Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters











Publication year range
1.
Mol Cell ; 84(15): 2966-2983.e9, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39089251

ABSTRACT

Defects in organellar acidification indicate compromised or infected compartments. Recruitment of the autophagy-related ATG16L1 complex to pathologically neutralized organelles targets ubiquitin-like ATG8 molecules to perturbed membranes. How this process is coupled to proton gradient disruption is unclear. Here, we reveal that the V1H subunit of the vacuolar ATPase (V-ATPase) proton pump binds directly to ATG16L1. The V1H/ATG16L1 interaction only occurs within fully assembled V-ATPases, allowing ATG16L1 recruitment to be coupled to increased V-ATPase assembly following organelle neutralization. Cells lacking V1H fail to target ATG8s during influenza infection or after activation of the immune receptor stimulator of interferon genes (STING). We identify a loop within V1H that mediates ATG16L1 binding. A neuronal V1H isoform lacks this loop and is associated with attenuated ATG8 targeting in response to ionophores in primary murine and human iPSC-derived neurons. Thus, V1H controls ATG16L1 recruitment following proton gradient dissipation, suggesting that the V-ATPase acts as a cell-intrinsic damage sensor.


Subject(s)
Autophagy-Related Proteins , Vacuolar Proton-Translocating ATPases , Vacuolar Proton-Translocating ATPases/metabolism , Vacuolar Proton-Translocating ATPases/genetics , Humans , Autophagy-Related Proteins/metabolism , Autophagy-Related Proteins/genetics , Animals , Mice , Protein Binding , Neurons/metabolism , Autophagy-Related Protein 8 Family/metabolism , Autophagy-Related Protein 8 Family/genetics , Autophagy , HEK293 Cells , Induced Pluripotent Stem Cells/metabolism , Influenza, Human/virology , Influenza, Human/metabolism , Influenza, Human/genetics , Mice, Inbred C57BL , Signal Transduction , Carrier Proteins/metabolism , Carrier Proteins/genetics , Mice, Knockout
2.
Stem Cell Res ; 74: 103291, 2024 02.
Article in English | MEDLINE | ID: mdl-38141358

ABSTRACT

The neuronal ceroid lipofuscinoses (NCLs) are a group of common inherited neurodegenerative disorders of childhood. All forms of NCLs are life-limiting with no curative treatments. Most of the 13 NCL genes encode proteins residing in endolysosomal pathways, such as CLN5, a potential lysosomal enzyme. Two induced pluripotent stem cell lines (hiPSCs) were generated from skin fibroblasts of CLN5 disease patients via non-integrating Sendai virus reprogramming. They demonstrate typical stem cell morphology, express pluripotency markers, exhibit trilineage differentiation potential and also successfully differentiate into neurons. These hiPSCs represent a potential resource to model CLN5 disease in a human context and investigate potential therapies.


Subject(s)
Induced Pluripotent Stem Cells , Neuronal Ceroid-Lipofuscinoses , Humans , Membrane Proteins/genetics , Neuronal Ceroid-Lipofuscinoses/genetics , Induced Pluripotent Stem Cells/metabolism , Lysosomal Membrane Proteins/genetics , Mutation/genetics , Fibroblasts/metabolism
3.
J Cell Sci ; 135(22)2022 11 15.
Article in English | MEDLINE | ID: mdl-36274588

ABSTRACT

Long-term changes in synaptic strength form the basis of learning and memory. These changes rely upon energy-demanding mechanisms, which are regulated by local Ca2+ signalling. Mitochondria are optimised for providing energy and buffering Ca2+. However, our understanding of the role of mitochondria in regulating synaptic plasticity is incomplete. Here, we have used optical and electrophysiological techniques in cultured hippocampal neurons and ex vivo hippocampal slices from mice with haploinsufficiency of the mitochondrial Ca2+ uniporter (MCU+/-) to address whether reducing mitochondrial Ca2+ uptake alters synaptic transmission and plasticity. We found that cultured MCU+/- hippocampal neurons have impaired Ca2+ clearance, and consequently enhanced synaptic vesicle fusion at presynapses occupied by mitochondria. Furthermore, long-term potentiation (LTP) at mossy fibre (MF) synapses, a process which is dependent on presynaptic Ca2+ accumulation, is enhanced in MCU+/- slices. Our results reveal a previously unrecognised role for mitochondria in regulating presynaptic plasticity of a major excitatory pathway involved in learning and memory.


Subject(s)
Long-Term Potentiation , Mossy Fibers, Hippocampal , Mice , Animals , Mossy Fibers, Hippocampal/metabolism , Long-Term Potentiation/physiology , Calcium/metabolism , Haploinsufficiency , Synapses/metabolism , Synaptic Transmission/physiology , Mitochondria/metabolism
6.
Cell Death Differ ; 27(10): 2781-2796, 2020 10.
Article in English | MEDLINE | ID: mdl-32341450

ABSTRACT

Protein aggregation and abnormal lipid homeostasis are both implicated in neurodegeneration through unknown mechanisms. Here we demonstrate that aggregate-membrane interaction is critical to induce a form of cell death called ferroptosis. Importantly, the aggregate-membrane interaction that drives ferroptosis depends both on the conformational structure of the aggregate, as well as the oxidation state of the lipid membrane. We generated human stem cell-derived models of synucleinopathy, characterized by the intracellular formation of α-synuclein aggregates that bind to membranes. In human iPSC-derived neurons with SNCA triplication, physiological concentrations of glutamate and dopamine induce abnormal calcium signaling owing to the incorporation of excess α-synuclein oligomers into membranes, leading to altered membrane conductance and abnormal calcium influx. α-synuclein oligomers further induce lipid peroxidation. Targeted inhibition of lipid peroxidation prevents the aggregate-membrane interaction, abolishes aberrant calcium fluxes, and restores physiological calcium signaling. Inhibition of lipid peroxidation, and reduction of iron-dependent accumulation of free radicals, further prevents oligomer-induced toxicity in human neurons. In summary, we report that peroxidation of polyunsaturated fatty acids underlies the incorporation of ß-sheet-rich aggregates into the membranes, and that additionally induces neuronal death. This suggests a role for ferroptosis in Parkinson's disease, and highlights a new mechanism by which lipid peroxidation causes cell death.


Subject(s)
Calcium/metabolism , Ferroptosis , Iron/metabolism , Lipid Peroxidation , Parkinson Disease , alpha-Synuclein/metabolism , Cells, Cultured , Human Embryonic Stem Cells , Humans , Induced Pluripotent Stem Cells , Parkinson Disease/metabolism , Parkinson Disease/pathology
7.
Methods Mol Biol ; 1994: 159-164, 2019.
Article in English | MEDLINE | ID: mdl-31124113

ABSTRACT

Induced pluripotent stem cell (iPSC) derived neurons are an excellent in vitro model of neurological diseases that are often used in early stage drug discovery projects. Thus far, the use of iPSC-derived cells in small molecule drug screening has been limited, and one of the reasons for this has been the challenge of miniaturization of iPSC culture and differentiation in low volume microwell plate formats. Here we describe a method of seeding iPSC-derived neurons into 384-well plates towards the end of the differentiation procedure. This method covers coating the plates with substrates to aid attachment, dissociation of the cells into a single cell suspension, and seeding onto 384-well plates to give an even distribution of neurons. This method facilitates the use of iPSC-derived neurons for high-content imaging, whole-well assays, and small-molecule drug screening.


Subject(s)
Induced Pluripotent Stem Cells/cytology , Neurons/cytology , Cell Culture Techniques/methods , Cell Differentiation , Cells, Cultured , Humans , Neurons/metabolism
8.
Methods Mol Biol ; 1994: 165-174, 2019.
Article in English | MEDLINE | ID: mdl-31124114

ABSTRACT

Autophagy is the process by which cellular proteins and organelles are degraded and recycled and is essential to the survival of cells. Defective autophagic degradation has been linked to many neurodegenerative diseases and in particular lysosomal storage diseases. Here we describe a high-content assay to detect defects in the autophagy pathway in induced pluripotent stem cell-derived neurons. This assay utilizes immunofluorescence to stain autophagosomes and uses automated image analysis to measure changes in autophagosome levels in response to modulators of autophagy.


Subject(s)
Autophagosomes/metabolism , Autophagy/physiology , Fluorescent Antibody Technique/methods , Induced Pluripotent Stem Cells/cytology , Neurons/cytology , Humans , Image Processing, Computer-Assisted , Microscopy, Fluorescence , Neurons/metabolism
9.
Methods Mol Biol ; 1994: 175-184, 2019.
Article in English | MEDLINE | ID: mdl-31124115

ABSTRACT

Mitochondrial dysfunction is linked to many neurological diseases; therefore, the ability to measure mitochondrial function is of great use for researching disease and testing potential therapeutics. Here we describe a high-content assay to simultaneously measure mitochondrial membrane potential, morphology and cell viability in iPSC-derived neurons. Neurons are seeded into plates suitable for fluorescent microscopy, stained with the mitochondrial membrane potential-dependent dye TMRM, cytoplasmic dye Calcein AM, and nuclear stain Hoechst 33342. Images are acquired in live cells and analyzed using automated image analysis software.


Subject(s)
Image Processing, Computer-Assisted/methods , Induced Pluripotent Stem Cells/cytology , Membrane Potential, Mitochondrial , Microscopy, Fluorescence , Neurons/ultrastructure , Cell Survival , Humans , Mitochondria/ultrastructure , Neurons/cytology , Neurons/physiology
10.
Neurobiol Aging ; 78: 130-141, 2019 06.
Article in English | MEDLINE | ID: mdl-30925301

ABSTRACT

Induced pluripotent stem cells and their derivatives have become an important tool for researching disease mechanisms. It is hoped that they could be used to discover new therapies by providing the most reliable and relevant human in vitro disease models for drug discovery. This review will summarize recent efforts to use stem cell-derived neurons for drug screening. We also explain the current hurdles to using these cells for high-throughput pharmaceutical screening and developments that may help overcome these hurdles. Finally, we critically discuss whether induced pluripotent stem cell-derived neurons will come to fruition as a model that is regularly used to screen for drugs to treat neurological diseases.


Subject(s)
Drug Evaluation, Preclinical/methods , High-Throughput Screening Assays/methods , Nervous System Diseases/drug therapy , Neural Stem Cells , Pluripotent Stem Cells , Drug Discovery , Humans
11.
Nat Nanotechnol ; 14(1): 80-88, 2019 01.
Article in English | MEDLINE | ID: mdl-30510280

ABSTRACT

Much of the functionality of multicellular systems arises from the spatial organization and dynamic behaviours within and between cells. Current single-cell genomic methods only provide a transcriptional 'snapshot' of individual cells. The real-time analysis and perturbation of living cells would generate a step change in single-cell analysis. Here we describe minimally invasive nanotweezers that can be spatially controlled to extract samples from living cells with single-molecule precision. They consist of two closely spaced electrodes with gaps as small as 10-20 nm, which can be used for the dielectrophoretic trapping of DNA and proteins. Aside from trapping single molecules, we also extract nucleic acids for gene expression analysis from living cells without affecting their viability. Finally, we report on the trapping and extraction of a single mitochondrion. This work bridges the gap between single-molecule/organelle manipulation and cell biology and can ultimately enable a better understanding of living cells.


Subject(s)
Nanotechnology , Optical Tweezers , Single-Cell Analysis , Animals , Axons/metabolism , Biopsy , Cell Line, Tumor , Cell Nucleus/metabolism , DNA/chemistry , Electricity , Electrodes , Fluorescence , Humans , Mice , Mitochondria/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Solutions
12.
Nat Commun ; 9(1): 2293, 2018 06 12.
Article in English | MEDLINE | ID: mdl-29895861

ABSTRACT

Protein aggregation causes α-synuclein to switch from its physiological role to a pathological toxic gain of function. Under physiological conditions, monomeric α-synuclein improves ATP synthase efficiency. Here, we report that aggregation of monomers generates beta sheet-rich oligomers that localise to the mitochondria in close proximity to several mitochondrial proteins including ATP synthase. Oligomeric α-synuclein impairs complex I-dependent respiration. Oligomers induce selective oxidation of the ATP synthase beta subunit and mitochondrial lipid peroxidation. These oxidation events increase the probability of permeability transition pore (PTP) opening, triggering mitochondrial swelling, and ultimately cell death. Notably, inhibition of oligomer-induced oxidation prevents the pathological induction of PTP. Inducible pluripotent stem cells (iPSC)-derived neurons bearing SNCA triplication, generate α-synuclein aggregates that interact with the ATP synthase and induce PTP opening, leading to neuronal death. This study shows how the transition of α-synuclein from its monomeric to oligomeric structure alters its functional consequences in Parkinson's disease.


Subject(s)
Induced Pluripotent Stem Cells/metabolism , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins , Mitochondrial Proton-Translocating ATPases/metabolism , Neurons/metabolism , Parkinson Disease/metabolism , alpha-Synuclein/metabolism , Animals , Coculture Techniques , Embryonic Stem Cells/metabolism , Humans , Lipid Peroxidation , Mitochondrial Permeability Transition Pore , Oxidation-Reduction , Patch-Clamp Techniques , Permeability , Proteomics , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism
13.
Sci Rep ; 8(1): 9033, 2018 06 13.
Article in English | MEDLINE | ID: mdl-29899557

ABSTRACT

Mitochondrial dysfunction is implicated in many neurodegenerative diseases including Parkinson's disease (PD). Induced pluripotent stem cells (iPSCs) provide a unique cell model for studying neurological diseases. We have established a high-content assay that can simultaneously measure mitochondrial function, morphology and cell viability in iPSC-derived dopaminergic neurons. iPSCs from PD patients with mutations in SNCA and unaffected controls were differentiated into dopaminergic neurons, seeded in 384-well plates and stained with the mitochondrial membrane potential dependent dye TMRM, alongside Hoechst-33342 and Calcein-AM. Images were acquired using an automated confocal screening microscope and single cells were analysed using automated image analysis software. PD neurons displayed reduced mitochondrial membrane potential and altered mitochondrial morphology compared to control neurons. This assay demonstrates that high content screening techniques can be applied to the analysis of mitochondria in iPSC-derived neurons. This technique could form part of a drug discovery platform to test potential new therapeutics for PD and other neurodegenerative diseases.


Subject(s)
Dopaminergic Neurons/metabolism , Induced Pluripotent Stem Cells/metabolism , Mitochondria/metabolism , Mutation , Single-Cell Analysis/methods , alpha-Synuclein/metabolism , Benzimidazoles , Cell Differentiation/genetics , Cell Survival/genetics , Cells, Cultured , Humans , Membrane Potential, Mitochondrial/genetics , Microscopy, Confocal , Mitochondria/genetics , Parkinson Disease/genetics , Parkinson Disease/metabolism , Parkinson Disease/pathology , Rhodamines , alpha-Synuclein/genetics
14.
Nat Rev Neurosci ; 19(2): 63-80, 2018 01 19.
Article in English | MEDLINE | ID: mdl-29348666

ABSTRACT

Synapses enable neurons to communicate with each other and are therefore a prerequisite for normal brain function. Presynaptically, this communication requires energy and generates large fluctuations in calcium concentrations. Mitochondria are optimized for supplying energy and buffering calcium, and they are actively recruited to presynapses. However, not all presynapses contain mitochondria; thus, how might synapses with and without mitochondria differ? Mitochondria are also increasingly recognized to serve additional functions at the presynapse. Here, we discuss the importance of presynaptic mitochondria in maintaining neuronal homeostasis and how dysfunctional presynaptic mitochondria might contribute to the development of disease.


Subject(s)
Mitochondria/physiology , Neurons/physiology , Presynaptic Terminals/physiology , Synaptic Transmission/physiology , Animals , Calcium Signaling/physiology , Homeostasis/physiology , Humans
15.
Acta Neuropathol ; 134(1): 129-149, 2017 07.
Article in English | MEDLINE | ID: mdl-28337542

ABSTRACT

α-Synuclein is strongly linked to Parkinson's disease but the molecular targets for its toxicity are not fully clear. However, many neuronal functions damaged in Parkinson's disease are regulated by signalling between the endoplasmic reticulum (ER) and mitochondria. This signalling involves close physical associations between the two organelles that are mediated by binding of the integral ER protein vesicle-associated membrane protein-associated protein B (VAPB) to the outer mitochondrial membrane protein, protein tyrosine phosphatase-interacting protein 51 (PTPIP51). VAPB and PTPIP51 thus act as a scaffold to tether the two organelles. Here we show that α-synuclein binds to VAPB and that overexpression of wild-type and familial Parkinson's disease mutant α-synuclein disrupt the VAPB-PTPIP51 tethers to loosen ER-mitochondria associations. This disruption to the VAPB-PTPIP51 tethers is also seen in neurons derived from induced pluripotent stem cells from familial Parkinson's disease patients harbouring pathogenic triplication of the α-synuclein gene. We also show that the α-synuclein induced loosening of ER-mitochondria contacts is accompanied by disruption to Ca2+ exchange between the two organelles and mitochondrial ATP production. Such disruptions are likely to be particularly damaging to neurons that are heavily dependent on correct Ca2+ signaling and ATP.


Subject(s)
Adenosine Triphosphate/metabolism , Calcium/metabolism , Homeostasis/physiology , Mitochondria/metabolism , Vesicular Transport Proteins/metabolism , alpha-Synuclein/metabolism , Animals , Cations, Divalent/metabolism , Cell Line, Tumor , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/pathology , Glycogen Synthase Kinase 3 beta/metabolism , HEK293 Cells , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology , Mitochondria/pathology , Mitochondrial Proteins/metabolism , Mutation , Parkinson Disease/genetics , Parkinson Disease/metabolism , Parkinson Disease/pathology , Protein Tyrosine Phosphatases/metabolism , Rats, Sprague-Dawley , alpha-Synuclein/genetics
16.
EMBO Rep ; 18(2): 231-240, 2017 02.
Article in English | MEDLINE | ID: mdl-28039205

ABSTRACT

Mitochondrial trafficking is influenced by neuronal activity, but it remains unclear how mitochondrial positioning influences neuronal transmission and plasticity. Here, we use live cell imaging with the genetically encoded presynaptically targeted Ca2+ indicator, SyGCaMP5, to address whether presynaptic Ca2+ responses are altered by mitochondria in synaptic terminals. We find that presynaptic Ca2+ signals, as well as neurotransmitter release, are significantly decreased in terminals containing mitochondria. Moreover, the localisation of mitochondria at presynaptic sites can be altered during long-term activity changes, dependent on the Ca2+-sensing function of the mitochondrial trafficking protein, Miro1. In addition, we find that Miro1-mediated activity-dependent synaptic repositioning of mitochondria allows neurons to homeostatically alter the strength of presynaptic Ca2+ signals in response to prolonged changes in neuronal activity. Our results support a model in which mitochondria are recruited to presynaptic terminals during periods of raised neuronal activity and are involved in rescaling synaptic signals during homeostatic plasticity.


Subject(s)
Calcium Signaling , Homeostasis , Mitochondria/metabolism , Neuronal Plasticity , Presynaptic Terminals/metabolism , rho GTP-Binding Proteins/metabolism , Animals , Calcium/metabolism , Cells, Cultured , Gene Expression , Mice , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Neurons/metabolism , Rats , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
17.
Stem Cell Res Ther ; 7(1): 166, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27846905

ABSTRACT

BACKGROUND: Cellular reprogramming is a stressful process, which requires cells to engulf somatic features and produce and maintain stemness machineries. Autophagy is a process to degrade unwanted proteins and is required for the derivation of induced pluripotent stem cells (iPSCs). However, the role of autophagy during iPSC maintenance remains undefined. METHODS: Human iPSCs were investigated by microscopy, immunofluorescence, and immunoblotting to detect autophagy machinery. Cells were treated with rapamycin to activate autophagy and with bafilomycin to block autophagy during iPSC maintenance. High concentrations of rapamycin treatment unexpectedly resulted in spontaneous formation of round floating spheres of uniform size, which were analyzed for differentiation into three germ layers. Mass spectrometry was deployed to reveal altered protein expression and pathways associated with rapamycin treatment. RESULTS: We demonstrate that human iPSCs express high basal levels of autophagy, including key components of APMKα, ULK1/2, BECLIN-1, ATG13, ATG101, ATG12, ATG3, ATG5, and LC3B. Block of autophagy by bafilomycin induces iPSC death and rapamycin attenuates the bafilomycin effect. Rapamycin treatment upregulates autophagy in iPSCs in a dose/time-dependent manner. High concentration of rapamycin reduces NANOG expression and induces spontaneous formation of round and uniformly sized embryoid bodies (EBs) with accelerated differentiation into three germ layers. Mass spectrometry analysis identifies actin cytoskeleton and adherens junctions as the major targets of rapamycin in mediating iPSC detachment and differentiation. CONCLUSIONS: High levels of basal autophagy activity are present during iPSC derivation and maintenance. Rapamycin alters expression of actin cytoskeleton and adherens junctions, induces uniform EB formation, and accelerates differentiation. IPSCs are sensitive to enzyme dissociation and require a lengthy differentiation time. The shape and size of EBs also play a role in the heterogeneity of end cell products. This research therefore highlights the potential of rapamycin in producing uniform EBs and in shortening iPSC differentiation duration.


Subject(s)
Autophagy/drug effects , Cell Adhesion/drug effects , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/physiology , Sirolimus/pharmacology , Autophagy/physiology , Cell Adhesion/physiology , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cells, Cultured , Cellular Reprogramming/drug effects , Cellular Reprogramming/physiology , Embryoid Bodies/drug effects , Embryoid Bodies/physiology , Germ Layers/drug effects , Germ Layers/physiology , Humans , Up-Regulation/drug effects , Up-Regulation/physiology
18.
ACS Chem Neurosci ; 7(3): 399-406, 2016 Mar 16.
Article in English | MEDLINE | ID: mdl-26800462

ABSTRACT

The misfolding and aggregation of proteins into amyloid fibrils characterizes many neurodegenerative disorders such as Parkinson's and Alzheimer's diseases. We report here a method, termed SAVE (single aggregate visualization by enhancement) imaging, for the ultrasensitive detection of individual amyloid fibrils and oligomers using single-molecule fluorescence microscopy. We demonstrate that this method is able to detect the presence of amyloid aggregates of α-synuclein, tau, and amyloid-ß. In addition, we show that aggregates can also be identified in human cerebrospinal fluid (CSF). Significantly, we see a twofold increase in the average aggregate concentration in CSF from Parkinson's disease patients compared to age-matched controls. Taken together, we conclude that this method provides an opportunity to characterize the structural nature of amyloid aggregates in a key biofluid, and therefore has the potential to study disease progression in both animal models and humans to enhance our understanding of neurodegenerative disorders.


Subject(s)
Amyloidogenic Proteins/cerebrospinal fluid , Diagnostic Imaging/methods , Microscopy, Fluorescence/methods , Parkinson Disease/cerebrospinal fluid , Aged , Aged, 80 and over , Benzothiazoles , Circular Dichroism/methods , Female , Humans , Male , Microscopy, Atomic Force/methods , Middle Aged , Thiazoles
19.
Antioxid Redox Signal ; 24(7): 376-91, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26564470

ABSTRACT

AIMS: Protein aggregation and oxidative stress are both key pathogenic processes in Parkinson's disease, although the mechanism by which misfolded proteins induce oxidative stress and neuronal death remains unknown. In this study, we describe how aggregation of alpha-synuclein (α-S) from its monomeric form to its soluble oligomeric state results in aberrant free radical production and neuronal toxicity. RESULTS: We first demonstrate excessive free radical production in a human induced pluripotent stem-derived α-S triplication model at basal levels and on application of picomolar doses of ß-sheet-rich α-S oligomers. We probed the effects of different structural species of α-S in wild-type rat neuronal cultures and show that both oligomeric and fibrillar forms of α-S are capable of generating free radical production, but that only the oligomeric form results in reduction of endogenous glutathione and subsequent neuronal toxicity. We dissected the mechanism of oligomer-induced free radical production and found that it was interestingly independent of several known cellular enzymatic sources. INNOVATION: The oligomer-induced reactive oxygen species (ROS) production was entirely dependent on the presence of free metal ions as addition of metal chelators was able to block oligomer-induced ROS production and prevent oligomer-induced neuronal death. CONCLUSION: Our findings further support the causative role of soluble amyloid oligomers in triggering neurodegeneration and shed light into the mechanisms by which these species cause neuronal damage, which, we show here, can be amenable to modulation through the use of metal chelation.


Subject(s)
Ions/metabolism , Metals/metabolism , Neurons/metabolism , Oxidative Stress , Parkinson Disease/metabolism , Protein Multimerization , alpha-Synuclein/metabolism , Amyloid beta-Peptides/metabolism , Animals , Apoptosis , Caspase 3/metabolism , Caspase 7/metabolism , Cell Differentiation , Enzyme Activation , Gene Duplication , Glutathione/metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Neurons/cytology , Neurons/pathology , Parkinson Disease/genetics , Parkinson Disease/pathology , Protein Aggregation, Pathological , Protein Conformation , Rats , Reactive Oxygen Species/metabolism , Structure-Activity Relationship , alpha-Synuclein/chemistry , alpha-Synuclein/genetics
20.
Neurobiol Dis ; 90: 27-34, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26707701

ABSTRACT

Neurons are highly polarised cells with an elaborate and diverse cytoarchitecture. But this complex architecture presents a major problem: how to appropriately distribute metabolic resources where they are most needed within the cell. The solution comes in the form of mitochondria: highly dynamic organelles subject to a repertoire of trafficking, fission/fusion and quality control systems which work in concert to orchestrate a precisely distributed and healthy mitochondrial network. Mitochondria are critical for maintaining local energy supply and buffering Ca(2+) flux within neurons, and are increasingly recognised as being essential for healthy neuronal function. Mitochondrial movements are facilitated by their coupling to microtubule-based transport via kinesin and dynein motors. Adaptor proteins are required for this coupling and the mitochondrial Rho GTPases Miro1 and Miro2 are core components of this machinery. Both Miros have Ca(2+)-sensing and GTPase domains, and are therefore ideally suited to coordinating mitochondrial dynamics with intracellular signalling pathways and local energy turnover. In this review, we focus on Miro's role in mediating mitochondrial transport in neurons, and the relevance of these mechanisms to neuronal health and disease.


Subject(s)
Mitochondrial Dynamics/physiology , Mitochondrial Proteins/metabolism , Neurons/metabolism , rho GTP-Binding Proteins/metabolism , Animals , Humans
SELECTION OF CITATIONS
SEARCH DETAIL