Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
J Biomed Sci ; 31(1): 24, 2024 Feb 23.
Article in English | MEDLINE | ID: mdl-38395904

ABSTRACT

BACKGROUND: Mutations in CHCHD2 have been linked to Parkinson's disease, however, their exact pathophysiologic roles are unclear. The p32 protein has been suggested to interact with CHCHD2, however, the physiological functions of such interaction in the context of PD have not been clarified. METHODS: Interaction between CHCHD2 and p32 was confirmed by co-immunoprecipitation experiments. We studied the effect of p32-knockdown in the transgenic Drosophila and Hela cells expressing the wild type and the pathogenic variants of hCHCHD2. We further investigated the rescue ability of a custom generated p32-inhibitor in these models as well as in the human fibroblast derived neural precursor cells and the dopaminergic neurons harboring hCHCHD2-Arg145Gln. RESULTS: Our results showed that wildtype and mutant hCHCHD2 could bind to p32 in vitro, supported by in vivo interaction between human CHCHD2 and Drosophila p32. Knockdown of p32 reduced mutant hCHCHD2 levels in Drosophila and in vitro. In Drosophila hCHCHD2 models, inhibition of p32 through genetic knockdown and pharmacological treatment using a customized p32-inhibitor restored dopaminergic neuron numbers and improved mitochondrial morphology. These were correlated with improved locomotor function, reduced oxidative stress and decreased mortality. Consistently, Hela cells expressing mutant hCHCHD2 showed improved mitochondrial morphology and function after treatment with the p32-inhibitor. As compared to the isogenic control cells, large percentage of the mutant neural precursor cells and dopaminergic neurons harboring hCHCHD2-Arg145Gln contained fragmented mitochondria which was accompanied by lower ATP production and cell viability. The NPCs harboring hCHCHD2-Arg145Gln also had a marked increase in α-synuclein expression. The p32-inhibitor was able to ameliorate the mitochondrial fragmentation, restored ATP levels, increased cell viability and reduced α-synuclein level in these cells. CONCLUSIONS: Our study identified p32 as a modulator of CHCHD2, possibly exerting its effects by reducing the toxic mutant hCHCHD2 expression and/or mitigating the downstream effects. Inhibition of the p32 pathway can be a potential therapeutic intervention for CHCHD2-linked PD and diseases involving mitochondrial dysfunction.


Subject(s)
Neural Stem Cells , Parkinson Disease , Animals , Humans , Adenosine Triphosphate/metabolism , alpha-Synuclein/genetics , alpha-Synuclein/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dopaminergic Neurons/metabolism , Drosophila/genetics , Drosophila/metabolism , HeLa Cells , Neural Stem Cells/metabolism , Parkinson Disease/drug therapy , Parkinson Disease/genetics , Parkinson Disease/metabolism , Phenotype , Transcription Factors/genetics , Transcription Factors/metabolism
2.
Hum Mutat ; 38(8): 978-987, 2017 08.
Article in English | MEDLINE | ID: mdl-28432706

ABSTRACT

Mutations and polymorphic risk variant of coiled-coil-helix-coiled-coil-helix domain containing 2 (CHCHD2) have been associated with late-onset Parkinson disease. In vivo pathological evidence of CHCHD2 mutations is currently lacking. Utilizing transgenic Drosophila model, we examined the relative pathophysiologic effect of the pathogenic (c.182C>T, p.Thr61Ile and c.434G>A, p.Arg145Gln) and the risk (c.5C>T, p.Pro2Leu) CHCHD2 variants. All the transgenic models exhibited locomotor dysfunction that could be exacerbated by rotenone exposure, dopaminergic neuron degeneration, reduction in lifespan, mitochondrial dysfunction, oxidative stress, and impairment in synaptic transmission. However, both mutants showed more severe early motor dysfunction, dopaminergic neuronal loss, and higher hydrogen peroxide production compared with the risk variant. p.Thr61Ile (co-segregated in three independent PD families) displayed the most severe phenotype followed by p.Arg145Gln (present only in index patient). We treated the transgenic flies with Ebselen, a mitochondrial hydrogen peroxide scavenger compound; Ebselen appears to be more effective in ameliorating motor function in the mutant than the risk variant models. We provide the first in vivo evidence of the pathological effects associated with CHCHD2 mutations. There was a difference in the pathological and drug response effects between the pathogenic and the risk variants. Ebselen may be a useful neuroprotective drug for carriers of CHCHD2 mutations.


Subject(s)
Drosophila Proteins/genetics , Mitochondrial Proteins/genetics , Animals , Blotting, Western , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Drosophila , Female , Immunohistochemistry , Locomotion/drug effects , Male , Microscopy, Electron, Transmission , Mutation/genetics , Oxidative Stress/drug effects , Oxidative Stress/genetics , Reverse Transcriptase Polymerase Chain Reaction , Rotenone/pharmacology , Synaptic Transmission/drug effects , Synaptic Transmission/genetics
3.
Hum Genet ; 135(11): 1223-1232, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27395408

ABSTRACT

Essential tremor (ET) is one of the most common adult-onset neurological disorders which produce motor and non-motor symptoms. To date, there are no gold standard pathological hallmarks of ET, and despite a strong genetic contribution toward ET development, only a few pathogenic mutations have been identified. Recently, a pathogenic FUS-Q290X mutation has been reported in a large ET-affected family; however, the pathophysiologic mechanism underlying FUS-linked ET is unknown. Here, we generated transgenic Drosophila expressing hFUS-WT and hFUS-Q290X and targeted their expression in different tissues. We found that the targeted expression of hFUS-Q290X in the dopaminergic and the serotonergic neurons did not cause obvious neuronal degeneration, but it resulted in motor dysfunction which was accompanied by impairment in the GABAergic pathway. The involvement of the GABAergic pathway was supported by rescue of motor symptoms with gabapentin. Interestingly, we observed gender specific downregulation of GABA-R and NMDA-R expression and reduction in serotonin level. Overexpression of hFUS-Q290X also caused an increase in longevity and this was accompanied by downregulation of the IIS/TOR signalling pathway. Our in vivo studies of the hFUS-Q290X mutation in Drosophila link motor dysfunction to impairment in the GABAergic pathway. Our findings would facilitate further efforts in unravelling the pathophysiology of ET.


Subject(s)
Essential Tremor/genetics , Longevity/genetics , Motor Disorders/genetics , RNA-Binding Protein FUS/genetics , Receptors, GABA/genetics , Amines/metabolism , Animals , Animals, Genetically Modified , Cyclohexanecarboxylic Acids/metabolism , Disease Models, Animal , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Drosophila melanogaster/genetics , Essential Tremor/pathology , GABAergic Neurons/metabolism , GABAergic Neurons/pathology , Gabapentin , Gene Expression Regulation, Developmental , Humans , Motor Disorders/pathology , Mutation , Organ Specificity , RNA-Binding Protein FUS/biosynthesis , Receptors, GABA/metabolism , Receptors, N-Methyl-D-Aspartate/genetics , Serotonergic Neurons/metabolism , Serotonergic Neurons/pathology , gamma-Aminobutyric Acid/genetics , gamma-Aminobutyric Acid/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...