SETBP1 oncoprotein has been involved in several myeloid disorders including atypical chronic myeloid leukemia (aCML), secondary acute myeloid leukemia, chronic myelomonocytic leukemia, juvenile myelomonocytic leukemia, chronic neutrophilic leukemia, as well as different myelodysplastic syndromes such as refractory anemia with excess blasts and refractory cytopenia with multilineage dysplasia. SETBP1 mutations usually co-occur with mutations in other driver genes and, at least in aCML, they appear as later events during oncogenesis. Nevertheless, Setbp1 overexpression confers self-renewal capability to myeloid progenitors in vitro. Thus, it is not yet understood whether SETBP1 can initiate leukemia in vivo. To answer this question, we generated a conditional mouse model expressing mutated SETBP1 in hematopoietic tissue. Mice developed a myeloid neoplasm featuring mature myeloid leukocytosis with dysplasia restricted to the megakaryocytic lineage, massive splenomegaly and bone marrow fibrosis. Single-cell transcriptome analysis revealed a striking alteration in the differentiation program of the hematopoietic progenitors, with profound dysregulation of several master regulators and effectors of the hematopoietic commitment such as Gata1, Mecom, Hoxa9. The unexpected finding of fibrotic bone marrow and massive splenomegaly in these mice prompted us to search for a possible driver role for SETBP1 mutations in the onset of human myelofibrosis. As most cases are already known to be driven by JAK2, CALR or MPL somatic mutations, we focused our attention on triple-negative primary myelofibrosis (TN-PMF). To this end, we selected a cohort of 36 TN-PMF cases showing no evidence of JAK2, CALR or MPL somatic variants, and characterized them by multi-omics analyses. We identified two clearly distinct subgroups, one with no clonal mutations and the other one characterized by somatic SETBP1 mutations, all occurring in the degron hotspot, a highly conserved mutational hotspot included in the SKI homologous region that controls the stability of SETBP1 protein. Clinically, a striking difference in disease aggressiveness was noted between the two groups, with SETBP1-mutated patients showing a much worse clinical course and severe prognosis. Since SETBP1 mutations are usually found as late events in MDS/MPN neoplasms, our hypothesis was that the diverging phenotype may be elucidated by different clonal architectures, considering the significant impact of clonal hierarchy on tumor phenotype and clinical severity. Hence, we applied single cell clonal hierarchy analysis; such approach allows to determine the lineage connections among somatic variants and unveil the order in which mutations arose in cells. This analysis showed that in these cases SETBP1 mutations represented an early driver event, suggesting that different SETBP1+ disorders are possibly triggered by an opposite hierarchy of the same clonal variants. These data indicate that (a) mutated SETBP1 is sufficient to induce a myeloproliferative disorder; and (b) early SETBP1 mutations mark an aggressive subtype of TN-PMF which could benefit from a more aggressive clinical approach.

Mutazioni sul gene SETBP1 e la relativa oncoproteina SETBP1 sono coinvolte in diversi disturbi mieloidi tra cui la leucemia mieloide cronica atipica (aCML), la leucemia mieloide acuta secondaria, la leucemia milomonocitica cronica, la leucemia mielomonocitica giovanile, la leucemia neutrofilica cronica; inoltre mutazioni di SETBP1 sono state riscontrate in diversi casi di sindromi mielodisplastiche tra cui anemia refrattaria con eccesso di blasti e citopenia refrattaria con displasia multilineare. Le mutazioni di SETBP1 di solito si verificano contemporaneamente a mutazioni in altri geni driver e, almeno nella aCML, sembrano essere eventi successivi durante l'oncogenesi. L’overespressione di Setbp1 conferisce la capacità di auto-rinnovamento ai progenitori mieloidi in vitro; pertanto, non è ancora chiaro se SETBP1 possa essere in grado di iniziare la patogenesi leucemica in vivo. Per rispondere a questa domanda, abbiamo generato un modello murino condizionale che esprime SETBP1 mutato nei tessuti ematopoietici. I topi hanno sviluppato una neoplasia mieloide caratterizzata da leucocitosi mieloide matura con displasia limitata alla linea megacariocitica, splenomegalia massiva e fibrosi del midollo osseo. L'analisi del trascrittoma a singola cellula ha rivelato una drastica alterazione del programma di differenziamento dei progenitori ematopoietici, con profonda deregolazione di diversi geni coinvolti nel differenziamento in senso ematopoietico come Gata1, Mecom, Hoxa9. La scoperta inaspettata di midollo osseo fibrotico e splenomegalia massiva in questi topi ci ha spinto a cercare un possibile ruolo di driver per le mutazioni di SETBP1 nell'insorgenza della mielofibrosi umana. Poiché la maggior parte dei casi sono già noti essere guidati da mutazioni somatiche di JAK2, CALR o MPL, abbiamo concentrato la nostra attenzione sulla mielofibrosi primaria tripla negativa (TN-PMF). A tal fine, abbiamo selezionato una coorte di 36 casi di TN-PMF che non mostravano evidenza di varianti somatiche di JAK2, CALR o MPL e li abbiamo caratterizzati con analisi multi-omiche. Abbiamo identificato due sottogruppi chiaramente distinti, uno senza mutazioni clonali e l'altro caratterizzato da mutazioni somatiche di SETBP1, tutte occorrenti in uno specifico hotspot mutazionale: il degron domain. Questo hotspot mutazionale altamente conservato incluso nella regione omologa SKI controlla la stabilità della proteina SETBP1. Clinicamente, è stata notata una differenza sorprendente nell'aggressività della malattia tra i due gruppi, con pazienti con mutazioni di SETBP1 che mostrano un decorso clinico molto peggiore e una prognosi grave. Poiché le mutazioni di SETBP1 di solito si trovano come eventi tardivi in neoplasie MDS/MPN, la nostra ipotesi era che il fenotipo divergente potesse essere spiegato da diverse architetture clonali, considerando l'impatto significativo di tale aspetto sul fenotipo tumorale e sulla gravità clinica. Pertanto, abbiamo applicato l'analisi gerarchica di cloni a singola cellula; tale approccio consente di determinare le connessioni di linea tra varianti somatiche e di svelare l'ordine in cui le mutazioni sono emerse nelle cellule. Questa analisi ha mostrato che in questi casi le mutazioni di SETBP1 rappresentavano un evento precoce nello sviluppo della malattia, suggerendo che diverse patologie associate a SETBP1 possano essere innescate da un'opposta gerarchia delle stesse varianti clonali.. Questi dati indicano che (a) SETBP1 mutato è sufficiente per indurre una malattia mieloproliferativa; e (b) mutazioni precoci di SETBP1 sono marker importanti di un sottotipo aggressivo di TN-PMF che potrebbe beneficiare di un approccio clinico più aggressivo.

(2023). First-hit SETBP1 mutations cause a myeloproliferative disorder with bone marrow fibrosis. (Tesi di dottorato, Università degli Studi di Milano-Bicocca, 2023).

First-hit SETBP1 mutations cause a myeloproliferative disorder with bone marrow fibrosis

CRESPIATICO, ILARIA
2023

Abstract

SETBP1 oncoprotein has been involved in several myeloid disorders including atypical chronic myeloid leukemia (aCML), secondary acute myeloid leukemia, chronic myelomonocytic leukemia, juvenile myelomonocytic leukemia, chronic neutrophilic leukemia, as well as different myelodysplastic syndromes such as refractory anemia with excess blasts and refractory cytopenia with multilineage dysplasia. SETBP1 mutations usually co-occur with mutations in other driver genes and, at least in aCML, they appear as later events during oncogenesis. Nevertheless, Setbp1 overexpression confers self-renewal capability to myeloid progenitors in vitro. Thus, it is not yet understood whether SETBP1 can initiate leukemia in vivo. To answer this question, we generated a conditional mouse model expressing mutated SETBP1 in hematopoietic tissue. Mice developed a myeloid neoplasm featuring mature myeloid leukocytosis with dysplasia restricted to the megakaryocytic lineage, massive splenomegaly and bone marrow fibrosis. Single-cell transcriptome analysis revealed a striking alteration in the differentiation program of the hematopoietic progenitors, with profound dysregulation of several master regulators and effectors of the hematopoietic commitment such as Gata1, Mecom, Hoxa9. The unexpected finding of fibrotic bone marrow and massive splenomegaly in these mice prompted us to search for a possible driver role for SETBP1 mutations in the onset of human myelofibrosis. As most cases are already known to be driven by JAK2, CALR or MPL somatic mutations, we focused our attention on triple-negative primary myelofibrosis (TN-PMF). To this end, we selected a cohort of 36 TN-PMF cases showing no evidence of JAK2, CALR or MPL somatic variants, and characterized them by multi-omics analyses. We identified two clearly distinct subgroups, one with no clonal mutations and the other one characterized by somatic SETBP1 mutations, all occurring in the degron hotspot, a highly conserved mutational hotspot included in the SKI homologous region that controls the stability of SETBP1 protein. Clinically, a striking difference in disease aggressiveness was noted between the two groups, with SETBP1-mutated patients showing a much worse clinical course and severe prognosis. Since SETBP1 mutations are usually found as late events in MDS/MPN neoplasms, our hypothesis was that the diverging phenotype may be elucidated by different clonal architectures, considering the significant impact of clonal hierarchy on tumor phenotype and clinical severity. Hence, we applied single cell clonal hierarchy analysis; such approach allows to determine the lineage connections among somatic variants and unveil the order in which mutations arose in cells. This analysis showed that in these cases SETBP1 mutations represented an early driver event, suggesting that different SETBP1+ disorders are possibly triggered by an opposite hierarchy of the same clonal variants. These data indicate that (a) mutated SETBP1 is sufficient to induce a myeloproliferative disorder; and (b) early SETBP1 mutations mark an aggressive subtype of TN-PMF which could benefit from a more aggressive clinical approach.
PIAZZA, ROCCO GIOVANNI
MOLOGNI, LUCA
EMATOLOGIA; MIELOFIBROSI; SETBP1; SINGLE CELL; MODELLO MURINO
HEMATOLOGY; MYELOFIBROSIS; SETBP1; SINGLE CELL; MOUSE MODEL
MED/15 - MALATTIE DEL SANGUE
English
4-lug-2023
35
2021/2022
embargoed_20260704
(2023). First-hit SETBP1 mutations cause a myeloproliferative disorder with bone marrow fibrosis. (Tesi di dottorato, Università degli Studi di Milano-Bicocca, 2023).
File in questo prodotto:
File Dimensione Formato  
phd_unimib_821737.pdf

embargo fino al 04/07/2026

Descrizione: Tesi di CRESPIATICO ILARIA-821737
Tipologia di allegato: Doctoral thesis
Dimensione 3.56 MB
Formato Adobe PDF
3.56 MB Adobe PDF   Visualizza/Apri   Richiedi una copia

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/10281/429863
Citazioni
  • Scopus ND
  • ???jsp.display-item.citation.isi??? ND
Social impact