Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
J Neuroinflammation ; 14(1): 128, 2017 06 26.
Article in English | MEDLINE | ID: mdl-28651603

ABSTRACT

BACKGROUND: Kv1.3 potassium channels regulate microglial functions and are overexpressed in neuroinflammatory diseases. Kv1.3 blockade may selectively inhibit pro-inflammatory microglia in neurological diseases but the molecular and cellular mechanisms regulated by Kv1.3 channels are poorly defined. METHODS: We performed immunoblotting and flow cytometry to confirm Kv1.3 channel upregulation in lipopolysaccharide (LPS)-activated BV2 microglia and in brain mononuclear phagocytes freshly isolated from LPS-treated mice. Quantitative proteomics was performed on BV2 microglia treated with control, LPS, ShK-223 (highly selective Kv1.3 blocker), and LPS+ShK-223. Gene ontology (GO) analyses of Kv1.3-dependent LPS-regulated proteins were performed, and the most representative proteins and GO terms were validated. Effects of Kv1.3-blockade on LPS-activated BV2 microglia were studied in migration, focal adhesion formation, reactive oxygen species production, and phagocytosis assays. In vivo validation of protein changes and predicted molecular pathways were performed in a model of systemic LPS-induced neuroinflammation, employing antigen presentation and T cell proliferation assays. Informed by pathway analyses of proteomic data, additional mechanistic experiments were performed to identify early Kv1.3-dependent signaling and transcriptional events. RESULTS: LPS-upregulated cell surface Kv1.3 channels in BV2 microglia and in microglia and CNS-infiltrating macrophages isolated from LPS-treated mice. Of 144 proteins differentially regulated by LPS (of 3141 proteins), 21 proteins showed rectification by ShK-223. Enriched cellular processes included MHCI-mediated antigen presentation (TAP1, EHD1), cell motility, and focal adhesion formation. In vitro, ShK-223 decreased LPS-induced focal adhesion formation, reversed LPS-induced inhibition of migration, and inhibited LPS-induced upregulation of EHD1, a protein involved in MHCI trafficking. In vivo, intra-peritoneal ShK-223 inhibited LPS-induced MHCI expression by CD11b+CD45low microglia without affecting MHCI expression or trafficking of CD11b+CD45high macrophages. ShK-223 inhibited LPS-induced MHCI-restricted antigen presentation to ovalbumin-specific CD8+ T cells both in vitro and in vivo. Kv1.3 co-localized with the LPS receptor complex and regulated LPS-induced early serine (S727) STAT1 phosphorylation. CONCLUSIONS: We have unraveled novel molecular and functional roles for Kv1.3 channels in pro-inflammatory microglial activation, including a Kv1.3 channel-regulated pathway that facilitates MHCI expression and MHCI-dependent antigen presentation by microglia to CD8+ T cells. We also provide evidence for neuro-immunomodulation by systemically administered ShK peptides. Our results further strengthen the therapeutic candidacy of microglial Kv1.3 channels in neurologic diseases.


Subject(s)
Kv1.3 Potassium Channel/biosynthesis , Lipopolysaccharides/pharmacology , Microglia/drug effects , Microglia/metabolism , Proteomics/methods , Animals , Cell Line , Cells, Cultured , Dose-Response Relationship, Drug , Female , Kv1.3 Potassium Channel/immunology , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Microglia/immunology
2.
Neurobiol Dis ; 54: 264-79, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23318928

ABSTRACT

Chronic activation of the NF-κB pathway is associated with progressive neurodegeneration in Parkinson's disease (PD). Given the role of neuronal RING finger protein 11 (RNF11) as a negative regulator of the NF-κB pathway, in this report we investigated the function of RNF11 in dopaminergic cells in PD-associated neurodegeneration. We found that RNF11 knockdown in an in vitro model of PD mediated protection against 6-OHDA-induced toxicity. In converse, over-expression of RNF11 enhanced 6-OHDA-induced dopaminergic cell death. Furthermore, by directly manipulating NF-κB signaling, we showed that the observed RNF11-enhanced 6-OHDA toxicity is mediated through inhibition of NF-κB-dependent transcription of TNF-α, antioxidants GSS and SOD1, and anti-apoptotic factor BCL2. Experiments in an in vivo 6-OHDA rat model of PD recapitulated the in vitro results. In vivo targeted RNF11 over-expression in nigral neurons enhanced 6-OHDA toxicity, as evident by increased amphetamine-induced rotations and loss of nigral dopaminergic neurons as compared to controls. This enhanced toxicity was coupled with the downregulation of NF-κB transcribed GSS, SOD1, BCL2, and neurotrophic factor BDNF mRNA levels, in addition to decreased TNF-α mRNA levels in ventral mesenchephalon samples. In converse, knockdown of RNF11 was associated with protective phenotypes and increased expression of above-mentioned NF-κB transcribed genes. Collectively, our in vitro and in vivo data suggest that RNF11-mediated inhibition of NF-κB in dopaminergic cells exaggerates 6-OHDA toxicity by inhibiting neuroprotective responses while loss of RNF11 inhibition on NF-κB activity promotes neuronal survival. The decreased expression of RNF11 in surviving cortical and nigral tissue detected in PD patients, thus implies a compensatory response in the diseased brain to PD-associated insults. In summary, our findings demonstrate that RNF11 in neurons can modulate susceptibility to 6-OHDA toxicity through NF-κB mediated responses. This neuron-specific role of RNF11 in the brain has important implications for targeted therapeutics aimed at preventing neurodegeneration.


Subject(s)
Carrier Proteins/metabolism , Dopaminergic Neurons/metabolism , NF-kappa B/metabolism , Parkinson Disease/metabolism , Substantia Nigra/metabolism , Adrenergic Agents/toxicity , Aged , Aged, 80 and over , Animals , DNA-Binding Proteins , Female , Humans , Immunohistochemistry , Male , Middle Aged , Nerve Degeneration/metabolism , Oxidopamine/toxicity , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Signal Transduction/physiology , Substantia Nigra/pathology , Transfection
3.
Alzheimers Dement (N Y) ; 9(2): e12394, 2023.
Article in English | MEDLINE | ID: mdl-37215505

ABSTRACT

Alzheimer's disease (AD) drug discovery has focused on a set of highly studied therapeutic hypotheses, with limited success. The heterogeneous nature of AD processes suggests that a more diverse, systems-integrated strategy may identify new therapeutic hypotheses. Although many target hypotheses have arisen from systems-level modeling of human disease, in practice and for many reasons, it has proven challenging to translate them into drug discovery pipelines. First, many hypotheses implicate protein targets and/or biological mechanisms that are under-studied, meaning there is a paucity of evidence to inform experimental strategies as well as high-quality reagents to perform them. Second, systems-level targets are predicted to act in concert, requiring adaptations in how we characterize new drug targets. Here we posit that the development and open distribution of high-quality experimental reagents and informatic outputs-termed target enabling packages (TEPs)-will catalyze rapid evaluation of emerging systems-integrated targets in AD by enabling parallel, independent, and unencumbered research.

4.
J Neuroinflammation ; 9: 67, 2012 Apr 16.
Article in English | MEDLINE | ID: mdl-22507528

ABSTRACT

BACKGROUND: The RING domain-containing protein RING finger protein 11 (RNF11) is a member of the A20 ubiquitin-editing protein complex and modulates peripheral NF-κB signaling. RNF11 is robustly expressed in neurons and colocalizes with a population of α-synuclein-positive Lewy bodies and neurites in Parkinson disease patients. The NF-κB pathway has an important role in the vertebrate nervous system, where the absence of NF-κB activity during development can result in learning and memory deficits, whereas chronic NF-κB activation is associated with persistent neuroinflammation. We examined the functional role of RNF11 with respect to canonical NF-κB signaling in neurons to gain understanding of the tight association of inflammatory pathways, including NF-κB, with the pathogenesis of neurodegenerative diseases. METHODS AND RESULTS: Luciferase assays were employed to assess NF-κB activity under targeted short hairpin RNA (shRNA) knockdown of RNF11 in human neuroblastoma cells and murine primary neurons, which suggested that RNF11 acts as a negative regulator of canonical neuronal NF-κB signaling. These results were further supported by analyses of p65 translocation to the nucleus following depletion of RNF11. Coimmunoprecipitation experiments indicated that RNF11 associates with members of the A20 ubiquitin-editing protein complex in neurons. Site-directed mutagenesis of the myristoylation domain, which is necessary for endosomal targeting of RNF11, altered the impact of RNF11 on NF-κB signaling and abrogated RNF11's association with the A20 ubiquitin-editing protein complex. A partial effect on canonical NF-κB signaling and an association with the A20 ubiquitin-editing protein complex was observed with mutagenesis of the PPxY motif, a proline-rich region involved in Nedd4-like protein interactions. Last, shRNA-mediated reduction of RNF11 in neurons and neuronal cell lines elevated levels of monocyte chemoattractant protein 1 and TNF-α mRNA and proteins, suggesting that NF-κB signaling and associated inflammatory responses are aberrantly regulated in the absence of RNF11. CONCLUSIONS: Our findings support the hypothesis that, in the nervous system, RNF11 negatively regulates canonical NF-κB signaling. Reduced or functionally compromised RNF11 could influence NF-κB-associated neuronal functions, including exaggerated inflammatory responses that may have implications for neurodegenerative disease pathogenesis and progression.


Subject(s)
Carrier Proteins/physiology , NF-kappa B/metabolism , Neurons/metabolism , Signal Transduction/physiology , Animals , Cell Line, Tumor , Cells, Cultured , DNA-Binding Proteins , Gene Knockdown Techniques , Humans , Mice , Mice, Inbred C57BL , NF-kappa B/physiology
5.
Nat Commun ; 13(1): 2927, 2022 05 25.
Article in English | MEDLINE | ID: mdl-35614064

ABSTRACT

Proteomic profiling of brain cell types using isolation-based strategies pose limitations in resolving cellular phenotypes representative of their native state. We describe a mouse line for cell type-specific expression of biotin ligase TurboID, for in vivo biotinylation of proteins. Using adenoviral and transgenic approaches to label neurons, we show robust protein biotinylation in neuronal soma and axons throughout the brain, allowing quantitation of over 2000 neuron-derived proteins spanning synaptic proteins, transporters, ion channels and disease-relevant druggable targets. Next, we contrast Camk2a-neuron and Aldh1l1-astrocyte proteomes and identify brain region-specific proteomic differences within both cell types, some of which might potentially underlie the selective vulnerability to neurological diseases. Leveraging the cellular specificity of proteomic labeling, we apply an antibody-based approach to uncover differences in neuron and astrocyte-derived signaling phospho-proteins and cytokines. This approach will facilitate the characterization of cell-type specific proteomes in a diverse number of tissues under both physiological and pathological states.


Subject(s)
Biotin , Proteomics , Animals , Astrocytes/metabolism , Biotin/metabolism , Biotinylation , Brain/metabolism , Mice , Neurons/metabolism , Proteome/metabolism
6.
Nat Neurosci ; 25(2): 213-225, 2022 02.
Article in English | MEDLINE | ID: mdl-35115731

ABSTRACT

The biological processes that are disrupted in the Alzheimer's disease (AD) brain remain incompletely understood. In this study, we analyzed the proteomes of more than 1,000 brain tissues to reveal new AD-related protein co-expression modules that were highly preserved across cohorts and brain regions. Nearly half of the protein co-expression modules, including modules significantly altered in AD, were not observed in RNA networks from the same cohorts and brain regions, highlighting the proteopathic nature of AD. Two such AD-associated modules unique to the proteomic network included a module related to MAPK signaling and metabolism and a module related to the matrisome. The matrisome module was influenced by the APOE ε4 allele but was not related to the rate of cognitive decline after adjustment for neuropathology. By contrast, the MAPK/metabolism module was strongly associated with the rate of cognitive decline. Disease-associated modules unique to the proteome are sources of promising therapeutic targets and biomarkers for AD.


Subject(s)
Alzheimer Disease , Cognitive Dysfunction , Alzheimer Disease/metabolism , Brain/metabolism , Cognitive Dysfunction/pathology , Humans , Proteome , Proteomics , RNA/metabolism
7.
Mol Neurodegener ; 15(1): 28, 2020 05 07.
Article in English | MEDLINE | ID: mdl-32381088

ABSTRACT

BACKGROUND: Proteomic characterization of microglia provides the most proximate assessment of functionally relevant molecular mechanisms of neuroinflammation. However, microglial proteomics studies have been limited by low cellular yield and contamination by non-microglial proteins using existing enrichment strategies. METHODS: We coupled magnetic-activated cell sorting (MACS) and fluorescence activated cell sorting (FACS) of microglia with tandem mass tag-mass spectrometry (TMT-MS) to obtain a highly-pure microglial proteome and identified a core set of highly-abundant microglial proteins in adult mouse brain. We interrogated existing human proteomic data for Alzheimer's disease (AD) relevance of highly-abundant microglial proteins and performed immuno-histochemical and in-vitro validation studies. RESULTS: Quantitative multiplexed proteomics by TMT-MS of CD11b + MACS-enriched (N = 5 mice) and FACS-isolated (N = 5 mice), from adult wild-type mice, identified 1791 proteins. A total of 203 proteins were highly abundant in both datasets, representing a core-set of highly abundant microglial proteins. In addition, we found 953 differentially enriched proteins comparing MACS and FACS-based approaches, indicating significant differences between both strategies. The FACS-isolated microglia proteome was enriched with cytosolic, endoplasmic reticulum, and ribosomal proteins involved in protein metabolism and immune system functions, as well as an abundance of canonical microglial proteins. Conversely, the MACS-enriched microglia proteome was enriched with mitochondrial and synaptic proteins and higher abundance of neuronal, oligodendrocytic and astrocytic proteins. From the 203 consensus microglial proteins with high abundance in both datasets, we confirmed microglial expression of moesin (Msn) in wild-type and 5xFAD mouse brains as well as in human AD brains. Msn expression is nearly exclusively found in microglia that surround Aß plaques in 5xFAD brains. In in-vitro primary microglial studies, Msn silencing by siRNA decreased Aß phagocytosis and increased lipopolysaccharide-induced production of the pro-inflammatory cytokine, tumor necrosis factor (TNF). In network analysis of human brain proteomic data, Msn was a hub protein of an inflammatory co-expression module positively associated with AD neuropathological features and cognitive dysfunction. CONCLUSIONS: Using FACS coupled with TMT-MS as the method of choice for microglial proteomics, we define a core set of highly-abundant adult microglial proteins. Among these, we validate Msn as highly-abundant in plaque-associated microglia with relevance to human AD.


Subject(s)
Alzheimer Disease/metabolism , Flow Cytometry , Macrophages/metabolism , Microfilament Proteins/metabolism , Microglia/metabolism , Animals , Brain/metabolism , Cognitive Dysfunction/pathology , Disease Models, Animal , Endoplasmic Reticulum/metabolism , Flow Cytometry/methods , Humans , Mice , Proteomics/methods
8.
Neurobiol Dis ; 34(3): 417-31, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19250966

ABSTRACT

More than 80 years after iron accumulation was initially described in the substantia nigra (SN) of Parkinson's disease (PD) patients, the mechanisms responsible for this phenomenon are still unknown. Similarly, how iron is delivered to its major recipients in the cell - mitochondria and the respiratory complexes - has yet to be elucidated. Here, we report a novel transferrin/transferrin receptor 2 (Tf/TfR2)-mediated iron transport pathway in mitochondria of SN dopamine neurons. We found that TfR2 has a previously uncharacterized mitochondrial targeting sequence that is sufficient to import the protein into these organelles. Importantly, the Tf/TfR2 pathway can deliver Tf bound iron to mitochondria and to the respiratory complex I as well. The pathway is redox-sensitive and oxidation of Tf thiols to disulfides induces release from Tf of highly reactive ferrous iron, which contributes to free radical production. In the rotenone model of PD, Tf accumulates in dopamine neurons, with much of it accumulating in the mitochondria. This is associated with iron deposition in SN, similar to what occurs in PD. In the human SN, TfR2 is also found in mitochondria of dopamine neurons, and in PD there is a dramatic increase of oxidized Tf in SN. Thus, we have discovered a novel mitochondrial iron transport system that goes awry in PD, and which may provide a new target for therapeutic intervention.


Subject(s)
Iron/metabolism , Mitochondria/physiology , Parkinson Disease, Secondary/metabolism , Receptors, Transferrin/metabolism , Substantia Nigra/physiopathology , Transferrin/metabolism , Aged , Animals , Dopamine/metabolism , Electron Transport Complex I/metabolism , Humans , Macaca fascicularis , Macaca mulatta , Neurons/physiology , Oxidation-Reduction , Parkinson Disease/physiopathology , Parkinson Disease, Secondary/chemically induced , Rats , Rats, Inbred Lew , Rotenone , Signal Transduction
9.
Neurobiol Dis ; 31(3): 309-15, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18573343

ABSTRACT

Fas-associated factor 1 or FAF1 is a Fas-binding protein implicated in apoptosis. FAF1 is the product of a gene at PARK 10 locus on chromosome 1p32, a locus associated with late-onset PD [Hicks, A.A., Petursson, H., Jonsson, T., Stefansson, H., Johannsdottir, H.S., Sainz, J., Frigge, M.L.et al., 2002. A susceptibility gene for late-onset idiopathic Parkinson's disease. Ann Neurol. 52, 549-555.]. In the present study we investigated the role of FAF1 in cell death and in Parkinson's disease (PD) pathogenesis. FAF1 levels were significantly increased in frontal cortex of PD as well as in PD cases with Alzheimer's disease (AD) pathology compared to control cases. Changes in FAF1 expression were specific to PD-related alpha-synuclein pathology and nigral cell loss. In addition, PD-related insults including, mitochondrial complex I inhibition, oxidative stress, and increased alpha-synuclein expression specifically increased endogenous FAF1 expression in vitro. Increased FAF1 levels induced cell death and significantly potentiated toxic effects of PD-related stressors including, oxidative stress, mitochondrial complex I inhibition and proteasomal inhibition. These studies, together with previous genetic linkage studies, highlight the potential significance of FAF1 in pathogenesis of idiopathic PD.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Brain/metabolism , Neurons/metabolism , Parkinson Disease/genetics , Parkinson Disease/metabolism , Aged , Aged, 80 and over , Apoptosis Regulatory Proteins , Brain/pathology , Brain/physiopathology , Cell Death/genetics , Cell Line , Chromosomes, Human, Pair 1/genetics , Electron Transport Complex I/metabolism , Energy Metabolism/genetics , Frontal Lobe/metabolism , Frontal Lobe/pathology , Frontal Lobe/physiopathology , Gene Expression Regulation/genetics , Genetic Predisposition to Disease , Humans , Middle Aged , Mitochondria/metabolism , Nerve Degeneration/genetics , Nerve Degeneration/metabolism , Nerve Degeneration/physiopathology , Neurons/pathology , Oxidative Stress/genetics , Parkinson Disease/physiopathology , Substantia Nigra/metabolism , Substantia Nigra/pathology , Substantia Nigra/physiopathology , alpha-Synuclein/metabolism
10.
Front Immunol ; 9: 405, 2018.
Article in English | MEDLINE | ID: mdl-29552013

ABSTRACT

In the central nervous system (CNS), microglia are innate immune mononuclear phagocytes (CNS MPs) that can phagocytose infectious particles, apoptotic cells, neurons, and pathological protein aggregates, such as Aß in Alzheimer's disease (AD). While CD11b+CD45low microglia account for the majority of CNS MPs, a small population of CD11b+CD45high CNS MPs is also recognized in AD that surround Aß plaques. These transcriptionally and pathologically unique CD45high cells have unclear origin and undefined phagocytic characteristics. We have comprehensively validated rapid flow cytometric assays of bulk-phase and amyloid ß fibril (fAß) phagocytosis and applied these to study acutely isolated CNS MPs. Using these methods, we provide novel insights into differential abilities of CD11b+ CD45low and CD45high CNS MPs to phagocytose macroparticles and fAß under normal, acute, and chronic neuroinflammatory states. CD45high CNS MPs also highly upregulate TREM2, CD11c, and several disease-associated microglia signature genes and have a higher phagocytic capacity for Aß as compared to CD45low microglia in the 5xFAD mouse model of AD that becomes more apparent with aging. Our data suggest an overall pro-phagocytic and protective role for CD11b+CD45high CNS MPs in neurodegeneration, which if promoted, could be beneficial.


Subject(s)
Aging/immunology , Alzheimer Disease/immunology , Amyloid beta-Peptides/metabolism , Brain/pathology , Microglia/immunology , Phagocytes/immunology , Phagocytosis , Amyloid beta-Peptides/immunology , Animals , Cell Line , Disease Models, Animal , Humans , Leukocyte Common Antigens/metabolism , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Mononuclear Phagocyte System , Receptors, Immunologic/metabolism
11.
Mol Neurodegener ; 13(1): 34, 2018 06 28.
Article in English | MEDLINE | ID: mdl-29954413

ABSTRACT

BACKGROUND: Microglia are innate immune cells of the brain that perform phagocytic and inflammatory functions in disease conditions. Transcriptomic studies of acutely-isolated microglia have provided novel insights into their molecular and functional diversity in homeostatic and neurodegenerative disease states. State-of-the-art mass spectrometry methods can comprehensively characterize proteomic alterations in microglia in neurodegenerative disorders, potentially providing novel functionally relevant molecular insights that are not provided by transcriptomics. However, comprehensive proteomic profiling of adult primary microglia in neurodegenerative disease conditions has not been performed. METHODS: We performed quantitative mass spectrometry based proteomic analyses of purified CD11b+ acutely-isolated microglia from adult (6 mo) mice in normal, acute neuroinflammatory (LPS-treatment) and chronic neurodegenerative states (5xFAD model of Alzheimer's disease [AD]). Differential expression analyses were performed to characterize specific microglial proteomic changes in 5xFAD mice and identify overlap with LPS-induced pro-inflammatory changes. Our results were also contrasted with existing proteomic data from wild-type mouse microglia and from existing microglial transcriptomic data from wild-type and 5xFAD mice. Neuropathological validation studies of select proteins were performed in human AD and 5xFAD brains. RESULTS: Of 4133 proteins identified, 187 microglial proteins were differentially expressed in the 5xFAD mouse model of AD pathology, including proteins with previously known (Apoe, Clu and Htra1) as well as previously unreported relevance to AD biology (Cotl1 and Hexb). Proteins upregulated in 5xFAD microglia shared significant overlap with pro-inflammatory changes observed in LPS-treated mice. Several proteins increased in human AD brain were also upregulated by 5xFAD microglia (Aß peptide, Apoe, Htra1, Cotl1 and Clu). Cotl1 was identified as a novel microglia-specific marker with increased expression and strong association with AD neuropathology. Apoe protein was also detected within plaque-associated microglia in which Apoe and Aß were highly co-localized, suggesting a role for Apoe in phagocytic clearance of Aß. CONCLUSIONS: We report a comprehensive proteomic study of adult mouse microglia derived from acute neuroinflammation and AD models, representing a valuable resource to the neuroscience research community. We highlight shared and unique microglial proteomic changes in acute neuroinflammation aging and AD mouse models and identify novel roles for microglial proteins in human neurodegeneration.


Subject(s)
Alzheimer Disease/immunology , Alzheimer Disease/metabolism , Microglia/immunology , Microglia/metabolism , Animals , Female , Mice , Mice, Inbred C57BL , Proteomics
12.
J Neuropathol Exp Neurol ; 66(10): 955-64, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17917589

ABSTRACT

The PARK10 locus is associated with idiopathic Parkinson disease (PD), but the responsible gene remains to be identified. Genes associated with familial PD, as well as biochemical evidence from sporadic PD and animal models, have implicated components of the ubiquitin-proteasome system in PD pathogenesis. One attractive candidate gene at the PARK10 locus is RING-Finger Protein 11 (RNF11), the deduced amino acid sequence of which predicts a RING-H2 domain common to E3 ubiquitin ligases such as parkin. To facilitate understanding of this protein and its possible role in PD, we characterized the expression and localization of RNF11 in brain. We detected RNF11 transcript and protein and provided the first direct evidence that RNF11 is expressed in brain. Immunohistochemical analysis of RNF11 protein in rat and human brain, using 2 different antibodies, corroborated the mRNA findings. Both antibodies show that RNF11 is restricted to neurons and excluded from white matter. Moreover, RNF11 is expressed by vulnerable neurons of the substantia nigra and sequestered into Lewy bodies in brains of patients with idiopathic PD. Collectively, these findings identify RNF11 as a strong candidate gene at the PARK10 locus and highlight its potential significance in the development of the common form of PD.


Subject(s)
Brain Chemistry/genetics , Carrier Proteins/genetics , Lewy Bodies/metabolism , Neurons/metabolism , Parkinson Disease/metabolism , Aged , Aged, 80 and over , Amino Acid Sequence , Animals , Autopsy , Blotting, Northern , DNA-Binding Proteins , Female , Gene Expression , Humans , Immunohistochemistry , Lewy Bodies/genetics , Male , Middle Aged , Molecular Sequence Data , Paraffin Embedding , Parkinson Disease/genetics , Plasmids/genetics , Rats
13.
J Neurosci ; 24(29): 6417-26, 2004 Jul 21.
Article in English | MEDLINE | ID: mdl-15269251

ABSTRACT

These experiments re-examined the notion that reduced activity in the external pallidal segment (GPe) results in the abnormalities of neuronal discharge in the subthalamic nucleus (STN) and the internal pallidal segment (GPi) and in the development of parkinsonian motor signs. Extracellular recording in two rhesus monkeys, which had been rendered parkinsonian by treatment with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), revealed that the average neuronal discharge rate decreased in GPe but increased in STN and GPi. After MPTP, neurons in all three nuclei tended to discharge in oscillatory bursts. In addition, GABA release in STN (measured with microdialysis) was reduced, indicative of reduced activity along the GPe-STN pathway. Finally, the concentration of glutamic acid dehydrogenase (GAD; measured with autoimmunoradiography) was increased in GPe and GPi, likely reflecting increased striatal input and increased activity of local axon collaterals, respectively. Surprisingly, GAD protein in STN remained unchanged, indicating that the usual assumption that GAD levels are determined primarily by the overall activity of GABAergic elements may be too simplistic. The results from the MPTP-treated animals were compared with results obtained in a second group of three animals with ibotenic acid lesions of GPe. GPe lesions resulted in increased discharge in STN and GPi, comparable with the changes seen after MPTP but did not induce oscillatory bursting and had no behavioral effects. The results indicate that a mere reduction of GPe activity does not produce parkinsonism. Other changes, such as altered discharge patterns in STN and GPi, may play an important role in the generation of parkinsonism.


Subject(s)
Globus Pallidus/physiopathology , MPTP Poisoning/etiology , Action Potentials , Animals , Basal Ganglia/chemistry , Basal Ganglia/pathology , Behavior, Animal , Globus Pallidus/metabolism , Globus Pallidus/pathology , MPTP Poisoning/pathology , MPTP Poisoning/physiopathology , Macaca mulatta , Microdialysis , Neurons/physiology , Oxidoreductases Acting on CH-NH Group Donors/metabolism , Subthalamic Nucleus/metabolism , Subthalamic Nucleus/physiopathology , gamma-Aminobutyric Acid/metabolism
14.
J Neurosci ; 22(16): 7006-15, 2002 Aug 15.
Article in English | MEDLINE | ID: mdl-12177198

ABSTRACT

Chronic systemic complex I inhibition caused by rotenone exposure induces features of Parkinson's disease (PD) in rats, including selective nigrostriatal dopaminergic degeneration and formation of ubiquitin- and alpha-synuclein-positive inclusions (Betarbet et al., 2000). To determine underlying mechanisms of rotenone-induced cell death, we developed a chronic in vitro model based on treating human neuroblastoma cells with 5 nm rotenone for 1-4 weeks. For up to 4 weeks, cells grown in the presence of rotenone had normal morphology and growth kinetics, but at this time point, approximately 5% of cells began to undergo apoptosis. Short-term rotenone treatment (1 week) elevated soluble alpha-synuclein protein levels without changing message levels, suggesting that alpha-synuclein degradation was retarded. Chronic rotenone exposure (4 weeks) increased levels of SDS-insoluble alpha-synuclein and ubiquitin. After a latency of >2 weeks, rotenone-treated cells showed evidence of oxidative stress, including loss of glutathione and increased oxidative DNA and protein damage. Chronic rotenone treatment (4 weeks) caused a slight elevation in basal apoptosis and markedly sensitized cells to further oxidative challenge. In response to H2O2, there was cytochrome c release from mitochondria, caspase-3 activation, and apoptosis, all of which occurred earlier and to a much greater extent in rotenone-treated cells; caspase inhibition provided substantial protection. These studies indicate that chronic low-grade complex I inhibition caused by rotenone exposure induces accumulation and aggregation of alpha-synuclein and ubiquitin, progressive oxidative damage, and caspase-dependent death, mechanisms that may be central to PD pathogenesis.


Subject(s)
Mitochondria/drug effects , NADH, NADPH Oxidoreductases/antagonists & inhibitors , Neurons/drug effects , Parkinson Disease/metabolism , Rotenone/pharmacology , Animals , Antiparkinson Agents/pharmacology , Apoptosis/drug effects , Caspase 3 , Caspase Inhibitors , Caspases/metabolism , Cell Respiration/drug effects , Cytochrome c Group/metabolism , DNA Damage/drug effects , Drug Synergism , Electron Transport Complex I , Enzyme Inhibitors/pharmacology , Glutathione/metabolism , Humans , Hydrogen Peroxide/pharmacology , Mitochondria/metabolism , Nerve Tissue Proteins/metabolism , Neuroblastoma/drug therapy , Neuroblastoma/metabolism , Neurons/metabolism , Neurons/pathology , Oxidants/pharmacology , Oxidation-Reduction/drug effects , Oxidative Stress/drug effects , Parkinson Disease/pathology , Parkinson Disease, Secondary/chemically induced , Synucleins , Time , Tumor Cells, Cultured , Ubiquitin/metabolism , Uncoupling Agents/pharmacology , alpha-Synuclein
15.
J Neurosci ; 23(34): 10756-64, 2003 Nov 26.
Article in English | MEDLINE | ID: mdl-14645467

ABSTRACT

Exposure of rats to the pesticide and complex I inhibitor rotenone reproduces features of Parkinson's disease, including selective nigrostriatal dopaminergic degeneration and alpha-synuclein-positive cytoplasmic inclusions (Betarbet et al., 2000; Sherer et al., 2003). Here, we examined mechanisms of rotenone toxicity using three model systems. In SK-N-MC human neuroblastoma cells, rotenone (10 nm to 1 microm) caused dose-dependent ATP depletion, oxidative damage, and death. To determine the molecular site of action of rotenone, cells were transfected with the rotenone-insensitive single-subunit NADH dehydrogenase of Saccharomyces cerevisiae (NDI1), which incorporates into the mammalian ETC and acts as a "replacement" for endogenous complex I. In response to rotenone, NDI1-transfected cells did not show mitochondrial impairment, oxidative damage, or death, demonstrating that these effects of rotenone were caused by specific interactions at complex I. Although rotenone caused modest ATP depletion, equivalent ATP loss induced by 2-deoxyglucose was without toxicity, arguing that bioenergetic defects were not responsible for cell death. In contrast, reducing oxidative damage with antioxidants, or by NDI1 transfection, blocked cell death. To determine the relevance of rotenone-induced oxidative damage to dopaminergic neuronal death, we used a chronic midbrain slice culture model. In this system, rotenone caused oxidative damage and dopaminergic neuronal loss, effects blocked by alpha-tocopherol. Finally, brains from rotenone-treated animals demonstrated oxidative damage, most notably in midbrain and olfactory bulb, dopaminergic regions affected by Parkinson's disease. These results, using three models of increasing complexity, demonstrate the involvement of oxidative damage in rotenone toxicity and support the evaluation of antioxidant therapies for Parkinson's disease.


Subject(s)
Parkinsonian Disorders/chemically induced , Parkinsonian Disorders/physiopathology , Rotenone/toxicity , Adenosine Triphosphate/deficiency , Adenosine Triphosphate/metabolism , Animals , Antioxidants/pharmacology , Cell Death/drug effects , Cell Line , Disease Models, Animal , Dopamine/metabolism , Dose-Response Relationship, Drug , Electron Transport Complex I/antagonists & inhibitors , Enzyme Inhibitors/toxicity , Humans , In Vitro Techniques , Mesencephalon/drug effects , Mesencephalon/metabolism , Mesencephalon/pathology , Neuroblastoma/drug therapy , Neuroblastoma/metabolism , Neuroblastoma/pathology , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Neuroprotective Agents/pharmacology , Olfactory Bulb/drug effects , Olfactory Bulb/pathology , Oxidative Stress/drug effects , Parkinsonian Disorders/pathology , Rats , Rats, Inbred Lew , Time
16.
Brain Res ; 1622: 328-38, 2015 Oct 05.
Article in English | MEDLINE | ID: mdl-26168900

ABSTRACT

The misfolding and aggregation of the Aß peptide - a fundamental event in the pathogenesis of Alzheimer׳s disease - can be instigated in the brains of experimental animals by the intracranial infusion of brain extracts that are rich in aggregated Aß. Recent experiments have found that the peripheral (intraperitoneal) injection of Aß seeds induces Aß deposition in the brains of APP-transgenic mice, largely in the form of cerebral amyloid angiopathy. Macrophage-type cells normally are involved in pathogen neutralization and antigen presentation, but under some circumstances, circulating monocytes have been found to act as vectors for the transport of pathogenic agents such as viruses and prions. The present study assessed the ability of peripheral monocytes to transport Aß aggregates from the peritoneal cavity to the brain. Our initial experiments showed that intravenously delivered macrophages that had previously ingested fluorescent nanobeads as tracers migrate primarily to peripheral organs such as spleen and liver, but that a small number also reach the brain parenchyma. We next injected CD45.1-expressing monocytes from donor mice intravenously into CD45.2-expressing host mice; after 24h, analysis by fluorescence-activated cell sorting (FACS) and histology confirmed that some CD45.1 monocytes enter the brain, particularly in the superficial cortex and around blood vessels. When the donor monocytes are first exposed to Aß-rich brain extracts from human AD cases, a subset of intravenously delivered Aß-containing cells migrate to the brain. These experiments indicate that, in mouse models, circulating monocytes are potential vectors by which exogenously delivered, aggregated Aß travels from periphery to brain, and more generally support the hypothesis that macrophage-type cells can participate in the dissemination of proteopathic seeds.


Subject(s)
Amyloid beta-Peptides/metabolism , Brain/metabolism , Monocytes/metabolism , Animals , Biological Transport , Brain/blood supply , Flow Cytometry , Humans , Immunohistochemistry , Leukocyte Common Antigens/metabolism , Mice, Inbred C57BL , Monocytes/transplantation , Spleen/metabolism
17.
Brain Pathol ; 12(4): 499-510, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12408237

ABSTRACT

Parkinson's disease (PD) is a progressive neurological disorder marked by nigrostriatal dopaminergic degeneration and development of cytoplasmic proteinaceous aggregates known as Lewy bodies. Although the pathogenic mechanisms responsible for PD are not completely understood, many clues have come from biochemical, epidemiological, and genetic studies. Mutations in certain genes found in rare, familial cases of PD, such as alpha-synuclein and parkin, suggest a role for the ubiquitin-proteosome system and aberrant protein aggregation. Biochemical analyses have implicated mitochondrial dysfunction in PD. Epidemiological and animal model studies point to a role for environmental toxins, some of which are mitochondrial inhibitors. Mitochondrial dysfunction, resulting from either genetic defects, environmental exposures or an interaction between the two, may cause alpha-synuclein aggregation or neurodegeneration through oxidative stress or excitotoxicity. A better understanding of the mechanisms underlying PD should reveal novel therapeutic targets.


Subject(s)
Environmental Exposure , Neurotoxins/toxicity , Parkinson Disease/etiology , Animals , Brain/drug effects , Brain/metabolism , Brain/physiopathology , Humans , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/pathology , Mutation/genetics , Nerve Tissue Proteins/drug effects , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Oxidative Stress/drug effects , Oxidative Stress/physiology , Parkinson Disease/genetics , Parkinson Disease/metabolism , Synucleins , alpha-Synuclein
18.
Neuroscientist ; 8(3): 192-7, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12061498

ABSTRACT

Parkinson's disease (PD) is a common and disabling neurodegenerative disease marked by progressive motor dysfunction, which results from selective degeneration of the nigrostriatal pathway. Epidemiological studies indicate that exposure to pesticides, rural living, farming, and drinking well water are associated with an increased risk of developing PD. Rare cases of PD are caused by mutations in nuclear genes, and there is increasing evidence for susceptibility genes that alter disease risk. Parkinson's disease is also associated with a systemic defect in mitochondrial complex I activity. Animal models indicate that exposure to inhibitors of mitochondrial complex I, including pesticides, is sufficient to reproduce the features of PD, but genetic factors clearly modulate susceptibility. Complex I defects may result in oxidative stress and increase the susceptibility of neurons to excitotoxic death. In this way, environmental exposures and mitochondrial dysfunction may interact and result in neurodegeneration.


Subject(s)
Environmental Exposure/adverse effects , Mitochondria/metabolism , Parkinson Disease, Secondary/chemically induced , Parkinson Disease/etiology , Parkinson Disease/metabolism , Animals , Electron Transport Complex I , Humans , Mutation , NADH, NADPH Oxidoreductases/metabolism , Oxidative Stress , Parkinson Disease/genetics , Parkinson Disease, Secondary/metabolism , Pesticides/adverse effects , Risk Factors
19.
Neurosci Lett ; 341(2): 87-90, 2003 May 01.
Article in English | MEDLINE | ID: mdl-12686372

ABSTRACT

Chronic rotenone exposure reproduces features of Parkinson's disease (PD) (Nat. Neurosci. 3 (2000) 1301; Exp. Neurol. 179 (2003) 9). We investigated the role of glial activation in rotenone toxicity in vivo. Male Lewis rats received 2-3 mg/kg rotenone per day for up to 4 weeks. In 50% of surviving rotenone-treated animals, there was nigrostriatal dopaminergic degeneration, marked by reduced tyrosine hydroxylase immunoreactivity). Extensive microglial activation, determined by OX-42-ir, occurred in striatum and nigra of rotenone-treated animals, and was prominent before anatomical evidence of dopaminergic lesions. Microglia enlarged and developed short, stubby processes in rotenone-treated animals. Rotenone-induced microglial activation was less pronounced in cortex. Reactive astrocytosis was minimal and limited to a thin rim around the lesion. Marked microglial activation with minimal astrocytosis is another pathological feature of PD reproduced by rotenone treatment.


Subject(s)
Antigens, CD , Antigens, Neoplasm , Antigens, Surface , Avian Proteins , Blood Proteins , Insecticides/toxicity , Microglia/metabolism , Parkinson Disease/metabolism , Rotenone/toxicity , Animals , Astrocytes/metabolism , Astrocytes/pathology , Basigin , Corpus Striatum/metabolism , Corpus Striatum/pathology , Disease Models, Animal , Environmental Exposure , Glial Fibrillary Acidic Protein/metabolism , Immunohistochemistry , Male , Membrane Glycoproteins/metabolism , Microglia/pathology , Parkinson Disease/enzymology , Parkinson Disease/pathology , Rats , Rats, Inbred Lew , Substantia Nigra/metabolism , Substantia Nigra/pathology , Tyrosine 3-Monooxygenase/metabolism
20.
Parkinsonism Relat Disord ; 9 Suppl 2: S59-64, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12915069

ABSTRACT

Parkinson's disease (PD) is occasionally caused by single gene mutations or by single toxic exposures, but most cases of PD are probably caused by some combination of genetic susceptibility and environmental exposure. Using rotenone as a prototype for an environmental toxicant, we argue here that genetic and environmental causes of PD converge on common pathogenic mechanisms. If so, protective strategies devised for one type of PD may be broadly useful for other forms of the disease.


Subject(s)
Insecticides/adverse effects , Parkinsonian Disorders/chemically induced , Parkinsonian Disorders/genetics , Rotenone/adverse effects , Environmental Exposure , Genetic Predisposition to Disease , Humans , Mitochondria/drug effects , Parkinsonian Disorders/etiology
SELECTION OF CITATIONS
SEARCH DETAIL