Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 191
Filter
1.
Stem Cell Res ; 76: 103375, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38490135

ABSTRACT

Myotonic dystrophy type 1 (DM1) is the most prevalent adult-onset muscular dystrophy affecting 1 in 8,000 individuals. It is characterized by multisystemic symptoms, primarily myopathy. The root cause of DM1 is a heterozygous CTG triplet expansion beyond the normal size threshold in the non-coding region of the DM1 protein kinase gene (DMPK). In our study, we generated and characterized three distinct DM1 induced pluripotent stem cell (iPSC) lines with CTG repeat expansions ranging from 900 to 2000 in the DMPK gene. These iPSC lines maintained normal karyotypes, exhibited distinctive colony morphology, robustly expressed pluripotency markers, differentiated into the three primary germ layers, and lacked residual viral vectors.


Subject(s)
Induced Pluripotent Stem Cells , Myotonic Dystrophy , Adult , Humans , Myotonic Dystrophy/genetics , Myotonic Dystrophy/metabolism , Induced Pluripotent Stem Cells/metabolism , Trinucleotide Repeat Expansion , Therapeutic Human Experimentation , Cell Line , Myotonin-Protein Kinase/genetics
2.
J Clin Invest ; 134(1)2024 Jan 02.
Article in English | MEDLINE | ID: mdl-38165037

ABSTRACT

Myotonic dystrophy type 1 (DM1) is an autosomal dominant disorder caused by an unstable expanded CTG repeat located in the 3'-UTR of the DM1 protein kinase (DMPK) gene. The pathogenic mechanism results in misregulated alternative splicing of hundreds of genes, creating the dilemma of establishing which genes contribute to the mechanism of DM1 skeletal muscle pathology. In this issue of the JCI, Cisco and colleagues systematically tested the combinatorial effects of DM1-relevant mis-splicing patterns in vivo and identified the synergistic effects of mis-spliced calcium and chloride channels as a major contributor to DM1 skeletal muscle impairment. The authors further demonstrated the therapeutic potential for calcium channel modulation to block the synergistic effects and rescue myopathy.


Subject(s)
Myotonic Dystrophy , Humans , Myotonic Dystrophy/metabolism , RNA Splicing , Muscle, Skeletal/metabolism , Alternative Splicing , Ion Channels/metabolism , Myotonin-Protein Kinase/genetics , Myotonin-Protein Kinase/metabolism , Trinucleotide Repeat Expansion
3.
Stem Cell Res ; 72: 103234, 2023 10.
Article in English | MEDLINE | ID: mdl-37871474

ABSTRACT

Congenital myotonic dystrophy (CDM) is an autosomal dominant multisystemic disorder attributed to a large expansion of CTG trinucleotide repeats within the myotonic dystrophy protein kinase (DMPK) gene. In this study, we successfully reprogrammed dermal fibroblasts derived from two pediatric CDM patients and two age-matched individuals into induced pluripotent stem cells (iPSCs) using a non-integrating viral vector. The resulting CDM iPSC lines harbored approximately 2000 CTG repeats in the mutated DMPK allele. These iPSC lines expressed pluripotency markers and exhibited the capacity to differentiate into cells representing all three germinal layers, confirming their reliability as a research tool for investigating CDM and therapeutic strategies.


Subject(s)
Induced Pluripotent Stem Cells , Myotonic Dystrophy , Humans , Child , Myotonic Dystrophy/genetics , Myotonic Dystrophy/metabolism , Induced Pluripotent Stem Cells/metabolism , Trinucleotide Repeat Expansion , Reproducibility of Results , Myotonin-Protein Kinase/genetics
4.
Int J Mol Sci ; 24(12)2023 Jun 14.
Article in English | MEDLINE | ID: mdl-37373276

ABSTRACT

Myotonic dystrophy type 1 (DM1) is an autosomal dominant multisystemic disease caused by a CTG repeat expansion in the 3'-untranslated region (UTR) of DMPK gene. DM1 alleles containing non-CTG variant repeats (VRs) have been described, with uncertain molecular and clinical consequences. The expanded trinucleotide array is flanked by two CpG islands, and the presence of VRs could confer an additional level of epigenetic variability. This study aims to investigate the association between VR-containing DMPK alleles, parental inheritance and methylation pattern of the DM1 locus. The DM1 mutation has been characterized in 20 patients using a combination of SR-PCR, TP-PCR, modified TP-PCR and LR-PCR. Non-CTG motifs have been confirmed by Sanger sequencing. The methylation pattern of the DM1 locus was determined by bisulfite pyrosequencing. We characterized 7 patients with VRs within the CTG tract at 5' end and 13 patients carrying non-CTG sequences at 3' end of the DM1 expansion. DMPK alleles with VRs at 5' end or 3' end were invariably unmethylated upstream of the CTG expansion. Interestingly, DM1 patients with VRs at the 3' end showed higher methylation levels in the downstream island of the CTG repeat tract, preferentially when the disease allele was maternally inherited. Our results suggest a potential correlation between VRs, parental origin of the mutation and methylation pattern of the DMPK expanded alleles. A differential CpG methylation status could play a role in the phenotypic variability of DM1 patients, representing a potentially useful diagnostic tool.


Subject(s)
Myotonic Dystrophy , Humans , Myotonic Dystrophy/genetics , Alleles , Myotonin-Protein Kinase/genetics , Trinucleotide Repeat Expansion , CpG Islands
5.
Stem Cell Res ; 71: 103148, 2023 09.
Article in English | MEDLINE | ID: mdl-37352653

ABSTRACT

Myotonic dystrophy type 1 (DM1) is a genetic neuromuscular disorder that affects many organs, including the heart. DM1 is caused by a heterozygous CTG triplet expansion exceeding the normal size threshold in the non-coding region of the DM1 protein kinase gene (DMPK). We generated and characterized a DM1 iPSC line carrying a 700 CTG repeat expansion as well as a control iPSC line from a healthy individual. The two iPSC lines expressed several pluripotency markers, had the capacity to differentiate into the three primary germ layers, had no residual viral vectors, had normal karyotypes, and had a typical colony morphology.


Subject(s)
Induced Pluripotent Stem Cells , Myotonic Dystrophy , Humans , Myotonic Dystrophy/genetics , Myotonic Dystrophy/metabolism , Trinucleotide Repeat Expansion/genetics , Induced Pluripotent Stem Cells/metabolism , Cell Line , Myotonin-Protein Kinase/genetics
6.
Acta Neuropathol Commun ; 11(1): 44, 2023 03 15.
Article in English | MEDLINE | ID: mdl-36922901

ABSTRACT

Reduced brain volume including atrophy in grey and white matter is commonly seen in myotonic dystrophy type 1 (DM1). DM1 is caused by an expansion of CTG trinucleotide repeats in the 3' untranslated region (UTR) of the Dystrophia Myotonica Protein Kinase (DMPK) gene. Mutant DMPK mRNA containing expanded CUG RNA (DMPK-CUGexp) sequesters cytoplasmic MBNL1, resulting in morphological impairment. How DMPK-CUGexp and loss of MBNL1 cause histopathological phenotypes in the DM1 brain remains elusive. Here, we show that BDNF-TrkB retrograde transport is impaired in neurons expressing DMPK-CUGexp due to loss of cytoplasmic MBNL1 function. We reveal that mature BDNF protein levels are reduced in the brain of the DM1 mouse model EpA960/CaMKII-Cre. Exogenous BDNF treatment did not rescue impaired neurite outgrowth in neurons expressing DMPK-CUGexp, whereas overexpression of the cytoplasmic MBNL1 isoform in DMPK-CUGexp-expressing neurons improved their responsiveness to exogenous BDNF. We identify dynein light chain LC8-type 2, DYNLL2, as an MBNL1-interacting protein and demonstrate that their interaction is RNA-independent. Using time-lapse imaging, we show that overexpressed MBNL1 and DYNLL2 move along axonal processes together and that MBNL1-knockdown impairs the motility of mCherry-tagged DYNLL2, resulting in a reduced percentage of retrograde DYNLL2 movement. Examination of the distribution of DYNLL2 and activated phospho-TrkB (pTrkB) receptor in EpA960/CaMKII-Cre brains revealed an increase in the postsynaptic membrane fraction (LP1), indicating impaired retrograde transport. Finally, our neuropathological analysis of postmortem DM1 tissue reveals that reduced cytoplasmic MBNL1 expression is associated with an increase in DYNLL2 and activated pTrkB receptor levels in the synaptosomal fraction. Together, our results support that impaired MBNL1-mediated retrograde BDNF-TrkB signaling may contribute to the histopathological phenotypes of DM1.


Subject(s)
Myotonic Dystrophy , Animals , Mice , Myotonic Dystrophy/genetics , Myotonic Dystrophy/metabolism , Myotonic Dystrophy/pathology , Brain-Derived Neurotrophic Factor/metabolism , Trinucleotide Repeat Expansion , Myotonin-Protein Kinase/genetics , Myotonin-Protein Kinase/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , RNA/genetics , Brain/pathology , DNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
7.
J Neurol ; 270(6): 3138-3158, 2023 Jun.
Article in English | MEDLINE | ID: mdl-36892629

ABSTRACT

BACKGROUND AND PURPOSE: Myotonic dystrophy type 1 (DM1) is the most common form of adult-onset muscular dystrophy and is caused by an repeat expansion [r(CUG)exp] located in the 3' untranslated region of the DMPK gene. Symptoms include skeletal and cardiac muscle dysfunction and fibrosis. In DM1, there is a lack of established biomarkers in routine clinical practice. Thus, we aimed to identify a blood biomarker with relevance for DM1-pathophysiology and clinical presentation. METHODS: We collected fibroblasts from 11, skeletal muscles from 27, and blood samples from 158 DM1 patients. Moreover, serum, cardiac, and skeletal muscle samples from DMSXL mice were included. We employed proteomics, immunostaining, qPCR and ELISA. Periostin level were correlated with CMRI-data available for some patients. RESULTS: Our studies identified Periostin, a modulator of fibrosis, as a novel biomarker candidate for DM1: proteomic profiling of human fibroblasts and murine skeletal muscles showed significant dysregulation of Periostin. Immunostaining on skeletal and cardiac muscles from DM1 patients and DMSXL mice showed an extracellular increase of Periostin, indicating fibrosis. qPCR studies indicated increased POSTN expression in fibroblasts and muscle. Quantification of Periostin in blood samples from DMSXL mice and two large validation cohorts of DM1 patients showed decreased levels in animals and diseased individuals correlating with repeat expansion and disease severity and presence of cardiac symptoms identified by MRI. Analyses of longitudinal blood samples revealed no correlation with disease progression. CONCLUSIONS: Periostin might serve as a novel stratification biomarker for DM1 correlating with disease severity, presence of cardiac malfunction and fibrosis.


Subject(s)
Cardiomyopathies , Myotonic Dystrophy , Adult , Humans , Mice , Animals , Myotonic Dystrophy/genetics , Trinucleotide Repeat Expansion , Proteomics , Muscle, Skeletal , Muscle Cells/metabolism , Cardiomyopathies/genetics , Cardiomyopathies/metabolism , Patient Acuity , Myotonin-Protein Kinase/genetics
8.
Structure ; 31(4): 435-446.e4, 2023 04 06.
Article in English | MEDLINE | ID: mdl-36854301

ABSTRACT

Protein kinases of the dystonia myotonica protein kinase (DMPK) family are critical regulators of actomyosin contractility in cells. The DMPK kinase MRCK1 is required for the activation of myosin, leading to the development of cortical tension, apical constriction, and early gastrulation. Here, we present the structure, conformation, and membrane-binding properties of Caenorhabditis elegans MRCK1. MRCK1 forms a homodimer with N-terminal kinase domains, a parallel coiled coil of 55 nm, and a C-terminal tripartite module of C1, pleckstrin homology (PH), and citron homology (CNH) domains. We report the high-resolution structure of the membrane-binding C1-PH-CNH module of MRCK1 and, using high-throughput and conventional liposome-binding assays, determine its binding to specific phospholipids. We further characterize the interaction of the C-terminal CRIB motif with Cdc42. The length of the coiled-coil domain of DMPK kinases is remarkably conserved over millions of years of evolution, suggesting that they may function as molecular rulers to position kinase activity at a fixed distance from the membrane.


Subject(s)
Myotonic Dystrophy , Protein Serine-Threonine Kinases , Animals , Protein Serine-Threonine Kinases/chemistry , Myotonin-Protein Kinase/genetics , Myotonin-Protein Kinase/metabolism , Amino Acid Sequence , Protein Kinases/metabolism , Caenorhabditis elegans/metabolism
9.
Stem Cell Res ; 67: 103037, 2023 03.
Article in English | MEDLINE | ID: mdl-36739767

ABSTRACT

Myotonic dystrophy Type 1 (DM1) is a severe inherited neuromuscular disease and is the most prevalent form of muscular dystrophy in adults. DM1 involves not only the striated muscles including skeletal, and cardiac but also other organs such as the eye, brain and gonads. We have generated and characterized 4 adult heterozygous DM1 iPSC lines carrying between 1300 and 1600 CTG repeat expansion in the DM1 protein kinase gene, and a control from an apparently healthy individual. They all show strong pluripotency markers, differentiation capacity, the absence of residual viral vectors as well as normal karyotypes and colony morphologies.


Subject(s)
Induced Pluripotent Stem Cells , Myotonic Dystrophy , Adult , Humans , Myotonic Dystrophy/metabolism , Induced Pluripotent Stem Cells/metabolism , Trinucleotide Repeat Expansion , Cell Line , Muscle, Skeletal/metabolism , Myotonin-Protein Kinase/genetics
10.
Hum Mol Genet ; 32(9): 1413-1428, 2023 04 20.
Article in English | MEDLINE | ID: mdl-36222125

ABSTRACT

Myotonic dystrophy type 1 (DM1) is a multi-systemic disorder caused by expansion of CTG microsatellite repeats within DMPK. The most severe form, congenital myotonic dystrophy (CDM), has symptom onset at birth due to large intergenerational repeat expansions. Despite a common mutation, CDM individuals present with a distinct clinical phenotype and absence of common DM1 symptoms. Given the clinical divergence, it is unknown if the hallmark of DM1 pathology, dysregulation of alternative splicing (AS) due to sequestration of MBNL proteins within toxic CUG repeat RNAs, contributes to disease throughout pediatric development. To evaluate global transcriptomic dysregulation, RNA-seq was performed on 36 CDM skeletal muscle biopsies ages 2 weeks to 16 years, including two longitudinal samples. Fifty DM1 and adult/pediatric controls were also sequenced as comparative groups. Despite a large CTG expansion and shared age of onset, CDM individuals presented with a heterogenous, MBNL-dependent mis-splicing signature. Estimation of intracellular MBNL concentrations from splicing responses of select events correlated with total spliceopathy and revealed a distinct, triphasic pattern of AS dysregulation across pediatric development. CDM infants (< 2 years) possess severe mis-splicing that significantly improves in early childhood (2-8 years) independent of sex or CTG repeat load. Adolescent individuals (8-16 years) stratified into two populations with a full range of global splicing dysregulation. DMPK expression changes correlated with alterations in splicing severity during development. This study reveals the complex dynamics of the CDM muscle transcriptome and provides insights into new therapeutic strategies, timing of therapeutic intervention, and biomarker development.


Subject(s)
Myotonic Dystrophy , Child, Preschool , Humans , Myotonic Dystrophy/pathology , Transcriptome/genetics , Myotonin-Protein Kinase/genetics , Myotonin-Protein Kinase/metabolism , Muscle, Skeletal/metabolism , RNA Splicing/genetics , Trinucleotide Repeat Expansion/genetics
11.
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
12.
Int J Mol Sci ; 23(21)2022 Nov 01.
Article in English | MEDLINE | ID: mdl-36362145

ABSTRACT

Myotonic dystrophy type 1 (DM1) is a dominant genetic disease in which the expansion of long CTG trinucleotides in the 3' UTR of the myotonic dystrophy protein kinase (DMPK) gene results in toxic RNA gain-of-function and gene mis-splicing affecting mainly the muscles, the heart, and the brain. The CUG-expanded transcripts are a suitable target for the development of antisense oligonucleotide (ASO) therapies. Various chemical modifications of the sugar-phosphate backbone have been reported to significantly enhance the affinity of ASOs for RNA and their resistance to nucleases, making it possible to reverse DM1-like symptoms following systemic administration in different transgenic mouse models. However, specific tissue delivery remains to be improved to achieve significant clinical outcomes in humans. Several strategies, including ASO conjugation to cell-penetrating peptides, fatty acids, or monoclonal antibodies, have recently been shown to improve potency in muscle and cardiac tissues in mice. Moreover, intrathecal administration of ASOs may be an advantageous complementary administration route to bypass the blood-brain barrier and correct defects of the central nervous system in DM1. This review describes the evolution of the chemical design of antisense oligonucleotides targeting CUG-expanded mRNAs and how recent advances in the field may be game-changing by forwarding laboratory findings into clinical research and treatments for DM1 and other microsatellite diseases.


Subject(s)
Myotonic Dystrophy , Mice , Humans , Animals , Myotonic Dystrophy/drug therapy , Myotonic Dystrophy/genetics , Myotonin-Protein Kinase/genetics , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/therapeutic use , Mice, Transgenic , Oligonucleotides/therapeutic use , 3' Untranslated Regions , Trinucleotide Repeat Expansion
13.
J Mol Diagn ; 24(11): 1143-1154, 2022 11.
Article in English | MEDLINE | ID: mdl-36084803

ABSTRACT

Myotonic dystrophy type 1 (DM1) exhibits highly heterogeneous clinical manifestations caused by an unstable CTG repeat expansion reaching up to 4000 CTG. The clinical variability depends on CTG repeat number, CNG repeat interruptions, and somatic mosaicism. Currently, none of these factors are simultaneously and accurately determined due to the limitations of gold standard methods used in clinical and research laboratories. An amplicon method for targeting the DMPK locus using single-molecule real-time sequencing was recently developed to accurately analyze expanded alleles. However, amplicon-based sequencing still depends on PCR, and the inherent bias toward preferential amplification of smaller repeats can be problematic in DM1. Thus, an amplification-free long-read sequencing method was developed by using CRISPR/Cas9 technology in DM1. This method was used to sequence the DMPK locus in patients with CTG repeat expansion ranging from 130 to >1000 CTG. We showed that elimination of PCR amplification improves the accuracy of measurement of inherited repeat number and somatic repeat variations, two key factors in DM1 severity and age at onset. For the first time, an expansion composed of >85% CCG repeats was identified by using this innovative method in a DM1 family with an atypical clinical profile. No-amplification targeted sequencing represents a promising method that can overcome research and diagnosis shortcomings, with translational implications for clinical and genetic counseling in DM1.


Subject(s)
Myotonic Dystrophy , Humans , Myotonic Dystrophy/diagnosis , Myotonic Dystrophy/genetics , Myotonin-Protein Kinase/genetics , Alleles , Trinucleotide Repeat Expansion/genetics , Genetic Counseling
14.
Intern Med ; 61(15): 2281-2286, 2022.
Article in English | MEDLINE | ID: mdl-35908960

ABSTRACT

Objective In myotonic dystrophy type 1 (DM1), the CTG repeat size in the dystrophia myotonica protein kinase gene has been shown to correlate with disease severity and is a potential predictive marker for respiratory decline. However, genetic testing can be challenging in some clinical situations. We developed a simple formula for estimating the CTG repeat size using a single spirometry test in patients with DM1. Methods In this single-center retrospective study, we reviewed 50 consecutive patients with genetically confirmed DM1 whose follow-up visits were at our hospital. The patients were randomly assigned to training and test analysis subsets. By applying a linear mixed model to the longitudinal spirometry results of the training set, we calculated the fixed effects on the annual respiratory decline. Subsequently, we derived a prediction formula to calculate the repeat size that incorporated %vital capacity (%VC) and the patient's age at the time of the spirometry evaluation; the results were validated by the test set. Results A total of 157 spirometry tests were recorded. The fixed effects on the annual %VC decline were =-0.90. The derived formula [repeat size=-16.8× (age+%VC/0.90) +2663] had a moderate predictive performance with a mean coefficient of determination of 0.41. Conclusion The CTG repeat size in patients with DM1 can be potentially predicted using a simple formula based on a single spirometry test conducted at any time over the disease course. It can be useful as a supportive tool for advance care planning when genetic testing is not available.


Subject(s)
Myotonic Dystrophy , Disease Progression , Humans , Myotonic Dystrophy/diagnosis , Myotonic Dystrophy/genetics , Myotonin-Protein Kinase/genetics , Randomized Controlled Trials as Topic , Retrospective Studies , Spirometry , Trinucleotide Repeat Expansion/genetics
15.
Genes (Basel) ; 13(6)2022 05 28.
Article in English | MEDLINE | ID: mdl-35741732

ABSTRACT

Myotonic dystrophy type 1 (DM1) is a multisystemic neuromuscular disorder caused by the expansion of a CTG repeat in the 3'-UTR of DMPK, which is transcribed to a toxic gain-of-function RNA that affects splicing of a range of genes. The expanded repeat is unstable in both germline and somatic cells. The variable age at disease onset and severity of symptoms have been linked to the inherited CTG repeat length, non-CTG interruptions, and methylation levels flanking the repeat. In general, the genetic biomarkers are investigated separately with specific methods, making it tedious to obtain an overall characterisation of the repeat for a given individual. In the present study, we employed Oxford nanopore sequencing in a pilot study to simultaneously determine the repeat lengths, investigate the presence and nature of repeat interruptions, and quantify methylation levels in the regions flanking the CTG-repeats in four patients with DM1. We determined the repeat lengths, and in three patients, we observed interruptions which were not detected using repeat-primed PCR. Interruptions may thus be more common than previously anticipated and should be investigated in larger cohorts. Allele-specific analyses enabled characterisation of aberrant methylation levels specific to the expanded allele, which greatly increased the sensitivity and resolved cases where the methylation levels were ambiguous.


Subject(s)
Myotonic Dystrophy , Myotonin-Protein Kinase , DNA Methylation , Humans , Myotonic Dystrophy/diagnosis , Myotonic Dystrophy/genetics , Myotonin-Protein Kinase/genetics , Pilot Projects , RNA Splicing , Trinucleotide Repeat Expansion
16.
Dis Model Mech ; 15(7)2022 07 01.
Article in English | MEDLINE | ID: mdl-35642886

ABSTRACT

RNA regulation in mammalian cells requires complex physical compartmentalisation, using structures thought to be formed by liquid-liquid phase separation. Disruption of these structures is implicated in numerous degenerative diseases. Myotonic dystrophy type 1 (DM1) is a multi-systemic trinucleotide repeat disorder resulting from an expansion of nucleotides CTG (CTGexp) in the DNA encoding DM1 protein kinase (DMPK). The cellular hallmark of DM1 is the formation of nuclear foci that contain expanded DMPK RNA (CUGexp) (with thymine instead of uracil). We report here the deregulation of stress granules (SGs) and processing bodies (P-bodies), two cytoplasmic structures key for mRNA regulation, in cell culture models of DM1. Alterations to the rates of formation and dispersal of SGs suggest an altered ability of cells to respond to stress associated with DM1, while changes to the structure and dynamics of SGs and P-bodies suggest that a widespread alteration to the biophysical properties of cellular structures is a consequence of the presence of CUGexp RNA.


Subject(s)
Myotonic Dystrophy , Animals , Mammals/metabolism , Myotonic Dystrophy/genetics , Myotonin-Protein Kinase/genetics , Myotonin-Protein Kinase/metabolism , Processing Bodies , RNA , Stress Granules , Trinucleotide Repeat Expansion/genetics
17.
Int J Mol Sci ; 23(9)2022 Apr 21.
Article in English | MEDLINE | ID: mdl-35563013

ABSTRACT

Myotonic dystrophy type 1 (DM1) is the most common muscular dystrophy affecting many different body tissues, predominantly skeletal and cardiac muscles and the central nervous system. The expansion of CTG repeats in the DM1 protein-kinase (DMPK) gene is the genetic cause of the disease. The pathogenetic mechanisms are mainly mediated by the production of a toxic expanded CUG transcript from the DMPK gene. With the availability of new knowledge, disease models, and technical tools, much progress has been made in the discovery of altered pathways and in the potential of therapeutic intervention, making the path to the clinic a closer reality. In this review, we describe and discuss the molecular therapeutic strategies for DM1, which are designed to directly target the CTG genomic tract, the expanded CUG transcript or downstream signaling molecules.


Subject(s)
Myotonic Dystrophy , Gene Editing , Humans , Myotonic Dystrophy/drug therapy , Myotonic Dystrophy/genetics , Myotonin-Protein Kinase/genetics , Myotonin-Protein Kinase/metabolism , Trinucleotide Repeat Expansion/genetics
18.
Mol Genet Genomic Med ; 10(5): e1881, 2022 05.
Article in English | MEDLINE | ID: mdl-35319816

ABSTRACT

BACKGROUND: Gastrointestinal stromal tumors (GISTs) are the most common type of mesenchymal tumor in gastrointestinal tract, with striking features of morphology and immunohistochemistry. But GISTs in pregnancy could seldom be found. Pathogenic activating mutations of the proto-oncogene KIT and PDGFRA are detected in majority GISTs, and adjuvant imatinib therapy targeting KIT and PDGFRA mutations is recommended for patients with high-risk GIST. However, some rare subgroups with distinct molecular features remain uncovered and more therapeutic targets need to be revealed. METHODS: The DNA/RNA samples were detected using the NGS-based YuanSu450 gene panel. After identifying the CDC42BPB-ALK fusion by NGS, this novel fusion was further confirmed by Sanger sequencing. Subsequently, FISH analysis was performed using the Vysis ALK Break Apart FISH Probe kit to testify the ALK status. ALK protein expression was confirmed by IHC (D5F3 and 5A4). RESULTS: Herein, we reported the first case of quadruple wild-type (WT) GIST with ALK-CDC42BPB fusion and ALK (D5F3) overexpression. In this study, we described a 33-year-old pregnant patient in lactation who had a massive space occupying lesion (with the maximum diameter of 22 cm) in the stomach and was eventually diagnosed as quadruple WT GIST (KITWT /PDGFRAWT /SDHWT /RAS-PWT ). CONCLUSION: We unexpectedly found that this GIST patient showed ALK (D5F3) overexpression and harbored a novel fusion CDC42BPB exon 24-ALK in exon 20.


Subject(s)
Gastrointestinal Stromal Tumors , Adult , Female , Gastrointestinal Stromal Tumors/genetics , Gastrointestinal Stromal Tumors/pathology , Humans , Mutation , Myotonin-Protein Kinase/genetics , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins c-kit/metabolism , Proto-Oncogene Proteins c-kit/therapeutic use , Receptor Protein-Tyrosine Kinases/genetics , Receptor, Platelet-Derived Growth Factor alpha/genetics , Receptor, Platelet-Derived Growth Factor alpha/metabolism
19.
Doc Ophthalmol ; 144(3): 217-226, 2022 06.
Article in English | MEDLINE | ID: mdl-35284965

ABSTRACT

BACKGROUND: Myotonic dystrophy type 1 (DM1) is an autosomal dominant genetic disorder that affects multiple organs, including the muscle and eye, caused by a CTG triplet expansion of the 3' untranslated region (UTR) of the DMPK gene. Cataracts and retinal degeneration are major eye complications in patients with DM1. We reported the case of a Japanese patient with DM1 who exhibited submacular hemorrhage unilaterally, rarely complicating DM1. CASE REPORT: A 56-year-old woman presented with loss of visual acuity in the left eye (LE). The patient was diagnosed with DM1, who carried expanded CTG repeats (1100) of the 3' UTR of DMPK. Her corrected visual acuities were 20/100 and 20/2000 in the right eye (RE) and LE, respectively. Cataracts were observed in both eyes. Fundoscopy and angiography revealed submacular hemorrhage in the LE due to polypoidal choroidal vasculopathy (PCV, also known as aneurysmal type 1 neovascularization). The patient underwent intravitreal injections of an anti-vascular endothelial growth factor drug and sulfur hexafluoride gas in the LE. Full-field electroretinography was performed, showing that the rod and standard-flash responses were reduced to 50% and below 10% in the RE and LE, whereas the cone and 30-Hz flicker responses were reduced to 40-50% and 15-20% in the RE and LE, respectively, compared with the controls. Multifocal electroretinography revealed that the overall responses were extinguished in the LE and considerably attenuated in the RE. CONCLUSIONS: This is the first patient with DM1 complicated with PCV. Widespread retinal dysfunction may be associated with expanded CTG repeats, which is significantly longer than the mean repeat number of patients with DM1.


Subject(s)
Cataract , Eye Diseases , Myotonic Dystrophy , Cataract/complications , Electroretinography , Eye Diseases/complications , Female , Humans , Intravitreal Injections , Middle Aged , Myotonic Dystrophy/complications , Myotonic Dystrophy/diagnosis , Myotonic Dystrophy/genetics , Myotonin-Protein Kinase/genetics , Retinal Hemorrhage/diagnosis , Retinal Hemorrhage/etiology
20.
Int J Mol Sci ; 23(2)2022 Jan 06.
Article in English | MEDLINE | ID: mdl-35054778

ABSTRACT

Myotonic dystrophy type 1 (DM1) is a severe neuromuscular disease mediated by a toxic gain of function of mutant RNAs. The neuropsychological manifestations affect multiple domains of cognition and behavior, but their etiology remains elusive. Transgenic DMSXL mice carry the DM1 mutation, show behavioral abnormalities, and express low levels of GLT1, a critical regulator of glutamate concentration in the synaptic cleft. However, the impact of glutamate homeostasis on neurotransmission in DM1 remains unknown. We confirmed reduced glutamate uptake in the DMSXL hippocampus. Patch clamp recordings in hippocampal slices revealed increased amplitude of tonic glutamate currents in DMSXL CA1 pyramidal neurons and DG granule cells, likely mediated by higher levels of ambient glutamate. Unexpectedly, extracellular GABA levels and tonic current were also elevated in DMSXL mice. Finally, we found evidence of synaptic dysfunction in DMSXL mice, suggestive of abnormal short-term plasticity, illustrated by an altered LTP time course in DG and in CA1. Synaptic dysfunction was accompanied by RNA foci accumulation in localized areas of the hippocampus and by the mis-splicing of candidate genes with relevant functions in neurotransmission. Molecular and functional changes triggered by toxic RNA may induce synaptic abnormalities in restricted brain areas that favor neuronal dysfunction.


Subject(s)
Hippocampus/metabolism , Myotonic Dystrophy/physiopathology , Myotonin-Protein Kinase/physiology , Neuronal Plasticity , Neurotransmitter Agents/metabolism , RNA Splicing , Animals , Disease Models, Animal , Excitatory Amino Acid Transporter 2 , Hippocampus/physiology , Homeostasis , Mice , Mice, Transgenic , Myotonic Dystrophy/metabolism , Myotonin-Protein Kinase/genetics , Pyramidal Cells/metabolism , Pyramidal Cells/physiology , RNA/metabolism , Synaptic Transmission
SELECTION OF CITATIONS
SEARCH DETAIL
...