Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 40
1.
Biomolecules ; 14(3)2024 Mar 04.
Article En | MEDLINE | ID: mdl-38540722

Schizophrenia is a complex mental condition, with key symptoms marked for diagnosis including delusions, hallucinations, disorganized thinking, reduced emotional expression, and social dysfunction. In the context of major developmental hypotheses of schizophrenia, notably those concerning maternal immune activation and neuroinflammation, we studied NLRP1 expression and content in the postmortem brain tissue of 10 schizophrenia and 10 control subjects. In the medial orbitofrontal cortex (Brodmann's area 11/12) and dorsolateral prefrontal cortex (area 46) from both hemispheres of six schizophrenia subjects, the NLRP1 mRNA expression was significantly higher than in six control brains (p < 0.05). As the expression difference was highest for the medial orbitofrontal cortex in the right hemisphere, we assessed NLRP1-immunoreactive pyramidal neurons in layers III, V, and VI in the medial orbitofrontal cortex in the right hemisphere of seven schizophrenia and five control brains. Compared to controls, we quantified a significantly higher number of NLRP1-positive pyramidal neurons in the schizophrenia brains (p < 0.01), suggesting NLRP1 inflammasome activation in schizophrenia subjects. Layer III pyramidal neuron dysfunction aligns with working memory deficits, while impairments of pyramidal neurons in layers V and VI likely disrupt predictive processing. We propose NLRP1 inflammasome as a potential biomarker and therapeutic target in schizophrenia.


Schizophrenia , Humans , Inflammasomes/genetics , Inflammasomes/metabolism , Cerebral Cortex/metabolism , Prefrontal Cortex/metabolism , Pyramidal Cells/metabolism , NLR Proteins/genetics , NLR Proteins/metabolism
2.
Neuromuscul Disord ; 38: 26-41, 2024 May.
Article En | MEDLINE | ID: mdl-38554696

LMNA-related congenital muscular dystrophy (L-CMD) is caused by mutations in the LMNA gene, encoding lamin A/C. To further understand the molecular mechanisms of L-CMD, proteomic profiling using DIA mass spectrometry was conducted on immortalized myoblasts and myotubes from controls and L-CMD donors each harbouring a different LMNA mutation (R249W, del.32 K and L380S). Compared to controls, 124 and 228 differentially abundant proteins were detected in L-CMD myoblasts and myotubes, respectively, and were associated with enriched canonical pathways including synaptogenesis and necroptosis in myoblasts, and Huntington's disease and insulin secretion in myotubes. Abnormal nuclear morphology and reduced lamin A/C and emerin abundance was evident in all L-CMD cell lines compared to controls, while nucleoplasmic aggregation of lamin A/C was restricted to del.32 K cells, and mislocalization of emerin was restricted to R249W cells. Abnormal nuclear morphology indicates loss of nuclear lamina integrity as a common feature of L-CMD, likely rendering muscle cells vulnerable to mechanically induced stress, while differences between L-CMD cell lines in emerin and lamin A localization suggests that some molecular alterations in L-CMD are mutation specific. Nonetheless, identifying common proteomic alterations and molecular pathways across all three L-CMD lines has highlighted potential targets for the development of non-mutation specific therapies.


Lamin Type A , Muscular Dystrophies , Proteomics , Humans , Lamin Type A/genetics , Lamin Type A/metabolism , Muscular Dystrophies/genetics , Muscular Dystrophies/metabolism , Muscular Dystrophies/pathology , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/pathology , Mutation , Myoblasts/metabolism , Male , Cell Line , Membrane Proteins/metabolism , Membrane Proteins/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism
3.
Hum Mol Genet ; 32(20): 2950-2965, 2023 10 04.
Article En | MEDLINE | ID: mdl-37498175

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.


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
4.
Gene Ther ; 30(12): 812-825, 2023 Dec.
Article En | MEDLINE | ID: mdl-37322133

Spinal muscular atrophy (SMA) is a neuromuscular disease particularly characterised by degeneration of ventral motor neurons. Survival motor neuron (SMN) 1 gene mutations cause SMA, and gene addition strategies to replace the faulty SMN1 copy are a therapeutic option. We have developed a novel, codon-optimised hSMN1 transgene and produced integration-proficient and integration-deficient lentiviral vectors with cytomegalovirus (CMV), human synapsin (hSYN) or human phosphoglycerate kinase (hPGK) promoters to determine the optimal expression cassette configuration. Integrating, CMV-driven and codon-optimised hSMN1 lentiviral vectors resulted in the highest production of functional SMN protein in vitro. Integration-deficient lentiviral vectors also led to significant expression of the optimised transgene and are expected to be safer than integrating vectors. Lentiviral delivery in culture led to activation of the DNA damage response, in particular elevating levels of phosphorylated ataxia telangiectasia mutated (pATM) and γH2AX, but the optimised hSMN1 transgene showed some protective effects. Neonatal delivery of adeno-associated viral vector (AAV9) vector encoding the optimised transgene to the Smn2B/- mouse model of SMA resulted in a significant increase of SMN protein levels in liver and spinal cord. This work shows the potential of a novel codon-optimised hSMN1 transgene as a therapeutic strategy for SMA.


Cytomegalovirus Infections , Muscular Atrophy, Spinal , Survival of Motor Neuron 1 Protein , Animals , Humans , Infant, Newborn , Mice , Cytomegalovirus Infections/genetics , Cytomegalovirus Infections/metabolism , Disease Models, Animal , DNA, Complementary/metabolism , Motor Neurons/metabolism , Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/therapy , Survival of Motor Neuron 1 Protein/genetics , Survival of Motor Neuron 1 Protein/metabolism , Transcription Factors/genetics , Transgenes
5.
Cells ; 11(24)2022 12 15.
Article En | MEDLINE | ID: mdl-36552829

Mutations in genes encoding proteins associated with the linker of nucleoskeleton and cytoskeleton (LINC) complex within the nuclear envelope cause different diseases with varying phenotypes including skeletal muscle, cardiac, metabolic, or nervous system pathologies. There is some understanding of the structure of LINC complex-associated proteins and how they interact, but it is unclear how mutations in genes encoding them can cause the same disease, and different diseases with different phenotypes. Here, published mutations in LINC complex-associated proteins were systematically reviewed and analyzed to ascertain whether patterns exist between the genetic sequence variants and clinical phenotypes. This revealed LMNA is the only LINC complex-associated gene in which mutations commonly cause distinct conditions, and there are no clear genotype-phenotype correlations. Clusters of LMNA variants causing striated muscle disease are located in exons 1 and 6, and metabolic disease-associated LMNA variants are frequently found in the tail of lamin A/C. Additionally, exon 6 of the emerin gene, EMD, may be a mutation "hot-spot", and diseases related to SYNE1, encoding nesprin-1, are most often caused by nonsense type mutations. These results provide insight into the diverse roles of LINC-complex proteins in human disease and provide direction for future gene-targeted therapy development.


Cytoskeleton , Microtubules , Humans , Cytoskeleton/genetics , Cytoskeleton/metabolism , Nuclear Envelope/metabolism , Nuclear Matrix , Mutation/genetics
6.
Cells ; 11(17)2022 08 23.
Article En | MEDLINE | ID: mdl-36078032

Most research to characterise the molecular consequences of spinal muscular atrophy (SMA) has focused on SMA I. Here, proteomic profiling of skin fibroblasts from severe (SMA I), intermediate (SMA II), and mild (SMA III) patients, alongside age-matched controls, was conducted using SWATH mass spectrometry analysis. Differentially expressed proteomic profiles showed limited overlap across each SMA type, and variability was greatest within SMA II fibroblasts, which was not explained by SMN2 copy number. Despite limited proteomic overlap, enriched canonical pathways common to two of three SMA severities with at least one differentially expressed protein from the third included mTOR signalling, regulation of eIF2 and eIF4 signalling, and protein ubiquitination. Network expression clustering analysis identified protein profiles that may discriminate or correlate with SMA severity. From these clusters, the differential expression of PYGB (SMA I), RAB3B (SMA II), and IMP1 and STAT1 (SMA III) was verified by Western blot. All SMA fibroblasts were transfected with an SMN-enhanced construct, but only RAB3B expression in SMA II fibroblasts demonstrated an SMN-dependent response. The diverse proteomic profiles and pathways identified here pave the way for studies to determine their utility as biomarkers for patient stratification or monitoring treatment efficacy and for the identification of severity-specific treatments.


Muscular Atrophy, Spinal , Proteome , Blotting, Western , Fibroblasts/metabolism , Humans , Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/metabolism , Proteome/metabolism , Proteomics
7.
Cytokine ; 159: 156011, 2022 11.
Article En | MEDLINE | ID: mdl-36067713

With the rising demand for improved COVID-19 disease monitoring and prognostic markers, studies have aimed to identify biomarkers using a range of screening methods. However, the selection of biomarkers for validation from large datasets may result in potentially important biomarkers being overlooked when datasets are considered in isolation. Here, we have utilized a meta-summary approach to investigate COVID-19 biomarker datasets to identify conserved biomarkers of COVID-19 severity. This approach identified a panel of 17 proteins that showed a consistent direction of change across two or more datasets. Furthermore, bioinformatics analysis of these proteins highlighted a range of enriched biological processes that include inflammatory responses and compromised integrity of physiological systems including cardiovascular, neurological, and metabolic. A panel of upstream regulators of the COVID-19 severity biomarkers were identified, including chemical compounds currently under investigation for COVID-19 treatment. One of the upstream regulators, interleukin 6 (IL6), was identified as a "master regulator" of the severity biomarkers. COVID-19 disease severity is intensified due to the extreme viral immunological reaction that results in increased inflammatory biomarkers and cytokine storm. Since IL6 is the primary stimulator of cytokines, it could be used independently as a biomarker in determining COVID-19 disease progression, in addition to a potential therapeutic approach targeting IL6. The array of upstream regulators of the severity biomarkers identified here serve as attractive candidates for the development of new therapeutic approaches to treating COVID-19. In addition, the findings from this study highlight COVID-19 severity biomarkers which represent promising, robust biomarkers for future validation studies for their use in defining and monitoring disease severity and patient prognosis.


COVID-19 Drug Treatment , COVID-19 , Biomarkers , COVID-19/diagnosis , Computational Biology , Cytokines , Humans , Interleukin-6 , Severity of Illness Index
8.
Spinal Cord ; 60(4): 320-325, 2022 04.
Article En | MEDLINE | ID: mdl-34601498

STUDY DESIGN: Explanatory and mechanistic study. OBJECTIVES: A better understanding of the 'whole-body' response following spinal cord injury (SCI) is needed to guide future research aimed at developing novel therapeutic interventions and identifying prognostic indicators for SCI. This study aimed to characterise the blood proteome following contusion or complete SCI compared to a sham injury in rat models. SETTING: United Kingdom. METHODS: Pooled blood samples from one and seven days after a contusion (serum; n = 5) or from 14 days and 112 days post-complete transection SCI (plasma; n = 8) and their sham-injured counterparts were subjected to independent iTRAQ nanoflow liquid chromatography tandem mass-spectrometry proteomic analyses. Pathway analyses of the proteins that were differentially abundant between SCI and their matched sham injured counterparts were completed to indicate biological pathways that may be changed in response to SCI. RESULTS: Eleven and 42 proteins were differentially abundant (≥±2.0 FC; p ≤ 0.05) between the contusion SCI and sham injured animals at 24 h and seven days post-injury, respectively. Seven and tweleve proteins were differentially abundant between complete and sham injured rats at 14 and 112 days post-injury, respectively. Acute-phase response signalling and Liver X Receptor/Retinoic X Receptor activation were identified as differentially regulated pathways in both models of SCI. CONCLUSIONS: We have utilised longitudinal preclinical SCI models to provide an insight into the blood proteome changes that result following SCI and to highlight a number of biological pathways of interest for future studies.


Contusions , Proteome , Spinal Cord Injuries , Animals , Contusions/blood , Proteomics/methods , Rats , Spinal Cord , Spinal Cord Injuries/blood
9.
BMC Mol Cell Biol ; 21(1): 88, 2020 Dec 01.
Article En | MEDLINE | ID: mdl-33261556

BACKGROUND: Popeye domain-containing proteins 1 and 2 (POPDC1 and POPDC2) are transmembrane proteins involved in cyclic AMP-mediated signalling processes and are required for normal cardiac pacemaking and conduction. In order to identify novel protein interaction partners, POPDC1 and 2 proteins were attached to beads and compared by proteomic analysis with control beads in the pull-down of proteins from cultured human skeletal myotubes. RESULTS: There were highly-significant interactions of both POPDC1 and POPDC2 with XIRP1 (Xin actin binding repeat-containing protein 1), actin and, to a lesser degree, annexin A5. In adult human skeletal muscle, both XIRP1 and POPDC1/2 were present at the sarcolemma and in T-tubules. The interaction of POPDC1 with XIRP1 was confirmed in adult rat heart extracts. Using new monoclonal antibodies specific for POPDC1 and POPDC2, both proteins, together with XIRP1, were found mainly at intercalated discs but also at T-tubules in adult rat and human heart. CONCLUSIONS: Mutations in human POPDC1, POPDC2 and in human XIRP1, all cause pathological cardiac arrhythmias, suggesting a possible role for POPDC1/2 and XIRP1 interaction in normal cardiac conduction.


Cell Adhesion Molecules/metabolism , DNA-Binding Proteins/metabolism , Heart Diseases/metabolism , Membrane Proteins/metabolism , Muscle Proteins/metabolism , Nuclear Proteins/metabolism , Sarcolemma/metabolism , Actins/metabolism , Adult , Animals , Annexin A5/metabolism , Antibodies, Monoclonal/metabolism , COS Cells , Chlorocebus aethiops , Humans , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/metabolism , Protein Binding , Rats, Sprague-Dawley
10.
Neuromuscul Disord ; 30(6): 443-456, 2020 06.
Article En | MEDLINE | ID: mdl-32522500

Emery-Dreifuss muscular dystrophy (EDMD) is a rare genetic disorder characterised by the early development of muscle contractures, progressive muscle weakness, and heart abnormalities. The latter may result in serious complications, or in severe cases, sudden death. Currently, there are very few effective treatment options available for EDMD and so there is a high clinical need for new therapies. Various genetic mutations have been identified in the development and causation of EDMD, each encoding proteins that are components of the Linker of Nucleoskeleton and Cytoskeleton (LINC) complex, which spans the nuclear envelope and serves to connect the nuclear lamina to the cytoskeleton. Within this review, we examine how mutations in the genes encoding these proteins, including lamins A/C, emerin, nesprins 1/2, FHL1, and SUN1/2 lead to muscle cell differentiation and development pathway defects. Further work to identify conserved molecular pathways downstream of these defective proteins may reveal potential targets for therapy design.


Cell Differentiation/physiology , Gene Expression Regulation, Developmental/genetics , Membrane Proteins/genetics , Muscle Cells/physiology , Muscle Proteins/genetics , Muscular Dystrophy, Emery-Dreifuss/genetics , Muscular Dystrophy, Emery-Dreifuss/physiopathology , Nuclear Proteins/genetics , Signal Transduction/genetics , Animals , Humans
11.
Neurosci Insights ; 15: 2633105520914301, 2020.
Article En | MEDLINE | ID: mdl-32285042

Most cases of spinal muscular atrophy are caused by functional loss of the survival of motor neuron 1 (SMN1) gene, while less than 5% of cases are attributed to genes other than SMN. Mutations in LMNA, the lamin A/C encoding gene, cause an adult form of spinal muscular atrophy (SMA), and in our recent work, we highlight a role for lamin A/C in SMN-related SMA pathways. Here, we discuss this apparent molecular crosstalk between different types of SMA in context with previous work, showing that dysregulation of proteins produced by other SMA-causing genes, including UBE1, GARS, and SETX, are also implicated in SMN-related SMA pathways. The perturbation of UBE1, GARS, and lamin A/C help explain mechanisms of tissue-specific pathology in SMA, and we propose Wnt/ß-catenin signalling as a common molecular pathway on which they each converge. Therapeutic strategies directed at these proteins, or their convergent pathways, may therefore offer a new approach to targeting tissue-specific pathology in SMN-related SMA.

12.
J Neurosci Res ; 98(7): 1417-1432, 2020 07.
Article En | MEDLINE | ID: mdl-32270889

There is a strong correlation between aging and onset of idiopathic Parkinson's disease, but little is known about whether cellular changes occur during normal aging that may explain this association. Here, proteomic and bioinformatic analysis was conducted on the substantia nigra (SN) of rats at four stages of life to identify and quantify protein changes throughout aging. This analysis revealed that proteins associated with cell adhesion, protein aggregation and oxidation-reduction are dysregulated as early as middle age in rats. Glial fibrillary acidic protein (GFAP) was identified as a network hub connecting the greatest number of proteins altered during aging. Furthermore, the isoform of GFAP expressed in the SN varied throughout life. However, the expression levels of the rate-limiting enzyme for dopamine production, tyrosine hydroxylase (TH), were maintained even in the oldest animals, despite a reduction in the number of dopamine neurons in the SN pars compact(SNc) as aging progressed. This age-related increase in TH expression per neuron would likely to increase the vulnerability of neurons, since increased dopamine production would be an additional source of oxidative stress. This, in turn, would place a high demand on support systems from local astrocytes, which themselves show protein changes that could affect their functionality. Taken together, this study highlights key processes that are altered with age in the rat SN, each of which converges upon GFAP. These findings offer insight into the relationship between aging and increased challenges to neuronal viability, and indicate an important role for glial cells in the aging process.


Aging/metabolism , Dopaminergic Neurons/metabolism , Glial Fibrillary Acidic Protein/metabolism , Parkinson Disease/metabolism , Substantia Nigra/metabolism , Animals , Astrocytes/metabolism , Female , Male , Proteomics , Rats , Rats, Sprague-Dawley
13.
Sci Rep ; 9(1): 14202, 2019 10 02.
Article En | MEDLINE | ID: mdl-31578382

Nesprins, nuclear envelope spectrin-repeat proteins encoded by the SYNE1 and SYNE2 genes, are involved in localization of nuclei. The short isoform, nesprin-1-alpha2, is required for relocation of the microtubule organizer function from centromeres to the nuclear rim during myogenesis. Using specific antibodies, we now show that both nesprin-1-alpha2 and nesprin-1-giant co-localize with kinesin at the junctions of concatenated nuclei and at the outer poles of nuclear chains in human skeletal myotubes. In adult muscle, nesprin-1-alpha2 was found, together with kinesin, only on nuclei associated with neuromuscular junctions, whereas all adult cardiomyocyte nuclei expressed nesprin-1-alpha2. In a proteomics study, kinesin heavy and light chains were the only significant proteins in myotube extracts pulled down by nesprin-1-alpha2, but not by a mutant lacking the highly-conserved STAR domain (18 amino-acids, including the LEWD motif). The results support a function for nesprin-1-alpha2 in the specific localization of skeletal muscle nuclei mediated by kinesins and suggest that its primary role is at the outer nuclear membrane.


Cell Nucleus/genetics , Cytoskeletal Proteins/genetics , Kinesins/genetics , Microfilament Proteins/genetics , Muscle Development/genetics , Nerve Tissue Proteins/genetics , Animals , Gene Expression Regulation, Developmental/genetics , Humans , Kinesins/chemistry , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/growth & development , Muscle, Skeletal/metabolism , Mutation/genetics , Neuromuscular Junction/genetics , Neuromuscular Junction/growth & development , Nuclear Envelope/genetics , Nuclear Envelope/metabolism , Protein Isoforms/genetics , Proteomics
14.
Hum Mol Genet ; 28(21): 3515-3527, 2019 11 01.
Article En | MEDLINE | ID: mdl-31397869

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.


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
15.
Brain Sci ; 8(12)2018 12 04.
Article En | MEDLINE | ID: mdl-30518112

Unravelling the complex molecular pathways responsible for motor neuron degeneration in amyotrophic lateral sclerosis (ALS) and spinal muscular atrophy (SMA) remains a persistent challenge. Interest is growing in the potential molecular similarities between these two diseases, with the hope of better understanding disease pathology for the guidance of therapeutic development. The aim of this study was to conduct a comparative analysis of published proteomic studies of ALS and SMA, seeking commonly dysregulated molecules to be prioritized as future therapeutic targets. Fifteen proteins were found to be differentially expressed in two or more proteomic studies of both ALS and SMA, and bioinformatics analysis identified over-representation of proteins known to associate in vesicles and molecular pathways, including metabolism of proteins and vesicle-mediated transport-both of which converge on endoplasmic reticulum (ER)-Golgi trafficking processes. Calreticulin, a calcium-binding chaperone found in the ER, was associated with both pathways and we independently confirm that its expression was decreased in spinal cords from SMA and increased in spinal cords from ALS mice. Together, these findings offer significant insights into potential common targets that may help to guide the development of new therapies for both diseases.

16.
World Neurosurg ; 120: e902-e920, 2018 Dec.
Article En | MEDLINE | ID: mdl-30193963

BACKGROUND: Malignant infarction of the middle cerebral artery (MCI) is life threatening. It is associated with a mortality as high as 80%, and survival often at the expense of serious disability. Limited success of medical therapies has resulted in decompressive craniectomy (DC) being increasingly used as a treatment for MCI, although evidence of its efficacy is inconclusive. In this study, the efficacy of DC in improving survival, or survival free of severe disability, was assessed. METHODS: A meta-analysis was performed to approximate the efficacy of DC for treating MCI, considering age and time to surgery. A systematic literature review was conducted on Medline, Embase, and Cochrane library databases to August 1, 2018. Death and severe disability at 3, 6, 12, and 36 months follow-up were assessed, comparing best medical therapy with DC. RESULTS: 18 studies were eligible for inclusion and represented 987 individuals who received DC. Nine of these were randomized controlled trials (RCTs) (n = 374 DC). Early DC (<48 hours from onset of stroke) reduced mortality (odds ratio [OR] = 0.18, 95% confidence interval [CI] = 0.11, 0.29; P < 0.00001) but not unfavourable outcome (modified Rankin Scale [mRS] >4) (OR = 1.38, 95% CI = 0.47, 4.11; P = 0.56) at 12 months follow-up. This survival benefit was maintained regardless of age. CONCLUSION: Early DC reduces mortality but does not appear to improve favourable outcomes in patients younger or older than 60 years after MCI. RCTs incorporating quality of life assessments are warranted for MCI patients, in addition to defining the optimal timing and benefits of DC in older patients.


Decompressive Craniectomy , Infarction, Middle Cerebral Artery/surgery , Humans , Infarction, Middle Cerebral Artery/mortality
17.
Arthritis Res Ther ; 20(1): 87, 2018 05 02.
Article En | MEDLINE | ID: mdl-29720234

BACKGROUND: Autologous chondrocyte implantation (ACI) has a failure rate of approximately 20%, but it is yet to be fully understood why. Biomarkers are needed that can pre-operatively predict in which patients it is likely to fail, so that alternative or individualised therapies can be offered. We previously used label-free quantitation (LF) with a dynamic range compression proteomic approach to assess the synovial fluid (SF) of ACI responders and non-responders. However, we were able to identify only a few differentially abundant proteins at baseline. In the present study, we built upon these previous findings by assessing higher-abundance proteins within this SF, providing a more global proteomic analysis on the basis of which more of the biology underlying ACI success or failure can be understood. METHODS: Isobaric tagging for relative and absolute quantitation (iTRAQ) proteomic analysis was used to assess SF from ACI responders (mean Lysholm improvement of 33; n = 14) and non-responders (mean Lysholm decrease of 14; n = 13) at the two stages of surgery (cartilage harvest and chondrocyte implantation). Differentially abundant proteins in iTRAQ and combined iTRAQ and LF datasets were investigated using pathway and network analyses. RESULTS: iTRAQ proteomic analysis confirmed our previous finding that there is a marked proteomic shift in response to cartilage harvest (70 and 54 proteins demonstrating ≥ 2.0-fold change and p < 0.05 between stages I and II in responders and non-responders, respectively). Further, it highlighted 28 proteins that were differentially abundant between responders and non-responders to ACI, which were not found in the LF study, 16 of which were altered at baseline. The differential expression of two proteins (complement C1s subcomponent and matrix metalloproteinase 3) was confirmed biochemically. Combination of the iTRAQ and LF proteomic datasets generated in-depth SF proteome information that was used to generate interactome networks representing ACI success or failure. Functional pathways that are dysregulated in ACI non-responders were identified, including acute-phase response signalling. CONCLUSIONS: Several candidate biomarkers for baseline prediction of ACI outcome were identified. A holistic overview of the SF proteome in responders and non-responders to ACI  has been profiled, providing a better understanding of the biological pathways underlying clinical outcome, particularly the differential response to cartilage harvest in non-responders.


Chondrocytes/transplantation , Proteome/metabolism , Proteomics/methods , Synovial Fluid/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged , Outcome Assessment, Health Care/methods , Outcome Assessment, Health Care/statistics & numerical data , Protein Interaction Maps , Transplantation, Autologous , Young Adult
18.
Exp Gerontol ; 108: 181-188, 2018 07 15.
Article En | MEDLINE | ID: mdl-29704639

Cerebrospinal fluid (CSF) circulates through the brain and has a unique composition reflecting the biological processes of the brain. Identifying ageing CSF biomarkers can aid in understanding the ageing process and interpreting CSF protein changes in neurodegenerative diseases. In this study, ovine CSF proteins from young (1-2 year old), middle aged (3-6 year old) and old (7-10 year old) sheep were systemically studied. CSF proteins were labelled with iTRAQ tagging reagents and fractionated by 2-dimensional high performance, liquid chromatography. Tryptic peptides were identified using MS/MS fragmentation ions for sequencing and quantified from iTRAQ reporter ion intensities at m/z 114, 115, 116 and 117. Two hundred thirty one peptides were detected, from which 143 proteins were identified. There were 52 proteins with >25% increase in concentrations in the old sheep compared to the young. 33 of them increased >25% but <50%, 13 increased >50% but <1 fold, 6 increased >1 fold [i.e. haptoglobin (Hp), haemoglobin, neuroendocrine protein 7B2, IgM, fibrous sheath interacting protein 1, vimentin]. There were 18 proteins with >25% decrease in concentrations in the old sheep compared to the young. 17 of them decreased >25% but <50%, and histone deacetylase 7 (HDAC7) was gradually decreased for over 80%. Glutathione S-transferase was decreased in middle aged CSF compared to both young and old CSF. The differential expressions of 3 proteins (Hp, neuroendocrine protein 7B2, IgM) were confirmed by immunoassays. These data expand our current knowledge regarding ovine CSF proteins, supply the necessary information to understand the ageing process in the brain and provide a basis for diagnosis of neurodegenerative diseases.


Aging/cerebrospinal fluid , Biomarkers/cerebrospinal fluid , Peptide Fragments/cerebrospinal fluid , Proteomics , Animals , Chromatography, High Pressure Liquid , Female , Neurodegenerative Diseases/cerebrospinal fluid , Neurodegenerative Diseases/diagnosis , Sheep , Tandem Mass Spectrometry
19.
J Neuromuscul Dis ; 4(4): 357-362, 2017.
Article En | MEDLINE | ID: mdl-29172006

Although primarily characterised by loss of motor neurons from the anterior horn of spinal cord and muscle atrophy, spinal muscular atrophy (SMA) is now recognised as a multi-systemic disorder. Here, we report two SMA Type II patients with eosinophilic oesophagitis (EoE), a rare, chronic immune/antigen-mediated condition. One patient presented with dysphagia and poor weight gain, and the second patient had symptoms of gastro-oesophageal reflux (GOR) and poor weight gain. In both patients, macroscopic observations during gastroscopy indicated typical signs of EoE, which were verified during histological examination of oesophageal biopsies. Given that there is a specific treatment strategy for EoE, these cases highlight the importance of considering this condition in clinical investigations - especially for patients with SMA - who have GOR, discomfort, and oral aversion.


Eosinophilic Esophagitis/complications , Eosinophilic Esophagitis/diagnosis , Spinal Muscular Atrophies of Childhood/complications , Spinal Muscular Atrophies of Childhood/diagnosis , Adolescent , Animals , Child , Diagnosis, Differential , Disease Models, Animal , Eosinophilic Esophagitis/pathology , Eosinophilic Esophagitis/therapy , Esophagus/pathology , Female , Humans , Male , Mice, Transgenic , Spinal Muscular Atrophies of Childhood/pathology , Spinal Muscular Atrophies of Childhood/therapy
20.
Sci Rep ; 7(1): 12412, 2017 09 29.
Article En | MEDLINE | ID: mdl-28963550

Synapses are an early pathological target in many neurodegenerative diseases ranging from well-known adult onset conditions such as Alzheimer and Parkinson disease to neurodegenerative conditions of childhood such as spinal muscular atrophy (SMA) and neuronal ceroid lipofuscinosis (NCLs). However, the reasons why synapses are particularly vulnerable to such a broad range of neurodegeneration inducing stimuli remains unknown. To identify molecular modulators of synaptic stability and degeneration, we have used the Cln3 -/- mouse model of a juvenile form of NCL. We profiled and compared the molecular composition of anatomically-distinct, differentially-affected pre-synaptic populations from the Cln3 -/- mouse brain using proteomics followed by bioinformatic analyses. Identified protein candidates were then tested using a Drosophila CLN3 model to study their ability to modify the CLN3-neurodegenerative phenotype in vivo. We identified differential perturbations in a range of molecular cascades correlating with synaptic vulnerability, including valine catabolism and rho signalling pathways. Genetic and pharmacological targeting of key 'hub' proteins in such pathways was sufficient to modulate phenotypic presentation in a Drosophila CLN3 model. We propose that such a workflow provides a target rich method for the identification of novel disease regulators which could be applicable to the study of other conditions where appropriate models exist.


Membrane Glycoproteins/metabolism , Membrane Proteins/metabolism , Neuronal Ceroid-Lipofuscinoses/pathology , Neurons , Proteomics/methods , Synapses , Animals , Disease Models, Animal , Drosophila/metabolism , Drosophila Proteins/metabolism , Humans , Mice, Inbred C57BL , Neurons/metabolism , Neurons/pathology , Synapses/metabolism , Synapses/pathology
...