Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
1.
Acta Neuropathol ; 146(3): 451-475, 2023 09.
Article in English | MEDLINE | ID: mdl-37488208

ABSTRACT

Amyotrophic Lateral Sclerosis (ALS) is a fatal neurodegenerative disease mainly affecting upper and lower motoneurons. Several functionally heterogeneous genes have been associated with the familial form of this disorder (fALS), depicting an extremely complex pathogenic landscape. This heterogeneity has limited the identification of an effective therapy, and this bleak prognosis will only improve with a greater understanding of convergent disease mechanisms. Recent evidence from human post-mortem material and diverse model systems has highlighted the synapse as a crucial structure actively involved in disease progression, suggesting that synaptic aberrations might represent a shared pathological feature across the ALS spectrum. To test this hypothesis, we performed the first comprehensive analysis of the synaptic proteome from post-mortem spinal cord and human iPSC-derived motoneurons carrying mutations in the major ALS genes. This integrated approach highlighted perturbations in the molecular machinery controlling vesicle release as a shared pathomechanism in ALS. Mechanistically, phosphoproteomic analysis linked the presynaptic vesicular phenotype to an accumulation of cytotoxic protein aggregates and to the pro-apoptotic activation of the transcription factor c-Jun, providing detailed insights into the shared pathobiochemistry in ALS. Notably, sub-chronic treatment of our iPSC-derived motoneurons with the fatty acid docosahexaenoic acid exerted a neuroprotective effect by efficiently rescuing the alterations revealed by our multidisciplinary approach. Together, this study provides strong evidence for the central and convergent role played by the synaptic microenvironment within the ALS spinal cord and highlights a potential therapeutic target that counteracts degeneration in a heterogeneous cohort of human motoneuron cultures.


Subject(s)
Amyotrophic Lateral Sclerosis , Neurodegenerative Diseases , Humans , Amyotrophic Lateral Sclerosis/pathology , Neurodegenerative Diseases/pathology , Proteomics , Superoxide Dismutase-1/genetics , Motor Neurons/metabolism
2.
Hum Mol Genet ; 29(16): 2674-2683, 2020 09 29.
Article in English | MEDLINE | ID: mdl-32644120

ABSTRACT

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


Subject(s)
Muscular Atrophy, Spinal/genetics , Myocardium/metabolism , Survival of Motor Neuron 1 Protein/genetics , Animals , Brain/metabolism , Disease Models, Animal , Heart/physiopathology , Humans , Liver/metabolism , Mice , Motor Neurons/metabolism , Motor Neurons/pathology , Muscular Atrophy, Spinal/diagnosis , Muscular Atrophy, Spinal/pathology , Myocardium/pathology , Phenotype , Prenatal Diagnosis , Proteomics , X-Ray Microtomography
3.
Neurobiol Dis ; 130: 104496, 2019 10.
Article in English | MEDLINE | ID: mdl-31176719

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.


Subject(s)
Mitochondria/metabolism , Neuromuscular Junction/metabolism , Oxidative Phosphorylation , Wallerian Degeneration/metabolism , Animals , Animals, Newborn , Mice , Mice, Inbred C57BL , Neuromuscular Junction/pathology , Wallerian Degeneration/pathology
4.
J Cell Sci ; 130(2): 325-331, 2017 01 15.
Article in English | MEDLINE | ID: mdl-28049716

ABSTRACT

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


Subject(s)
Cation Transport Proteins/metabolism , Drosophila Proteins/metabolism , Membrane Proteins/metabolism , Mitochondrial Proteins/metabolism , Synapses/metabolism , alpha-Synuclein/metabolism , Animals , Drosophila melanogaster/metabolism , Energy Metabolism , Gene Ontology , Humans , Mice, Inbred C57BL , Mice, Knockout , Mitochondrial Membranes/metabolism , Neuromuscular Junction/metabolism
5.
Nat Chem Biol ; 13(5): 514-521, 2017 05.
Article in English | MEDLINE | ID: mdl-28288108

ABSTRACT

Inducing macromolecular interactions with small molecules to activate cellular signaling is a challenging goal. PROTACs (proteolysis-targeting chimeras) are bifunctional molecules that recruit a target protein in proximity to an E3 ubiquitin ligase to trigger protein degradation. Structural elucidation of the key ternary ligase-PROTAC-target species and its impact on target degradation selectivity remain elusive. We solved the crystal structure of Brd4 degrader MZ1 in complex with human VHL and the Brd4 bromodomain (Brd4BD2). The ligand folds into itself to allow formation of specific intermolecular interactions in the ternary complex. Isothermal titration calorimetry studies, supported by surface mutagenesis and proximity assays, are consistent with pronounced cooperative formation of ternary complexes with Brd4BD2. Structure-based-designed compound AT1 exhibits highly selective depletion of Brd4 in cells. Our results elucidate how PROTAC-induced de novo contacts dictate preferential recruitment of a target protein into a stable and cooperative complex with an E3 ligase for selective degradation.


Subject(s)
Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Proteolysis/drug effects , Small Molecule Libraries/pharmacology , Transcription Factors/chemistry , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/metabolism , Amino Acid Sequence , Cell Cycle Proteins , Crystallography, X-Ray , Dipeptides/chemistry , Dipeptides/pharmacology , Elongin , Heterocyclic Compounds, 3-Ring/chemistry , Heterocyclic Compounds, 3-Ring/pharmacology , Humans , Models, Molecular , Protein Binding , Protein Conformation , Small Molecule Libraries/chemistry , Structure-Activity Relationship , Thermodynamics , Von Hippel-Lindau Tumor Suppressor Protein/chemistry , Von Hippel-Lindau Tumor Suppressor Protein/metabolism
6.
Brain ; 141(10): 2878-2894, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30239612

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.


Subject(s)
Amino Acyl-tRNA Synthetases/metabolism , Muscular Atrophy, Spinal/pathology , Neural Pathways/pathology , Ubiquitin-Activating Enzymes/metabolism , 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
8.
Mol Cell Proteomics ; 14(11): 3072-86, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26364976

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.


Subject(s)
Amyloid beta-Protein Precursor/genetics , Horse Diseases/genetics , Neurodegenerative Diseases/genetics , Proteostasis Deficiencies/genetics , Ubiquitin/genetics , tau Proteins/genetics , 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
9.
Proc Natl Acad Sci U S A ; 111(49): 17432-7, 2014 Dec 09.
Article in English | MEDLINE | ID: mdl-25326418

ABSTRACT

The siRNA knockdown of IFN Regulatory Factor 5 (IRF5) in the human plasmacytoid dendritic cell line Gen2.2 prevented IFNß production induced by compound CL097, a ligand for Toll-like receptor 7 (TLR7). CL097 also stimulated the phosphorylation of IRF5 at Ser462 and stimulated the nuclear translocation of wild-type IRF5, but not the IRF5[Ser462Ala] mutant. The CL097-stimulated phosphorylation of IRF5 at Ser462 and its nuclear translocation was prevented by the pharmacological inhibition of protein kinase IKKß or the siRNA knockdown of IKKß or its "upstream" activator, the protein kinase TAK1. Similar results were obtained in a murine macrophage cell line stimulated with the TLR7 agonist compound R848 or the nucleotide oligomerization domain 1 (NOD1) agonist KF-1B. IKKß phosphorylated IRF5 at Ser462 in vitro and induced the dimerization of wild-type IRF5 but not the IRF5[S462A] mutant. These findings demonstrate that IKKß activates two "master" transcription factors of the innate immune system, IRF5 and NF-κB.


Subject(s)
Gene Expression Regulation, Enzymologic , I-kappa B Proteins/metabolism , Interferon Regulatory Factors/metabolism , NF-kappa B p50 Subunit/metabolism , Active Transport, Cell Nucleus , Animals , Cell Line , Humans , Immunity, Innate , Inflammation , Interferon-beta/metabolism , Ligands , Mice , Microscopy, Fluorescence , Mutation , Phosphorylation , Protein Multimerization , Serine/chemistry , Transcription, Genetic , Transfection
10.
Kidney Int ; 89(5): 1125-1135, 2016 May.
Article in English | MEDLINE | ID: mdl-27083286

ABSTRACT

Many diabetic patients suffer from declining renal function without developing albuminuria. To identify alternative biomarkers for diabetic nephropathy (DN) we performed urinary peptidomic analysis in a rodent model in which hyperglycemia and hypertension synergize to promote renal pathologic changes consistent with human DN. We identified 297 increased and 15 decreased peptides in the urine of rats with DN compared with controls, including peptides derived from proteins associated with DN and novel candidate biomarkers. We confirmed by ELISA that one of the parent proteins, urinary epidermal growth factor (uEGF), was more than 2-fold reduced in rats with DN in comparison with controls. To assess the clinical utility of uEGF we examined renal outcomes in 642 participants from the Edinburgh Type 2 Diabetes Study who were normoalbuminuric and had preserved renal function at baseline. After adjustment for established renal risk factors, a lower uEGF to creatinine ratio was associated with new-onset estimated glomerular filtration rate less than 60 ml/min per 1.73m(2) (odds ratio 0.48; 95% confidence interval, 0.26-0.90), rapid (over 5% per annum) decline in renal function (odds ratio 0.44; 95% confidence interval, 0.27-0.72) or the composite of both outcomes (odds ratio 0.38; 95% confidence interval, 0.24-0.62). Thus, the utility of a low uEGF to creatinine ratio as a biomarker of progressive decline in renal function in normoalbuminuric patients should be assessed in additional populations.


Subject(s)
Diabetes Mellitus, Type 2/complications , Diabetic Nephropathies/urine , ErbB Receptors/urine , Proteinuria/urine , Proteomics , Receptor, ErbB-2/urine , Aged , Animals , Biomarkers/urine , Case-Control Studies , Chi-Square Distribution , Creatinine/urine , Diabetes Mellitus, Type 2/diagnosis , Diabetic Nephropathies/diagnosis , Diabetic Nephropathies/etiology , Diabetic Nephropathies/physiopathology , Disease Models, Animal , Disease Progression , Enzyme-Linked Immunosorbent Assay , Female , Glomerular Filtration Rate , Humans , Hypertension/complications , Kaplan-Meier Estimate , Kidney/physiopathology , Logistic Models , Male , Mass Spectrometry , Middle Aged , Multivariate Analysis , Odds Ratio , Predictive Value of Tests , Proteomics/methods , Rats, Transgenic , Risk Factors , Scotland , Urinalysis
11.
J Neurosci ; 34(38): 12904-18, 2014 Sep 17.
Article in English | MEDLINE | ID: mdl-25232125

ABSTRACT

Postnatal synapse elimination plays a critical role in sculpting and refining neural connectivity throughout the central and peripheral nervous systems, including the removal of supernumerary axonal inputs from neuromuscular junctions (NMJs). Here, we reveal a novel and important role for myelinating glia in regulating synapse elimination at the mouse NMJ, where loss of a single glial cell protein, the glial isoform of neurofascin (Nfasc155), was sufficient to disrupt postnatal remodeling of synaptic circuitry. Neuromuscular synapses were formed normally in mice lacking Nfasc155, including the establishment of robust neuromuscular synaptic transmission. However, loss of Nfasc155 was sufficient to cause a robust delay in postnatal synapse elimination at the NMJ across all muscle groups examined. Nfasc155 regulated neuronal remodeling independently of its canonical role in forming paranodal axo-glial junctions, as synapse elimination occurred normally in mice lacking the axonal paranodal protein Caspr. Rather, high-resolution proteomic screens revealed that loss of Nfasc155 from glial cells was sufficient to disrupt neuronal cytoskeletal organization and trafficking pathways, resulting in reduced levels of neurofilament light (NF-L) protein in distal axons and motor nerve terminals. Mice lacking NF-L recapitulated the delayed synapse elimination phenotype observed in mice lacking Nfasc155, suggesting that glial cells regulate synapse elimination, at least in part, through modulation of the axonal cytoskeleton. Together, our study reveals a glial cell-dependent pathway regulating the sculpting of neuronal connectivity and synaptic circuitry in the peripheral nervous system.


Subject(s)
Cell Adhesion Molecules/deficiency , Cell Adhesion Molecules/physiology , Nerve Growth Factors/deficiency , Nerve Growth Factors/physiology , Neuromuscular Junction/physiology , Synapses/physiology , Animals , Axons/metabolism , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules, Neuronal/genetics , Cell Adhesion Molecules, Neuronal/physiology , Cytoskeleton/metabolism , Mice , Mice, Knockout , Motor Endplate/growth & development , Motor Neurons/metabolism , Nerve Growth Factors/genetics , Neural Conduction/genetics , Neural Conduction/physiology , Neurofilament Proteins/metabolism , Neuroglia/metabolism , Neuromuscular Junction/growth & development , Protein Isoforms/genetics , Proteomics , Schwann Cells/metabolism , Synapses/genetics , Synaptic Transmission/physiology
12.
BMC Cancer ; 15: 885, 2015 Nov 10.
Article in English | MEDLINE | ID: mdl-26555036

ABSTRACT

BACKGROUND: Cyclin-dependent protein kinase-5 (CDK5) is an unusual member of the CDK family as it is not cell cycle regulated. However many of its substrates have roles in cell growth and oncogenesis, raising the possibility that CDK5 modulation could have therapeutic benefit. In order to establish whether changes in CDK5 activity are associated with oncogenesis one could quantify phosphorylation of CDK5 targets in disease tissue in comparison to appropriate controls. However the identity of physiological and pathophysiological CDK5 substrates remains the subject of debate, making the choice of CDK5 activity biomarkers difficult. METHODS: Here we use in vitro and in cell phosphorylation assays to identify novel features of CDK5 target sequence determinants that confer enhanced CDK5 selectivity, providing means to select substrate biomarkers of CDK5 activity with more confidence. We then characterize tools for the best CDK5 substrate we identified to monitor its phosphorylation in human tissue and use these to interrogate human tumour arrays. RESULTS: The close proximity of Arg/Lys amino acids and a proline two residues N-terminal to the phosphorylated residue both improve recognition of the substrate by CDK5. In contrast the presence of a proline two residues C-terminal to the target residue dramatically reduces phosphorylation rate. Serine-522 of Collapsin Response Mediator-2 (CRMP2) is a validated CDK5 substrate with many of these structural criteria. We generate and characterise phosphospecific antibodies to Ser522 and show that phosphorylation appears in human tumours (lung, breast, and lymphoma) in stark contrast to surrounding non-neoplastic tissue. In lung cancer the anti-phospho-Ser522 signal is positive in squamous cell carcinoma more frequently than adenocarcinoma. Finally we demonstrate that it is a specific and unusual splice variant of CRMP2 (CRMP2A) that is phosphorylated in tumour cells. CONCLUSIONS: For the first time this data associates altered CDK5 substrate phosphorylation with oncogenesis in some but not all tumour types, implicating altered CDK5 activity in aspects of pathogenesis. These data identify a novel oncogenic mechanism where CDK5 activation induces CRMP2A phosphorylation in the nuclei of tumour cells.


Subject(s)
Carcinogenesis/genetics , Cyclin-Dependent Kinase 5/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Neoplasms/genetics , Nerve Tissue Proteins/metabolism , Amino Acid Sequence , Biomarkers, Tumor , Cyclin-Dependent Kinase 5/genetics , Humans , Intercellular Signaling Peptides and Proteins/genetics , Neoplasms/metabolism , Neoplasms/pathology , Nerve Tissue Proteins/genetics , Phosphorylation , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA Splicing/genetics , Serine/metabolism
13.
PLoS Genet ; 8(8): e1002936, 2012.
Article in English | MEDLINE | ID: mdl-22952455

ABSTRACT

Degeneration of synaptic and axonal compartments of neurons is an early event contributing to the pathogenesis of many neurodegenerative diseases, but the underlying molecular mechanisms remain unclear. Here, we demonstrate the effectiveness of a novel "top-down" approach for identifying proteins and functional pathways regulating neurodegeneration in distal compartments of neurons. A series of comparative quantitative proteomic screens on synapse-enriched fractions isolated from the mouse brain following injury identified dynamic perturbations occurring within the proteome during both initiation and onset phases of degeneration. In silico analyses highlighted significant clustering of proteins contributing to functional pathways regulating synaptic transmission and neurite development. Molecular markers of degeneration were conserved in injury and disease, with comparable responses observed in synapse-enriched fractions isolated from mouse models of Huntington's disease (HD) and spinocerebellar ataxia type 5. An initial screen targeting thirteen degeneration-associated proteins using mutant Drosophila lines revealed six potential regulators of synaptic and axonal degeneration in vivo. Mutations in CALB2, ROCK2, DNAJC5/CSP, and HIBCH partially delayed injury-induced neurodegeneration. Conversely, mutations in DNAJC6 and ALDHA1 led to spontaneous degeneration of distal axons and synapses. A more detailed genetic analysis of DNAJC5/CSP mutants confirmed that loss of DNAJC5/CSP was neuroprotective, robustly delaying degeneration in axonal and synaptic compartments. Our study has identified conserved molecular responses occurring within synapse-enriched fractions of the mouse brain during the early stages of neurodegeneration, focused on functional networks modulating synaptic transmission and incorporating molecular chaperones, cytoskeletal modifiers, and calcium-binding proteins. We propose that the proteins and functional pathways identified in the current study represent attractive targets for developing therapeutics aimed at modulating synaptic and axonal stability and neurodegeneration in vivo.


Subject(s)
Brain Injuries , Drosophila , Nerve Degeneration , Synapses , Aldehyde Dehydrogenase/genetics , Aldehyde Dehydrogenase/metabolism , Animals , Axons/metabolism , Axons/pathology , Axons/physiology , Brain Injuries/metabolism , Brain Injuries/pathology , Calbindin 2 , Drosophila/genetics , Drosophila/physiology , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , HSP40 Heat-Shock Proteins/genetics , HSP40 Heat-Shock Proteins/metabolism , Huntington Disease/genetics , Huntington Disease/metabolism , Mice , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Mutation , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Proteomics , S100 Calcium Binding Protein G/genetics , S100 Calcium Binding Protein G/metabolism , Spinocerebellar Ataxias/genetics , Spinocerebellar Ataxias/metabolism , Synapses/metabolism , Synapses/pathology , Thiolester Hydrolases/genetics , Thiolester Hydrolases/metabolism , Wallerian Degeneration/metabolism , Wallerian Degeneration/pathology , rho-Associated Kinases/genetics , rho-Associated Kinases/metabolism
14.
J Proteome Res ; 13(11): 4546-57, 2014 Nov 07.
Article in English | MEDLINE | ID: mdl-25151848

ABSTRACT

Low levels of survival of motor neuron (SMN) protein cause the neuromuscular disease spinal muscular atrophy (SMA), characterized by degeneration of lower motor neurons and atrophy of skeletal muscle. Recent work demonstrated that low levels of SMN also trigger pathological changes in Schwann cells, leading to abnormal axon myelination and disrupted deposition of extracellular matrix proteins in peripheral nerve. However, the molecular pathways linking SMN depletion to intrinsic defects in Schwann cells remained unclear. Label-free proteomics analysis of Schwann cells isolated from SMA mouse peripheral nerve revealed widespread changes to the Schwann cell proteome, including disruption to growth/proliferation, cell death/survival, and molecular transport pathways. Functional clustering analyses revealed significant disruption to a number of proteins contributing to ubiquitination pathways, including reduced levels of ubiquitin-like modifier activating enzyme 1 (Uba1). Pharmacological suppression of Uba1 in Schwann cells was sufficient to reproduce the defective myelination phenotype seen in SMA. These findings demonstrate an important role for SMN protein and ubiquitin-dependent pathways in maintaining Schwann cell homeostasis and provide significant additional experimental evidence supporting a key role for ubiquitin pathways and, Uba1 in particular, in driving SMA pathogenesis across a broad range of cells and tissues.


Subject(s)
Homeostasis/physiology , Muscular Atrophy, Spinal/metabolism , Muscular Atrophy, Spinal/pathology , Proteomics/methods , Schwann Cells/pathology , Ubiquitin/metabolism , Animals , Cluster Analysis , Homeostasis/genetics , Mice , Peripheral Nerves/cytology , Peripheral Nerves/metabolism , Schwann Cells/metabolism , Ubiquitin-Activating Enzymes/antagonists & inhibitors
15.
Anal Chem ; 86(20): 10296-302, 2014 Oct 21.
Article in English | MEDLINE | ID: mdl-25233145

ABSTRACT

Reproducible, comprehensive phosphopeptide enrichment is essential for studying phosphorylation-regulated processes. Here, we describe the application of hyper-porous magnetic TiO2 and Ti-IMAC microspheres for uniform automated phosphopeptide enrichment. Combining magnetic microspheres with a magnetic particle-handling robot enables rapid (45 min), reproducible (r2 ≥ 0.80) and high-fidelity (>90% purity) phosphopeptide purification in a 96-well format. Automated phosphopeptide enrichment demonstrates reproducible synthetic phosphopeptide recovery across 2 orders of magnitude, "well-to-well" quantitative reproducibility indistinguishable to internal SILAC standards, and robust "plate-to-plate" reproducibility across 5 days of independent enrichments. As a result, automated phosphopeptide enrichment enables statistical analysis of label-free phosphoproteomic samples in a high-throughput manner. This technique uses commercially available, off-the-shelf components and can be easily adopted by any laboratory interested in phosphoproteomic analysis. We provide a free downloadable automated phosphopeptide enrichment program to facilitate uniform interlaboratory collaboration and exchange of phosphoproteomic data sets.


Subject(s)
Imidazoles/chemistry , Magnetics , Phosphopeptides/chemistry , Proteomics/methods , Titanium/chemistry , Automation , Multivariate Analysis , Phosphopeptides/isolation & purification , Reproducibility of Results
16.
Nat Commun ; 15(1): 1221, 2024 Feb 09.
Article in English | MEDLINE | ID: mdl-38336824

ABSTRACT

Exposure of plants to ultraviolet-B (UV-B) radiation initiates transcriptional responses that modify metabolism, physiology and development to enhance viability in sunlight. Many of these regulatory responses to UV-B radiation are mediated by the photoreceptor UV RESISTANCE LOCUS 8 (UVR8). Following photoreception, UVR8 interacts directly with multiple proteins to regulate gene expression, but the mechanisms that control differential protein binding to initiate distinct responses are unknown. Here we show that UVR8 is phosphorylated at several sites and that UV-B stimulates phosphorylation at Serine 402. Site-directed mutagenesis to mimic Serine 402 phosphorylation promotes binding of UVR8 to REPRESSOR OF UV-B PHOTOMORPHOGENESIS (RUP) proteins, which negatively regulate UVR8 action. Complementation of the uvr8 mutant with phosphonull or phosphomimetic variants suggests that phosphorylation of Serine 402 modifies UVR8 activity and promotes flavonoid biosynthesis, a key UV-B-stimulated response that enhances plant protection and crop nutritional quality. This research provides a basis to understand how UVR8 interacts differentially with effector proteins to regulate plant responses to UV-B radiation.


Subject(s)
Arabidopsis Proteins , Arabidopsis , Chromosomal Proteins, Non-Histone , Ultraviolet Rays , Arabidopsis/metabolism , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Gene Expression Regulation, Plant , Phosphorylation , Serine/metabolism
17.
Hum Mol Genet ; 20(22): 4334-44, 2011 Nov 15.
Article in English | MEDLINE | ID: mdl-21840928

ABSTRACT

Low levels of full-length survival motor neuron (SMN) protein cause the motor neuron disease, spinal muscular atrophy (SMA). Although motor neurons undoubtedly contribute directly to SMA pathogenesis, the role of muscle is less clear. We demonstrate significant disruption to the molecular composition of skeletal muscle in pre-symptomatic severe SMA mice, in the absence of any detectable degenerative changes in lower motor neurons and with a molecular profile distinct from that of denervated muscle. Functional cluster analysis of proteomic data and phospho-histone H2AX labelling of DNA damage revealed increased activity of cell death pathways in SMA muscle. Robust upregulation of voltage-dependent anion-selective channel protein 2 (Vdac2) and downregulation of parvalbumin in severe SMA mice was confirmed in a milder SMA mouse model and in human patient muscle biopsies. Molecular pathology of skeletal muscle was ameliorated in mice treated with the FDA-approved histone deacetylase inhibitor, suberoylanilide hydroxamic acid. We conclude that intrinsic pathology of skeletal muscle is an important and reversible event in SMA and also suggest that muscle proteins have the potential to act as novel biomarkers in SMA.


Subject(s)
Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Atrophy, Spinal/metabolism , Muscular Atrophy, Spinal/pathology , SMN Complex Proteins/metabolism , Animals , Blotting, Western , Histone Deacetylase Inhibitors/therapeutic use , Humans , Hydroxamic Acids/therapeutic use , Immunohistochemistry , In Vitro Techniques , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Muscle, Skeletal/drug effects , Muscular Atrophy, Spinal/drug therapy , SMN Complex Proteins/genetics , Survival of Motor Neuron 1 Protein/genetics , Survival of Motor Neuron 1 Protein/metabolism , Survival of Motor Neuron 2 Protein/genetics , Survival of Motor Neuron 2 Protein/metabolism , Vorinostat
18.
Proc Natl Acad Sci U S A ; 107(44): 18838-43, 2010 Nov 02.
Article in English | MEDLINE | ID: mdl-20956331

ABSTRACT

Recognition and repair of cellular damage is crucial if organisms are to survive harmful environmental conditions. In mammals, the Keap1 protein orchestrates this response, but how it perceives adverse circumstances is not fully understood. Herein, we implicate NO, Zn(2+), and alkenals, endogenously occurring chemicals whose concentrations increase during stress, in this process. By combining molecular modeling with phylogenetic, chemical, and functional analyses, we show that Keap1 directly recognizes NO, Zn(2+), and alkenals through three distinct sensors. The C288 alkenal sensor is of ancient origin, having evolved in a common ancestor of bilaterans. The Zn(2+) sensor minimally comprises H225, C226, and C613. The most recent sensor, the NO sensor, emerged coincident with an expansion of the NOS gene family in vertebrates. It comprises a cluster of basic amino acids (H129, K131, R135, K150, and H154) that facilitate S-nitrosation of C151. Taken together, our data suggest that Keap1 is a specialized sensor that quantifies stress by monitoring the intracellular concentrations of NO, Zn(2+), and alkenals, which collectively serve as second messengers that may signify danger and/or damage.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cytoskeletal Proteins/metabolism , Models, Biological , Nitric Oxide/metabolism , Signal Transduction/physiology , Stress, Physiological/physiology , Zinc/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , COS Cells , Chlorocebus aethiops , Cytoskeletal Proteins/genetics , Kelch-Like ECH-Associated Protein 1 , Mice , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase/metabolism
19.
Aging Cell ; 22(5): e13814, 2023 05.
Article in English | MEDLINE | ID: mdl-36973898

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.


Subject(s)
Necroptosis , Neurodegenerative Diseases , Humans , Mice , Animals , Aged , Proteomics , Rejuvenation , Aging/physiology , Brain , Memory Disorders
20.
Hum Mol Genet ; 19(21): 4216-28, 2010 Nov 01.
Article in English | MEDLINE | ID: mdl-20705736

ABSTRACT

Reduced expression of the survival motor neuron (SMN) gene causes the childhood motor neuron disease spinal muscular atrophy (SMA). Low levels of ubiquitously expressed SMN protein result in the degeneration of lower motor neurons, but it remains unclear whether other regions of the nervous system are also affected. Here we show that reduced levels of SMN lead to impaired perinatal brain development in a mouse model of severe SMA. Regionally selective changes in brain morphology were apparent in areas normally associated with higher SMN levels in the healthy postnatal brain, including the hippocampus, and were associated with decreased cell density, reduced cell proliferation and impaired hippocampal neurogenesis. A comparative proteomics analysis of the hippocampus from SMA and wild-type littermate mice revealed widespread modifications in expression levels of proteins regulating cellular proliferation, migration and development when SMN levels were reduced. This study reveals novel roles for SMN protein in brain development and maintenance and provides the first insights into cellular and molecular pathways disrupted in the brain in a severe form of SMA.


Subject(s)
Disease Models, Animal , Hippocampus/growth & development , Muscular Atrophy, Spinal/genetics , Survival of Motor Neuron 1 Protein/genetics , Animals , Cell Movement , Cell Proliferation , Mice , Mice, Knockout , Proteomics
SELECTION OF CITATIONS
SEARCH DETAIL