Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 165
Filter
Add more filters

Publication year range
1.
Hum Mol Genet ; 32(20): 2950-2965, 2023 10 04.
Article in English | MEDLINE | ID: mdl-37498175

ABSTRACT

Structural, functional and molecular cardiac defects have been reported in spinal muscular atrophy (SMA) patients and mouse models. Previous quantitative proteomics analyses demonstrated widespread molecular defects in the severe Taiwanese SMA mouse model. Whether such changes are conserved across different mouse models, including less severe forms of the disease, has yet to be established. Here, using the same high-resolution proteomics approach in the less-severe Smn2B/- SMA mouse model, 277 proteins were found to be differentially abundant at a symptomatic timepoint (post-natal day (P) 18), 50 of which were similarly dysregulated in severe Taiwanese SMA mice. Bioinformatics analysis linked many of the differentially abundant proteins to cardiovascular development and function, with intermediate filaments highlighted as an enriched cellular compartment in both datasets. Lamin A/C was increased in the cardiac tissue, whereas another intermediate filament protein, desmin, was reduced. The extracellular matrix (ECM) protein, elastin, was also robustly decreased in the heart of Smn2B/- mice. AAV9-SMN1-mediated gene therapy rectified low levels of survival motor neuron protein and restored desmin levels in heart tissues of Smn2B/- mice. In contrast, AAV9-SMN1 therapy failed to correct lamin A/C or elastin levels. Intermediate filament proteins and the ECM have key roles in cardiac function and their dysregulation may explain cardiac impairment in SMA, especially since mutations in genes encoding these proteins cause other diseases with cardiac aberration. Cardiac pathology may need to be considered in the long-term care of SMA patients, as it is unclear whether currently available treatments can fully rescue peripheral pathology in SMA.


Subject(s)
Motor Neurons , Muscular Atrophy, Spinal , Humans , Mice , Animals , Motor Neurons/metabolism , Desmin/genetics , Desmin/metabolism , Elastin/genetics , Lamin Type A/genetics , Lamin Type A/metabolism , Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/therapy , Muscular Atrophy, Spinal/pathology , Genetic Therapy , Disease Models, Animal , Survival of Motor Neuron 1 Protein/genetics , Survival of Motor Neuron 1 Protein/metabolism
2.
Prostate ; 84(5): 491-501, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38173273

ABSTRACT

BACKGROUND: Radical prostatectomy remains the main choice of treatment for prostate cancer. However, despite improvements in surgical techniques and neurovascular sparing procedures, rates of erectile dysfunction, and urinary incontinence remain variable. This is due, at least in part, to an incomplete understanding of neurovascular structures associated with the prostate. The objective of this study was to provide a comprehensive, detailed histological overview of the distribution of nerves and blood vessels within the prostate, facilitating subsequent correlation of prostatic neurovascular structures with patients' clinical outcomes after radical prostatectomy. METHODS: Neurovascular structures within the prostate were investigated in a total of 309 slides obtained from 15 patients who underwent non-nerve-sparing radical prostatectomy. Immunohistochemical staining was performed to identify and distinguish between parasympathetic and sympathetic nerves, whereas hematoxylin and eosin staining was used to identify blood vessels. The total number, density, and relative position of nerves and blood vessels were established using quantitative morphometry and illustrated using visualization approaches. Patient-specific outcome data were then used to establish whether the internal distribution of nerves and blood vessels within the prostate correlated with the nature and extent of complications after surgery. One-way analysis of variance tests and unpaired t tests were applied to establish statistically significant differences across the measured variables. RESULTS: Nerves and blood vessels were present across all prostatic levels and regions. However, their number and density varied considerably between regions. Assessment of the precise positioning of neurovascular structures revealed that the majority of nerve fibers were located within the dorsal and peripheral aspects of the gland. In contrast, blood vessels were predominantly located within ventral and dorsal midline regions. The number of intraprostatic nerves was found to be significantly lower in patients who recovered their continence within 12 months of surgery, compared to those whose recovery took 12 months or longer. CONCLUSION: We report an unexpected disconnect between the localization and positioning of nerve fibers and blood vessels within the prostate. Moreover, individual variability in the density of intraprostatic neurovascular structures appears to correlate with the successful recovery of urinary continence after radical prostatectomy, suggesting that differences in intrinsic neurovascular arrangements of the prostate influence postoperative outcomes.


Subject(s)
Erectile Dysfunction , Prostatic Neoplasms , Urinary Incontinence , Male , Humans , Prostate/pathology , Prostatectomy/adverse effects , Prostatectomy/methods , Erectile Dysfunction/etiology , Prostatic Neoplasms/pathology , Urinary Incontinence/etiology , Postoperative Complications/surgery
3.
Nucleic Acids Res ; 50(4): e23, 2022 02 28.
Article in English | MEDLINE | ID: mdl-34850942

ABSTRACT

Most RNA footprinting approaches that require ribonuclease cleavage generate RNA fragments bearing a phosphate or cyclic phosphate group at their 3' end. Unfortunately, current library preparation protocols rely only on a 3' hydroxyl group for adaptor ligation or poly-A tailing. Here, we developed circAID-p-seq, a PCR-free library preparation for selective 3' phospho-RNA sequencing. As a proof of concept, we applied circAID-p-seq to ribosome profiling, which is based on sequencing of RNA fragments protected by ribosomes after endonuclease digestion. CircAID-p-seq, combined with the dedicated computational pipeline circAidMe, facilitates accurate, fast and highly efficient sequencing of phospho-RNA fragments from eukaryotic cells and tissues. We used circAID-p-seq to portray ribosome occupancy in transcripts, providing a versatile and PCR-free strategy to possibly unravel any endogenous 3'-phospho RNA molecules.


Subject(s)
RNA , Ribosomes , Gene Library , High-Throughput Nucleotide Sequencing/methods , Phosphates , RNA/genetics , Ribosomes/genetics , Sequence Analysis, RNA/methods
4.
Neuropathol Appl Neurobiol ; 49(4): e12925, 2023 08.
Article in English | MEDLINE | ID: mdl-37465879

ABSTRACT

AIMS: Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease with complex aetiology. Despite evidence of neuromuscular junction (NMJ) denervation and 'dying-back' pathology in models of SOD1-dependent ALS, evidence in other genetic forms of ALS is limited by a lack of suitable animal models. TDP-43, a key mediator protein in ALS, is overexpressed in neurons in Thy1-hTDP-43WT mice. We therefore aimed to comprehensively analyse NMJ pathology in this model of ALS. METHODS: Expression of TDP-43 was assessed via western blotting. Immunohistochemistry techniques, alongside NMJ-morph quantification, were used to analyse motor neuron number, NMJ denervation status and terminal Schwann cell morphology. RESULTS: We present a time course of progressive, region-specific motor neuron pathology in Thy1-hTDP-43WT mice. Thy1-driven hTDP-43 expression increased steadily, correlating with developing hindlimb motor weakness and associated motor neuron loss in the spinal cord with a median survival of 21 days. Pronounced NMJ denervation was observed in hindlimb muscles, mild denervation in cranial muscles but no evidence of denervation in either forelimb or trunk muscles. NMJ pathology was restricted to motor nerve terminals, with denervation following the same time course as motor neuron loss. Terminal Schwann cells were lost from NMJs in hindlimb muscles, directly correlating with denervation status. CONCLUSIONS: Thy1-hTDP-43WT mice represent a severe model of ALS, with NMJ pathology/denervation of distal muscles and motor neuron loss, as observed in ALS patients. This model therefore provides an ideal platform to investigate mechanisms of dying-back pathology, as well as NMJ-targeting disease-modifying therapies in ALS.


Subject(s)
Amyotrophic Lateral Sclerosis , Neurodegenerative Diseases , Mice , Animals , Amyotrophic Lateral Sclerosis/pathology , Neurodegenerative Diseases/pathology , Neuromuscular Junction/pathology , Motor Neurons/pathology , Schwann Cells/metabolism , Schwann Cells/pathology , Denervation , DNA-Binding Proteins/metabolism , Mice, Transgenic , Disease Models, Animal
5.
Hum Mol Genet ; 29(16): 2647-2661, 2020 09 29.
Article in English | MEDLINE | ID: mdl-32686835

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is an incurable neurological disease with progressive loss of motor neuron (MN) function in the brain and spinal cord. Mutations in TARDBP, encoding the RNA-binding protein TDP-43, are one cause of ALS, and TDP-43 mislocalization in MNs is a key pathological feature of >95% of ALS cases. While numerous studies support altered RNA regulation by TDP-43 as a major cause of disease, specific changes within MNs that trigger disease onset remain unclear. Here, we combined translating ribosome affinity purification (TRAP) with RNA sequencing to identify molecular changes in spinal MNs of TDP-43-driven ALS at motor symptom onset. By comparing the MN translatome of hTDP-43A315T mice to littermate controls and to mice expressing wild type hTDP-43, we identified hundreds of mRNAs that were selectively up- or downregulated in MNs. We validated the deregulated candidates Tex26, Syngr4, and Plekhb1 mRNAs in an independent TRAP experiment. Moreover, by quantitative immunostaining of spinal cord MNs, we found corresponding protein level changes for SYNGR4 and PLEKHB1. We also observed these changes in spinal MNs of an independent ALS mouse model caused by a different patient mutant allele of TDP-43, suggesting that they are general features of TDP-43-driven ALS. Thus, we identified SYNGR4 and PLEKHB1 to be deregulated in MNs at motor symptom onset in TDP-43-driven ALS models. This spatial and temporal pattern suggests that these proteins could be functionally important for driving the transition to the symptomatic phase of the disease.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , DNA-Binding Proteins/genetics , Membrane Proteins/genetics , Synaptogyrins/genetics , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Animals , Brain/metabolism , Brain/pathology , Disease Models, Animal , Humans , Mice , Mice, Transgenic , Motor Neurons/metabolism , Motor Neurons/pathology , Protein Biosynthesis/genetics , RNA-Seq , Spinal Cord/metabolism , Spinal Cord/pathology
6.
Hum Mol Genet ; 29(14): 2365-2378, 2020 08 11.
Article in English | MEDLINE | ID: mdl-32588893

ABSTRACT

Spinal muscular atrophy (SMA) occurs as a result of cell-ubiquitous depletion of the essential survival motor neuron (SMN) protein. Characteristic disease pathology is driven by a particular vulnerability of the ventral motor neurons of the spinal cord to decreased SMN. Perhaps not surprisingly, many other organ systems are also impacted by SMN depletion. The normal kidney expresses very high levels of SMN protein, equivalent to those found in the nervous system and liver, and levels are dramatically lowered by ~90-95% in mouse models of SMA. Taken together, these data suggest that renal pathology may be present in SMA. We have addressed this using an established mouse model of severe SMA. Nephron number, as assessed by gold standard stereological techniques, was significantly reduced. In addition, morphological assessment showed decreased renal vasculature, particularly of the glomerular capillary knot, dysregulation of nephrin and collagen IV, and ultrastructural changes in the trilaminar filtration layers of the nephron. To explore the molecular drivers underpinning this process, we correlated these findings with quantitative PCR measurements and protein analyses of glial cell-line-derived neurotrophic factor, a crucial factor in ureteric bud branching and subsequent nephron development. Glial cell-line-derived neurotrophic factor levels were significantly reduced at early stages of disease in SMA mice. Collectively, these findings reveal significant renal pathology in a mouse model of severe SMA, further reinforcing the need to develop and administer systemic therapies for this neuromuscular disease.


Subject(s)
Glial Cell Line-Derived Neurotrophic Factor/genetics , Motor Neurons/metabolism , Muscular Atrophy, Spinal/genetics , Neuromuscular Diseases/genetics , Animals , Disease Models, Animal , Humans , Kidney/metabolism , Kidney/pathology , Mice , Motor Neurons/pathology , Muscular Atrophy, Spinal/metabolism , Muscular Atrophy, Spinal/pathology , Nephrons/metabolism , Nephrons/pathology , Neuromuscular Diseases/metabolism , Neuromuscular Diseases/pathology , Spinal Cord/metabolism , Spinal Cord/pathology
7.
Hum Mol Genet ; 29(16): 2674-2683, 2020 09 29.
Article in English | MEDLINE | ID: mdl-32644120

ABSTRACT

Spinal muscular atrophy (SMA) is a neuromuscular disease caused by mutations in survival motor neuron 1 (SMN1). SMN-restoring therapies have recently emerged; however, preclinical and clinical studies revealed a limited therapeutic time window and systemic aspects of the disease. This raises a fundamental question of whether SMA has presymptomatic, developmental components to disease pathogenesis. We have addressed this by combining micro-computed tomography (µCT) and comparative proteomics to examine systemic pre-symptomatic changes in a prenatal mouse model of SMA. Quantitative µCT analyses revealed that SMA embryos were significantly smaller than littermate controls, indicative of general developmental delay. More specifically, cardiac ventricles were smaller in SMA hearts, whilst liver and brain remained unaffected. In order to explore the molecular consequences of SMN depletion during development, we generated comprehensive, high-resolution, proteomic profiles of neuronal and non-neuronal organs in SMA mouse embryos. Significant molecular perturbations were observed in all organs examined, highlighting tissue-specific prenatal molecular phenotypes in SMA. Together, our data demonstrate considerable systemic changes at an early, presymptomatic stage in SMA mice, revealing a significant developmental component to SMA pathogenesis.


Subject(s)
Muscular Atrophy, Spinal/genetics , Myocardium/metabolism , Survival of Motor Neuron 1 Protein/genetics , Animals , Brain/metabolism , Disease Models, Animal , Heart/physiopathology , Humans , Liver/metabolism , Mice , Motor Neurons/metabolism , Motor Neurons/pathology , Muscular Atrophy, Spinal/diagnosis , Muscular Atrophy, Spinal/pathology , Myocardium/pathology , Phenotype , Prenatal Diagnosis , Proteomics , X-Ray Microtomography
8.
J Anat ; 240(4): 761-771, 2022 04.
Article in English | MEDLINE | ID: mdl-34725812

ABSTRACT

The glenohumeral joint is the most mobile joint in the human skeleton, supported by both active and passive stabilisers. As one of the passive stabilisers, the glenoid labrum has increasingly been recognised to play an important role in stability of the glenohumeral joint, acting to maintain intraarticular pressure, centralise the humeral head and contribute to concavity-compression stability. Several studies have investigated the macro- and micro-anatomical features of the labrum as well as its biomechanical function. However, in order to better understand the role of the labrum and its mechanics, a comprehensive anatomical, functional and biomechanical review of these studies is needed. Therefore, this article reviews the current literature detailing anatomical descriptions of the glenoid labrum, with an emphasis on its function(s) and biomechanics, as well as its interaction with neighbouring structures. The intimate relationship between the labrum and the surrounding structures was found to be important in glenohumeral stability, which owes further investigation into the microanatomy of labrum to better understand this relationship.


Subject(s)
Shoulder Joint , Biomechanical Phenomena , Cadaver , Humans , Humeral Head/anatomy & histology , Movement , Shoulder Joint/anatomy & histology
9.
J Anat ; 241(5): 1133-1147, 2022 11.
Article in English | MEDLINE | ID: mdl-36087283

ABSTRACT

Morphological study of the neuromuscular junction (NMJ), a specialised peripheral synapse formed between a lower motor neuron and skeletal muscle fibre, has significantly contributed to the understanding of synaptic biology and neuromuscular disease pathogenesis. Rodent NMJs are readily accessible, and research into conditions such as amyotrophic lateral sclerosis (ALS), Charcot-Marie-Tooth disease (CMT), and spinal muscular atrophy (SMA) has relied heavily on experimental work in these small mammals. However, given that nerve length dependency is an important feature of many peripheral neuropathies, these rodent models have clear shortcomings; large animal models might be preferable, but their size presents novel anatomical challenges. Overcoming these constraints to study the NMJ morphology of large mammalian distal limb muscles is of prime importance to increase cross-species translational neuromuscular research potential, particularly in the study of long motor units. In the past, NMJ phenotype analysis of large muscle bodies within the equine distal pelvic limb, such as the tibialis cranialis, or within muscles of high fibrous content, such as the soleus, has posed a distinct experimental hurdle. We optimised a technique for NMJ location and dissection from equine pelvic limb muscles. Using a quantification method validated in smaller species, we demonstrate their morphology and show that equine NMJs can be reliably dissected, stained and analysed. We reveal that the NMJs within the equine soleus have distinctly different morphologies when compared to the extensor digitorum longus and tibialis cranialis muscles. Overall, we demonstrate that equine distal pelvic limb muscles can be regionally dissected, with samples whole-mounted and their innervation patterns visualised. These methods will allow the localisation and analysis of neuromuscular junctions within the muscle bodies of large mammals to identify neuroanatomical and neuropathological features.


Subject(s)
Coloring Agents , Peripheral Nervous System Diseases , Animals , Horses , Mammals , Motor Neurons/pathology , Muscle Fibers, Skeletal , Muscle, Skeletal/pathology , Neuromuscular Junction/pathology , Peripheral Nervous System Diseases/pathology
10.
Acta Neuropathol ; 144(3): 437-464, 2022 09.
Article in English | MEDLINE | ID: mdl-35876881

ABSTRACT

Dysfunction and degeneration of synapses is a common feature of amyotrophic lateral sclerosis and frontotemporal dementia (ALS/FTD). A GGGGCC hexanucleotide repeat expansion in the C9ORF72 gene is the main genetic cause of ALS/FTD (C9ALS/FTD). The repeat expansion leads to reduced expression of the C9orf72 protein. How C9orf72 haploinsufficiency contributes to disease has not been resolved. Here we identify the synapsin family of synaptic vesicle proteins, the most abundant group of synaptic phosphoproteins, as novel interactors of C9orf72 at synapses and show that C9orf72 plays a cell-autonomous role in the regulation of excitatory synapses. We mapped the interaction of C9orf72 and synapsin to the N-terminal longin domain of C9orf72 and the conserved C domain of synapsin, and show interaction of the endogenous proteins in synapses. Functionally, C9orf72 deficiency reduced the number of excitatory synapses and decreased synapsin levels at remaining synapses in vitro in hippocampal neuron cultures and in vivo in the hippocampal mossy fibre system of C9orf72 knockout mice. Consistent with synaptic dysfunction, electrophysiological recordings identified impaired excitatory neurotransmission and network function in hippocampal neuron cultures with reduced C9orf72 expression, which correlated with a severe depletion of synaptic vesicles from excitatory synapses in the hippocampus of C9orf72 knockout mice. Finally, neuropathological analysis of post-mortem sections of C9ALS/FTD patient hippocampus with C9orf72 haploinsufficiency revealed a marked reduction in synapsin, indicating that disruption of the interaction between C9orf72 and synapsin may contribute to ALS/FTD pathobiology. Thus, our data show that C9orf72 plays a cell-autonomous role in the regulation of neurotransmission at excitatory synapses by interaction with synapsin and modulation of synaptic vesicle pools, and identify a novel role for C9orf72 haploinsufficiency in synaptic dysfunction in C9ALS/FTD.


Subject(s)
Amyotrophic Lateral Sclerosis , C9orf72 Protein/metabolism , Frontotemporal Dementia , Synapsins/metabolism , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Animals , C9orf72 Protein/genetics , DNA Repeat Expansion , Frontotemporal Dementia/metabolism , Frontotemporal Dementia/pathology , Mice , Mice, Knockout , Synapses/pathology
11.
Cell Mol Life Sci ; 78(10): 4785-4804, 2021 May.
Article in English | MEDLINE | ID: mdl-33821292

ABSTRACT

Spinal muscular atrophy (SMA) is an autosomal recessive motor neuron disease of variable clinical severity that is caused by mutations in the survival motor neuron 1 (SMN1) gene. Despite its name, SMN is a ubiquitous protein that functions within and outside the nervous system and has multiple cellular roles in transcription, translation, and proteostatic mechanisms. Encouragingly, several SMN-directed therapies have recently reached the clinic, albeit this has highlighted the increasing need to develop combinatorial therapies for SMA to achieve full clinical efficacy. As a subcellular site of dysfunction in SMA, mitochondria represents a relevant target for a combinatorial therapy. Accordingly, we will discuss our current understanding of mitochondrial dysfunction in SMA, highlighting mitochondrial-based pathways that offer further mechanistic insights into the involvement of mitochondria in SMA. This may ultimately facilitate translational development of targeted mitochondrial therapies for SMA. Due to clinical and mechanistic overlaps, such strategies may also benefit other motor neuron diseases and related neurodegenerative disorders.


Subject(s)
Mitochondria/physiology , Muscular Atrophy, Spinal/physiopathology , Animals , Humans , Mitochondria/genetics , Motor Neurons/physiology , Muscular Atrophy, Spinal/genetics , Mutation/genetics , SMN Complex Proteins/genetics , Signal Transduction/genetics , Signal Transduction/physiology
12.
Hum Mol Genet ; 28(21): 3515-3527, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31397869

ABSTRACT

Cardiac pathology is emerging as a prominent systemic feature of spinal muscular atrophy (SMA), but little is known about the underlying molecular pathways. Using quantitative proteomics analysis, we demonstrate widespread molecular defects in heart tissue from the Taiwanese mouse model of severe SMA. We identify increased levels of lamin A/C as a robust molecular phenotype in the heart of SMA mice and show that lamin A/C dysregulation is also apparent in SMA patient fibroblast cells and other tissues from SMA mice. Lamin A/C expression was regulated in vitro by knockdown of the E1 ubiquitination factor ubiquitin-like modifier activating enzyme 1, a key downstream mediator of SMN-dependent disease pathways, converging on ß-catenin signaling. Increased levels of lamin A are known to increase the rigidity of nuclei, inevitably disrupting contractile activity in cardiomyocytes. The increased lamin A/C levels in the hearts of SMA mice therefore provide a likely mechanism explaining morphological and functional cardiac defects, leading to blood pooling. Therapeutic strategies directed at lamin A/C may therefore offer a new approach to target cardiac pathology in SMA.


Subject(s)
Lamin Type A/metabolism , Muscular Atrophy, Spinal/metabolism , Myocardium/pathology , Animals , Disease Models, Animal , Humans , Lamin Type A/genetics , Male , Mice , Mice, Transgenic , Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/pathology , Myocardium/metabolism
13.
J Anat ; 239(5): 1221-1225, 2021 11.
Article in English | MEDLINE | ID: mdl-34633083

ABSTRACT

Teaching and learning anatomy by using human cadaveric specimens has been a foundation of medical and biomedical teaching for hundreds of years. Therefore, the majority of institutions that teach topographical anatomy rely on body donation programmes to provide specimens for both undergraduate and postgraduate teaching of gross anatomy. The COVID-19 pandemic has posed an unprecedented challenge to anatomy teaching because of the suspension of donor acceptance at most institutions. This was largely due to concerns about the potential transmissibility of the SARS-CoV-2 virus and the absence of data about the ability of embalming solutions to neutralise the virus. Twenty embalming solutions commonly used in institutions in the United Kingdom and Ireland were tested for their ability to neutralise SARS-CoV-2, using an established cytotoxicity assay. All embalming solutions tested neutralised SARS-CoV-2, with the majority of solutions being effective at high-working dilutions. These results suggest that successful embalming with the tested solutions can neutralise the SARS-CoV-2 virus, thereby facilitating the safe resumption of body donation programmes and cadaveric anatomy teaching.


Subject(s)
COVID-19/virology , Disease Transmission, Infectious/prevention & control , Embalming/methods , Formaldehyde/pharmacology , Pandemics , SARS-CoV-2 , Tissue Fixation/methods , COVID-19/transmission , Cadaver , Cells, Cultured , Fixatives/pharmacology , Humans
14.
Hum Mol Genet ; 27(16): 2851-2862, 2018 08 15.
Article in English | MEDLINE | ID: mdl-29790918

ABSTRACT

Spinal muscular atrophy (SMA) is a progressive motor neuron disease caused by deleterious variants in SMN1 that lead to a marked decrease in survival motor neuron (SMN) protein expression. Humans have a second SMN gene (SMN2) that is almost identical to SMN1. However, due to alternative splicing the majority of SMN2 messenger ribonucleic acid (mRNA) is translated into a truncated, unstable protein that is quickly degraded. Because the presence of SMN2 provides a unique opportunity for therapy development in SMA patients, the mechanisms that regulate SMN2 splicing and mRNA expression have been elucidated in great detail. In contrast, how much SMN protein is produced at different developmental time points and in different tissues remains under-characterized. In this study, we addressed this issue by determining SMN protein expression levels at three developmental time points across six different mouse tissues and in two distinct mouse models of SMA ('severe' Taiwanese and 'intermediate' Smn2B/- mice). We found that, in healthy control mice, SMN protein expression was significantly influenced by both age and tissue type. When comparing mouse models of SMA, we found that, despite being transcribed from genetically different alleles, control SMN levels were relatively similar. In contrast, the degree of SMN depletion between tissues in SMA varied substantially over time and between the two models. These findings offer an explanation for the differential vulnerability of tissues and organs observed in SMA and further our understanding of the systemic and temporal requirements for SMN with direct relevance for developing effective therapies for SMA.


Subject(s)
Muscular Atrophy, Spinal/genetics , Survival of Motor Neuron 1 Protein/genetics , Alternative Splicing/genetics , Animals , Disease Models, Animal , Exons , Humans , Mice , Motor Neurons/metabolism , Motor Neurons/pathology , Muscular Atrophy, Spinal/physiopathology , RNA Splicing/genetics , Spinal Cord/physiopathology , Survival of Motor Neuron 2 Protein/genetics
15.
J Cell Sci ; 131(8)2018 04 17.
Article in English | MEDLINE | ID: mdl-29507115

ABSTRACT

Spinal muscular atrophy (SMA) is an inherited neurodegenerative condition caused by a reduction in the amount of functional survival motor neuron (SMN) protein. SMN has been implicated in transport of mRNA in neural cells for local translation. We previously identified microtubule-dependent mobile vesicles rich in SMN and SNRPB, a member of the Sm family of small nuclear ribonucleoprotein (snRNP)-associated proteins, in neural cells. By comparing the interactomes of SNRPB and SNRPN, a neural-specific Sm protein, we now show that the essential neural protein neurochondrin (NCDN) interacts with Sm proteins and SMN in the context of mobile vesicles in neurites. NCDN has roles in protein localisation in neural cells and in maintenance of cell polarity. NCDN is required for the correct localisation of SMN, suggesting they may both be required for formation and transport of trafficking vesicles. NCDN may have potential as a therapeutic target for SMA together with, or in place of the targeting of SMN expression.This article has an associated First Person interview with the first author of the paper.


Subject(s)
Muscular Atrophy, Spinal/pathology , Nerve Tissue Proteins/metabolism , SMN Complex Proteins/metabolism , Cells, Cultured , Humans
16.
J Anat ; 237(3): 393-403, 2020 09.
Article in English | MEDLINE | ID: mdl-32628795

ABSTRACT

The outbreak of COVID-19, resulting from widespread transmission of the SARS-CoV-2 virus, represents one of the foremost current challenges to societies across the globe, with few areas of life remaining untouched. Here, we detail the immediate impact that COVID-19 has had on the teaching and practice of anatomy, providing specific examples of the varied responses from several UK, Irish and German universities and medical schools. Alongside significant issues for, and suspension of, body donation programmes, the widespread closure of university campuses has led to challenges in delivering anatomy education via online methods, a particular problem for a practical, experience-based subject such as anatomy. We discuss the short-term consequences of COVID-19 for body donation programmes and anatomical education, and highlight issues and challenges that will need to be addressed in the medium to long term in order to restore anatomy education and practice throughout the world.


Subject(s)
Anatomy/education , COVID-19 , Education, Medical , Humans , Pandemics , SARS-CoV-2 , Universities
17.
J Anat ; 237(5): 827-836, 2020 11.
Article in English | MEDLINE | ID: mdl-32573802

ABSTRACT

The neuromuscular junction (NMJ)-a synapse formed between lower motor neuron and skeletal muscle fibre-represents a major focus of both basic neuroscience research and clinical neuroscience research. Although the NMJ is known to play an important role in many neurodegenerative conditions affecting humans, the vast majority of anatomical and physiological data concerning the NMJ come from lower mammalian (e.g. rodent) animal models. However, recent findings have demonstrated major differences between the cellular anatomy and molecular anatomy of human and rodent NMJs. Therefore, we undertook a comparative morphometric analysis of the NMJ across several larger mammalian species in order to generate baseline inter-species anatomical reference data for the NMJ and to identify animal models that better represent the morphology of the human NMJ in vivo. Using a standardized morphometric platform ('NMJ-morph'), we analysed 5,385 individual NMJs from lower/pelvic limb muscles (EDL, soleus and peronei) of 6 mammalian species (mouse, cat, dog, sheep, pig and human). There was marked heterogeneity of NMJ morphology both within and between species, with no overall relationship found between NMJ morphology and muscle fibre diameter or body size. Mice had the largest NMJs on the smallest muscle fibres; cats had the smallest NMJs on the largest muscle fibres. Of all the species examined, the sheep NMJ had the most closely matched morphology to that found in humans. Taken together, we present a series of comprehensive baseline morphometric data for the mammalian NMJ and suggest that ovine models are likely to best represent the human NMJ in health and disease.


Subject(s)
Mammals/anatomy & histology , Neuromuscular Junction/anatomy & histology , Animals , Cats , Dogs , Humans , Mice
18.
PLoS Genet ; 13(4): e1006744, 2017 04.
Article in English | MEDLINE | ID: mdl-28426667

ABSTRACT

Degeneration and loss of lower motor neurons is the major pathological hallmark of spinal muscular atrophy (SMA), resulting from low levels of ubiquitously-expressed survival motor neuron (SMN) protein. One remarkable, yet unresolved, feature of SMA is that not all motor neurons are equally affected, with some populations displaying a robust resistance to the disease. Here, we demonstrate that selective vulnerability of distinct motor neuron pools arises from fundamental modifications to their basal molecular profiles. Comparative gene expression profiling of motor neurons innervating the extensor digitorum longus (disease-resistant), gastrocnemius (intermediate vulnerability), and tibialis anterior (vulnerable) muscles in mice revealed that disease susceptibility correlates strongly with a modified bioenergetic profile. Targeting of identified bioenergetic pathways by enhancing mitochondrial biogenesis rescued motor axon defects in SMA zebrafish. Moreover, targeting of a single bioenergetic protein, phosphoglycerate kinase 1 (Pgk1), was found to modulate motor neuron vulnerability in vivo. Knockdown of pgk1 alone was sufficient to partially mimic the SMA phenotype in wild-type zebrafish. Conversely, Pgk1 overexpression, or treatment with terazosin (an FDA-approved small molecule that binds and activates Pgk1), rescued motor axon phenotypes in SMA zebrafish. We conclude that global bioenergetics pathways can be therapeutically manipulated to ameliorate SMA motor neuron phenotypes in vivo.


Subject(s)
Motor Neurons/metabolism , Muscular Atrophy, Spinal/metabolism , Phosphoglycerate Kinase/genetics , Spinal Cord/metabolism , Survival of Motor Neuron 1 Protein/genetics , Adenosine Triphosphate/metabolism , Animals , Axons/metabolism , Axons/pathology , Disease Models, Animal , Disease Susceptibility , Energy Metabolism , Gene Expression Regulation, Developmental , Humans , Mice , Mitochondria/metabolism , Motor Neurons/drug effects , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/physiopathology , Phosphoglycerate Kinase/antagonists & inhibitors , Prazosin/administration & dosage , Prazosin/analogs & derivatives , Spinal Cord/growth & development , Spinal Cord/pathology , Survival of Motor Neuron 1 Protein/metabolism , Zebrafish/genetics , Zebrafish/growth & development
19.
J Cell Sci ; 130(2): 325-331, 2017 01 15.
Article in English | MEDLINE | ID: mdl-28049716

ABSTRACT

α-Synuclein plays a central role in Parkinson's disease, where it contributes to the vulnerability of synapses to degeneration. However, the downstream mechanisms through which α-synuclein controls synaptic stability and degeneration are not fully understood. Here, comparative proteomics on synapses isolated from α-synuclein-/- mouse brain identified mitochondrial proteins as primary targets of α-synuclein, revealing 37 mitochondrial proteins not previously linked to α-synuclein or neurodegeneration pathways. Of these, sideroflexin 3 (SFXN3) was found to be a mitochondrial protein localized to the inner mitochondrial membrane. Loss of SFXN3 did not disturb mitochondrial electron transport chain function in mouse synapses, suggesting that its function in mitochondria is likely to be independent of canonical bioenergetic pathways. In contrast, experimental manipulation of SFXN3 levels disrupted synaptic morphology at the Drosophila neuromuscular junction. These results provide novel insights into α-synuclein-dependent pathways, highlighting an important influence on mitochondrial proteins at the synapse, including SFXN3. We also identify SFXN3 as a new mitochondrial protein capable of regulating synaptic morphology in vivo.


Subject(s)
Cation Transport Proteins/metabolism , Drosophila Proteins/metabolism , Membrane Proteins/metabolism , Mitochondrial Proteins/metabolism , Synapses/metabolism , alpha-Synuclein/metabolism , Animals , Drosophila melanogaster/metabolism , Energy Metabolism , Gene Ontology , Humans , Mice, Inbred C57BL , Mice, Knockout , Mitochondrial Membranes/metabolism , Neuromuscular Junction/metabolism
20.
PLoS Comput Biol ; 14(8): e1006169, 2018 08.
Article in English | MEDLINE | ID: mdl-30102689

ABSTRACT

Ribosome profiling is a powerful technique used to study translation at the genome-wide level, generating unique information concerning ribosome positions along RNAs. Optimal localization of ribosomes requires the proper identification of the ribosome P-site in each ribosome protected fragment, a crucial step to determine the trinucleotide periodicity of translating ribosomes, and draw correct conclusions concerning where ribosomes are located. To determine the P-site within ribosome footprints at nucleotide resolution, the precise estimation of its offset with respect to the protected fragment is necessary. Here we present riboWaltz, an R package for calculation of optimal P-site offsets, diagnostic analysis and visual inspection of ribosome profiling data. Compared to existing tools, riboWaltz shows improved accuracies for P-site estimation and neat ribosome positioning in multiple case studies. riboWaltz was implemented in R and is available as an R package at https://github.com/LabTranslationalArchitectomics/RiboWaltz.


Subject(s)
Computational Biology/methods , Ribosomes/physiology , Sequence Analysis, RNA/methods , High-Throughput Nucleotide Sequencing/methods , Protein Biosynthesis/physiology , RNA, Messenger/metabolism , Ribosomes/metabolism , Software
SELECTION OF CITATIONS
SEARCH DETAIL