intronic splicing enhancer
Recently Published Documents


TOTAL DOCUMENTS

20
(FIVE YEARS 3)

H-INDEX

11
(FIVE YEARS 1)

2021 ◽  
Vol 12 (1) ◽  
Author(s):  
Masahiko Ajiro ◽  
Tomonari Awaya ◽  
Young Jin Kim ◽  
Kei Iida ◽  
Masatsugu Denawa ◽  
...  

AbstractApproximately half of genetic disease-associated mutations cause aberrant splicing. However, a widely applicable therapeutic strategy to splicing diseases is yet to be developed. Here, we analyze the mechanism whereby IKBKAP-familial dysautonomia (FD) exon 20 inclusion is specifically promoted by a small molecule splice modulator, RECTAS, even though IKBKAP-FD exon 20 has a suboptimal 5′ splice site due to the IVS20 + 6 T > C mutation. Knockdown experiments reveal that exon 20 inclusion is suppressed in the absence of serine/arginine-rich splicing factor 6 (SRSF6) binding to an intronic splicing enhancer in intron 20. We show that RECTAS directly interacts with CDC-like kinases (CLKs) and enhances SRSF6 phosphorylation. Consistently, exon 20 splicing is bidirectionally manipulated by targeting cellular CLK activity with RECTAS versus CLK inhibitors. The therapeutic potential of RECTAS is validated in multiple FD disease models. Our study indicates that small synthetic molecules affecting phosphorylation state of SRSFs is available as a new therapeutic modality for mechanism-oriented precision medicine of splicing diseases.


2018 ◽  
Vol 92 (8) ◽  
pp. e02050-17 ◽  
Author(s):  
Safder S. Ganaie ◽  
Aaron Yun Chen ◽  
Chun Huang ◽  
Peng Xu ◽  
Steve Kleiboeker ◽  
...  

ABSTRACTHuman parvovirus B19 (B19V) expresses a single precursor mRNA (pre-mRNA), which undergoes alternative splicing and alternative polyadenylation to generate 12 viral mRNA transcripts that encode two structural proteins (VP1 and VP2) and three nonstructural proteins (NS1, 7.5-kDa protein, and 11-kDa protein). Splicing at the second 5′ donor site (D2 site) of the B19V pre-mRNA is essential for the expression of VP2 and the 11-kDa protein. We previously identified thatcis-acting intronic splicing enhancer 2 (ISE2) that lies immediately after the D2 site facilitates the recognition of the D2 donor for its efficient splicing. In this study, we report that ISE2 is critical for the expression of the 11-kDa viral nonstructural protein. We found that ISE2 harbors a consensus RNA binding motif protein 38 (RBM38) binding sequence, 5′-UGUGUG-3′. RBM38 is expressed during the middle stage of erythropoiesis. We first confirmed that RBM38 binds specifically with the ISE2 elementin vitro. The knockdown of RBM38 significantly decreases the level of spliced mRNA at D2 that encodes the 11-kDa protein but not that of the D2-spliced mRNA that encodes VP2. Importantly, we found that the 11-kDa protein enhances viral DNA replication and virion release. Accordingly, the knockdown of RBM38 decreases virus replication via downregulating 11-kDa protein expression. Taken together, these results suggest that the 11-kDa protein facilitates B19V DNA replication and that RBM38 is an essential host factor for B19V pre-mRNA splicing and for the expression of the 11-kDa protein.IMPORTANCEB19V is a human pathogen that can cause fifth disease, arthropathy, anemia in immunocompromised patients and sickle cell disease patients, myocarditis, and hydrops fetalis in pregnant women. Human erythroid progenitor cells (EPCs) are most susceptible to B19V infection and fully support viral DNA replication. The exclusive tropism of B19V for erythroid-lineage cells is dependent not only on the expression of viral receptors and coreceptors on the cell surface but also on the intracellular host factors that support B19V replication. Our present study shows that B19V uses a host factor, RNA binding motif protein 38 (RBM38), for the processing of its pre-mRNA during virus replication. Specifically, RBM38 interacts with the intronic splicing enhancer 2 (ISE2) element of B19V pre-mRNA and promotes 11-kDa protein expression, thereby regulating the 11-kDa protein-mediated augmentation of B19V replication. The identification of this novel host-pathogen interaction will provide mechanistic insights into B19V replication and aid in finding new targets for anti-B19V therapeutics.


2009 ◽  
Vol 37 (21) ◽  
pp. 7258-7267 ◽  
Author(s):  
Tibor Pastor ◽  
Gabriele Talotti ◽  
Marzena Anna Lewandowska ◽  
Franco Pagani

2009 ◽  
Vol 83 (12) ◽  
pp. 6306-6311 ◽  
Author(s):  
Eun-Young Choi ◽  
David Pintel

ABSTRACT The essential proteins NS1 and NS2 of minute virus of mice are encoded by mRNAs R1 and R2, respectively. R2 is derived from R1 by excision of a large intron and thus splicing governs the relative ratios of NS1 and NS2. Excision of the large intron utilizes a nonconsensus 5′ donor site. We identified a U-rich and A-rich intronic sequence immediately downstream of the nonconsensus 5′ donor site that functions as an intronic splicing enhancer (ISE) required for efficient large-intron excision. The ISE binds the cellular RNA-processing proteins TIA-1 and TIAR, which enhance usage of the nonconsensus donor.


2008 ◽  
Vol 283 (15) ◽  
pp. 10058-10067 ◽  
Author(s):  
Puneet Seth ◽  
Heather B. Miller ◽  
Erika L. Lasda ◽  
James L. Pearson ◽  
Mariano A. Garcia-Blanco

Sign in / Sign up

Export Citation Format

Share Document