Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Cell ; 157(2): 472-485, 2014 Apr 10.
Article in English | MEDLINE | ID: mdl-24725412

ABSTRACT

Mutations in leucine-rich repeat kinase 2 (LRRK2) are a common cause of familial and sporadic Parkinson's disease (PD). Elevated LRRK2 kinase activity and neurodegeneration are linked, but the phosphosubstrate that connects LRRK2 kinase activity to neurodegeneration is not known. Here, we show that ribosomal protein s15 is a key pathogenic LRRK2 substrate in Drosophila and human neuron PD models. Phosphodeficient s15 carrying a threonine 136 to alanine substitution rescues dopamine neuron degeneration and age-related locomotor deficits in G2019S LRRK2 transgenic Drosophila and substantially reduces G2019S LRRK2-mediated neurite loss and cell death in human dopamine and cortical neurons. Remarkably, pathogenic LRRK2 stimulates both cap-dependent and cap-independent mRNA translation and induces a bulk increase in protein synthesis in Drosophila, which can be prevented by phosphodeficient T136A s15. These results reveal a novel mechanism of PD pathogenesis linked to elevated LRRK2 kinase activity and aberrant protein synthesis in vivo.


Subject(s)
Neurons/metabolism , Parkinson Disease/metabolism , Protein Serine-Threonine Kinases/metabolism , Ribosomal Proteins/metabolism , Amino Acid Sequence , Animals , Drosophila melanogaster , Humans , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Molecular Sequence Data , Neurons/pathology , Parkinson Disease/pathology , Ribosomal Proteins/chemistry
2.
Proc Natl Acad Sci U S A ; 115(7): 1635-1640, 2018 02 13.
Article in English | MEDLINE | ID: mdl-29386392

ABSTRACT

Mutations in LRRK2 are known to be the most common genetic cause of sporadic and familial Parkinson's disease (PD). Multiple lines of LRRK2 transgenic or knockin mice have been developed, yet none exhibit substantial dopamine (DA)-neuron degeneration. Here we develop human tyrosine hydroxylase (TH) promoter-controlled tetracycline-sensitive LRRK2 G2019S (GS) and LRRK2 G2019S kinase-dead (GS/DA) transgenic mice and show that LRRK2 GS expression leads to an age- and kinase-dependent cell-autonomous neurodegeneration of DA and norepinephrine (NE) neurons. Accompanying the loss of DA neurons are DA-dependent behavioral deficits and α-synuclein pathology that are also LRRK2 GS kinase-dependent. Transmission EM reveals that that there is an LRRK2 GS kinase-dependent significant reduction in synaptic vesicle number and a greater abundance of clathrin-coated vesicles in DA neurons. These transgenic mice indicate that LRRK2-induced DA and NE neurodegeneration is kinase-dependent and can occur in a cell-autonomous manner. Moreover, these mice provide a substantial advance in animal model development for LRRK2-associated PD and an important platform to investigate molecular mechanisms for how DA neurons degenerate as a result of expression of mutant LRRK2.


Subject(s)
Disease Models, Animal , Dopamine/metabolism , Dopaminergic Neurons/pathology , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/physiology , Neurodegenerative Diseases/pathology , Norepinephrine/metabolism , Age Factors , Animals , Behavior, Animal , Dopaminergic Neurons/metabolism , Humans , Male , Mice , Mice, Transgenic , Motor Activity , Mutation , Neurodegenerative Diseases/metabolism , alpha-Synuclein/metabolism
3.
EMBO Rep ; 16(9): 1114-30, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26162776

ABSTRACT

Mutations in PINK1 and PARKIN cause recessive, early-onset Parkinson's disease (PD). Together, these two proteins orchestrate a protective mitophagic response that ensures the safe disposal of damaged mitochondria. The kinase PINK1 phosphorylates ubiquitin (Ub) at the conserved residue S65, in addition to modifying the E3 ubiquitin ligase Parkin. The structural and functional consequences of Ub phosphorylation (pS65-Ub) have already been suggested from in vitro experiments, but its (patho-)physiological significance remains unknown. We have generated novel antibodies and assessed pS65-Ub signals in vitro and in cells, including primary neurons, under endogenous conditions. pS65-Ub is dependent on PINK1 kinase activity as confirmed in patient fibroblasts and postmortem brain samples harboring pathogenic mutations. We show that pS65-Ub is reversible and barely detectable under basal conditions, but rapidly induced upon mitochondrial stress in cells and amplified in the presence of functional Parkin. pS65-Ub accumulates in human brain during aging and disease in the form of cytoplasmic granules that partially overlap with mitochondrial, lysosomal, and total Ub markers. Additional studies are now warranted to further elucidate pS65-Ub functions and fully explore its potential for biomarker or therapeutic development.


Subject(s)
Protein Kinases/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitin/metabolism , Animals , Antibodies , Biomarkers , Brain/cytology , Fibroblasts , HeLa Cells , Humans , Mice , Mitochondria/physiology , Mitophagy/genetics , Mutation , Neurons/metabolism , Neurons/ultrastructure , Parkinson Disease/genetics , Parkinson Disease/physiopathology , Phosphorylation , Protein Kinases/genetics , Ubiquitin/genetics , Ubiquitin/immunology , Ubiquitination
4.
Acta Neuropathol ; 128(4): 505-24, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25173361

ABSTRACT

The occurrence of repeat-associated non-ATG (RAN) translation, an atypical form of translation of expanded repeats that results in the synthesis of homopolymeric expansion proteins, is becoming more widely appreciated among microsatellite expansion disorders. Such disorders include amyotrophic lateral sclerosis and frontotemporal dementia caused by a hexanucleotide repeat expansion in the C9ORF72 gene (c9FTD/ALS). We and others have recently shown that this bidirectionally transcribed repeat is RAN translated, and the "c9RAN proteins" thusly produced form neuronal inclusions throughout the central nervous system of c9FTD/ALS patients. Nonetheless, the potential contribution of c9RAN proteins to disease pathogenesis remains poorly understood. In the present study, we demonstrate that poly(GA) c9RAN proteins are neurotoxic and may be implicated in the neurodegenerative processes of c9FTD/ALS. Specifically, we show that expression of poly(GA) proteins in cultured cells and primary neurons leads to the formation of soluble and insoluble high molecular weight species, as well as inclusions composed of filaments similar to those observed in c9FTD/ALS brain tissues. The expression of poly(GA) proteins is accompanied by caspase-3 activation, impaired neurite outgrowth, inhibition of proteasome activity, and evidence of endoplasmic reticulum (ER) stress. Of importance, ER stress inhibitors, salubrinal and TUDCA, provide protection against poly(GA)-induced toxicity. Taken together, our data provide compelling evidence towards establishing RAN translation as a pathogenic mechanism of c9FTD/ALS, and suggest that targeting the ER using small molecules may be a promising therapeutic approach for these devastating diseases.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , DNA Repeat Expansion/genetics , Endoplasmic Reticulum Stress/physiology , Frontotemporal Dementia/metabolism , Proteins/metabolism , Adult , Aged , Aged, 80 and over , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Animals , Antibodies/pharmacology , Brain/metabolism , Brain/pathology , Brain/ultrastructure , C9orf72 Protein , Cell Nucleolus/metabolism , Cell Nucleolus/ultrastructure , Cells, Cultured , Cholagogues and Choleretics/pharmacology , DNA Repeat Expansion/immunology , Embryo, Mammalian , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/genetics , Female , Frontotemporal Dementia/genetics , Frontotemporal Dementia/pathology , Gene Expression Regulation/drug effects , Gene Expression Regulation/radiation effects , HEK293 Cells , Humans , Male , Mice , Middle Aged , Nerve Tissue Proteins/metabolism , Protein Structure, Secondary , Proteins/chemistry
5.
Mov Disord ; 28(1): 51-60, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22927178

ABSTRACT

The defining anatomical feature of Parkinson's disease (PD) is the degeneration of substantia nigra pars compacta (SNc) neurons, resulting in striatal dopamine (DA) deficiency and in the subsequent alteration of basal ganglia physiology. Treatments targeting the dopaminergic system alleviate PD symptoms but are not able to slow the neurodegenerative process that underlies PD progression. The nucleus striatum comprises a complex network of projecting neurons and interneurons that integrates different neural signals to modulate the activity of the basal ganglia circuitry. In this review we describe new potential molecular and synaptic striatal targets for the development of both symptomatic and neuroprotective strategies for PD. In particular, we focus on the interaction between adenosine A2A receptors and dopamine D2 receptors, on the role of a correct assembly of NMDA receptors, and on the sGC/cGMP/PKG pathway. Moreover, we also discuss the possibility to target the cell death program parthanatos and the kinase LRRK2 in order to develop new putative neuroprotective agents for PD acting on dopaminergic nigral neurons as well as on other basal ganglia structures.


Subject(s)
Neurons/drug effects , Neuroprotective Agents/therapeutic use , Parkinson Disease , Synapses/drug effects , Humans , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Neurons/pathology , Parkinson Disease/drug therapy , Parkinson Disease/genetics , Parkinson Disease/pathology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Receptor, Adenosine A2A/metabolism , Receptors, Dopamine/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Signal Transduction/drug effects , Synapses/pathology
6.
Biochim Biophys Acta ; 1802(11): 1095-104, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20656023

ABSTRACT

Prior exposure to sub toxic insults can induce a powerful endogenous neuroprotective program known as ischemic preconditioning. Current models typically rely on a single stress episode to induce neuroprotection whereas the clinical reality is that patients may experience multiple transient ischemic attacks (TIAs) prior to suffering a stroke. We sought to develop a neuron-enriched preconditioning model using multiple oxygen glucose deprivation (OGD) episodes to assess the endogenous protective mechanisms neurons implement at the metabolic and cellular level. We found that neurons exposed to a five minute period of glucose deprivation recovered oxygen utilization and lactate production using novel microphysiometry techniques. Using the non-toxic and energetically favorable five minute exposure, we developed a preconditioning paradigm where neurons are exposed to this brief OGD for three consecutive days. These cells experienced a 45% greater survival following an otherwise lethal event and exhibited a longer lasting window of protection in comparison to our previous in vitro preconditioning model using a single stress. As in other models, preconditioned cells exhibited mild caspase activation, an increase in oxidized proteins and a requirement for reactive oxygen species for neuroprotection. Heat shock protein 70 was upregulated during preconditioning, yet the majority of this protein was released extracellularly. We believe coupling this neuron-enriched multi-day model with microphysiometry will allow us to assess neuronal specific real-time metabolic adaptations necessary for preconditioning.


Subject(s)
Adaptation, Physiological , Glucose/metabolism , Neurons/metabolism , Oxygen/metabolism , Animals , Blotting, Western , Caspase 3/metabolism , Cell Hypoxia , Cells, Cultured , Energy Metabolism/drug effects , Enzyme Activation/drug effects , Glucose/pharmacology , HSP70 Heat-Shock Proteins/metabolism , Immunohistochemistry , Neurons/cytology , Neurons/drug effects , Oxygen/pharmacology , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Stress, Physiological , Time Factors
7.
J Neurochem ; 119(3): 604-16, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21838782

ABSTRACT

Fatty acids such as eicosapentaenoic acid (EPA) have been shown to be beneficial for neurological function and human health. It is widely thought that oxidation products of EPA are responsible for biological activity, although the specific EPA peroxidation product(s) which exert these responses have not yet been identified. In this work we provide the first evidence that the synthesized representative cyclopentenone IsoP, 15-A(3t)-IsoP, serves as a potent inhibitor of lipopolysaccharide-stimulated macrophage activation. The anti-inflammatory activities of 15-A(3t)-IsoP were observed in response not only to lipopolysaccharide, but also to tumor necrosis factor alpha and IL-1b stimulation. Subsequently, this response blocked the ability of these compounds to stimulate nuclear factor kappa b (NFκB) activation and production of proinflammatory cytokines. The bioactivity of 15-A(3t)-IsoP was shown to be dependent upon an unsaturated carbonyl residue which transiently adducts to free thiols. Site directed mutagenesis of the redox sensitive C179 site of the Ikappa kinase beta subunit, blocked the biological activity of 15-A(3t)-IsoP and NFκB activation. The vasoprotective potential of 15-A(3t)-IsoP was underscored by the ability of this compound to block oxidized lipid accumulation, a critical step in foam cell transformation and atherosclerotic plaque formation. Taken together, these are the first data identifying the biological activity of a specific product of EPA peroxidation, which is formed in abundance in vivo. The clear mechanism linking 15-A(3t)-IsoP to redox control of NFκB transcription, and the compound's ability to block foam cell transformation suggest that 15-A(3t)-IsoP provides a unique and potent tool to provide vaso- and cytoprotection under conditions of oxidative stress.


Subject(s)
Fatty Acids/metabolism , Isoprostanes/chemistry , Isoprostanes/pharmacology , Macrophage Activation/physiology , Macrophages/metabolism , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Transcription, Genetic/physiology , Animals , Cell Line , Fatty Acids/physiology , Isoprostanes/physiology , Macrophage Activation/drug effects , Macrophages/drug effects , Mice , NF-kappa B/genetics , Oxidation-Reduction , Oxidative Stress/drug effects , Oxidative Stress/physiology , Transcription, Genetic/drug effects
8.
J Neurochem ; 110(1): 378-89, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19457100

ABSTRACT

Manganese is an essential nutrient, integral to proper metabolism of amino acids, proteins and lipids. Excessive environmental exposure to manganese can produce extrapyramidal symptoms similar to those observed in Parkinson's disease (PD). We used in vivo and in vitro models to examine cellular and circuitry alterations induced by manganese exposure. Primary mesencephalic cultures were treated with 10-800 microM manganese chloride which resulted in dramatic changes in the neuronal cytoskeleton even at subtoxic concentrations. Using cultures from mice with red fluorescent protein driven by the tyrosine hydroxylase (TH) promoter, we found that dopaminergic neurons were more susceptible to manganese toxicity. To understand the vulnerability of dopaminergic cells to chronic manganese exposure, mice were given i.p. injections of MnCl(2) for 30 days. We observed a 20% reduction in TH-positive neurons in the substantia nigra pars compacta (SNpc) following manganese treatment. Quantification of Nissl bodies revealed a widespread reduction in SNpc cell numbers. Other areas of the basal ganglia were also altered by manganese as evidenced by the loss of glutamic acid decarboxylase 67 in the striatum. These studies suggest that acute manganese exposure induces cytoskeletal dysfunction prior to degeneration and that chronic manganese exposure results in neurochemical dysfunction with overlapping features to PD.


Subject(s)
Dopamine/metabolism , Manganese Poisoning/metabolism , Manganese/toxicity , Neurons/metabolism , Substantia Nigra/metabolism , gamma-Aminobutyric Acid/metabolism , Animals , Basal Ganglia/drug effects , Basal Ganglia/metabolism , Basal Ganglia/physiopathology , Cytoskeleton/drug effects , Cytoskeleton/metabolism , Cytoskeleton/pathology , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/pathology , Glutamate Decarboxylase/drug effects , Glutamate Decarboxylase/metabolism , Magnesium Chloride/toxicity , Manganese Poisoning/physiopathology , Mice , Neurons/drug effects , Neurotoxins/toxicity , Rats , Substantia Nigra/drug effects , Substantia Nigra/physiopathology , Synaptic Transmission/drug effects , Synaptic Transmission/physiology , Tyrosine 3-Monooxygenase/genetics , Tyrosine 3-Monooxygenase/metabolism
9.
Mol Neurodegener ; 14(1): 9, 2019 02 15.
Article in English | MEDLINE | ID: mdl-30767771

ABSTRACT

BACKGROUND: A G4C2 hexanucleotide repeat expansion in the noncoding region of C9orf72 is the major genetic cause of frontotemporal dementia and amyotrophic lateral sclerosis (c9FTD/ALS). Putative disease mechanisms underlying c9FTD/ALS include toxicity from sense G4C2 and antisense G2C4 repeat-containing RNA, and from dipeptide repeat (DPR) proteins unconventionally translated from these RNA products. METHODS: Intracerebroventricular injections with adeno-associated virus (AAV) encoding 2 or 149 G4C2 repeats were performed on postnatal day 0, followed by assessment of behavioral and neuropathological phenotypes. RESULTS: Relative to control mice, gliosis and neurodegeneration accompanied by cognitive and motor deficits were observed in (G4C2)149 mice by 6 months of age. Recapitulating key pathological hallmarks, we also demonstrate that sense and antisense RNA foci, inclusions of poly(GA), poly(GP), poly(GR), poly(PR), and poly(PA) DPR proteins, and inclusions of endogenous phosphorylated TDP-43 (pTDP-43) developed in (G4C2)149 mice but not control (G4C2)2 mice. Notably, proteins that play a role in the regulation of stress granules - RNA-protein assemblies that form in response to translational inhibition and that have been implicated in c9FTD/ALS pathogenesis - were mislocalized in (G4C2)149 mice as early as 3 months of age. Specifically, we observed the abnormal deposition of stress granule components within inclusions immunopositive for poly(GR) and pTDP-43, as well as evidence of nucleocytoplasmic transport defects. CONCLUSIONS: Our in vivo model of c9FTD/ALS is the first to robustly recapitulate hallmark features derived from both sense and antisense C9orf72 repeat-associated transcripts complete with neurodegeneration and behavioral impairments. More importantly, the early appearance of persistent pathological stress granules prior to significant pTDP-43 deposition implicates an aberrant stress granule response as a key disease mechanism driving TDP-43 proteinopathy in c9FTD/ALS.


Subject(s)
C9orf72 Protein/genetics , Heat-Shock Proteins/metabolism , Nerve Degeneration/pathology , TDP-43 Proteinopathies/pathology , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Animals , Frontotemporal Dementia/genetics , Frontotemporal Dementia/metabolism , Frontotemporal Dementia/pathology , Mice , Nerve Degeneration/genetics , Nerve Degeneration/metabolism , TDP-43 Proteinopathies/genetics , TDP-43 Proteinopathies/metabolism , Trinucleotide Repeat Expansion
10.
Mol Neurodegener ; 12(1): 55, 2017 07 25.
Article in English | MEDLINE | ID: mdl-28743268

ABSTRACT

Haploinsufficiency of GRN, the gene encoding progranulin (PGRN), causes frontotemporal lobar degeneration (FTLD), the second most common cause of early-onset dementia. Receptor-mediated lysosomal targeting has been shown to regulate brain PGRN levels, and complete deficiency of PGRN is a direct cause of neuronal ceroid lipofuscinosis (NCL), a lysosomal storage disease. Here we show that the lysosomal cysteine protease cathepsin L (Cat L) can mediate the proteolytic cleavage of intracellular PGRN into poly-granulin and granulin fragments. Further, PGRN and Cat L co-localize in lysosomes of HEK293 cells, iPSC-derived neurons and human cortical neurons from human postmortem tissue. These data identify Cat L as a key intracellular lysosomal PGRN protease, and provides an intriguing new link between lysosomal dysfunction and FTLD.


Subject(s)
Cathepsin L/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Lysosomes/metabolism , Proteins/metabolism , Cells, Cultured , Frontotemporal Lobar Degeneration/metabolism , Humans , Neurons/metabolism , Progranulins
11.
eNeuro ; 4(2)2017.
Article in English | MEDLINE | ID: mdl-28321439

ABSTRACT

Mutations in the leucine-rich repeat kinase 2 (LRRK2) gene have been identified as an unambiguous cause of late-onset, autosomal-dominant familial Parkinson's disease (PD) and LRRK2 mutations are the strongest genetic risk factor for sporadic PD known to date. A number of transgenic mice expressing wild-type or mutant LRRK2 have been described with varying degrees of LRRK2-related abnormalities and modest pathologies. None of these studies directly addressed the role of the kinase domain in the changes observed and none of the mice present with robust features of the human disease. In an attempt to address these issues, we created a conditional LRRK2 G2019S (LRRK2 GS) mutant and a functionally negative control, LRRK2 G2019S/D1994A (LRRK2 GS/DA). Expression of LRRK2 GS or LRRK2 GS/DA was conditionally controlled using the tet-off system in which the presence of tetracycline-transactivator protein (tTA) with a CAMKIIα promoter (CAMKIIα-tTA) induced expression of TetP-LRRK2 GS or TetP-LRRK2 GS/DA in the mouse forebrain. Overexpression of LRRK2 GS in mouse forebrain induced behavioral deficits and α-synuclein pathology in a kinase-dependent manner. Similar to other genetically engineered LRRK2 GS mice, there was no significant loss of dopaminergic neurons. These mice provide an important new tool to study neurobiological changes associated with the increased kinase activity from the LRRK2 G2019S mutation, which may ultimately lead to a better understanding of not only the physiologic actions of LRRK2, but also potential pathologic actions that underlie LRRK2 GS-associated PD.


Subject(s)
Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/metabolism , Parkinsonian Disorders/metabolism , Parkinsonian Disorders/pathology , Prosencephalon/metabolism , Prosencephalon/pathology , alpha-Synuclein/metabolism , Amphetamine/pharmacology , Animals , Central Nervous System Stimulants/pharmacology , Exploratory Behavior/drug effects , Female , Humans , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Male , Mice, Transgenic , Motor Activity/drug effects , Mutation , Parkinsonian Disorders/psychology , Prosencephalon/drug effects , Protein Aggregation, Pathological/metabolism , Protein Aggregation, Pathological/pathology , Random Allocation , Tyrosine 3-Monooxygenase/metabolism
12.
Sci Transl Med ; 9(383)2017 03 29.
Article in English | MEDLINE | ID: mdl-28356511

ABSTRACT

There is no effective treatment for amyotrophic lateral sclerosis (ALS), a devastating motor neuron disease. However, discovery of a G4C2 repeat expansion in the C9ORF72 gene as the most common genetic cause of ALS has opened up new avenues for therapeutic intervention for this form of ALS. G4C2 repeat expansion RNAs and proteins of repeating dipeptides synthesized from these transcripts are believed to play a key role in C9ORF72-associated ALS (c9ALS). Therapeutics that target G4C2 RNA, such as antisense oligonucleotides (ASOs) and small molecules, are thus being actively investigated. A limitation in moving such treatments from bench to bedside is a lack of pharmacodynamic markers for use in clinical trials. We explored whether poly(GP) proteins translated from G4C2 RNA could serve such a purpose. Poly(GP) proteins were detected in cerebrospinal fluid (CSF) and in peripheral blood mononuclear cells from c9ALS patients and, notably, from asymptomatic C9ORF72 mutation carriers. Moreover, CSF poly(GP) proteins remained relatively constant over time, boding well for their use in gauging biochemical responses to potential treatments. Treating c9ALS patient cells or a mouse model of c9ALS with ASOs that target G4C2 RNA resulted in decreased intracellular and extracellular poly(GP) proteins. This decrease paralleled reductions in G4C2 RNA and downstream G4C2 RNA-mediated events. These findings indicate that tracking poly(GP) proteins in CSF could provide a means to assess target engagement of G4C2 RNA-based therapies in symptomatic C9ORF72 repeat expansion carriers and presymptomatic individuals who are expected to benefit from early therapeutic intervention.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Biomarkers/metabolism , C9orf72 Protein/genetics , Dinucleotide Repeats/genetics , Adult , Aged , Amyotrophic Lateral Sclerosis/cerebrospinal fluid , Amyotrophic Lateral Sclerosis/pathology , Animals , Brain/metabolism , Brain/pathology , Cell Line , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Longitudinal Studies , Mice , Middle Aged , Neurons/metabolism , Oligonucleotides, Antisense/pharmacology , Prognosis , RNA/genetics
13.
Nat Neurosci ; 19(5): 668-677, 2016 05.
Article in English | MEDLINE | ID: mdl-26998601

ABSTRACT

Neuronal inclusions of poly(GA), a protein unconventionally translated from G4C2 repeat expansions in C9ORF72, are abundant in patients with frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS) caused by this mutation. To investigate poly(GA) toxicity, we generated mice that exhibit poly(GA) pathology, neurodegeneration and behavioral abnormalities reminiscent of FTD and ALS. These phenotypes occurred in the absence of TDP-43 pathology and required poly(GA) aggregation. HR23 proteins involved in proteasomal degradation and proteins involved in nucleocytoplasmic transport were sequestered by poly(GA) in these mice. HR23A and HR23B similarly colocalized to poly(GA) inclusions in C9ORF72 expansion carriers. Sequestration was accompanied by an accumulation of ubiquitinated proteins and decreased xeroderma pigmentosum C (XPC) levels in mice, indicative of HR23A and HR23B dysfunction. Restoring HR23B levels attenuated poly(GA) aggregation and rescued poly(GA)-induced toxicity in neuronal cultures. These data demonstrate that sequestration and impairment of nuclear HR23 and nucleocytoplasmic transport proteins is an outcome of, and a contributor to, poly(GA) pathology.


Subject(s)
Carrier Proteins/metabolism , DNA-Binding Proteins/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Neurons/pathology , Proteins/toxicity , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Animals , Atrophy/pathology , Behavior, Animal , Brain/metabolism , Brain/pathology , Brain/ultrastructure , C9orf72 Protein , Frontotemporal Dementia/metabolism , Frontotemporal Dementia/pathology , Gene Expression/genetics , Humans , Inclusion Bodies/metabolism , Inclusion Bodies/ultrastructure , Mice , Mutation , Nerve Degeneration/pathology , Neurons/metabolism , Primary Cell Culture , Proteins/genetics , Proteins/metabolism , Ubiquitinated Proteins/metabolism
14.
Antioxid Redox Signal ; 23(6): 535-49, 2015 Aug 20.
Article in English | MEDLINE | ID: mdl-25602369

ABSTRACT

AIMS: Determine the mechanism by which C-terminus of HSC70-interacting protein (CHIP) induction alters neuronal survival under conditions of mitochondrial stress induced by oxygen glucose deprivation. RESULTS: We report that animals deficient in the E3 ubiquitin ligase, CHIP, have high baseline levels of central nervous system protein oxidation and lipid peroxidation, reduced antioxidant defenses, and decreased energetic status. Stress-associated molecules typically linked to Parkinson's disease such as the mitochondrial kinase, PTEN-inducible putative kinase 1 (PINK1), and another E3 ligase, Parkin, are upregulated in brains from CHIP knockout (KO) animals. Utilizing a novel biotin-avidin capture technique, we found that the oxidation status of Parkin and the mitochondrial fission protein, dynamin-related protein 1 (Drp1), are altered in a CHIP-dependent manner. We also found that following oxygen-glucose deprivation (OGD), the expression of CHIP, PINK1, and the autophagic marker, LC3, increase and there is activation of the redox-sensitive kinase p66(shc). Under conditions of OGD, CHIP relocalizes from the cytosol to mitochondria. Mitochondria from CHIP KO mice have profound impairments in stress response induced by calcium overload, resulting in accelerated permeability transition activity. While CHIP-deficient neurons are morphologically intact, they are more susceptible to OGD consistent with a previously unknown neuroprotective role for CHIP in maintaining mitochondrial homeostasis. INNOVATION: CHIP relocalization to the mitochondria is essential for the regulation of mitochondrial integrity and neuronal survival following OGD. CONCLUSIONS: CHIP is an essential regulator of neuronal bioenergetics and redox tone. Altering the expression of this protein has profound effects on neuronal survival when cells are exposed to OGD.


Subject(s)
Mitochondria/metabolism , Neurons/metabolism , Ubiquitin-Protein Ligases/physiology , Animals , Autophagy , Cell Hypoxia , Cells, Cultured , Glucose/metabolism , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mitochondrial Dynamics , Oxidation-Reduction , Protein Biosynthesis , Rats, Sprague-Dawley , Signal Transduction , Stress, Physiological
15.
Science ; 348(6239): 1151-4, 2015 Jun 05.
Article in English | MEDLINE | ID: mdl-25977373

ABSTRACT

The major genetic cause of frontotemporal dementia and amyotrophic lateral sclerosis is a G4C2 repeat expansion in C9ORF72. Efforts to combat neurodegeneration associated with "c9FTD/ALS" are hindered by a lack of animal models recapitulating disease features. We developed a mouse model to mimic both neuropathological and clinical c9FTD/ALS phenotypes. We expressed (G4C2)66 throughout the murine central nervous system by means of somatic brain transgenesis mediated by adeno-associated virus. Brains of 6-month-old mice contained nuclear RNA foci, inclusions of poly(Gly-Pro), poly(Gly-Ala), and poly(Gly-Arg) dipeptide repeat proteins, as well as TDP-43 pathology. These mouse brains also exhibited cortical neuron and cerebellar Purkinje cell loss, astrogliosis, and decreased weight. (G4C2)66 mice also developed behavioral abnormalities similar to clinical symptoms of c9FTD/ALS patients, including hyperactivity, anxiety, antisocial behavior, and motor deficits.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , DNA-Binding Proteins/genetics , Disease Models, Animal , Frontotemporal Dementia/genetics , Mice , Proteins/genetics , Amyotrophic Lateral Sclerosis/pathology , Animals , Antisocial Personality Disorder/genetics , Antisocial Personality Disorder/pathology , C9orf72 Protein , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Dependovirus , Dipeptides/metabolism , Frontotemporal Dementia/pathology , Gene Transfer Techniques , HEK293 Cells , Humans , Purkinje Cells/metabolism , Purkinje Cells/pathology , RNA, Nuclear/metabolism
16.
Alzheimers Res Ther ; 6(3): 29, 2014.
Article in English | MEDLINE | ID: mdl-25031639

ABSTRACT

The identification of tau protein as a major constituent of neurofibrillary tangles spurred considerable effort devoted to identifying and validating pathways through which therapeutics may alleviate tau burden in Alzheimer's disease and related tauopathies, including chronic traumatic encephalopathy associated with sport- and military-related injuries. Most tau-based therapeutic strategies have previously focused on modulating tau phosphorylation, given that tau species present within neurofibrillary tangles are hyperphosphorylated on a number of different residues. However, the recent discovery that tau is modified by acetylation necessitates additional research to provide greater mechanistic insight into the spectrum of physiological consequences of tau acetylation, which may hold promise as a novel therapeutic target. In this review, we discuss recent findings evaluating tau acetylation in the context of previously accepted notions regarding tau biology and pathophysiology. We also examine the evidence demonstrating the neuroprotective and beneficial consequences of inhibiting histone deacetylase (HDAC)6, a tau deacetylase, including its effect on microtubule stabilization. We also discuss the rationale for pharmacologically modulating HDAC6 in tau-based pathologies as a novel therapeutic strategy.

17.
CNS Neurol Disord Drug Targets ; 12(6): 849-60, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23469839

ABSTRACT

The isocitrate dehydrogenase (IDH) enzymes were initially identified as essential components of the Krebs cycle. IDH mutations were thought to be incompatible with cell survival. However, 90% of glioblastomas were recently shown to be associated with somatic mutations in these enzymes, indicating a possible role for IDH in promoting cellular survival in hypoxic environments. Our proteomic analysis of rats given 10 minutes of middle cerebral artery occlusion to induce transient ischemia demonstrates a significant decrease in IDH expression. We have recapitulated this decrease in an in vitro model using primary cortical neurons exposed to acute oxygen and glucose deprivation. Given the role of IDHs in energy metabolism and antioxidant production, we hypothesize that the IDHs may serve as first-line, rapid-response enzymes that regulate survival in environments of energetic or oxidative stress. In order to identify the specific events that regulate IDH enzymes, HT-22 neural cells were subjected to either a selective energetic challenge or a pure oxidative stress. In response to the non-lethal energetic challenge induced by substituting galactose for glucose, we observed increased IDH1, 2, and 3 expression and cessation of cellular proliferation. No change in expression of any IDH isoform was observed when neural cells were subjected to subtoxic oxidative stress via glutathione depletion. Taken together, these data imply that IDH expression rapidly responds to changes in energetic status, but not to oxidative stress. These data also suggest that IDH enzymes respond not only to allosteric modulation, but can also change patterns of expression in response to moderate stress in an effort to maximize ATP production and survival.


Subject(s)
Adaptation, Physiological/physiology , Brain Ischemia/enzymology , Cerebral Cortex/enzymology , Energy Metabolism/physiology , Isocitrate Dehydrogenase/metabolism , Neurons/enzymology , Acute Disease , Animals , Brain Ischemia/pathology , Cell Line, Transformed , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/pathology , Mice , Neurons/pathology , Neurons/physiology , Rats , Rats, Sprague-Dawley
18.
PLoS One ; 7(5): e36340, 2012.
Article in English | MEDLINE | ID: mdl-22606257

ABSTRACT

The multifunctional E3 ubiquitin ligase CHIP is an essential interacting partner of HSP70, which together promote the proteasomal degradation of client proteins. Acute CHIP overexpression provides neuroprotection against neurotoxic mitochondrial stress, glucocorticoids, and accumulation of toxic amyloid fragments, as well as genetic mutations in other E3 ligases, which have been shown to result in familial Parkinson's disease. These studies have created a great deal of interest in understanding CHIP activity, expression and modulation. While CHIP knockout mice have the potential to provide essential insights into the molecular control of cell fate and survival, the animals have been difficult to characterize in vivo due to severe phenotypic and behavioral dysfunction, which have thus far been poorly characterized. Therefore, in the present study we conducted a battery of neurobehavioral and physiological assays of adult CHIP heterozygotic (HET) mutant mice to provide a better understanding of the functional consequence of CHIP deficiency. We found that CHIP HET mice had normal body and brain weight, body temperature, muscle tone and breathing patterns, but do have a significant elevation in baseline heart rate. Meanwhile basic behavioral screens of sensory, motor, emotional and cognitive functions were normative. We observed no alterations in performance in the elevated plus maze, light-dark preference and tail suspension assays, or two simple cognitive tasks: novel object recognition and spontaneous alternation in a Y maze. Significant deficits were found, however, when CHIP HET mice performed wire hang, inverted screen, wire maneuver, and open field tasks. Taken together, our data indicate a clear subset of behaviors that are altered at baseline in CHIP deficient animals, which will further guide whole animal studies of the effects of CHIP dysregulation on cardiac function, brain circuitry and function, and responsiveness to environmental and cellular stress.


Subject(s)
Behavior, Animal/physiology , Ubiquitin-Protein Ligases/deficiency , Animals , Anxiety/genetics , Anxiety/physiopathology , Emotions/physiology , Haploinsufficiency , Male , Maze Learning/physiology , Mice , Mice, Mutant Strains , Motivation/genetics , Motivation/physiology , Motor Activity/physiology , Motor Skills/physiology , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/physiology
19.
Antioxid Redox Signal ; 14(10): 1841-51, 2011 May 15.
Article in English | MEDLINE | ID: mdl-20626319

ABSTRACT

The development of a suitable neuroprotective agent to treat ischemic stroke has failed when transitioned to the clinical setting. An understanding of the molecular mechanisms involved in neuronal injury during ischemic stroke is important, but must be placed in the clinical context. Current therapeutic targets have focused on the preservation of the ischemic penumbra in the hope of improving clinical outcomes. Unfortunately, most patients in the ultra-early time windows harbor penumbra but have tremendous variability in the size of the core infarct, the ultimate predictor of prognosis. Understanding this variability may allow for proper patient selection that may better correlate to bench models. Reperfusion therapies are rapidly evolving and have been shown to improve clinical outcomes. The use of neuroprotective agents to prolong time windows prior to reperfusion or to prevent reperfusion injury may present future therapeutic targets for the treatment of ischemic stroke. We review the molecular pathways and the clinical context from which future targets may be identified.


Subject(s)
Neuroprotective Agents/therapeutic use , Stroke/drug therapy , Humans , Oxidative Stress/physiology , Reactive Oxygen Species/metabolism , Stroke/metabolism , Stroke/pathology , Stroke/physiopathology
20.
Methods Mol Biol ; 758: 215-35, 2011.
Article in English | MEDLINE | ID: mdl-21815069

ABSTRACT

Drug discovery and therapeutic development for disorders of the central nervous system (CNS) represents one of the largest unmet markets in modern medicine. We have increasingly recognized that the lack of stringent assessment of mitochondrial function during the discovery process has resulted in drug recalls, black box warnings, and an urgent need to understand the metabolic liability of small molecules in neural systems. Given that the brain is the most energetically demanding organ, even modest perturbations in neuronal energetic pathways have been shown to impact growth, signaling, connectivity, and the restorative capacity of the CNS. In this work, we describe several tools to assess metabolic activity of primary neuronal cultures and neural cell lines using an acute model of injury induced by oxygen glucose deprivation. Methods include the measurement of total ATP and NADH, enzymatic assessment of lactate production by anaerobic respiration, as well as viability assays. We also present a modified screening method for assessing aerobic respiration of immortalized cell lines using galactose challenge.


Subject(s)
Energy Metabolism , Neurons/metabolism , Adenosine Triphosphate/chemistry , Adenosine Triphosphate/metabolism , Animals , Brain/cytology , Brain/metabolism , Cell Extracts/chemistry , Cell Hypoxia , Cell Line, Tumor , Cell Survival , Embryo, Mammalian/cytology , Female , Galactose/chemistry , Galactose/metabolism , Glucose/chemistry , Glucose/metabolism , Humans , L-Lactate Dehydrogenase/chemistry , L-Lactate Dehydrogenase/metabolism , Lactic Acid/chemistry , Lactic Acid/metabolism , Pregnancy , Pyruvic Acid/chemistry , Pyruvic Acid/metabolism , Rats
SELECTION OF CITATIONS
SEARCH DETAIL