83 results on '"Wishart TM"'
Search Results
2. Delayed synaptic degeneration in the CNS of Wlds mice after cortical lesion.
- Author
-
Gillingwater TH, Ingham CA, Parry KE, Wright AK, Haley JE, Wishart TM, Arbuthnott GW, Ribchester RR, Gillingwater, Thomas H, Ingham, Cali A, Parry, Katherine E, Wright, Ann K, Haley, Jane E, Wishart, Thomas M, Arbuthnott, Gordon W, and Ribchester, Richard R
- Published
- 2006
- Full Text
- View/download PDF
3. Expression of the neuroprotective slow Wallerian degeneration (WldS) gene in non-neuronal tissues.
- Author
-
Wishart TM, Brownstein DG, Thomson D, Tabakova AM, Boothe KM, Tsao JW, Gillingwater TH, Wishart, Thomas M, Brownstein, David G, Thomson, Derek, Tabakova, Anca M, Boothe, Katherine M, Tsao, Jack W, and Gillingwater, Thomas H
- Abstract
Background: The slow Wallerian Degeneration (Wld(S)) gene specifically protects axonal and synaptic compartments of neurons from a wide variety of degeneration-inducing stimuli, including; traumatic injury, Parkinson's disease, demyelinating neuropathies, some forms of motor neuron disease and global cerebral ischemia. The Wld(S) gene encodes a novel Ube4b-Nmnat1 chimeric protein (Wld(S) protein) that is responsible for conferring the neuroprotective phenotype. How the chimeric Wld(S) protein confers neuroprotection remains controversial, but several studies have shown that expression in neurons in vivo and in vitro modifies key cellular pathways, including; NAD biosynthesis, ubiquitination, the mitochondrial proteome, cell cycle status and cell stress. Whether similar changes are induced in non-neuronal tissue and organs at a basal level in vivo remains to be determined. This may be of particular importance for the development and application of neuroprotective therapeutic strategies based around Wld(S)-mediated pathways designed for use in human patients.Results: We have undertaken a detailed analysis of non-neuronal Wld(S) expression in Wld(S) mice, alongside gravimetric and histological analyses, to examine the influence of Wld(S) expression in non-neuronal tissues. We show that expression of Wld(S) RNA and protein are not restricted to neuronal tissue, but that the relative RNA and protein expression levels rarely correlate in these non-neuronal tissues. We show that Wld(S) mice have normal body weight and growth characteristics as well as gravimetrically and histologically normal organs, regardless of Wld(S) protein levels. Finally, we demonstrate that previously reported Wld(S)-induced changes in cell cycle and cell stress status are neuronal-specific, not recapitulated in non-neuronal tissues at a basal level.Conclusions: We conclude that expression of Wld(S) protein has no adverse effects on non-neuronal tissue at a basal level in vivo, supporting the possibility of its safe use in future therapeutic strategies targeting axonal and/or synaptic compartments in patients with neurodegenerative disease. Future experiments determining whether Wld(S) protein can modify responses to injury in non-neuronal tissue are now required. [ABSTRACT FROM AUTHOR]- Published
- 2009
- Full Text
- View/download PDF
4. Modelling neurological diseases in large animals: Criteria for model selection and clinical assessment
- Author
-
Eaton, SL, Murdoch, F, Rzechorzek, NM, Thompson, G, Hartley, C, Blacklock, BT, Proudfoot, C, Lillico, SG, Tennant, P, Ritchie, A, Nixon, J, Brennan, PM, Guido, S, Mitchell, Nadia, Palmer, DN, Whitelaw, CBA, Cooper, JD, and Wishart, TM
- Full Text
- View/download PDF
5. ATP-binding cassette family C member 1 constrains metabolic responses to high-fat diet in male mice.
- Author
-
Villalobos E, Miguelez-Crespo A, Morgan RA, Ivatt L, Paul M, Simpson JP, Homer NZM, Kurian D, Aguilar J, Kline RA, Wishart TM, Morton NM, Stimson RH, Andrew R, Walker BR, and Nixon M
- Subjects
- Animals, Male, Mice, Mice, Knockout, Mice, Inbred C57BL, Glucose metabolism, Diet, High-Fat adverse effects, Obesity metabolism, Obesity genetics, Obesity etiology, Adipose Tissue metabolism, Insulin Resistance physiology, Corticosterone blood, Corticosterone metabolism, Muscle, Skeletal metabolism, Multidrug Resistance-Associated Proteins metabolism, Multidrug Resistance-Associated Proteins genetics
- Abstract
Glucocorticoids modulate glucose homeostasis, acting on metabolically active tissues such as liver, skeletal muscle, and adipose tissue. Intracellular regulation of glucocorticoid action in adipose tissue impacts metabolic responses to obesity. ATP-binding cassette family C member 1 (ABCC1) is a transmembrane glucocorticoid transporter known to limit the accumulation of exogenously administered corticosterone in adipose tissue. However, the role of ABCC1 in the regulation of endogenous glucocorticoid action and its impact on fuel metabolism has not been studied. Here, we investigate the impact of Abcc1 deficiency on glucocorticoid action and high-fat-diet (HFD)-induced obesity. In lean male mice, deficiency of Abcc1 increased endogenous corticosterone levels in skeletal muscle and adipose tissue but did not impact insulin sensitivity. In contrast, Abcc1-deficient male mice on HFD displayed impaired glucose and insulin tolerance, and fasting hyperinsulinaemia, without alterations in tissue corticosterone levels. Proteomics and bulk RNA sequencing revealed that Abcc1 deficiency amplified the transcriptional response to an obesogenic diet in adipose tissue but not in skeletal muscle. Moreover, Abcc1 deficiency impairs key signalling pathways related to glucose metabolism in both skeletal muscle and adipose tissue, in particular those related to OXPHOS machinery and Glut4. Together, our results highlight a role for ABCC1 in regulating glucose homeostasis, demonstrating diet-dependent effects that are not associated with altered tissue glucocorticoid concentrations.
- Published
- 2024
- Full Text
- View/download PDF
6. Hyaluronan in mesenchymal stromal cell lineage differentiation from human pluripotent stem cells: application in serum free culture.
- Author
-
De Sousa PA, Perfect L, Ye J, Samuels K, Piotrowska E, Gordon M, Mate R, Abranches E, Wishart TM, Dockrell DH, and Courtney A
- Subjects
- Humans, Culture Media, Serum-Free pharmacology, Cell Lineage, Cells, Cultured, Cell Culture Techniques methods, Coculture Techniques, Mesenchymal Stem Cells metabolism, Mesenchymal Stem Cells cytology, Hyaluronic Acid pharmacology, Hyaluronic Acid metabolism, Cell Differentiation, Pluripotent Stem Cells metabolism, Pluripotent Stem Cells cytology
- Abstract
Background: Hyaluronan (HA) is an extracellular glycosaminoglycan polysaccharide with widespread roles throughout development and in healthy and neoplastic tissues. In pluripotent stem cell culture it can support both stem cell renewal and differentiation. However, responses to HA in culture are influenced by interaction with a range of cognate factors and receptors including components of blood serum supplements, which alter results. These may contribute to variation in cell batch production yield and phenotype as well as heighten the risks of adventitious pathogen transmission in the course of cell processing for therapeutic applications. MAIN: Here we characterise differentiation of a human embryo/pluripotent stem cell derived Mesenchymal Stromal Cell (hESC/PSC-MSC)-like cell population by culture on a planar surface coated with HA in serum-free media qualified for cell production for therapy. Resulting cells met minimum criteria of the International Society for Cellular Therapy for identification as MSC by expression of. CD90, CD73, CD105, and lack of expression for CD34, CD45, CD14 and HLA-II. They were positive for other MSC associated markers (i.e.CD166, CD56, CD44, HLA 1-A) whilst negative for others (e.g. CD271, CD71, CD146). In vitro co-culture assessment of MSC associated functionality confirmed support of growth of hematopoietic progenitors and inhibition of mitogen activated proliferation of lymphocytes from umbilical cord and adult peripheral blood mononuclear cells, respectively. Co-culture with immortalized THP-1 monocyte derived macrophages (Mɸ) concurrently stimulated with lipopolysaccharide as a pro-inflammatory stimulus, resulted in a dose dependent increase in pro-inflammatory IL6 but negligible effect on TNFα. To further investigate these functionalities, a bulk cell RNA sequence comparison with adult human bone marrow derived MSC and hESC substantiated a distinctive genetic signature more proximate to the former., Conclusion: Cultivation of human pluripotent stem cells on a planar substrate of HA in serum-free culture media systems is sufficient to yield a distinctive developmental mesenchymal stromal cell lineage with potential to modify the function of haematopoietic lineages in therapeutic applications., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
7. Assessment of the alpha 7 nicotinic acetylcholine receptor as an imaging marker of cardiac repair-associated processes using NS14490.
- Author
-
Reid VJM, McLoughlin WKX, Pandya K, Stott H, Iškauskienė M, Šačkus A, Marti JA, Kurian D, Wishart TM, Lucatelli C, Peters D, Gray GA, Baker AH, Newby DE, Hadoke PWF, Tavares AAS, and MacAskill MG
- Abstract
Background: Cardiac repair and remodeling following myocardial infarction (MI) is a multifactorial process involving pro-reparative inflammation, angiogenesis and fibrosis. Noninvasive imaging using a radiotracer targeting these processes could be used to elucidate cardiac wound healing mechanisms. The alpha7 nicotinic acetylcholine receptor (ɑ7nAChR) stimulates pro-reparative macrophage activity and angiogenesis, making it a potential imaging biomarker in this context. We investigated this by assessing in vitro cellular expression of ɑ7nAChR, and by using a tritiated version of the PET radiotracer [
18 F]NS14490 in tissue autoradiography studies., Results: ɑ7nAChR expression in human monocyte-derived macrophages and vascular cells showed the highest relative expression was within macrophages, but only endothelial cells exhibited a proliferation and hypoxia-driven increase in expression. Using a mouse model of inflammatory angiogenesis following sponge implantation, specific binding of [3 H]NS14490 increased from 3.6 ± 0.2 µCi/g at day 3 post-implantation to 4.9 ± 0.2 µCi/g at day 7 (n = 4, P < 0.01), followed by a reduction at days 14 and 21. This peak matched the onset of vessel formation, macrophage infiltration and sponge fibrovascular encapsulation. In a rat MI model, specific binding of [3 H]NS14490 was low in sham and remote MI myocardium. Specific binding within the infarct increased from day 14 post-MI (33.8 ± 14.1 µCi/g, P ≤ 0.01 versus sham), peaking at day 28 (48.9 ± 5.1 µCi/g, P ≤ 0.0001 versus sham). Histological and proteomic profiling of ɑ7nAChR positive tissue revealed strong associations between ɑ7nAChR and extracellular matrix deposition, and rat cardiac fibroblasts expressed ɑ7nAChR protein under normoxic and hypoxic conditions., Conclusion: ɑ7nAChR is highly expressed in human macrophages and showed proliferation and hypoxia-driven expression in human endothelial cells. While NS14490 imaging displays a pattern that coincides with vessel formation, macrophage infiltration and fibrovascular encapsulation in the sponge model, this is not the case in the MI model where the ɑ7nAChR imaging signal was strongly associated with extracellular matrix deposition which could be explained by ɑ7nAChR expression in fibroblasts. Overall, these findings support the involvement of ɑ7nAChR across several processes central to cardiac repair, with fibrosis most closely associated with ɑ7nAChR following MI., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
8. Necroptosis inhibition counteracts neurodegeneration, memory decline, and key hallmarks of aging, promoting brain rejuvenation.
- Author
-
Arrázola MS, Lira M, Véliz-Valverde F, Quiroz G, Iqbal S, Eaton SL, Lamont DJ, Huerta H, Ureta G, Bernales S, Cárdenas JC, Cerpa W, Wishart TM, and Court FA
- Subjects
- Humans, Mice, Animals, Aged, Proteomics, Rejuvenation, Aging physiology, Brain, Memory Disorders, Necroptosis, Neurodegenerative Diseases
- Abstract
Age is the main risk factor for the development of neurodegenerative diseases. In the aged brain, axonal degeneration is an early pathological event, preceding neuronal dysfunction, and cognitive disabilities in humans, primates, rodents, and invertebrates. Necroptosis mediates degeneration of injured axons, but whether necroptosis triggers neurodegeneration and cognitive impairment along aging is unknown. Here, we show that the loss of the necroptotic effector Mlkl was sufficient to delay age-associated axonal degeneration and neuroinflammation, protecting against decreased synaptic transmission and memory decline in aged mice. Moreover, short-term pharmacologic inhibition of necroptosis targeting RIPK3 in aged mice, reverted structural and functional hippocampal impairment, both at the electrophysiological and behavioral level. Finally, a quantitative proteomic analysis revealed that necroptosis inhibition leads to an overall improvement of the aged hippocampal proteome, including a subclass of molecular biofunctions associated with brain rejuvenation, such as long-term potentiation and synaptic plasticity. Our results demonstrate that necroptosis contributes to age-dependent brain degeneration, disturbing hippocampal neuronal connectivity, and cognitive function. Therefore, necroptosis inhibition constitutes a potential geroprotective strategy to treat age-related disabilities associated with memory impairment and cognitive decline., (© 2023 The Authors. Aging Cell published by Anatomical Society and John Wiley & Sons Ltd.)
- Published
- 2023
- Full Text
- View/download PDF
9. Synaptic proteomics reveal distinct molecular signatures of cognitive change and C9ORF72 repeat expansion in the human ALS cortex.
- Author
-
Laszlo ZI, Hindley N, Sanchez Avila A, Kline RA, Eaton SL, Lamont DJ, Smith C, Spires-Jones TL, Wishart TM, and Henstridge CM
- Subjects
- Humans, C9orf72 Protein genetics, C9orf72 Protein metabolism, DNA Repeat Expansion genetics, Proteomics, Proteome genetics, Cognition, Amyotrophic Lateral Sclerosis pathology, Frontotemporal Dementia genetics
- Abstract
Increasing evidence suggests synaptic dysfunction is a central and possibly triggering factor in Amyotrophic Lateral Sclerosis (ALS). Despite this, we still know very little about the molecular profile of an ALS synapse. To address this gap, we designed a synaptic proteomics experiment to perform an unbiased assessment of the synaptic proteome in the ALS brain. We isolated synaptoneurosomes from fresh-frozen post-mortem human cortex (11 controls and 18 ALS) and stratified the ALS group based on cognitive profile (Edinburgh Cognitive and Behavioural ALS Screen (ECAS score)) and presence of a C9ORF72 hexanucleotide repeat expansion (C9ORF72-RE). This allowed us to assess regional differences and the impact of phenotype and genotype on the synaptic proteome, using Tandem Mass Tagging-based proteomics. We identified over 6000 proteins in our synaptoneurosomes and using robust bioinformatics analysis we validated the strong enrichment of synapses. We found more than 30 ALS-associated proteins in synaptoneurosomes, including TDP-43, FUS, SOD1 and C9ORF72. We identified almost 500 proteins with altered expression levels in ALS, with region-specific changes highlighting proteins and pathways with intriguing links to neurophysiology and pathology. Stratifying the ALS cohort by cognitive status revealed almost 150 specific alterations in cognitively impaired ALS synaptic preparations. Stratifying by C9ORF72-RE status revealed 330 protein alterations in the C9ORF72-RE +ve group, with KEGG pathway analysis highlighting strong enrichment for postsynaptic dysfunction, related to glutamatergic receptor signalling. We have validated some of these changes by western blot and at a single synapse level using array tomography imaging. In summary, we have generated the first unbiased map of the human ALS synaptic proteome, revealing novel insight into this key compartment in ALS pathophysiology and highlighting the influence of cognitive decline and C9ORF72-RE on synaptic composition., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
10. Cross-species efficacy of enzyme replacement therapy for CLN1 disease in mice and sheep.
- Author
-
Nelvagal HR, Eaton SL, Wang SH, Eultgen EM, Takahashi K, Le SQ, Nesbitt R, Dearborn JT, Siano N, Puhl AC, Dickson PI, Thompson G, Murdoch F, Brennan PM, Gray M, Greenhalgh SN, Tennant P, Gregson R, Clutton E, Nixon J, Proudfoot C, Guido S, Lillico SG, Whitelaw CBA, Lu JY, Hofmann SL, Ekins S, Sands MS, Wishart TM, and Cooper JD
- Subjects
- Animals, Child, Disease Models, Animal, Enzyme Replacement Therapy, Humans, Mice, Mutation, Sheep, Neuronal Ceroid-Lipofuscinoses drug therapy, Neuronal Ceroid-Lipofuscinoses genetics
- Abstract
CLN1 disease, also called infantile neuronal ceroid lipofuscinosis (NCL) or infantile Batten disease, is a fatal neurodegenerative lysosomal storage disorder resulting from mutations in the CLN1 gene encoding the soluble lysosomal enzyme palmitoyl-protein thioesterase 1 (PPT1). Therapies for CLN1 disease have proven challenging because of the aggressive disease course and the need to treat widespread areas of the brain and spinal cord. Indeed, gene therapy has proven less effective for CLN1 disease than for other similar lysosomal enzyme deficiencies. We therefore tested the efficacy of enzyme replacement therapy (ERT) by administering monthly infusions of recombinant human PPT1 (rhPPT1) to PPT1-deficient mice (Cln1-/-) and CLN1R151X sheep to assess how to potentially scale up for translation. In Cln1-/- mice, intracerebrovascular (i.c.v.) rhPPT1 delivery was the most effective route of administration, resulting in therapeutically relevant CNS levels of PPT1 activity. rhPPT1-treated mice had improved motor function, reduced disease-associated pathology, and diminished neuronal loss. In CLN1R151X sheep, i.c.v. infusions resulted in widespread rhPPT1 distribution and positive treatment effects measured by quantitative structural MRI and neuropathology. This study demonstrates the feasibility and therapeutic efficacy of i.c.v. rhPPT1 ERT. These findings represent a key step toward clinical testing of ERT in children with CLN1 disease and highlight the importance of a cross-species approach to developing a successful treatment strategy.
- Published
- 2022
- Full Text
- View/download PDF
11. An Optimized Comparative Proteomic Approach as a Tool in Neurodegenerative Disease Research.
- Author
-
Kline RA, Lößlein L, Kurian D, Aguilar Martí J, Eaton SL, Court FA, Gillingwater TH, and Wishart TM
- Subjects
- Humans, Mass Spectrometry methods, Proteome analysis, Neurodegenerative Diseases, Proteomics methods
- Abstract
Recent advances in proteomic technologies now allow unparalleled assessment of the molecular composition of a wide range of sample types. However, the application of such technologies and techniques should not be undertaken lightly. Here, we describe why the design of a proteomics experiment itself is only the first step in yielding high-quality, translatable results. Indeed, the effectiveness and/or impact of the majority of contemporary proteomics screens are hindered not by commonly considered technical limitations such as low proteome coverage but rather by insufficient analyses. Proteomic experimentation requires a careful methodological selection to account for variables from sample collection, through to database searches for peptide identification to standardised post-mass spectrometry options directed analysis workflow, which should be adjusted for each study, from determining when and how to filter proteomic data to choosing holistic versus trend-wise analyses for biologically relevant patterns. Finally, we highlight and discuss the difficulties inherent in the modelling and study of the majority of progressive neurodegenerative conditions. We provide evidence (in the context of neurodegenerative research) for the benefit of undertaking a comparative approach through the application of the above considerations in the alignment of publicly available pre-existing data sets to identify potential novel regulators of neuronal stability.
- Published
- 2022
- Full Text
- View/download PDF
12. Modelling Neurological Diseases in Large Animals: Criteria for Model Selection and Clinical Assessment.
- Author
-
Eaton SL, Murdoch F, Rzechorzek NM, Thompson G, Hartley C, Blacklock BT, Proudfoot C, Lillico SG, Tennant P, Ritchie A, Nixon J, Brennan PM, Guido S, Mitchell NL, Palmer DN, Whitelaw CBA, Cooper JD, and Wishart TM
- Subjects
- Animals, Disease Progression, Humans, Mammals, Models, Animal, Nervous System Diseases
- Abstract
Issue: The impact of neurological disorders is recognised globally, with one in six people affected in their lifetime and few treatments to slow or halt disease progression. This is due in part to the increasing ageing population, and is confounded by the high failure rate of translation from rodent-derived therapeutics to clinically effective human neurological interventions. Improved translation is demonstrated using higher order mammals with more complex/comparable neuroanatomy. These animals effectually span this translational disparity and increase confidence in factors including routes of administration/dosing and ability to scale, such that potential therapeutics will have successful outcomes when moving to patients. Coupled with advancements in genetic engineering to produce genetically tailored models, livestock are increasingly being used to bridge this translational gap. Approach: In order to aid in standardising characterisation of such models, we provide comprehensive neurological assessment protocols designed to inform on neuroanatomical dysfunction and/or lesion(s) for large animal species. We also describe the applicability of these exams in different large animals to help provide a better understanding of the practicalities of cross species neurological disease modelling. Recommendation: We would encourage the use of these assessments as a reference framework to help standardise neurological clinical scoring of large animal models.
- Published
- 2022
- Full Text
- View/download PDF
13. The Proteome Signatures of Fibroblasts from Patients with Severe, Intermediate and Mild Spinal Muscular Atrophy Show Limited Overlap.
- Author
-
Brown SJ, Kline RA, Synowsky SA, Shirran SL, Holt I, Sillence KA, Claus P, Wirth B, Wishart TM, and Fuller HR
- Subjects
- Blotting, Western, Fibroblasts metabolism, Humans, Proteomics, Muscular Atrophy, Spinal genetics, Muscular Atrophy, Spinal metabolism, Proteome metabolism
- Abstract
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.
- Published
- 2022
- Full Text
- View/download PDF
14. Effects of chronic cannabidiol in a mouse model of naturally occurring neuroinflammation, neurodegeneration, and spontaneous seizures.
- Author
-
Dearborn JT, Nelvagal HR, Rensing NR, Takahashi K, Hughes SM, Wishart TM, Cooper JD, Wong M, and Sands MS
- Subjects
- Animals, Disease Models, Animal, Mice, Neuroinflammatory Diseases, Neuronal Ceroid-Lipofuscinoses, Proteomics, Cannabidiol pharmacology, Cannabidiol therapeutic use, Graft vs Host Disease drug therapy
- Abstract
Cannabidiol (CBD) has gained attention as a therapeutic agent and is purported to have immunomodulatory, neuroprotective, and anti-seizure effects. Here, we determined the effects of chronic CBD administration in a mouse model of CLN1 disease (Cln1
-/- ) that simultaneously exhibits neuroinflammation, neurodegeneration, and spontaneous seizures. Proteomic analysis showed that putative CBD receptors are expressed at similar levels in the brains of Cln1-/- mice compared to normal animals. Cln1-/- mice received an oral dose (100 mg/kg/day) of CBD for six months and were evaluated for changes in pathological markers of disease and seizures. Chronic cannabidiol administration was well-tolerated, high levels of CBD were detected in the brain, and markers of astrocytosis and microgliosis were reduced. However, CBD had no apparent effect on seizure frequency or neuron survival. These data are consistent with CBD having immunomodulatory effects. It is possible that a higher dose of CBD could also reduce neurodegeneration and seizure frequency., (© 2022. The Author(s).)- Published
- 2022
- Full Text
- View/download PDF
15. The mitochondrial protein Sideroflexin 3 (SFXN3) influences neurodegeneration pathways in vivo.
- Author
-
Ledahawsky LM, Terzenidou ME, Edwards R, Kline RA, Graham LC, Eaton SL, van der Hoorn D, Chaytow H, Huang YT, Groen EJN, Motyl AAL, Lamont DJ, Tokatlidis K, Wishart TM, and Gillingwater TH
- Subjects
- Animals, Mice, Mitochondrial Membranes metabolism, Mitochondrial Proteins genetics, Mitochondrial Proteins metabolism, Nerve Degeneration pathology, Parkinson Disease pathology, Synapses metabolism, alpha-Synuclein genetics, alpha-Synuclein metabolism, Cation Transport Proteins metabolism, Nerve Degeneration metabolism
- Abstract
Synapses are a primary pathological target in neurodegenerative diseases. Identifying therapeutic targets at the synapse could delay progression of numerous conditions. The mitochondrial protein SFXN3 is a neuronally enriched protein expressed in synaptic terminals and regulated by key synaptic proteins, including α-synuclein. We first show that SFXN3 uses the carrier import pathway to insert into the inner mitochondrial membrane. Using high-resolution proteomics on Sfxn3-KO mice synapses, we then demonstrate that SFXN3 influences proteins and pathways associated with neurodegeneration and cell death (including CSPα and Caspase-3), as well as neurological conditions (including Parkinson's disease and Alzheimer's disease). Overexpression of SFXN3 orthologues in Drosophila models of Parkinson's disease significantly reduced dopaminergic neuron loss. In contrast, the loss of SFXN3 was insufficient to trigger neurodegeneration in mice, indicating an anti- rather than pro-neurodegeneration role for SFXN3. Taken together, these results suggest a potential role for SFXN3 in the regulation of neurodegeneration pathways., (© 2022 The Authors. The FEBS Journal published by John Wiley & Sons Ltd on behalf of Federation of European Biochemical Societies.)
- Published
- 2022
- Full Text
- View/download PDF
16. Training associated alterations in equine respiratory immunity using a multiomics comparative approach.
- Author
-
Karagianni AE, Kurian D, Cillán-Garcia E, Eaton SL, Wishart TM, and Pirie RS
- Subjects
- Animals, Gene Expression Profiling, Male, Respiratory System cytology, Horses immunology, Physical Conditioning, Animal, Proteome, Respiratory System immunology, Transcriptome
- Abstract
Neutrophilic airway inflammation is highly prevalent in racehorses in training, with the term mild to moderate equine asthma (MMEA) being applied to the majority of such cases. Our proposed study is largely derived from the strong association between MMEA in racehorses and their entry into a race training program. The objectives of this study are to characterise the effect of training on the local pulmonary immune system by defining the gene and protein expression of tracheal wash (TW) derived samples from Thoroughbred racehorses prior to and following commencement of race training. Multiomics analysis detected 2138 differentially expressed genes and 260 proteins during the training period. Gene and protein sets were enriched for biological processes related to acute phase response, oxidative stress, haemopoietic processes, as well as to immune response and inflammation. This study demonstrated TW samples to represent a rich source of airway cells, protein and RNA to study airway immunity in the horse and highlighted the benefits of a multiomics methodological approach to studying the dynamics of equine airway immunity. Findings likely reflect the known associations between race-training and both airway inflammation and bleeding, offering further insight into the potential mechanisms which underpin training associated airway inflammation., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
17. Temporal Profiling of the Cortical Synaptic Mitochondrial Proteome Identifies Ageing Associated Regulators of Stability.
- Author
-
Graham LC, Kline RA, Lamont DJ, Gillingwater TH, Mabbott NA, Skehel PA, and Wishart TM
- Subjects
- Animals, Brain metabolism, Brain pathology, Disease Models, Animal, Drosophila genetics, Drosophila physiology, Gene Expression Regulation genetics, Humans, Mice, Mitochondria genetics, Muscular Dystrophies pathology, Neuromuscular Junction genetics, Neuromuscular Junction pathology, Neurons metabolism, Aging genetics, Mitochondrial Proteins genetics, Muscular Dystrophies genetics, Proteome genetics, Synapses genetics
- Abstract
Synapses are particularly susceptible to the effects of advancing age, and mitochondria have long been implicated as organelles contributing to this compartmental vulnerability. Despite this, the mitochondrial molecular cascades promoting age-dependent synaptic demise remain to be elucidated. Here, we sought to examine how the synaptic mitochondrial proteome (including strongly mitochondrial associated proteins) was dynamically and temporally regulated throughout ageing to determine whether alterations in the expression of individual candidates can influence synaptic stability/morphology. Proteomic profiling of wild-type mouse cortical synaptic and non-synaptic mitochondria across the lifespan revealed significant age-dependent heterogeneity between mitochondrial subpopulations, with aged organelles exhibiting unique protein expression profiles. Recapitulation of aged synaptic mitochondrial protein expression at the Drosophila neuromuscular junction has the propensity to perturb the synaptic architecture, demonstrating that temporal regulation of the mitochondrial proteome may directly modulate the stability of the synapse in vivo.
- Published
- 2021
- Full Text
- View/download PDF
18. Confocal Endomicroscopy of Neuromuscular Junctions Stained with Physiologically Inert Protein Fragments of Tetanus Toxin.
- Author
-
Roesl C, Evans ER, Dissanayake KN, Boczonadi V, Jones RA, Jordan GR, Ledahawsky L, Allen GCC, Scott M, Thomson A, Wishart TM, Hughes DI, Mead RJ, Shone CC, Slater CR, Gillingwater TH, Skehel PA, and Ribchester RR
- Subjects
- Animals, Animals, Newborn, Axons drug effects, Axons metabolism, Binding Sites, Fluorescence, Green Fluorescent Proteins metabolism, Humans, Mice, Inbred C57BL, Motor Neurons drug effects, Motor Neurons metabolism, Nerve Tissue drug effects, Nerve Tissue metabolism, Neuromuscular Junction drug effects, Neuromuscular Junction pathology, Synapses drug effects, Synapses metabolism, Synaptic Transmission drug effects, Mice, Microscopy, Confocal, Neuromuscular Junction diagnostic imaging, Tetanus Toxin toxicity
- Abstract
Live imaging of neuromuscular junctions (NMJs) in situ has been constrained by the suitability of ligands for inert vital staining of motor nerve terminals. Here, we constructed several truncated derivatives of the tetanus toxin C-fragment (TetC) fused with Emerald Fluorescent Protein (emGFP). Four constructs, namely full length emGFP-TetC (emGFP-865:TetC) or truncations comprising amino acids 1066-1315 (emGFP-1066:TetC), 1093-1315 (emGFP-1093:TetC) and 1109-1315 (emGFP-1109:TetC), produced selective, high-contrast staining of motor nerve terminals in rodent or human muscle explants. Isometric tension and intracellular recordings of endplate potentials from mouse muscles indicated that neither full-length nor truncated emGFP-TetC constructs significantly impaired NMJ function or transmission. Motor nerve terminals stained with emGFP-TetC constructs were readily visualised in situ or in isolated preparations using fibre-optic confocal endomicroscopy (CEM). emGFP-TetC derivatives and CEM also visualised regenerated NMJs. Dual-waveband CEM imaging of preparations co-stained with fluorescent emGFP-TetC constructs and Alexa647-α-bungarotoxin resolved innervated from denervated NMJs in axotomized Wld
S mouse muscle and degenerating NMJs in transgenic SOD1G93A mouse muscle. Our findings highlight the region of the TetC fragment required for selective binding and visualisation of motor nerve terminals and show that fluorescent derivatives of TetC are suitable for in situ morphological and physiological characterisation of healthy, injured and diseased NMJs.- Published
- 2021
- Full Text
- View/download PDF
19. Application across species of a one health approach to liquid sample handling for respiratory based -omics analysis.
- Author
-
Karagianni AE, Eaton SL, Kurian D, Cillán-Garcia E, Twynam-Perkins J, Raper A, Wishart TM, and Pirie RS
- Subjects
- Allergy and Immunology, Animals, Asthma diagnosis, Bronchoalveolar Lavage, Bronchoalveolar Lavage Fluid, Chromatography, Liquid, Computational Biology methods, Female, Horse Diseases diagnosis, Horses, Inflammation veterinary, Macrophages metabolism, Male, Mass Spectrometry, Mice, Mice, Inbred BALB C, Species Specificity, Trachea metabolism, Trachea physiology, Genomics instrumentation, Lung metabolism, Lung physiology, Metabolomics instrumentation, One Health, Proteomics instrumentation, Respiration, Specimen Handling methods
- Abstract
Airway inflammation is highly prevalent in horses, with the majority of non-infectious cases being defined as equine asthma. Currently, cytological analysis of airway derived samples is the principal method of assessing lower airway inflammation. Samples can be obtained by tracheal wash (TW) or by lavage of the lower respiratory tract (bronchoalveolar lavage (BAL) fluid; BALF). Although BALF cytology carries significant diagnostic advantages over TW cytology for the diagnosis of equine asthma, sample acquisition is invasive, making it prohibitive for routine and sequential screening of airway health. However, recent technological advances in sample collection and processing have made it possible to determine whether a wider range of analyses might be applied to TW samples. Considering that TW samples are relatively simple to collect, minimally invasive and readily available in the horse, it was considered appropriate to investigate whether, equine tracheal secretions represent a rich source of cells and both transcriptomic and proteomic data. Similar approaches have already been applied to a comparable sample set in humans; namely, induced sputum. Sputum represents a readily available source of airway biofluids enriched in proteins, changes in the expression of which may reveal novel mechanisms in the pathogenesis of respiratory diseases, such as asthma and chronic obstructive pulmonary disease. The aim of this study was to establish a robust protocol to isolate macrophages, protein and RNA for molecular characterization of TW samples and demonstrate the applicability of sample handling to rodent and human pediatric bronchoalveolar lavage fluid isolates. TW samples provided a good quality and yield of both RNA and protein for downstream transcriptomic/proteomic analyses. The sample handling methodologies were successfully applicable to BALF for rodent and human research. TW samples represent a rich source of airway cells, and molecular analysis to facilitate and study airway inflammation, based on both transcriptomic and proteomic analysis. This study provides a necessary methodological platform for future transcriptomic and/or proteomic studies on equine lower respiratory tract secretions and BALF samples from humans and mice., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
20. Microarray profiling emphasizes transcriptomic differences between hippocampal in vivo tissue and in vitro cultures.
- Author
-
King D, Skehel PA, Dando O, Emelianova K, Barron R, and Wishart TM
- Abstract
Primary hippocampal cell cultures are routinely used as an experimentally accessible model platform for the hippocampus and brain tissue in general. Containing multiple cell types including neurons, astrocytes and microglia in a state that can be readily analysed optically, biochemically and electrophysiologically, such cultures have been used in many in vitro studies. To what extent the in vivo environment is recapitulated in primary cultures is an on-going question. Here, we compare the transcriptomic profiles of primary hippocampal cell cultures and intact hippocampal tissue. In addition, by comparing profiles from wild type and the PrP 101LL transgenic model of prion disease, we also demonstrate that gene conservation is predominantly conserved across genetically altered lines., (© The Author(s) (2021). Published by Oxford University Press on behalf of the Guarantors of Brain.)
- Published
- 2021
- Full Text
- View/download PDF
21. SMN Depleted Mice Offer a Robust and Rapid Onset Model of Nonalcoholic Fatty Liver Disease.
- Author
-
Deguise MO, Pileggi C, De Repentigny Y, Beauvais A, Tierney A, Chehade L, Michaud J, Llavero-Hurtado M, Lamont D, Atrih A, Wishart TM, Gillingwater TH, Schneider BL, Harper ME, Parson SH, and Kothary R
- Subjects
- Animals, Fatty Liver pathology, Mice, Mice, Inbred C57BL, Mice, Knockout, Non-alcoholic Fatty Liver Disease pathology, Survival of Motor Neuron 1 Protein genetics, Disease Models, Animal, Fatty Liver metabolism, Non-alcoholic Fatty Liver Disease metabolism, Survival of Motor Neuron 1 Protein metabolism
- Abstract
Background & Aims: Nonalcoholic fatty liver disease (NAFLD) is considered a health epidemic with potential devastating effects on the patients and the healthcare systems. Current preclinical models of NAFLD are invariably imperfect and generally take a long time to develop. A mouse model of survival motor neuron (SMN) depletion (Smn
2B/- mice) was recently shown to develop significant hepatic steatosis in less than 2 weeks from birth. The rapid onset of fatty liver in Smn2B/- mice provides an opportunity to identify molecular markers of NAFLD. Here, we investigated whether Smn2B/- mice display typical features of NAFLD/nonalcoholic steatohepatitis (NASH)., Methods: Biochemical, histologic, electron microscopy, proteomic, and high-resolution respirometry were used., Results: The Smn2B/- mice develop microvesicular steatohepatitis within 2 weeks, a feature prevented by AAV9-SMN gene therapy. Although fibrosis is not overtly apparent in histologic sections of the liver, there is molecular evidence of fibrogenesis and presence of stellate cell activation. The consequent liver damage arises from mitochondrial reactive oxygen species production and results in hepatic dysfunction in protein output, complement, coagulation, iron homeostasis, and insulin-like growth factor-1 metabolism. The NAFLD phenotype is likely due to non-esterified fatty acid overload from peripheral lipolysis subsequent to hyperglucagonemia compounded by reduced muscle use and insulin resistance. Despite the low hepatic mitochondrial content, isolated mitochondria show enhanced β-oxidation, likely as a compensatory response, resulting in the production of reactive oxygen species. In contrast to typical NAFLD/NASH, the Smn2B/- mice lose weight because of their associated neurological condition (spinal muscular atrophy) and develop hypoglycemia., Conclusions: The Smn2B/- mice represent a good model of microvesicular steatohepatitis. Like other models, it is not representative of the complete NAFLD/NASH spectrum. Nevertheless, it offers a reliable, low-cost, early-onset model that is not dependent on diet to identify molecular players in NAFLD pathogenesis and can serve as one of the very few models of microvesicular steatohepatitis for both adult and pediatric populations., (Copyright © 2021 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
22. Comparative anatomy of the mammalian neuromuscular junction.
- Author
-
Boehm I, Alhindi A, Leite AS, Logie C, Gibbs A, Murray O, Farrukh R, Pirie R, Proudfoot C, Clutton R, Wishart TM, Jones RA, and Gillingwater TH
- Subjects
- Animals, Cats, Dogs, Humans, Mice, Mammals anatomy & histology, Neuromuscular Junction anatomy & histology
- 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., (© 2020 The Authors. Journal of Anatomy published by John Wiley & Sons Ltd on behalf of Anatomical Society.)
- Published
- 2020
- Full Text
- View/download PDF
23. Collateral Sprouting of Peripheral Sensory Neurons Exhibits a Unique Transcriptomic Profile.
- Author
-
Lemaitre D, Hurtado ML, De Gregorio C, Oñate M, Martínez G, Catenaccio A, Wishart TM, and Court FA
- Subjects
- Animals, Cell Proliferation, Female, Ganglia, Spinal pathology, Gene Expression Regulation, Lumbar Vertebrae pathology, Mice, Inbred C57BL, Myelin Sheath metabolism, Peripheral Nerve Injuries genetics, Peripheral Nerve Injuries pathology, Peripheral Nerves ultrastructure, Sciatic Nerve metabolism, Sciatic Nerve pathology, Sensory Receptor Cells ultrastructure, Wallerian Degeneration genetics, Wallerian Degeneration pathology, Gene Expression Profiling, Neurogenesis genetics, Peripheral Nerves physiology, Sensory Receptor Cells physiology, Transcriptome genetics
- Abstract
Peripheral nerve injuries result in motor and sensory dysfunction which can be recovered by compensatory or regenerative processes. In situations where axonal regeneration of injured neurons is hampered, compensation by collateral sprouting from uninjured neurons contributes to target reinnervation and functional recovery. Interestingly, this process of collateral sprouting from uninjured neurons has been associated with the activation of growth-associated programs triggered by Wallerian degeneration. Nevertheless, the molecular alterations at the transcriptomic level associated with these compensatory growth mechanisms remain to be fully elucidated. We generated a surgical model of partial sciatic nerve injury in mice to mechanistically study degeneration-induced collateral sprouting from spared fibers in the peripheral nervous system. Using next-generation sequencing and Ingenuity Pathway Analysis, we described the sprouting-associated transcriptome of uninjured sensory neurons and compare it with the activated by regenerating neurons. In vitro approaches were used to functionally assess sprouting gene candidates in the mechanisms of axonal growth. Using a novel animal model, we provide the first description of the sprouting transcriptome observed in uninjured sensory neurons after nerve injury. This collateral sprouting-associated transcriptome differs from that seen in regenerating neurons, suggesting a molecular program distinct from axonal growth. We further demonstrate that genetic upregulation of novel sprouting-associated genes activates a specific growth program in vitro, leading to increased neuronal branching. These results contribute to our understanding of the molecular mechanisms associated with collateral sprouting in vivo. The data provided here will therefore be instrumental in developing therapeutic strategies aimed at promoting functional recovery after injury to the nervous system.
- Published
- 2020
- Full Text
- View/download PDF
24. Pre-natal manifestation of systemic developmental abnormalities in spinal muscular atrophy.
- Author
-
Motyl AAL, Faller KME, Groen EJN, Kline RA, Eaton SL, Ledahawsky LM, Chaytow H, Lamont DJ, Wishart TM, Huang YT, and Gillingwater TH
- Subjects
- 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, Muscular Atrophy, Spinal genetics, Myocardium metabolism, Survival of Motor Neuron 1 Protein genetics
- 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., (© The Author(s) 2020. Published by Oxford University Press.)
- Published
- 2020
- Full Text
- View/download PDF
25. Comparative proteomic profiling reveals mechanisms for early spinal cord vulnerability in CLN1 disease.
- Author
-
Nelvagal HR, Hurtado ML, Eaton SL, Kline RA, Lamont DJ, Sands MS, Wishart TM, and Cooper JD
- Subjects
- Animals, Disease Models, Animal, Disease Progression, Humans, Mice, Mice, Inbred C57BL, Mice, Knockout, Neuronal Ceroid-Lipofuscinoses pathology, Protein Array Analysis, Proteome genetics, Proteome metabolism, Spinal Cord pathology, Thiolester Hydrolases deficiency, Membrane Proteins genetics, Neuronal Ceroid-Lipofuscinoses genetics, Neuronal Ceroid-Lipofuscinoses metabolism, Spinal Cord metabolism, Thiolester Hydrolases genetics
- Abstract
CLN1 disease is a fatal inherited neurodegenerative lysosomal storage disease of early childhood, caused by mutations in the CLN1 gene, which encodes the enzyme Palmitoyl protein thioesterase-1 (PPT-1). We recently found significant spinal pathology in Ppt1-deficient (Ppt1
-/- ) mice and human CLN1 disease that contributes to clinical outcome and precedes the onset of brain pathology. Here, we quantified this spinal pathology at 3 and 7 months of age revealing significant and progressive glial activation and vulnerability of spinal interneurons. Tandem mass tagged proteomic analysis of the spinal cord of Ppt1-/- and control mice at these timepoints revealed a significant neuroimmune response and changes in mitochondrial function, cell-signalling pathways and developmental processes. Comparing proteomic changes in the spinal cord and cortex at 3 months revealed many similarly affected processes, except the inflammatory response. These proteomic and pathological data from this largely unexplored region of the CNS may help explain the limited success of previous brain-directed therapies. These data also fundamentally change our understanding of the progressive, site-specific nature of CLN1 disease pathogenesis, and highlight the importance of the neuroimmune response. This should greatly impact our approach to the timing and targeting of future therapeutic trials for this and similar disorders.- Published
- 2020
- Full Text
- View/download PDF
26. Applying modern Omic technologies to the Neuronal Ceroid Lipofuscinoses.
- Author
-
Kline RA, Wishart TM, Mills K, and Heywood WE
- Subjects
- Animals, Biomarkers, Humans, Genomics, Neuronal Ceroid-Lipofuscinoses genetics, Neuronal Ceroid-Lipofuscinoses metabolism, Proteomics
- Abstract
The Neuronal Ceroid Lipofuscinoses are a group of severe and progressive neurodegenerative disorders, which generally present during childhood. With new treatments emerging on the horizon, there is a growing need to understand the specific disease mechanisms as well as identify prospective biomarkers for use to stratify patients and monitor treatment. The use of Omics technologies to NCLs has the potential to address this need. We discuss the recent use and outcomes of Omics to various forms of NCL including identification of interactomes, affected biological pathways and potential biomarker candidates. We also identify common pathways affected in NCL across the reviewed studies., (Copyright © 2019 Elsevier B.V. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
27. Neuromuscular junctions are stable in patients with cancer cachexia.
- Author
-
Boehm I, Miller J, Wishart TM, Wigmore SJ, Skipworth RJ, Jones RA, and Gillingwater TH
- Subjects
- Female, Humans, Male, Cachexia metabolism, Cachexia pathology, Gastrointestinal Neoplasms metabolism, Gastrointestinal Neoplasms pathology, Neuromuscular Junction metabolism, Neuromuscular Junction pathology, Rectus Abdominis metabolism, Rectus Abdominis pathology
- Abstract
Cancer cachexia is a major cause of patient morbidity and mortality, with no efficacious treatment or management strategy. Despite cachexia sharing pathophysiological features with a number of neuromuscular wasting conditions, including age-related sarcopenia, the mechanisms underlying cachexia remain poorly understood. Studies of related conditions suggest that pathological targeting of the neuromuscular junction (NMJ) may play a key role in cachexia, but this has yet to be investigated in human patients. Here, high-resolution morphological analyses were undertaken on NMJs of rectus abdominis obtained from patients undergoing upper GI cancer surgery compared with controls (N = 30; n = 1,165 NMJs). Cancer patients included those with cachexia and weight-stable disease. Despite the low skeletal muscle index and significant muscle fiber atrophy (P < 0.0001) in patients with cachexia, NMJ morphology was fully conserved. No significant differences were observed in any of the pre- and postsynaptic variables measured. We conclude that NMJs remain structurally intact in rectus abdominis in both cancer and cachexia, suggesting that denervation of skeletal muscle is not a major driver of pathogenesis. The absence of NMJ pathology is in stark contrast to what is found in related conditions, such as age-related sarcopenia, and supports the hypothesis that intrinsic changes within skeletal muscle, independent of any changes in motor neurons, represent the primary locus of neuromuscular pathology in cancer cachexia.
- Published
- 2020
- Full Text
- View/download PDF
28. Comparative profiling of the synaptic proteome from Alzheimer's disease patients with focus on the APOE genotype.
- Author
-
Hesse R, Hurtado ML, Jackson RJ, Eaton SL, Herrmann AG, Colom-Cadena M, Tzioras M, King D, Rose J, Tulloch J, McKenzie CA, Smith C, Henstridge CM, Lamont D, Wishart TM, and Spires-Jones TL
- Subjects
- Adult, Aged, 80 and over, Alzheimer Disease pathology, Apolipoprotein E4 metabolism, Brain pathology, Female, Humans, Male, Middle Aged, Mitochondria metabolism, Neurons pathology, Proteomics, Synapses pathology, Alzheimer Disease metabolism, Apolipoproteins E metabolism, Brain metabolism, Neurons metabolism, Proteome, Synapses metabolism
- Abstract
Degeneration of synapses in Alzheimer's disease (AD) strongly correlates with cognitive decline, and synaptic pathology contributes to disease pathophysiology. We recently observed that the strongest genetic risk factor for sporadic AD, apolipoprotein E epsilon 4 (APOE4), is associated with exacerbated synapse loss and synaptic accumulation of oligomeric amyloid beta in human AD brain. To begin to understand the molecular cascades involved in synapse loss in AD and how this is mediated by APOE, and to generate a resource of knowledge of changes in the synaptic proteome in AD, we conducted a proteomic screen and systematic in silico analysis of synaptoneurosome preparations from temporal and occipital cortices of human AD and control subjects with known APOE gene status. We examined brain tissue from 33 subjects (7-10 per group). We pooled tissue from all subjects in each group for unbiased proteomic analyses followed by validation with individual case samples. Our analysis identified over 5500 proteins in human synaptoneurosomes and highlighted disease, brain region, and APOE-associated changes in multiple molecular pathways including a decreased abundance in AD of proteins important for synaptic and mitochondrial function and an increased abundance of proteins involved in neuroimmune interactions and intracellular signaling.
- Published
- 2019
- Full Text
- View/download PDF
29. Altered mitochondrial bioenergetics are responsible for the delay in Wallerian degeneration observed in neonatal mice.
- Author
-
Kline RA, Dissanayake KN, Hurtado ML, Martínez NW, Ahl A, Mole AJ, Lamont DJ, Court FA, Ribchester RR, Wishart TM, and Murray LM
- Subjects
- Animals, Animals, Newborn, Mice, Mice, Inbred C57BL, Neuromuscular Junction pathology, Wallerian Degeneration pathology, Mitochondria metabolism, Neuromuscular Junction metabolism, Oxidative Phosphorylation, Wallerian Degeneration metabolism
- Abstract
Neurodegenerative and neuromuscular disorders can manifest throughout the lifespan of an individual, from infant to elderly individuals. Axonal and synaptic degeneration are early and critical elements of nearly all human neurodegenerative diseases and neural injury, however the molecular mechanisms which regulate this process are yet to be fully elucidated. Furthermore, how the molecular mechanisms governing degeneration are impacted by the age of the individual is poorly understood. Interestingly, in mice which are under 3 weeks of age, the degeneration of axons and synapses following hypoxic or traumatic injury is significantly slower. This process, known as Wallerian degeneration (WD), is a molecularly and morphologically distinct subtype of neurodegeneration by which axons and synapses undergo distinct fragmentation and death following a range of stimuli. In this study, we first use an ex-vivo model of axon injury to confirm the significant delay in WD in neonatal mice. We apply tandem mass-tagging quantitative proteomics to profile both nerve and muscle between P12 and P24 inclusive. Application of unbiased in silico workflows to relevant protein identifications highlights a steady elevation in oxidative phosphorylation cascades corresponding to the accelerated degeneration rate. We demonstrate that inhibition of Complex I prevents the axotomy-induced rise in reactive oxygen species and protects axons following injury. Furthermore, we reveal that pharmacological activation of oxidative phosphorylation significantly accelerates degeneration at the neuromuscular junction in neonatal mice. In summary, we reveal dramatic changes in the neuromuscular proteome during post-natal maturation of the neuromuscular system, and demonstrate that endogenous dynamics in mitochondrial bioenergetics during this time window have a functional impact upon regulating the stability of the neuromuscular system., (Copyright © 2019 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
30. CRISPR/Cas9 mediated generation of an ovine model for infantile neuronal ceroid lipofuscinosis (CLN1 disease).
- Author
-
Eaton SL, Proudfoot C, Lillico SG, Skehel P, Kline RA, Hamer K, Rzechorzek NM, Clutton E, Gregson R, King T, O'Neill CA, Cooper JD, Thompson G, Whitelaw CB, and Wishart TM
- Subjects
- Animals, Female, Male, Neuronal Ceroid-Lipofuscinoses genetics, Neuronal Ceroid-Lipofuscinoses metabolism, Sheep, Thiolester Hydrolases genetics, CRISPR-Cas Systems, Disease Models, Animal, Mutation, Neuronal Ceroid-Lipofuscinoses pathology, Phenotype, Thiolester Hydrolases antagonists & inhibitors
- Abstract
The neuronal ceroid lipofuscinoses (NCLs) are a group of devastating monogenetic lysosomal disorders that affect children and young adults with no cure or effective treatment currently available. One of the more severe infantile forms of the disease (INCL or CLN1 disease) is due to mutations in the palmitoyl-protein thioesterase 1 (PPT1) gene and severely reduces the child's lifespan to approximately 9 years of age. In order to better translate the human condition than is possible in mice, we sought to produce a large animal model employing CRISPR/Cas9 gene editing technology. Three PPT1 homozygote sheep were generated by insertion of a disease-causing PPT1 (R151X) human mutation into the orthologous sheep locus. This resulted in a morphological, anatomical and biochemical disease phenotype that closely resembles the human condition. The homozygous sheep were found to have significantly reduced PPT1 enzyme activity and accumulate autofluorescent storage material, as is observed in CLN1 patients. Clinical signs included pronounced behavioral deficits as well as motor deficits and complete loss of vision, with a reduced lifespan of 17 ± 1 months at a humanely defined terminal endpoint. Magnetic resonance imaging (MRI) confirmed a significant decrease in motor cortical volume as well as increased ventricular volume corresponding with observed brain atrophy and a profound reduction in brain mass of 30% at necropsy, similar to alterations observed in human patients. In summary, we have generated the first CRISPR/Cas9 gene edited NCL model. This novel sheep model of CLN1 disease develops biochemical, gross morphological and in vivo brain alterations confirming the efficacy of the targeted modification and potential relevance to the human condition.
- Published
- 2019
- Full Text
- View/download PDF
31. Regional Molecular Mapping of Primate Synapses during Normal Healthy Aging.
- Author
-
Graham LC, Naldrett MJ, Kohama SG, Smith C, Lamont DJ, McColl BW, Gillingwater TH, Skehel P, Urbanski HF, and Wishart TM
- Subjects
- Animals, Hippocampus metabolism, Humans, Macaca mulatta, Occipital Lobe metabolism, Proteomics, Signal Transduction, Transforming Growth Factor beta1 metabolism, Transforming Growth Factor beta1 physiology, Aging, Proteome, Synapses metabolism
- Abstract
Normal mammalian brain aging is characterized by the selective loss of discrete populations of dendritic spines and synapses, particularly affecting neuroanatomical regions such as the hippocampus. Although previous investigations have quantified this morphologically, the molecular pathways orchestrating preferential synaptic vulnerability remain to be elucidated. Using quantitative proteomics and healthy rhesus macaque and human patient brain regional tissues, we have comprehensively profiled the temporal expression of the synaptic proteome throughout the adult lifespan in differentially vulnerable brain regions. Comparative profiling of hippocampal (age vulnerable) and occipital cortex (age resistant) synapses revealed discrete and dynamic alterations in the synaptic proteome, which appear unequivocally conserved between species. The generation of these unique and important datasets will aid in delineating the molecular mechanisms underpinning primate brain aging, in addition to deciphering the regulatory biochemical cascades governing neurodegenerative disease pathogenesis., (Copyright © 2019 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
32. UBA1/GARS-dependent pathways drive sensory-motor connectivity defects in spinal muscular atrophy.
- Author
-
Shorrock HK, van der Hoorn D, Boyd PJ, Llavero Hurtado M, Lamont DJ, Wirth B, Sleigh JN, Schiavo G, Wishart TM, Groen EJN, and Gillingwater TH
- Subjects
- Animals, Ganglia, Spinal metabolism, Ganglia, Spinal pathology, Gene Expression Regulation physiology, HEK293 Cells, Humans, Mice, Motor Neurons metabolism, Motor Neurons pathology, Muscular Atrophy, Spinal metabolism, Neural Pathways metabolism, Sensory Receptor Cells metabolism, Sensory Receptor Cells pathology, Signal Transduction physiology, Spinal Cord metabolism, Spinal Cord pathology, Amino Acyl-tRNA Synthetases metabolism, Muscular Atrophy, Spinal pathology, Neural Pathways pathology, Ubiquitin-Activating Enzymes metabolism
- Abstract
Deafferentation of motor neurons as a result of defective sensory-motor connectivity is a critical early event in the pathogenesis of spinal muscular atrophy, but the underlying molecular pathways remain unknown. We show that restoration of ubiquitin-like modifier-activating enzyme 1 (UBA1) was sufficient to correct sensory-motor connectivity in the spinal cord of mice with spinal muscular atrophy. Aminoacyl-tRNA synthetases, including GARS, were identified as downstream targets of UBA1. Regulation of GARS by UBA1 occurred via a non-canonical pathway independent of ubiquitylation. Dysregulation of UBA1/GARS pathways in spinal muscular atrophy mice disrupted sensory neuron fate, phenocopying GARS-dependent defects associated with Charcot-Marie-Tooth disease. Sensory neuron fate was corrected following restoration of UBA1 expression and UBA1/GARS pathways in spinal muscular atrophy mice. We conclude that defective sensory motor connectivity in spinal muscular atrophy results from perturbations in a UBA1/GARS pathway that modulates sensory neuron fate, thereby highlighting significant molecular and phenotypic overlap between spinal muscular atrophy and Charcot-Marie-Tooth disease.
- Published
- 2018
- Full Text
- View/download PDF
33. Cellular and Molecular Anatomy of the Human Neuromuscular Junction.
- Author
-
Jones RA, Harrison C, Eaton SL, Llavero Hurtado M, Graham LC, Alkhammash L, Oladiran OA, Gale A, Lamont DJ, Simpson H, Simmen MW, Soeller C, Wishart TM, and Gillingwater TH
- Subjects
- Aging physiology, Animals, Humans, Motor Neurons metabolism, Muscle, Skeletal metabolism, Nervous System metabolism, Neuromuscular Junction metabolism, Proteomics, Synapses metabolism, Synaptic Transmission physiology, Neuromuscular Junction anatomy & histology, Neuromuscular Junction cytology
- Abstract
The neuromuscular junction (NMJ) plays a fundamental role in transferring information from lower motor neuron to skeletal muscle to generate movement. It is also an experimentally accessible model synapse routinely studied in animal models to explore fundamental aspects of synaptic form and function. Here, we combined morphological techniques, super-resolution imaging, and proteomic profiling to reveal the detailed cellular and molecular architecture of the human NMJ. Human NMJs were significantly smaller, less complex, and more fragmented than mouse NMJs. In contrast to mice, human NMJs were also remarkably stable across the entire adult lifespan, showing no signs of age-related degeneration or remodeling. Super-resolution imaging and proteomic profiling revealed distinctive distribution of active zone proteins and differential expression of core synaptic proteins and molecular pathways at the human NMJ. Taken together, these findings reveal human-specific cellular and molecular features of the NMJ that distinguish them from comparable synapses in other mammalian species., (Copyright © 2017 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
34. Molecular analysis of axonal-intrinsic and glial-associated co-regulation of axon degeneration.
- Author
-
Catenaccio A, Llavero Hurtado M, Diaz P, Lamont DJ, Wishart TM, and Court FA
- Subjects
- Animals, Cell Dedifferentiation drug effects, Cells, Cultured, Contractile Proteins antagonists & inhibitors, Contractile Proteins genetics, Contractile Proteins metabolism, Peptidyl-Prolyl Isomerase F, Cyclophilins deficiency, Cyclophilins genetics, Dactinomycin pharmacology, Ganglia, Spinal cytology, Ganglia, Spinal metabolism, Mice, Mice, Inbred C57BL, Mice, Knockout, Myelin Sheath physiology, Neuroglia cytology, Proteomics, RNA Interference, Rats, Rats, Sprague-Dawley, Sciatic Nerve drug effects, Sciatic Nerve injuries, Wallerian Degeneration metabolism, Wallerian Degeneration pathology, rho-Associated Kinases metabolism, Axons metabolism, Neuroglia metabolism
- Abstract
Wallerian degeneration is an active program tightly associated with axonal degeneration, required for axonal regeneration and functional recovery after nerve damage. Here we provide a functional molecular foundation for our undertstanding of the complex non-cell autonomous role of glial cells in the regulation of axonal degeneration. To shed light on the complexity of the molecular machinery governing axonal degeneration we employ a multi-model, unbiased, in vivo approach combining morphological assesment and quantitative proteomics with in silico-based higher order functional clustering to genetically uncouple the intrinsic and extrinsic processes governing Wallerian degeneration. Highlighting a pivotal role for glial cells in the early stages fragmenting the axon by a cytokinesis-like process and a cell autonomous stage of axonal disintegration associated to mitochondrial dysfunction.
- Published
- 2017
- Full Text
- View/download PDF
35. Proteomic profiling of neuronal mitochondria reveals modulators of synaptic architecture.
- Author
-
Graham LC, Eaton SL, Brunton PJ, Atrih A, Smith C, Lamont DJ, Gillingwater TH, Pennetta G, Skehel P, and Wishart TM
- Subjects
- Animals, Drosophila, Female, Humans, Male, Mice, Mitochondria pathology, Nerve Degeneration metabolism, Nerve Degeneration pathology, Neurons pathology, Proteomics, Rats, Rats, Sprague-Dawley, Sheep, Synapses pathology, Mitochondria metabolism, Nerve Degeneration physiopathology, Neurons metabolism, Synapses metabolism
- Abstract
Background: Neurons are highly polarized cells consisting of three distinct functional domains: the cell body (and associated dendrites), the axon and the synapse. Previously, it was believed that the clinical phenotypes of neurodegenerative diseases were caused by the loss of entire neurons, however it has recently become apparent that these neuronal sub-compartments can degenerate independently, with synapses being particularly vulnerable to a broad range of stimuli. Whilst the properties governing the differential degenerative mechanisms remain unknown, mitochondria consistently appear in the literature, suggesting these somewhat promiscuous organelles may play a role in affecting synaptic stability. Synaptic and non-synaptic mitochondrial subpools are known to have different enzymatic properties (first demonstrated by Lai et al., 1977). However, the molecular basis underpinning these alterations, and their effects on morphology, has not been well documented., Methods: The current study has employed electron microscopy, label-free proteomics and in silico analyses to characterize the morphological and biochemical properties of discrete sub-populations of mitochondria. The physiological relevance of these findings was confirmed in-vivo using a molecular genetic approach at the Drosophila neuromuscular junction., Results: Here, we demonstrate that mitochondria at the synaptic terminal are indeed morphologically different to non-synaptic mitochondria, in both rodents and human patients. Furthermore, generation of proteomic profiles reveals distinct molecular fingerprints - highlighting that the properties of complex I may represent an important specialisation of synaptic mitochondria. Evidence also suggests that at least 30% of the mitochondrial enzymatic activity differences previously reported can be accounted for by protein abundance. Finally, we demonstrate that the molecular differences between discrete mitochondrial sub-populations are capable of selectively influencing synaptic morphology in-vivo. We offer several novel mitochondrial candidates that have the propensity to significantly alter the synaptic architecture in-vivo., Conclusions: Our study demonstrates discrete proteomic profiles exist dependent upon mitochondrial subcellular localization and selective alteration of intrinsic mitochondrial proteins alters synaptic morphology in-vivo.
- Published
- 2017
- Full Text
- View/download PDF
36. Proteomic mapping of differentially vulnerable pre-synaptic populations identifies regulators of neuronal stability in vivo.
- Author
-
Llavero Hurtado M, Fuller HR, Wong AMS, Eaton SL, Gillingwater TH, Pennetta G, Cooper JD, and Wishart TM
- Subjects
- Animals, Disease Models, Animal, Drosophila metabolism, Drosophila Proteins metabolism, Humans, Mice, Inbred C57BL, Membrane Glycoproteins metabolism, Membrane Proteins metabolism, Neuronal Ceroid-Lipofuscinoses pathology, Neurons metabolism, Neurons pathology, Proteomics methods, Synapses metabolism, Synapses pathology
- Abstract
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.- Published
- 2017
- Full Text
- View/download PDF
37. Bridging the gap: large animal models in neurodegenerative research.
- Author
-
Eaton SL and Wishart TM
- Subjects
- Animals, Animals, Genetically Modified, Genetic Predisposition to Disease, Humans, Nervous System Diseases etiology, Neurodegenerative Diseases diagnosis, Neurodegenerative Diseases metabolism, Neurodegenerative Diseases therapy, Workflow, Disease Models, Animal, Neurodegenerative Diseases etiology, Research
- Abstract
The world health organisation has declared neurological disorders as one of the greatest public health risks in the world today. Yet, despite this growing concern, the mechanisms underpinning many of these conditions are still poorly understood. This may in part be due to the seemingly diverse nature of the initiating insults ranging from genetic (such as the Ataxia's and Lysosomal storage disorders) through to protein misfolding and aggregation (i.e. Prions), and those of a predominantly unknown aetiology (i.e. Alzheimer's and Parkinson's disease). However, efforts to elucidate mechanistic regulation are also likely to be hampered because of the complexity of the human nervous system, the apparent selective regional vulnerability and differential degenerative progression. The key to elucidating these aetiologies is determining the regional molecular cascades, which are occurring from the early through to terminal stages of disease progression. Whilst much molecular data have been captured at the end stage of disease from post-mortem analysis in humans, the very early stages of disease are often conspicuously asymptomatic, and even if they were not, repeated sampling from multiple brain regions of "affected" patients and "controls" is neither ethical nor possible. Model systems therefore become fundamental for elucidating the mechanisms governing these complex neurodegenerative conditions. However, finding a model that precisely mimics the human condition can be challenging and expensive. Whilst cellular and invertebrate models are frequently used in neurodegenerative research and have undoubtedly yielded much useful data, the comparatively simplistic nature of these systems makes insights gained from such a stand alone model limited when it comes to translation. Given the recent advances in gene editing technology, the options for novel model generation in higher order species have opened up new and exciting possibilities for the field. In this review, we therefore explain some of the reasons why larger animal models often appear to give a more robust recapitulation of human neurological disorders and why they may be a critical stepping stone for effective therapeutic translation.
- Published
- 2017
- Full Text
- View/download PDF
38. Pro-death NMDA receptor signaling is promoted by the GluN2B C-terminus independently of Dapk1.
- Author
-
McQueen J, Ryan TJ, McKay S, Marwick K, Baxter P, Carpanini SM, Wishart TM, Gillingwater TH, Manson JC, Wyllie DJA, Grant SGN, McColl BW, Komiyama NH, and Hardingham GE
- Subjects
- Animals, Brain Ischemia drug therapy, Brain Ischemia metabolism, Brain Ischemia pathology, Cell Death, Cells, Cultured, Cerebral Cortex drug effects, Cerebral Cortex metabolism, Cerebral Cortex pathology, Death-Associated Protein Kinases antagonists & inhibitors, Male, Mice, Mice, Knockout, Neurons drug effects, Neurons metabolism, Neuroprotective Agents pharmacology, Phosphorylation, Protein Subunits, Serine chemistry, Serine metabolism, Signal Transduction, Death-Associated Protein Kinases physiology, Neurons pathology, Neuropeptides pharmacology, Receptors, N-Methyl-D-Aspartate metabolism
- Abstract
Aberrant NMDA receptor (NMDAR) activity contributes to several neurological disorders, but direct antagonism is poorly tolerated therapeutically. The GluN2B cytoplasmic C-terminal domain (CTD) represents an alternative therapeutic target since it potentiates excitotoxic signaling. The key GluN2B CTD-centred event in excitotoxicity is proposed to involve its phosphorylation at Ser-1303 by Dapk1, that is blocked by a neuroprotective cell-permeable peptide mimetic of the region. Contrary to this model, we find that excitotoxicity can proceed without increased Ser-1303 phosphorylation, and is unaffected by Dapk1 deficiency in vitro or following ischemia in vivo. Pharmacological analysis of the aforementioned neuroprotective peptide revealed that it acts in a sequence-independent manner as an open-channel NMDAR antagonist at or near the Mg
2+ site, due to its high net positive charge. Thus, GluN2B-driven excitotoxic signaling can proceed independently of Dapk1 or altered Ser-1303 phosphorylation.- Published
- 2017
- Full Text
- View/download PDF
39. Bioenergetic status modulates motor neuron vulnerability and pathogenesis in a zebrafish model of spinal muscular atrophy.
- Author
-
Boyd PJ, Tu WY, Shorrock HK, Groen EJN, Carter RN, Powis RA, Thomson SR, Thomson D, Graham LC, Motyl AAL, Wishart TM, Highley JR, Morton NM, Becker T, Becker CG, Heath PR, and Gillingwater TH
- Subjects
- 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, Motor Neurons metabolism, Muscular Atrophy, Spinal metabolism, Phosphoglycerate Kinase genetics, Spinal Cord metabolism, Survival of Motor Neuron 1 Protein genetics
- 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.
- Published
- 2017
- Full Text
- View/download PDF
40. Analysis of gene expression in the nervous system identifies key genes and novel candidates for health and disease.
- Author
-
Carpanini SM, Wishart TM, Gillingwater TH, Manson JC, and Summers KM
- Subjects
- Animals, Gene Expression Regulation, Gene Ontology, Gene Regulatory Networks, Health, Humans, Male, Mice, Mice, Inbred C57BL, Molecular Sequence Annotation, Nervous System pathology, Nervous System Diseases pathology, Gene Expression Profiling, Genetic Association Studies, Nervous System metabolism, Nervous System Diseases genetics, Transcriptome
- Abstract
The incidence of neurodegenerative diseases in the developed world has risen over the last century, concomitant with an increase in average human lifespan. A major challenge is therefore to identify genes that control neuronal health and viability with a view to enhancing neuronal health during ageing and reducing the burden of neurodegeneration. Analysis of gene expression data has recently been used to infer gene functions for a range of tissues from co-expression networks. We have now applied this approach to transcriptomic datasets from the mammalian nervous system available in the public domain. We have defined the genes critical for influencing neuronal health and disease in different neurological cell types and brain regions. The functional contribution of genes in each co-expression cluster was validated using human disease and knockout mouse phenotypes, pathways and gene ontology term annotation. Additionally a number of poorly annotated genes were implicated by this approach in nervous system function. Exploiting gene expression data available in the public domain allowed us to validate key nervous system genes and, importantly, to identify additional genes with minimal functional annotation but with the same expression pattern. These genes are thus novel candidates for a role in neurological health and disease and could now be further investigated to confirm their function and regulation during ageing and neurodegeneration.
- Published
- 2017
- Full Text
- View/download PDF
41. Sideroflexin 3 is an α-synuclein-dependent mitochondrial protein that regulates synaptic morphology.
- Author
-
Amorim IS, Graham LC, Carter RN, Morton NM, Hammachi F, Kunath T, Pennetta G, Carpanini SM, Manson JC, Lamont DJ, Wishart TM, and Gillingwater TH
- Subjects
- Animals, Drosophila melanogaster metabolism, Energy Metabolism, Gene Ontology, Humans, Mice, Inbred C57BL, Mice, Knockout, Mitochondrial Membranes metabolism, Neuromuscular Junction metabolism, Cation Transport Proteins metabolism, Drosophila Proteins metabolism, Membrane Proteins metabolism, Mitochondrial Proteins metabolism, Synapses metabolism, alpha-Synuclein metabolism
- 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., Competing Interests: I.S.A. and T.H.G. received funding from a CASE Studentship Award supported by GlaxoSmithKline., (© 2017. Published by The Company of Biologists Ltd.)- Published
- 2017
- Full Text
- View/download PDF
42. Equine grass sickness, but not botulism, causes autonomic and enteric neurodegeneration and increases soluble N-ethylmaleimide-sensitive factor attachment receptor protein expression within neuronal perikarya.
- Author
-
McGorum BC, Scholes S, Milne EM, Eaton SL, Wishart TM, Poxton IR, Moss S, Wernery U, Davey T, Harris JB, and Pirie RS
- Subjects
- Animals, Gene Expression Regulation, Horses, N-Ethylmaleimide-Sensitive Proteins genetics, SNARE Proteins genetics, Autonomic Nervous System Diseases veterinary, Botulism veterinary, Horse Diseases physiopathology, N-Ethylmaleimide-Sensitive Proteins metabolism, Neurons metabolism, SNARE Proteins metabolism
- Abstract
Reasons for Performing Study: Equine grass sickness (EGS) is of unknown aetiology. Despite some evidence suggesting that it represents a toxico-infection with Clostridium botulinum types C and/or D, the effect of EGS on the functional targets of botulinum neurotoxins, namely the soluble N-ethylmaleimide-sensitive factor attachment receptor (SNARE) proteins, is unknown. Further, while it is commonly stated that, unlike EGS, equine botulism is not associated with autonomic and enteric neurodegeneration, this has not been definitively assessed., Objectives: To determine: 1) whether botulism causes autonomic and enteric neurodegeneration; and 2) the effect of EGS on the expression of SNARE proteins within cranial cervical ganglion (CCG) and enteric neuronal perikarya., Study Design: Descriptive study., Methods: Light microscopy was used to compare the morphology of neurons in haematoxylin-eosin stained sections of CCG and ileum from 6 EGS horses, 5 botulism horses and 6 control horses. Immunohistochemistry was used to compare the expression of synaptosomal-associated protein-25, synaptobrevin (Syb) and syntaxin within CCG neurons, and of Syb in enteric neurons, from horses with EGS, horses with botulism and control horses. The concentrations of these SNARE proteins in extracts of CCG from EGS and control horses were compared using quantitative fluorescent western blotting., Results: EGS, but not botulism, was associated with autonomic and enteric neurodegeneration and with increased immunoreactivity for SNARE proteins within neuronal perikarya. Quantitative fluorescent western blotting confirmed increased concentrations of synaptosomal-associated protein-25, Syb and syntaxin within CCG extracts from EGS vs. control horses, with the increases in the latter 2 proteins being statistically significant., Conclusions: The occurrence of autonomic and enteric neurodegeneration, and increased expression of SNARE proteins within neuronal perikarya, in EGS but not botulism, suggests that EGS may not be caused by botulinum neurotoxins. Further investigation of the aetiology of EGS is therefore warranted., (© 2015 EVJ Ltd.)
- Published
- 2016
- Full Text
- View/download PDF
43. Commonality amid diversity: Multi-study proteomic identification of conserved disease mechanisms in spinal muscular atrophy.
- Author
-
Fuller HR, Gillingwater TH, and Wishart TM
- Subjects
- Animals, Humans, Muscular Atrophy, Spinal genetics, Muscular Atrophy, Spinal metabolism, Proteome
- Abstract
The neuromuscular disease spinal muscular atrophy (SMA) is a leading genetic cause of infant mortality, resulting from low levels of full-length survival motor neuron (SMN) protein. Despite having a good understanding of the underlying genetics of SMA, the molecular pathways downstream of SMN that regulate disease pathogenesis remain unclear. The identification of molecular perturbations downstream of SMN is required in order to fully understand the fundamental biological role(s) for SMN in cells and tissues of the body, as well as to develop a range of therapeutic targets for developing novel treatments for SMA. Recent developments in proteomic screening technologies have facilitated proteome-wide investigations of a range of SMA models and tissues, generating novel insights into disease mechanisms by highlighting conserved changes in a range of molecular pathways. Comparative analysis of distinct proteomic datasets reveals conserved changes in pathways converging on GAP43, GAPDH, NCAM, UBA1, LMNA, ANXA2 and COL6A3. Proteomic studies therefore represent a leading tool with which to dissect the molecular mechanisms of disease pathogenesis in SMA, serving to identify potentially attractive targets for the development of novel therapies., (Copyright © 2016 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
44. Systemic restoration of UBA1 ameliorates disease in spinal muscular atrophy.
- Author
-
Powis RA, Karyka E, Boyd P, Côme J, Jones RA, Zheng Y, Szunyogova E, Groen EJ, Hunter G, Thomson D, Wishart TM, Becker CG, Parson SH, Martinat C, Azzouz M, and Gillingwater TH
- Subjects
- Animals, Gene Knockdown Techniques, Homeostasis, Humans, Mice, Mice, Knockout, Motor Neurons cytology, Zebrafish, Genetic Therapy, Muscular Atrophy, Spinal therapy, Ubiquitin-Activating Enzymes genetics
- Abstract
The autosomal recessive neuromuscular disease spinal muscular atrophy (SMA) is caused by loss of survival motor neuron (SMN) protein. Molecular pathways that are disrupted downstream of SMN therefore represent potentially attractive therapeutic targets for SMA. Here, we demonstrate that therapeutic targeting of ubiquitin pathways disrupted as a consequence of SMN depletion, by increasing levels of one key ubiquitination enzyme (ubiquitin-like modifier activating enzyme 1 [UBA1]), represents a viable approach for treating SMA. Loss of UBA1 was a conserved response across mouse and zebrafish models of SMA as well as in patient induced pluripotent stem cell-derive motor neurons. Restoration of UBA1 was sufficient to rescue motor axon pathology and restore motor performance in SMA zebrafish. Adeno-associated virus serotype 9-UBA1 (AAV9-UBA1) gene therapy delivered systemic increases in UBA1 protein levels that were well tolerated over a prolonged period in healthy control mice. Systemic restoration of UBA1 in SMA mice ameliorated weight loss, increased survival and motor performance, and improved neuromuscular and organ pathology. AAV9-UBA1 therapy was also sufficient to reverse the widespread molecular perturbations in ubiquitin homeostasis that occur during SMA. We conclude that UBA1 represents a safe and effective therapeutic target for the treatment of both neuromuscular and systemic aspects of SMA.
- Published
- 2016
- Full Text
- View/download PDF
45. Understanding the molecular consequences of inherited muscular dystrophies: advancements through proteomic experimentation.
- Author
-
Fuller HR, Graham LC, Llavero Hurtado M, and Wishart TM
- Subjects
- Humans, Muscular Dystrophies pathology, Biomarkers, Muscular Dystrophies genetics, Proteome genetics, Proteomics
- Abstract
Introduction: Proteomic techniques offer insights into the molecular perturbations occurring in muscular-dystrophies (MD). Revisiting published datasets can highlight conserved downstream molecular alterations, which may be worth re-assessing to determine whether their experimental manipulation is capable of modulating disease severity., Areas Covered: Here, we review the MD literature, highlighting conserved molecular insights warranting mechanistic investigation for therapeutic potential. We also describe a workflow currently proving effective for efficient identification of biomarkers & therapeutic targets in other neurodegenerative conditions, upon which future MD proteomic investigations could be modelled. Expert commentary: Studying disease models can be useful for identifying biomarkers and model specific degenerative cascades, but rarely offer translatable mechanistic insights into disease pathology. Conversely, direct analysis of human samples undergoing degeneration presents challenges derived from complex chronic degenerative molecular processes. This requires a carefully planed & reproducible experimental paradigm accounting for patient selection through to grouping by disease severity and ending with proteomic data filtering and processing.
- Published
- 2016
- Full Text
- View/download PDF
46. Quantitative imaging of tissue sections using infrared scanning technology.
- Author
-
Eaton SL, Cumyn E, King D, Kline RA, Carpanini SM, Del-Pozo J, Barron R, and Wishart TM
- Subjects
- Animals, Mice, Mice, Inbred BALB C, Models, Animal, Brain anatomy & histology, Image Processing, Computer-Assisted methods, Immunohistochemistry methods, Infrared Rays, Microscopy methods
- Abstract
Quantification of immunohistochemically (IHC) labelled tissue sections typically yields semi-quantitative results. Visualising infrared (IR) 'tags', with an appropriate scanner, provides an alternative system where the linear nature of the IR fluorophore emittance enables realistic quantitative fluorescence IHC (QFIHC). Importantly, this new technology enables entire tissue sections to be scanned, allowing accurate area and protein abundance measurements to be calculated from rapidly acquired images. Here, some of the potential benefits of using IR-based tissue imaging are examined, and the following are demonstrated. Firstly, image capture and analysis using IR-based scanning technology yields comparable area-based quantification to those obtained from a modern high-resolution digital slide scanner. Secondly, IR-based dual target visualisation and expression-based quantification is rapid and simple. Thirdly, IR-based relative protein abundance QIHC measurements are an accurate reflection of tissue sample protein abundance, as demonstrated by comparison with quantitative fluorescent Western blotting data. In summary, it is proposed that IR-based QFIHC provides an alternative method of rapid whole-tissue section low-resolution imaging for the production of reliable and accurate quantitative data., (© 2015 The Authors. Journal of Anatomy published by John Wiley & Sons Ltd on behalf of Anatomical Society.)
- Published
- 2016
- Full Text
- View/download PDF
47. Proteomic Profiling of Cranial (Superior) Cervical Ganglia Reveals Beta-Amyloid and Ubiquitin Proteasome System Perturbations in an Equine Multiple System Neuropathy.
- Author
-
McGorum BC, Pirie RS, Eaton SL, Keen JA, Cumyn EM, Arnott DM, Chen W, Lamont DJ, Graham LC, Llavero Hurtado M, Pemberton A, and Wishart TM
- Subjects
- Amyloid beta-Protein Precursor metabolism, Animals, Female, Ganglia, Sensory chemistry, Ganglia, Sensory metabolism, Ganglia, Sensory pathology, Gene Expression Profiling, Gene Expression Regulation, Gene Ontology, Horse Diseases diagnosis, Horse Diseases metabolism, Horse Diseases pathology, Horses, Male, Molecular Sequence Annotation, Neurodegenerative Diseases diagnosis, Neurodegenerative Diseases metabolism, Neurodegenerative Diseases pathology, Proteasome Endopeptidase Complex metabolism, Proteomics, Proteostasis Deficiencies diagnosis, Proteostasis Deficiencies metabolism, Proteostasis Deficiencies pathology, Ubiquitin metabolism, tau Proteins metabolism, Amyloid beta-Protein Precursor genetics, Horse Diseases genetics, Neurodegenerative Diseases genetics, Proteostasis Deficiencies genetics, Ubiquitin genetics, tau Proteins genetics
- Abstract
Equine grass sickness (EGS) is an acute, predominantly fatal, multiple system neuropathy of grazing horses with reported incidence rates of ∼2%. An apparently identical disease occurs in multiple species, including but not limited to cats, dogs, and rabbits. Although the precise etiology remains unclear, ultrastructural findings have suggested that the primary lesion lies in the glycoprotein biosynthetic pathway of specific neuronal populations. The goal of this study was therefore to identify the molecular processes underpinning neurodegeneration in EGS. Here, we use a bottom-up approach beginning with the application of modern proteomic tools to the analysis of cranial (superior) cervical ganglion (CCG, a consistently affected tissue) from EGS-affected patients and appropriate control cases postmortem. In what appears to be the proteomic application of modern proteomic tools to equine neuronal tissues and/or to an inherent neurodegenerative disease of large animals (not a model of human disease), we identified 2,311 proteins in CCG extracts, with 320 proteins increased and 186 decreased by greater than 20% relative to controls. Further examination of selected proteomic candidates by quantitative fluorescent Western blotting (QFWB) and subcellular expression profiling by immunohistochemistry highlighted a previously unreported dysregulation in proteins commonly associated with protein misfolding/aggregation responses seen in a myriad of human neurodegenerative conditions, including but not limited to amyloid precursor protein (APP), microtubule associated protein (Tau), and multiple components of the ubiquitin proteasome system (UPS). Differentially expressed proteins eligible for in silico pathway analysis clustered predominantly into the following biofunctions: (1) diseases and disorders, including; neurological disease and skeletal and muscular disorders and (2) molecular and cellular functions, including cellular assembly and organization, cell-to-cell signaling and interaction (including epinephrine, dopamine, and adrenergic signaling and receptor function), and small molecule biochemistry. Interestingly, while the biofunctions identified in this study may represent pathways underpinning EGS-induced neurodegeneration, this is also the first demonstration of potential molecular conservation (including previously unreported dysregulation of the UPS and APP) spanning the degenerative cascades from an apparently unrelated condition of large animals, to small animal models with altered neuronal vulnerability, and human neurological conditions. Importantly, this study highlights the feasibility and benefits of applying modern proteomic techniques to veterinary investigations of neurodegenerative processes in diseases of large animals., (© 2015 by The American Society for Biochemistry and Molecular Biology, Inc.)
- Published
- 2015
- Full Text
- View/download PDF
48. Molecular neuropathology of the synapse in sheep with CLN5 Batten disease.
- Author
-
Amorim IS, Mitchell NL, Palmer DN, Sawiak SJ, Mason R, Wishart TM, and Gillingwater TH
- Subjects
- Animals, Cerebellum pathology, Cerebral Cortex pathology, Female, Male, Sheep, Disease Models, Animal, Neuronal Ceroid-Lipofuscinoses pathology, Sheep Diseases pathology, Synapses pathology
- Abstract
Aims: Synapses represent a major pathological target across a broad range of neurodegenerative conditions. Recent studies addressing molecular mechanisms regulating synaptic vulnerability and degeneration have relied heavily on invertebrate and mouse models. Whether similar molecular neuropathological changes underpin synaptic breakdown in large animal models and in human patients with neurodegenerative disease remains unclear. We therefore investigated whether molecular regulators of synaptic pathophysiology, previously identified in Drosophila and mouse models, are similarly present and modified in the brain of sheep with CLN5 Batten disease., Methods: Gross neuropathological analysis of CLN5 Batten disease sheep and controls was used alongside postmortem MRI imaging to identify affected brain regions. Synaptosome preparations were then generated and quantitative fluorescent Western blotting used to determine and compare levels of synaptic proteins., Results: The cortex was particularly affected by regional neurodegeneration and synaptic loss in CLN5 sheep, whilst the cerebellum was relatively spared. Quantitative assessment of the protein content of synaptosome preparations revealed significant changes in levels of seven out of eight synaptic neurodegeneration proteins investigated in the motor cortex, but not cerebellum, of CLN5 sheep (α-synuclein, CSP-α, neurofascin, ROCK2, calretinin, SIRT2, and UBR4)., Conclusions: Synaptic pathology is a robust correlate of region-specific neurodegeneration in the brain of CLN5 sheep, driven by molecular pathways similar to those reported in Drosophila and rodent models. Thus, large animal models, such as sheep, represent ideal translational systems to develop and test therapeutics aimed at delaying or halting synaptic pathology for a range of human neurodegenerative conditions.
- Published
- 2015
- Full Text
- View/download PDF
49. Increased levels of UCHL1 are a compensatory response to disrupted ubiquitin homeostasis in spinal muscular atrophy and do not represent a viable therapeutic target.
- Author
-
Powis RA, Mutsaers CA, Wishart TM, Hunter G, Wirth B, and Gillingwater TH
- Subjects
- Animals, Cells, Cultured, Disease Models, Animal, Fibroblasts drug effects, Fibroblasts enzymology, Homeostasis drug effects, Humans, Indoles administration & dosage, Indoles adverse effects, Mice, Motor Activity drug effects, Motor Neurons drug effects, Motor Neurons metabolism, Muscular Atrophy, Spinal pathology, Oximes administration & dosage, Oximes adverse effects, Indoles therapeutic use, Muscular Atrophy, Spinal drug therapy, Muscular Atrophy, Spinal enzymology, Oximes therapeutic use, Survival of Motor Neuron 1 Protein metabolism, Ubiquitin Thiolesterase antagonists & inhibitors, Ubiquitin Thiolesterase metabolism
- Abstract
Aim: Levels of ubiquitin carboxyl-terminal hydrolase L1 (UCHL1) are robustly increased in spinal muscular atrophy (SMA) patient fibroblasts and mouse models. We therefore wanted to establish whether changes in UCHL1 contribute directly to disease pathogenesis, and to assess whether pharmacological inhibition of UCHL1 represents a viable therapeutic option for SMA., Methods: SMA mice and control littermates received a pharmacological UCHL1 inhibitor (LDN-57444) or DMSO vehicle. Survival and weight were monitored daily, a righting test of motor performance was performed, and motor neurone loss, muscle fibre atrophy and neuromuscular junction pathology were all quantified. Ubiquitin-like modifier activating enzyme 1 (Uba1) was then pharmacologically inhibited in neurones in vitro to examine the relationship between Uba1 levels and UCHL1 in SMA., Results: Pharmacological inhibition of UCHL1 failed to improve survival, motor symptoms or neuromuscular pathology in SMA mice and actually precipitated the onset of weight loss. LDN-57444 treatment significantly decreased spinal cord mono-ubiquitin levels, further exacerbating ubiquitination defects in SMA mice. Pharmacological inhibition of Uba1, levels of which are robustly reduced in SMA, was sufficient to induce accumulation of UCHL1 in primary neuronal cultures., Conclusion: Pharmacological inhibition of UCHL1 exacerbates rather than ameliorates disease symptoms in a mouse model of SMA. Thus, pharmacological inhibition of UCHL1 is not a viable therapeutic target for SMA. Moreover, increased levels of UCHL1 in SMA likely represent a downstream consequence of decreased Uba1 levels, indicative of an attempted supportive compensatory response to defects in ubiquitin homeostasis caused by low levels of SMN protein., (© 2014 British Neuropathological Society.)
- Published
- 2014
- Full Text
- View/download PDF
50. A guide to modern quantitative fluorescent western blotting with troubleshooting strategies.
- Author
-
Eaton SL, Hurtado ML, Oldknow KJ, Graham LC, Marchant TW, Gillingwater TH, Farquharson C, and Wishart TM
- Subjects
- Animals, Brain Chemistry, Coloring Agents, Fluorescence, Horses, Mice, Muscle Proteins analysis, Muscle, Skeletal chemistry, Nerve Tissue Proteins analysis, Sheep, Blotting, Western methods, Proteins analysis
- Abstract
The late 1970s saw the first publicly reported use of the western blot, a technique for assessing the presence and relative abundance of specific proteins within complex biological samples. Since then, western blotting methodology has become a common component of the molecular biologists experimental repertoire. A cursory search of PubMed using the term "western blot" suggests that in excess of two hundred and twenty thousand published manuscripts have made use of this technique by the year 2014. Importantly, the last ten years have seen technical imaging advances coupled with the development of sensitive fluorescent labels which have improved sensitivity and yielded even greater ranges of linear detection. The result is a now truly Quantifiable Fluorescence based Western Blot (QFWB) that allows biologists to carry out comparative expression analysis with greater sensitivity and accuracy than ever before. Many "optimized" western blotting methodologies exist and are utilized in different laboratories. These often prove difficult to implement due to the requirement of subtle but undocumented procedural amendments. This protocol provides a comprehensive description of an established and robust QFWB method, complete with troubleshooting strategies.
- Published
- 2014
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.