Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
Add more filters

Publication year range
1.
Hum Mol Genet ; 32(4): 621-631, 2023 01 27.
Article in English | MEDLINE | ID: mdl-36099027

ABSTRACT

Myotonic dystrophy type 1 is a complex disease caused by a genetically unstable CTG repeat expansion in the 3'-untranslated region of the DMPK gene. Age-dependent, tissue-specific somatic instability has confounded genotype-phenotype associations, but growing evidence suggests that it also contributes directly toward disease progression. Using a well-characterized clinical cohort of DM1 patients from Costa Rica, we quantified somatic instability in blood, buccal cells, skin and skeletal muscle. Whilst skeletal muscle showed the largest expansions, modal allele lengths in skin were also very large and frequently exceeded 2000 CTG repeats. Similarly, the degree of somatic expansion in blood, muscle and skin were associated with each other. Notably, we found that the degree of somatic expansion in skin was highly predictive of that in skeletal muscle. More importantly, we established that individuals whose repeat expanded more rapidly than expected in one tissue (after correction for progenitor allele length and age) also expanded more rapidly than expected in other tissues. We also provide evidence suggesting that individuals in whom the repeat expanded more rapidly than expected in skeletal muscle have an earlier age at onset than expected (after correction for the progenitor allele length). Pyrosequencing analyses of the genomic DNA flanking the CTG repeat revealed that the degree of methylation in muscle was well predicted by the muscle modal allele length and age, but that neither methylation of the flanking DNA nor levels of DMPK sense and anti-sense transcripts could obviously explain individual- or tissue-specific patterns of somatic instability.


Subject(s)
Myotonic Dystrophy , Humans , Myotonic Dystrophy/genetics , Trinucleotide Repeat Expansion/genetics , Mouth Mucosa , Alleles , DNA/genetics , Myotonin-Protein Kinase/genetics
2.
Hum Mol Genet ; 31(2): 262-274, 2021 12 27.
Article in English | MEDLINE | ID: mdl-34432028

ABSTRACT

Myotonic dystrophy type 1 (DM1) is a complex disease with a wide spectrum of symptoms. The exact relationship between mutant CTG repeat expansion size and clinical outcome remains unclear. DM1 congenital patients (CDM) inherit the largest expanded alleles, which are associated with abnormal and increased DNA methylation flanking the CTG repeat. However, DNA methylation at the DMPK locus remains understudied. Its relationship to DM1 clinical subtypes, expansion size and age-at-onset is not yet completely understood. Using pyrosequencing-based methylation analysis on 225 blood DNA samples from Costa Rican DM1 patients, we determined that the size of the estimated progenitor allele length (ePAL) is not only a good discriminator between CDM and non-CDM cases (with an estimated threshold at 653 CTG repeats), but also for all DM1 clinical subtypes. Secondly, increased methylation at both CTCF sites upstream and downstream of the expansion was almost exclusively present in CDM cases. Thirdly, levels of abnormal methylation were associated with clinical subtype, age and ePAL, with strong correlations between these variables. Fourthly, both ePAL and the intergenerational expansion size were significantly associated with methylation status. Finally, methylation status was associated with ePAL and maternal inheritance, with almost exclusively maternal transmission of CDM. In conclusion, increased DNA methylation at the CTCF sites flanking the DM1 expansion could be linked to ePAL, and both increased methylation and the ePAL could be considered biomarkers for the CDM phenotype.


Subject(s)
Myotonic Dystrophy , Alleles , CCCTC-Binding Factor , DNA Methylation/genetics , Humans , Myotonic Dystrophy/genetics , Myotonin-Protein Kinase/genetics , Trinucleotide Repeat Expansion/genetics
3.
Hum Mol Genet ; 29(15): 2496-2507, 2020 08 29.
Article in English | MEDLINE | ID: mdl-32601694

ABSTRACT

In myotonic dystrophy type 1 (DM1), somatic mosaicism of the (CTG)n repeat expansion is age-dependent, tissue-specific and expansion-biased. These features contribute toward variation in disease severity and confound genotype-to-phenotype analyses. To investigate how the (CTG)n repeat expansion changes over time, we collected three longitudinal blood DNA samples separated by 8-15 years and used small pool and single-molecule PCR in 43 DM1 patients. We used the lower boundary of the allele length distribution as the best estimate for the inherited progenitor allele length (ePAL), which is itself the best predictor of disease severity. Although in most patients the lower boundary of the allele length distribution was conserved over time, in many this estimate also increased with age, suggesting samples for research studies and clinical trials should be obtained as early as possible. As expected, the modal allele length increased over time, driven primarily by ePAL, age-at-sampling and the time interval. As expected, small expansions <100 repeats did not expand as rapidly as larger alleles. However, the rate of expansion of very large alleles was not obviously proportionally higher. This may, at least in part, be a result of the allele length-dependent increase in large contractions that we also observed. We also determined that individual-specific variation in the increase of modal allele length over time not accounted for by ePAL, age-at-sampling and time was inversely associated with individual-specific variation in age-at-onset not accounted for by ePAL, further highlighting somatic expansion as a therapeutic target in DM1.


Subject(s)
DNA/genetics , Mosaicism , Myotonic Dystrophy/genetics , Trinucleotide Repeats/genetics , Adolescent , Age Factors , Age of Onset , Alleles , Child , Child, Preschool , Female , Humans , Male , Myotonic Dystrophy/pathology , Phenotype , Trinucleotide Repeat Expansion
4.
Proc Natl Acad Sci U S A ; 115(9): 2198-2203, 2018 02 27.
Article in English | MEDLINE | ID: mdl-29440484

ABSTRACT

TP53 mutations occur in ∼50% of all human tumors, with increased frequency in aggressive cancers that are notoriously difficult to treat. Additionally, p53 missense mutations are remarkably predictive of refractoriness to chemo/radiotherapy in various malignancies. These observations have led to the development of mutant p53-targeting agents that restore p53 function. An important unknown is which p53-mutant tumors will respond to p53 reactivation-based therapies. Here, we found a heterogeneous impact on therapeutic response to p53 restoration, suggesting that it will unlikely be effective as a monotherapy. Through gene expression profiling of p53R172H -mutant lymphomas, we identified retinoic acid receptor gamma (RARγ) as an actionable target and demonstrated that pharmacological activation of RARγ with a synthetic retinoid sensitizes resistant p53-mutant lymphomas to p53 restoration, while additively improving outcome and survival in inherently sensitive tumors.


Subject(s)
Gene Expression Regulation, Neoplastic/drug effects , Neoplasms, Experimental/drug therapy , Retinoids/pharmacology , Tamoxifen/pharmacology , Tumor Suppressor Protein p53/metabolism , Animals , Antineoplastic Agents, Hormonal/administration & dosage , Antineoplastic Agents, Hormonal/pharmacokinetics , Antineoplastic Agents, Hormonal/pharmacology , Drug Resistance, Neoplasm , Drug Synergism , Mice , Mice, Inbred Strains , Mutation, Missense , Retinoids/administration & dosage , Retinoids/pharmacokinetics , Tamoxifen/administration & dosage , Tamoxifen/pharmacokinetics , Transcriptome , Tumor Necrosis Factor-alpha/metabolism , Tumor Suppressor Protein p53/genetics
5.
BMC Genomics ; 18(1): 740, 2017 Sep 19.
Article in English | MEDLINE | ID: mdl-28927378

ABSTRACT

BACKGROUND: Nearly 6 million deaths and over a half trillion dollars in healthcare costs worldwide are attributed to tobacco smoking each year. Extensive research efforts have been pursued to elucidate the molecular underpinnings of smoking addiction and facilitate cessation. In this study, we genotyped and obtained both resting state and task-based functional magnetic resonance imaging from 64 non-smokers and 42 smokers. Smokers were imaged after having smoked normally ("sated") and after having not smoked for at least 12 h ("abstinent"). RESULTS: While abstinent smokers did not differ from non-smokers with respect to pairwise resting state functional connectivities (RSFCs) between 12 brain regions of interest, RSFCs involving the caudate and putamen of sated smokers significantly differed from those of non-smokers (P < 0.01). Further analyses of caudate and putamen activity during elicited experiences of reward and disappointment show that caudate activity during reward (CR) correlated with smoking status (P = 0.015). Moreover, abstinent smokers with lower CR experienced greater withdrawal symptoms (P = 0.024), which suggests CR may be related to smoking urges. Associations between genetic variants and CR, adjusted for smoking status, were identified by genome-wide association study (GWAS). Genes containing or exhibiting caudate-specific expression regulation by these variants were enriched within Gene Ontology terms that describe cytoskeleton functions, synaptic organization, and injury response (P < 0.001, FDR < 0.05). CONCLUSIONS: By integrating genomic and imaging data, novel insights into potential mechanisms of caudate activation and homeostasis are revealed that may guide new directions of research toward improving our understanding of addiction pathology.


Subject(s)
Behavior, Addictive/diagnostic imaging , Caudate Nucleus/pathology , Genome-Wide Association Study , Homeostasis , Magnetic Resonance Imaging , Neuroglia/metabolism , Smoking/genetics , Adult , Behavior, Addictive/genetics , Behavior, Addictive/metabolism , Behavior, Addictive/pathology , Emotions , Female , Humans , Male , Reward , Signal Transduction , Smoking/metabolism , Smoking/psychology
6.
FASEB J ; 29(6): 2555-65, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25746793

ABSTRACT

Previous studies showed that nucleolar protein 66 (NO66), the Jumonji C-domain-containing histone demethylase for methylated histone H3K4 and H3K36 (H3K36me), negatively regulates osteoblast differentiation in vitro by inhibiting the activity of transcription factor osterix (Osx). However, whether NO66 affects mammalian skeletogenesis in vivo is not yet known. Here, we generated transgenic (TG) mice overexpressing a flag-tagged NO66 transgene driven by the Prx1 (paired related homeobox 1) promoter. We found that NO66 overexpression in Prx1-expressing mesenchymal cells inhibited skeletal growth and bone formation. The inhibitory phenotype was associated with >50% decreases in chondrocyte/osteoblast proliferation and differentiation. Moreover, we found that in bones of NO66-TG mice, expression of Igf1, Igf1 receptor (Igf1r), runt-related transcription factor 2, and Osx was significantly down-regulated (P < 0.05). Consistent with these results, we observed >50% reduction in levels of phosphorylated protein kinase B (Akt) and H3K36me3 in bones of NO66-TG mice, suggesting an inverse correlation between NO66 histone demethylase and the activity of IGF1R/Akt signaling. This correlation was further confirmed by in vitro assays of C2C12 cells with NO66 overexpression. We propose that the decrease in the IGF1R/Akt signaling pathway in mice with mesenchymal overexpression of NO66 may contribute in part to the inhibition of skeletal growth and bone formation.


Subject(s)
Bone and Bones/metabolism , Gene Expression Regulation, Developmental , Jumonji Domain-Containing Histone Demethylases/genetics , Mesoderm/metabolism , Skull/metabolism , Animals , Blotting, Western , Bone and Bones/cytology , Bone and Bones/embryology , Cell Differentiation/genetics , Cell Line , Cell Proliferation/genetics , Chondrocytes/cytology , Chondrocytes/metabolism , Immunohistochemistry , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Jumonji Domain-Containing Histone Demethylases/metabolism , Mesoderm/cytology , Mesoderm/embryology , Mice, Transgenic , Microscopy, Fluorescence , Osteoblasts/cytology , Osteoblasts/metabolism , Osteogenesis/genetics , Proto-Oncogene Proteins c-akt/metabolism , Receptor, IGF Type 1/genetics , Receptor, IGF Type 1/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics , Skull/embryology , Sp7 Transcription Factor , Transcription Factors/genetics , Transcription Factors/metabolism
7.
Am J Pathol ; 184(8): 2322-32, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24907641

ABSTRACT

Myotonic dystrophy type 2 (DM2) is a multisystemic disorder caused by a (CCTG)n repeat expansion in intron 1 of CNBP. Transcription of the repeats causes a toxic RNA gain of function involving their accumulation in ribonuclear foci. This leads to sequestration of splicing factors and alters pre-mRNA splicing in a range of downstream effector genes, which is thought to contribute to the diverse DM2 clinical features. Hyperlipidemia is frequent in DM2 patients, but the treatment is problematic because of an increased risk of statin-induced adverse reactions. Hypothesizing that shared pathways lead to the increased risk, we compared the skeletal muscle expression profiles of DM2 patients and controls with patients with hyperlipidemia on statin therapy. Neural precursor cell expressed, developmentally downregulated-4 (NEDD4), an ubiquitin ligase, was one of the dysregulated genes identified in DM2 patients and patients with statin-treated hyperlipidemia. In DM2 muscle, NEDD4 mRNA was abnormally spliced, leading to aberrant NEDD4 proteins. NEDD4 was down-regulated in persons taking statins, and simvastatin treatment of C2C12 cells suppressed NEDD4 transcription. Phosphatase and tensin homologue (PTEN), an established NEDD4 target, was increased and accumulated in highly atrophic DM2 muscle fibers. PTEN ubiquitination was reduced in DM2 myofibers, suggesting that the NEDD4-PTEN pathway is dysregulated in DM2 skeletal muscle. Thus, this pathway may contribute to the increased risk of statin-adverse reactions in patients with DM2.


Subject(s)
Endosomal Sorting Complexes Required for Transport/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/adverse effects , Muscle, Skeletal/metabolism , Myotonic Dystrophy/metabolism , Ubiquitin-Protein Ligases/metabolism , Adult , Blotting, Western , Endosomal Sorting Complexes Required for Transport/genetics , Female , Fluorescent Antibody Technique , Genotype , Humans , Hyperlipidemias/drug therapy , Hyperlipidemias/etiology , Immunohistochemistry , Liver-Specific Organic Anion Transporter 1 , Male , Middle Aged , Myotonic Dystrophy/complications , Myotonic Dystrophy/genetics , Nedd4 Ubiquitin Protein Ligases , Oligonucleotide Array Sequence Analysis , Organic Anion Transporters/genetics , PTEN Phosphohydrolase/metabolism , RNA Splicing , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/physiology , Transcriptome , Ubiquitin-Protein Ligases/genetics
8.
J Osteoporos ; 2023: 8998556, 2023.
Article in English | MEDLINE | ID: mdl-36660551

ABSTRACT

In previous study, we showed that nucleolar protein 66 (NO66) is a chromatin modifier and negatively regulates Osterix activity as well as mesenchymal progenitor differentiation. Genetic ablation of the NO66 (RIOX1) gene in cells of the Prx1-expressing mesenchymal lineage leads to acceleration of osteochondrogenic differentiation and a larger skeleton in adult mice, whereas mesenchyme-specific overexpression of NO66 inhibits osteochondrogenesis resulting in dwarfism and osteopenia. However, the impact of NO66 overexpression in cells of the osteoblast lineage in vivo remains largely undefined. Here, we generated osteoblast-specific transgenic mice overexpressing a FLAG-tagged NO66 transgene driven by the 2.3 kB alpha-1type I collagen (Col1a1) promoter. We found that overexpression of NO66 in cells of the osteoblast lineage did not cause overt defects in developmental bones but led to osteoporosis in the long bones of adult mice. This includes decreased bone volume (BV), bone volume density (bone volume/total volume, BV/TV), and bone mineral density (BMD) in cancellous compartment of long bones, along with the accumulation of fatty droplets in bone marrow. Ex vivo culture of the bone marrow mesenchymal stem/stromal cells (BMSCs) from adult Col1a1-NO66 transgenic mice showed an increase in adipogenesis and a decrease in osteogenesis. Taken together, these data demonstrate a crucial role for NO66 in adult bone formation and homeostasis. Our Col1a1-NO66 transgenic mice provide a novel animal model for the mechanistic and therapeutic study of NO66 in osteoporosis.

9.
Mol Carcinog ; 51(11): 895-906, 2012 Nov.
Article in English | MEDLINE | ID: mdl-21976412

ABSTRACT

Aberrant RNA splicing is thought to play a key role in tumorigenesis. The assessment of its specific contributions is limited by the complexity of information derived from genome-wide array-based approaches. We describe how performing splicing factor-specific comparisons using both tumor and cell line data sets may more readily identify physiologically relevant tumor-specific splicing events. Affymetrix exon array data derived from glioblastoma (GBM) tumor samples with defined polypyrimidine tract-binding protein 1 (PTBP1) levels were compared with data from U251 GBM cells with and without PTBP1 knockdown. This comparison yielded overlapping gene sets that comprised only a minor fraction of each data set. The identification of a novel GBM-specific splicing event involving the USP5 gene led us to further examine its role in tumorigenesis. In GBM, USP5 generates a shorter isoform 2 through recognition of a 5' splice site within exon 15. Production of the USP5 isoform 2 was strongly correlated with PTBP1 expression in GBM tumor samples and cell lines. Splicing regulation was consistent with the presence of an intronic PTBP1 binding site and could be modulated through antisense targeting of the isoform 2 splice site to force expression of isoform 1 in GBM cells. The forced expression of USP5 isoform 1 in two GBM cell lines inhibited cell growth and migration, implying an important role for USP5 splicing in gliomagenesis. These results support a role for aberrant RNA splicing in tumorigenesis and suggest that changes in relatively few genes may be sufficient to drive the process.


Subject(s)
Alternative Splicing , Endopeptidases/genetics , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Polypyrimidine Tract-Binding Protein/metabolism , Base Sequence , Binding Sites , Cell Line, Tumor , Cell Movement , Cell Proliferation , Exons , Gene Knockdown Techniques , Glioblastoma/metabolism , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Humans , Molecular Sequence Data , Polypyrimidine Tract-Binding Protein/genetics , Protein Isoforms/genetics
10.
Proc Natl Acad Sci U S A ; 106(9): 3270-5, 2009 Mar 03.
Article in English | MEDLINE | ID: mdl-19218442

ABSTRACT

All DNA repeats known to undergo expansion leading to human neurodegenerative disease can form one, or several, alternative conformations, including hairpin, slipped strand, triplex, quadruplex, or unwound DNA structures. These alternative structures may interfere with the normal cellular processes of transcription, DNA repair, replication initiation, or polymerase elongation and thereby contribute to the genetic instability of these repeat tracts. We show that (CCTG) x (CAGG) repeats, in the first intron of the ZNF9 gene associated with myotonic dystrophy type 2, form slipped-strand DNA structures in a length-dependent fashion upon reduplexing. The threshold for structure formation on reduplexing is between 36 and 42 repeats in length. Alternative DNA structures also form in (CCTG)(58) x (CAGG)(58) and larger repeat tracts in plasmids at physiological superhelical densities. This represents an example of a sequence that forms slipped-strand DNA from the energy of DNA supercoiling. Moreover, Z-DNA forms in a (TG) x (CA) tract within the complex repeat sequence 5' of the (CCTG)(n) x (CAGG)(n) repeat in the ZNF9 gene. Upon reduplexing, the presence of the flanking sequence containing the Z-DNA-forming tract reduced the extent of slipped-strand DNA formation by 62% for (CCTG)(57) x (CAGG)(57) compared with 58 pure repeats without the flanking sequence. This finding suggests that the Z-DNA-forming sequence in the DM2 gene locus may have a protective effect of reducing the potential for slipped-strand DNA formation in (CCTG)(n) x (CAGG)(n) repeats.


Subject(s)
DNA, Z-Form/genetics , Myotonic Dystrophy/genetics , 5' Flanking Region , Base Sequence , Humans , Molecular Sequence Data , Myotonic Dystrophy/classification , Sequence Alignment
11.
Hum Genet ; 130(6): 789-94, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21688173

ABSTRACT

Germline mutations in the tumor suppressor gene TP53 occur in the majority of families with Li-Fraumeni syndrome, who are at an increased risk for a wide spectrum of early onset cancers. Several genetic polymorphisms in TP53 modify its effect on cancer risk. While some studies indicate that the TP53 PIN3 deletion allele (D) accelerate tumor onset in carriers with TP53 germline mutations, other studies have shown that the TP53 PIN3 insertion allele (I) confers a significantly higher risk of developing cancer than D allele. To further determine the effects of the TP53 PIN3 polymorphism on cancer development among TP53 germline mutations and to evaluate if those are differenence between male and female carriers, we studied a total of 152 germline mutation carriers with available DNA samples that can be used for genotyping. Our results indicate that the TP53 PIN3 polymorphism has a sex-specific effect on the risk of cancer in TP53 mutation carriers, conferring cancer risk in men (P = 0.0041) but not women with DI or II genotypes.


Subject(s)
Germ-Line Mutation , Li-Fraumeni Syndrome/genetics , Neoplasms/genetics , Tumor Suppressor Protein p53/genetics , Adult , Age Factors , Age of Onset , Alleles , Cohort Studies , Female , Genetic Predisposition to Disease , Genotype , Haplotypes , Heterozygote , Humans , Male , Middle Aged , Multivariate Analysis , Polymorphism, Genetic , Risk Factors , Sex Factors , Young Adult
12.
Hum Genet ; 129(6): 663-73, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21305319

ABSTRACT

Li-Fraumeni syndrome (LFS) is a rare familial cancer syndrome characterized by early cancer onset, diverse tumor types, and multiple primary tumors. Germ-line TP53 mutations have been identified in most LFS families. A high-frequency single-nucleotide polymorphism, SNP309 (rs2279744), in MDM2 was recently confirmed to be a modifier of cancer risk in several case-series studies: substantially earlier cancer onset was observed in SNP309 G-allele carriers than in wild-type individuals by 7-16 years. However, cancer risk analyses that jointly account for measured hereditary TP53 mutations and MDM2 SNP309 have not been systematically investigated in familial cases. Here, we determined the combined effects of measured TP53 mutations, MDM2 SNP309, and gender and their interactions simultaneously in LFS families. We used the method that is designed for extended pedigrees and structured for age-specific risk models based on Cox proportional hazards regression. We analyzed the cancer incidence in 19 extended pedigrees with germ-line TP53 mutations ascertained through the clinical LFS phenotype. The dataset consisted of 463 individuals with 129 TP53 mutation carriers. Our analyses showed that the TP53 germ-line mutation and its interaction with gender were strongly associated with familial cancer incidence and that the association between MDM2 SNP309 and increased cancer risk was modest. In contrast with several case-series studies, the interaction between MDM2 SNP309 and TP53 mutation was not statistically significant in our LFS family cohort. Our results showed that SNP309 G-alleles were associated with accelerated tumor formation in both carriers and non-carriers of germ-line TP53 mutations.


Subject(s)
Genetic Predisposition to Disease , Germ-Line Mutation , Li-Fraumeni Syndrome/genetics , Polymorphism, Single Nucleotide , Proto-Oncogene Proteins c-mdm2/genetics , Tumor Suppressor Protein p53/genetics , Adolescent , Adult , Aged , Child , Child, Preschool , Female , Humans , Infant , Infant, Newborn , Male , Middle Aged , Pedigree
13.
Am J Pathol ; 177(6): 3025-36, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20971734

ABSTRACT

The mutation that underlies myotonic dystrophy type 2 (DM2) is a (CCTG)n expansion in intron 1 of zinc finger protein 9 (ZNF9). It has been suggested that ZNF9 is of no consequence for disease pathogenesis. We determined the expression levels of ZNF9 during muscle cell differentiation and in DM2 muscle by microarray profiling, real-time RT-PCR, splice variant analysis, immunofluorescence, and Western blotting. Our results show that in differentiating myoblasts, ZNF9 protein was localized primarily to the nucleus, whereas in mature muscle fibers, it was cytoplasmic and organized in sarcomeric striations at the Z-disk. In patients with DM2, ZNF9 was abnormally expressed. First, there was an overall reduction in both the mRNA and protein levels. Second, the subcellular localization of the ZNF9 protein was somewhat less cytoplasmic and more membrane-bound. Third, our splice variant analysis revealed retention of intron 3 in an aberrant isoform, and fourth quantitative allele-specific expression analysis showed the persistence of intron 1 sequences from the abnormal allele, further suggesting that the mutant allele is incompletely spliced. Thus, the decrease in total expression appears to be due to impaired splicing of the mutant transcript. Our data indicate that ZNF9 expression in DM2 patients is altered at multiple levels. Although toxic RNA effects likely explain overlapping phenotypic manifestations between DM1 and DM2, abnormal ZNF9 levels in DM2 may account for the differences in DM1.


Subject(s)
DNA Repeat Expansion/physiology , RNA-Binding Proteins/genetics , Adult , Aged , Female , Gene Expression Profiling , Gene Expression Regulation/genetics , Humans , Male , Microarray Analysis , Middle Aged , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Mutation/physiology , Myotonic Disorders/genetics , Myotonic Disorders/metabolism , Myotonic Disorders/pathology , Myotonic Dystrophy , RNA-Binding Proteins/metabolism , Tissue Distribution/genetics , Young Adult
14.
Am J Pathol ; 175(2): 748-62, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19590039

ABSTRACT

Myotonic dystrophy 2 (DM2) is a multisystem skeletal muscle disease caused by an expansion of tetranucleotide CCTG repeats, the transcription of which results in the accumulation of untranslated CCUG RNA. In this study, we report that CCUG repeats both bind to and misregulate the biological functions of cytoplasmic multiprotein complexes. Two CCUG-interacting complexes were subsequently purified and analyzed. A major component of one of the complexes was found to be the 20S catalytic core complex of the proteasome. The second complex was found to contain CUG triplet repeat RNA-binding protein 1 (CUGBP1) and the translation initiation factor eIF2. Consistent with the biological functions of the 20S proteasome and the CUGBP1-eIF2 complexes, the stability of short-lived proteins and the levels of the translational targets of CUGBP1 were shown to be elevated in DM2 myoblasts. We found that the overexpression of CCUG repeats in human myoblasts from unaffected patients, in C2C12 myoblasts, and in a DM2 mouse model alters protein translation and degradation, similar to the alterations observed in DM2 patients. Taken together, these findings show that RNA CCUG repeats misregulate protein turnover on both the levels of translation and proteasome-mediated protein degradation.


Subject(s)
Microsatellite Repeats , Myotonic Dystrophy/metabolism , Protein Biosynthesis , Proteins/metabolism , Untranslated Regions/metabolism , Animals , Base Sequence , Cell Line , Cell Nucleus/metabolism , Cytoplasm/metabolism , Eukaryotic Initiation Factor-2/metabolism , Humans , Mice , Myoblasts/metabolism , Myotonic Dystrophy/genetics , Proteasome Endopeptidase Complex/metabolism , Untranslated Regions/genetics
15.
Acta Neuropathol ; 119(4): 465-79, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20066428

ABSTRACT

Aberrant transcription and mRNA processing of multiple genes due to RNA-mediated toxic gain-of-function has been suggested to cause the complex phenotype in myotonic dystrophies type 1 and 2 (DM1 and DM2). However, the molecular basis of muscle weakness and wasting and the different pattern of muscle involvement in DM1 and DM2 are not well understood. We have analyzed the mRNA expression of genes encoding muscle-specific proteins and transcription factors by microarray profiling and studied selected genes for abnormal splicing. A subset of the abnormally regulated genes was further analyzed at the protein level. TNNT3 and LDB3 showed abnormal splicing with significant differences in proportions between DM2 and DM1. The differential abnormal splicing patterns for TNNT3 and LDB3 appeared more pronounced in DM2 relative to DM1 and are among the first molecular differences reported between the two diseases. In addition to these specific differences, the majority of the analyzed genes showed an overall increased expression at the mRNA level. In particular, there was a more global abnormality of all different myosin isoforms in both DM1 and DM2 with increased transcript levels and a differential pattern of protein expression. Atrophic fibers in DM2 patients expressed only the fast myosin isoform, while in DM1 patients they co-expressed fast and slow isoforms. However, there was no increase of total myosin protein levels, suggesting that aberrant protein translation and/or turnover may also be involved.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Muscle Fibers, Skeletal/metabolism , Muscular Atrophy/genetics , Myosins/genetics , Myotonic Dystrophy/genetics , Troponin T/genetics , Adaptor Proteins, Signal Transducing/metabolism , Alternative Splicing , Female , Gene Expression Regulation , Humans , LIM Domain Proteins , Male , Muscle Fibers, Skeletal/pathology , Muscular Atrophy/metabolism , Muscular Atrophy/pathology , Myosins/metabolism , Myotonic Dystrophy/metabolism , Myotonic Dystrophy/pathology , Oligonucleotide Array Sequence Analysis , Protein Isoforms/genetics , Protein Isoforms/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Troponin T/metabolism
16.
Muscle Nerve ; 42(6): 856-63, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21104860

ABSTRACT

Because of their central role in muscle development and maintenance, MEF2 family members represent excellent candidate effectors of the muscle pathology in myotonic dystrophy (DM). We investigated the expression and alternative splicing of all four MEF2 genes in muscle from neuromuscular disorder (NMD) patients, including DM1 and DM2. We observed MEF2A and MEF2C overexpression in all NMD muscle, including 12 MEF2-interacting genes. Exon 4 and 5 usage in MEF2A and MEF2C was different between DM and normal muscle, with DM showing the embryonic isoform. Similar splicing differences were observed in other NMD muscle. For MEF2C, missplicing was more pronounced in DM than in other dystrophies. Our data confirm dysregulation of MEF2A and MEF2C expression and splicing in several NMD, including DM. Our findings demonstrate that aberrant splicing in NMD is independent from expression of mutant repeats, and suggests that some aberrant splicing, even in DM, may be compensatory rather than primary.


Subject(s)
MADS Domain Proteins/genetics , Muscle, Skeletal/metabolism , Muscular Dystrophies/genetics , Myogenic Regulatory Factors/genetics , Myotonia Congenita/genetics , Gene Expression , Humans , MADS Domain Proteins/metabolism , MEF2 Transcription Factors , Muscle, Skeletal/pathology , Muscular Dystrophies/metabolism , Muscular Dystrophies/pathology , Myogenic Regulatory Factors/metabolism , Myotonia Congenita/metabolism , Myotonia Congenita/pathology , Oligonucleotide Array Sequence Analysis , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA Splicing , Reverse Transcriptase Polymerase Chain Reaction , Tissue Array Analysis
17.
Brain ; 132(Pt 8): 2277-88, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19506066

ABSTRACT

Polypyrimidine tract-binding protein 1 (PTBP1) is a multi-functional RNA-binding protein that is aberrantly overexpressed in glioma. PTBP1 and its brain-specific homologue polypyrimidine tract-binding protein 2 (PTBP2) regulate neural precursor cell differentiation. However, the overlapping and non-overlapping target transcripts involved in this process are still unclear. To determine why PTBP1 and not PTBP2 would promote glial cell-derived tumours, both PTBP1 and PTBP2 were knocked down in the human glioma cell lines U251 and LN229 to determine the role of these proteins in cell proliferation, migration, and adhesion. Surprisingly, removal of both PTBP1 and PTBP2 slowed cell proliferation, with the double knockdown having no additive effects. Decreased expression of both proteins individually and in combination inhibited cell migration and increased adhesion of cells to fibronectin and vitronectin. A global survey of differential exon expression was performed following PTBP1 knockdown in U251 cells using the Affymetrix Exon Array to identify PTBP1-specific splicing targets that enhance gliomagenesis. In the PTBP1 knockdown, previously determined targets were unaltered in their splicing patterns. A single gene, RTN4 (Nogo) had significantly enhanced inclusion of exon 3 when PTBP1 was removed. Overexpression of the splice isoform containing exon 3 decreased cell proliferation to a similar degree as the removal of PTBP1. These results provide the first evidence that RNA-binding proteins affect the invasive and rapid growth characteristics of glioma cell lines. Its actions on proliferation appear to be mediated, in part, through alternative splicing of RTN4.


Subject(s)
Glioma/pathology , Heterogeneous-Nuclear Ribonucleoproteins/physiology , Neoplasm Proteins/physiology , Nerve Tissue Proteins/physiology , Polypyrimidine Tract-Binding Protein/physiology , Actin Cytoskeleton/pathology , Alternative Splicing/physiology , Cell Adhesion/physiology , Cell Movement/physiology , Cell Proliferation , Exons/genetics , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques/methods , Glioma/genetics , Glioma/metabolism , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Humans , Myelin Proteins/biosynthesis , Myelin Proteins/genetics , Nerve Tissue Proteins/genetics , Nogo Proteins , Polypyrimidine Tract-Binding Protein/genetics , Tumor Cells, Cultured
18.
PLoS One ; 15(4): e0231000, 2020.
Article in English | MEDLINE | ID: mdl-32287265

ABSTRACT

Myotonic dystrophy type 1 (DM1) is a rare genetic disorder, characterised by muscular dystrophy, myotonia, and other symptoms. DM1 is caused by the expansion of a CTG repeat in the 3'-untranslated region of DMPK. Longer CTG expansions are associated with greater symptom severity and earlier age at onset. The primary mechanism of pathogenesis is thought to be mediated by a gain of function of the CUG-containing RNA, that leads to trans-dysregulation of RNA metabolism of many other genes. Specifically, the alternative splicing (AS) and alternative polyadenylation (APA) of many genes is known to be disrupted. In the context of clinical trials of emerging DM1 treatments, it is important to be able to objectively quantify treatment efficacy at the level of molecular biomarkers. We show how previously described candidate mRNA biomarkers can be used to model an effective reduction in CTG length, using modern high-dimensional statistics (machine learning), and a blood and muscle mRNA microarray dataset. We show how this model could be used to detect treatment effects in the context of a clinical trial.


Subject(s)
Myotonic Dystrophy/genetics , Myotonic Dystrophy/therapy , RNA, Messenger/genetics , Alternative Splicing , Biostatistics , Clinical Trials as Topic/methods , Clinical Trials as Topic/statistics & numerical data , Databases, Nucleic Acid/statistics & numerical data , Genetic Markers , Humans , Least-Squares Analysis , Machine Learning , Models, Genetic , Muscles/metabolism , Myotonic Dystrophy/metabolism , Myotonin-Protein Kinase/genetics , Oligonucleotide Array Sequence Analysis/statistics & numerical data , Polyadenylation , RNA, Messenger/metabolism , Treatment Outcome , Trinucleotide Repeat Expansion
19.
PLoS Med ; 6(5): e1000068, 2009 May 26.
Article in English | MEDLINE | ID: mdl-19536326

ABSTRACT

BACKGROUND: Breast cancer in young women tends to have a natural history of aggressive disease for which rates of recurrence are higher than in breast cancers detected later in life. Little is known about the genetic pathways that underlie early-onset breast cancer. Here we report the discovery of DEAR1 (ductal epithelium-associated RING Chromosome 1), a novel gene encoding a member of the TRIM (tripartite motif) subfamily of RING finger proteins, and provide evidence for its role as a dominant regulator of acinar morphogenesis in the mammary gland and as an independent predictor of local recurrence-free survival in early-onset breast cancer. METHODS AND FINDINGS: Suppression subtractive hybridization identified DEAR1 as a novel gene mapping to a region of high-frequency loss of heterozygosity (LOH) in a number of histologically diverse human cancers within Chromosome 1p35.1. In the breast epithelium, DEAR1 expression is limited to the ductal and glandular epithelium and is down-regulated in transition to ductal carcinoma in situ (DCIS), an early histologic stage in breast tumorigenesis. DEAR1 missense mutations and homozygous deletion (HD) were discovered in breast cancer cell lines and tumor samples. Introduction of the DEAR1 wild type and not the missense mutant alleles to complement a mutation in a breast cancer cell line, derived from a 36-year-old female with invasive breast cancer, initiated acinar morphogenesis in three-dimensional (3D) basement membrane culture and restored tissue architecture reminiscent of normal acinar structures in the mammary gland in vivo. Stable knockdown of DEAR1 in immortalized human mammary epithelial cells (HMECs) recapitulated the growth in 3D culture of breast cancer cell lines containing mutated DEAR1, in that shDEAR1 clones demonstrated disruption of tissue architecture, loss of apical basal polarity, diffuse apoptosis, and failure of lumen formation. Furthermore, immunohistochemical staining of a tissue microarray from a cohort of 123 young female breast cancer patients with a 20-year follow-up indicated that in early-onset breast cancer, DEAR1 expression serves as an independent predictor of local recurrence-free survival and correlates significantly with strong family history of breast cancer and the triple-negative phenotype (ER(-), PR(-), HER-2(-)) of breast cancers with poor prognosis. CONCLUSIONS: Our data provide compelling evidence for the genetic alteration and loss of expression of DEAR1 in breast cancer, for the functional role of DEAR1 in the dominant regulation of acinar morphogenesis in 3D culture, and for the potential utility of an immunohistochemical assay for DEAR1 expression as an independent prognostic marker for stratification of early-onset disease.


Subject(s)
Biomarkers, Tumor/biosynthesis , Breast Neoplasms/pathology , Breast/growth & development , Neoplasm Recurrence, Local/diagnosis , Tumor Suppressor Proteins/biosynthesis , Biomarkers, Tumor/genetics , Breast/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/mortality , Cell Line, Tumor , Disease-Free Survival , Down-Regulation/genetics , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Gene Knockdown Techniques , Humans , Morphogenesis/genetics , Mutation/genetics , Neoplasm Recurrence, Local/genetics , Prognosis , Tripartite Motif Proteins , Tumor Suppressor Proteins/genetics , Ubiquitin-Protein Ligases
20.
PLoS One ; 14(5): e0216407, 2019.
Article in English | MEDLINE | ID: mdl-31048891

ABSTRACT

Genotype-to-phenotype correlation studies in myotonic dystrophy type 1 (DM1) have been confounded by the age-dependent, tissue-specific and expansion-biased features of somatic mosaicism of the expanded CTG repeat. Previously, we showed that by controlling for the confounding effects of somatic instability to estimate the progenitor allele CTG length in blood DNA, age at onset correlations could be significantly improved. To determine the suitability of saliva DNA as a source for genotyping, we used small pool-PCR to perform a detailed quantitative study of the somatic mutational dynamics of the CTG repeat in saliva and blood DNA from 40 DM1 patients. Notably, the modal allele length in saliva was only moderately higher in saliva and not as large as previously observed in most other tissues. The lower boundary of the allele distribution was also slightly higher in saliva than it was in blood DNA. However, the progenitor allele length estimated in blood explained more of the variation in age at onset than that estimated from saliva. Interestingly, although the modal allele length was slightly higher in saliva, the overall degree of somatic variation was typically lower than in blood DNA, revealing new insights into the tissue-specific dynamics of somatic mosaicism. These data indicate that saliva constitutes an accessible, non-invasive and suitable DNA sample source for performing genetic studies in DM1.


Subject(s)
DNA/genetics , Genetic Loci , Myotonic Dystrophy , Saliva , Trinucleotide Repeat Expansion , Trinucleotide Repeats , Age of Onset , DNA Mutational Analysis , Female , Humans , Male , Myotonic Dystrophy/diagnosis , Myotonic Dystrophy/genetics , Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL