Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 2 de 2
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Genet Genomic Med ; 6(6): 887-897, 2018 11.
Article in English | MEDLINE | ID: mdl-30209894

ABSTRACT

BACKGROUND: Hereditary myopathy with lactic acidosis (HML) is an autosomal recessive disease caused by an intron mutation in the iron-sulfur cluster assembly (ISCU) gene. The mutation results in aberrant splicing, where part of the intron is retained in the final mRNA transcript, giving rise to a truncated nonfunctional ISCU protein. Using an ISCU mini-gene system, we have previously shown that PTBP1 can act as a repressor of the mis-splicing of ISCU, where overexpression of PTBP1 resulted in a decrease of the incorrect splicing. In this study, we wanted to, in more detail, analyze the role of PTBP1 in the regulation of endogenous ISCU mis-splicing. METHODS: Overexpression and knockdown of PTBP1 was performed in myoblasts from two HML patients and a healthy control. Quantification of ISCU mis-splicing was done by qRTPCR. Biotinylated ISCU RNA, representing wildtype and mutant intron sequence, was used in a pull-down assay with nuclear extracts from myoblasts. Levels of PTBP1 in human cell lines and mice tissues were analyzed by qRTPCR and western blot. RESULTS: PTBP1 overexpression in HML patient myoblasts resulted in a substantial decrease of ISCU mis-splicing while knockdown of PTBP1 resulted in a drastic increase. The effect could be observed in both patient and control myoblasts. We could also show that PTBP1 interacts with both the mutant and wild-type ISCU intron sequence, but with a higher affinity to the mutant sequence. Furthermore, low levels of PTBP1 among examined mouse tissues correlated with high levels of incorrect splicing of ISCU. CONCLUSION: Our results show that PTBP1 acts as a dominant repressor of ISCU mis-splicing. We also show an inverse correlation between the levels of PTBP1 and ISCU mis-splicing, suggesting that the high level of mis-splicing in the skeletal muscle is primarily due to the low levels of PTBP1.


Subject(s)
Acidosis, Lactic/congenital , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Iron-Sulfur Proteins/genetics , Muscular Diseases/congenital , Polypyrimidine Tract-Binding Protein/genetics , RNA Splicing , Acidosis, Lactic/genetics , Animals , Cells, Cultured , Genes, Dominant , HEK293 Cells , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Humans , Iron-Sulfur Proteins/metabolism , Mice , Mice, Inbred CBA , Muscular Diseases/genetics , Myoblasts/metabolism , Polypyrimidine Tract-Binding Protein/metabolism , Suppression, Genetic
2.
PLoS One ; 11(10): e0165453, 2016.
Article in English | MEDLINE | ID: mdl-27783661

ABSTRACT

Hereditary myopathy with lactic acidosis (HML) is an autosomal recessive disease caused by an intronic one-base mutation in the iron-sulfur cluster assembly (ISCU) gene, resulting in aberrant splicing. The incorrectly spliced transcripts contain a 100 or 86 bp intron sequence encoding a non-functional ISCU protein, which leads to defects in several Fe-S containing proteins in the respiratory chain and the TCA cycle. The symptoms in HML are restricted to skeletal muscle, and it has been proposed that this effect is due to higher levels of incorrectly spliced ISCU in skeletal muscle compared with other energy-demanding tissues. In this study, we confirm that skeletal muscle contains the highest levels of incorrect ISCU splice variants compared with heart, brain, liver and kidney using a transgenic mouse model expressing human HML mutated ISCU. We also show that incorrect splicing occurs to a significantly higher extent in the slow-twitch soleus muscle compared with the gastrocnemius and quadriceps. The splicing factor serine/arginine-rich splicing factor 3 (SRSF3) was identified as a potential candidate for the slow fiber specific regulation of ISCU splicing since this factor was expressed at higher levels in the soleus compared to the gastrocnemius and quadriceps. We identified an interaction between SRSF3 and the ISCU transcript, and by overexpressing SRSF3 in human myoblasts we observed increased levels of incorrectly spliced ISCU, while knockdown of SRSF3 resulted in decreased levels. We therefore suggest that SRSF3 may participate in the regulation of the incorrect splicing of mutant ISCU and may, at least partially, explain the muscle-specific symptoms of HML.


Subject(s)
Acidosis, Lactic/congenital , Iron-Sulfur Proteins/genetics , Muscle, Skeletal/metabolism , Muscular Diseases/congenital , RNA Splicing , Serine-Arginine Splicing Factors/genetics , Acidosis, Lactic/genetics , Acidosis, Lactic/pathology , Animals , Cells, Cultured , Humans , Iron-Sulfur Proteins/metabolism , Mice , Mice, Inbred CBA , Mice, Transgenic , Muscular Diseases/genetics , Muscular Diseases/pathology , Mutagenesis, Site-Directed , Myoblasts/cytology , Myoblasts/metabolism , RNA/isolation & purification , RNA/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Real-Time Polymerase Chain Reaction , Serine-Arginine Splicing Factors/antagonists & inhibitors , Serine-Arginine Splicing Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...