Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
FEBS J ; 289(13): 3894-3914, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35092170

RESUMEN

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.


Asunto(s)
Proteínas de Transporte de Catión , Degeneración Nerviosa/metabolismo , Animales , Proteínas de Transporte de Catión/metabolismo , Ratones , Membranas Mitocondriales/metabolismo , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Degeneración Nerviosa/patología , Enfermedad de Parkinson/patología , Sinapsis/metabolismo , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo
2.
Cells ; 10(12)2021 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-34943911

RESUMEN

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.


Asunto(s)
Envejecimiento/genética , Proteínas Mitocondriales/genética , Distrofias Musculares/genética , Proteoma/genética , Sinapsis/genética , Animales , Encéfalo/metabolismo , Encéfalo/patología , Modelos Animales de Enfermedad , Drosophila/genética , Drosophila/fisiología , Regulación de la Expresión Génica/genética , Humanos , Ratones , Mitocondrias/genética , Distrofias Musculares/patología , Unión Neuromuscular/genética , Unión Neuromuscular/patología , Neuronas/metabolismo
3.
Cell Rep ; 27(4): 1018-1026.e4, 2019 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-31018120

RESUMEN

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.


Asunto(s)
Envejecimiento , Proteoma , Sinapsis/metabolismo , Animales , Hipocampo/metabolismo , Humanos , Macaca mulatta , Lóbulo Occipital/metabolismo , Proteómica , Transducción de Señal , Factor de Crecimiento Transformador beta1/metabolismo , Factor de Crecimiento Transformador beta1/fisiología
4.
Cell Rep ; 21(9): 2348-2356, 2017 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-29186674

RESUMEN

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.


Asunto(s)
Unión Neuromuscular/anatomía & histología , Unión Neuromuscular/citología , Envejecimiento/fisiología , Animales , Humanos , Neuronas Motoras/metabolismo , Músculo Esquelético/metabolismo , Sistema Nervioso/metabolismo , Unión Neuromuscular/metabolismo , Proteómica , Sinapsis/metabolismo , Transmisión Sináptica/fisiología
5.
Mol Neurodegener ; 12(1): 77, 2017 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-29078798

RESUMEN

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.


Asunto(s)
Mitocondrias/metabolismo , Degeneración Nerviosa/fisiopatología , Neuronas/metabolismo , Sinapsis/metabolismo , Animales , Drosophila , Femenino , Humanos , Masculino , Ratones , Mitocondrias/patología , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/patología , Neuronas/patología , Proteómica , Ratas , Ratas Sprague-Dawley , Ovinos , Sinapsis/patología
6.
PLoS Genet ; 13(4): e1006744, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28426667

RESUMEN

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.


Asunto(s)
Neuronas Motoras/metabolismo , Atrofia Muscular Espinal/metabolismo , Fosfoglicerato Quinasa/genética , Médula Espinal/metabolismo , Proteína 1 para la Supervivencia de la Neurona Motora/genética , Adenosina Trifosfato/metabolismo , Animales , Axones/metabolismo , Axones/patología , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Metabolismo Energético , Regulación del Desarrollo de la Expresión Génica , Humanos , Ratones , Mitocondrias/metabolismo , Neuronas Motoras/efectos de los fármacos , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Atrofia Muscular Espinal/genética , Atrofia Muscular Espinal/fisiopatología , Fosfoglicerato Quinasa/antagonistas & inhibidores , Prazosina/administración & dosificación , Prazosina/análogos & derivados , Médula Espinal/crecimiento & desarrollo , Médula Espinal/patología , Proteína 1 para la Supervivencia de la Neurona Motora/metabolismo , Pez Cebra/genética , Pez Cebra/crecimiento & desarrollo
7.
J Cell Sci ; 130(2): 325-331, 2017 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-28049716

RESUMEN

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.


Asunto(s)
Proteínas de Transporte de Catión/metabolismo , Proteínas de Drosophila/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Mitocondriales/metabolismo , Sinapsis/metabolismo , alfa-Sinucleína/metabolismo , Animales , Drosophila melanogaster/metabolismo , Metabolismo Energético , Ontología de Genes , Humanos , Ratones Endogámicos C57BL , Ratones Noqueados , Membranas Mitocondriales/metabolismo , Unión Neuromuscular/metabolismo
8.
Expert Rev Proteomics ; 13(7): 659-71, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27329572

RESUMEN

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.


Asunto(s)
Biomarcadores , Distrofias Musculares/genética , Proteoma/genética , Proteómica , Humanos , Distrofias Musculares/patología
9.
Mol Cell Proteomics ; 14(11): 3072-86, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26364976

RESUMEN

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.


Asunto(s)
Precursor de Proteína beta-Amiloide/genética , Enfermedades de los Caballos/genética , Enfermedades Neurodegenerativas/genética , Deficiencias en la Proteostasis/genética , Ubiquitina/genética , Proteínas tau/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Femenino , Ganglios Sensoriales/química , Ganglios Sensoriales/metabolismo , Ganglios Sensoriales/patología , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Ontología de Genes , Enfermedades de los Caballos/diagnóstico , Enfermedades de los Caballos/metabolismo , Enfermedades de los Caballos/patología , Caballos , Masculino , Anotación de Secuencia Molecular , Enfermedades Neurodegenerativas/diagnóstico , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/patología , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteómica , Deficiencias en la Proteostasis/diagnóstico , Deficiencias en la Proteostasis/metabolismo , Deficiencias en la Proteostasis/patología , Ubiquitina/metabolismo , Proteínas tau/metabolismo
10.
J Vis Exp ; (93): e52099, 2014 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-25490604

RESUMEN

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.


Asunto(s)
Western Blotting/métodos , Proteínas/análisis , Animales , Química Encefálica , Colorantes , Fluorescencia , Caballos , Ratones , Proteínas Musculares/análisis , Músculo Esquelético/química , Proteínas del Tejido Nervioso/análisis , Ovinos
11.
J Proteome Res ; 13(11): 4546-57, 2014 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-25151848

RESUMEN

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.


Asunto(s)
Homeostasis/fisiología , Atrofia Muscular Espinal/metabolismo , Atrofia Muscular Espinal/patología , Proteómica/métodos , Células de Schwann/patología , Ubiquitina/metabolismo , Animales , Análisis por Conglomerados , Homeostasis/genética , Ratones , Nervios Periféricos/citología , Nervios Periféricos/metabolismo , Células de Schwann/metabolismo , Enzimas Activadoras de Ubiquitina/antagonistas & inhibidores
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...