Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
Add more filters

Publication year range
1.
Nucleic Acids Res ; 52(5): 2355-2371, 2024 Mar 21.
Article in English | MEDLINE | ID: mdl-38180815

ABSTRACT

The yeast Rif2 protein is known to inhibit Mre11 nuclease and the activation of Tel1 kinase through a short motif termed MIN, which binds the Rad50 subunit and simulates its ATPase activity in vitro. The mechanism by which Rif2 restrains Tel1 activation and the consequences of this inhibition at DNA double-strand breaks (DSBs) are poorly understood. In this study, we employed AlphaFold-Multimer modelling to pinpoint and validate the interaction surface between Rif2 MIN and Rad50. We also engineered the rif2-S6E mutation that enhances the inhibitory effect of Rif2 by increasing Rif2-Rad50 interaction. Unlike rif2Δ, the rif2-S6E mutation impairs hairpin cleavage. Furthermore, it diminishes Tel1 activation by inhibiting Tel1 binding to DSBs while leaving MRX association unchanged, indicating that Rif2 can directly inhibit Tel1 recruitment to DSBs. Additionally, Rif2S6E reduces Tel1-MRX interaction and increases stimulation of ATPase by Rad50, indicating that Rif2 binding to Rad50 induces an ADP-bound MRX conformation that is not suitable for Tel1 binding. The decreased Tel1 recruitment to DSBs in rif2-S6E cells impairs DSB end-tethering and this bridging defect is suppressed by expressing a Tel1 mutant variant that increases Tel1 persistence at DSBs, suggesting a direct role for Tel1 in the bridging of DSB ends.


Subject(s)
DNA-Binding Proteins , Protein Serine-Threonine Kinases , Saccharomyces cerevisiae Proteins , Telomere-Binding Proteins , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , DNA/genetics , DNA/metabolism , DNA Repair , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Endodeoxyribonucleases/genetics , Endodeoxyribonucleases/metabolism , Exodeoxyribonucleases/genetics , Exodeoxyribonucleases/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Telomere-Binding Proteins/metabolism
2.
Nucleic Acids Res ; 51(4): 1783-1802, 2023 02 28.
Article in English | MEDLINE | ID: mdl-36762474

ABSTRACT

DNA double-strand breaks (DSBs) can be repaired by either homologous recombination (HR) or non-homologous end-joining (NHEJ). NHEJ is induced by the binding to DSBs of the Ku70-Ku80 heterodimer, which acts as a hub for the recruitment of downstream NHEJ components. An important issue in DSB repair is the maintenance of the DSB ends in close proximity, a function that in yeast involves the MRX complex and Sae2. Here, we provide evidence that Ku contributes to keep the DNA ends tethered to each other. The ku70-C85Y mutation, which increases Ku affinity for DNA and its persistence very close to the DSB ends, enhances DSB end-tethering and suppresses the end-tethering defect of sae2Δ cells. Impairing histone removal around DSBs either by eliminating Tel1 kinase activity or nucleosome remodelers enhances Ku persistence at DSBs and DSB bridging, suggesting that Tel1 antagonizes the Ku function in supporting end-tethering by promoting nucleosome removal and possibly Ku sliding inwards. As Ku provides a block to DSB resection, this Tel1 function can be important to regulate the mode by which DSBs are repaired.


Subject(s)
DNA-Binding Proteins , Protein Serine-Threonine Kinases , Saccharomyces cerevisiae Proteins , DNA/metabolism , DNA Breaks, Double-Stranded , DNA End-Joining Repair , DNA Repair , DNA-Binding Proteins/genetics , Intracellular Signaling Peptides and Proteins/genetics , Ku Autoantigen/metabolism , Nucleosomes , Protein Serine-Threonine Kinases/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics
3.
Nucleic Acids Res ; 50(10): 5443-5466, 2022 06 10.
Article in English | MEDLINE | ID: mdl-35061895

ABSTRACT

Although recent regulatory approval of splice-switching oligonucleotides (SSOs) for the treatment of neuromuscular disease such as Duchenne muscular dystrophy has been an advance for the splice-switching field, current SSO chemistries have shown limited clinical benefit due to poor pharmacology. To overcome limitations of existing technologies, we engineered chimeric stereopure oligonucleotides with phosphorothioate (PS) and phosphoryl guanidine-containing (PN) backbones. We demonstrate that these chimeric stereopure oligonucleotides have markedly improved pharmacology and efficacy compared with PS-modified oligonucleotides, preventing premature death and improving median survival from 49 days to at least 280 days in a dystrophic mouse model with an aggressive phenotype. These data demonstrate that chemical optimization alone can profoundly impact oligonucleotide pharmacology and highlight the potential for continued innovation around the oligonucleotide backbone. More specifically, we conclude that chimeric stereopure oligonucleotides are a promising splice-switching modality with potential for the treatment of neuromuscular and other genetic diseases impacting difficult to reach tissues such as the skeletal muscle and heart.


Subject(s)
Muscular Dystrophy, Duchenne , Oligonucleotides, Antisense/chemistry , Phosphorothioate Oligonucleotides/chemistry , Animals , Exons , Mice , Muscle, Skeletal , Muscular Dystrophy, Duchenne/drug therapy , Muscular Dystrophy, Duchenne/therapy , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/pharmacology , Phosphorothioate Oligonucleotides/pharmacology , RNA Splicing/drug effects
4.
Pract Neurol ; 2024 Mar 14.
Article in English | MEDLINE | ID: mdl-38485225

ABSTRACT

A 57-year-old man developed worsening early morning headaches, muscle cramps and falls over 12 months. He had widespread fasciculation and was diagnosed with motor neurone disease, and treated with nocturnal hypoventilation. Based on this diagnosis, he made significant personal and financial decisions including retiring and selling his house. He subsequently developed a lump in his right breast and was found to have gynaecomastia. This triggered genetic testing for Kennedy's disease leading to the correct diagnosis. This case highlights an unusual presentation of a rare disease leading to misdiagnosis and major repercussions for the patient. Recent genetic analysis from the 100 000 genome project suggests Kennedy's disease may be four times more prevalent in the population than previously thought, highlighting the need to consider genetic testing, especially if there is a suggestion of multisystem disease.

5.
Acta Neuropathol ; 143(6): 713-731, 2022 06.
Article in English | MEDLINE | ID: mdl-35522298

ABSTRACT

Androgens and androgen-related molecules exert a plethora of functions across different tissues, mainly through binding to the transcription factor androgen receptor (AR). Despite widespread therapeutic use and misuse of androgens as potent anabolic agents, the molecular mechanisms of this effect on skeletal muscle are currently unknown. Muscle mass in adulthood is mainly regulated by the bone morphogenetic protein (BMP) axis of the transforming growth factor (TGF)-ß pathway via recruitment of mothers against decapentaplegic homolog 4 (SMAD4) protein. Here we show that, upon activation, AR forms a transcriptional complex with SMAD4 to orchestrate a muscle hypertrophy programme by modulating SMAD4 chromatin binding dynamics and enhancing its transactivation activity. We challenged this mechanism of action using spinal and bulbar muscular atrophy (SBMA) as a model of study. This adult-onset neuromuscular disease is caused by a polyglutamine expansion (polyQ) in AR and is characterized by progressive muscle weakness and atrophy secondary to a combination of lower motor neuron degeneration and primary muscle atrophy. Here we found that the presence of an elongated polyQ tract impairs AR cooperativity with SMAD4, leading to an inability to mount an effective anti-atrophy gene expression programme in skeletal muscle in response to denervation. Furthermore, adeno-associated virus, serotype 9 (AAV9)-mediated muscle-restricted delivery of BMP7 is able to rescue the muscle atrophy in SBMA mice, supporting the development of treatments able to fine-tune AR-SMAD4 transcriptional cooperativity as a promising target for SBMA and other conditions associated with muscle loss.


Subject(s)
Muscular Atrophy, Spinal , Receptors, Androgen , Androgens/metabolism , Androgens/pharmacology , Animals , Homeostasis , Mice , Mice, Transgenic , Muscle, Skeletal/pathology , Muscular Atrophy/metabolism , Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/metabolism , Receptors, Androgen/genetics , Smad4 Protein
6.
Mol Psychiatry ; 26(10): 5636-5657, 2021 10.
Article in English | MEDLINE | ID: mdl-31942037

ABSTRACT

Alzheimer's disease (AD) is the most common age-related form of dementia, associated with deposition of intracellular neuronal tangles consisting primarily of hyperphosphorylated microtubule-associated protein tau (p-tau) and extracellular plaques primarily comprising amyloid- ß (Aß) peptide. The p-tau tangle unit is a posttranslational modification of normal tau protein. Aß is a neurotoxic peptide excised from the amyloid-ß precursor protein (APP) by ß-site APP-cleaving enzyme 1 (BACE1) and the γ-secretase complex. MicroRNAs (miRNAs) are short, single-stranded RNAs that modulate protein expression as part of the RNA-induced silencing complex (RISC). We identified miR-298 as a repressor of APP, BACE1, and the two primary forms of Aß (Aß40 and Aß42) in a primary human cell culture model. Further, we discovered a novel effect of miR-298 on posttranslational levels of two specific tau moieties. Notably, miR-298 significantly reduced levels of ~55 and 50 kDa forms of the tau protein without significant alterations of total tau or other forms. In vivo overexpression of human miR-298 resulted in nonsignificant reduction of APP, BACE1, and tau in mice. Moreover, we identified two miR-298 SNPs associated with higher cerebrospinal fluid (CSF) p-tau and lower CSF Aß42 levels in a cohort of human AD patients. Finally, levels of miR-298 varied in postmortem human temporal lobe between AD patients and age-matched non-AD controls. Our results suggest that miR-298 may be a suitable target for AD therapy.


Subject(s)
Alzheimer Disease , MicroRNAs , Alzheimer Disease/genetics , Amyloid Precursor Protein Secretases/genetics , Amyloid beta-Peptides , Amyloid beta-Protein Precursor/genetics , Animals , Aspartic Acid Endopeptidases/genetics , Humans , Mice , MicroRNAs/genetics , tau Proteins/genetics
7.
Nucleic Acids Res ; 48(5): 2424-2441, 2020 03 18.
Article in English | MEDLINE | ID: mdl-31879780

ABSTRACT

The cellular response to DNA double-strand breaks (DSBs) is initiated by the Mre11-Rad50-Xrs2 (MRX) complex that has structural and catalytic functions. MRX association at DSBs is counteracted by Rif2, which is known to interact with Rap1 that binds telomeric DNA through two tandem Myb-like domains. Whether and how Rap1 acts at DSBs is unknown. Here we show that Rif2 inhibits MRX association to DSBs in a manner dependent on Rap1, which binds to DSBs and promotes Rif2 association to them. Rap1 in turn can negatively regulate MRX function at DNA ends also independently of Rif2. In fact, a characterization of Rap1 mutant variants shows that Rap1 binding to DNA through both Myb-like domains results in formation of Rap1-DNA complexes that control MRX functions at both DSBs and telomeres primarily through Rif2. By contrast, Rap1 binding to DNA through a single Myb-like domain results in formation of high stoichiometry complexes that act at DNA ends mostly in a Rif2-independent manner. Altogether these findings indicate that the DNA binding modes of Rap1 influence its functional properties, thus highlighting the structural plasticity of this protein.


Subject(s)
DNA, Fungal/metabolism , Multiprotein Complexes/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Telomere Homeostasis , Telomere-Binding Proteins/metabolism , Telomere/metabolism , Transcription Factors/metabolism , Alleles , DNA Breaks, Double-Stranded , DNA Damage , Models, Biological , Mutation/genetics , Protein Binding , Saccharomyces cerevisiae/cytology , Shelterin Complex , Transcription, Genetic
8.
Hum Mol Genet ; 25(10): 1979-1989, 2016 05 15.
Article in English | MEDLINE | ID: mdl-26962150

ABSTRACT

Spinal and bulbar muscular atrophy (SBMA, also known as Kennedy's disease) is one of nine neurodegenerative disorders that are caused by expansion of polyglutamine-encoding CAG repeats. Intracellular accumulation of abnormal proteins in these diseases, a pathological hallmark, is associated with defects in protein homeostasis. Enhancement of the cellular proteostasis capacity with small molecules has therefore emerged as a promising approach to treatment. Here, we characterize a novel curcumin analog, ASC-JM17, as an activator of central pathways controlling protein folding, degradation and oxidative stress resistance. ASC-JM17 acts on Nrf1, Nrf2 and Hsf1 to increase the expression of proteasome subunits, antioxidant enzymes and molecular chaperones. We show that ASC-JM17 ameliorates toxicity of the mutant androgen receptor (AR) responsible for SBMA in cell, fly and mouse models. Knockdown of the Drosophila Nrf1 and Nrf2 ortholog cap 'n' collar isoform-C, but not Hsf1, blocks the protective effect of ASC-JM17 on mutant AR-induced eye degeneration in flies. Our observations indicate that activation of the Nrf1/Nrf2 pathway is a viable option for pharmacological intervention in SBMA and potentially other polyglutamine diseases.


Subject(s)
Bulbo-Spinal Atrophy, X-Linked/genetics , Curcumin/analogs & derivatives , DNA-Binding Proteins/genetics , Drosophila Proteins/genetics , Muscular Disorders, Atrophic/genetics , NF-E2-Related Factor 1/genetics , NF-E2-Related Factor 2/genetics , Receptors, Androgen/genetics , Transcription Factors/genetics , Trinucleotide Repeat Expansion/genetics , Animals , Bulbo-Spinal Atrophy, X-Linked/drug therapy , Bulbo-Spinal Atrophy, X-Linked/pathology , Curcumin/administration & dosage , Curcumin/chemistry , Disease Models, Animal , Drosophila melanogaster/genetics , Gene Knockdown Techniques , Heat Shock Transcription Factors , Humans , Mice , Muscular Disorders, Atrophic/drug therapy , Muscular Disorders, Atrophic/pathology , Oxidative Stress/drug effects , Peptides/genetics , Proteasome Endopeptidase Complex/drug effects , Protein Aggregation, Pathological/genetics , Protein Folding/drug effects , Signal Transduction/drug effects , Small Molecule Libraries/administration & dosage
9.
J Neurol Neurosurg Psychiatry ; 89(8): 808-812, 2018 08.
Article in English | MEDLINE | ID: mdl-29353237

ABSTRACT

Kennedy's disease, or spinal and bulbar muscular atrophy (SBMA), is an X-linked neuromuscular condition clinically characterised by weakness, atrophy and fasciculations of the limb and bulbar muscles, as a result of lower motor neuron degeneration. The disease is caused by an abnormally expanded triplet repeat expansions in the ubiquitously expressed androgen receptor gene, through mechanisms which are not entirely elucidated. Over the years studies from both humans and animal models have highlighted the involvement of cell populations other than motor neurons in SBMA, widening the disease phenotype. The most compelling aspect of these findings is their potential for therapeutic impact: muscle, for example, which is primarily affected in the disease, has been recently shown to represent a valid alternative target for therapy to motor neurons. In this review, we discuss the emerging study of the extra-motor neuron involvement in SBMA, which, besides increasingly pointing towards a multidisciplinary approach for affected patients, deepens our understanding of the pathogenic mechanisms and holds potential for providing new therapeutic targets for this disease.


Subject(s)
Autonomic Nervous System Diseases/pathology , Bulbo-Spinal Atrophy, X-Linked/pathology , Motor Neurons/pathology , Muscular Atrophy/pathology , Urinary Bladder Neck Obstruction/pathology , Autonomic Nervous System Diseases/genetics , Bulbo-Spinal Atrophy, X-Linked/genetics , Humans , Muscular Atrophy/genetics , Phenotype , Trinucleotide Repeat Expansion , Urinary Bladder Neck Obstruction/genetics
10.
Mol Ther ; 24(5): 937-45, 2016 05.
Article in English | MEDLINE | ID: mdl-26755334

ABSTRACT

Spinal and bulbar muscular atrophy (SBMA) is a currently untreatable adult-onset neuromuscular disease caused by expansion of a polyglutamine repeat in the androgen receptor (AR). In SBMA, as in other polyglutamine diseases, a toxic gain of function in the mutant protein is an important factor in the disease mechanism; therefore, reducing the mutant protein holds promise as an effective treatment strategy. In this work, we evaluated a microRNA (miRNA) to reduce AR expression. From a list of predicted miRNAs that target human AR, we selected microRNA-298 (miR-298) for its ability to downregulate AR mRNA and protein levels when transfected in cells overexpressing wild-type and mutant AR and in SBMA patient-derived fibroblasts. We showed that miR-298 directly binds to the 3'-untranslated region of the human AR transcript, and counteracts AR toxicity in vitro. Intravenous delivery of miR-298 with adeno-associated virus serotype 9 vector resulted in efficient transduction of muscle and spinal cord and amelioration of the disease phenotype in SBMA mice. Our findings support the development of miRNAs as a therapeutic strategy for SBMA and other neurodegenerative disorders caused by toxic proteins.


Subject(s)
Down-Regulation , Genetic Therapy/methods , MicroRNAs/genetics , Muscular Atrophy, Spinal/therapy , Receptors, Androgen/genetics , 3' Untranslated Regions , Administration, Intravenous , Animals , Cell Line , Dependovirus/genetics , Disease Models, Animal , Genetic Vectors/administration & dosage , Humans , MCF-7 Cells , Mice , Muscular Atrophy, Spinal/genetics
11.
Am J Hum Genet ; 91(6): 1095-102, 2012 Dec 07.
Article in English | MEDLINE | ID: mdl-23217327

ABSTRACT

Cowchock syndrome (CMTX4) is a slowly progressive X-linked recessive disorder with axonal neuropathy, deafness, and cognitive impairment. The disease locus was previously mapped to an 11 cM region at chromosome X: q24-q26. Exome sequencing of an affected individual from the originally described family identified a missense change c.1478A>T (p.Glu493Val) in AIFM1, the gene encoding apoptosis-inducing factor (AIF) mitochondrion-associated 1. The change is at a highly conserved residue and cosegregated with the phenotype in the family. AIF is an FAD-dependent NADH oxidase that is imported into mitochondria. With apoptotic insults, a N-terminal transmembrane linker is cleaved off, producing a soluble fragment that is released into the cytosol and then transported into the nucleus, where it triggers caspase-independent apoptosis. Another AIFM1 mutation that predicts p.Arg201del has recently been associated with severe mitochondrial encephalomyopathy in two infants by impairing oxidative phosphorylation. The c.1478A>T (p.Glu493Val) mutation found in the family reported here alters the redox properties of the AIF protein and results in increased cell death via apoptosis, without affecting the activity of the respiratory chain complexes. Our findings expand the spectrum of AIF-related disease and provide insight into the effects of AIFM1 mutations.


Subject(s)
Apoptosis Inducing Factor/genetics , Charcot-Marie-Tooth Disease/genetics , Hearing Loss, Sensorineural/genetics , Mental Retardation, X-Linked/genetics , Mutation , Apoptosis/genetics , Apoptosis Inducing Factor/chemistry , Apoptosis Inducing Factor/metabolism , Base Sequence , Brain/pathology , Cell Nucleus/genetics , Cell Nucleus/metabolism , Charcot-Marie-Tooth Disease/diagnosis , Charcot-Marie-Tooth Disease/metabolism , Exons , Hearing Loss, Sensorineural/diagnosis , Hearing Loss, Sensorineural/metabolism , Humans , Magnetic Resonance Imaging , Male , Mental Retardation, X-Linked/diagnosis , Mental Retardation, X-Linked/metabolism , Mitochondria/genetics , Mitochondria/metabolism , Mitochondria/ultrastructure , Models, Molecular , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscle, Skeletal/ultrastructure , Neuroimaging , Oxidation-Reduction , Pedigree , Protein Conformation , Protein Transport
13.
Neurobiol Dis ; 70: 12-20, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24925468

ABSTRACT

Spinal and bulbar muscular atrophy (SBMA, Kennedy's disease) is a motor neuron disease caused by polyglutamine repeat expansion in the androgen receptor. Although degeneration occurs in the spinal cord and muscle, the exact mechanism is not clear. Induced pluripotent stem cells from spinal and bulbar muscular atrophy patients provide a useful model for understanding the disease mechanism and designing effective therapy. Stem cells were generated from six patients and compared to control lines from three healthy individuals. Motor neurons from four patients were differentiated from stem cells and characterized to understand disease-relevant phenotypes. Stem cells created from patient fibroblasts express less androgen receptor than control cells, but show androgen-dependent stabilization and nuclear translocation. The expanded repeat in several stem cell clones was unstable, with either expansion or contraction. Patient stem cell clones produced a similar number of motor neurons compared to controls, with or without androgen treatment. The stem cell-derived motor neurons had immunoreactivity for HB9, Isl1, ChAT, and SMI-32, and those with the largest repeat expansions were found to have increased acetylated α-tubulin and reduced HDAC6. Reduced HDAC6 was also found in motor neuron cultures from two other patients with shorter repeats. Evaluation of stably transfected mouse cells and SBMA spinal cord showed similar changes in acetylated α-tubulin and HDAC6. Perinuclear lysosomal enrichment, an HDAC6 dependent process, was disrupted in motor neurons from two patients with the longest repeats. SBMA stem cells present new insights into the disease, and the observations of reduced androgen receptor levels, repeat instability, and reduced HDAC6 provide avenues for further investigation of the disease mechanism and development of effective therapy.


Subject(s)
Bulbo-Spinal Atrophy, X-Linked/physiopathology , Induced Pluripotent Stem Cells/physiology , Motor Neurons/physiology , Acetylation , Adult , Aged , Bulbo-Spinal Atrophy, X-Linked/genetics , Cells, Cultured , DNA Repeat Expansion , Female , Fibroblasts/physiology , Histone Deacetylase 6 , Histone Deacetylases/deficiency , Humans , Male , Middle Aged , Neurogenesis/physiology , Receptors, Androgen/metabolism , Tubulin/metabolism , Young Adult
14.
JCI Insight ; 9(13)2024 May 30.
Article in English | MEDLINE | ID: mdl-38973610

ABSTRACT

Spinal and bulbar muscular atrophy (SBMA) is a slowly progressing disease with limited sensitive biomarkers that support clinical research. We analyzed plasma and serum samples from patients with SBMA and matched healthy controls in multiple cohorts, identifying 40 highly reproducible SBMA-associated proteins out of nearly 3,000 measured. These proteins were robustly enriched in gene sets of skeletal muscle expression and processes related to mitochondria and calcium signaling. Many proteins outperformed currently used clinical laboratory tests (e.g., creatine kinase [CK]) in distinguishing patients from controls and in their correlations with clinical and functional traits in patients. Two of the 40 proteins, Ectodysplasin A2 receptor (EDA2R) and Repulsive guidance molecule A (RGMA), were found to be associated with decreased survival and body weight in a mouse model of SBMA. In summary, we identified what we believe to be a robust and novel set of fluid protein biomarkers in SBMA that are linked with relevant disease features in patients and in a mouse model of disease. Changes in these SBMA-associated proteins could be used as an early predictor of treatment effects in clinical trials.


Subject(s)
Biomarkers , Humans , Animals , Biomarkers/blood , Biomarkers/metabolism , Mice , Male , Female , Middle Aged , Disease Models, Animal , Muscle, Skeletal/metabolism , Adult , Case-Control Studies , Aged , GPI-Linked Proteins/blood , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism
15.
Hum Mutat ; 34(10): 1357-60, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23857908

ABSTRACT

We report here the genetic basis for a form of progressive hereditary spastic paraplegia (SPG43) previously described in two Malian sisters. Exome sequencing revealed a homozygous missense variant (c.187G>C; p.Ala63Pro) in C19orf12, a gene recently implicated in neurodegeneration with brain iron accumulation (NBIA). The same mutation was subsequently also found in a Brazilian family with features of NBIA, and we identified another NBIA patient with a three-nucleotide deletion (c.197_199del; p.Gly66del). Haplotype analysis revealed that the p.Ala63Pro mutations have a common origin, but MRI scans showed no brain iron deposition in the Malian SPG43 subjects. Heterologous expression of these SPG43 and NBIA variants resulted in similar alterations in the subcellular distribution of C19orf12. The SPG43 and NBIA variants reported here as well as the most common C19orf12 missense mutation reported in NBIA patients are found within a highly conserved, extended hydrophobic domain in C19orf12, underscoring the functional importance of this domain.


Subject(s)
Mitochondrial Proteins/genetics , Mutation , Spastic Paraplegia, Hereditary/diagnosis , Spastic Paraplegia, Hereditary/genetics , Adolescent , Amino Acid Sequence , Brain/metabolism , Brain/pathology , Homozygote , Humans , Intracellular Space/metabolism , Magnetic Resonance Imaging , Male , Mitochondrial Proteins/chemistry , Mitochondrial Proteins/metabolism , Molecular Sequence Data , Protein Transport , Sequence Alignment , Sequence Deletion , Spastic Paraplegia, Hereditary/metabolism
16.
J Neuromuscul Dis ; 10(4): 473-482, 2023.
Article in English | MEDLINE | ID: mdl-37182892

ABSTRACT

Three decades since the Human Genome Project began, scientists have now identified more then 25,000 protein coding genes in the human genome. The vast majority of the protein coding genes (> 90%) are multi-exonic, with the coding DNA being interrupted by intronic sequences, which are removed from the pre-mRNA transcripts before being translated into proteins, a process called splicing maturation. Variations in this process, i.e. by exon skipping, intron retention, alternative 5' splice site (5'ss), 3' splice site (3'ss), or polyadenylation usage, lead to remarkable transcriptome and proteome diversity in human tissues. Given its critical biological importance, alternative splicing is tightly regulated in a tissue- and developmental stage-specific manner. The central nervous system and skeletal muscle are amongst the tissues with the highest number of differentially expressed alternative exons, revealing a remarkable degree of transcriptome complexity. It is therefore not surprising that splicing mis-regulation is causally associated with a myriad of neuromuscular diseases, including but not limited to amyotrophic lateral sclerosis (ALS), spinal muscular atrophy (SMA), Duchenne muscular dystrophy (DMD), and myotonic dystrophy type 1 and 2 (DM1, DM2). A gene's transcript diversity has since become an integral and an important consideration for drug design, development and therapy. In this review, we will discuss transcript diversity in the context of neuromuscular diseases and current approaches to address splicing mis-regulation.


Subject(s)
Alternative Splicing , RNA Splice Sites , Humans , RNA Splicing/genetics , Exons , Introns
17.
Front Cell Dev Biol ; 11: 1250264, 2023.
Article in English | MEDLINE | ID: mdl-37771378

ABSTRACT

Replicative senescence is an essential cellular process playing important physiological functions, but it is better known for its implications in aging, cancer, and other pathologies. One of the main triggers of replicative senescence is telomere shortening and/or its dysfunction and, therefore, a deep understanding of the molecular determinants is crucial. However, replicative senescence is a heterogeneous and hard to study process, especially in mammalian cells, and some important questions still need an answer. These questions concern i) the exact molecular causes triggering replicative senescence, ii) the role of DNA repair mechanisms and iii) the importance of R-loops at telomeres in regulating senescence onset, and iv) the mechanisms underlying the bypass of replicative senescence. In this review, we will report and discuss recent findings about these mechanisms both in mammalian cells and in the model organism Saccharomyces cerevisiae.

18.
Cell Rep ; 42(11): 113360, 2023 11 28.
Article in English | MEDLINE | ID: mdl-38007689

ABSTRACT

DNA damage elicits a checkpoint response depending on the Mec1/ATR kinase, which detects the presence of single-stranded DNA and activates the effector kinase Rad53/CHK2. In Saccharomyces cerevisiae, one of the signaling circuits leading to Rad53 activation involves the evolutionarily conserved 9-1-1 complex, which acts as a platform for the binding of Dpb11 and Rad9 (referred to as the 9-1-1 axis) to generate a protein complex that allows Mec1 activation. By examining the effects of both loss-of-function and hypermorphic mutations, here, we show that the Cdc55 and Tpd3 subunits of the PP2A phosphatase counteract activation of the 9-1-1 axis. The lack of this inhibitory function results in DNA-damage sensitivity, sustained checkpoint-mediated cell-cycle arrest, and impaired resection of DNA double-strand breaks. This PP2A anti-checkpoint role depends on the capacity of Cdc55 to interact with Ddc1 and to counteract Ddc1-Dpb11 complex formation by preventing Dpb11 recognition of Ddc1 phosphorylated on Thr602.


Subject(s)
Protein Serine-Threonine Kinases , Saccharomyces cerevisiae Proteins , Protein Serine-Threonine Kinases/metabolism , Cell Cycle Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , DNA Damage , Phosphorylation , DNA/metabolism , Checkpoint Kinase 2/genetics
19.
Nanoscale Adv ; 5(11): 2941-2949, 2023 May 30.
Article in English | MEDLINE | ID: mdl-37260495

ABSTRACT

Nucleic acid therapeutics require delivery systems to reach their targets. Key challenges to be overcome include avoidance of accumulation in cells of the mononuclear phagocyte system and escape from the endosomal pathway. Spherical nucleic acids (SNAs), in which a gold nanoparticle supports a corona of oligonucleotides, are promising carriers for nucleic acids with valuable properties including nuclease resistance, sequence-specific loading and control of receptor-mediated endocytosis. However, SNAs accumulate in the endosomal pathway and are thus vulnerable to lysosomal degradation or recycling exocytosis. Here, an alternative SNA core based on diblock copolymer PMPC25-PDPA72 is investigated. This pH-sensitive polymer self-assembles into vesicles with an intrinsic ability to escape endosomes via osmotic shock triggered by acidification-induced disassembly. DNA oligos conjugated to PMPC25-PDPA72 molecules form vesicles, or polymersomes, with DNA coronae on luminal and external surfaces. Nucleic acid cargoes or nucleic acid-tagged targeting moieties can be attached by hybridization to the coronal DNA. These polymeric SNAs are used to deliver siRNA duplexes against C9orf72, a genetic target with therapeutic potential for amyotrophic lateral sclerosis, to motor neuron-like cells. By attaching a neuron-specific targeting peptide to the PSNA corona, effective knock-down is achieved at doses of 2 particles per cell.

20.
EMBO Mol Med ; 15(11): e17683, 2023 11 08.
Article in English | MEDLINE | ID: mdl-37724723

ABSTRACT

Spinal muscular atrophy (SMA) is a leading genetic cause of infant mortality. The advent of approved treatments for this devastating condition has significantly changed SMA patients' life expectancy and quality of life. Nevertheless, these are not without limitations, and research efforts are underway to develop new approaches for improved and long-lasting benefits for patients. Protein arginine methyltransferases (PRMTs) are emerging as druggable epigenetic targets, with several small-molecule PRMT inhibitors already in clinical trials. From a screen of epigenetic molecules, we have identified MS023, a potent and selective type I PRMT inhibitor able to promote SMN2 exon 7 inclusion in preclinical SMA models. Treatment of SMA mice with MS023 results in amelioration of the disease phenotype, with strong synergistic amplification of the positive effect when delivered in combination with the antisense oligonucleotide nusinersen. Moreover, transcriptomic analysis revealed that MS023 treatment has minimal off-target effects, and the added benefit is mainly due to targeting neuroinflammation. Our study warrants further clinical investigation of PRMT inhibition both as a stand-alone and add-on therapy for SMA.


Subject(s)
Muscular Atrophy, Spinal , Quality of Life , Animals , Humans , Infant , Mice , Exons , Muscular Atrophy, Spinal/drug therapy , Muscular Atrophy, Spinal/genetics , Oligonucleotides/pharmacology , Oligonucleotides/therapeutic use , Survival of Motor Neuron 2 Protein/genetics , Survival of Motor Neuron 2 Protein/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL