Acute lymphoblastic leukemia (ALL) is the most common childhood malignancy. About 80% of the cases originates from precursor B cells (BCP-ALL), which abnormally accumulate as a consequence of genetic alterations associated to differentiation inhibition and abnormal expansion. Despite cure rates exceeding 90% in children, relapse still occurs in about 20% of patients because of the emergence of therapy resistance. In the last years, an increasing number of new molecular targets is emerging from studies on the “leukemic niche” where stromal cells and, in particular, mesenchymal stromal cells (MSCs), could confer support and chemoprotection to leukemic cells. This pro-tumoral effect is the consequence of the corruption of the bone marrow (BM) stroma operated by leukemic cells. In present work, we focused our attention on stroma-derived Activin A, a member of the TGF-β superfamily, well known for its role as a regulator of immune system but also for its tumor-promoting role in several type of solid cancers. Here we found that Activin A could be considered a new crucial factor exploited by leukemic cells to create a self-reinforcing microenvironment: indeed, Activin A was highly expressed in BM plasma of BCP-ALL patients, compared to healthy donors, and its expression was specifically confined to leukemic infiltrated area. In particular, we demonstrated that the engagement of bone marrow stromal cells by leukemic blasts, along with a pro-inflammatory milieu, that characterized the leukemic microenvironment, enhanced Activin A secretion. The main effect exerted by this molecules was the regulation of cell motility associated pathways: indeed, both gene expression analysis and functional assays for evaluation of cell motility, chemotaxis and invasion revealed the ability of Activin A to promote basal and CXCL12-driven migration and invasion of leukemic cells, while it impaired CXCL12-induced migration of healthy CD34+. We found out, according to previous published data, a strong reduction in CXCL12 levels in BM plasma of BCP-ALL patients. It is worth to notice that Activin A was able to increase the responsiveness of leukemic cells even to very low levels of CXCL12. Overall these data suggest a possible mechanism by which leukemic cells could persist in the BM niche, concomitantly displacing healthy heamatopoiesis. Activin A did not alter the expression of CXCL12 receptors, CXCR4 and CXCR7, however it was able to increase both the intracellular calcium content and actin polymerization in leukemic cells. Since Ca2+ signaling is intrinsically related to actin cytoskeletal reorganization and effective cell movement, we hypothesized that Activin A enhanced migration could be ascribed to a calcium-dependent mechanism. Moreover, long-lasting effect of Activin A could be mediated by the regulation of genes associated with Ras and PI3K/AKT pathways as well as calcium homeostatic mechanisms. These molecules may account for the pro-migratory and pro-invasive effects of Activin A. Finally, in vivo model further supported in vitro data about Activin A pro-migratory and pro-invasive role. Overall, our data suggest that Activin A could confer a migratory advantage to leukemic cells to generate a malignant BM niche at the detriment of the healthy hematopoiesis. Therefore, our work will pave the way for the design of new therapeutically approaches for BCP-ALL treatment aimed to target the interplay between leukemic cells and BM niche.

La Leucemia Linfoblastica Acuta (LLA) è il più comune tumore in età pediatrica. La percentuale di successo terapeutico è del 90%, tuttavia la recidiva si può presentare in quasi il 20% dei casi per l’insorgenza di meccanismi di resistenza farmacologica. Lo studio della “nicchia leucemica" ha permesso di individuare potenziali bersagli molecolari grazie allo studio delle modalità con cui le cellule stromali mesenchimali (MSC), riprogrammate dalle cellule leucemiche, conferiscono supporto e chemio resistenza. In questo lavoro ci siamo focalizzati sul ruolo di Attivina A, molecola della superfamiglia del TGF-β, nell’interazione tra cellule leucemiche e stroma midollare. Recentemente Attivina A è stata identificata in diversi tipi di tumori solidi come molecola capace di promuovere migrazione e invasione, le quali possono favorire la progressione tumorale. Il presente lavoro definisce per la prima volta il ruolo di Attivina A come molecola chiave nell’interazione tra cellule leucemiche e MSC capace di contribuire alla generazione di un microambiente in grado di sostenere la progressione leucemica. Attivina A è altamente espressa nel midollo dei pazienti LLA-B e, nello specifico, tale molecola è prodotta all’interno del midollo leucemico nelle aree infiltrate dai blasti leucemici. Inoltre, la stimolazione da parte di blasti leucemici e di molecole pro-infiammatorie, tipiche del microambiente midollare leucemico, è in grado di aumentare la produzione di Attivina A nelle cellule mesenchimali stromali. Tale molecola agisce principalmente regolando i meccanismi di motilità cellulare e di omeostasi dello ione calcio. Sia l’analisi dell’espressione genica sia i saggi funzionali hanno messo in luce la capacità di Attivina A di promuovere nelle cellule leucemiche i processi di migrazione e di invasività, in presenza o meno di un fattore chemotattico, quale CXCL12. Tale molecola è fondamentale nella localizzazione delle cellule staminali ematopoietiche e delle cellule leucemiche all’interno della nicchia midollare. Pertanto, la ridotta espressione di CXCL12 nel midollo leucemico insieme all’aumentata espressione di Attivina A, associata sia ad un aumento della sensibilità delle cellule leucemiche a CXCL12 sia a una riduzione della risposta delle cellule staminali sane a CXCL12, possono essere considerati come meccanismo di persistenza delle cellule leucemiche all’interno della nicchia, a discapito dell’ematopoiesi sana. Il meccanismo di azione di Attivina A non dipende dalla regolazione dell’espressione dei recettori di CXCL12, CXCR4 e CXCR7, ma è mediato dall’incremento dei livelli di calcio intracellulare, il quale ha un ruolo chiave nella riorganizzazione citoscheletrica e nel movimento cellulare. Inoltre, è possibile identificare un effetto a lungo termine determinato dalla regolazione di geni associati ad assi molecolari, quali Ras e PI3K/AKT, e a meccanismi di regolazione del calcio, i quali possono spiegare l’effetto pro-migratorio di Attivina A. Infine, il modello in vivo di malattia ha permesso di confermare il ruolo pro-invasivo di Attivina A. Complessivamente, questo lavoro definisce il meccanismo con cui Attivina A è in grado di conferire un vantaggio alle cellule leucemiche, tale da renderle in grado di generare una nicchia maligna a discapito dell’ematopoiesi sana. Pertanto, in futuro, tale meccanismo potrebbe essere bersagliato con nuovi approcci terapeutici per il trattamento della LLA-B.

(2018). Activin A as a new key factor in the leukemic bone marrow niche. (Tesi di dottorato, Università degli Studi di Milano-Bicocca, 2018).

Activin A as a new key factor in the leukemic bone marrow niche

PORTALE, FEDERICA
2018

Abstract

Acute lymphoblastic leukemia (ALL) is the most common childhood malignancy. About 80% of the cases originates from precursor B cells (BCP-ALL), which abnormally accumulate as a consequence of genetic alterations associated to differentiation inhibition and abnormal expansion. Despite cure rates exceeding 90% in children, relapse still occurs in about 20% of patients because of the emergence of therapy resistance. In the last years, an increasing number of new molecular targets is emerging from studies on the “leukemic niche” where stromal cells and, in particular, mesenchymal stromal cells (MSCs), could confer support and chemoprotection to leukemic cells. This pro-tumoral effect is the consequence of the corruption of the bone marrow (BM) stroma operated by leukemic cells. In present work, we focused our attention on stroma-derived Activin A, a member of the TGF-β superfamily, well known for its role as a regulator of immune system but also for its tumor-promoting role in several type of solid cancers. Here we found that Activin A could be considered a new crucial factor exploited by leukemic cells to create a self-reinforcing microenvironment: indeed, Activin A was highly expressed in BM plasma of BCP-ALL patients, compared to healthy donors, and its expression was specifically confined to leukemic infiltrated area. In particular, we demonstrated that the engagement of bone marrow stromal cells by leukemic blasts, along with a pro-inflammatory milieu, that characterized the leukemic microenvironment, enhanced Activin A secretion. The main effect exerted by this molecules was the regulation of cell motility associated pathways: indeed, both gene expression analysis and functional assays for evaluation of cell motility, chemotaxis and invasion revealed the ability of Activin A to promote basal and CXCL12-driven migration and invasion of leukemic cells, while it impaired CXCL12-induced migration of healthy CD34+. We found out, according to previous published data, a strong reduction in CXCL12 levels in BM plasma of BCP-ALL patients. It is worth to notice that Activin A was able to increase the responsiveness of leukemic cells even to very low levels of CXCL12. Overall these data suggest a possible mechanism by which leukemic cells could persist in the BM niche, concomitantly displacing healthy heamatopoiesis. Activin A did not alter the expression of CXCL12 receptors, CXCR4 and CXCR7, however it was able to increase both the intracellular calcium content and actin polymerization in leukemic cells. Since Ca2+ signaling is intrinsically related to actin cytoskeletal reorganization and effective cell movement, we hypothesized that Activin A enhanced migration could be ascribed to a calcium-dependent mechanism. Moreover, long-lasting effect of Activin A could be mediated by the regulation of genes associated with Ras and PI3K/AKT pathways as well as calcium homeostatic mechanisms. These molecules may account for the pro-migratory and pro-invasive effects of Activin A. Finally, in vivo model further supported in vitro data about Activin A pro-migratory and pro-invasive role. Overall, our data suggest that Activin A could confer a migratory advantage to leukemic cells to generate a malignant BM niche at the detriment of the healthy hematopoiesis. Therefore, our work will pave the way for the design of new therapeutically approaches for BCP-ALL treatment aimed to target the interplay between leukemic cells and BM niche.
BIONDI, ANDREA
D'AMICO, GIOVANNA
ALL;; Activin-A;; MSC;; migration;; invasion
ALL;; Activin-A;; MSC;; migration;; invasion
MED/38 - PEDIATRIA GENERALE E SPECIALISTICA
English
16-mar-2018
MEDICINA TRASLAZIONALE E MOLECOLARE - DIMET - 76R
30
2016/2017
open
(2018). Activin A as a new key factor in the leukemic bone marrow niche. (Tesi di dottorato, Università degli Studi di Milano-Bicocca, 2018).
File in questo prodotto:
File Dimensione Formato  
phd_unimib_798733.pdf

Accesso Aperto

Descrizione: tesi di dottorato
Tipologia di allegato: Doctoral thesis
Dimensione 4.1 MB
Formato Adobe PDF
4.1 MB Adobe PDF Visualizza/Apri

I documenti in IRIS 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/10281/199027
Citazioni
  • Scopus ND
  • ???jsp.display-item.citation.isi??? ND
Social impact