Calcific aortic valve disease (CAVD) is a degenerative, inflammatory process characterized by a progressive calcification of aortic valve (AV). It is the third leading cause of death among cardiovascular diseases (CVD) for which the only effective therapeutic solution is surgical valve replacement. Inflammation plays a key role in AV pathological remodeling; hence, anti-inflammatory therapy has been proposed as a strategy to prevent CAVD. Cyclooxygenase 2 (COX-2) is a key mediator of the inflammation, and it is the target of widely used anti-inflammatory drugs. The COX-2 inhibitor celecoxib was first shown to reduce calcification of AV. However, a recent retrospective clinical analysis found an association between CAVD and celecoxib use. Several studies have also shown that dysregulation of the Notch pathway in aortic valve interstitial cells (AVICs) and valve endothelial cells (VECs) is involved in the pathogenesis of CAVD. However, the precise mechanisms and the role of Notch components pathway in the calcific valve degeneration remain unknown. In the context of CVD, recent data show that somatic mutations leading to clonal expansion of hematopoietic stem cells, known as clonal hematopoiesis of indeterminate potential (CHIP), correlates with an increased risk of CVD such as atherosclerosis, and coronary-artery calcification. The aims of this study were i) to investigate the role of COX-2 and Notch ligands Delta-like 4 (Dll4) and Jagged1 in CAVD by establishing whether a variation in the expression levels of these molecules in specific cell type in human AVs may be linked to CAVD, ii) to assess whether CHIP is related to an increase in the incidence of CAVD and uncover the molecular mechanism by which these mutations could affect the aortic valve environment. For the first aim we extracted total RNA from surgically explanted AVs from patients without or with CAVD and we isolated human AVICs. We then quantified by qRT-PCR the levels of COX-2 and Notch ligands and we explored how COX-2 inhibition affects AVICs trans-differentiation towards a myofibroblast phenotype. In addition, we transduced AVICs in presence of osteogenic medium with lentivirus bearing Dll4, Jagged1 or their inhibitors, and calcium deposition was analyzed through Alizarine red staining. For the second aim we extracted DNA from blood samples of CAVD patients undergoing aortic valve replacement surgery or aortic transcatheter valve transplantation (TAVI) and from patients without CAVD but with severe aortic insufficiency or aneurysm of the aortic root; DNA was analyzed through new generation sequencing techniques (NGS). Then, we performed RNA-seq analyses from RNA extracted from CHIP and non-CHIP patients. We found that: i) COX-2 inhibition with celecoxib promote AVICs trans-differentiation towards a myofibroblast phenotype and increases the levels of TGF-β-induced apoptosis, leading to progression of aortic valve calcification; ii) Jagged1 and COX-2 expression was higher in non-calcific compared to CAVD samples. Conversely, Dll4 expression was lower in non-calcific vs CAVD samples. Of note, COX-2 expression correlated, directly or inversely, with Jagged1 or Dll4 levels, respectively; iii) Furthermore, Alizarin red staining suggested that inhibition of Jagged1 reduced calcium deposition. On the contrary, inhibition of Dll4 induced calcification; iv) the prevalence of CHIP in our population is 30,4% and the most frequently mutated genes are TET2 and DNMT3A; v) CHIP and non-CHIP patients shared altered expression of several genes, include genes already implicated in CAVD and related to extracellular matrix and osteo-chondrocyte transition and highlight that the involvement of adaptive immune activation is augmented in CHIP carriers and frequently involves humoral responses. These findings could be relevant since the identification of the major players involved in CAVD can pave the way for the development of biologically based tailored therapeutic approach.

La stenosi della valvola aortica (AVS) è la terza causa di morte tra le malattie cardiovascolari. Non esistono terapie farmacologiche efficaci per prevenire o rallentare la progressione di questa patologia e l’unico rimedio è la sostituzione della valvola tramite chirurgia. L'infiammazione ha un ruolo fondamentale nella patogenesi di AVS, per cui, la terapia antinfiammatoria è stata proposta come possibile strategia terapeutica. La cicloossigenasi- 2 (COX-2) è un mediatore chiave dell'infiammazione. È noto che il celecoxib, inibitore di COX-2, riduce la calcificazione della valvola aortica. Tuttavia, un recente studio clinico retrospettivo ha riscontrato un’associazione tra l’uso di celecoxib e AVS. È noto che la disregolazione del pathway di Notch nelle cellule interstiziali della valvola aortica (AVICs) e nelle cellule endoteliali della valvola (VEC) è coinvolta nella patogenesi di AVS. Tuttavia, il ruolo preciso dei ligandi e recettori di Notch in AVS non è del tutto chiarito. Dati recenti mostrano che la presenza di mutazioni somatiche a carico di cellule staminali ematopoietiche, condizione nota come ematopoiesi clonale di potenziale indeterminato (CHIP), correla con un aumentato rischio di malattie cardiovascolari ma non è noto se CHIP sia associato o meno ad AVS. Questo studio si propone di: i) studiare il coinvolgimento di COX-2 e dei ligandi di Notch, Jagged1 e Dll4, nella patogenesi di AVS; ii) valutare se CHIP è associato ad un aumento dell'incidenza di AVS ed analizzare le differenze nel trascrittoma delle valvole calcifiche derivanti da pazienti CHIP e non-CHIP e da valvole non calcifiche. Per il primo scopo, è stato estratto l'RNA da valvole aortiche calcifiche e non, e sono stati quantificati mediante qRT-PCR i livelli di Jagged1, Dll4 e di COX-2. È stato inoltre indagato se l'inibizione di COX-2 influenzi la trans-differenziazione delle AVICs verso un fenotipo miofibroblastico. Inoltre, le AVICs derivanti da valvole di pazienti AVS sono state transfettate con vettori lentivirali che overesprimono Dll4, Jagged1 o i loro inibitori e la deposizione di calcio è stata analizzata mediante il saggio di Alizarina Red. Per il secondo scopo è stato estratto il DNA da campioni di sangue di pazienti sottoposti a sostituzione della valvola aortica tramite chirurgia o tramite metodo transcatetere (TAVI) e un gruppo di controllo non affetti da AVS. Il DNA è stato analizzato mediante tecniche di sequenziamento di nuova generazione (NGS). Inoltre, sono state eseguite analisi di RNA-seq da RNA estratto da valvole derivanti da pazienti affetti da mutazioni CHIP per i geni DNMT3A e TET2, valvole non calcifiche o calcifiche ma non CHIP. I risultati ottenuti hanno mostrato che: i) l'inibizione di COX-2 promuove la trans-differenziazione delle AVICs in un fenotipo miofibroblastico e aumenta i livelli di apoptosi indotta da TGF-β, contribuendo alla progressione della calcificazione; ii) i livelli di espressione di Jagged1 e di COX-2 sono più elevati nelle valvole non calcifiche rispetto a quelle AVS. Al contrario i livelli di espressione di Dll4 sono inferiori nelle valvole non calcifiche rispetto a quelle AVS. Inoltre, abbiamo osservato che i livelli di espressione di COX-2 correlano, direttamente o inversamente, rispettivamente con i livelli di Jagged1 o Dll4; iii) le analisi di Alizarina Red hanno suggerito un ruolo pro-calcifico per Jagged1 e anti-calcifico per Dll4; iv) la prevalenza di CHIP in AVS è del 30,4% e i geni più frequentemente mutati sono TET2 e DNMT3A; v) i pazienti CHIP e non-CHIP condividono l’alterata espressione di geni correlati al differenziamento osteo-condrogenico e, inoltre, nei pazienti CHIP è stato evidenziato un aumento del coinvolgimento dell’immunità adattativa. Questi risultati hanno rilevanza traslazionale poiché l'identificazione dei principali attori coinvolti in AVS può aprire la strada allo sviluppo di un approccio biologico terapeutico personalizzato.

Study of factors involved in the calcific aortic valve disease

MARRACINO, Luisa
2022

Abstract

Calcific aortic valve disease (CAVD) is a degenerative, inflammatory process characterized by a progressive calcification of aortic valve (AV). It is the third leading cause of death among cardiovascular diseases (CVD) for which the only effective therapeutic solution is surgical valve replacement. Inflammation plays a key role in AV pathological remodeling; hence, anti-inflammatory therapy has been proposed as a strategy to prevent CAVD. Cyclooxygenase 2 (COX-2) is a key mediator of the inflammation, and it is the target of widely used anti-inflammatory drugs. The COX-2 inhibitor celecoxib was first shown to reduce calcification of AV. However, a recent retrospective clinical analysis found an association between CAVD and celecoxib use. Several studies have also shown that dysregulation of the Notch pathway in aortic valve interstitial cells (AVICs) and valve endothelial cells (VECs) is involved in the pathogenesis of CAVD. However, the precise mechanisms and the role of Notch components pathway in the calcific valve degeneration remain unknown. In the context of CVD, recent data show that somatic mutations leading to clonal expansion of hematopoietic stem cells, known as clonal hematopoiesis of indeterminate potential (CHIP), correlates with an increased risk of CVD such as atherosclerosis, and coronary-artery calcification. The aims of this study were i) to investigate the role of COX-2 and Notch ligands Delta-like 4 (Dll4) and Jagged1 in CAVD by establishing whether a variation in the expression levels of these molecules in specific cell type in human AVs may be linked to CAVD, ii) to assess whether CHIP is related to an increase in the incidence of CAVD and uncover the molecular mechanism by which these mutations could affect the aortic valve environment. For the first aim we extracted total RNA from surgically explanted AVs from patients without or with CAVD and we isolated human AVICs. We then quantified by qRT-PCR the levels of COX-2 and Notch ligands and we explored how COX-2 inhibition affects AVICs trans-differentiation towards a myofibroblast phenotype. In addition, we transduced AVICs in presence of osteogenic medium with lentivirus bearing Dll4, Jagged1 or their inhibitors, and calcium deposition was analyzed through Alizarine red staining. For the second aim we extracted DNA from blood samples of CAVD patients undergoing aortic valve replacement surgery or aortic transcatheter valve transplantation (TAVI) and from patients without CAVD but with severe aortic insufficiency or aneurysm of the aortic root; DNA was analyzed through new generation sequencing techniques (NGS). Then, we performed RNA-seq analyses from RNA extracted from CHIP and non-CHIP patients. We found that: i) COX-2 inhibition with celecoxib promote AVICs trans-differentiation towards a myofibroblast phenotype and increases the levels of TGF-β-induced apoptosis, leading to progression of aortic valve calcification; ii) Jagged1 and COX-2 expression was higher in non-calcific compared to CAVD samples. Conversely, Dll4 expression was lower in non-calcific vs CAVD samples. Of note, COX-2 expression correlated, directly or inversely, with Jagged1 or Dll4 levels, respectively; iii) Furthermore, Alizarin red staining suggested that inhibition of Jagged1 reduced calcium deposition. On the contrary, inhibition of Dll4 induced calcification; iv) the prevalence of CHIP in our population is 30,4% and the most frequently mutated genes are TET2 and DNMT3A; v) CHIP and non-CHIP patients shared altered expression of several genes, include genes already implicated in CAVD and related to extracellular matrix and osteo-chondrocyte transition and highlight that the involvement of adaptive immune activation is augmented in CHIP carriers and frequently involves humoral responses. These findings could be relevant since the identification of the major players involved in CAVD can pave the way for the development of biologically based tailored therapeutic approach.
RIZZO, Paola
File in questo prodotto:
File Dimensione Formato  
Thesis Marracino Luisa.pdf

Open Access dal 16/06/2023

Descrizione: Thesis Marracino Luisa
Tipologia: Tesi di dottorato
Dimensione 13.1 MB
Formato Adobe PDF
13.1 MB Adobe PDF Visualizza/Apri

I documenti in SFERA sono protetti da copyright e tutti i diritti sono riservati, salvo diversa indicazione.

Utilizza questo identificativo per citare o creare un link a questo documento: https://hdl.handle.net/11392/2490102
 Attenzione

Attenzione! I dati visualizzati non sono stati sottoposti a validazione da parte dell'ateneo

Citazioni
  • ???jsp.display-item.citation.pmc??? ND
  • Scopus ND
  • ???jsp.display-item.citation.isi??? ND
social impact