Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
1.
Sci Rep ; 11(1): 7696, 2021 04 08.
Article in English | MEDLINE | ID: mdl-33833316

ABSTRACT

Retinitis pigmentosa (RP) is a clinically and genetically heterogeneous group of inherited retinal degenerations. The ortholog of Drosophila eyes shut/spacemaker, EYS on chromosome 6q12 is a major genetic cause of recessive RP worldwide, with prevalence of 5 to 30%. In this study, by using targeted NGS, MLPA and Sanger sequencing we uncovered the EYS gene as one of the most common genetic cause of autosomal recessive RP in northern Sweden accounting for at least 16%. The most frequent pathogenic variant was c.8648_8655del that in some patients was identified in cis with c.1155T>A, indicating Finnish ancestry. We also showed that two novel EYS variants, c.2992_2992+6delinsTG and c.3877+1G>A caused exon skipping in human embryonic kidney cells, HEK293T and in retinal pigment epithelium cells, ARPE-19 demonstrating that in vitro minigene assay is a straightforward tool for the analysis of intronic variants. We conclude, that whenever it is possible, functional testing is of great value for classification of intronic EYS variants and the following molecular testing of family members, their genetic counselling, and inclusion of RP patients to future treatment studies.


Subject(s)
Eye Proteins/genetics , Retinitis Pigmentosa/classification , Adult , Aged , Aged, 80 and over , Alleles , Cohort Studies , Female , Founder Effect , Genes, Recessive , HEK293 Cells , Humans , Male , Middle Aged , Mutation , RNA Splicing , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/metabolism , Retinitis Pigmentosa/genetics , Sweden , Young Adult
2.
EBioMedicine ; 45: 192-207, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31262711

ABSTRACT

BACKGROUND: Tumour necrosis factor receptor associated factor 6 (TRAF6) promotes inflammation in response to various cytokines. Aberrant Wnt3a signals promotes cancer progression through accumulation of ß-Catenin. Here we investigated a potential role for TRAF6 in Wnt signaling. METHODS: TRAF6 expression was silenced by siRNA in human prostate cancer (PC3U) and human colorectal SW480 cells and by CRISPR/Cas9 in zebrafish. Several biochemical methods and analyses of mutant phenotype in zebrafish were used to analyse the function of TRAF6 in Wnt signaling. FINDINGS: Wnt3a-treatment promoted binding of TRAF6 to the Wnt co-receptors LRP5/LRP6 in PC3U and LNCaP cells in vitro. TRAF6 positively regulated mRNA expression of ß-Catenin and subsequent activation of Wnt target genes in PC3U cells. Wnt3a-induced invasion of PC3U and SW480 cells were significantly reduced when TRAF6 was silenced by siRNA. Database analysis revealed a correlation between TRAF6 mRNA and Wnt target genes in patients with prostate cancer, and high expression of LRP5, TRAF6 and c-Myc correlated with poor prognosis. By using CRISPR/Cas9 to silence TRAF6 in zebrafish, we confirm TRAF6 as a key molecule in Wnt3a signaling for expression of Wnt target genes. INTERPRETATION: We identify TRAF6 as an important component in Wnt3a signaling to promote activation of Wnt target genes, a finding important for understanding mechanisms driving prostate cancer progression. FUND: KAW 2012.0090, CAN 2017/544, Swedish Medical Research Council (2016-02513), Prostatacancerförbundet, Konung Gustaf V:s Frimurarestiftelse and Cancerforskningsfonden Norrland. The funders did not play a role in manuscript design, data collection, data analysis, interpretation nor writing of the manuscript.


Subject(s)
Inflammation/genetics , Prostatic Neoplasms/genetics , TNF Receptor-Associated Factor 6/genetics , Wnt3A Protein/genetics , Zebrafish Proteins/genetics , Animals , CRISPR-Cas Systems/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Inflammation/pathology , Low Density Lipoprotein Receptor-Related Protein-5/genetics , Male , Prostatic Neoplasms/pathology , Wnt Signaling Pathway/genetics , Zebrafish , beta Catenin/genetics
3.
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
4.
Acta Ophthalmol ; 96(7): 737-743, 2018 Nov.
Article in English | MEDLINE | ID: mdl-29461686

ABSTRACT

PURPOSE: Inherited retinal dystrophies (IRDs) represent a group of progressive conditions affecting the retina. There is a great genetic heterogeneity causing IRDs, and to date, more than 260 genes are associated with IRDs. Stargardt disease, type 1 (STGD1) or macular degeneration with flecks, STGD1 represents a disease with early onset, central visual impairment, frequent appearance of yellowish flecks and mutations in the ATP-binding cassette subfamily A, member 4 (ABCA4) gene. A large number of intronic sequence variants in ABCA4 have been considered pathogenic although their functional effect was seldom demonstrated. In this study, we aimed to reveal how intronic variants present in patients with Stargardt from the same Swedish family affect splicing. METHODS: The splicing of the ABCA4 gene was studied in human embryonic kidney cells, HEK293T, and in human retinal pigment epithelium cells, ARPE-19, using a minigene system containing variants c.4773+3A>G and c.5461-10T>C. RESULTS: We showed that both ABCA4 variants, c.4773+3A>G and c.5461-10T>C, cause aberrant splicing of the ABCA4 minigene resulting in exon skipping. We also demonstrated that splicing of ABCA4 has different outcomes depending on transfected cell type. CONCLUSION: Two intronic variants c.4773+3A>G and c.5461-10T>C, both predicted to affect splicing, are indeed disease-causing mutations due to skipping of exons 33, 34, 39 and 40 of ABCA4 gene. The experimental proof that ABCA4 mutations in STGD patients affect protein function is crucial for their inclusion to future clinical trials; therefore, functional testing of all ABCA4 intronic variants associated with Stargardt disease by minigene technology is desirable.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Introns/genetics , Macular Degeneration/congenital , Polymorphism, Single Nucleotide , RNA Splice Sites/genetics , Adult , DNA Mutational Analysis , Exons/genetics , Female , HEK293 Cells/metabolism , Humans , Macular Degeneration/diagnosis , Macular Degeneration/genetics , Male , Mutation , Pedigree , Real-Time Polymerase Chain Reaction , Retinal Pigment Epithelium/metabolism , Stargardt Disease , Transfection
5.
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
...