Skip to main content
Erschienen in: Indian Journal of Thoracic and Cardiovascular Surgery 2/2023

Open Access 25.05.2023 | Review Article

The role of transforming growth factor beta in bicuspid aortic valve aortopathy

verfasst von: Nimrat Grewal, Onur Dolmaci, Arthur Klautz, Juno Legue, Antoine Driessen, Robert Klautz, Robert Poelmann

Erschienen in: Indian Journal of Thoracic and Cardiovascular Surgery | Sonderheft 2/2023

Abstract

A bicuspid aortic valve (BAV) is the most prevalent congenital cardiac deformity, which is associated with an increased risk to develop a thoracic aortic aneurysm and/or an aortic dissection as compared to persons with a tricuspid aortic valve. Due to the high prevalence of a BAV in the general population and the associated life-long increased risk for adverse vascular events, BAV disease places a considerable burden on the public health. The aim of the present review is to discuss the role of transforming growth factor beta (TGF-β) signaling in the development of the vascular wall and on how this complex signaling pathway may be involved in thoracic aortic aneurysm formation in tricuspid and BAV patients.
Hinweise

Publisher's note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Introduction

A bicuspid aortic valve (BAV) is the most prevalent congenital cardiac deformity defined by an aortic valve which consists of two, instead of the normal three, leaflets. BAV patients have an 80-fold increased risk to develop a thoracic aortic aneurysm and/or an aortic dissection as compared to persons with a tricuspid aortic valve (TAV) [14]. An aortic aneurysm is a pathological widening of the aorta, which according to its location can be classified as thoracic or abdominal. Due to the high prevalence of a BAV in the general population and the associated life-long increased risk for adverse vascular events, BAV disease places a considerable burden on the public health.
To prevent the occurrence of lethal acute aortic complications, current aortic surgical guidelines recommend prophylactic vascular replacement surgery in BAV patients based on the size and progression rate of the aortic diameter [5, 6]. Aortic dissections however frequently occur in aortic dimensions below the recommended surgical threshold [7, 8]. The geometrical criterion to intervene thus is not sufficient to timely identify patients with an increased risk for thoracic aortopathy [7, 911]. Considering the limitations of diameter-based risk assessment in BAV, there is an unmet medical need to identify genetic and/or molecular markers to improve patient-tailored treatment strategies, including pharmaceutical stabilization. So far, no drugs have been found that can halt aneurysm growth to prevent aortic events such as dissection or rupture. Consequently, identification of the pathogenetic mechanisms leading to thoracic aortopathy is essential.
A BAV is a complex congenital malformation, with genetically an autosomal dominant inheritance pattern with reduced penetrance and variable expressivity. BAV is therefore characterized by a heterogeneous phenotype. Many studies have focused on the clinical sequelae in BAV related to the genetic background [1215], hemodynamics [1619], animal models [20, 21], and biomarkers [2225]. Despite enormous efforts and great progress in research in the abovementioned areas, the exact underlying pathological pathways leading to aortic dilatation remain unknown. Connective tissue disorders, such as Marfan syndrome and Ehlers-Danlos, are monogenic syndromes which are also associated with an increased risk for aortopathy. These conditions are characterized by an increased transforming growth factor beta (TGF-β) activity [26]. Some similarities in ascending aortic wall pathology have recently been noted between BAV patients and patients with Marfan syndrome [2729]. Considering these overlapping pathological mechanisms, it is interesting to comprehend the role of TGF-β in BAV aortopathy, which has not yet been elucidated. Stressing the need to understand the substrate of aortic complications in BAV patients, this review focuses on the fundamental ascending aortic wall pathology in BAV aortopathy and our current understanding of the role of TGF-β. We present a detailed histopathological overview of the ascending aortic wall in healthy, non-dilated and in pathologically dilated TAV individuals and compare those to the pathological conditions as seen in the BAV patients. We discuss the role of TGF-β in the development of thoracic aortopathy.

The ascending aortic wall development and histology in the tricuspid aortic valve

The intimal layer in tricuspid aortic valve

The aorta is an elastic artery consisting of three layers: the tunica intima, media, and adventitia (Fig. 1). The intima is the innermost layer and endothelial cells form the border of the luminal surface. The formation of the intima starts in the early phases of embryonic development. It has been demonstrated that the intimal layer in a TAV individual at 10 weeks gestational age consists of a single endothelial cell layer which is closely adherent to a thick internal elastic lamina (lamina elastica interna). The internal elastic lamina separates the intimal layer from the middle layer of the ascending aorta [30]. The intimal layer contains only a single elastic lamina until the age of 4 months, after which the intimal thickness increases between the endothelial layer and the internal elastic lamina, forming the so-called sub-endothelial layer. This sub-endothelium consists of fine, thin, and fragmented elastic fibers, with large amounts of extracellular matrix, particularly collagen types I and III and is devoid of vascular smooth muscle cells (SMCs) until the age of 6 months [31]. During growth, a further increase in thickness of the intimal layer is seen [30], due to a process of low-grade injury and repair (Fig. 2) [32]. It is believed that vascular injuries transform vascular SMCs from a metabolic quiescent but contractile to a synthetic and proliferative phenotype through a process called phenotypical switching [33]. This differentiation of vascular SMCs forms an integral part in embryonic vascular development and adult vascular pathology. TGF-β signaling plays pivotal roles in SMC differentiation during vascular development as well as phenotypic switching in disease states [34]. TGF-β is the founding member of the TGF-β superfamily that comprise TGF-βs, activins, bone morphogenetic proteins (BMPs), and growth and differentiation factors (GDFs) [35]. The TGF-β signaling pathway is regulated in multiple steps by several factors which can be summarized as the canonical and non-canonical pathway [36]. In the canonical pathway, TGF-β function is directly mediated by the transcription of Suppressor of Mothers Against Decapentaplegic (SMAD)–dependent target genes. In the non-canonical pathway, TGF-β function is mediated by SMAD-independent pathways including mitogen-activated protein kinase (MAPK) signaling pathways and Wingless-related integration (Wnt) signaling.
The thickened intimal layer consists of extracellular matrix proteins and vascular SMCs besides fine elastic lamellae. Ageing may further lead to the development of atherosclerosis in the intimal layer, a chronic progressive inflammatory disease which can be variably present and in different stages (Fig. 1) [37]. Studies using lineage tracing and vascular SMC–specific manipulation have suggested that phenotypic switching of SMCs might also play a role in atherosclerosis and plaque stability [33]. The role TGF-β plays in the development of atherosclerosis has not yet been fully elucidated. According to recent literature, this signaling pathway cannot be considered as fully “atheroprotective” or “atherogenic”; TGF-β is more likely to play a central role in both normal and pathological vascular repair [38].

The medial layer in tricuspid aortic valve

The medial layer is defined as the area between the internal elastic lamina and the outermost layer, the adventitia. In the premature TAV, the medial layer consists of elastic lamellae without any form of pathology such as elastic fiber thinning, fragmentation, or degeneration. The number of lamellae in the media increases significantly between the premature and neonate life phase. Interestingly, the number of lamellae increases until individuals are 6 years old, after which the number stabilizes for a few years [30, 32].
In adulthood, the number decreases significantly [30, 39].
Apart from elastic lamellae, the medial layer consists of extracellular matrix, vascular SMCs, and vasa vasorum [30, 40]. With increasing age, a decrease of the elastin to collagen ratio is seen in the aorta. The elastin content decreases, whereas other matrix components, primarily collagen which is absent to rare in young aortas, increase [4143]. This process of vascular remodeling alters the vessel wall architecture in response to hemodynamic changes or various forms of vascular injury and serves to maintain the aortic diameter and consistent blood flow under normal physiological conditions. Matrix metalloproteinases (MMPs) play an important role in vascular remodeling and control degradation of extracellular matrix proteins, such as elastin and collagen. Histopathological analysis of tricuspid aneurysmal tissue has shown a characteristic pathological remodeling of collagen and elastin, driven by enhanced production of extracellular proteases and loss of vascular SMCs. The integrity of the aortic extracellular matrix may therefore be compromised by an enhanced MMP activity. Overactivity of MMPs is possible by two means: either due to overproduction by vascular smooth muscle and inflammatory cells in response to vascular injury inflammation, oxidative stresses, matrix degradation products or due to an increased TGF-β activity [44, 45] (Fig. 3).
The major constituents of the aortic media are the vascular SMCs. These are highly specialized cells with contraction and regulation of blood vessel tone-diameter as their main function. During development, SMCs play a key role in the morphogenesis of blood vessels and exhibit high rates of proliferation and production of extracellular matrix components (i.e., collagen, elastin, and proteoglycans), while at the same time acquiring contractile properties [33]. Several studies have demonstrated the importance of TGF-β signaling pathway in SMC differentiation during embryonic development [35]. Adult vascular SMCs are not terminally differentiated and retain the possibility to undergo a phenotypic switch in response to local environmental cues [46]. In adult human blood vessels, the vascular SMCs are characterized by a low rate of proliferation/turnover and a very low rate of synthesis of extracellular matrix components and thus are completely dedicated to carry out their contractile function. However, in response to vascular injury, SMCs dramatically increase their rate of proliferation, migration, and synthetic capacity, playing a crucial role in vascular repair [33, 47]. The ability to phenotypically switch between a “contractile” and “synthetic” phenotype is an important feature of the differentiated vascular SMCs. As discussed above, TGF-β plays a major role in their ability to differentiate and switch between phenotypical states. The possibility to undergo a phenotypical switch in response to environmental cues also plays a major role in the development of cardiovascular diseases, such as atherosclerosis, hypertension, and aneurysm formation [4749]. As mentioned above, in TAV individuals, the number of vascular SMCs decreases in the ageing aorta through apoptosis in the aortic media [39, 42].

The adventitial layer in tricuspid aortic valve

The media to adventitia border is marked by the last, outermost elastic lamella of the media, the external elastic lamina. The adventitia is the surrounding loose fibrous layer that contains connective tissue, the vasa vasorum, adipocytes, and nerve cells (Fig. 1). This layer varies in thickness and composition depending on the site of the ascending aorta, being thick at the aortic root level and thin in the epicardium-covered part of the ascending aorta inside the pericardial cavity. Its variable thickness across the ascending aorta and its rich collagen content give the adventitia the greatest tensile strength of the three aortic wall layers [32]. Distal to the pericardial reflection, the adventitia merges with the surrounding mesenchyme of the thorax. The vasa vasorum normally extends into the outer third of the medial layer. Extension of the vasa vasorum deeper into the media helps to preserve the integrity of the vascular SMCs with ageing, as diffusion of reagents from the lumen into the vascular SMCs becomes difficult with increasing size [50].

The ascending aortic wall development and histology in the bicuspid aortic valve

Patients with a BAV often develop life-threatening thoracic aortopathy. Histopathological differences have mainly been concluded by studying dilated end-stage ascending aortic specimen in BAV and TAV. Recently, focus has shifted towards the structural make-up of the non-dilated ascending aortic wall in BAV patients [37]. These studies analyzed the ascending aortic wall in the premature and adult phase and revealed several differences between BAV and TAV patients [30, 37].

The intimal layer in bicuspid aortic valve

The intimal layer in the premature BAV comprises an endothelial cell layer and internal elastic lamina. In contrast to the TAV however, the sub-endothelial layer already shows thickening before birth [30], leading to a significantly thicker intimal layer. Comparable to the TAV, the sub-endothelial layer in the premature BAV consists of fine elastic lamellae and some extracellular matrix. No intimal vascular SMC expression is seen at this age. The intimal layer becomes significantly thinner directly after birth, with only a single internal elastic lamina in both the non-dilated and dilated BAV patients (Fig. 4A, B). Interestingly, a significantly thin intimal layer has also been described in patients with Marfan syndrome [28, 29]. Marfan syndrome is characterized by a defect in the TGF-β signaling pathway. Considering the prominent role of TGF-β in the development of the intimal layer, it is plausible that in the BAV patients, a defect in the TGF-β signaling pathway is responsible for the lack of intimal thickening after birth and during their lifetime. Our previous work has further shown that the intimal layer in the BAV is completely devoid of TGF-β expression and the downstream signaling factor phosphorylated SMAD2 (pSMAD2), in both the non-dilated and dilated ascending aortic wall, whereas the expression was significantly higher in all TAV patients. Clinical and histopathological studies have also shown that the BAV ascending aorta does also not show any signs of (age-dependent) atherosclerosis, which is commonly seen in the TAV [13, 14, 37].

The medial layer in bicuspid aortic valve

The premature BAV medial layer consists of elastic lamellae, vascular SMCs, and mucoid extracellular matrix, which is similar to the TAV. Differences are however noted in the development of the vascular wall elements. Elastic lamellae in BAV are significantly thinner in both the non- and dilated BAV and hardly show any fragmentation (Fig. 4C). The number of lamellae remains stable throughout most of their lifetime, without observing a decrease in adulthood or difference between non-aneurysmal and aneurysmal tissue.
Vascular SMCs are critical in the differences between BAV and TAV histopathology. Not only have these cells been found in apoptosis [51, 52], they are also morphologically distinct in the BAV population. Vascular SMCs in BAV patients are less well differentiated in both non-dilated and dilated BAV patients as revealed by a significantly lower expression of differentiated vascular SMC markers: smoothelin, calponin, and SM22alpa. Furthermore, Lamin A/C, playing a key role in the differentiation of vascular SMCs, is significantly expressed lower in the non-dilated and dilated BAV as compared to the TAV population [53, 54]. Their immaturity is noted in both the non- and dilated BAV population, excluding aortic dilatation as the consequence of the observed phenotypic switch of the vascular SMCs. Recent studies have emphasized that an early developmental defect could be held responsible for the formation of both a BAV and related aortic wall complications [55].
During embryogenesis, the heart and vessels are the first organ to form. The cardiac development is characterized by a staged formation of all elements which contribute to the normal functioning of the heart. The valves are formed between the fifth and the eight weeks of fetal life. Three distinct cardiac progenitor cells are involved in the formation of the semilunar valves through a process of endocardial cushion formation, being the neural crest, second heart field, and endocardial cushion–derived cells [55, 56]. The three leaflets of the aortic valve are expected to have a different origin: the two main facing cushions are filled by mesenchymal cells, derived both from the endocardium, and later arriving neural crest cells. The third leaflet has a more second heart field–related etiology [57]. The vascular SMCs of the great arteries are also derived from the abovementioned progenitor cell lines. An early embryonic defect could therefore simultaneously lead to a malformed aortic valve and immaturity of the vascular SMCs. Murine genetics studies have shown that a loss of TGF-β signaling components generally leads to abnormal differentiation and maturation of the primitive vascular network, including a failure of SMC recruitment and/or differentiation [58]. Medial accumulation of mucoid extracellular matrix components is significantly higher in non-dilated and dilated BAV patients as compared to the TAV (Fig. 4D) [37, 59].

The adventitial layer in bicuspid aortic valve

The BAV adventitial layer consists of similar elements as seen in the TAV population, being loose fibrous tissue, large nerve fibers, vasa vasorum, and adipocytes. Differences in the vasa vasorum function in BAV have been suggested recently. The adventitia varies in thickness in the BAV too, independent of age and gender.
Histopathological findings in tricuspid, bicuspid and Marfan syndrome patients are summarized in Table 1.
Table 1
Histopathological findings in tricuspid, bicuspid and Marfan syndrome patients
 
Tricuspid aortic valve
Bicuspid aortic valve
Marfan syndrome
Tunica intima
• Thick intimal layer
• Atherosclerosis + 
• Histopathologically
TGF-β and pSMAD2 expression + 
• Significantly thinner intimal layer
• Atherosclerosis –
• Defective TGF-β signaling
• Histopathologically
TGF-β and pSMAD2 expression –
• Significantly thinner intimal layer
• Atherosclerosis –
• Defective TGF-β signaling
• Histopathologically
• TGF-β and pSMAD2 expression –
Tunica media
• Differentiated VSMCs
• TGF-β expression + 
• Immature VSMCs
• Loss of TGF-β expression
• Immature VSMCs
• Loss of TGF-β expression
Tunica adventitia
 
• Reported difference in vasa vasorum
 
Legend: pSMAD2, phosphorylated SMAD2; TGF-β, transforming growth factor beta; VSMCs, vascular smooth muscle cells

Discussion

A BAV is the most common congenital cardiac malformation, which carries an extremely high risk for thoracic aortic aneurysm development. Other conditions which also carry an increased susceptibility for thoracic aortopathy are related to specific genetic conditions, such as Marfan syndrome, Loeys-Dietz syndrome, Ehlers-Danlos syndrome, and familial thoracic aortic aneurysms and dissections. Thoracic aortopathy is however most commonly seen in patients with a TAV, in the form of degenerative thoracic aortopathy. Many studies have recently compared the ascending aortic wall pathology in abovementioned conditions and discovered specific histopathological features in the aortic wall make-up. The disease-of-interest, thoracic aortopathy, is a very heterogeneous disorder with multiple distinct underlying genetic mutations but common clinical phenotypes and histopathologic and molecular findings [27, 53, 60, 61]. As a thoracic aortic aneurysm does not have any preceding symptoms, the first manifestation often is chest pain due to a ruptured aorta. Management of thoracic aortopathy therefore remains a challenge in elective as well as emergency cases. The identification of common pathological mechanisms would significantly improve the individual cardiovascular risk stratification and thus will result in a major improvement in the field of personalized medicine. The aim of the present review is to discuss the role of TGF-β signaling in both the development of the vascular wall and on how this complex signaling pathway may be involved in thoracic aortic aneurysm formation in TAV and BAV patients.
The TGF-β signaling pathway not only plays a crucial role in vascular development, but also in degenerative thoracic aortic aneurysm formation in the TAV population. Thoracic aortopathy develops as a result of maladaptive remodeling of the vascular extracellular matrix. In normal conditions, a balance exists in vascular remodeling with matrix deposition and degradation resulting in maintenance of the structural integrity of the vascular wall. Recent evidence highlights that a dysregulation of TGF-β signaling disrupts the balance in favor of enhanced proteolysis resulting in pathological remodeling of the vascular extracellular matrix [62]. A dysregulation of the TGF-β canonical signaling pathway has further been shown to lead to a fragmentation of the elastic lamellae [63] causing weakening of the aortic architecture and increased susceptibility for aortic dilatation and dissection [64]. In the bicuspid research field, recent focus has shifted towards a common embryonic origin of both valvular and vascular development. Genetic defects in early development can account for both a deformed aortic valve and defective composition of the ascending aortic wall. Progenitor cells are responsible for region-specific vascular SMCs in the aortic wall. Therefore, defects in the neural crest and second heart field signaling in the BAV patients lead to BAV-related aortopathy in the proximal thoracic aorta that is rarely found in the descending thoracic aorta [27]. In comparison in Marfan patients, defects are seen in the neural crest, second heart field, and paraxial mesoderm which cause aortopathy in both the ascending and descending aorta. In this review, we discussed the characteristic features of the bicuspid aortic wall, including a thin intimal layer, a phenotypical switch defect resulting in less differentiated SMCs, excessive mucoid extracellular matrix accumulation, and lack of atherosclerosis. Previous studies have shown that the ascending aortic wall in Marfan syndrome and a dissected aortic wall share many features in common with the BAV, in form of a thin intimal layer, immature vascular SMCs, and lack of atherosclerosis [4, 65] (Table 1). Considering the functions of TGF-β in the development of the intimal layer and normal vascular remodeling and phenotypical switch of vascular SMCs, it is plausible that a defect of this signaling pathway is responsible for the aortic pathology encountered in patients with a BAV. Expression of TGF-β and downstream signalers has been studied in several ways. Our group investigated the expression of TGF-β and pSMAD2 in the ascending aortic wall in the non- and dilated BAV and TAV patients. We found that the intima lacked TGF-β and pSMAD expression and the medial expression was significantly lower in the BAV as compared to the TAV dilated specimen. Our findings are in line with many other studies which found a decreased expression of TGF-β in the BAV aorta as compared to the TAV [63, 66], also investigating the canonical SMAD-mediated pathway. In murine aneurysmal studies, an increased TGF-β signal through the non-canonical pathway has been described leading to extracellular matrix degradation [67, 68]. Unfortunately, special attention to the intimal layer has not been given by any other study. Besides differences in expression between valve morphologies, we were able to identify non-dilated BAVs with increased susceptibility for future complications on basis of the medial expression of TGF-β, pSMAD2, and MMP9 [25]. Ikonomidis et al. also suggested in their study that a unique profile of plasma MMPs, tissue inhibitors of MMPs, and micro ribonucleic acids (microRNAs) could possibly predict the increased risk for thoracic aortopathy in BAV and TAV using a plasma multianalyte regression strategy [24]. Expression of TGF-β and markers of the non- and canonical pathway should therefore be further investigated as a marker to distinguish BAV patients with an increased risk for future aortic complications. Differences in the amount of TGF-β in the aortic wall have further been reported to correlate well with the levels of shear stress on the wall [69]; therefore, in future studies, the role of shear stress on the aortic wall should also be considered. It is interesting to note that a specific group of animals like reptiles normally present with bicuspid semilunar valves. Evidently, these have a different physiology, being “cold-blooded” as well as anatomy, as they present two aortas [70]. Nevertheless, here the bicuspid valves function efficiently, even until advanced ages without signs of thoracic aortopathy. It would therefore be interesting to study the TGF-β expression in reptiles where BAV prevails under normal conditions.

Conclusion and future directions

In most of the thoracic aortic aneurysm cases, an increased TGF-β activity has been identified. Characteristics in BAV point at a decreased or defective signaling in the TGF-β signaling pathway. A defect in early embryogenesis is most likely responsible for the development of both a BAV and an abnormal ascending aortic wall, which is susceptible for future aortic complication. The characteristic BAV findings, which can be related to a defective TGF-β signaling are, however, generic to all BAV patients and cannot be applied to identify the high-risk subset. Furthermore, a BAV shares many histopathological features with Marfan syndrome characterized by a defect in the TGF-β signaling pathway. In Marfan however, a BAV is not an obligatory clinical manifestation, indicating that a defective TGF-β signaling is at least not the main factor causing BAV formation. Even though many histopathological features in BAV can be explained by a decreased TGF-β activation, future studies will have to focus on differences in expression in the non-dilated BAV groups to be able to distinguish cause and effect of expression and identify patients with an increased vulnerability for future thoracic aortopathy.

Declarations

Institutional review board approval

Not applicable. The research does not involve human participants.
Not applicable.

Conflict of interest

The authors declare no conflict of interest.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​.

Publisher's note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Unsere Produktempfehlungen

Die Chirurgie

Print-Titel

Das Abo mit mehr Tiefe

Mit der Zeitschrift Die Chirurgie erhalten Sie zusätzlich Online-Zugriff auf weitere 43 chirurgische Fachzeitschriften, CME-Fortbildungen, Webinare, Vorbereitungskursen zur Facharztprüfung und die digitale Enzyklopädie e.Medpedia.

Bis 30. April 2024 bestellen und im ersten Jahr nur 199 € zahlen!

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

Literatur
1.
Zurück zum Zitat Michelena HI, Della Corte A, Prakash SK, Milewicz DM, Evangelista A, Enriquez-Sarano M. Bicuspid aortic valve aortopathy in adults: incidence, etiology, and clinical significance. Int J Cardiol. 2015;201:400–7.PubMedCrossRef Michelena HI, Della Corte A, Prakash SK, Milewicz DM, Evangelista A, Enriquez-Sarano M. Bicuspid aortic valve aortopathy in adults: incidence, etiology, and clinical significance. Int J Cardiol. 2015;201:400–7.PubMedCrossRef
2.
Zurück zum Zitat Sherrah AG, Andvik S, van der Linde D, Davies L, Bannon PG, Padang R, et al. Nonsyndromic thoracic aortic aneurysm and dissection: outcomes with Marfan syndrome versus bicuspid aortic valve aneurysm. J Am Coll Cardiol. 2016;67:618–26.PubMedCrossRef Sherrah AG, Andvik S, van der Linde D, Davies L, Bannon PG, Padang R, et al. Nonsyndromic thoracic aortic aneurysm and dissection: outcomes with Marfan syndrome versus bicuspid aortic valve aneurysm. J Am Coll Cardiol. 2016;67:618–26.PubMedCrossRef
3.
Zurück zum Zitat Michelena HI, Prakash SK, Della Corte A, Bissell MM, Anavekar N, Mathieu P, et al. Bicuspid aortic valve: identifying knowledge gaps and rising to the challenge from the International Bicuspid Aortic Valve Consortium (BAVCon). Circulation. 2014;129:2691–704.PubMedPubMedCentralCrossRef Michelena HI, Prakash SK, Della Corte A, Bissell MM, Anavekar N, Mathieu P, et al. Bicuspid aortic valve: identifying knowledge gaps and rising to the challenge from the International Bicuspid Aortic Valve Consortium (BAVCon). Circulation. 2014;129:2691–704.PubMedPubMedCentralCrossRef
4.
Zurück zum Zitat Grewal N, Velders BJJ, Gittenberger-de Groot AC, Poelmann R, Klautz RJM, Van Brakel TJ, et al. A systematic histopathologic evaluation of type-A aortic dissections implies a uniform multiple-hit causation. J Cardiovasc Dev Dis. 2021;8:12. Grewal N, Velders BJJ, Gittenberger-de Groot AC, Poelmann R, Klautz RJM, Van Brakel TJ, et al. A systematic histopathologic evaluation of type-A aortic dissections implies a uniform multiple-hit causation. J Cardiovasc Dev Dis. 2021;8:12.
5.
Zurück zum Zitat Hardikar AA, Marwick TH. Surgical thresholds for bicuspid aortic valve associated aortopathy. JACC Cardiovasc Imaging. 2013;6:1311–20.PubMedCrossRef Hardikar AA, Marwick TH. Surgical thresholds for bicuspid aortic valve associated aortopathy. JACC Cardiovasc Imaging. 2013;6:1311–20.PubMedCrossRef
6.
Zurück zum Zitat Hiratzka LF, Creager MA, Isselbacher EM, Svensson LG, Nishimura RA, Bonow RO, et al. Surgery for aortic dilatation in patients with bicuspid aortic valves: a statement of clarification from the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. J Thorac Cardiovasc Surg. 2016;151:959–66.PubMedCrossRef Hiratzka LF, Creager MA, Isselbacher EM, Svensson LG, Nishimura RA, Bonow RO, et al. Surgery for aortic dilatation in patients with bicuspid aortic valves: a statement of clarification from the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. J Thorac Cardiovasc Surg. 2016;151:959–66.PubMedCrossRef
7.
Zurück zum Zitat Pape LA, Tsai TT, Isselbacher EM, Oh JK, O’Gara PT, Evangelista A, 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;116:1120–7.PubMedCrossRef Pape LA, Tsai TT, Isselbacher EM, Oh JK, O’Gara PT, Evangelista A, 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;116:1120–7.PubMedCrossRef
8.
Zurück zum Zitat Girdauskas E, Disha K, Raisin HH, Secknus MA, Borger MA, Kuntze T. Risk of late aortic events after an isolated aortic valve replacement for bicuspid aortic valve stenosis with concomitant ascending aortic dilation. Eur J Cardiothorac Surg. 2012;42:832–7. Girdauskas E, Disha K, Raisin HH, Secknus MA, Borger MA, Kuntze T. Risk of late aortic events after an isolated aortic valve replacement for bicuspid aortic valve stenosis with concomitant ascending aortic dilation. Eur J Cardiothorac Surg. 2012;42:832–7.
9.
Zurück zum Zitat Rylski B, Blanke P, Beyersdorf F, Desai ND, Milewski RK, Siepe M, et al. How does the ascending aorta geometry change when it dissects? J Am Coll Cardiol. 2014;63:1311–9.PubMedCrossRef Rylski B, Blanke P, Beyersdorf F, Desai ND, Milewski RK, Siepe M, et al. How does the ascending aorta geometry change when it dissects? J Am Coll Cardiol. 2014;63:1311–9.PubMedCrossRef
10.
Zurück zum Zitat Erbel R, Aboyans V, Boileau C, Bossone E, Di Bartolomeo R, Eggebrecht H, et al. ESC Guidelines on the diagnosis and treatment of aortic diseases: document covering acute and chronic aortic diseases of the thoracic and abdominal aorta of the adult. The Task Force for the Diagnosis and Treatment of Aortic Diseases of the European Society of Cardiology (ESC). Eur Heart J. 2014;35:2873–926. Erbel R, Aboyans V, Boileau C, Bossone E, Di Bartolomeo R, Eggebrecht H, et al. ESC Guidelines on the diagnosis and treatment of aortic diseases: document covering acute and chronic aortic diseases of the thoracic and abdominal aorta of the adult. The Task Force for the Diagnosis and Treatment of Aortic Diseases of the European Society of Cardiology (ESC). Eur Heart J. 2014;35:2873–926.
11.
Zurück zum Zitat Neri E, Barabesi L, Buklas D, Vricella LA, Benvenuti A, Tucci E, et al. Limited role of aortic size in the genesis of acute type A aortic dissection. Eur J Cardiothorac Surg. 2005;28:857–63.PubMedCrossRef Neri E, Barabesi L, Buklas D, Vricella LA, Benvenuti A, Tucci E, et al. Limited role of aortic size in the genesis of acute type A aortic dissection. Eur J Cardiothorac Surg. 2005;28:857–63.PubMedCrossRef
12.
Zurück zum Zitat Koenraadt WMC, Grewal N, Gaidoukevitch OY, DeRuiter MC, Gittenberger-de Groot AC, Bartelings MM, et al. The extent of the raphe in bicuspid aortic valves is associated with aortic regurgitation and aortic root dilatation. Neth Heart J. 2016;24:127–33. Koenraadt WMC, Grewal N, Gaidoukevitch OY, DeRuiter MC, Gittenberger-de Groot AC, Bartelings MM, et al. The extent of the raphe in bicuspid aortic valves is associated with aortic regurgitation and aortic root dilatation. Neth Heart J. 2016;24:127–33.
13.
Zurück zum Zitat Dolmaci OB, Legue J, Lindeman JHN, Driessen AHG, Klautz RJM, van Brakel TJ, et al. Extent of coronary artery disease in patients with stenotic bicuspid versus tricuspid aortic valves. J Am Heart Assoc. 2021;10:e020080. Dolmaci OB, Legue J, Lindeman JHN, Driessen AHG, Klautz RJM, van Brakel TJ, et al. Extent of coronary artery disease in patients with stenotic bicuspid versus tricuspid aortic valves. J Am Heart Assoc. 2021;10:e020080.
14.
Zurück zum Zitat Dolmaci OB, Driessen AHG, Klautz RJM, Poelmann R, Lindeman JHN, Grewal N. Comparative evaluation of coronary disease burden: bicuspid valve disease is not atheroprotective. Open heart. 2021;8:e001772. Dolmaci OB, Driessen AHG, Klautz RJM, Poelmann R, Lindeman JHN, Grewal N. Comparative evaluation of coronary disease burden: bicuspid valve disease is not atheroprotective. Open heart. 2021;8:e001772.
15.
Zurück zum Zitat Sequeira Gross TM, Lindner D, Ojeda FM, Neumann J, Grewal N, Kuntze T, et al. Comparison of microstructural alterations in the proximal aorta between aortic stenosis and regurgitation. J Thorac Cardiovasc Surg. 2021;162:1684–95.PubMedCrossRef Sequeira Gross TM, Lindner D, Ojeda FM, Neumann J, Grewal N, Kuntze T, et al. Comparison of microstructural alterations in the proximal aorta between aortic stenosis and regurgitation. J Thorac Cardiovasc Surg. 2021;162:1684–95.PubMedCrossRef
16.
Zurück zum Zitat Girdauskas E, Disha K, Borger MA, Kuntze T. Relation of bicuspid aortic valve morphology to the dilatation pattern of the proximal aorta: focus on the transvalvular flow. Cardiol Res Pract. 2012;2012: 478259.PubMedPubMedCentralCrossRef Girdauskas E, Disha K, Borger MA, Kuntze T. Relation of bicuspid aortic valve morphology to the dilatation pattern of the proximal aorta: focus on the transvalvular flow. Cardiol Res Pract. 2012;2012: 478259.PubMedPubMedCentralCrossRef
17.
Zurück zum Zitat Youssefi P, Gomez A, He T, Anderson L, Bunce N, Sharma R, et al. Patient-specific computational fluid dynamics-assessment of aortic hemodynamics in a spectrum of aortic valve pathologies. J Thorac Cardiovasc Surg. 2017;153:8-20.e3.PubMedCrossRef Youssefi P, Gomez A, He T, Anderson L, Bunce N, Sharma R, et al. Patient-specific computational fluid dynamics-assessment of aortic hemodynamics in a spectrum of aortic valve pathologies. J Thorac Cardiovasc Surg. 2017;153:8-20.e3.PubMedCrossRef
18.
19.
Zurück zum Zitat Grewal N, Girdauskas E, DeRuiter M, Goumans MJ, Poelmann RE, Klautz RJM, et al. The role of hemodynamics in bicuspid aortopathy: a histopathologic study. Cardiovasc Pathol. 2019;41:29–37.PubMedCrossRef Grewal N, Girdauskas E, DeRuiter M, Goumans MJ, Poelmann RE, Klautz RJM, et al. The role of hemodynamics in bicuspid aortopathy: a histopathologic study. Cardiovasc Pathol. 2019;41:29–37.PubMedCrossRef
20.
Zurück zum Zitat Fernandez B, Duran AC, Fernandez-Gallego T, Fernandez MC, Such M, Arque JM, et al. Bicuspid aortic valves with different spatial orientations of the leaflets are distinct etiological entities. J Am Coll Cardiol. 2009;54:2312–8.PubMedCrossRef Fernandez B, Duran AC, Fernandez-Gallego T, Fernandez MC, Such M, Arque JM, et al. Bicuspid aortic valves with different spatial orientations of the leaflets are distinct etiological entities. J Am Coll Cardiol. 2009;54:2312–8.PubMedCrossRef
21.
Zurück zum Zitat Detaint D, Aegerter P, Tubach F, Hoffman I, Plauchu H, Dulac Y, et al. Rationale and design of a randomized clinical trial (Marfan Sartan) of angiotensin II receptor blocker therapy versus placebo in individuals with Marfan syndrome. Arch Cardiovasc Dis. 2010;103:317–25.PubMedCrossRef Detaint D, Aegerter P, Tubach F, Hoffman I, Plauchu H, Dulac Y, et al. Rationale and design of a randomized clinical trial (Marfan Sartan) of angiotensin II receptor blocker therapy versus placebo in individuals with Marfan syndrome. Arch Cardiovasc Dis. 2010;103:317–25.PubMedCrossRef
22.
Zurück zum Zitat Black KM, Masuzawa A, Hagberg RC, Khabbaz KR, Trovato ME, Rettagliati VM, et al. Preliminary biomarkers for identification of human ascending thoracic aortic aneurysm. J Am Heart Assoc. 2013;2: e000138.PubMedPubMedCentralCrossRef Black KM, Masuzawa A, Hagberg RC, Khabbaz KR, Trovato ME, Rettagliati VM, et al. Preliminary biomarkers for identification of human ascending thoracic aortic aneurysm. J Am Heart Assoc. 2013;2: e000138.PubMedPubMedCentralCrossRef
23.
Zurück zum Zitat Jones JA, Stroud RE, O’Quinn EC, Black LE, Barth JL, Elefteriades JA, et al. Selective microRNA suppression in human thoracic aneurysms: relationship of miR-29a to aortic size and proteolytic induction. Circ Cardiovasc Genet. 2011;4:605–13.PubMedPubMedCentralCrossRef Jones JA, Stroud RE, O’Quinn EC, Black LE, Barth JL, Elefteriades JA, et al. Selective microRNA suppression in human thoracic aneurysms: relationship of miR-29a to aortic size and proteolytic induction. Circ Cardiovasc Genet. 2011;4:605–13.PubMedPubMedCentralCrossRef
24.
Zurück zum Zitat Ikonomidis JS, Ivey CR, Wheeler JB, Akerman AW, Rice A, Patel RK, et al. Plasma biomarkers for distinguishing etiologic subtypes of thoracic aortic aneurysm disease. J Thorac Cardiovasc Surg. 2013;145:1326–33.PubMedPubMedCentralCrossRef Ikonomidis JS, Ivey CR, Wheeler JB, Akerman AW, Rice A, Patel RK, et al. Plasma biomarkers for distinguishing etiologic subtypes of thoracic aortic aneurysm disease. J Thorac Cardiovasc Surg. 2013;145:1326–33.PubMedPubMedCentralCrossRef
25.
Zurück zum Zitat Grewal N, Gittenberger-de Groot AC, DeRuiter MC, Klautz RJ, Poelmann RE, Duim S, et al. Bicuspid aortic valve: phosphorylation of c-Kit and downstream targets are prognostic for future aortopathy. Eur J Cardiothorac Surg. 2014;46:831–9. Grewal N, Gittenberger-de Groot AC, DeRuiter MC, Klautz RJ, Poelmann RE, Duim S, et al. Bicuspid aortic valve: phosphorylation of c-Kit and downstream targets are prognostic for future aortopathy. Eur J Cardiothorac Surg. 2014;46:831–9.
26.
Zurück zum Zitat Loeys BL, Schwarze U, Holm T, Callewaert BL, Thomas GH, Pannu H, et al. Aneurysm syndromes caused by mutations in the TGF-beta receptor. N Engl J Med. 2006;355:788–98.PubMedCrossRef Loeys BL, Schwarze U, Holm T, Callewaert BL, Thomas GH, Pannu H, et al. Aneurysm syndromes caused by mutations in the TGF-beta receptor. N Engl J Med. 2006;355:788–98.PubMedCrossRef
27.
Zurück zum Zitat Grewal N, Franken R, Mulder BJM, Goumans MJ, Lindeman JHN, Jongbloed MRM, et al. Histopathology of aortic complications in bicuspid aortic valve versus Marfan syndrome: relevance for therapy? Heart Vessels. 2016;31:795–806. Grewal N, Franken R, Mulder BJM, Goumans MJ, Lindeman JHN, Jongbloed MRM, et al. Histopathology of aortic complications in bicuspid aortic valve versus Marfan syndrome: relevance for therapy? Heart Vessels. 2016;31:795–806.
28.
Zurück zum Zitat Grewal NK, Goumans MJ, deRuiter MC, Klautz RJM, Poelmann RE, Mulder BJM, et al. Aortopathy in bicuspid aortic valve and marfan syndrome is characterized by a lack of activation potential of the epicardium in the ascending aorta. Int J Pathol Clin Res. 2017. https://doi.org/10.23937/2469-5807/1510051. Grewal NK, Goumans MJ, deRuiter MC, Klautz RJM, Poelmann RE, Mulder BJM, et al. Aortopathy in bicuspid aortic valve and marfan syndrome is characterized by a lack of activation potential of the epicardium in the ascending aorta. Int J Pathol Clin Res. 2017. https://​doi.​org/​10.​23937/​2469-5807/​1510051.
29.
Zurück zum Zitat Grewal N, Gittenberger-de Groot AC. Pathogenesis of aortic wall complications in Marfan syndrome. Cardiovasc Pathol. 2018;33:62–9.PubMedCrossRef Grewal N, Gittenberger-de Groot AC. Pathogenesis of aortic wall complications in Marfan syndrome. Cardiovasc Pathol. 2018;33:62–9.PubMedCrossRef
30.
Zurück zum Zitat Grewal N, Gittenberger-de Groot AC, Von der Thusen J, Wisse LJ, Bartelings MM, DeRuiter MC, et al. The development of the ascending aortic wall in tricuspid and bicuspid aortic valve: a process from maturation to degeneration. J Clin Med. 2020;9:908. Grewal N, Gittenberger-de Groot AC, Von der Thusen J, Wisse LJ, Bartelings MM, DeRuiter MC, et al. The development of the ascending aortic wall in tricuspid and bicuspid aortic valve: a process from maturation to degeneration. J Clin Med. 2020;9:908.
31.
Zurück zum Zitat Wang M, Zhang J, Jiang LQ, Spinetti G, Pintus G, Monticone R, et al. Proinflammatory profile within the grossly normal aged human aortic wall. Hypertension. 2007;50:219–27.PubMedCrossRef Wang M, Zhang J, Jiang LQ, Spinetti G, Pintus G, Monticone R, et al. Proinflammatory profile within the grossly normal aged human aortic wall. Hypertension. 2007;50:219–27.PubMedCrossRef
32.
Zurück zum Zitat Halushka MK, Angelini A, Bartoloni G, Basso C, Batoroeva L, Bruneval P, et al. Consensus statement on surgical pathology of the aorta from the Society for Cardiovascular Pathology and the Association For European Cardiovascular Pathology: II. Noninflammatory degenerative diseases - nomenclature and diagnostic criteria. Cardiovasc Pathol. 2016;25:247–57.PubMedCrossRef Halushka MK, Angelini A, Bartoloni G, Basso C, Batoroeva L, Bruneval P, et al. Consensus statement on surgical pathology of the aorta from the Society for Cardiovascular Pathology and the Association For European Cardiovascular Pathology: II. Noninflammatory degenerative diseases - nomenclature and diagnostic criteria. Cardiovasc Pathol. 2016;25:247–57.PubMedCrossRef
33.
Zurück zum Zitat Owens GK, Kumar MS, Wamhoff BR. Molecular regulation of vascular smooth muscle cell differentiation in development and disease. Physiol Rev. 2004;84:767–801.PubMedCrossRef Owens GK, Kumar MS, Wamhoff BR. Molecular regulation of vascular smooth muscle cell differentiation in development and disease. Physiol Rev. 2004;84:767–801.PubMedCrossRef
34.
Zurück zum Zitat Sinha S, Hoofnagle MH, Kingston PA, McCanna ME, Owens GK. Transforming growth factor-beta1 signaling contributes to development of smooth muscle cells from embryonic stem cells. Am J Physiol Cell Physiol. 2004;287:C1560–8.PubMedCrossRef Sinha S, Hoofnagle MH, Kingston PA, McCanna ME, Owens GK. Transforming growth factor-beta1 signaling contributes to development of smooth muscle cells from embryonic stem cells. Am J Physiol Cell Physiol. 2004;287:C1560–8.PubMedCrossRef
35.
Zurück zum Zitat Guo X, Chen S-Y. Transforming growth factor-β and smooth muscle differentiation. World J Biol Chem. 2012;3:41–52. Guo X, Chen S-Y. Transforming growth factor-β and smooth muscle differentiation. World J Biol Chem. 2012;3:41–52.
36.
Zurück zum Zitat Chakrabarti M, Al-Sammarraie N, Gebere MG, Bhattacharya A, Chopra S, Johnson J, et al. Transforming growth factor beta3 is required for cardiovascular development. J Cardiovasc Dev Dis. 2020;7:19. Chakrabarti M, Al-Sammarraie N, Gebere MG, Bhattacharya A, Chopra S, Johnson J, et al. Transforming growth factor beta3 is required for cardiovascular development. J Cardiovasc Dev Dis. 2020;7:19.
37.
Zurück zum Zitat Grewal N, Girdauskas E, Idhrees M, Velayudhan B, Klautz R, Driessen A, et al. Structural abnormalities in the non-dilated ascending aortic wall of bicuspid aortic valve patients. Cardiovasc Pathol. 2023;62:107478. Grewal N, Girdauskas E, Idhrees M, Velayudhan B, Klautz R, Driessen A, et al. Structural abnormalities in the non-dilated ascending aortic wall of bicuspid aortic valve patients. Cardiovasc Pathol. 2023;62:107478.
38.
Zurück zum Zitat Toma I, McCaffrey TA. Transforming growth factor-β and atherosclerosis: interwoven atherogenic and atheroprotective aspects. Cell Tissue Res. 2012;347:155–75.PubMedCrossRef Toma I, McCaffrey TA. Transforming growth factor-β and atherosclerosis: interwoven atherogenic and atheroprotective aspects. Cell Tissue Res. 2012;347:155–75.PubMedCrossRef
39.
Zurück zum Zitat Fritze O, Romero B, Schleicher M, Jacob MP, Oh DY, Starcher B, et al. Age-related changes in the elastic tissue of the human aorta. J Vasc Res. 2012;49:77–86.PubMedCrossRef Fritze O, Romero B, Schleicher M, Jacob MP, Oh DY, Starcher B, et al. Age-related changes in the elastic tissue of the human aorta. J Vasc Res. 2012;49:77–86.PubMedCrossRef
40.
Zurück zum Zitat Wolinsky H, Glagov S. A lamellar unit of aortic medial structure and function in mammals. Circ Res. 1967;20:99–111.PubMedCrossRef Wolinsky H, Glagov S. A lamellar unit of aortic medial structure and function in mammals. Circ Res. 1967;20:99–111.PubMedCrossRef
41.
Zurück zum Zitat Virmani R, Avolio AP, Mergner WJ, Robinowitz M, Herderick EE, Cornhill JF, et al. Effect of aging on aortic morphology in populations with high and low prevalence of hypertension and atherosclerosis. Comparison between occidental and Chinese communities. Am J Pathol. 1991;139:1119–29. Virmani R, Avolio AP, Mergner WJ, Robinowitz M, Herderick EE, Cornhill JF, et al. Effect of aging on aortic morphology in populations with high and low prevalence of hypertension and atherosclerosis. Comparison between occidental and Chinese communities. Am J Pathol. 1991;139:1119–29. 
42.
Zurück zum Zitat Sawabe M. Vascular aging: from molecular mechanism to clinical significance. Geriatr Gerontol Int. 2010;10:S213–20.PubMedCrossRef Sawabe M. Vascular aging: from molecular mechanism to clinical significance. Geriatr Gerontol Int. 2010;10:S213–20.PubMedCrossRef
43.
Zurück zum Zitat Tsamis A, Krawiec JT, Vorp DA. Elastin and collagen fibre microstructure of the human aorta in ageing and disease: a review. J R Soc Interface. 2013;10:20121004.PubMedPubMedCentralCrossRef Tsamis A, Krawiec JT, Vorp DA. Elastin and collagen fibre microstructure of the human aorta in ageing and disease: a review. J R Soc Interface. 2013;10:20121004.PubMedPubMedCentralCrossRef
44.
Zurück zum Zitat Curci JA, Liao S, Huffman MD, Shapiro SD, Thompson RW. Expression and localization of macrophage elastase (matrix metalloproteinase-12) in abdominal aortic aneurysms. J Clin Invest. 1998;102:1900–10. Curci JA, Liao S, Huffman MD, Shapiro SD, Thompson RW. Expression and localization of macrophage elastase (matrix metalloproteinase-12) in abdominal aortic aneurysms. J Clin Invest. 1998;102:1900–10.
45.
Zurück zum Zitat Jacob MP. Extracellular matrix remodeling and matrix metalloproteinases in the vascular wall during aging and in pathological conditions. Biomed Pharmacother. 2003;57:195–202.PubMedCrossRef Jacob MP. Extracellular matrix remodeling and matrix metalloproteinases in the vascular wall during aging and in pathological conditions. Biomed Pharmacother. 2003;57:195–202.PubMedCrossRef
46.
Zurück zum Zitat Owens GK. Regulation of differentiation of vascular smooth muscle cells. Physiol Rev. 1995;75:487–517.PubMedCrossRef Owens GK. Regulation of differentiation of vascular smooth muscle cells. Physiol Rev. 1995;75:487–517.PubMedCrossRef
48.
Zurück zum Zitat Gabbiani G, Schmid E, Winter S, Chaponnier C, de Ckhastonay C, Vandekerckhove J, et al. Vascular smooth muscle cells differ from other smooth muscle cells: predominance of vimentin filaments and a specific alpha-type actin. Proc Natl Acad Sci USA. 1981;78:298–302.PubMedPubMedCentralCrossRef Gabbiani G, Schmid E, Winter S, Chaponnier C, de Ckhastonay C, Vandekerckhove J, et al. Vascular smooth muscle cells differ from other smooth muscle cells: predominance of vimentin filaments and a specific alpha-type actin. Proc Natl Acad Sci USA. 1981;78:298–302.PubMedPubMedCentralCrossRef
49.
Zurück zum Zitat van der Loop FT, Gabbiani G, Kohnen G, Ramaekers FC, van Eys GJ. Differentiation of smooth muscle cells in human blood vessels as defined by smoothelin, a novel marker for the contractile phenotype. Arterioscler Thromb Vasc Biol. 1997;17:665–71.PubMedCrossRef van der Loop FT, Gabbiani G, Kohnen G, Ramaekers FC, van Eys GJ. Differentiation of smooth muscle cells in human blood vessels as defined by smoothelin, a novel marker for the contractile phenotype. Arterioscler Thromb Vasc Biol. 1997;17:665–71.PubMedCrossRef
50.
Zurück zum Zitat Mulligan-Kehoe MJ. The vasa vasorum in diseased and nondiseased arteries. Am J Physiol Heart Circ Physiol. 2010;298:H295-305.PubMedCrossRef Mulligan-Kehoe MJ. The vasa vasorum in diseased and nondiseased arteries. Am J Physiol Heart Circ Physiol. 2010;298:H295-305.PubMedCrossRef
51.
Zurück zum Zitat Mohamed SA, Misfeld M, Hanke T, Charitos EI, Bullerdiek J, Belge G, et al. Inhibition of caspase-3 differentially affects vascular smooth muscle cell apoptosis in the concave versus convex aortic sites in ascending aneurysms with a bicuspid aortic valve. Ann Anat. 2010;192:145–50. Mohamed SA, Misfeld M, Hanke T, Charitos EI, Bullerdiek J, Belge G, et al. Inhibition of caspase-3 differentially affects vascular smooth muscle cell apoptosis in the concave versus convex aortic sites in ascending aneurysms with a bicuspid aortic valve. Ann Anat. 2010;192:145–50.
52.
Zurück zum Zitat Della Corte A, Bancone C, Quarto C, Dialetto G, Covino FE, Scardone M, et al. Predictors of ascending aortic dilatation with bicuspid aortic valve: a wide spectrum of disease expression. Eur J Cardiothorac Surg. 2007;31:397–404. Della Corte A, Bancone C, Quarto C, Dialetto G, Covino FE, Scardone M, et al. Predictors of ascending aortic dilatation with bicuspid aortic valve: a wide spectrum of disease expression. Eur J Cardiothorac Surg. 2007;31:397–404.
53.
Zurück zum Zitat Grewal N, Gittenberger-de Groot AC, Poelmann RE, Klautz RJM, Lindeman JHN, Goumans MJ, et al. Ascending aorta dilation in association with bicuspid aortic valve: a maturation defect of the aortic wall. J Thorac Cardiovasc Surg. 2014;148:1583–90. Grewal N, Gittenberger-de Groot AC, Poelmann RE, Klautz RJM, Lindeman JHN, Goumans MJ, et al. Ascending aorta dilation in association with bicuspid aortic valve: a maturation defect of the aortic wall. J Thorac Cardiovasc Surg. 2014;148:1583–90.
54.
55.
Zurück zum Zitat Grewal N, Gittenberger-de Groot AC, Lindeman JH, Klautz A, Driessen A, Klautz RJM, et al. Normal and abnormal development of the aortic valve and ascending aortic wall: a comprehensive overview of the embryology and pathology of the bicuspid aortic valve. Ann Cardiothorac Surg. 2022;11:380–8. Grewal N, Gittenberger-de Groot AC, Lindeman JH, Klautz A, Driessen A, Klautz RJM, et al. Normal and abnormal development of the aortic valve and ascending aortic wall: a comprehensive overview of the embryology and pathology of the bicuspid aortic valve. Ann Cardiothorac Surg. 2022;11:380–8.
56.
Zurück zum Zitat Gittenberger-de Groot AC, Bartelings MM, Deruiter MC, Poelmann RE. Basics of cardiac development for the understanding of congenital heart malformations. Pediatr Res. 2005;57:169–76.PubMedCrossRef Gittenberger-de Groot AC, Bartelings MM, Deruiter MC, Poelmann RE. Basics of cardiac development for the understanding of congenital heart malformations. Pediatr Res. 2005;57:169–76.PubMedCrossRef
57.
Zurück zum Zitat Soto-Navarrete MT, López-Unzu MA, Durán AC, Fernández B. Embryonic development of bicuspid aortic valves. Prog Cardiovasc Dis. 2020;63:407–18. Soto-Navarrete MT, López-Unzu MA, Durán AC, Fernández B. Embryonic development of bicuspid aortic valves. Prog Cardiovasc Dis. 2020;63:407–18.
58.
Zurück zum Zitat Goumans MJ, Mummery C. Functional analysis of the TGFbeta receptor/Smad pathway through gene ablation in mice. Int J Dev Biol. 2000;44:253–65.PubMed Goumans MJ, Mummery C. Functional analysis of the TGFbeta receptor/Smad pathway through gene ablation in mice. Int J Dev Biol. 2000;44:253–65.PubMed
59.
Zurück zum Zitat Balint B, Federspiel J, Kollmann C, Teping P, Schwab T, Schäfers HJ. SMAD3 contributes to ascending aortic dilatation independent of transforming growth factor-beta in bicuspid and unicuspid aortic valve disease. Sci Rep. 2022;12:15476.PubMedPubMedCentralCrossRef Balint B, Federspiel J, Kollmann C, Teping P, Schwab T, Schäfers HJ. SMAD3 contributes to ascending aortic dilatation independent of transforming growth factor-beta in bicuspid and unicuspid aortic valve disease. Sci Rep. 2022;12:15476.PubMedPubMedCentralCrossRef
60.
61.
Zurück zum Zitat Creamer TJ, Bramel EE, MacFarlane EG. Insights on the pathogenesis of aneurysm through the study of hereditary aortopathies. Genes. 2021;12:183. Creamer TJ, Bramel EE, MacFarlane EG. Insights on the pathogenesis of aneurysm through the study of hereditary aortopathies. Genes. 2021;12:183.
62.
Zurück zum Zitat Forte A, Bancone C, Cobellis G, Buonocore M, Santarpino G, Fischlein TJM, et al. A possible early biomarker for bicuspid aortopathy: circulating transforming growth factor β-1 to soluble endoglin ratio. Circ Res. 2017;120:1800–11.PubMedCrossRef Forte A, Bancone C, Cobellis G, Buonocore M, Santarpino G, Fischlein TJM, et al. A possible early biomarker for bicuspid aortopathy: circulating transforming growth factor β-1 to soluble endoglin ratio. Circ Res. 2017;120:1800–11.PubMedCrossRef
63.
Zurück zum Zitat Paloschi V, Gadin JR, Khan S, Bjorck HM, Du L, Maleki S, et al. Aneurysm development in patients with a bicuspid aortic valve is not associated with transforming growth factor-beta activation. Arterioscler Thromb Vasc Biol. 2015;35:973–80.PubMedCrossRef Paloschi V, Gadin JR, Khan S, Bjorck HM, Du L, Maleki S, et al. Aneurysm development in patients with a bicuspid aortic valve is not associated with transforming growth factor-beta activation. Arterioscler Thromb Vasc Biol. 2015;35:973–80.PubMedCrossRef
64.
Zurück zum Zitat Grewal N, Klautz R, Poelmann RE. Can transforming growth factor beta and downstream signalers distinguish bicuspid aortic valve patients susceptible for future aortic complications? Cardiovasc Pathol. 2023;63:107498. Grewal N, Klautz R, Poelmann RE. Can transforming growth factor beta and downstream signalers distinguish bicuspid aortic valve patients susceptible for future aortic complications? Cardiovasc Pathol. 2023;63:107498.
65.
Zurück zum Zitat Dolmaci OB, Ayyildiz T, Poelmann RE, Driessen AHG, Koolbergen DR, Klautz RJM, et al. Risk for acquired coronary artery disease in genetic vs. congenital thoracic aortopathy. Front Cardiovasc Med. 2023;9:1036522. Dolmaci OB, Ayyildiz T, Poelmann RE, Driessen AHG, Koolbergen DR, Klautz RJM, et al. Risk for acquired coronary artery disease in genetic vs. congenital thoracic aortopathy. Front Cardiovasc Med. 2023;9:1036522.
66.
Zurück zum Zitat Gomez D, Al Haj Zen A, Borges LF, Philippe M, Gutierrez PS, Jondeau G, et al. Syndromic and non-syndromic aneurysms of the human ascending aorta share activation of the Smad2 pathway. J Pathol. 2009;218:131–42.PubMedCrossRef Gomez D, Al Haj Zen A, Borges LF, Philippe M, Gutierrez PS, Jondeau G, et al. Syndromic and non-syndromic aneurysms of the human ascending aorta share activation of the Smad2 pathway. J Pathol. 2009;218:131–42.PubMedCrossRef
67.
Zurück zum Zitat Jones JA, Barbour JR, Stroud RE, Bouges S, Stephens SL, Spinale FG, et al. Altered transforming growth factor-beta signaling in a murine model of thoracic aortic aneurysm. J Vasc Res. 2008;45:457–68.PubMedCrossRef Jones JA, Barbour JR, Stroud RE, Bouges S, Stephens SL, Spinale FG, et al. Altered transforming growth factor-beta signaling in a murine model of thoracic aortic aneurysm. J Vasc Res. 2008;45:457–68.PubMedCrossRef
68.
Zurück zum Zitat Wheeler JB, Ikonomidis JS, Jones JA. Connective tissue disorders and cardiovascular complications: the indomitable role of transforming growth factor-beta signaling. Adv Exp Med Biol. 2014;802:107–27.PubMedPubMedCentralCrossRef Wheeler JB, Ikonomidis JS, Jones JA. Connective tissue disorders and cardiovascular complications: the indomitable role of transforming growth factor-beta signaling. Adv Exp Med Biol. 2014;802:107–27.PubMedPubMedCentralCrossRef
69.
Zurück zum Zitat Guzzardi DG, Barker AJ, van Ooij P, Malaisrie SC, Puthumana JJ, Belke DD, et al. Valve-related hemodynamics mediate human bicuspid aortopathy: insights from wall shear stress mapping. J Am Coll Cardiol. 2015;66:892–900.PubMedPubMedCentralCrossRef Guzzardi DG, Barker AJ, van Ooij P, Malaisrie SC, Puthumana JJ, Belke DD, et al. Valve-related hemodynamics mediate human bicuspid aortopathy: insights from wall shear stress mapping. J Am Coll Cardiol. 2015;66:892–900.PubMedPubMedCentralCrossRef
70.
Zurück zum Zitat Poelmann RE, Gittenberger-de Groot AC, Goerdajal C, Grewal N, De Bakker MAG, Richardson MK. Ventricular septation and outflow tract development in crocodilians result in two aortas with bicuspid semilunar valves. J Cardiovasc Dev Dis. 2021;8:132. Poelmann RE, Gittenberger-de Groot AC, Goerdajal C, Grewal N, De Bakker MAG, Richardson MK. Ventricular septation and outflow tract development in crocodilians result in two aortas with bicuspid semilunar valves. J Cardiovasc Dev Dis. 2021;8:132.
Metadaten
Titel
The role of transforming growth factor beta in bicuspid aortic valve aortopathy
verfasst von
Nimrat Grewal
Onur Dolmaci
Arthur Klautz
Juno Legue
Antoine Driessen
Robert Klautz
Robert Poelmann
Publikationsdatum
25.05.2023
Verlag
Springer Nature Singapore
Erschienen in
Indian Journal of Thoracic and Cardiovascular Surgery / Ausgabe Sonderheft 2/2023
Print ISSN: 0970-9134
Elektronische ISSN: 0973-7723
DOI
https://doi.org/10.1007/s12055-023-01513-8

Weitere Artikel der Sonderheft 2/2023

Indian Journal of Thoracic and Cardiovascular Surgery 2/2023 Zur Ausgabe

Vorsicht, erhöhte Blutungsgefahr nach PCI!

10.05.2024 Koronare Herzerkrankung Nachrichten

Nach PCI besteht ein erhöhtes Blutungsrisiko, wenn die Behandelten eine verminderte linksventrikuläre Ejektionsfraktion aufweisen. Das Risiko ist umso höher, je stärker die Pumpfunktion eingeschränkt ist.

Darf man die Behandlung eines Neonazis ablehnen?

08.05.2024 Gesellschaft Nachrichten

In einer Leseranfrage in der Zeitschrift Journal of the American Academy of Dermatology möchte ein anonymer Dermatologe bzw. eine anonyme Dermatologin wissen, ob er oder sie einen Patienten behandeln muss, der eine rassistische Tätowierung trägt.

Deutlich weniger Infektionen: Wundprotektoren schützen!

08.05.2024 Postoperative Wundinfektion Nachrichten

Der Einsatz von Wundprotektoren bei offenen Eingriffen am unteren Gastrointestinaltrakt schützt vor Infektionen im Op.-Gebiet – und dient darüber hinaus der besseren Sicht. Das bestätigt mit großer Robustheit eine randomisierte Studie im Fachblatt JAMA Surgery.

Chirurginnen und Chirurgen sind stark suizidgefährdet

07.05.2024 Suizid Nachrichten

Der belastende Arbeitsalltag wirkt sich negativ auf die psychische Gesundheit der Angehörigen ärztlicher Berufsgruppen aus. Chirurginnen und Chirurgen bilden da keine Ausnahme, im Gegenteil.

Update Chirurgie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.

S3-Leitlinie „Diagnostik und Therapie des Karpaltunnelsyndroms“

Karpaltunnelsyndrom BDC Leitlinien Webinare
CME: 2 Punkte

Das Karpaltunnelsyndrom ist die häufigste Kompressionsneuropathie peripherer Nerven. Obwohl die Anamnese mit dem nächtlichen Einschlafen der Hand (Brachialgia parästhetica nocturna) sehr typisch ist, ist eine klinisch-neurologische Untersuchung und Elektroneurografie in manchen Fällen auch eine Neurosonografie erforderlich. Im Anfangsstadium sind konservative Maßnahmen (Handgelenksschiene, Ergotherapie) empfehlenswert. Bei nicht Ansprechen der konservativen Therapie oder Auftreten von neurologischen Ausfällen ist eine Dekompression des N. medianus am Karpaltunnel indiziert.

Prof. Dr. med. Gregor Antoniadis
Berufsverband der Deutschen Chirurgie e.V.

S2e-Leitlinie „Distale Radiusfraktur“

Radiusfraktur BDC Leitlinien Webinare
CME: 2 Punkte

Das Webinar beschäftigt sich mit Fragen und Antworten zu Diagnostik und Klassifikation sowie Möglichkeiten des Ausschlusses von Zusatzverletzungen. Die Referenten erläutern, welche Frakturen konservativ behandelt werden können und wie. Das Webinar beantwortet die Frage nach aktuellen operativen Therapiekonzepten: Welcher Zugang, welches Osteosynthesematerial? Auf was muss bei der Nachbehandlung der distalen Radiusfraktur geachtet werden?

PD Dr. med. Oliver Pieske
Dr. med. Benjamin Meyknecht
Berufsverband der Deutschen Chirurgie e.V.

S1-Leitlinie „Empfehlungen zur Therapie der akuten Appendizitis bei Erwachsenen“

Appendizitis BDC Leitlinien Webinare
CME: 2 Punkte

Inhalte des Webinars zur S1-Leitlinie „Empfehlungen zur Therapie der akuten Appendizitis bei Erwachsenen“ sind die Darstellung des Projektes und des Erstellungswegs zur S1-Leitlinie, die Erläuterung der klinischen Relevanz der Klassifikation EAES 2015, die wissenschaftliche Begründung der wichtigsten Empfehlungen und die Darstellung stadiengerechter Therapieoptionen.

Dr. med. Mihailo Andric
Berufsverband der Deutschen Chirurgie e.V.