputation of “silent killer” is to be ascribed to its characteristic features, including its slow and gradual formation and the absence of visible signs, with patients remaining asymptomatic. This condition is elusive and yet potentially life-threatening, as it manifests itself only once the aneurysm is large enough to lead to an acute and devastating aortic event, with a significant percentage of patients dying before reaching the hospital. As a result, it is of the utmost relevance to identify biomarkers for the early identification of asymptomatic patients, a task which is both essential and challenging. In this regard, there’s an important distinction to be made between the TAD management within the emergency department, in which the room to maneuver is objectively limited, and those other situations in which the fatal event has not happened yet. In the first case, as Mehta and co-workers pointed out in a very recent review, the margin of intervention is essentially directed to improving the patient’s outcome by different means, including the multidisciplinary collaboration between specialists (emergency physicians, surgeons, radiologists) and identification of the optimal interventional treatment and post-operative care [229]. These goals are basically pursued by an accurate evaluation of the aortic pathology, the “nature of the pain” described by patients, the malperfusion at different locations (neurologic, spinal cord, visceral, renal), and the correct interpretation of diagnostic-imaging findings, while laboratory tests are essentially represented by basic metabolic panels, cardiac biomarkers, DD, troponin T, and complete blood count; those measurements, however, are insufficient in acute aortic pathologies diagnosis and, in most cases, are non-specific. On the other hand, for risk-stratification, risk-prediction in first-degree family members, disease management, and follow-up purposes, the identification of affordable and reliable markers is critical, challenging, and yet practicable. In this respect, efforts have been directed over the years to the evaluation of circulating biomarkers that could be used in the TAA diagnostic path to discriminate those patients who are more prone to develop a potentially fatal complication. A number of circulating biomarkers have been suggested to be useful, alone or in combination, essentially in ruling out aortic dissection (DD, MMP8, smMHC, sELAF, PC1). According to the 2014 European guidelines, in case of the suspicion of aortic dissection, part of these molecules is currently tested, yet they have not entered the clinical arena [11]. Traditional circulating biomarkers do not represent a satisfactory and reliable support in the initial patient screening as well, in which, on the contrary, the molecular/genetic evaluation can be diriment. Genetic testing, especially that which interrogates several genes at once in a parallel approach that is, at present, undoubtedly preferable to the cascade one, has long been included in the diagnostic flowchart for TAA diagnosis. First of all, it allows for the identification of a co-existing condition with TAA, such as MFS or LDS, thus directing the most appropriate management in terms of periodic check-ups, time of intervention, risk-recurrence calculation for pregnancies, and screening for first-degree relatives. In addition, the constant implementation of molecular methodologies allowing for the interrogation of the entire genome or transcriptome in an “omic” approach could be, undoubtedly, beneficial for patients’ stratification. In fact, the combination of the data derived from the WES/WGS/RNA-seq approaches can help define profiles that could be highly specific for subgroups of TAA patients, not to mention the potential use of those data in deepening knowledge about the disease’s onset and progression as well as for identifying new targets for therapy. Even with the considerable limitations characterizing the omic approaches (production of a large amount of bioinformatic data that need to be correctly interpreted, safely stored, and validated through functional studies; the possibility of VUSs and incidental findings), the future benefits they may represent for the improvement of the TAA diagnostic work-up have to be considered and perhaps should be addressed more closely and in greater detail in the international guidelines.

Funding

This paper received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

AAAabdominal aortic aneurysm
AAADacute Stanford type A aortic dissection
ADAMTSA Disintegrin and Metalloproteinase with Thrombospondin motifs
BAVbicuspid aortic valve
circRNAscircular RNAs
CK-MMCreatine-kinase isozyme MM
CRPC-reactive protein
DDd-Dimer
ECMextracellular matrix
ESCEuropean Society of Cardiology
FTAADfamilial thoracic aortic aneurysm and dissection
Hcyhomocysteine
LAPlatency-associated peptide
LDSLoeys-Dietz syndrome
lncRNAslong non-coding RNAs
LTBPlatent TGF? binding protein
MFSMarfan syndrome
miRNAsmicro-RNAs
MMPsmetalloproteinases
MPVmean platelet volume
ncRNAsnon-coding RNAs
NGSnext-generation sequencing
PC1Polycistin 1
piRNAsPiwi-interacting RNAs
PLTplatelet
qRT-PCRQuantitative Reverse Transcription Polymerase Chain Reaction
rasiRNAsrepeat associated small interfering RNAs
sELAFssoluble elastin fragments
SMCssmooth muscle cells
smMHCsmooth muscle myosin heavy chain
TAA/Dthoracic aortic aneurysm and dissection
TAAthoracic aortic aneurys
TADthoracic aortic dissection
TEVARthoracic endovascular aortic repair
TGFβTransforming Growth Factor-β
tHcytotal homocysteine
TIMPstissue inhibitors of metalloproteinases
vEDSvascular Ehlers-Danlos syndrome
VEGFvascular endothelial growth factor
VSMCsvascular smooth muscle cells
VUSvariant of uncertain significance
WESwhole exome sequencing
WGSwhole genome sequencing

References

  1. Bossone, E.; Eagle, K.A. Epidemiology and Management of Aortic Disease: Aortic Aneurysms and Acute Aortic Syndromes. Nat. Rev. Cardiol. 2021, 18, 331–348. [Google Scholar] [CrossRef]
  2. Kuzmik, G.A.; Sang, A.X.; Elefteriades, J.A. Natural History of Thoracic Aortic Aneurysms. J. Vasc. Surg. 2012, 56, 565–571. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Clouse, W.D.; Hallett, J.W.; Schaff, H.V.; Spittell, P.C.; Rowland, C.M.; Ilstrup, D.M.; Melton, L.J. Acute Aortic Dissection: Population-Based Incidence Compared with Degenerative Aortic Aneurysm Rupture. Mayo Clin. Proc. 2004, 79, 176–180. [Google Scholar] [CrossRef] [PubMed]
  4. Nienaber, C.A.; Clough, R.E.; Sakalihasan, N.; Suzuki, T.; Gibbs, R.; Mussa, F.; Jenkins, M.P.; Thompson, M.M.; Evangelista, A.; Yeh, J.S.M.; et al. Aortic Dissection. Nat. Rev. Dis. Primer 2016, 2, 16053. [Google Scholar] [CrossRef] [PubMed]
  5. Smedberg, C.; Steuer, J.; Leander, K.; Hultgren, R. Sex Differences and Temporal Trends in Aortic Dissection: A Population-Based Study of Incidence, Treatment Strategies, and Outcome in Swedish Patients during 15 Years. Eur. Heart J. 2020, 41, 2430–2438. [Google Scholar] [CrossRef] [PubMed]
  6. Faggion Vinholo, T.; Zafar, M.A.; Ziganshin, B.A.; Elefteriades, J.A. Nonsyndromic Thoracic Aortic Aneurysms and Dissections—Is Screening Possible? Semin. Thorac. Cardiovasc. Surg. 2019, 31, 628–634. [Google Scholar] [CrossRef]
  7. Salameh, M.J.; Black, J.H.; Ratchford, E.V. Thoracic Aortic Aneurysm. Vasc. Med. 2018, 23, 573–578. [Google Scholar] [CrossRef]
  8. Monda, E.; Fusco, A.; Della Corte, A.; Caiazza, M.; Cirillo, A.; Gragnano, F.; Giugliano, M.P.; Citro, R.; Rubino, M.; Esposito, A.; et al. Impact of Regular Physical Activity on Aortic Diameter Progression in Paediatric Patients with Bicuspid Aortic Valve. Pediatr. Cardiol. 2021, 42, 1133–1140. [Google Scholar] [CrossRef]
  9. Rohde, S.; Zafar, M.A.; Ziganshin, B.A.; Elefteriades, J.A. Thoracic Aortic Aneurysm Gene Dictionary. Asian Cardiovasc. Thorac. Ann. 2021, 29, 682–696. [Google Scholar] [CrossRef]
  10. Chou, E.L.; Lindsay, M.E. The Genetics of Aortopathies: Hereditary Thoracic Aortic Aneurysms and Dissections. Am. J. Med. Genet. C Semin. Med. Genet. 2020, 184, 136–148. [Google Scholar] [CrossRef]
  11. Erbel, R.; Aboyans, V.; Boileau, C.; Bossone, E.; Di Bartolomeo, R.; Eggebrecht, H.; Evangelista, A.; Falk, V.; Frank, H.; Gaemperli, O.; et al. 2014 ESC Guidelines on the diagnosis and treatment of aortic diseases. Kardiol. Pol. 2014, 72, 1169–1252. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Harris, S.L.; Lindsay, M.E. Role of Clinical Genetic Testing in the Management of Aortopathies. Curr. Cardiol. Rep. 2021, 23, 10. [Google Scholar] [CrossRef] [PubMed]
  13. Elefteriades, J.A.; Sang, A.; Kuzmik, G.; Hornick, M. Guilt by Association: Paradigm for Detecting a Silent Killer (Thoracic Aortic Aneurysm). Open Heart 2015, 2, e000169. [Google Scholar] [CrossRef]
  14. Quintana, R.A.; Taylor, W.R. Cellular Mechanisms of Aortic Aneurysm Formation. Circ. Res. 2019, 124, 607–618. [Google Scholar] [CrossRef]
  15. Albornoz, G.; Coady, M.A.; Roberts, M.; Davies, R.R.; Tranquilli, M.; Rizzo, J.A.; Elefteriades, J.A. Familial Thoracic Aortic Aneurysms and Dissections—Incidence, Modes of Inheritance, and Phenotypic Patterns. Ann. Thorac. Surg. 2006, 82, 1400–1405. [Google Scholar] [CrossRef] [PubMed]
  16. Biddinger, A.; Rocklin, M.; Coselli, J.; Milewicz, D.M. Familial Thoracic Aortic Dilatations and Dissections: A Case Control Study. J. Vasc. Surg. 1997, 25, 506–511. [Google Scholar] [CrossRef] [Green Version]
  17. Coady, M.A.; Davies, R.R.; Roberts, M.; Goldstein, L.J.; Rogalski, M.J.; Rizzo, J.A.; Hammond, G.L.; Kopf, G.S.; Elefteriades, J.A. Familial Patterns of Thoracic Aortic Aneurysms. Arch. Surg. 1999, 134, 361–367. [Google Scholar] [CrossRef] [Green Version]
  18. Cannon Albright, L.A.; Camp, N.J.; Farnham, J.M.; MacDonald, J.; Abtin, K.; Rowe, K.G. A Genealogical Assessment of Heritable Predisposition to Aneurysms. J. Neurosurg. 2003, 99, 637–643. [Google Scholar] [CrossRef]
  19. Meester, J.A.N.; Vandeweyer, G.; Pintelon, I.; Lammens, M.; Van Hoorick, L.; De Belder, S.; Waitzman, K.; Young, L.; Markham, L.W.; Vogt, J.; et al. Loss-of-Function Mutations in the X-Linked Biglycan Gene Cause a Severe Syndromic Form of Thoracic Aortic Aneurysms and Dissections. Genet. Med. 2017, 19, 386–395. [Google Scholar] [CrossRef] [Green Version]
  20. Takeda, N.; Komuro, I. Genetic Basis of Hereditary Thoracic Aortic Aneurysms and Dissections. J. Cardiol. 2019, 74, 136–143. [Google Scholar] [CrossRef]
  21. Baldwin, A.K.; Simpson, A.; Steer, R.; Cain, S.A.; Kielty, C.M. Elastic Fibres in Health and Disease. Expert Rev. Mol. Med. 2013, 15, e8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Szabo, Z. Aortic Aneurysmal Disease and Cutis Laxa Caused by Defects in the Elastin Gene. J. Med. Genet. 2005, 43, 255–258. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Guemann, A.-S.; Andrieux, J.; Petit, F.; Halimi, E.; Bouquillon, S.; Manouvrier-Hanu, S.; Van De Kamp, J.; Boileau, C.; Hanna, N.; Jondeau, G.; et al. ELN Gene Triplication Responsible for Familial Supravalvular Aortic Aneurysm. Cardiol. Young 2015, 25, 712–717. [Google Scholar] [CrossRef]
  24. Kent, K.C.; Crenshaw, M.L.; Goh, D.L.M.; Dietz, H.C. Genotype-Phenotype Correlation in Patients with Bicuspid Aortic Valve and Aneurysm. J. Thorac. Cardiovasc. Surg. 2013, 146, 158–165.e1. [Google Scholar] [CrossRef] [Green Version]
  25. Weerakkody, R.; Ross, D.; Parry, D.A.; Ziganshin, B.; Vandrovcova, J.; Gampawar, P.; Abdullah, A.; Biggs, J.; Dumfarth, J.; Ibrahim, Y.; et al. Targeted Genetic Analysis in a Large Cohort of Familial and Sporadic Cases of Aneurysm or Dissection of the Thoracic Aorta. Genet. Med. Off. J. Am. Coll. Med. Genet. 2018, 20, 1414–1422. [Google Scholar] [CrossRef] [PubMed]
  26. Ostberg, N.P.; Zafar, M.A.; Ziganshin, B.A.; Elefteriades, J.A. The Genetics of Thoracic Aortic Aneurysms and Dissection: A Clinical Perspective. Biomolecules 2020, 10, 182. [Google Scholar] [CrossRef] [Green Version]
  27. Franken, R.; Groenink, M.; de Waard, V.; Feenstra, H.M.A.; Scholte, A.J.; van den Berg, M.P.; Pals, G.; Zwinderman, A.H.; Timmermans, J.; Mulder, B.J.M. Genotype Impacts Survival in Marfan Syndrome. Eur. Heart J. 2016, 37, 3285–3290. [Google Scholar] [CrossRef]
  28. Guo, D.; Regalado, E.S.; Gong, L.; Duan, X.; Santos-Cortez, R.L.P.; Arnaud, P.; Ren, Z.; Cai, B.; Hostetler, E.M.; Moran, R.; et al. LOX Mutations Predispose to Thoracic Aortic Aneurysms and Dissections. Circ. Res. 2016, 118, 928–934. [Google Scholar] [CrossRef] [Green Version]
  29. Lee, V.S.; Halabi, C.M.; Hoffman, E.P.; Carmichael, N.; Leshchiner, I.; Lian, C.G.; Bierhals, A.J.; Vuzman, D.; Brigham Genomic Medicine; Mecham, R.P.; et al. Loss of Function Mutation in LOX Causes Thoracic Aortic Aneurysm and Dissection in Humans. Proc. Natl. Acad. Sci. USA 2016, 113, 8759–8764. [Google Scholar] [CrossRef] [Green Version]
  30. Kuang, S.-Q.; Medina-Martinez, O.; Guo, D.-C.; Gong, L.; Regalado, E.S.; Reynolds, C.L.; Boileau, C.; Jondeau, G.; Prakash, S.K.; Kwartler, C.S.; et al. FOXE3 Mutations Predispose to Thoracic Aortic Aneurysms and Dissections. J. Clin. Investig. 2016, 126, 948–961. [Google Scholar] [CrossRef] [Green Version]
  31. Schubert, J.A.; Landis, B.J.; Shikany, A.R.; Hinton, R.B.; Ware, S.M. Clinically Relevant Variants Identified in Thoracic Aortic Aneurysm Patients by Research Exome Sequencing. Am. J. Med. Genet. A 2016, 170, 1288–1294. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Milewicz, D.M.; Guo, D.-C.; Tran-Fadulu, V.; Lafont, A.L.; Papke, C.L.; Inamoto, S.; Kwartler, C.S.; Pannu, H. Genetic Basis of Thoracic Aortic Aneurysms and Dissections: Focus on Smooth Muscle Cell Contractile Dysfunction. Annu. Rev. Genom. Hum. Genet. 2008, 9, 283–302. [Google Scholar] [CrossRef] [PubMed]
  33. Chen, M.H.; Choudhury, S.; Hirata, M.; Khalsa, S.; Chang, B.; Walsh, C.A. Thoracic Aortic Aneurysm in Patients with Loss of Function Filamin A Mutations: Clinical Characterization, Genetics, and Recommendations. Am. J. Med. Genet. A. 2018, 176, 337–350. [Google Scholar] [CrossRef] [PubMed]
  34. Zhu, L.; Vranckx, R.; Khau Van Kien, P.; Lalande, A.; Boisset, N.; Mathieu, F.; Wegman, M.; Glancy, L.; Gasc, J.-M.; Brunotte, F.; et al. Mutations in Myosin Heavy Chain 11 Cause a Syndrome Associating Thoracic Aortic Aneurysm/Aortic Dissection and Patent Ductus Arteriosus. Nat. Genet. 2006, 38, 343–349. [Google Scholar] [CrossRef]
  35. Luyckx, I.; Loeys, B.L. Curriculum topic: Disease of the aorta and trauma to the aorta and heart The Genetic Architecture of Non-Syndromic Thoracic Aortic Aneurysm. Heart Br. Card. Soc. 2015, 101, 1678–1684. [Google Scholar] [CrossRef]
  36. Renard, M.; Francis, C.; Ghosh, R.; Scott, A.F.; Witmer, P.D.; Adès, L.C.; Andelfinger, G.U.; Arnaud, P.; Boileau, C.; Callewaert, B.L.; et al. Clinical Validity of Genes for Heritable Thoracic Aortic Aneurysm and Dissection. J. Am. Coll. Cardiol. 2018, 72, 605–615. [Google Scholar] [CrossRef]
  37. Ponińska, J.K.; Bilińska, Z.T.; Truszkowska, G.; Michalak, E.; Podgórska, A.; Stępień-Wojno, M.; Chmielewski, P.; Lutyńska, A.; Płoski, R. Good Performance of the Criteria of American College of Medical Genetics and Genomics/Association for Molecular Pathology in Prediction of Pathogenicity of Genetic Variants Causing Thoracic Aortic Aneurysms and Dissections. J. Transl. Med. 2022, 20, 42. [Google Scholar] [CrossRef]
  38. Quiñones-Pérez, B.; VanNoy, G.E.; Towne, M.C.; Shen, Y.; Singh, M.N.; Agrawal, P.B.; Smith, S.E. Three-Generation Family with Novel Contiguous Gene Deletion on Chromosome 2p22 Associated with Thoracic Aortic Aneurysm Syndrome. Am. J. Med. Genet. A 2018, 176, 560–569. [Google Scholar] [CrossRef]
  39. MacCarrick, G.; Black, J.H.; Bowdin, S.; El-Hamamsy, I.; Frischmeyer-Guerrerio, P.A.; Guerrerio, A.L.; Sponseller, P.D.; Loeys, B.; Dietz, H.C. Loeys–Dietz Syndrome: A Primer for Diagnosis and Management. Genet. Med. 2014, 16, 576–587. [Google Scholar] [CrossRef]
  40. Milewicz, D.M.; Regalado, E. Heritable Thoracic Aortic Disease Overview. In GeneReviews®; Adam, M.P., Ardinger, H.H., Pagon, R.A., Wallace, S.E., Bean, L.J., Gripp, K.W., Mirzaa, G.M., Amemiya, A., Eds.; University of Washington: Seattle, WA, USA, 1993. [Google Scholar]
  41. Wooten, E.C.; Iyer, L.K.; Montefusco, M.C.; Hedgepeth, A.K.; Payne, D.D.; Kapur, N.K.; Housman, D.E.; Mendelsohn, M.E.; Huggins, G.S. Application of Gene Network Analysis Techniques Identifies AXIN1/PDIA2 and Endoglin Haplotypes Associated with Bicuspid Aortic Valve. PLoS ONE 2010, 5, e8830. [Google Scholar] [CrossRef]
  42. Takeda, N.; Morita, H.; Fujita, D.; Inuzuka, R.; Taniguchi, Y.; Imai, Y.; Hirata, Y.; Komuro, I. Congenital Contractural Arachnodactyly Complicated with Aortic Dilatation and Dissection: Case Report and Review of Literature. Am. J. Med. Genet. A 2015, 167, 2382–2387. [Google Scholar] [CrossRef]
  43. Guo, D.; Gong, L.; Regalado, E.S.; Santos-Cortez, R.L.; Zhao, R.; Cai, B.; Veeraraghavan, S.; Prakash, S.K.; Johnson, R.J.; Muilenburg, A.; et al. MAT2A Mutations Predispose Individuals to Thoracic Aortic Aneurysms. Am. J. Hum. Genet. 2015, 96, 170–177. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Callewaert, B.; De Paepe, A.; Coucke, P. Arterial Tortuosity Syndrome. In GeneReviews®; Adam, M.P., Ardinger, H.H., Pagon, R.A., Wallace, S.E., Bean, L.J., Gripp, K.W., Mirzaa, G.M., Amemiya, A., Eds.; University of Washington: Seattle, WA, USA, 1993. [Google Scholar]
  45. Lillie, M.A.; David, G.J.; Gosline, J.M. Mechanical Role of Elastin-Associated Microfibrils in Pig Aortic Elastic Tissue. Connect. Tissue Res. 1998, 37, 121–141. [Google Scholar] [CrossRef] [PubMed]
  46. Creamer, T.J.; Bramel, E.E.; MacFarlane, E.G. Insights on the Pathogenesis of Aneurysm through the Study of Hereditary Aortopathies. Genes 2021, 12, 183. [Google Scholar] [CrossRef] [PubMed]
  47. Dietz, H.C.; Cutting, C.R.; Pyeritz, R.E.; Maslen, C.L.; Sakai, L.Y.; Corson, G.M.; Puffenberger, E.G.; Hamosh, A.; Nanthakumar, E.J.; Curristin, S.M.; et al. Marfan Syndrome Caused by a Recurrent de Novo Missense Mutation in the Fibrillin Gene. Nature 1991, 352, 337–339. [Google Scholar] [CrossRef]
  48. Csiszar, K. Lysyl Oxidases: A Novel Multifunctional Amine Oxidase Family. In Progress in Nucleic Acid Research and Molecular Biology; Elsevier: Amsterdam, The Netherlands, 2001; Volume 70l, pp. 1–32. ISBN 978-0-12-540070-1. [Google Scholar]
  49. Zentner, D.; James, P.; Bannon, P.; Jeremy, R. Familial Aortopathies—State of the Art Review. Heart Lung Circ. 2020, 29, 607–618. [Google Scholar] [CrossRef] [Green Version]
  50. Papke, C.L.; Yanagisawa, H. Fibulin-4 and Fibulin-5 in Elastogenesis and beyond: Insights from Mouse and Human Studies. Matrix Biol. 2014, 37, 142–149. [Google Scholar] [CrossRef]
  51. De Figueiredo Borges, L.; Jaldin, R.G.; Dias, R.R.; Stolf, N.A.G.; Michel, J.-B.; Gutierrez, P.S. Collagen Is Reduced and Disrupted in Human Aneurysms and Dissections of Ascending Aorta. Hum. Pathol. 2008, 39, 437–443. [Google Scholar] [CrossRef]
  52. Combs, M.D.; Knutsen, R.H.; Broekelmann, T.J.; Toennies, H.M.; Brett, T.J.; Miller, C.A.; Kober, D.L.; Craft, C.S.; Atkinson, J.J.; Shipley, J.M.; et al. Microfibril-Associated Glycoprotein 2 (MAGP2) Loss of Function Has Pleiotropic Effects in Vivo. J. Biol. Chem. 2013, 288, 28869–28880. [Google Scholar] [CrossRef] [Green Version]
  53. Kolb, M.; Margetts, P.J.; Sime, P.J.; Gauldie, J. Proteoglycans Decorin and Biglycan Differentially Modulate TGF-β-Mediated Fibrotic Responses in the Lung. Am. J. Physiol. Lung Cell. Mol. Physiol. 2001, 280, L1327–L1334. [Google Scholar] [CrossRef]
  54. Guo, D.-C.; Pannu, H.; Tran-Fadulu, V.; Papke, C.L.; Yu, R.K.; Avidan, N.; Bourgeois, S.; Estrera, A.L.; Safi, H.J.; Sparks, E.; et al. Mutations in Smooth Muscle α-Actin (ACTA2) Lead to Thoracic Aortic Aneurysms and Dissections. Nat. Genet. 2007, 39, 1488–1493. [Google Scholar] [CrossRef] [PubMed]
  55. Morisaki, H.; Akutsu, K.; Ogino, H.; Kondo, N.; Yamanaka, I.; Tsutsumi, Y.; Yoshimuta, T.; Okajima, T.; Matsuda, H.; Minatoya, K.; et al. Mutation of ACTA2 Gene as an Important Cause of Familial and Nonfamilial Nonsyndromatic Thoracic Aortic Aneurysm and/or Dissection (TAAD). Hum. Mutat. 2009, 30, 1406–1411. [Google Scholar] [CrossRef] [PubMed]
  56. Disabella, E.; Grasso, M.; Gambarin, F.I.; Narula, N.; Dore, R.; Favalli, V.; Serio, A.; Antoniazzi, E.; Mosconi, M.; Pasotti, M.; et al. Risk of Dissection in Thoracic Aneurysms Associated with Mutations of Smooth Muscle Alpha-Actin 2 (ACTA2). Heart Br. Card. Soc. 2011, 97, 321–326. [Google Scholar] [CrossRef]
  57. Lu, H.; Fagnant, P.M.; Bookwalter, C.S.; Joel, P.; Trybus, K.M. Vascular Disease-Causing Mutation R258C in ACTA2 Disrupts Actin Dynamics and Interaction with Myosin. Proc. Natl. Acad. Sci. USA 2015, 112, E4168–E4177. [Google Scholar] [CrossRef] [Green Version]
  58. Kim, H.; McCulloch, C.A. Filamin A Mediates Interactions between Cytoskeletal Proteins That Control Cell Adhesion. FEBS Lett. 2011, 585, 18–22. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Siddiqui, S.T.; Fisher, S.D. Heritable FLNA Gene Mutation in a Patient with Thoracic Aortic Aneurysm. JACC Case Rep. 2022, 4, 87–90. [Google Scholar] [CrossRef] [PubMed]
  60. Pomianowski, P.; Elefteriades, J.A. The Genetics and Genomics of Thoracic Aortic Disease. Ann. Cardiothorac. Surg. 2013, 2, 271–279. [Google Scholar] [CrossRef]
  61. Daugherty, A.; Chen, Z.; Sawada, H.; Rateri, D.L.; Sheppard, M.B. Transforming Growth Factor-β in Thoracic Aortic Aneurysms: Good, Bad, or Irrelevant? J. Am. Heart Assoc. 2017, 6, e005221. [Google Scholar] [CrossRef]
  62. Michel, J.-B.; Jondeau, G.; Milewicz, D.M. From Genetics to Response to Injury: Vascular Smooth Muscle Cells in Aneurysms and Dissections of the Ascending Aorta. Cardiovasc. Res. 2018, 114, 578–589. [Google Scholar] [CrossRef]
  63. MacFarlane, E.G.; Parker, S.J.; Shin, J.Y.; Ziegler, S.G.; Creamer, T.J.; Bagirzadeh, R.; Bedja, D.; Chen, Y.; Calderon, J.F.; Weissler, K.; et al. Lineage-Specific Events Underlie Aortic Root Aneurysm Pathogenesis in Loeys-Dietz Syndrome. J. Clin. Investig. 2019, 129, 659–675. [Google Scholar] [CrossRef]
  64. Lindsay, M.E.; Dietz, H.C. Lessons on the Pathogenesis of Aneurysm from Heritable Conditions. Nature 2011, 473, 308–316. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Guo, D.-C.; Regalado, E.S.; Pinard, A.; Chen, J.; Lee, K.; Rigelsky, C.; Zilberberg, L.; Hostetler, E.M.; Aldred, M.; Wallace, S.E.; et al. LTBP3 Pathogenic Variants Predispose Individuals to Thoracic Aortic Aneurysms and Dissections. Am. J. Hum. Genet. 2018, 102, 706–712. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Schepers, D.; Tortora, G.; Morisaki, H.; MacCarrick, G.; Lindsay, M.; Liang, D.; Mehta, S.G.; Hague, J.; Verhagen, J.; van de Laar, I.; et al. A Mutation Update on the LDS-Associated Genes TGFB2/3 and SMAD2/3. Hum. Mutat. 2018, 39, 621–634. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Cecconi, M.; Manfrin, M.; Moraca, A.; Zanoli, R.; Colonna, P.L.; Bettuzzi, M.G.; Moretti, S.; Gabrielli, D.; Perna, G.P. Aortic Dimensions in Patients with Bicuspid Aortic Valve without Significant Valve Dysfunction. Am. J. Cardiol. 2005, 95, 292–294. [Google Scholar] [CrossRef]
  68. Pepe, G.; Nistri, S.; Giusti, B.; Sticchi, E.; Attanasio, M.; Porciani, C.; Abbate, R.; Bonow, R.O.; Yacoub, M.; Gensini, G.F. Identification of Fibrillin 1 Gene Mutations in Patients with Bicuspid Aortic Valve (BAV) without Marfan Syndrome. BMC Med. Genet. 2014, 15, 23. [Google Scholar] [CrossRef] [Green Version]
  69. Boyum, J.; Fellinger, E.K.; Schmoker, J.D.; Trombley, L.; McPartland, K.; Ittleman, F.P.; Howard, A.B. Matrix Metalloproteinase Activity in Thoracic Aortic Aneurysms Associated with Bicuspid and Tricuspid Aortic Valves. J. Thorac. Cardiovasc. Surg. 2004, 127, 686–691. [Google Scholar] [CrossRef] [Green Version]
  70. Rabkin, S.W. Differential Expression of MMP-2, MMP-9 and TIMP Proteins in Thoracic Aortic Aneurysm - Comparison with and without Bicuspid Aortic Valve: A Meta-Analysis. Vasa 2014, 43, 433–442. [Google Scholar] [CrossRef]
  71. Martin-Blazquez, A.; Heredero, A.; Aldamiz-Echevarria, G.; Martin-Lorenzo, M.; Alvarez-Llamas, G. Non-Syndromic Thoracic Aortic Aneurysm: Cellular and Molecular Insights. J. Pathol. 2021, 254, 229–238. [Google Scholar] [CrossRef]
  72. Geng, L.; Wang, W.; Chen, Y.; Cao, J.; Lu, L.; Chen, Q.; He, R.; Shen, W. Elevation of ADAM10, ADAM17, MMP-2 and MMP-9 Expression with Media Degeneration Features CaCl2-Induced Thoracic Aortic Aneurysm in a Rat Model. Exp. Mol. Pathol. 2010, 89, 72–81. [Google Scholar] [CrossRef]
  73. Ren, P.; Zhang, L.; Xu, G.; Palmero, L.C.; Albini, P.T.; Coselli, J.S.; Shen, Y.H.; LeMaire, S.A. ADAMTS-1 and ADAMTS-4 Levels Are Elevated in Thoracic Aortic Aneurysms and Dissections. Ann. Thorac. Surg. 2013, 95, 570–577. [Google Scholar] [CrossRef] [Green Version]
  74. Wilton, E.; Bland, M.; Thompson, M.; Jahangiri, M. Matrix Metalloproteinase Expression in the Ascending Aorta and Aortic Valve. Interact. Cardiovasc. Thorac. Surg. 2008, 7, 37–40. [Google Scholar] [CrossRef] [PubMed]
  75. Evangelista, A.; Isselbacher, E.M.; Bossone, E.; Gleason, T.G.; Eusanio, M.D.; Sechtem, U.; Ehrlich, M.P.; Trimarchi, S.; Braverman, A.C.; Myrmel, T.; et al. Insights from the International Registry of Acute Aortic Dissection: A 20-Year Experience of Collaborative Clinical Research. Circulation 2018, 137, 1846–1860. [Google Scholar] [CrossRef] [PubMed]
  76. Silaschi, M.; Byrne, J.; Wendler, O. Aortic Dissection: Medical, Interventional and Surgical Management. Heart 2017, 103, 78–87. [Google Scholar] [CrossRef] [PubMed]
  77. Hagan, P.G.; Nienaber, C.A.; Isselbacher, E.M.; Bruckman, D.; Karavite, D.J.; Russman, P.L.; Evangelista, A.; Fattori, R.; Suzuki, T.; Oh, J.K.; et al. The International Registry of Acute Aortic Dissection (IRAD): New Insights into an Old Disease. JAMA 2000, 283, 897. [Google Scholar] [CrossRef]
  78. Vilacosta, I.; San Román, J.A.; di Bartolomeo, R.; Eagle, K.; Estrera, A.L.; Ferrera, C.; Kaji, S.; Nienaber, C.A.; Riambau, V.; Schäfers, H.-J.; et al. Acute Aortic Syndrome Revisited: JACC State-of-the-Art Review. J. Am. Coll. Cardiol. 2021, 78, 2106–2125. [Google Scholar] [CrossRef]
  79. White, A.; Broder, J.; Mando-Vandrick, J.; Wendell, J.; Crowe, J. Acute Aortic Emergencies—Part 2 Aortic Dissections. Adv. Emerg. Nurs. J. 2013, 35, 28–52. [Google Scholar] [CrossRef]
  80. Von Kodolitsch, Y.; Csösz, S.K.; Koschyk, D.H.; Schalwat, I.; Loose, R.; Karck, M.; Dieckmann, C.; Fattori, R.; Haverich, A.; Berger, J.; et al. Intramural Hematoma of the Aorta: Predictors of Progression to Dissection and Rupture. Circulation 2003, 107, 1158–1163. [Google Scholar] [CrossRef]
  81. Manabe, T.; Imoto, K.; Uchida, K.; Doi, C.; Takanashi, Y. Decreased Tissue Inhibitor of Metalloproteinase-2/Matrix Metalloproteinase Ratio in the Acute Phase of Aortic Dissection. Surg. Today 2004, 34, 220–225. [Google Scholar] [CrossRef]
  82. Koullias, G.J.; Ravichandran, P.; Korkolis, D.P.; Rimm, D.L.; Elefteriades, J.A. Increased Tissue Microarray Matrix Metalloproteinase Expression Favors Proteolysis in Thoracic Aortic Aneurysms and Dissections. Ann. Thorac. Surg. 2004, 78, 2106–2110. [Google Scholar] [CrossRef]
  83. Del Porto, F.; di Gioia, C.; Tritapepe, L.; Ferri, L.; Leopizzi, M.; Nofroni, I.; De Santis, V.; Della Rocca, C.; Mitterhofer, A.P.; Bruno, G.; et al. The Multitasking Role of Macrophages in Stanford Type A Acute Aortic Dissection. Cardiology 2014, 127, 123–129. [Google Scholar] [CrossRef]
  84. Landenhed, M.; Engström, G.; Gottsäter, A.; Caulfield, M.P.; Hedblad, B.; Newton-Cheh, C.; Melander, O.; Smith, J.G. Risk Profiles for Aortic Dissection and Ruptured or Surgically Treated Aneurysms: A Prospective Cohort Study. J. Am. Heart Assoc. 2015, 4, e001513. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Hahn, A.W.A.; Jonas, U.; Bühler, F.R.; Resink, T.J. Activation of Human Peripheral Monocytes by Angiotensin II. FEBS Lett. 1994, 347, 178–180. [Google Scholar] [CrossRef] [Green Version]
  86. Stumpf, C.; Jukic, J.; Yilmaz, A.; Raaz, D.; Schmieder, R.E.; Daniel, W.G.; Garlichs, C.D. Elevated VEGF-Plasma Levels in Young Patients with Mild Essential Hypertension. Eur. J. Clin. Investig. 2009, 39, 31–36. [Google Scholar] [CrossRef] [PubMed]
  87. Derosa, G.; D’Angelo, A.; Ciccarelli, L.; Piccinni, M.N.; Pricolo, F.; Salvadeo, S.; Montagna, L.; Gravina, A.; Ferrari, I.; Galli, S.; et al. Matrix Metalloproteinase-2, -9, and Tissue Inhibitor of Metalloproteinase-1 in Patients with Hypertension. Endothel. J. Endothel. Cell Res. 2006, 13, 227–231. [Google Scholar] [CrossRef]
  88. Elefteriades, J.A. Natural History of Thoracic Aortic Aneurysms: Indications for Surgery, and Surgical versus Nonsurgical Risks. Ann. Thorac. Surg. 2002, 74, S1877–S1880, discussion S1892–S1898. [Google Scholar] [CrossRef]
  89. Pape, L.A.; Tsai, T.T.; Isselbacher, E.M.; Oh, J.K.; O’gara, P.T.; Evangelista, A.; Fattori, R.; Meinhardt, G.; Trimarchi, S.; Bossone, E.; et al. Aortic Diameter > or =5.5 Cm Is Not a Good Predictor of Type A Aortic Dissection: Observations from the International Registry of Acute Aortic Dissection (IRAD). Circulation 2007, 116, 1120–1127. [Google Scholar] [CrossRef] [Green Version]
  90. Davies, R.R.; Goldstein, L.J.; Coady, M.A.; Tittle, S.L.; Rizzo, J.A.; Kopf, G.S.; Elefteriades, J.A. Yearly Rupture or Dissection Rates for Thoracic Aortic Aneurysms: Simple Prediction Based on Size. Ann. Thorac. Surg. 2002, 73, 17–27, discussion 27–28. [Google Scholar] [CrossRef]
  91. Gawinecka, J.; Schönrath, F.; von Eckardstein, A. Acute Aortic Dissection: Pathogenesis, Risk Factors and Diagnosis. Swiss Med. Wkly. 2017, 147, w14489. [Google Scholar] [CrossRef]
  92. Pyeritz, R.E. Recent Progress in Understanding the Natural and Clinical Histories of the Marfan Syndrome. Trends Cardiovasc. Med. 2016, 26, 423–428. [Google Scholar] [CrossRef]
  93. Weinsaft, J.W.; Devereux, R.B.; Preiss, L.R.; Feher, A.; Roman, M.J.; Basson, C.T.; Geevarghese, A.; Ravekes, W.; Dietz, H.C.; Holmes, K.; et al. Aortic Dissection in Patients with Genetically Mediated Aneurysms. J. Am. Coll. Cardiol. 2016, 67, 2744–2754. [Google Scholar] [CrossRef]
  94. Den Hartog, A.W.; Franken, R.; Zwinderman, A.H.; Timmermans, J.; Scholte, A.J.; van den Berg, M.P.; de Waard, V.; Pals, G.; Mulder, B.J.M.; Groenink, M. The Risk for Type B Aortic Dissection in Marfan Syndrome. J. Am. Coll. Cardiol. 2015, 65, 246–254. [Google Scholar] [CrossRef]
  95. Pape, L.A.; Awais, M.; Woznicki, E.M.; Suzuki, T.; Trimarchi, S.; Evangelista, A.; Myrmel, T.; Larsen, M.; Harris, K.M.; Greason, K.; et al. Presentation, Diagnosis, and Outcomes of Acute Aortic Dissection: 17-Year Trends from the International Registry of Acute Aortic Dissection. J. Am. Coll. Cardiol. 2015, 66, 350–358. [Google Scholar] [CrossRef] [Green Version]
  96. Tran-Fadulu, V.; Pannu, H.; Kim, D.H.; Vick, G.W.; Lonsford, C.M.; Lafont, A.L.; Boccalandro, C.; Smart, S.; Peterson, K.L.; Hain, J.Z.; et al. Analysis of Multigenerational Families with Thoracic Aortic Aneurysms and Dissections Due to TGFBR1 or TGFBR2 Mutations. J. Med. Genet. 2009, 46, 607–613. [Google Scholar] [CrossRef] [PubMed]
  97. Jondeau, G.; Ropers, J.; Regalado, E.; Braverman, A.; Evangelista, A.; Teixedo, G.; De Backer, J.; Muiño-Mosquera, L.; Naudion, S.; Zordan, C.; et al. International Registry of Patients Carrying TGFBR1 or TGFBR2 Mutations: Results of the MAC (Montalcino Aortic Consortium). Circ. Cardiovasc. Genet. 2016, 9, 548–558. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Van de Laar, I.M.B.H.; Oldenburg, R.A.; Pals, G.; Roos-Hesselink, J.W.; de Graaf, B.M.; Verhagen, J.M.A.; Hoedemaekers, Y.M.; Willemsen, R.; Severijnen, L.-A.; Venselaar, H.; et al. Mutations in SMAD3 Cause a Syndromic Form of Aortic Aneurysms and Dissections with Early-Onset Osteoarthritis. Nat. Genet. 2011, 43, 121–126. [Google Scholar] [CrossRef] [PubMed]
  99. Laterza, D.; Ritelli, M.; Zini, A.; Colombi, M.; Dell’Acqua, M.L.; Vandelli, L.; Bigliardi, G.; Verganti, L.; Vallone, S.; Vincenzi, C.; et al. Novel Pathogenic TGFBR1 and SMAD3 Variants Identified after Cerebrovascular Events in Adult Patients with Loeys-Dietz Syndrome. Eur. J. Med. Genet. 2019, 62, 103727. [Google Scholar] [CrossRef]
  100. Zhao, H.; Yang, Y.; Pan, X.; Li, W.; Sun, L.; Guo, J. Identification of Clinically Relevant Variants by Whole Exome Sequencing in Chinese Patients with Sporadic Non-Syndromic Type A Aortic Dissection. Clin. Chim. Acta 2020, 506, 160–165. [Google Scholar] [CrossRef]
  101. Hostetler, E.M.; Regalado, E.S.; Guo, D.-C.; Hanna, N.; Arnaud, P.; Muiño-Mosquera, L.; Callewaert, B.L.; Lee, K.; Leal, S.M.; Wallace, S.E.; et al. SMAD3 Pathogenic Variants: Risk for Thoracic Aortic Disease and Associated Complications from the Montalcino Aortic Consortium. J. Med. Genet. 2019, 56, 252–260. [Google Scholar] [CrossRef]
  102. Hilhorst-Hofstee, Y.; Scholte, A.J.H.A.; Rijlaarsdam, M.E.B.; van Haeringen, A.; Kroft, L.J.; Reijnierse, M.; Ruivenkamp, C.A.L.; Versteegh, M.I.M.; Pals, G.; Breuning, M.H. An Unanticipated Copy Number Variant of Chromosome 15 Disrupting SMAD3 Reveals a Three-Generation Family at Serious Risk for Aortic Dissection. Clin. Genet. 2013, 83, 337–344. [Google Scholar] [CrossRef]
  103. Lee, S.-T.; Kim, J.-A.; Jang, S.-Y.; Kim, D.-K.; Kim, J.-W.; Ki, C.-S. A Novel COL3A1 Gene Mutation in Patient with Aortic Dissected Aneurysm and Cervical Artery Dissections. Heart Vessels 2008, 23, 144–148. [Google Scholar] [CrossRef]
  104. Koitabashi, N.; Yamaguchi, T.; Fukui, D.; Nakano, T.; Umeyama, A.; Toda, K.; Funada, R.; Ishikawa, M.; Kawamura, R.; Okada, K.; et al. Peripartum Iliac Arterial Aneurysm and Rupture in a Patient with Vascular Ehlers-Danlos Syndrome Diagnosed by Next-Generation Sequencing. Int. Heart J. 2018, 59, 1180–1185. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Makrygiannis, G.; Loeys, B.; Defraigne, J.-O.; Sakalihasan, N. Cervical Artery Dissections and Type A Aortic Dissection in a Family with a Novel Missense COL3A1 Mutation of Vascular Type Ehlers–Danlos Syndrome. Eur. J. Med. Genet. 2015, 58, 634–636. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Nakamura, M.; Yajima, J.; Oikawa, Y.; Ogasawara, K.; Uejima, T.; Abe, K.; Aizawa, T. Vascular Ehlers-Danlos Syndrome. J. Cardiol. 2009, 53, 458–462. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Shields, L.B.E.; Rolf, C.M.; Davis, G.J.; Hunsaker III, J.C. Sudden and Unexpected Death in Three Cases of Ehlers-Danlos Syndrome Type IV*: SUDDEN DEATH IN EHLERS-DANLOS TYPE IV. J. Forensic Sci. 2010, 55, 1641–1645. [Google Scholar] [CrossRef]
  108. Schwarze, U.; Goldstein, J.A.; Byers, P.H. Splicing Defects in the COL3A1 Gene: Marked Preference for 5′ (Donor) Spice-Site Mutations in Patients with Exon-Skipping Mutations and Ehlers-Danlos Syndrome Type IV. Am. J. Hum. Genet. 1997, 61, 1276–1286. [Google Scholar] [CrossRef] [Green Version]
  109. Sakai, K.; Toda, M.; Kyoyama, H.; Nishimura, H.; Kojima, A.; Kuwabara, Y.; Kobayashi, Y.; Kikuchi, S.; Hirata, Y.; Moriyama, G.; et al. Vascular Ehlers-Danlos Syndrome with a Novel Missense Mutation in COL3A1: A Man in His 50s with Aortic Dissection after Interventional Treatment for Hemothorax as the First Manifestation. Intern. Med. Tokyo Jpn. 2019, 58, 3441–3447. [Google Scholar] [CrossRef] [Green Version]
  110. Shalhub, S.; Black, J.H.; Cecchi, A.C.; Xu, Z.; Griswold, B.F.; Safi, H.J.; Milewicz, D.M.; McDonnell, N.B. Molecular Diagnosis in Vascular Ehlers-Danlos Syndrome Predicts Pattern of Arterial Involvement and Outcomes. J. Vasc. Surg. 2014, 60, 160–169. [Google Scholar] [CrossRef] [Green Version]
  111. Meienberg, J.; Rohrbach, M.; Neuenschwander, S.; Spanaus, K.; Giunta, C.; Alonso, S.; Arnold, E.; Henggeler, C.; Regenass, S.; Patrignani, A.; et al. Hemizygous Deletion of COL3A1, COL5A2, and MSTN Causes a Complex Phenotype with Aortic Dissection: A Lesson for and from True Haploinsufficiency. Eur. J. Hum. Genet. 2010, 18, 1315–1321. [Google Scholar] [CrossRef] [Green Version]
  112. Wang, Z.; Zhuang, X.; Chen, B.; Wen, J.; Peng, F.; Liu, X.; Wei, M. 99-Case Study of Sporadic Aortic Dissection by Whole Exome Sequencing Indicated Novel Disease-Associated Genes and Variants in Chinese Population. BioMed Res. Int. 2020, 2020, 7857043. [Google Scholar] [CrossRef]
  113. Chen, Y.; Sun, Y.; Li, Z.; Li, C.; Xiao, L.; Dai, J.; Li, S.; Liu, H.; Hu, D.; Wu, D.; et al. Identification of COL3A1 Variants Associated with Sporadic Thoracic Aortic Dissection: A Case-Control Study. Front. Med. 2021, 15, 438–447. [Google Scholar] [CrossRef]
  114. Frank, M.; Albuisson, J.; Ranque, B.; Golmard, L.; Mazzella, J.-M.; Bal-Theoleyre, L.; Fauret, A.-L.; Mirault, T.; Denarié, N.; Mousseaux, E.; et al. The Type of Variants at the COL3A1 Gene Associates with the Phenotype and Severity of Vascular Ehlers–Danlos Syndrome. Eur. J. Hum. Genet. 2015, 23, 1657–1664. [Google Scholar] [CrossRef] [PubMed]
  115. Pepin, M.G.; Schwarze, U.; Rice, K.M.; Liu, M.; Leistritz, D.; Byers, P.H. Survival Is Affected by Mutation Type and Molecular Mechanism in Vascular Ehlers-Danlos Syndrome (EDS Type IV). Genet. Med. Off. J. Am. Coll. Med. Genet. 2014, 16, 881–888. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Saratzis, A.; Bown, M.J. The Genetic Basis for Aortic Aneurysmal Disease. Heart Br. Card. Soc. 2014, 100, 916–922. [Google Scholar] [CrossRef] [PubMed]
  117. Guo, D.; Regalado, E.; Casteel, D.E.; Santos-Cortez, R.L.; Gong, L.; Kim, J.J.; Dyack, S.; Horne, S.G.; Chang, G.; Jondeau, G.; et al. Recurrent Gain-of-Function Mutation in PRKG1 Causes Thoracic Aortic Aneurysms and Acute Aortic Dissections. Am. J. Hum. Genet. 2013, 93, 398–404. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Mokashi, S.A.; Svensson, L.G. Guidelines for the Management of Thoracic Aortic Disease in 2017. Gen. Thorac. Cardiovasc. Surg. 2019, 67, 59–65. [Google Scholar] [CrossRef]
  119. Elefteriades, J.A. Thoracic Aortic Aneurysm: Reading the Enemy’s Playbook. Curr. Probl. Cardiol. 2008, 33, 203–277. [Google Scholar] [CrossRef]
  120. Rogers, A.M.; Hermann, L.K.; Booher, A.M.; Nienaber, C.A.; Williams, D.M.; Kazerooni, E.A.; Froehlich, J.B.; O’Gara, P.T.; Montgomery, D.G.; Cooper, J.V.; et al. Sensitivity of the Aortic Dissection Detection Risk Score, a Novel Guideline-Based Tool for Identification of Acute Aortic Dissection at Initial Presentation: Results from the International Registry of Acute Aortic Dissection. Circulation 2011, 123, 2213–2218. [Google Scholar] [CrossRef] [Green Version]
  121. Morello, F.; Piler, P.; Novak, M.; Kruzliak, P. Biomarkers for Diagnosis and Prognostic Stratification of Aortic Dissection: Challenges and Perspectives. Biomark. Med. 2014, 8, 931–941. [Google Scholar] [CrossRef]
  122. Nazerian, P.; Giachino, F.; Vanni, S.; Veglio, M.G.; Castelli, M.; Lison, D.; Bitossi, L.; Moiraghi, C.; Grifoni, S.; Morello, F. Diagnostic Performance of the Aortic Dissection Detection Risk Score in Patients with Suspected Acute Aortic Dissection. Eur. Heart J. Acute Cardiovasc. Care 2014, 3, 373–381. [Google Scholar] [CrossRef]
  123. Torbicki, A.; Perrier, A.; Konstantinides, S.; Agnelli, G.; Galiè, N.; Pruszczyk, P.; Bengel, F.; Brady, A.J.B.; Ferreira, D.; Janssens, U.; et al. Guidelines on the Diagnosis and Management of Acute Pulmonary Embolism: The Task Force for the Diagnosis and Management of Acute Pulmonary Embolism of the European Society of Cardiology (ESC). Eur. Heart J. 2008, 29, 2276–2315. [Google Scholar] [CrossRef] [Green Version]
  124. Weber, T.; Högler, S.; Auer, J.; Berent, R.; Lassnig, E.; Kvas, E.; Eber, B. D-Dimer in Acute Aortic Dissection. Chest 2003, 123, 1375–1378. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Eggebrecht, H.; Naber, C.K.; Bruch, C.; Kröger, K.; von Birgelen, C.; Schmermund, A.; Wichert, M.; Bartel, T.; Mann, K.; Erbel, R. Value of Plasma Fibrin D-Dimers for Detection of Acute Aortic Dissection. J. Am. Coll. Cardiol. 2004, 44, 804–809. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Suzuki, T.; Distante, A.; Zizza, A.; Trimarchi, S.; Villani, M.; Salerno Uriarte, J.A.; De Luca Tupputi Schinosa, L.; Renzulli, A.; Sabino, F.; Nowak, R.; et al. Diagnosis of Acute Aortic Dissection by D-Dimer: The International Registry of Acute Aortic Dissection Substudy on Biomarkers (IRAD-Bio) Experience. Circulation 2009, 119, 2702–2707. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Shimony, A.; Filion, K.B.; Mottillo, S.; Dourian, T.; Eisenberg, M.J. Meta-Analysis of Usefulness of d-Dimer to Diagnose Acute Aortic Dissection. Am. J. Cardiol. 2011, 107, 1227–1234. [Google Scholar] [CrossRef] [PubMed]
  128. Brown, M.D.; Newman, D.H. Evidence-Based Emergency Medicine. Can a Negative D-Dimer Result Rule out Acute Aortic Dissection? Ann. Emerg. Med. 2011, 58, 375–376. [Google Scholar] [CrossRef] [PubMed]
  129. Schmoker, J.D.; McPartland, K.J.; Fellinger, E.K.; Boyum, J.; Trombley, L.; Ittleman, F.P.; Terrien, C.; Stanley, A.; Howard, A. Matrix Metalloproteinase and Tissue Inhibitor Expression in Atherosclerotic and Nonatherosclerotic Thoracic Aortic Aneurysms. J. Thorac. Cardiovasc. Surg. 2007, 133, 155–161. [Google Scholar] [CrossRef] [Green Version]
  130. Sinha, I.; Bethi, S.; Cronin, P.; Williams, D.M.; Roelofs, K.; Ailawadi, G.; Henke, P.K.; Eagleton, M.J.; Deeb, G.M.; Patel, H.J.; et al. A Biologic Basis for Asymmetric Growth in Descending Thoracic Aortic Aneurysms: A Role for Matrix Metalloproteinase 9 and 2. J. Vasc. Surg. 2006, 43, 342–348. [Google Scholar] [CrossRef] [Green Version]
  131. Giachino, F.; Loiacono, M.; Lucchiari, M.; Manzo, M.; Battista, S.; Saglio, E.; Lupia, E.; Moiraghi, C.; Hirsch, E.; Mengozzi, G.; et al. Rule out of Acute Aortic Dissection with Plasma Matrix Metalloproteinase 8 in the Emergency Department. Crit. Care 2013, 17, R33. [Google Scholar] [CrossRef] [Green Version]
  132. Li, T.; Jiang, B.; Li, X.; Sun, H.; Li, X.; Jing, J.; Yang, J. Serum Matrix Metalloproteinase-9 Is a Valuable Biomarker for Identification of Abdominal and Thoracic Aortic Aneurysm: A Case-Control Study. BMC Cardiovasc. Disord. 2018, 18, 202. [Google Scholar] [CrossRef] [Green Version]
  133. Song, Y.; Xie, Y.; Liu, F.; Zhao, C.; Yu, R.; Ban, S.; Ye, Q.; Wen, J.; Wan, H.; Li, X.; et al. Expression of Matrix Metalloproteinase-12 in Aortic Dissection. BMC Cardiovasc. Disord. 2013, 13, 34. [Google Scholar] [CrossRef] [Green Version]
  134. Van Bogerijen, G.H.W.; Tolenaar, J.L.; Grassi, V.; Lomazzi, C.; Segreti, S.; Rampoldi, V.; Elefteriades, J.A.; Trimarchi, S. Biomarkers in TAA-the Holy Grail. Prog. Cardiovasc. Dis. 2013, 56, 109–115. [Google Scholar] [CrossRef] [PubMed]
  135. Wilson, K.A.; Lindholt, J.S.; Hoskins, P.R.; Heickendorff, L.; Vammen, S.; Bradbury, A.W. The Relationship Between Abdominal Aortic Aneurysm Distensibility and Serum Markers of Elastin and Collagen Metabolism. Eur. J. Vasc. Endovasc. Surg. 2001, 21, 175–178. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Shinohara, T.; Suzuki, K.; Okada, M.; Shiigai, M.; Shimizu, M.; Maehara, T.; Ohsuzu, F. Soluble Elastin Fragments in Serum Are Elevated in Acute Aortic Dissection. Arterioscler. Thromb. Vasc. Biol. 2003, 23, 1839–1844. [Google Scholar] [CrossRef] [PubMed]
  137. Yuan, S.-M.; Shi, Y.-H.; Wang, J.-J.; Lü, F.-Q.; Gao, S. Elevated Plasma D-Dimer and Hypersensitive C-Reactive Protein Levels May Indicate Aortic Disorders. Rev. Bras. Cir. Cardiovasc. Orgao Off. Soc. Bras. Cir. Cardiovasc. 2011, 26, 573–581. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. Wen, D.; Du, X.; Dong, J.-Z.; Zhou, X.-L.; Ma, C.-S. Value of D-Dimer and C Reactive Protein in Predicting Inhospital Death in Acute Aortic Dissection. Heart Br. Card. Soc. 2013, 99, 1192–1197. [Google Scholar] [CrossRef] [Green Version]
  139. Wen, D.; Zhou, X.-L.; Li, J.-J.; Luo, F.; Zhang, L.; Gao, L.-G.; Wang, L.-P.; Song, L.; Sun, K.; Zou, Y.-B.; et al. Plasma Concentrations of Interleukin-6, C-Reactive Protein, Tumor Necrosis Factor-α and Matrix Metalloproteinase-9 in Aortic Dissection. Clin. Chim. Acta Int. J. Clin. Chem. 2012, 413, 198–202. [Google Scholar] [CrossRef]
  140. Erdolu, B.; As, A.K. C-Reactive Protein and Neutrophil to Lymphocyte Ratio Values in Predicting Inhospital Death in Patients with Stanford Type A Acute Aortic Dissection. Heart Surg. Forum 2020, 23, E488–E492. [Google Scholar] [CrossRef]
  141. Sbarouni, E.; Georgiadou, P.; Analitis, A.; Chaidaroglou, A.; Marathias, A.; Degiannis, D.; Voudris, V. High Homocysteine and Low Folate Concentrations in Acute Aortic Dissection. Int. J. Cardiol. 2013, 168, 463–466. [Google Scholar] [CrossRef]
  142. Roohi, J.; Kang, B.; Bernard, D.; Bedja, D.; Dietz, H.C.; Brody, L.C. Moderately Elevated Homocysteine Does Not Contribute to Thoracic Aortic Aneurysm in Mice. J. Nutr. 2017, 147, 1290–1295. [Google Scholar] [CrossRef] [Green Version]
  143. Giusti, B.; Porciani, M.C.; Brunelli, T.; Evangelisti, L.; Fedi, S.; Gensini, G.F.; Abbate, R.; Sani, G.; Yacoub, M.; Pepe, G. Phenotypic Variability of Cardiovascular Manifestations in Marfan Syndrome. Possible Role of Hyperhomocysteinemia and C677T MTHFR Gene Polymorphism. Eur. Heart J. 2003, 24, 2038–2045. [Google Scholar] [CrossRef] [Green Version]
  144. Giusti, B.; Marcucci, R.; Lapini, I.; Sestini, I.; Lenti, M.; Yacoub, M.; Pepe, G. Role of Hyperhomocysteinemia in Aortic Disease. Cell. Mol. Biol. Noisy Gd. Fr. 2004, 50, 945–952. [Google Scholar]
  145. Loeys, B.L.; Schwarze, U.; Holm, T.; Callewaert, B.L.; Thomas, G.H.; Pannu, H.; De Backer, J.F.; Oswald, G.L.; Symoens, S.; Manouvrier, S.; et al. Aneurysm Syndromes Caused by Mutations in the TGF-Beta Receptor. N. Engl. J. Med. 2006, 355, 788–798. [Google Scholar] [CrossRef] [PubMed]
  146. Braverman, A.C. Transforming Growth Factor-β: A Biomarker in Marfan Syndrome? Circulation 2009, 120, 464–466. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Stengl, R.; Ágg, B.; Pólos, M.; Mátyás, G.; Szabó, G.; Merkely, B.; Radovits, T.; Szabolcs, Z.; Benke, K. Potential Predictors of Severe Cardiovascular Involvement in Marfan Syndrome: The Emphasized Role of Genotype–Phenotype Correlations in Improving Risk Stratification—A Literature Review. Orphanet J. Rare Dis. 2021, 16, 245. [Google Scholar] [CrossRef]
  148. Matt, P.; Habashi, J.; Carrel, T.; Cameron, D.E.; Van Eyk, J.E.; Dietz, H.C. Recent Advances in Understanding Marfan Syndrome: Should We Now Treat Surgical Patients with Losartan? J. Thorac. Cardiovasc. Surg. 2008, 135, 389–394. [Google Scholar] [CrossRef] [Green Version]
  149. Franken, R.; den Hartog, A.W.; de Waard, V.; Engele, L.; Radonic, T.; Lutter, R.; Timmermans, J.; Scholte, A.J.; van den Berg, M.P.; Zwinderman, A.H.; et al. Circulating Transforming Growth Factor-β as a Prognostic Biomarker in Marfan Syndrome. Int. J. Cardiol. 2013, 168, 2441–2446. [Google Scholar] [CrossRef]
  150. Ogawa, N.; Imai, Y.; Nishimura, H.; Kato, M.; Takeda, N.; Nawata, K.; Taketani, T.; Morota, T.; Takamoto, S.; Nagai, R.; et al. Circulating Transforming Growth Factor β-1 Level in Japanese Patients with Marfan Syndrome. Int. Heart. J. 2013, 54, 23–26. [Google Scholar] [CrossRef] [Green Version]
  151. Forteza, A.; Evangelista, A.; Sánchez, V.; Teixidó, G.; García, D.; Sanz, P.; Gutiérrez, L.; Centeno, J.; Rodríguez-Palomares, J.; Cortina, J.; et al. Valoración de la eficacia y la seguridad del losartán frente al atenolol en la prevención de la dilatación de la aorta en el síndrome de Marfan. Rev. Esp. Cardiol. 2011, 64, 492–498. [Google Scholar] [CrossRef]
  152. Suzuki, T.; Trimarchi, S.; Sawaki, D.; Grassi, V.; Costa, E.; Rampoldi, V.; Nagai, R.; Eagle, K. Circulating Transforming Growth Factor-Beta Levels in Acute Aortic Dissection. J. Am. Coll. Cardiol. 2011, 58, 775. [Google Scholar] [CrossRef] [Green Version]
  153. Yuan, S.-M.; Lin, H. Expressions of Transforming Growth Factor Β1 Signaling Cytokines in Aortic Dissection. Braz. J. Cardiovasc. Surg. 2018, 33, 597–602. [Google Scholar] [CrossRef]
  154. Tellides, G. Further Evidence Supporting a Protective Role of Transforming Growth Factor-β (TGFβ) in Aortic Aneurysm and Dissection. Arterioscler. Thromb. Vasc. Biol. 2017, 37, 1983–1986. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Hara, H.; Takeda, N.; Fujiwara, T.; Yagi, H.; Maemura, S.; Kanaya, T.; Nawata, K.; Morita, H.; Komuro, I. Activation of TGF-β Signaling in an Aortic Aneurysm in a Patient with Loeys-Dietz Syndrome Caused by a Novel Loss-of-Function Variant of TGFBR1. Hum. Genome Var. 2019, 6, 6. [Google Scholar] [CrossRef] [PubMed]
  156. Lane, B.A.; Chakrabarti, M.; Ferruzzi, J.; Azhar, M.; Eberth, J.F. Mechanics of Ascending Aortas from TGFβ-1, -2, -3 Haploinsufficient Mice and Elastase-Induced Aortopathy. J. Biomech. 2021, 125, 110543. [Google Scholar] [CrossRef] [PubMed]
  157. Hillebrand, M.; Millot, N.; Sheikhzadeh, S.; Rybczynski, M.; Gerth, S.; Kölbel, T.; Keyser, B.; Kutsche, K.; Robinson, P.N.; Berger, J.; et al. Total Serum Transforming Growth Factor-Β1 Is Elevated in the Entire Spectrum of Genetic Aortic Syndromes: TGF-Β1 Serum Levels in GAS. Clin. Cardiol. 2014, 37, 672–679. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  158. Katoh, H.; Suzuki, T.; Hiroi, Y.; Ohtaki, E.; Suzuki, S.; Yazaki, Y.; Nagai, R. Diagnosis of Aortic Dissection by Immunoassay for Circulating Smooth Muscle Myosin. Lancet 1995, 345, 191–192. [Google Scholar] [CrossRef]
  159. Suzuki, T.; Katoh, H.; Watanabe, M.; Kurabayashi, M.; Hiramori, K.; Hori, S.; Nobuyoshi, M.; Tanaka, H.; Kodama, K.; Sato, H.; et al. Novel Biochemical Diagnostic Method for Aortic Dissection: Results of a Prospective Study Using an Immunoassay of Smooth Muscle Myosin Heavy Chain. Circulation 1996, 93, 1244–1249. [Google Scholar] [CrossRef]
  160. Suzuki, T.; Katoh, H.; Tsuchio, Y.; Hasegawa, A.; Kurabayashi, M.; Ohira, A.; Hiramori, K.; Sakomura, Y.; Kasanuki, H.; Hori, S.; et al. Diagnostic Implications of Elevated Levels of Smooth-Muscle Myosin Heavy-Chain Protein in Acute Aortic Dissection. The Smooth Muscle Myosin Heavy Chain Study. Ann. Intern. Med. 2000, 133, 537–541. [Google Scholar] [CrossRef]
  161. Davidson, E.; Weinberger, I.; Rotenberg, Z.; Fuchs, J.; Maler, S.; Agmon, J. Elevated Serum Creatine Kinase Levels. An Early Diagnostic Sign of Acute Dissection of the Aorta. Arch. Intern. Med. 1988, 148, 2184–2186. [Google Scholar] [CrossRef]
  162. Suzuki, T.; Katoh, H.; Kurabayashi, M.; Yazaki, Y.; Nagai, R. Biochemical Diagnosis of Aortic Dissection by Raised Concentrations of Creatine Kinase BB-Isozyme. Lancet 1997, 350, 784–785. [Google Scholar] [CrossRef]
  163. Suzuki, T.; Distante, A.; Zizza, A.; Trimarchi, S.; Villani, M.; Salerno Uriarte, J.A.; de Luca Tupputi Schinosa, L.; Renzulli, A.; Sabino, F.; Nowak, R.; et al. Preliminary Experience with the Smooth Muscle Troponin-like Protein, Calponin, as a Novel Biomarker for Diagnosing Acute Aortic Dissection. Eur. Heart J. 2008, 29, 1439–1445. [Google Scholar] [CrossRef] [Green Version]
  164. Peng, W.; Peng, Z.; Chai, X.; Zhu, Q.; Yang, G.; Zhao, Q.; Zhou, S. Potential Biomarkers for Early Diagnosis of Acute Aortic Dissection. Heart Lung J. Crit. Care 2015, 44, 205–208. [Google Scholar] [CrossRef] [PubMed]
  165. Zhang, S.; Qian, H.; Yang, Q.; Hu, J.; Gan, C.; Meng, W. Relationship between the Extent of Dissection and Platelet Activation in Acute Aortic Dissection. J. Cardiothorac. Surg. 2015, 10, 162. [Google Scholar] [CrossRef] [Green Version]
  166. Sbarouni, E.; Georgiadou, P.; Analitis, A.; Voudris, V. Significant Changes in Platelet Count, Volume and Size in Acute Aortic Dissection. Int. J. Cardiol. 2013, 168, 4349–4350. [Google Scholar] [CrossRef]
  167. Huang, B.; Tian, L.; Fan, X.; Zhu, J.; Liang, Y.; Yang, Y. Low Admission Platelet Counts Predicts Increased Risk of In-Hospital Mortality in Patients with Type A Acute Aortic Dissection. Int. J. Cardiol. 2014, 172, e484–e486. [Google Scholar] [CrossRef] [PubMed]
  168. König, K.C.; Lahm, H.; Dreßen, M.; Doppler, S.A.; Eichhorn, S.; Beck, N.; Kraehschuetz, K.; Doll, S.; Holdenrieder, S.; Kastrati, A.; et al. Aggrecan: A New Biomarker for Acute Type A Aortic Dissection. Sci. Rep. 2021, 11, 10371. [Google Scholar] [CrossRef] [PubMed]
  169. Li, G.; Wu, X.-W.; Lu, W.-H.; Cheng, J.; Wu, X.-Y.; Ai, R.; Zhou, Z.-H.; Tang, Z.-Z.; Liao, Y.-H. High-Sensitivity Cardiac Troponin T: A Biomarker for the Early Risk Stratification of Type-A Acute Aortic Dissection? Arch. Cardiovasc. Dis. 2016, 109, 163–170. [Google Scholar] [CrossRef] [PubMed]
  170. Yang, Y.; Jiao, X.; Li, L.; Hu, C.; Zhang, X.; Pan, L.; Yu, H.; Li, J.; Chen, D.; Du, J.; et al. Increased Circulating Angiopoietin-Like Protein 8 Levels Are Associated with Thoracic Aortic Dissection and Higher Inflammatory Conditions. Cardiovasc. Drugs Ther. 2020, 34, 65–77. [Google Scholar] [CrossRef] [Green Version]
  171. Jerves, T.; Beaton, A.; Kruszka, P. The Genetic Workup for Structural Congenital Heart Disease. Am. J. Med. Genet. C Semin. Med. Genet. 2020, 184, 178–186. [Google Scholar] [CrossRef]
  172. Rigelsky, C.M.; Moran, R.T. Genetics of Syndromic and Nonsyndromic Aortopathies. Curr. Opin. Pediatr. 2019, 31, 694–701. [Google Scholar] [CrossRef]
  173. SEC Working Group for ESC 2014 Guidelines on Diagnosis and Treatment of Aortic Diseases; Expert Reviewers for ESC 2014 Guidelines on Diagnosis and Treatment of Aortic Diseases; SEC Clinical Practice Guidelines Committee. Comments on the 2014 ESC Guidelines on the Diagnosis and Treatment of Aortic Diseases. Rev. Esp. Cardiol. Engl. Ed. 2015, 68, 179–184. [Google Scholar] [CrossRef]
  174. Writing Group Members; Hiratzka, L.F.; Bakris, G.L.; Beckman, J.A.; Bersin, R.M.; Carr, V.F.; Casey, D.E.; Eagle, K.A.; Hermann, L.K.; Isselbacher, E.M.; et al. 2010 ACCF/AHA/AATS/ACR/ASA/SCA/SCAI/SIR/STS/SVM Guidelines for the Diagnosis and Management of Patients with Thoracic Aortic Disease: A Report of the American College of Cardiology Foundation/American Heart Association Task Force on Practice Guidelines, American Association for Thoracic Surgery, American College of Radiology, American Stroke Association, Society of Cardiovascular Anesthesiologists, Society for Cardiovascular Angiography and Interventions, Society of Interventional Radiology, Society of Thoracic Surgeons, and Society for Vascular Medicine. Circulation 2010, 121, e266–e369. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  175. Guo, D.-C.; Hostetler, E.M.; Fan, Y.; Kulmacz, R.J.; Zhang, D.; GenTAC Investigators; Nickerson, D.A.; Leal, S.M.; LeMaire, S.A.; Regalado, E.S.; et al. Heritable Thoracic Aortic Disease Genes in Sporadic Aortic Dissection. J. Am. Coll. Cardiol. 2017, 70, 2728–2730. [Google Scholar] [CrossRef] [PubMed]
  176. Hicks, K.L.; Byers, P.H.; Quiroga, E.; Pepin, M.G.; Shalhub, S. Testing Patterns for Genetically Triggered Aortic and Arterial Aneurysms and Dissections at an Academic Center. J. Vasc. Surg. 2018, 68, 701–711. [Google Scholar] [CrossRef] [PubMed]
  177. Ripperger, T.; Tröger, H.D.; Schmidtke, J. The Genetic Message of a Sudden, Unexpected Death Due to Thoracic Aortic Dissection. Forensic Sci. Int. 2009, 187, 1–5. [Google Scholar] [CrossRef]
  178. Boileau, A.; Lindsay, M.; Michel, J.-B.; Devaux, Y. Epigenetics in Ascending Thoracic Aortic Aneurysm and Dissection. AORTA 2018, 06, 001–012. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  179. Wang, Y.; Barbacioru, C.C.; Shiffman, D.; Balasubramanian, S.; Iakoubova, O.; Tranquilli, M.; Albornoz, G.; Blake, J.; Mehmet, N.N.; Ngadimo, D.; et al. Gene Expression Signature in Peripheral Blood Detects Thoracic Aortic Aneurysm. PLoS ONE 2007, 2, e1050. [Google Scholar] [CrossRef] [PubMed]
  180. Boon, R.A.; Seeger, T.; Heydt, S.; Fischer, A.; Hergenreider, E.; Horrevoets, A.J.G.; Vinciguerra, M.; Rosenthal, N.; Sciacca, S.; Pilato, M.; et al. MicroRNA-29 in Aortic Dilation: Implications for Aneurysm Formation. Circ. Res. 2011, 109, 1115–1119. [Google Scholar] [CrossRef] [Green Version]
  181. Merk, D.R.; Chin, J.T.; Dake, B.A.; Maegdefessel, L.; Miller, M.O.; Kimura, N.; Tsao, P.S.; Iosef, C.; Berry, G.J.; Mohr, F.W.; et al. MiR-29b Participates in Early Aneurysm Development in Marfan Syndrome. Circ. Res. 2012, 110, 312–324. [Google Scholar] [CrossRef]
  182. Van Rooij, E. The Art of MicroRNA Research. Circ. Res. 2011, 108, 219–234. [Google Scholar] [CrossRef]
  183. Elia, L.; Quintavalle, M.; Zhang, J.; Contu, R.; Cossu, L.; Latronico, M.V.G.; Peterson, K.L.; Indolfi, C.; Catalucci, D.; Chen, J.; et al. The Knockout of MiR-143 and -145 Alters Smooth Muscle Cell Maintenance and Vascular Homeostasis in Mice: Correlates with Human Disease. Cell Death Differ. 2009, 16, 1590–1598. [Google Scholar] [CrossRef]
  184. Cheng, Y.; Liu, X.; Yang, J.; Lin, Y.; Xu, D.-Z.; Lu, Q.; Deitch, E.A.; Huo, Y.; Delphin, E.S.; Zhang, C. MicroRNA-145, a Novel Smooth Muscle Cell Phenotypic Marker and Modulator, Controls Vascular Neointimal Lesion Formation. Circ. Res. 2009, 105, 158–166. [Google Scholar] [CrossRef] [PubMed]
  185. Jones, J.A.; Stroud, R.E.; O’Quinn, E.C.; Black, L.E.; Barth, J.L.; Elefteriades, J.A.; Bavaria, J.E.; Gorman, J.H.; Gorman, R.C.; Spinale, F.G.; et al. Selective MicroRNA Suppression in Human Thoracic Aneurysms: Relationship of MiR-29a to Aortic Size and Proteolytic Induction. Circ. Cardiovasc. Genet. 2011, 4, 605–613. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  186. Venkatesh, P.; Phillippi, J.; Chukkapalli, S.; Rivera-Kweh, M.; Velsko, I.; Gleason, T.; VanRyzin, P.; Aalaei-Andabili, S.; Ghanta, R.; Beaver, T.; et al. Aneurysm-Specific MiR-221 and MiR-146a Participates in Human Thoracic and Abdominal Aortic Aneurysms. Int. J. Mol. Sci. 2017, 18, 875. [Google Scholar] [CrossRef]
  187. Patuzzo, C.; Pasquali, A.; Trabetti, E.; Malerba, G.; Pignatii, P.; Tessari, M.; Faggian, G. A Preliminary MicroRNA Analysis of Non Syndromic Thoracic Aortic Aneurysms. Balk. J. Med. Genet. 2012, 15, 51–55. [Google Scholar] [CrossRef] [Green Version]
  188. Pei, H.; Tian, C.; Sun, X.; Qian, X.; Liu, P.; Liu, W.; Chang, Q. Overexpression of MicroRNA-145 Promotes Ascending Aortic Aneurysm Media Remodeling through TGF-Β1. Eur. J. Vasc. Endovasc. Surg. Off. J. Eur. Soc. Vasc. Surg. 2015, 49, 52–59. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  189. Liao, M.; Zou, S.; Weng, J.; Hou, L.; Yang, L.; Zhao, Z.; Bao, J.; Jing, Z. A MicroRNA Profile Comparison between Thoracic Aortic Dissection and Normal Thoracic Aorta Indicates the Potential Role of MicroRNAs in Contributing to Thoracic Aortic Dissection Pathogenesis. J. Vasc. Surg. 2011, 53, 1341–1349.e3. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  190. Xie, C.; Huang, H.; Sun, X.; Guo, Y.; Hamblin, M.; Ritchie, R.P.; Garcia-Barrio, M.T.; Zhang, J.; Chen, Y.E. MicroRNA-1 Regulates Smooth Muscle Cell Differentiation by Repressing Kruppel-Like Factor 4. Stem Cells Dev. 2011, 20, 205–210. [Google Scholar] [CrossRef] [Green Version]
  191. Torella, D.; Iaconetti, C.; Catalucci, D.; Ellison, G.M.; Leone, A.; Waring, C.D.; Bochicchio, A.; Vicinanza, C.; Aquila, I.; Curcio, A.; et al. MicroRNA-133 Controls Vascular Smooth Muscle Cell Phenotypic Switch in Vitro and Vascular Remodeling In Vivo. Circ. Res. 2011, 109, 880–893. [Google Scholar] [CrossRef] [Green Version]
  192. Li, P.; Liu, Y.; Yi, B.; Wang, G.; You, X.; Zhao, X.; Summer, R.; Qin, Y.; Sun, J. MicroRNA-638 Is Highly Expressed in Human Vascular Smooth Muscle Cells and Inhibits PDGF-BB-Induced Cell Proliferation and Migration through Targeting Orphan Nuclear Receptor NOR1. Cardiovasc. Res. 2013, 99, 185–193. [Google Scholar] [CrossRef] [Green Version]
  193. Merlet, E.; Atassi, F.; Motiani, R.K.; Mougenot, N.; Jacquet, A.; Nadaud, S.; Capiod, T.; Trebak, M.; Lompré, A.-M.; Marchand, A. MiR-424/322 Regulates Vascular Smooth Muscle Cell Phenotype and Neointimal Formation in the Rat. Cardiovasc. Res. 2013, 98, 458–468. [Google Scholar] [CrossRef] [Green Version]
  194. Wang, Y.-S.; Wang, H.-Y.J.; Liao, Y.-C.; Tsai, P.-C.; Chen, K.-C.; Cheng, H.-Y.; Lin, R.-T.; Juo, S.-H.H. MicroRNA-195 Regulates Vascular Smooth Muscle Cell Phenotype and Prevents Neointimal Formation. Cardiovasc. Res. 2012, 95, 517–526. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  195. Liu, X.; Cheng, Y.; Yang, J.; Xu, L.; Zhang, C. Cell-Specific Effects of MiR-221/222 in Vessels: Molecular Mechanism and Therapeutic Application. J. Mol. Cell. Cardiol. 2012, 52, 245–255. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  196. Leeper, N.J.; Raiesdana, A.; Kojima, Y.; Chun, H.J.; Azuma, J.; Maegdefessel, L.; Kundu, R.K.; Quertermous, T.; Tsao, P.S.; Spin, J.M. MicroRNA-26a Is a Novel Regulator of Vascular Smooth Muscle Cell Function. J. Cell. Physiol. 2011, 226, 1035–1043. [Google Scholar] [CrossRef] [Green Version]
  197. Dong, S.; Xiong, W.; Yuan, J.; Li, J.; Liu, J.; Xu, X. MiRNA-146a Regulates the Maturation and Differentiation of Vascular Smooth Muscle Cells by Targeting NF-ΚB Expression. Mol. Med. Rep. 2013, 8, 407–412. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  198. Zhang, J.; Zhao, F.; Yu, X.; Lu, X.; Zheng, G. MicroRNA-155 Modulates the Proliferation of Vascular Smooth Muscle Cells by Targeting Endothelial Nitric Oxide Synthase. Int. J. Mol. Med. 2015, 35, 1708–1714. [Google Scholar] [CrossRef] [Green Version]
  199. Liu, X.; Cheng, Y.; Chen, X.; Yang, J.; Xu, L.; Zhang, C. MicroRNA-31 Regulated by the Extracellular Regulated Kinase Is Involved in Vascular Smooth Muscle Cell Growth via Large Tumor Suppressor Homolog 2. J. Biol. Chem. 2011, 286, 42371–42380. [Google Scholar] [CrossRef] [Green Version]
  200. Li, T.-J.; Chen, Y.-L.; Gua, C.-J.; Xue, S.-J.; Ma, S.-M.; Li, X.-D. MicroRNA 181b Promotes Vascular Smooth Muscle Cells Proliferation through Activation of PI3K and MAPK Pathways. Int. J. Clin. Exp. Pathol. 2015, 8, 10375–10384. [Google Scholar]
  201. Ji, R.; Cheng, Y.; Yue, J.; Yang, J.; Liu, X.; Chen, H.; Dean, D.B.; Zhang, C. MicroRNA Expression Signature and Antisense-Mediated Depletion Reveal an Essential Role of MicroRNA in Vascular Neointimal Lesion Formation. Circ. Res. 2007, 100, 1579–1588. [Google Scholar] [CrossRef]
  202. Yang, J.; Chen, L.; Ding, J.; Fan, Z.; Li, S.; Wu, H.; Zhang, J.; Yang, C.; Wang, H.; Zeng, P.; et al. MicroRNA-24 Inhibits High Glucose-Induced Vascular Smooth Muscle Cell Proliferation and Migration by Targeting HMGB1. Gene 2016, 586, 268–273. [Google Scholar] [CrossRef]
  203. Moushi, A.; Michailidou, K.; Soteriou, M.; Cariolou, M.; Bashiardes, E. MicroRNAs as Possible Biomarkers for Screening of Aortic Aneurysms: A Systematic Review and Validation Study. Biomarkers 2018, 23, 253–264. [Google Scholar] [CrossRef] [Green Version]
  204. D’Amico, F.; Doldo, E.; Pisano, C.; Scioli, M.G.; Centofanti, F.; Proietti, G.; Falconi, M.; Sangiuolo, F.; Ferlosio, A.; Ruvolo, G.; et al. Specific MiRNA and Gene Deregulation Characterize the Increased Angiogenic Remodeling of Thoracic Aneurysmatic Aortopathy in Marfan Syndrome. Int. J. Mol. Sci. 2020, 21, 6886. [Google Scholar] [CrossRef] [PubMed]
  205. Patamsytė, V.; Žukovas, G.; Gečys, D.; Žaliaduonytė, D.; Jakuška, P.; Benetis, R.; Lesauskaitė, V. Long Noncoding RNAs CARMN, LUCAT1, SMILR, and MALAT1 in Thoracic Aortic Aneurysm: Validation of Biomarkers in Clinical Samples. Dis. Markers 2020, 2020, 8521899. [Google Scholar] [CrossRef] [PubMed]
  206. Sun, J.; Chen, G.; Jing, Y.; He, X.; Dong, J.; Zheng, J.; Zou, M.; Li, H.; Wang, S.; Sun, Y.; et al. LncRNA Expression Profile of Human Thoracic Aortic Dissection by High-Throughput Sequencing. Cell. Physiol. Biochem. 2018, 46, 1027–1041. [Google Scholar] [CrossRef]
  207. Xiao, W.; Li, X.; Ji, C.; Shi, J.; Pan, Y. LncRNA Sox2ot Modulates the Progression of Thoracic Aortic Aneurysm by Regulating MiR-330-5p/Myh11. Biosci. Rep. 2020, 40, BSR20194040. [Google Scholar] [CrossRef]
  208. Zhao, X.; Cheng, S.; Li, S.; Li, J.; Bai, X.; Xi, J. CDKN2B-AS1 Aggravates the Pathogenesis of Human Thoracic Aortic Dissection by Sponge to MiR-320d. J. Cardiovasc. Pharmacol. 2020, 76, 592–601. [Google Scholar] [CrossRef] [PubMed]
  209. Liang, K.; Cui, M.; Fu, X.; Ma, J.; Zhang, K.; Zhang, D.; Zhai, S. LncRNA Xist Induces Arterial Smooth Muscle Cell Apoptosis in Thoracic Aortic Aneurysm through MiR-29b-3p/Eln Pathway. Biomed. Pharmacother. 2021, 137, 111163. [Google Scholar] [CrossRef]
  210. Ren, M.; Wang, T.; Wei, X.; Wang, Y.; Ouyang, C.; Xie, Y.; Ye, X.; Han, Z. LncRNA H19 Regulates Smooth Muscle Cell Functions and Participates in the Development of Aortic Dissection through Sponging MiR-193b-3p. Biosci. Rep. 2021, 41, BSR20202298. [Google Scholar] [CrossRef]
  211. Wang, P.; Wang, Z.; Zhang, M.; Wu, Q.; Shi, F. Lnc-OIP5-AS1 Exacerbates Aorta Wall Injury during the Development of Aortic Dissection through Upregulating TUB via Sponging MiR-143-3p. Life Sci. 2021, 271, 119199. [Google Scholar] [CrossRef]
  212. Boeckel, J.-N.; Jaé, N.; Heumüller, A.W.; Chen, W.; Boon, R.A.; Stellos, K.; Zeiher, A.M.; John, D.; Uchida, S.; Dimmeler, S. Identification and Characterization of Hypoxia-Regulated Endothelial Circular RNA. Circ. Res. 2015, 117, 884–890. [Google Scholar] [CrossRef] [Green Version]
  213. Tian, C.; Tang, X.; Zhu, X.; Zhou, Q.; Guo, Y.; Zhao, R.; Wang, D.; Gong, B. Expression Profiles of CircRNAs and the Potential Diagnostic Value of Serum CircMARK3 in Human Acute Stanford Type A Aortic Dissection. PLoS ONE 2019, 14, e0219013. [Google Scholar] [CrossRef] [Green Version]
  214. Cheng, M.; Yang, Y.; Xin, H.; Li, M.; Zong, T.; He, X.; Yu, T.; Xin, H. Non-Coding RNAs in Aortic Dissection: From Biomarkers to Therapeutic Targets. J. Cell. Mol. Med. 2020, 24, 11622–11637. [Google Scholar] [CrossRef] [PubMed]
  215. Gago-Díaz, M.; Blanco-Verea, A.; Teixidó-Turà, G.; Valenzuela, I.; Del Campo, M.; Borregan, M.; Sobrino, B.; Amigo, J.; García-Dorado, D.; Evangelista, A.; et al. Whole Exome Sequencing for the Identification of a New Mutation in TGFB2 Involved in a Familial Case of Non-Syndromic Aortic Disease. Clin. Chim. Acta 2014, 437, 88–92. [Google Scholar] [CrossRef] [PubMed]
  216. Milewicz, D.M.; Regalado, E.S.; Shendure, J.; Nickerson, D.A.; Guo, D. Successes and Challenges of Using Whole Exome Sequencing to Identify Novel Genes Underlying an Inherited Predisposition for Thoracic Aortic Aneurysms and Acute Aortic Dissections. Trends Cardiovasc. Med. 2014, 24, 53–60. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  217. Han, Q.; Zhang, W.; Liu, C.; Zhou, M.; Ran, F.; Yi, L.; Sun, X.; Liu, Z. Whole Exome Sequencing Identifies FBN1 Mutations in Two Patients with Early-onset Type B Aortic Dissection. Mol. Med. Rep. 2017, 16, 6620–6625. [Google Scholar] [CrossRef] [Green Version]
  218. Regalado, E.S.; Guo, D.C.; Santos-Cortez, R.L.P.; Hostetler, E.; Bensend, T.A.; Pannu, H.; Estrera, A.; Safi, H.; Mitchell, A.L.; Evans, J.P.; et al. Pathogenic FBN1 Variants in Familial Thoracic Aortic Aneurysms and Dissections. Clin. Genet. 2016, 89, 719–723. [Google Scholar] [CrossRef] [Green Version]
  219. Ziganshin, B.A.; Bailey, A.E.; Coons, C.; Dykas, D.; Charilaou, P.; Tanriverdi, L.H.; Liu, L.; Tranquilli, M.; Bale, A.E.; Elefteriades, J.A. Routine Genetic Testing for Thoracic Aortic Aneurysm and Dissection in a Clinical Setting. Ann. Thorac. Surg. 2015, 100, 1604–1611. [Google Scholar] [CrossRef] [Green Version]
  220. Landis, B.J.; Schubert, J.A.; Lai, D.; Jegga, A.G.; Shikany, A.R.; Foroud, T.; Ware, S.M.; Hinton, R.B. Exome Sequencing Identifies Candidate Genetic Modifiers of Syndromic and Familial Thoracic Aortic Aneurysm Severity. J Cardiovasc. Transl. Res. 2017, 10, 423–432. [Google Scholar] [CrossRef]
  221. Li, Y.; Gao, S.; Han, Y.; Song, L.; Kong, Y.; Jiao, Y.; Huang, S.; Du, J.; Li, Y. Variants of Focal Adhesion Scaffold Genes Cause Thoracic Aortic Aneurysm. Circ. Res. 2021, 128, 8–23. [Google Scholar] [CrossRef]
  222. Li, Y.; Fang, M.; Yang, J.; Yu, C.; Kuang, J.; Sun, T.; Fan, R. Analysis of the Contribution of 129 Candidate Genes to Thoracic Aortic Aneurysm or Dissection of a Mixed Cohort of Sporadic and Familial Cases in South China. Am. J. Transl. Res. 2021, 13, 4281–4295. [Google Scholar]
  223. Erhart, P.; Brandt, T.; Straub, B.K.; Hausser, I.; Hentze, S.; Böckler, D.; Grond-Ginsbach, C. Familial Aortic Disease and a Large Duplication in Chromosome 16p13.1. Mol. Genet. Genomic Med. 2018, 6, 441–445. [Google Scholar] [CrossRef] [Green Version]
  224. Wu, B.; Li, J.; Wang, Y.; Cheng, Y.; Wang, C.; Shu, X. Recurrent Germline Mutations as Genetic Markers for Aortic Root Dilatation in Bicuspid Aortic Valve Patients. Heart Vessels 2021, 36, 530–540. [Google Scholar] [CrossRef] [PubMed]
  225. Overwater, E.; Marsili, L.; Baars, M.J.H.; Baas, A.F.; van de Beek, I.; Dulfer, E.; van Hagen, J.M.; Hilhorst-Hofstee, Y.; Kempers, M.; Krapels, I.P.; et al. Results of Next-Generation Sequencing Gene Panel Diagnostics Including Copy-Number Variation Analysis in 810 Patients Suspected of Heritable Thoracic Aortic Disorders. Hum. Mutat. 2018, 39, 1173–1192. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  226. Shen, M.; Hu, M.; Fedak, P.W.M.; Oudit, G.Y.; Kassiri, Z. Cell-Specific Functions of ADAM17 Regulate the Progression of Thoracic Aortic Aneurysm. Circ. Res. 2018, 123, 372–388. [Google Scholar] [CrossRef]
  227. Zhou, Z.; Liu, Y.; Gao, S.; Zhou, M.; Qi, F.; Ding, N.; Zhang, J.; Li, R.; Wang, J.; Shi, J.; et al. Excessive DNA Damage Mediates ECM Degradation via the RBBP8/NOTCH1 Pathway in Sporadic Aortic Dissection. Biochim. Biophys. Acta BBA Mol. Basis Dis. 2022, 1868, 166303. [Google Scholar] [CrossRef] [PubMed]
  228. Mohammed, S.; Lim, Z.; Dean, P.H.; Potts, J.E.; Tang, J.N.C.; Etheridge, S.P.; Lara, A.; Husband, P.; Sherwin, E.D.; Ackerman, M.J.; et al. Genetic Insurance Discrimination in Sudden Arrhythmia Death Syndromes: Empirical Evidence from a Cross-Sectional Survey in North America. Circ. Cardiovasc. Genet. 2017, 10, e001442. [Google Scholar] [CrossRef] [Green Version]
  229. Mehta, C.K.; Son, A.Y.; Chia, M.C.; Budd, A.N.; Allen, B.D.; Vassallo, P.; Hoel, A.W.; Brady, W.J.; Nable, J.V. Management of Acute Aortic Syndromes from Initial Presentation to Definitive Treatment. Am. J. Emerg. Med. 2022, 51, 108–113. [Google Scholar] [CrossRef]
Figure 1. Schematization of the ECM main components. Genes codifying each component are reported in red (created with BioRender.com (accessed on 13 May 2022)).
Figure 1. Schematization of the ECM main components. Genes codifying each component are reported in red (created with BioRender.com (accessed on 13 May 2022)).
Diagnostics 12 01785 g001
Figure 2. Schematization of the main components of the SMCs compartment. Genes codifying each component are reported in red (created in BioRender.com (accessed on 13 May 2022)).
Figure 2. Schematization of the main components of the SMCs compartment. Genes codifying each component are reported in red (created in BioRender.com (accessed on 13 May 2022)).
Diagnostics 12 01785 g002
Figure 3. Schematization of the main components of TGF-β signaling. Genes codifying each component are reported in red (created in BioRender.com (accessed on 13 May 2022)).
Figure 3. Schematization of the main components of TGF-β signaling. Genes codifying each component are reported in red (created in BioRender.com (accessed on 13 May 2022)).
Diagnostics 12 01785 g003
Table 1. Genes associated with TAA/D (syndromic and non-syndromic).
Table 1. Genes associated with TAA/D (syndromic and non-syndromic).
Biological Process/Cellular CompartmentGeneProteinOMIMSyndromic TAA/DNon-Syndromic FTAA/DAssociated Syndrome/Diseases
Extracellular matrix/remodelingBGNBiglycan300,989+Meester-Loeys syndrome. ARD, TAAD, pulmonary artery aneurysm, IA, arterial tortuosity [19].
COL3A1Collagen Type III α1 Chain130,050+EDS, vascular type IV. TAAD, early aortic dissection, visceral arterial dissection, vessel fragility [20].
EFEMP2EGF Containing Fibulin Extracellular Matrix Protein 2614,437+Cutis laxa, AR type Ib. Ascending aortic aneurysms, other arterial aneurysms, arterial tortuosity, stenosis [21].
ELNElastin123,700
185,500
+Cutis laxa. AD ARD, ascending aortic aneurysm and dissection [22], TAA [23,24], BAV, IA possibly associated with SVAS.
FBN1Fibrillin-1154,700++Marfan syndrome. ARD, TAA [25], TAAD [26], AAA, other arterial aneurysms, pulmonary artery dilatation, arterial tortuosity [27].
LOXProtein-lysine 6-oxidase617,168+AAT10. AAA, hepatic artery aneurysm, BAV, CAD, TAAD [28,29].
MFAP5Microfibril Associated Protein 5616,166+AAT9. ARD, TAA [30,31].
Smooth muscle cellsACTA2Smooth muscle α-actin611,788
613,834
614,042
++AAT6, multisystemic smooth muscle dysfunction, MYMY5. Early aortic dissection, CAD, stroke (moyamoya disease), PDA, pulmonary artery dilation, BAV, TAAD, TAA [24,32].
FLNAFilamin A300,049+Periventricular nodular heterotopia and otopalatodigital syndrome. Aortic dilatation/aneurysms, peripheral arterial dilatation, PDA, IA, BAV, TAA [32,33].
MYH11Smooth muscle myosin heavy chain132,900+AAT4. PDA, CAD, peripheral vascular occlusive disease, carotid IA, TAAD, early aortic dissection [32,34,35].
MYLKMyosin light chain kinase613,780+AAT7. TAAD, early aortic dissections [36,37].
TGF-β signalingLTBP1Latent TGF-β binding protein 1150,390+Aortic dilation with associated musculoskeletal findings. Dental anomalies, short stature. TAAD,
AAA, visceral and peripheral arterial aneurysm [38].
LTBP3Latent TGF-β binding protein 3602,090
SMAD2SMAD2619,657
619,656
+-Unidentified CTD with arterial aneurysm/dissections. ARD, ascending aortic aneurysms, vertebral/carotid aneurysms and dissections [39], AAA.
SMAD3SMAD3613,795++LDS type III. ARD, TAAD [40], early aortic dissection [39], AAA, arterial tortuosity, other arterial aneurysms/dissections [9], IA, BAV.
SMAD4SMAD4175,050+-JP/HHT syndrome. ARD, TAAD [39], AVMs, IA.
SMAD6SMAD6602,931-+AOVD2. BAV/TAA [24].
TGFB2TGF-β2614,816++LDS type IV. ARD, TAA [40], TAAD, arterial tortuosity [39], other arterial aneurysms, BAV.
TGFB3TGF-β3615,582+-LDS type V. ARD, TAAD, AAA/dissection, other arterial aneurysms, IA/dissection [39].
TGFBR1TGF-β receptor
type 1
609,192++LDS type I+AAT5. TAAD [40], early aortic dissection, AAA, arterial tortuosity, other arterial aneurysms/dissection [9], IA, PDA, BAV.
TGFBR2TGF-β receptor
type 2
610,168++LDS type II+AAT3. TAAD [40], early aortic dissection, AAA, arterial tortuosity, other arterial aneurysms/dissection [9], IA, PDA, BAV.
OthersAXIN1/PDIA2 locus+BAV. BAV/TAA [41].
FBN2Fibrillin-2121,050+Contractual arachnodactyly. Rare ARD and aortic dissection [42], BAV, PDA.
FOXE3Forkhead box 3617,349+AAT11. TAAD [30] (primarily type A dissection).
MAT2AMethionine adenosyl-transferase II αn.a.+FTAA Thoracic aortic aneurysms [30,43]. BAV.
NOTCH1NOTCH1109,730+AOVD1. BAV/TAAD [24].
PRKG1Type 1 cGMP-dependent protein kinase615,436+AAT8. TAAD [28,43], early aortic dissection, AAA, coronary artery aneurysm/dissection, aortic tortuosity, small vessel, CVD.
ROBO4Roundabout guidance receptor 4607,528+BAV. BAV/TAA [24].
SKISloan Kettering proto-oncoprotein182,212+Shprintzen–Goldberg syndrome. ARD, arterial tortuosity, pulmonary artery dilation, other (splenic) arterial aneurysms [36].
SLC2A10Glucose transporter 10208,050+Arterial tortuosity syndrome. ARD, ascending aortic aneurysms [36], other arterial aneurysms, arterial tortuosity [44], elongated arteries, aortic/pulmonary artery stenosis.
In bold: genes associated with dissection. AAA: abdominal aortic aneurysm; AAT/TAA: aortic aneurysm, thoracic; AD: autosomal dominant; AOVD: aortic valve disease; ARD: aortic root dilatation; AVM: arteriovenous malformation; BAV: bicuspid aortic valve; CAD: coronary artery disease; CTD: connective tissue disease; CVD: cerebrovascular disease; EDS: Ehlers-Danlos syndrome; FTAA: familial thoracic aortic aneurysm; FTAAD: familial thoracic aortic aneurysm and/or dissection; HHT: hereditary hemorrhagic telangiectasia; IA: intracranial aneurysm; JP: juvenile polyposis; LDS:, Loeys-Dietz syndrome; n.a.: not applicable; PDA: patent ductus arteriosus; SVAS: supravalvular aortic stenosis; TGF: transforming growth factor; TAAD: thoracic aortic aneurysm and/or dissection.
Table 2. Proposed/suggested circulating biomarkers for TAAD.
Table 2. Proposed/suggested circulating biomarkers for TAAD.
MarkerAnimal ModelsHuman CohortTAATAAD
ANGPTL8-78 patients with AD and 72 controls [170]
Calponin-217 patients with AD [163]++
CK-BB-10 patients with AAD [162]
CK-MM-22 patients with AAD [161]
CRP-49 patients with aortic disorders [130]++
-114 patients with AAD [139]
-118 patients with AAD [140]
CSPCP (aggrecan)-33 patients with AAD [168]++
cTnT-103 patients with AAD [169]
DD-24 patients with AD/TAAD [124] +
-64 patients with AD [125]
-220 patients with AAD [126]
Hcy-31 patients with AAD [141] +
C57BL/6J mice [142]-
MMP8-186 patients suspected AAD [131]
MMP9-105 patients with AAA, 79 with TAA, 112 controls [132]+
MMP12-15 patients with AAD, 10 controls [133]
MPV/PLT-300 patients with aortic disorders [166] +
-183 patients with AAD [167]
sELAFs-62 patients with AAA [135]
-25 patients with AAD [136]
smMHCMice [158]
-27 patients with AD [159] +
TIMP1-93 patients with TAA and 24 controls [70]+
TIMP2-93 patients with TAA and 24 controls [70]+
TGF-β-50 families with LDS [145] +
-28 patients with AAD [152]
-40 patients with aortic disorders [153]++
-1 patient with LDS [155]+
AAA: abdominal aortic aneurysm; AAD: acute aortic dissection; AD: aortic dissection; ANGPTL8: angiopoietin-like protein 8; CK-BB: isozyme BB of creatine kinase; CK-MM: isozyme MM of creatine kinase; CRP: C-reactive protein; CSPCS: cartilage-specific proteoglycan core protein; cTnT: cardiac troponin T; Hcy: homocysteine; LDS: Loeys–Dietz syndrome; MMP: metalloproteinase; sELAFs: soluble elastin fragments; smMHC: smooth muscle myosin heavy chain; TAA: aortic aneurysm, thoracic; TAAD: thoracic aortic aneurysm and/or dissection.
Table 4. Novel potential TAAD-associated genes.
Table 4. Novel potential TAAD-associated genes.
Study/MethodologyGenes IdentifiedAnimal ModelsHuman Cohort
WES (WHOLE EXOME
SEQUENCING)
MLX, DAB2IP, EP300, ZFYVE9, PML, PRKCD-99 patients with TAA [112]
ADCK4, COL15A1-27 patients with fTAA [220]
TES, TLN1, ZYXC57/BL6 mice556 patients with sporadic TAA and 1092 controls [221]
MCTP2-151 patients with TAAD [222]
16p13.1 duplication-1 patient with fTAAD [223]
C1R-13 patients with BAV [224]
NGS PANELSSCARF2-810 cases of suspected TAA [225]
MOUSE MODELSADAM17Sm22α-Cre mice [226]-
RBBP8Male C57/BL6 mice12 Aortic aneurysm/dissection samples [227]
AAT/TAA: aortic aneurysm, thoracic; BAV: bicuspid aortic valve; FTAAD: familial thoracic aortic aneurysm and/or dissection; TAAD: thoracic aortic aneurysm and/or dissection.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

De Cario, R.; Giannini, M.; Cassioli, G.; Kura, A.; Gori, A.M.; Marcucci, R.; Nistri, S.; Pepe, G.; Giusti, B.; Sticchi, E. Tracking an Elusive Killer: State of the Art of Molecular-Genetic Knowledge and Laboratory Role in Diagnosis and Risk Stratification of Thoracic Aortic Aneurysm and Dissection. Diagnostics 2022, 12, 1785. https://doi.org/10.3390/diagnostics12081785

AMA Style

De Cario R, Giannini M, Cassioli G, Kura A, Gori AM, Marcucci R, Nistri S, Pepe G, Giusti B, Sticchi E. Tracking an Elusive Killer: State of the Art of Molecular-Genetic Knowledge and Laboratory Role in Diagnosis and Risk Stratification of Thoracic Aortic Aneurysm and Dissection. Diagnostics. 2022; 12(8):1785. https://doi.org/10.3390/diagnostics12081785

Chicago/Turabian Style

De Cario, Rosina, Marco Giannini, Giulia Cassioli, Ada Kura, Anna Maria Gori, Rossella Marcucci, Stefano Nistri, Guglielmina Pepe, Betti Giusti, and Elena Sticchi. 2022. "Tracking an Elusive Killer: State of the Art of Molecular-Genetic Knowledge and Laboratory Role in Diagnosis and Risk Stratification of Thoracic Aortic Aneurysm and Dissection" Diagnostics 12, no. 8: 1785. https://doi.org/10.3390/diagnostics12081785

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