splicing factor
Recently Published Documents


TOTAL DOCUMENTS

1189
(FIVE YEARS 301)

H-INDEX

90
(FIVE YEARS 10)

2022 ◽  
Author(s):  
Adria Closa ◽  
Marina Reixachs-Solé ◽  
Antonio Fuentes-Fayos ◽  
Katharina Hayer ◽  
Juan Melero ◽  
...  

Abstract A significant proportion of B-cell acute lymphoblastic leukemia (B-ALL) patients remains with a dismal prognosis due to yet undetermined mechanisms. We performed a comprehensive multicohort analysis of gene fusions, gene expression, and RNA splicing alterations to uncover molecular signatures potentially linked to the observed poor outcome. We identified 84 fusions with significant allele frequency across patients. We identified an expression signature that predicts high risk independently of the gene fusion background. This signature includes the upregulation of the splicing factor SRRM1, which potentially impacts splicing events associated with poor outcomes through protein-protein interactions with other splicing factors. Experiments in B-ALL cell lines provided further evidence for the role of SRRM1 on cell survival, proliferation, and invasion. Our findings reveal a convergent mechanism of aberrant RNA processing that sustains a malignant phenotype independently of gene fusions and could complement current clinical strategies in B-ALL.


RNA Biology ◽  
2021 ◽  
pp. 1-13
Author(s):  
Sansi Xing ◽  
Jane Wang ◽  
Ruilin Wu ◽  
Marco M. Hefti ◽  
John F. Crary ◽  
...  

2021 ◽  
Author(s):  
Ziva Pogacar ◽  
Kelvin Groot ◽  
Fleur Jochems ◽  
Matheus Dos Santos Dias ◽  
Ben Morris ◽  
...  

Discovering biomarkers of drug response and finding powerful drug combinations can support the reuse of previously abandoned cancer drugs in the clinic. Indisulam is an abandoned drug that acts as a molecular glue, inducing degradation of splicing factor RBM39 through interaction with CRL4DCAF15. Here, we performed genetic and compound screens to uncover factors mediating indisulam sensitivity and resistance. First, a dropout CRISPR screen identified SRPK1 loss as a synthetic lethal interaction with indisulam that can be exploited therapeutically by the SRPK1 inhibitor SPHINX31. Moreover, a CRISPR resistance screen identified components of the degradation complex that mediate resistance to indisulam: DCAF15, DDA1, and CAND1. Lastly, we show that cancer cells readily acquire spontaneous resistance to indisulam. Upon acquiring indisulam resistance, pancreatic cancer (Panc10.05) cells still degrade RBM39 and are vulnerable to BCL-xL inhibition. The better understanding of the factors that influence the response to indisulam can assist rational reuse of this drug in the clinic.


2021 ◽  
Author(s):  
Adria Closa ◽  
Marina Reixachs-Sole ◽  
Antonio C Fuentes-Fayos ◽  
Katharina E Hayer ◽  
Juan L Melero ◽  
...  

A significant proportion of B-cell acute lymphoblastic leukemia (B-ALL) patients remains with a dismal prognosis due to yet undetermined mechanisms. We performed a comprehensive multicohort analysis of gene fusions, gene expression, and RNA splicing alterations to uncover molecular signatures potentially linked to the observed poor outcome. We identified 84 fusions with significant allele frequency across patients. We identified an expression signature that predicts high risk independently of the gene fusion background. This signature includes the upregulation of the splicing factor SRRM1, which potentially impacts splicing events associated with poor outcomes through protein-protein interactions with other splicing factors. Experiments in B-ALL cell lines provided further evidence for the role of SRRM1 on cell survival, proliferation, and invasion. Our findings reveal a convergent mechanism of aberrant RNA processing that sustains a malignant phenotype independently of gene fusions and could complement current clinical strategies in B-ALL.


2021 ◽  
Vol 11 ◽  
Author(s):  
Chen Zhao ◽  
Yingwei Li ◽  
Chunping Qiu ◽  
Jingying Chen ◽  
Huan Wu ◽  
...  

Ovarian carcinoma remains the most lethal gynecological carcinoma. Abnormal expression of splicing factors is closely related to the occurrence and development of tumors. The DEAD-box RNA helicases are important members of the splicing factor family. However, their role in the occurrence and progression of ovarian cancer is still unclear. In this study, we identified DEAD-box helicase 23 (DDX23) as a key DEAD-box RNA helicase in ovarian cancer using bioinformatics methods. We determined that DDX23 was upregulated in ovarian cancer and its high expression predicted poor prognosis. Functional assays indicated that DDX23 silencing significantly impeded cell proliferation/invasion in vitro and tumor growth in vivo. Mechanistically, transcriptomic analysis showed that DDX23 was involved in mRNA processing in ovarian cancer cells. Specifically, DDX23 regulated the mRNA processing of FOXM1. DDX23 silencing reduced the production of FOXM1C, the major oncogenic transcript of FOXM1 in ovarian cancer, thereby decreasing the FOXM1 protein expression and attenuating the malignant progression of ovarian cancer. Rescue assays indicated that FOXM1 was a key executor in DDX23-induced malignant phenotype of ovarian cancer. Furthermore, we confirmed that DDX23 was transcriptionally activated by the transcription factor (TF) E2F1 in ovarian cancer using luciferase reporter assays and chromatin immunoprecipitation (ChIP) assays. In conclusion, our study demonstrates that high DDX23 expression is involved in malignant behavior of ovarian cancer and DDX23 may become a potential target for precision therapy of ovarian cancer.


Biomedicines ◽  
2021 ◽  
Vol 9 (12) ◽  
pp. 1844
Author(s):  
Carmelo Gurnari ◽  
Simona Pagliuca ◽  
Valeria Visconte

Alternative RNA splicing (AS) is an essential physiologic function that diversifies the human proteome. AS also has a crucial role during cellular development. In fact, perturbations in RNA-splicing have been implicated in the development of several cancers, including myeloid malignancies. Splicing dysfunction can be independent of genetic lesions or appear as a direct consequence of mutations in components of the RNA-splicing machinery, such as in the case of mutations occurring in splicing factor genes (i.e., SF3B1, SRSF2, U2AF1) and their regulators. In addition, cancer cells exhibit marked gene expression alterations, including different usage of AS isoforms, possibly causing tissue-specific effects and perturbations of downstream pathways. This review summarizes several modalities leading to splicing diversity in myeloid malignancies.


Author(s):  
Emilia Alors-Perez ◽  
Ricardo Blázquez-Encinas ◽  
Sonia Alcalá ◽  
Cristina Viyuela-García ◽  
Sergio Pedraza-Arevalo ◽  
...  

Abstract Background Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal cancer, requiring novel treatments to target both cancer cells and cancer stem cells (CSCs). Altered splicing is emerging as both a novel cancer hallmark and an attractive therapeutic target. The core splicing factor SF3B1 is heavily altered in cancer and can be inhibited by Pladienolide-B, but its actionability in PDAC is unknown. We explored the presence and role of SF3B1 in PDAC and interrogated its potential as an actionable target. Methods SF3B1 was analyzed in PDAC tissues, an RNA-seq dataset, and publicly available databases, examining associations with splicing alterations and key features/genes. Functional assays in PDAC cell lines and PDX-derived CSCs served to test Pladienolide-B treatment effects in vitro, and in vivo in zebrafish and mice. Results SF3B1 was overexpressed in human PDAC and associated with tumor grade and lymph-node involvement. SF3B1 levels closely associated with distinct splicing event profiles and expression of key PDAC players (KRAS, TP53). In PDAC cells, Pladienolide-B increased apoptosis and decreased multiple tumor-related features, including cell proliferation, migration, and colony/sphere formation, altering AKT and JNK signaling, and favoring proapoptotic splicing variants (BCL-XS/BCL-XL, KRASa/KRAS, Δ133TP53/TP53). Importantly, Pladienolide-B similarly impaired CSCs, reducing their stemness capacity and increasing their sensitivity to chemotherapy. Pladienolide-B also reduced PDAC/CSCs xenograft tumor growth in vivo in zebrafish and in mice. Conclusion SF3B1 overexpression represents a therapeutic vulnerability in PDAC, as altered splicing can be targeted with Pladienolide-B both in cancer cells and CSCs, paving the way for novel therapies for this lethal cancer.


Sign in / Sign up

Export Citation Format

Share Document