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

2.
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
3.
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
4.
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
5.
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
6.
Neurol Clin Pract ; 9(4): 343-353, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31583190

ABSTRACT

PURPOSE OF REVIEW: Myotonic dystrophy type 2 (DM2) is a rare, progressive multisystem disease particularly affecting the skeletal muscle. A causal therapy is not yet available; however, prompt, appropriate symptomatic treatments are essential to limit disease-related complications. Evidence-based guidelines to assist medical practitioners in the care of DM2 patients do not exist. RECENT FINDINGS: The Myotonic Dystrophy Foundation (MDF) previously worked with an international group of 66 clinicians to develop consensus-based care recommendations for myotonic dystrophy type 1. Following a similar approach, the MDF recruited 15 international clinicians with long-standing experience in the care of DM2 patients to develop consensus-based care recommendations. The single text procedure was adopted. This process generated a 4-page Quick Reference Guide and a comprehensive 55-page document that provides care recommendations for DM2 patients. SUMMARY: The resulting recommendations will help standardize and improve care for DM2 patients and facilitate appropriate management in centers without neuromuscular specialists.

7.
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
8.
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
9.
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
10.
Dis Model Mech ; 10(8): 993-1003, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28623239

ABSTRACT

The myotonic dystrophies are prototypic toxic RNA gain-of-function diseases. Myotonic dystrophy type 1 (DM1) and type 2 (DM2) are caused by different unstable, noncoding microsatellite repeat expansions - (CTG)DM1 in DMPK and (CCTG)DM2 in CNBP Although transcription of mutant repeats into (CUG)DM1 or (CCUG)DM2 appears to be necessary and sufficient to cause disease, their pathomechanisms remain incompletely understood. To study the mechanisms of (CCUG)DM2 toxicity and develop a convenient model for drug screening, we generated a transgenic DM2 model in the fruit fly Drosophila melanogaster with (CCUG)n repeats of variable length (n=16 and 106). Expression of noncoding (CCUG)106, but not (CCUG)16, in muscle and retinal cells led to the formation of ribonuclear foci and mis-splicing of genes implicated in DM pathology. Mis-splicing could be rescued by co-expression of human MBNL1, but not by CUGBP1 (CELF1) complementation. Flies with (CCUG)106 displayed strong disruption of external eye morphology and of the underlying retina. Furthermore, expression of (CCUG)106 in developing retinae caused a strong apoptotic response. Inhibition of apoptosis rescued the retinal disruption in (CCUG)106 flies. Finally, we tested two chemical compounds that have shown therapeutic potential in DM1 models. Whereas treatment of (CCUG)106 flies with pentamidine had no effect, treatment with a PKR inhibitor blocked both the formation of RNA foci and apoptosis in retinae of (CCUG)106 flies. Our data indicate that expression of expanded (CCUG)DM2 repeats is toxic, causing inappropriate cell death in affected fly eyes. Our Drosophila DM2 model might provide a convenient tool for in vivo drug screening.


Subject(s)
Apoptosis/drug effects , DNA Repeat Expansion/genetics , Drosophila melanogaster/genetics , Myotonic Dystrophy/genetics , RNA/toxicity , Animals , Apoptosis/genetics , Disease Models, Animal , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/drug effects , Humans , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Atrophy/pathology , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Pentamidine/pharmacology , Photoreceptor Cells, Invertebrate/drug effects , Photoreceptor Cells, Invertebrate/metabolism , Photoreceptor Cells, Invertebrate/pathology , Protein Kinase Inhibitors/pharmacology , RNA Splicing/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Retina/abnormalities , Retina/drug effects , Retina/pathology , eIF-2 Kinase/antagonists & inhibitors , eIF-2 Kinase/metabolism
11.
Curr Protoc Hum Genet ; 91: 9.29.1-9.29.19, 2016 10 11.
Article in English | MEDLINE | ID: mdl-27727437

ABSTRACT

Myotonic dystrophy types 1 (DM1) and 2 (DM2) are autosomal dominant, microsatellite repeat expansion disorders that affect muscle function. Myotonic dystrophy type 1 is caused by CTG repeat expansion in the 3' UTR region of the DMPK gene. Patients with DM2 have expansion of CCTG repeats in intron 1 of the CNBP gene. In this unit, we review and discuss the clinical phenotypes, genetic mutations causing the diseases, and the molecular diagnostic approaches and tools that are used to determine repeat sizes in DM1/2. In summary, the goal of this chapter is to provide the reader with a basic understanding of the clinical, genetic and diagnostic aspects of these disorders. © 2016 by John Wiley & Sons, Inc.


Subject(s)
Mutation/genetics , Myotonic Dystrophy/diagnosis , Myotonic Dystrophy/genetics , Pathology, Molecular/methods , Trinucleotide Repeat Expansion/genetics , Humans , Phenotype
12.
J Bone Miner Res ; 30(9): 1608-17, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25736226

ABSTRACT

Our previous studies indicated that the Jumonji C (JmjC)-domain-containing NO66 is a histone demethylase with specificity for methylated histone H3K4 and H3K36. NO66 binds to the transcription factor Osterix (Osx) and inhibits its transcriptional activity in promoter assays. However, the physiological role of NO66 in formation of mammalian bones is unknown. Here, using a genetically engineered mouse model, we show that during early skeletal development, Prx1-Cre-dependent mesenchymal deletion of NO66 promotes osteogenesis and formation of both endochondral as well as intramembranous skeletal elements, leading to a larger skeleton and a high bone mass phenotype in adult mice. The excess bone formation in mice where NO66 was deleted in cells of mesenchymal origin is associated with an increase in the number of preosteoblasts and osteoblasts. Further analysis revealed that in the embryonic limbs and adult calvaria of mice with deletion of NO66 in cells of mesenchymal origin, expression of several genes including bone morphogenetic protein 2 (Bmp2), insulin-like growth factor 1 (Igf1), and osteoclast inhibitor osteoprotegerin was increased, concurrent with an increase in expression of bone formation markers such as osterix (Osx), type I collagen, and bone sialoprotein (Bsp). Taken together, our results provide the first in vivo evidence that NO66 histone demethylase plays an important role in mammalian osteogenesis during early development as well as in adult bone homeostasis. We postulate that NO66 regulates bone formation, at least in part, via regulating the number of bone-forming cells and expression of multiple genes that are critical for these processes.


Subject(s)
Bone and Bones/metabolism , Gene Deletion , Jumonji Domain-Containing Histone Demethylases/metabolism , Mesoderm/metabolism , Osteogenesis/genetics , Transcription Factors/metabolism , 3T3 Cells , Animals , Bone Morphogenetic Protein 2/metabolism , Cell Differentiation , Collagen Type I/metabolism , Female , Fluoresceins/chemistry , Gene Expression Regulation , Genotype , Histones/metabolism , Insulin-Like Growth Factor I/metabolism , Integrin-Binding Sialoprotein/metabolism , Jumonji Domain-Containing Histone Demethylases/genetics , Male , Mice , Mice, Knockout , Osteoblasts/metabolism , Phenotype , Sp7 Transcription Factor , X-Ray Microtomography
13.
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
14.
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
15.
Neuromuscul Disord ; 24(3): 227-40, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24332166

ABSTRACT

The prevailing pathomechanistic paradigm for myotonic dystrophy (DM) is that aberrant expression of embryonic/fetal mRNA/protein isoforms accounts for most aspects of the pleiotropic phenotype. To identify aberrant isoforms in skeletal muscle of DM1 and DM2 patients, we performed exon-array profiling and RT-PCR validation on the largest DM sample set to date, including Duchenne, Becker and tibial muscular dystrophy (NMD) patients as disease controls, and non-disease controls. Strikingly, most expression and splicing changes in DM patients were shared with NMD controls. Comparison between DM and NMD identified almost no significant differences. We conclude that DM1 and DM2 are essentially identical for dysregulation of gene expression, and DM expression changes represent a subset of broader spectrum dystrophic changes. We found no evidence for qualitative splicing differences between DM1 and DM2. While some DM-specific splicing differences exist, most of the DM splicing differences were also seen in NMD controls. SSBP3 exon 6 missplicing was observed in all diseased muscle and led to reduced protein. We conclude there is no widespread DM-specific spliceopathy in skeletal muscle and suggest that missplicing in DM (and NMD) may not be the driving mechanism for the muscle pathology, since the same pathways show expression changes unrelated to splicing.


Subject(s)
Gene Expression , Muscle, Skeletal/metabolism , Muscular Dystrophies/genetics , Myotonic Disorders/genetics , Myotonic Dystrophy/genetics , RNA Splicing , Adult , Aged , Aged, 80 and over , Child , Exons , Female , Humans , Male , Middle Aged , Muscular Dystrophies/metabolism , Myotonic Disorders/metabolism , Myotonic Dystrophy/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Young Adult
16.
Lancet Neurol ; 11(10): 891-905, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22995693

ABSTRACT

Myotonic dystrophy is the most common type of muscular dystrophy in adults and is characterised by progressive myopathy, myotonia, and multiorgan involvement. Two genetically distinct entities have been identified. Myotonic dystrophy type 1 (also known as Steinert's disease) was first described more than 100 years ago, whereas myotonic dystrophy type 2 was identified only 18 years ago, after genetic testing for type 1 disease could be applied. Both diseases are caused by autosomal dominant nucleotide repeat expansions. In patients with myotonic dystrophy type 1, a (CTG)(n) expansion is present in DMPK, whereas in patients with type 2 disease, there is a (CCTG)(n) expansion in CNBP. When transcribed into CUG-containing RNA, mutant transcripts aggregate as nuclear foci that sequester RNA-binding proteins, resulting in a spliceopathy of downstream effector genes. The prevailing paradigm therefore is that both disorders are toxic RNA diseases. However, research indicates several additional pathogenic effects take place with respect to protein translation and turnover. Despite clinical and genetic similarities, myotonic dystrophy type 1 and type 2 are distinct disorders requiring different diagnostic and management strategies.


Subject(s)
Myotonic Disorders/genetics , Myotonic Dystrophy/genetics , Humans , Myotonic Disorders/classification , Myotonic Disorders/physiopathology , Myotonic Disorders/therapy , Myotonic Dystrophy/classification , Myotonic Dystrophy/physiopathology , Myotonic Dystrophy/therapy
17.
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
18.
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
19.
Eur J Hum Genet ; 19(7): 776-82, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21364698

ABSTRACT

Myotonic dystrophy (DM) is the most common adult-onset muscular dystrophy with an estimated prevalence of 1/8000. There are two genetically distinct types, DM1 and DM2. DM2 is generally milder with more phenotypic variability than the classic DM1. Our previous data on co-segregation of heterozygous recessive CLCN1 mutations in DM2 patients indicated a higher than expected DM2 prevalence. The aim of this study was to determine the DM2 and DM1 frequency in the general population, and to explore whether the DM2 mutation functions as a modifier in other neuromuscular diseases (NMD) to account for unexplained phenotypic variability. We genotyped 5535 Finnish individuals: 4532 normal blood donors, 606 patients with various non-myotonic NMD, 221 tibial muscular dystrophy patients and their 176 healthy relatives for the DM2 and DM1 mutations. We also genotyped an Italian idiopathic non-myotonic proximal myopathy cohort (n = 93) for the DM2 mutation. In 5496 samples analyzed for DM2, we found three DM2 mutations and two premutations. In 5511 samples analyzed for DM1, we found two DM1 mutations and two premutations. In the Italian cohort, we identified one patient with a DM2 mutation. We conclude that the DM2 mutation frequency is significantly higher in the general population (1/1830; P-value = 0.0326) than previously estimated. The identification of DM2 mutations in NMD patients with clinical phenotypes not previously associated with DM2 is of particular interest and is in accord with the high overall prevalence. On the basis of our results, DM2 appears more frequent than DM1, with most DM2 patients currently undiagnosed with symptoms frequently occurring in the elderly population.


Subject(s)
Gene Frequency/genetics , Mutation/genetics , Myotonic Disorders/epidemiology , Myotonic Disorders/genetics , Myotonic Dystrophy/epidemiology , Myotonic Dystrophy/genetics , Alleles , Finland/epidemiology , Genetics, Population , Genotype , Humans , Phenotype , Prevalence
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...