Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 71
Filter
1.
Nutrients ; 13(7)2021 Jul 13.
Article in English | MEDLINE | ID: mdl-34371910

ABSTRACT

The management of patients with spinal muscular atrophy type 1 (SMA1) is constantly evolving. In just a few decades, the medical approach has switched from an exclusively palliative therapy to a targeted therapy, transforming the natural history of the disease, improving survival time and quality of life and creating new challenges and goals. Many nutritional problems, gastrointestinal disorders and metabolic and endocrine alterations are commonly identified in patients affected by SMA1 during childhood and adolescence. For this reason, a proper pediatric multidisciplinary approach is then required in the clinical care of these patients, with a specific focus on the prevention of most common complications. The purpose of this narrative review is to provide the clinician with a practical and usable tool about SMA1 patients care, through a comprehensive insight into the nutritional, gastroenterological, metabolic and endocrine management of SMA1. Considering the possible horizons opened thanks to new therapeutic frontiers, a nutritional and endo-metabolic surveillance is a crucial element to be considered for a proper clinical care of these patients.


Subject(s)
Child Nutritional Physiological Phenomena , Energy Metabolism , Enteric Nervous System/physiopathology , Gastrointestinal Tract/innervation , Nutritional Status , Spinal Muscular Atrophies of Childhood/therapy , Adolescent , Animals , Child , Child, Preschool , Combined Modality Therapy , Dietary Supplements , Enteral Nutrition , Gastrointestinal Absorption , Humans , Infant , Neuroprotective Agents/therapeutic use , Spinal Muscular Atrophies of Childhood/metabolism , Spinal Muscular Atrophies of Childhood/physiopathology , Treatment Outcome
2.
Arch Pediatr ; 27(7S): 7S3-7S8, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33357595

ABSTRACT

Autosomal-recessive spinal muscular atrophy (SMA) is characterized by the loss of specific motor neurons of the spinal cord and skeletal muscle atrophy. SMA is caused by mutations or deletions of the survival motor neuron 1 (SMN1) gene, and disease severity correlates with the expression levels of the nearly identical copy gene, SMN2. Both genes ubiquitously express SMN protein, but SMN2 generates only low levels of protein that do not fully compensate for the loss-of-function of SMN1. SMN protein forms a multiprotein complex essential for the cellular assembly of ribonucleoprotein particles involved in diverse aspects of RNA metabolism. Other studies using animal models revealed a spatio-temporal requirement of SMN that is high during the development of the neuromuscular system and later, in the general maintenance of cellular and tissues homeostasis. These observations define a period for maximum therapeutic efficiency of SMN restoration, and suggest that cells outside the central nervous system may also participate in the pathogenesis of SMA. Finally, recent innovative therapies have been shown to mitigate SMN deficiency and have been approved to treat SMA patients. We briefly review major findings from the past twenty-five years of SMA research. © 2020 French Society of Pediatrics. Published by Elsevier Masson SAS. All rights reserved.


Subject(s)
Genetic Therapy/methods , Neuromuscular Agents/therapeutic use , Spinal Muscular Atrophies of Childhood/genetics , Spinal Muscular Atrophies of Childhood/therapy , Survival of Motor Neuron 1 Protein/genetics , Animals , Biomarkers/metabolism , Genetic Markers , Humans , Mutation , RNA/metabolism , Spinal Muscular Atrophies of Childhood/metabolism , Spinal Muscular Atrophies of Childhood/physiopathology , Survival of Motor Neuron 1 Protein/metabolism , Survival of Motor Neuron 2 Protein/genetics , Survival of Motor Neuron 2 Protein/metabolism
3.
Ann Clin Transl Neurol ; 7(7): 1158-1165, 2020 07.
Article in English | MEDLINE | ID: mdl-32558393

ABSTRACT

OBJECTIVE: Recent advances in therapeutics have improved prognosis for severely affected spinal muscular atrophy (SMA) type 1 and 2 patients, while the best method of treatment for SMA type 3 patients with later onset of disease is unknown. To better characterize the SMA type 3 population and provide potential therapeutic targets, we aimed to understand gene expression differences in whole blood of SMA type 3 patients (n = 31) and age- and gender-matched controls (n = 34). METHODS: We performed the first large-scale whole blood transcriptomic screen with L1000, a rapid, high-throughput gene expression profiling technology that uses 978 landmark genes to capture a representation of the transcriptome and predict expression of 9196 additional genes. RESULTS: The primary downregulated KEGG pathway in adult SMA type 3 patients was "Regulation of Actin Cytoskeleton," and downregulated expression of key genes in this pathway, including ROCK1, RHOA, and ACTB, was confirmed in the same whole blood samples using RT-qPCR. SMA type 3 patient-derived fibroblasts had lower expression of these genes compared to control fibroblasts from unaffected first-degree relatives. Overexpression of SMN levels using an AAV vector in fibroblasts did not normalize ROCK1, RHOA, and ACTB mRNA expression, indicating the involvement of additional genes in cytoskeleton dynamic regulation. INTERPRETATION: Our findings from whole blood and patient-derived fibroblasts suggest SMA type 3 patients have decreased expression of actin cytoskeleton regulators. These observations provide new insights and potential therapeutic targets for SMA patients with longstanding denervation and secondary musculoskeletal pathophysiology.


Subject(s)
Actin Cytoskeleton/genetics , Gene Expression Profiling , Gene Expression Regulation , Spinal Muscular Atrophies of Childhood/genetics , Spinal Muscular Atrophies of Childhood/metabolism , Adult , Family , Female , Fibroblasts , Humans , Male , Middle Aged , Spinal Muscular Atrophies of Childhood/blood
4.
Am J Clin Nutr ; 111(5): 983-996, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32145012

ABSTRACT

BACKGROUND: Knowledge on resting energy expenditure (REE) in spinal muscular atrophy type I (SMAI) is still limited. The lack of a population-specific REE equation has led to poor nutritional support and impairment of nutritional status. OBJECTIVE: To identify the best predictors of measured REE (mREE) among simple bedside parameters, to include these predictors in population-specific equations, and to compare such models with the common predictive equations. METHODS: Demographic, clinical, anthropometric, and treatment variables were examined as potential predictors of mREE by indirect calorimetry (IC) in 122 SMAI children consecutively enrolled in an ongoing longitudinal observational study. Parameters predicting REE were identified, and prespecified linear regression models adjusted for nusinersen treatment (discrete: 0 = no; 1 = yes) were used to develop predictive equations, separately in spontaneously breathing and mechanically ventilated patients. RESULTS: In naïve patients, the median (25th, 75th percentile) mREE was 480 (412, 575) compared with 394 (281, 554) kcal/d in spontaneously breathing and mechanically ventilated patients, respectively (P = 0.009).In nusinersen-treated patients, the median (25th, 75th percentile) mREE was 609 (592, 702) compared with 639 (479, 723) kcal/d in spontaneously breathing and mechanically ventilated patients, respectively (P = 0.949).Both in spontaneously breathing and mechanically ventilated patients, the best prediction of REE was obtained from 3 models, all using as predictors: 1 body size related measurement and nusinersen treatment status. Nusinersen treatment was correlated with higher REE both in spontaneously breathing and mechanically ventilated patients. The population-specific equations showed a lower interindividual variability of the bias than the other equation tested, however, they showed a high root mean squared error. CONCLUSIONS: We demonstrated that ventilatory status, nusinersen treatment, demographic, and anthropometric characteristics determine energy requirements in SMAI. Our SMAI-specific equations include variables available in clinical practice and were generally more accurate than previously published equations. At the individual level, however, IC is strongly recommended for assessing energy requirements. Further research is needed to externally validate these predictive equations.


Subject(s)
Spinal Muscular Atrophies of Childhood/metabolism , Basal Metabolism , Calorimetry, Indirect , Child , Child, Preschool , Energy Metabolism , Female , Humans , Infant , Longitudinal Studies , Male , Nutritional Requirements , Nutritional Status , Oligonucleotides/administration & dosage , Spinal Muscular Atrophies of Childhood/genetics , Spinal Muscular Atrophies of Childhood/therapy , Ventilators, Mechanical
5.
Brain ; 143(1): 69-93, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31828288

ABSTRACT

Spinal muscular atrophy (SMA) is a neuromuscular disorder caused by homozygous mutation or deletion of the survival motor neuron 1 (SMN1) gene. A second copy, SMN2, is similar to SMN1 but produces ∼10% SMN protein because of a single-point mutation that causes splicing defects. Chronic low levels of SMN cause accumulation of co-transcriptional R-loops and DNA damage leading to genomic instability and neurodegeneration in SMA. Severity of SMA disease correlates inversely with SMN levels. SMN2 is a promising target to produce higher levels of SMN by enhancing its expression. Mechanisms that regulate expression of SMN genes are largely unknown. We report that zinc finger protein ZPR1 binds to RNA polymerase II, interacts in vivo with SMN locus and upregulates SMN2 expression in SMA mice and patient cells. Modulation of ZPR1 levels directly correlates and influences SMN2 expression levels in SMA patient cells. ZPR1 overexpression in vivo results in a systemic increase of SMN levels and rescues severe to moderate disease in SMA mice. ZPR1-dependent rescue improves growth and motor function and increases the lifespan of male and female SMA mice. ZPR1 reduces neurodegeneration in SMA mice and prevents degeneration of cultured primary spinal cord neurons derived from SMA mice. Further, we show that the low levels of ZPR1 associated with SMA pathogenesis cause accumulation of co-transcriptional RNA-DNA hybrids (R-loops) and DNA damage leading to genomic instability in SMA mice and patient cells. Complementation with ZPR1 elevates senataxin levels, reduces R-loop accumulation and rescues DNA damage in SMA mice, motor neurons and patient cells. In conclusion, ZPR1 is critical for preventing accumulation of co-transcriptional R-loops and DNA damage to avert genomic instability and neurodegeneration in SMA. ZPR1 enhances SMN2 expression and leads to SMN-dependent rescue of SMA. ZPR1 represents a protective modifier and a therapeutic target for developing a new method for the treatment of SMA.


Subject(s)
DNA Damage , Fibroblasts/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Membrane Transport Proteins/genetics , R-Loop Structures , Spinal Muscular Atrophies of Childhood/genetics , Survival of Motor Neuron 1 Protein/genetics , Survival of Motor Neuron 2 Protein/genetics , Animals , DNA Helicases/metabolism , Disease Models, Animal , Female , HeLa Cells , Humans , Immunohistochemistry , In Vitro Techniques , Male , Mice , Mice, Transgenic , Motor Neurons/metabolism , Multifunctional Enzymes/metabolism , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Primary Cell Culture , RNA Helicases/metabolism , RNA Polymerase II/metabolism , Severity of Illness Index , Spinal Cord/metabolism , Spinal Cord/pathology , Spinal Muscular Atrophies of Childhood/metabolism , Spinal Muscular Atrophies of Childhood/pathology , Spinal Muscular Atrophies of Childhood/physiopathology , Survival of Motor Neuron 2 Protein/metabolism , Up-Regulation
6.
J Neurol ; 267(4): 898-912, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31776722

ABSTRACT

Only few studies have reported muscle involvement in spinal muscular atrophy using muscle MRI but this has not been systematically investigated in a large cohort of both pediatric and adult patients with type 2 and type 3 spinal muscular atrophy. The aim of the present study was to define possible patterns of muscle involvement on MRI, assessing both fatty replacement and muscle atrophy, in a cohort of type 2 and type 3 spinal muscular atrophy children and adults (age range 2-45 years), including both ambulant and non-ambulant patients. Muscle MRI protocol consisted in T1-weighted sequences acquired on axial plane covering the pelvis, the thigh, and the leg with contiguous slices. Each muscle was examined through its whole extension using a grading system that allows a semiquantitative evaluation of fatty infiltration. Thigh muscles were also grouped in anterior, posterior, and medial compartment for classification of global atrophy. The results showed a large variability in both type 2 and type 3 spinal muscular atrophy, with a various degree of proximal to distal gradient. Some muscles, such us the adductor longus and gracilis were always selectively spared. In all patients, the involvement was a combination of muscle atrophy and muscle infiltration. The variability observed may help to better understand both natural history and response to new treatments.


Subject(s)
Muscle, Skeletal/diagnostic imaging , Muscle, Skeletal/pathology , Muscular Atrophy, Spinal/diagnostic imaging , Muscular Atrophy, Spinal/pathology , Adiposity/physiology , Adolescent , Adult , Atrophy/pathology , Child , Child, Preschool , Female , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Muscle, Skeletal/metabolism , Muscular Atrophy, Spinal/metabolism , Prospective Studies , Spinal Muscular Atrophies of Childhood/diagnostic imaging , Spinal Muscular Atrophies of Childhood/metabolism , Spinal Muscular Atrophies of Childhood/pathology , Young Adult
7.
Neuromuscul Disord ; 29(7): 525-532, 2019 07.
Article in English | MEDLINE | ID: mdl-31266719

ABSTRACT

With recent advances in the treatment of Spinal Muscular Atrophy (SMA), there is a strong need to increase knowledge on the involvement of organs and systems outside the central nervous system. We investigated bone metabolism, bone mineral density (BMD) and fractures, and their possible correlation with age and motor capacities. Thirty-two children with SMA (27 type 2, 5 type 3), mean age 40 ±â€¯32.3 months, underwent two evaluations at an 18-month interval (V1 and V2). Twelve of these children also underwent a third evaluation at month 36 (V3). Diet, bone metabolism, BMD, X-rays, and motor function (by the Hammersmith Functional Motor Scale Expanded - HFMSE - and the Upper Limb Module - ULM) were assessed. At V1, 25-OH vitamin D3 (25OH D) therapy was started, and dietary calcium intake adjusted according to the recommended dietary allowance. Low 25OH D levels and asymptomatic vertebral fractures were mainly observed at V1. At all visits, bone resorption markers were higher than normal. At V2 and V3, decreased BMD was observed. Higher spine BMD values at follow-up were associated with HFMSE score >12 at baseline (p<0.03). This study suggests that even young children with SMA are at risk of severe bone fragility. Further investigations of the molecular mechanisms leading to altered bone metabolism in SMA could help identify novel therapeutic targets and establish better guidelines for bone fragility management.


Subject(s)
Bone Density , Bone and Bones/metabolism , Bone and Bones/pathology , Spinal Muscular Atrophies of Childhood/metabolism , Spinal Muscular Atrophies of Childhood/pathology , Absorptiometry, Photon , Adolescent , Bone Remodeling , Calcium, Dietary/administration & dosage , Calcium, Dietary/therapeutic use , Child , Child, Preschool , Diet , Female , Humans , Hydroxycholecalciferols/therapeutic use , Male , Movement , Prospective Studies , Recommended Dietary Allowances , Spine/metabolism , Spine/pathology , Vitamins/therapeutic use
8.
Sci Rep ; 7(1): 3672, 2017 06 16.
Article in English | MEDLINE | ID: mdl-28623256

ABSTRACT

Spinal muscular atrophy (SMA) is an autosomal recessive disorder affecting motor neurons, and is currently the most frequent genetic cause of infant mortality. SMA is caused by a loss-of-function mutation in the survival motor neuron 1 (SMN1) gene. SMN2 is an SMN1 paralogue, but cannot compensate for the loss of SMN1 since exon 7 in SMN2 mRNA is excluded (spliced out) due to a single C-to-T nucleotide transition in the exon 7. One of the most promising strategies to treat SMA is antisense oligonucleotide (AON)-mediated therapy. AONs are utilized to block intronic splicing silencer number 1 (ISS-N1) on intron 7 of SMN2, which causes exon 7 inclusion of the mRNA and the recovery of the expression of functional SMN protein from the endogenous SMN2 gene. We developed novel locked nucleic acid (LNA)-based antisense oligonucleotides (LNA/DNA mixmers), which efficiently induce exon 7 inclusion in SMN2 and restore the SMN protein production in SMA patient fibroblasts. The mixmers are highly specific to the targeted sequence, and showed significantly higher efficacy than an all-LNA oligonucleotide with the equivalent sequence. These data suggest that use of LNA/DNA mixmer-based AONs may be an attractive therapeutic strategy to treat SMA.


Subject(s)
Alternative Splicing , Fibroblasts/metabolism , Oligodeoxyribonucleotides, Antisense , Oligonucleotides , Spinal Muscular Atrophies of Childhood/genetics , Gene Expression Regulation , Humans , Spinal Muscular Atrophies of Childhood/metabolism , Survival of Motor Neuron 2 Protein/genetics , Survival of Motor Neuron 2 Protein/metabolism
9.
Orphanet J Rare Dis ; 12(1): 67, 2017 04 11.
Article in English | MEDLINE | ID: mdl-28399889

ABSTRACT

BACKGROUND: Hereditary proximal spinal muscular atrophy (SMA) is a severe neuromuscular disease of childhood caused by homozygous loss of function of the survival motor neuron (SMN) 1 gene. The presence of a second, nearly identical SMN gene (SMN2) in the human genome ensures production of residual levels of the ubiquitously expressed SMN protein. Alpha-motor neurons in the ventral horns of the spinal cord are most vulnerable to reduced SMN concentrations but the development or function of other tissues may also be affected, and cardiovascular abnormalities have frequently been reported both in patients and SMA mouse models. METHODS: We systematically reviewed reported cardiac pathology in relation to SMN deficiency. To investigate the relevance of the possible association in more detail, we used clinical classification systems to characterize structural cardiac defects and arrhythmias. CONCLUSIONS: Seventy-two studies with a total of 264 SMA patients with reported cardiac pathology were identified, along with 14 publications on SMA mouse models with abnormalities of the heart. Structural cardiac pathology, mainly septal defects and abnormalities of the cardiac outflow tract, was reported predominantly in the most severely affected patients (i.e. SMA type 1). Cardiac rhythm disorders were most frequently reported in patients with milder SMA types (e.g. SMA type 3). All included studies lacked control groups and a standardized approach for cardiac evaluation. The convergence to specific abnormalities of cardiac structure and function may indicate vulnerability of specific cell types or developmental processes relevant for cardiogenesis. Future studies would benefit from a controlled and standardized approach for cardiac evaluation in patients with SMA.


Subject(s)
Muscular Atrophy, Spinal/physiopathology , Heart/physiopathology , Humans , Motor Neurons/metabolism , Muscular Atrophy, Spinal/metabolism , Spinal Muscular Atrophies of Childhood/metabolism , Survival of Motor Neuron 1 Protein/metabolism , Survival of Motor Neuron 2 Protein/metabolism
10.
Biochim Biophys Acta Gene Regul Mech ; 1860(3): 299-315, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28095296

ABSTRACT

The multifunctional Survival Motor Neuron (SMN) protein is required for the survival of all organisms of the animal kingdom. SMN impacts various aspects of RNA metabolism through the formation and/or interaction with ribonucleoprotein (RNP) complexes. SMN regulates biogenesis of small nuclear RNPs, small nucleolar RNPs, small Cajal body-associated RNPs, signal recognition particles and telomerase. SMN also plays an important role in DNA repair, transcription, pre-mRNA splicing, histone mRNA processing, translation, selenoprotein synthesis, macromolecular trafficking, stress granule formation, cell signaling and cytoskeleton maintenance. The tissue-specific requirement of SMN is dictated by the variety and the abundance of its interacting partners. Reduced expression of SMN causes spinal muscular atrophy (SMA), a leading genetic cause of infant mortality. SMA displays a broad spectrum ranging from embryonic lethality to an adult onset. Aberrant expression and/or localization of SMN has also been associated with male infertility, inclusion body myositis, amyotrophic lateral sclerosis and osteoarthritis. This review provides a summary of various SMN functions with implications to a better understanding of SMA and other pathological conditions.


Subject(s)
Gene Expression Regulation , Survival of Motor Neuron 1 Protein/metabolism , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Animals , Coiled Bodies/genetics , Coiled Bodies/metabolism , Cytoskeleton/genetics , Cytoskeleton/metabolism , Female , Humans , Infertility, Male/genetics , Infertility, Male/metabolism , Male , Myositis, Inclusion Body/genetics , Myositis, Inclusion Body/metabolism , RNA, Small Nuclear/genetics , RNA, Small Nuclear/metabolism , Spinal Muscular Atrophies of Childhood/genetics , Spinal Muscular Atrophies of Childhood/metabolism , Survival of Motor Neuron 1 Protein/genetics
11.
J Neuromuscul Dis ; 3(3): 419-423, 2016 08 30.
Article in English | MEDLINE | ID: mdl-27854232

ABSTRACT

Ketoacidosis is an important but under-recognized complication of neuromuscular disease, in particular spinal muscular atrophy. This easily treatable condition is largely overlooked in best practice guidelines, and lack of awareness contributes to adverse outcomes in this patient population. Neuromyopathy associated ketosis should be considered in all patients with severe muscle wasting presenting with an elevated anion gap metabolic ketoacidosis. Treatment is simple, effective, and should be instituted early. Our report of a 50-year-old patient with type 2 spinal muscular atrophy who presents with recurrent ketoacidosis aims to increase awareness of neuromyopathy associated ketosis as a clinical entity, and to enhance its early recognition and timely treatment in order to improve patient outcomes.


Subject(s)
Ketosis/metabolism , Spinal Muscular Atrophies of Childhood/metabolism , Stress, Physiological , Anti-Bacterial Agents/therapeutic use , Glucose/therapeutic use , Humans , Hypoglycemia/drug therapy , Hypoglycemia/etiology , Hypoglycemia/metabolism , Ketosis/drug therapy , Ketosis/etiology , Male , Middle Aged , Spinal Muscular Atrophies of Childhood/complications , Urinary Tract Infections/complications , Urinary Tract Infections/drug therapy
12.
Neuromuscul Disord ; 26(9): 570-5, 2016 09.
Article in English | MEDLINE | ID: mdl-27450922

ABSTRACT

Biallelic mutations in IGHMBP2 cause spinal muscular atrophy with respiratory distress type 1 (SMARD1) or Charcot-Marie-Tooth type 2S (CMT2S). We report three families variably affected by IGHMBP2 mutations. Patient 1, an 8-year-old boy with two homozygous variants: c.2T>C and c.861C>G, was wheelchair bound due to sensorimotor axonal neuropathy and chronic respiratory failure. Patient 2 and his younger sister, Patient 3, had compound heterozygous variants: c.983_987delAAGAA and c.1478C>T. However, clinical phenotypes differed markedly as the elder with sensorimotor axonal neuropathy had still unaffected respiratory function at 4.5 years, whereas the younger presented as infantile spinal muscular atrophy and died from relentless respiratory failure at 11 months. Patient 4, a 6-year-old girl homozygous for IGHMBP2 c.449+1G>T documented to result in two aberrant transcripts, was wheelchair dependent due to axonal polyneuropathy. The clinical presentation in Patients 1 and 3 were consistent with SMARD1, whereas Patients 2 and 4 were in agreement with CMT2S.


Subject(s)
DNA-Binding Proteins/genetics , Mutation , Spinal Muscular Atrophies of Childhood/genetics , Spinal Muscular Atrophies of Childhood/metabolism , Transcription Factors/genetics , Child , Child, Preschool , Fatal Outcome , Female , Humans , Infant , Male , Phenotype , Respiratory Insufficiency/genetics , Respiratory Insufficiency/metabolism , Siblings
13.
PLoS One ; 10(10): e0138893, 2015.
Article in English | MEDLINE | ID: mdl-26473610

ABSTRACT

We report on the derivation of a diploid 46(XX) human embryonic stem cell (HESC) line that is homozygous for the common deletion associated with Spinal muscular atrophy type 1 (SMA) from a pathenogenetic embryo. By characterizing the methylation status of three different imprinted loci (MEST, SNRPN and H19), monitoring the expression of two parentally imprinted genes (SNRPN and H19) and carrying out genome-wide SNP analysis, we provide evidence that this cell line was established from the activation of a mutant oocyte by diploidization of the entire genome. Therefore, our SMA parthenogenetic HESC (pHESC) line provides a proof-of-principle for the establishment of diseased HESC lines without the need for gene manipulation. As mutant oocytes are easily obtained and readily available during preimplantation genetic diagnosis (PGD) cycles, this approach should provide a powerful tool for disease modelling and is especially advantageous since it can be used to induce large or complex mutations in HESCs, including gross DNA alterations and chromosomal rearrangements, which are otherwise hard to achieve.


Subject(s)
Genomic Imprinting , Homozygote , Human Embryonic Stem Cells , Mutation , Parthenogenesis , Spinal Muscular Atrophies of Childhood , Human Embryonic Stem Cells/metabolism , Human Embryonic Stem Cells/pathology , Humans , Polymorphism, Single Nucleotide , Proteins/genetics , Proteins/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Spinal Muscular Atrophies of Childhood/genetics , Spinal Muscular Atrophies of Childhood/metabolism , Spinal Muscular Atrophies of Childhood/pathology , snRNP Core Proteins/genetics , snRNP Core Proteins/metabolism
14.
Pediatr Neurol ; 52(6): 638-41, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25838041

ABSTRACT

BACKGROUND: Spinal muscular atrophy is caused by survival motor neuron gene SMN1 mutations. SMN1 produces a full-length SMN1 protein isoform encoded by exons 1-7, and an axonal-SMN protein isoform encoded by exons 1-3 and intron 3. The axonal-SMN protein is expressed only in the embryonic period and plays a significant role in axonal growth. However, there has been no report on contribution of axonal-SMN to spinal muscular atrophy severity until now. PATIENTS: Two Japanese boys with spinal muscular atrophy type 1 in our study presented with generalized muscle weakness and respiratory insufficiency soon after birth and required an artificial ventilator from early infancy. Patient 1 was compound heterozygous for two SMN1 mutations, whole-gene deletion, and an intragenic mutation (c.819_820insT). He retained one copy of SMN1 producing the N-terminal part of SMN1 including axonal-SMN. On the other hand, patient 2 was homozygous for SMN1 deletion. Both of them showed the same copy number of spinal muscular atrophy-modifying genes, NAIP and SMN2. These findings suggested that the C-terminal domain of full-length SMN1 determined the severity, irrespective of presence or absence of axonal-SMN expression. CONCLUSION: In patient 1, the C-terminal domain of full-length SMN1 determined spinal muscular atrophy severity, rather than the axonal-SMN, one copy of which could be present and intact. The presence or absence of axonal-SMN may not impact disease severity in spinal muscular atrophy type 1 patients.


Subject(s)
Axons/metabolism , Spinal Muscular Atrophies of Childhood/genetics , Survival of Motor Neuron 1 Protein/genetics , Child , DNA Mutational Analysis , Exons , Humans , Infant , Male , Mutation , Severity of Illness Index , Spinal Muscular Atrophies of Childhood/diagnosis , Spinal Muscular Atrophies of Childhood/metabolism
15.
Stem Cell Reports ; 4(4): 561-8, 2015 Apr 14.
Article in English | MEDLINE | ID: mdl-25801509

ABSTRACT

Spinal muscular atrophy (SMA) is a neuromuscular disorder caused by mutations of the survival of motor neuron 1 (SMN1) gene. In the pathogenesis of SMA, pathological changes of the neuromuscular junction (NMJ) precede the motor neuronal loss. Therefore, it is critical to evaluate the NMJ formed by SMA patients' motor neurons (MNs), and to identify drugs that can restore the normal condition. We generated NMJ-like structures using MNs derived from SMA patient-specific induced pluripotent stem cells (iPSCs), and found that the clustering of the acetylcholine receptor (AChR) is significantly impaired. Valproic acid and antisense oligonucleotide treatment ameliorated the AChR clustering defects, leading to an increase in the level of full-length SMN transcripts. Thus, the current in vitro model of AChR clustering using SMA patient-derived iPSCs is useful to dissect the pathophysiological mechanisms underlying the development of SMA, and to evaluate the efficacy of new therapeutic approaches.


Subject(s)
Induced Pluripotent Stem Cells/metabolism , Muscular Atrophy, Spinal/metabolism , Neuromuscular Junction/metabolism , Phenotype , Animals , Cell Culture Techniques , Cell Differentiation , Cell Line , Coculture Techniques , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/drug effects , Mice , Morpholinos/pharmacology , Motor Neurons/cytology , Motor Neurons/metabolism , Muscular Atrophy, Spinal/genetics , Neuromuscular Junction/drug effects , Neuromuscular Junction/pathology , Spinal Muscular Atrophies of Childhood/genetics , Spinal Muscular Atrophies of Childhood/metabolism , Valproic Acid/pharmacology
16.
J Neurosci ; 35(3): 936-42, 2015 Jan 21.
Article in English | MEDLINE | ID: mdl-25609612

ABSTRACT

Spinal muscular atrophy with pontocerebellar hypoplasia (SMA-PCH) is an infantile SMA variant with additional manifestations, particularly severe microcephaly. We previously identified a nonsense mutation in Vaccinia-related kinase 1 (VRK1), R358X, as a cause of SMA-PCH. VRK1-R358X is a rare founder mutation in Ashkenazi Jews, and additional mutations in patients of different origins have recently been identified. VRK1 is a nuclear serine/threonine protein kinase known to play multiple roles in cellular proliferation, cell cycle regulation, and carcinogenesis. However, VRK1 was not known to have neuronal functions before its identification as a gene mutated in SMA-PCH. Here we show that VRK1-R358X homozygosity results in lack of VRK1 protein, and demonstrate a role for VRK1 in neuronal migration and neuronal stem cell proliferation. Using shRNA in utero electroporation in mice, we show that Vrk1 knockdown significantly impairs cortical neuronal migration, and affects the cell cycle of neuronal progenitors. Expression of wild-type human VRK1 rescues both proliferation and migration phenotypes. However, kinase-dead human VRK1 rescues only the migration impairment, suggesting the role of VRK1 in neuronal migration is partly noncatalytic. Furthermore, we found that VRK1 deficiency in human and mouse leads to downregulation of amyloid-ß precursor protein (APP), a known neuronal migration gene. APP overexpression rescues the phenotype caused by Vrk1 knockdown, suggesting that VRK1 affects neuronal migration through an APP-dependent mechanism.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Cell Movement/genetics , Cerebellum/abnormalities , Intracellular Signaling Peptides and Proteins/metabolism , Nervous System Malformations/metabolism , Neurons/cytology , Protein Serine-Threonine Kinases/metabolism , Spinal Muscular Atrophies of Childhood/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Cell Cycle/genetics , Cell Line, Tumor , Cerebellar Diseases/genetics , Cerebellar Diseases/metabolism , Cerebellar Diseases/pathology , Cerebellum/metabolism , Cerebellum/pathology , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Developmental Disabilities/genetics , Developmental Disabilities/metabolism , Developmental Disabilities/pathology , Humans , Intracellular Signaling Peptides and Proteins/genetics , Magnetic Resonance Imaging , Mice , Nervous System Malformations/genetics , Nervous System Malformations/pathology , Neurons/metabolism , Protein Serine-Threonine Kinases/genetics , Spinal Muscular Atrophies of Childhood/genetics , Spinal Muscular Atrophies of Childhood/pathology
17.
J Pediatr ; 164(5): 1228-30, 2014 May.
Article in English | MEDLINE | ID: mdl-24423433

ABSTRACT

We investigated in children with spinal muscular atrophy type 2 the consistency of 4 different equations for predicting resting energy expenditure (REE) compared with measured REE by using indirect calorimetry. In patients with spinal muscular atrophy type 2, measured REE was lower than predicted. We also found a correlation between energy consumption and motor skills.


Subject(s)
Basal Metabolism , Spinal Muscular Atrophies of Childhood/metabolism , Adolescent , Calorimetry, Indirect/methods , Case-Control Studies , Child , Child, Preschool , Female , Humans , Infant , Male
18.
J Child Neurol ; 29(3): 374-80, 2014 Mar.
Article in English | MEDLINE | ID: mdl-23334077

ABSTRACT

Children with type I spinal muscular atrophy commonly demonstrate reduced bone mineral density. Our objectives were to evaluate and assess adequacy of vitamin D intake, serum levels, and association with bone mineral density. Assessments were completed using 3-day food records and dual energy x-ray absorptiometry scans. The spinal muscular atrophy type I cohort included 22 males and 18 females (N = 40), with a mean age of 18.6 months. Data collection occurred from 2001 to 2011. Seventy-five percent of patients had inadequate intake of vitamin D at the initial visit. Using mixed-effects analyses, vitamin D and calcium intakes correlated positively with bone mineral density (r = 0.31 and r = 0.53, respectively). Increased vitamin D and calcium consumption were associated with an increase in bone mineral density (P = .04 and P = .01, respectively). Vitamin D intake correlated positively with serum levels (r = 0.65). Further study is needed to determine optimal intakes of vitamin D and calcium in the spinal muscular atrophy type I population.


Subject(s)
Bone and Bones/metabolism , Spinal Muscular Atrophies of Childhood/metabolism , Vitamin D/administration & dosage , Absorptiometry, Photon , Adolescent , Bone Density , Calcium, Dietary/administration & dosage , Child , Child, Preschool , Cohort Studies , Diet Records , Eating , Female , Follow-Up Studies , Humans , Infant , Infant, Newborn , Male , Spinal Muscular Atrophies of Childhood/physiopathology , Vitamin D/blood
20.
Brain Pathol ; 23(6): 613-22, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23750936

ABSTRACT

Understanding the events that are responsible for a disease is mandatory for setting up a therapeutic strategy. Although spinal muscular atrophy (SMA) is considered a rare neurodegenerative pathology, its impact in our society is really devastating as it strikes young people from birth onward, and it affects their families either emotionally or financially. Moreover, it requires intensive care for the children, and this diverts both parents and relatives from their occupations. Each neuron is very different from one another; therefore, in a neurodegenerative disease, the population of axons, synapses and cell bodies degenerate asynchronously, and subpopulations of neurons have different vulnerabilities. The knowledge of the sequence of events along the lengths of individual neurons is crucial to understand if each synapse degenerates before the corresponding axon, or if each axon degenerates before the corresponding cell body. Early degeneration of one neuronal compartment in disease often reflects molecular defects somewhere else. Up until now, SMA is considered mostly a lower motor neuron disease caused by the loss-of-function mutations in the SMN1 gene; here, we inspect other features that can be altered by this defect, such as the cross talk between muscle and motor neuron and the role of physical inactivity.


Subject(s)
Spinal Muscular Atrophies of Childhood/etiology , Humans , Motor Neurons/metabolism , Neurons/metabolism , Spinal Muscular Atrophies of Childhood/genetics , Spinal Muscular Atrophies of Childhood/metabolism , Survival of Motor Neuron 1 Protein/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...