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

Country/Region as subject
Publication year range
1.
Glia ; 70(5): 842-857, 2022 05.
Article in English | MEDLINE | ID: mdl-34978340

ABSTRACT

In amyotrophic lateral sclerosis (ALS) caused by SOD1 gene mutations, both cell-autonomous and noncell-autonomous mechanisms lead to the selective degeneration of motoneurons (MN). Here, we evaluate the therapeutic potential of gene therapy targeting mutated SOD1 in mature astrocytes using mice expressing the mutated SOD1G93A protein. An AAV-gfaABC1 D vector encoding an artificial microRNA is used to deliver RNA interference against mutated SOD1 selectively in astrocytes. The treatment leads to the progressive rescue of neuromuscular junction occupancy, to the recovery of the compound muscle action potential in the gastrocnemius muscle, and significantly improves neuromuscular function. In the spinal cord, gene therapy targeting astrocytes protects a small pool of the most vulnerable fast-fatigable MN until disease end stage. In the gastrocnemius muscle of the treated SOD1G93A mice, the fast-twitch type IIB muscle fibers are preserved from atrophy. Axon collateral sprouting is observed together with muscle fiber type grouping indicative of denervation/reinnervation events. The transcriptome profiling of spinal cord MN shows changes in the expression levels of factors regulating the dynamics of microtubules. Gene therapy delivering RNA interference against mutated SOD1 in astrocytes protects fast-fatigable motor units and thereby improves neuromuscular function in ALS mice.


Subject(s)
Amyotrophic Lateral Sclerosis , Superoxide Dismutase-1/metabolism , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/therapy , Animals , Astrocytes/metabolism , Disease Models, Animal , Mice , Mice, Transgenic , Motor Neurons/metabolism , RNA Interference , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Superoxide Dismutase-1/genetics
2.
Hum Mol Genet ; 26(3): 582-598, 2017 02 01.
Article in English | MEDLINE | ID: mdl-28053050

ABSTRACT

To understand the cause of Parkinson's disease (PD), it is important to determine the functional interactions between factors linked to the disease. Parkin is associated with autosomal recessive early-onset PD, and controls the transcription of PGC-1α, a master regulator of mitochondrial biogenesis. These two factors functionally interact to regulate the turnover and quality of mitochondria, by increasing both mitophagic activity and mitochondria biogenesis. In cortical neurons, co-expressing PGC-1α and Parkin increases the number of mitochondria, enhances maximal respiration, and accelerates the recovery of the mitochondrial membrane potential following mitochondrial uncoupling. PGC-1α enhances Mfn2 transcription, but also leads to increased degradation of the Mfn2 protein, a key ubiquitylation target of Parkin on mitochondria. In vivo, Parkin has significant protective effects on the survival and function of nigral dopaminergic neurons in which the chronic expression of PGC-1α is induced. Ultrastructural analysis shows that these two factors together control the density of mitochondria and their interaction with the endoplasmic reticulum. These results highlight the combined effects of Parkin and PGC-1α in the maintenance of mitochondrial homeostasis in dopaminergic neurons. These two factors synergistically control the quality and function of mitochondria, which is important for the survival of neurons in Parkinson's disease.


Subject(s)
GTP Phosphohydrolases/genetics , Mitochondria/genetics , Mitochondrial Proteins/genetics , Parkinsonian Disorders/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Ubiquitin-Protein Ligases/genetics , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Dopaminergic Neurons/ultrastructure , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , Gene Expression Regulation , Humans , Membrane Potential, Mitochondrial/genetics , Mitochondria/pathology , Mitochondria/ultrastructure , Organelle Biogenesis , Oxidative Stress/genetics , Parkinsonian Disorders/metabolism , Parkinsonian Disorders/pathology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Proteolysis , Ubiquitin-Protein Ligases/metabolism
3.
Brain ; 139(Pt 5): 1587-604, 2016 05.
Article in English | MEDLINE | ID: mdl-26956423

ABSTRACT

Passive immunization against misfolded toxic proteins is a promising approach to treat neurodegenerative disorders. For effective immunotherapy against Alzheimer's disease, recent clinical data indicate that monoclonal antibodies directed against the amyloid-ß peptide should be administered before the onset of symptoms associated with irreversible brain damage. It is therefore critical to develop technologies for continuous antibody delivery applicable to disease prevention. Here, we addressed this question using a bioactive cellular implant to deliver recombinant anti-amyloid-ß antibodies in the subcutaneous tissue. An encapsulating device permeable to macromolecules supports the long-term survival of myogenic cells over more than 10 months in immunocompetent allogeneic recipients. The encapsulated cells are genetically engineered to secrete high levels of anti-amyloid-ß antibodies. Peripheral implantation leads to continuous antibody delivery to reach plasma levels that exceed 50 µg/ml. In a proof-of-concept study, we show that the recombinant antibodies produced by this system penetrate the brain and bind amyloid plaques in two mouse models of the Alzheimer's pathology. When encapsulated cells are implanted before the onset of amyloid plaque deposition in TauPS2APP mice, chronic exposure to anti-amyloid-ß antibodies dramatically reduces amyloid-ß40 and amyloid-ß42 levels in the brain, decreases amyloid plaque burden, and most notably, prevents phospho-tau pathology in the hippocampus. These results support the use of encapsulated cell implants for passive immunotherapy against the misfolded proteins, which accumulate in Alzheimer's disease and other neurodegenerative disorders.


Subject(s)
Alzheimer Disease/prevention & control , Amyloid beta-Peptides/antagonists & inhibitors , Antibodies, Monoclonal/pharmacology , Drug Implants , Immunization, Passive/methods , Tauopathies/prevention & control , Amyloid beta-Peptides/immunology , Amyloid beta-Peptides/metabolism , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/blood , Antibodies, Monoclonal/pharmacokinetics , Brain/metabolism , Cells, Cultured , Mice , Mice, Transgenic , Neuroprotection , Peptide Fragments/metabolism , Plaque, Amyloid/metabolism , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacokinetics , Recombinant Proteins/pharmacology , Subcutaneous Absorption
4.
Int J Neurosci ; 127(1): 28-36, 2017 Jan.
Article in English | MEDLINE | ID: mdl-26856727

ABSTRACT

Neuromodulation by spinal cord stimulation has been proposed as a symptomatic treatment for Parkinson's disease. We tested the chronic effects of spinal cord stimulation in a progressive model of Parkinson's based on overexpression of alpha-synuclein in the substantia nigra. Adult Sprague Dawley rats received unilateral injections of adeno-associated virus serotype 6 (AAV6) in the substantia nigra to express alpha-synuclein. Locomotion and forepaw use of the rats were evaluated during the next 10 weeks. Starting on week 6, a group of AAV6-injected rats received spinal cord stimulation once a week. At the end of the experiment, tyrosine hydroxylase and alpha-synuclein immunostaining were performed. Rats with unilateral alpha-synuclein expression showed a significant decrease in the use of the contralateral forepaw, which was mildly but significantly reverted by spinal cord stimulation applied once a week from the 6th to the 10th week after the AAV6 injection. Long-term spinal cord stimulation proved to be effective to suppress or delay motor symptoms in a sustained and progressive model of Parkinson's and might become an alternative, less invasive neuromodulation option to treat this disease.


Subject(s)
Behavior, Animal/physiology , Parkinson Disease/therapy , Spinal Cord Stimulation/methods , Animals , Disease Models, Animal , Male , Rats , Rats, Sprague-Dawley , Substantia Nigra/metabolism , alpha-Synuclein/metabolism
5.
J Neurosci ; 35(10): 4168-78, 2015 Mar 11.
Article in English | MEDLINE | ID: mdl-25762664

ABSTRACT

Excitatory synaptic transmission is accompanied by a local surge in interstitial lactate that occurs despite adequate oxygen availability, a puzzling phenomenon termed aerobic glycolysis. In addition to its role as an energy substrate, recent studies have shown that lactate modulates neuronal excitability acting through various targets, including NMDA receptors and G-protein-coupled receptors specific for lactate, but little is known about the cellular and molecular mechanisms responsible for the increase in interstitial lactate. Using a panel of genetically encoded fluorescence nanosensors for energy metabolites, we show here that mouse astrocytes in culture, in cortical slices, and in vivo maintain a steady-state reservoir of lactate. The reservoir was released to the extracellular space immediately after exposure of astrocytes to a physiological rise in extracellular K(+) or cell depolarization. Cell-attached patch-clamp analysis of cultured astrocytes revealed a 37 pS lactate-permeable ion channel activated by cell depolarization. The channel was modulated by lactate itself, resulting in a positive feedback loop for lactate release. A rapid fall in intracellular lactate levels was also observed in cortical astrocytes of anesthetized mice in response to local field stimulation. The existence of an astrocytic lactate reservoir and its quick mobilization via an ion channel in response to a neuronal cue provides fresh support to lactate roles in neuronal fueling and in gliotransmission.


Subject(s)
Astrocytes/drug effects , Ion Channels/physiology , Lactic Acid/metabolism , Potassium/pharmacology , Animals , Animals, Newborn , Barium/pharmacology , Cadmium/pharmacology , Cells, Cultured , Cerebral Cortex/cytology , Female , Fluoresceins/metabolism , Glycogen/metabolism , Humans , In Vitro Techniques , Ion Channels/drug effects , Ions/pharmacology , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mice , Mice, Inbred C57BL , Neurons/drug effects , Neurons/physiology , Pyruvic Acid/pharmacology , Somatosensory Cortex/cytology , Somatosensory Cortex/physiology , Transfection
6.
Hum Mol Genet ; 23(18): 4887-905, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-24794857

ABSTRACT

Mutations in LRRK2 are one of the primary genetic causes of Parkinson's disease (PD). LRRK2 contains a kinase and a GTPase domain, and familial PD mutations affect both enzymatic activities. However, the signaling mechanisms regulating LRRK2 and the pathogenic effects of familial mutations remain unknown. Identifying the signaling proteins that regulate LRRK2 function and toxicity remains a critical goal for the development of effective therapeutic strategies. In this study, we apply systems biology tools to human PD brain and blood transcriptomes to reverse-engineer a LRRK2-centered gene regulatory network. This network identifies several putative master regulators of LRRK2 function. In particular, the signaling gene RGS2, which encodes for a GTPase-activating protein (GAP), is a key regulatory hub connecting the familial PD-associated genes DJ-1 and PINK1 with LRRK2 in the network. RGS2 expression levels are reduced in the striata of LRRK2 and sporadic PD patients. We identify RGS2 as a novel interacting partner of LRRK2 in vivo. RGS2 regulates both the GTPase and kinase activities of LRRK2. We show in mammalian neurons that RGS2 regulates LRRK2 function in the control of neuronal process length. RGS2 is also protective against neuronal toxicity of the most prevalent mutation in LRRK2, G2019S. We find that RGS2 regulates LRRK2 function and neuronal toxicity through its effects on kinase activity and independently of GTPase activity, which reveals a novel mode of action for GAP proteins. This work identifies RGS2 as a promising target for interfering with neurodegeneration due to LRRK2 mutations in PD patients.


Subject(s)
Gene Regulatory Networks , Neurons/pathology , Parkinson Disease/genetics , Parkinson Disease/pathology , Protein Serine-Threonine Kinases/metabolism , RGS Proteins/metabolism , Animals , Brain/metabolism , Caenorhabditis elegans/metabolism , Gene Expression Regulation , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Neurons/metabolism , Oncogene Proteins/metabolism , Parkinson Disease/blood , Protein Deglycase DJ-1 , Protein Kinases/metabolism , Systems Biology/methods , Transcriptome
7.
Proc Natl Acad Sci U S A ; 110(41): E3945-54, 2013 Oct 08.
Article in English | MEDLINE | ID: mdl-23983262

ABSTRACT

An increase in α-synuclein levels due to gene duplications/triplications or impaired degradation is sufficient to trigger its aggregation and cause familial Parkinson disease (PD). Therefore, lowering α-synuclein levels represents a viable therapeutic strategy for the treatment of PD and related synucleinopathies. Here, we report that Polo-like kinase 2 (PLK2), an enzyme up-regulated in synucleinopathy-diseased brains, interacts with, phosphorylates and enhances α-synuclein autophagic degradation in a kinase activity-dependent manner. PLK2-mediated degradation of α-synuclein requires both phosphorylation at S129 and PLK2/α-synuclein complex formation. In a rat genetic model of PD, PLK2 overexpression reduces intraneuronal human α-synuclein accumulation, suppresses dopaminergic neurodegeneration, and reverses hemiparkinsonian motor impairments induced by α-synuclein overexpression. This PLK2-mediated neuroprotective effect is also dependent on PLK2 activity and α-synuclein phosphorylation. Collectively, our findings demonstrate that PLK2 is a previously undescribed regulator of α-synuclein turnover and that modulating its kinase activity could be a viable target for the treatment of synucleinopathies.


Subject(s)
Gene Expression Regulation/physiology , Parkinson Disease/metabolism , Protein Serine-Threonine Kinases/metabolism , Proteolysis , alpha-Synuclein/metabolism , Analysis of Variance , Animals , DNA Primers/genetics , Electrophoresis, Polyacrylamide Gel , HEK293 Cells , Humans , Immunohistochemistry , Immunoprecipitation , Phosphorylation , Rats
8.
Mol Ther ; 21(8): 1602-10, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23712039

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease characterized by the progressive loss of motor neurons in the brain and spinal cord. We have recently shown that human mesenchymal stem cells (hMSCs) modified to release glial cell line-derived neurotrophic factor (GDNF) decrease disease progression in a rat model of ALS when delivered to skeletal muscle. In the current study, we determined whether or not this effect could be enhanced by delivering GDNF in concert with other trophic factors. hMSC engineered to secrete GDNF (hMSC-GDNF), vascular endothelial growth factor (hMSC-VEGF), insulin-like growth factor-I (hMSC-IGF-I), or brain-derived neurotrophic factor (hMSC-BDNF), were prepared and transplanted bilaterally into three muscle groups. hMSC-GDNF and hMSC-VEGF prolonged survival and slowed the loss of motor function, but hMSC-IGF-I and hMSC-BDNF did not have any effect. We then tested the efficacy of a combined ex vivo delivery of GDNF and VEGF in extending survival and protecting neuromuscular junctions (NMJs) and motor neurons. Interestingly, the combined delivery of these neurotrophic factors showed a strong synergistic effect. These studies further support ex vivo gene therapy approaches for ALS that target skeletal muscle.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Glial Cell Line-Derived Neurotrophic Factor/genetics , Longevity/genetics , Vascular Endothelial Growth Factor A/genetics , Amyotrophic Lateral Sclerosis/mortality , Animals , Cell Survival/genetics , Disease Models, Animal , Disease Progression , Female , Gene Expression , Gene Transfer Techniques , Genetic Therapy , Humans , Male , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Motor Neurons/metabolism , Muscle, Skeletal/metabolism , Neuromuscular Junction/metabolism , Rats
9.
J Neurosci ; 32(5): 1536-44, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22302797

ABSTRACT

Several lines of evidence suggest that phosphorylation of α-synuclein (α-syn) at S87 or S129 may play an important role in regulating its aggregation, fibrillogenesis, Lewy body formation, and neurotoxicity in vivo. However, whether phosphorylation at these residues enhances or protects against α-syn toxicity in vivo remains unknown. In this study, we investigated the cellular and behavioral effect of overexpression of wild-type (WT), S87A, and S87E α-syn to block or to mimic S87 phosphorylation, respectively, in the substantia nigra of Wistar rats using recombinant adeno-associated vectors. Our results revealed that WT and S87A overexpression induced α-syn aggregation, loss of dopaminergic neurons, and fiber pathology. These neuropathological effects correlated well with the induction of hemi-parkinsonian motor symptoms. Strikingly, overexpression of the phosphomimic mutant S87E did not show any toxic effect on dopaminergic neurons and resulted in significantly less α-syn aggregates, dystrophic fibers, and motor impairment. Together, our data demonstrate, for the first time, that mimicking phosphorylation at S87 inhibits α-syn aggregation and protects against α-syn-induced toxicity in vivo, suggesting that phosphorylation at this residue would play an important role in controlling α-syn neuropathology. In addition, our results provide strong evidence for a direct correlation between α-syn-induced neurotoxicity, fiber pathology, and motor impairment and the extent of α-syn aggregation in vivo, suggesting that lowering α-syn levels and/or blocking its aggregation are viable therapeutic strategies for the treatment of Parkinson's disease and related synucleinopathies.


Subject(s)
Disease Models, Animal , Parkinson Disease/metabolism , Serine/metabolism , alpha-Synuclein/metabolism , Animals , Corpus Striatum/metabolism , Corpus Striatum/pathology , Gene Transfer Techniques , Humans , Male , Mutation/genetics , Parkinson Disease/pathology , Parkinson Disease/prevention & control , Phosphorylation/physiology , Rats , Rats, Wistar , Serine/genetics , alpha-Synuclein/genetics , alpha-Synuclein/toxicity
10.
J Biol Chem ; 287(19): 15345-64, 2012 May 04.
Article in English | MEDLINE | ID: mdl-22315227

ABSTRACT

Since the discovery and isolation of α-synuclein (α-syn) from human brains, it has been widely accepted that it exists as an intrinsically disordered monomeric protein. Two recent studies suggested that α-syn produced in Escherichia coli or isolated from mammalian cells and red blood cells exists predominantly as a tetramer that is rich in α-helical structure (Bartels, T., Choi, J. G., and Selkoe, D. J. (2011) Nature 477, 107-110; Wang, W., Perovic, I., Chittuluru, J., Kaganovich, A., Nguyen, L. T. T., Liao, J., Auclair, J. R., Johnson, D., Landeru, A., Simorellis, A. K., Ju, S., Cookson, M. R., Asturias, F. J., Agar, J. N., Webb, B. N., Kang, C., Ringe, D., Petsko, G. A., Pochapsky, T. C., and Hoang, Q. Q. (2011) Proc. Natl. Acad. Sci. 108, 17797-17802). However, it remains unknown whether or not this putative tetramer is the main physiological form of α-syn in the brain. In this study, we investigated the oligomeric state of α-syn in mouse, rat, and human brains. To assess the conformational and oligomeric state of native α-syn in complex mixtures, we generated α-syn standards of known quaternary structure and conformational properties and compared the behavior of endogenously expressed α-syn to these standards using native and denaturing gel electrophoresis techniques, size-exclusion chromatography, and an oligomer-specific ELISA. Our findings demonstrate that both human and rodent α-syn expressed in the central nervous system exist predominantly as an unfolded monomer. Similar results were observed when human α-syn was expressed in mouse and rat brains as well as mammalian cell lines (HEK293, HeLa, and SH-SY5Y). Furthermore, we show that α-syn expressed in E. coli and purified under denaturing or nondenaturing conditions, whether as a free protein or as a fusion construct with GST, is monomeric and adopts a disordered conformation after GST removal. These results do not rule out the possibility that α-syn becomes structured upon interaction with other proteins and/or biological membranes.


Subject(s)
Brain/metabolism , Erythrocytes/metabolism , Recombinant Proteins/metabolism , alpha-Synuclein/metabolism , Amino Acid Sequence , Animals , Cell Line, Tumor , Central Nervous System/metabolism , Chromatography, Gel , Enzyme-Linked Immunosorbent Assay , Escherichia coli/genetics , HEK293 Cells , HeLa Cells , Humans , Immunoblotting , Mice , Mice, Transgenic , Molecular Sequence Data , Mutation , Protein Conformation , Protein Structure, Secondary , Protein Unfolding , Rats , Rats, Sprague-Dawley , Recombinant Proteins/chemistry , alpha-Synuclein/chemistry , alpha-Synuclein/genetics
12.
J Neurosci ; 31(3): 907-12, 2011 Jan 19.
Article in English | MEDLINE | ID: mdl-21248115

ABSTRACT

The G2019S mutation in the leucine-rich repeat kinase 2 (LRRK2) gene is the most common genetic cause of Parkinson's disease (PD), accounting for a significant proportion of both autosomal dominant familial and sporadic PD cases. Our aim in the present study is to generate a mammalian model of mutant G2019S LRRK2 pathogenesis, which reproduces the robust nigral neurodegeneration characteristic of PD. We developed adenoviral vectors to drive neuron-specific expression of full-length wild-type or mutant G2019S human LRRK2 in the nigrostriatal system of adult rats. Wild-type LRRK2 did not induce any significant neuronal loss. In contrast, under the same conditions and levels of expression, G2019S mutant LRRK2 causes a progressive degeneration of nigral dopaminergic neurons. Our data provide a novel rat model of PD, based on a prevalent genetic cause, that reproduces a cardinal feature of the disease within a rapid time frame suitable for testing of neuroprotective strategies.


Subject(s)
Brain/pathology , Disease Models, Animal , Nerve Degeneration/pathology , Neurons/pathology , Parkinson Disease/pathology , Protein Serine-Threonine Kinases/genetics , Analysis of Variance , Animals , Blotting, Western , Brain/metabolism , Cell Count , Dopamine/metabolism , Female , Immunohistochemistry , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Mutation , Nerve Degeneration/genetics , Nerve Degeneration/metabolism , Neurons/metabolism , Parkinson Disease/genetics , Parkinson Disease/metabolism , Protein Serine-Threonine Kinases/metabolism , Rats , Rats, Wistar
13.
Neurobiol Dis ; 48(2): 189-201, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22227451

ABSTRACT

Parkinson's disease is a neurodegenerative disorder characterized by the progressive loss of dopaminergic neurons in the substantia nigra. While sporadic in the majority of cases, PD-linked dominant mutations in the α-synuclein and LRRK-2 genes, and recessive mutations in the parkin, DJ-1 and PINK-1 genes have been identified in PD families in recent years. In this review we describe viral animal models for PD, i.e. models that are based on PD-associated mutations, and have been generated by viral delivery of the respective disease genes to the substantia nigra of rodents and non-human primates. To date, viral PD models comprise α-synuclein and LRRK-2-based overexpression models, as well as models that mimic parkin loss of function by overexpression of the parkin substrates Pael-R, CDCrel-1, p38/JTV or synphilin-1. These viral models provide valuable insights into Parkinson disease mechanisms, help to identify therapeutic targets and may contribute to the development of therapeutic approaches.


Subject(s)
Genetic Therapy/methods , Genetic Vectors/genetics , Parkinson Disease/genetics , Parkinson Disease/therapy , Viruses/genetics , Animals , Behavior, Animal/physiology , Carrier Proteins/genetics , Cell Cycle Proteins/genetics , Disease Models, Animal , Humans , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Nerve Tissue Proteins/genetics , Parkinson Disease/physiopathology , Parkinson Disease/psychology , Protein Serine-Threonine Kinases/genetics , Receptors, G-Protein-Coupled/genetics , Septins/genetics , Ubiquitin-Protein Ligases/genetics , alpha-Synuclein/genetics , p38 Mitogen-Activated Protein Kinases/genetics
14.
Acta Neuropathol ; 123(5): 653-69, 2012 May.
Article in English | MEDLINE | ID: mdl-22361813

ABSTRACT

α-Synuclein (α-syn) is a presynaptic protein present at most nerve terminals, but its function remains largely unknown. The familial forms of Parkinson's disease associated with multiplications of the α-syn gene locus indicate that overabundance of this protein might have a detrimental effect on dopaminergic transmission. To investigate this hypothesis, we use adeno-associated viral (AAV) vectors to overexpress human α-syn in the rat substantia nigra. Moderate overexpression of either wild-type (WT) or A30P α-syn differs in the motor phenotypes induced, with only the WT form generating hemiparkinsonian impairments. Wild-type α-syn causes a reduction of dopamine release in the striatum that exceeds the loss of dopaminergic neurons, axonal fibers, and the reduction in total dopamine. At the ultrastructural level, the reduced dopamine release corresponds to a decreased density of dopaminergic vesicles and synaptic contacts in striatal terminals. Interestingly, the membrane-binding-deficient A30P mutant does neither notably reduce dopamine release nor it cause ultrastructural changes in dopaminergic axons, showing that α-syn's membrane-binding properties are critically involved in the presynaptic defects. To further determine if the affinity of the protein for membranes determines the extent of motor defects, we compare three forms of α-syn in conditions leading to pronounced degeneration. While membrane-binding α-syns (wild-type and A53T) induce severe motor impairments, an N-terminal deleted form with attenuated affinity for membranes is inefficient in inducing motor defects. Overall, these results demonstrate that α-syn overabundance is detrimental to dopamine neurotransmission at early stages of the degeneration of nigrostriatal dopaminergic axons.


Subject(s)
Corpus Striatum/metabolism , Dopamine/deficiency , Intermediate Filament Proteins/metabolism , Motor Activity/physiology , Substantia Nigra/metabolism , Synaptic Vesicles/metabolism , 3,4-Dihydroxyphenylacetic Acid/metabolism , Amphetamine/pharmacology , Analysis of Variance , Animals , Apomorphine/pharmacology , Chromatography, High Pressure Liquid/methods , Corpus Striatum/drug effects , Dopamine Agonists/pharmacology , Dopamine Uptake Inhibitors/pharmacology , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/ultrastructure , Electrochemistry , Enzyme-Linked Immunosorbent Assay/methods , Exploratory Behavior/drug effects , Exploratory Behavior/physiology , Female , Functional Laterality , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Genetic Vectors/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Homovanillic Acid/metabolism , Humans , In Vitro Techniques , Intermediate Filament Proteins/genetics , Intermediate Filament Proteins/ultrastructure , Microscopy, Electron, Transmission , Motor Activity/drug effects , Mutation/genetics , Potassium/pharmacology , Rats , Rats, Sprague-Dawley , Substantia Nigra/drug effects , Substantia Nigra/ultrastructure , Synaptic Vesicles/ultrastructure , Time Factors , Transduction, Genetic , Vesicular Monoamine Transport Proteins/metabolism
15.
Nat Med ; 11(4): 423-8, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15768028

ABSTRACT

Mutations in Cu/Zn superoxide dismutase (encoded by SOD1), one of the causes of familial amyotrophic lateral sclerosis (ALS), lead to progressive death of motoneurons through a gain-of-function mechanism. RNA interference (RNAi) mediated by viral vectors allows for long-term reduction in gene expression and represents an attractive therapeutic approach for genetic diseases characterized by acquired toxic properties. We report that in SOD1(G93A) transgenic mice, a model for familial ALS, intraspinal injection of a lentiviral vector that produces RNAi-mediated silencing of SOD1 substantially retards both the onset and the progression rate of the disease.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , RNA Interference , Superoxide Dismutase/genetics , Animals , Disease Models, Animal , Disease Progression , Genetic Vectors , Humans , Lentivirus , Mice , Mice, Transgenic , Molecular Sequence Data , Mutation , RNA, Small Interfering
16.
Mol Ther ; 19(2): 274-83, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21102563

ABSTRACT

A major challenge in neurological gene therapy is delivery of the transgene to sufficient cell numbers in an atraumatic manner. This is particularly difficult for motor neuron (MN) diseases that have cells located across the entire spinal cord, brain stem, and cortex. We have used the familial mouse model of amyotrophic lateral sclerosis (ALS) to examine the feasibility of body-wide intramuscular injections of adeno-associated virus serotype 6 (AAV6), a vector capable of axonal retrograde transport, to deliver therapeutic genetic information across the lower MN axis. Neonatal muscle delivery of AAV expressing small hairpin RNAs (shRNAs) against the toxic transgene in this model, human mutant superoxide dismutase 1 (mSOD1), led to significant mSOD1 knockdown in the muscle as well as innervating MNs. This knockdown conferred neuroprotection and halted muscle atrophy in individually targeted MN pools. However, despite the vector being targeted to MNs that innervate muscle groups controlling eating, breathing, and locomotion, this approach was unable to therapeutically impact on disease progression in the ALS mouse model. These results stress the complexity of gene delivery for mSOD1 silencing and suggest that critical thresholds of protein knockdown and transduction across various cell types are required to translate local neuroprotective effects into functional improvements.


Subject(s)
Dependovirus/genetics , Genetic Vectors/genetics , Motor Neurons/metabolism , Superoxide Dismutase/genetics , Amyotrophic Lateral Sclerosis/therapy , Animals , Disease Models, Animal , Electromyography , Genetic Therapy/methods , Humans , Injections, Intramuscular , Mice , Mice, Transgenic , RNA Interference/physiology , RNA, Small Interfering/genetics , RNA, Small Interfering/physiology , Superoxide Dismutase-1
17.
Cell Rep Med ; 3(5): 100633, 2022 05 17.
Article in English | MEDLINE | ID: mdl-35584623

ABSTRACT

Targeting mitophagy to activate the recycling of faulty mitochondria during aging is a strategy to mitigate muscle decline. We present results from a randomized, placebo-controlled trial in middle-aged adults where we administer a postbiotic compound Urolithin A (Mitopure), a known mitophagy activator, at two doses for 4 months (NCT03464500). The data show significant improvements in muscle strength (∼12%) with intake of Urolithin A. We observe clinically meaningful improvements with Urolithin A on aerobic endurance (peak oxygen oxygen consumption [VO2]) and physical performance (6 min walk test) but do not notice a significant improvement on peak power output (primary endpoint). Levels of plasma acylcarnitines and C-reactive proteins are significantly lower with Urolithin A, indicating higher mitochondrial efficiency and reduced inflammation. We also examine expression of proteins linked to mitophagy and mitochondrial metabolism in skeletal muscle and find a significant increase with Urolithin A administration. This study highlights the benefit of Urolithin A to improve muscle performance.


Subject(s)
Mitophagy , Muscle Strength , Biomarkers , Coumarins , Mitochondria
18.
JAMA Netw Open ; 5(1): e2144279, 2022 01 04.
Article in English | MEDLINE | ID: mdl-35050355

ABSTRACT

Importance: Aging is associated with a decline in mitochondrial function and reduced exercise capacity. Urolithin A is a natural gut microbiome-derived food metabolite that has been shown to stimulate mitophagy and improve muscle function in older animals and to induce mitochondrial gene expression in older humans. Objective: To investigate whether oral administration of urolithin A improved the 6-minute walk distance, muscle endurance in hand and leg muscles, and biomarkers associated with mitochondrial and cellular health. Design, Setting, and Participants: This double-blind, placebo-controlled randomized clinical trial in adults aged 65 to 90 years was conducted at a medical center and a cancer research center in Seattle, Washington, from March 1, 2018, to July 30, 2020. Muscle fatigue tests and plasma analysis of biomarkers were assessed at baseline, 2 months, and 4 months. Six-minute walk distance and maximal ATP production were assessed using magnetic resonance spectroscopy at baseline and at the end of study at 4 months. The analysis used an intention-to-treat approach. Interventions: Participants were randomized to receive daily oral supplementation with either 1000 mg urolithin A or placebo for 4 months. Main Outcomes and Measures: The primary end point was change from baseline in the 6-minute walk distance and change from baseline to 4 months in maximal ATP production in the hand skeletal muscle. The secondary end points were change in muscle endurance of 2 skeletal muscles (tibialis anterior [TA] in the leg and first dorsal interosseus [FDI] in the hand). Cellular health biomarkers were investigated via plasma metabolomics. Adverse events were recorded and compared between the 2 groups during the intervention period. Results: A total of 66 participants were randomized to either the urolithin A (n = 33) or the placebo (n = 33) intervention group. These participants had a mean (SD) age of 71.7 (4.94) years, were predominantly women (50 [75.8%]), and were all White individuals. Urolithin A, compared with placebo, significantly improved muscle endurance (ie, increase in the number of muscle contractions until fatigue from baseline) in the FDI and TA at 2 months (urolithin A: FDI, 95.3 [115.5] and TA, 41.4 [65.5]; placebo: FDI, 11.6 [147.4] and TA, 5.7 [127.1]). Plasma levels of several acylcarnitines, ceramides, and C-reactive protein were decreased by urolithin A, compared with placebo, at 4 months (baseline vs 4 mo: urolithin A, 2.14 [2.15] vs 2.07 [1.46]; placebo, 2.17 [2.52] vs 2.65 [1.86]). The mean (SD) increase from baseline in the 6-minute walk distance was 60.8 (67.2) m in the urolithin A group and 42.5 (73.3) m in the placebo group. The mean (SD) change from baseline to 4 months in maximal ATP production in the FDI was 0.07 (0.23) mM/s in the urolithin A group and 0.06 (0.20) mM/s in the placebo group; for the TA, it was -0.03 (0.10) mM/s in the urolithin A group and 0.03 (0.10) mM/s in the placebo group. These results showed no significant improvement with urolithin A supplementation compared with placebo. No statistical differences in adverse events were observed between the 2 groups. Conclusions and Relevance: This randomized clinical trial found that urolithin A supplementation was safe and well tolerated in the assessed population. Although the improvements in the 6-minute walk distance and maximal ATP production in the hand muscle were not significant in the urolithin A group vs the placebo group, long-term urolithin A supplementation was beneficial for muscle endurance and plasma biomarkers, suggesting that urolithin A may counteract age-associated muscle decline; however, future work is needed to confirm this finding. Trial Registration: ClinicalTrials.gov Identifier: NCT03283462.


Subject(s)
Coumarins/therapeutic use , Dietary Supplements , Muscle, Skeletal/metabolism , Vital Capacity/drug effects , Walk Test , Adult , Antioxidants/therapeutic use , Double-Blind Method , Female , Humans , Male , Middle Aged , Walking
19.
Eur J Clin Nutr ; 76(2): 297-308, 2022 02.
Article in English | MEDLINE | ID: mdl-34117375

ABSTRACT

BACKGROUND: Urolithin A (UA) is produced by gut microflora from foods rich in ellagitannins. UA has been shown to improve mitochondrial health preclinically and in humans. Not everyone has a microbiome capable of producing UA, making supplementation with UA an appealing strategy. OBJECTIVE: This is the first detailed investigation of the prevalence of UA producers in a healthy population and the ability of direct UA supplementation to overcome both microbiome and dietary variability. Dietary intake of a glass of pomegranate juice (PJ) was used to assess UA producer status (n = 100 participants) and to characterize differences in gut microbiome between UA producers from non-producers. METHODS: Subjects were randomized (1:1) to either PJ or a food product containing UA (500 mg). Prevalence of UA producers and non-producers were determined in the PJ group. Diet questionnaires and fecal samples were collected to compare differences between UA producers and non-producers along with plasma samples at different time points to assess levels of UA and its conjugates between the interventions. RESULTS: Only 12% of subjects had detectable levels of UA at baseline. Following PJ intake ~40% of the subjects converted significantly the precursor compounds into UA. UA producers were distinguished by a significantly higher gut microbiome diversity and ratio of Firmicutes to Bacteroides. Direct supplementation with UA significantly increased plasma levels and provided a >6-fold exposure to UA vs. PJ (p < 0.0001). CONCLUSIONS: Differences in gut microbiome and diet that dictate natural exposure to UA can be overcome via direct dietary UA supplementation.


Subject(s)
Gastrointestinal Microbiome , Adult , Coumarins/pharmacology , Dietary Exposure , Dietary Supplements , Humans
20.
J Neurosci ; 30(2): 785-96, 2010 Jan 13.
Article in English | MEDLINE | ID: mdl-20071543

ABSTRACT

Embryonic motoneurons from mutant SOD1 (mSOD1) mouse models of amyotrophic lateral sclerosis (ALS), but not wild-type motoneurons, can be triggered to die by exposure to nitric oxide (NO), leading to activation of a motoneuron-specific signaling pathway downstream of the death receptor Fas/CD95. To identify effectors of mSOD1-dependent cell death, we performed a proteomic analysis. Treatment of cultured mSOD1 motoneurons with NO led to a 2.5-fold increase in levels of collapsin response mediator protein 4a (CRMP4a). In vivo, the percentage of mSOD1 lumbar motoneurons expressing CRMP4 in mSOD1 mice increased progressively from presymptomatic to early-onset stages, reaching a maximum of 25%. Forced adeno-associated virus (AAV)-mediated expression of CRMP4a in wild-type motoneurons in vitro triggered a process of axonal degeneration and cell death affecting 60% of motoneurons, whereas silencing of CRMP4a in mSOD1 motoneurons protected them from NO-induced death. In vivo, AAV-mediated overexpression of CRMP4a but not CRMP2 led to the death of 30% of the lumbar motoneurons and an 18% increase in denervation of neuromuscular junctions in the gastrocnemius muscle. Our data identify CRMP4a as a potential early effector in the neurodegenerative process in ALS.


Subject(s)
Amyotrophic Lateral Sclerosis , Motor Neurons/metabolism , Nerve Degeneration/genetics , Nerve Tissue Proteins/metabolism , Superoxide Dismutase/genetics , Up-Regulation/genetics , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Amyotrophic Lateral Sclerosis/physiopathology , Animals , Axons/physiology , Cell Death/genetics , Cells, Cultured , Disease Models, Animal , Electroporation/methods , Embryo, Mammalian , Green Fluorescent Proteins/genetics , Humans , Mice , Mice, Mutant Strains , Motor Neurons/pathology , Nerve Degeneration/etiology , Nerve Tissue Proteins/genetics , Nitric Oxide/pharmacology , Proteomics/methods , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Spinal Cord/cytology , Up-Regulation/drug effects , Vesicular Acetylcholine Transport Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL