Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Brain ; 144(6): 1661-1669, 2021 07 28.
Article in English | MEDLINE | ID: mdl-33760024

ABSTRACT

α-Synuclein aggregation at the synapse is an early event in Parkinson's disease and is associated with impaired striatal synaptic function and dopaminergic neuronal death. The cysteine string protein (CSPα) and α-synuclein have partially overlapping roles in maintaining synaptic function and mutations in each cause neurodegenerative diseases. CSPα is a member of the DNAJ/HSP40 family of co-chaperones and like α-synuclein, chaperones the SNARE complex assembly and controls neurotransmitter release. α-Synuclein can rescue neurodegeneration in CSPαKO mice. However, whether α-synuclein aggregation alters CSPα expression and function is unknown. Here we show that α-synuclein aggregation at the synapse is associated with a decrease in synaptic CSPα and a reduction in the complexes that CSPα forms with HSC70 and STGa. We further show that viral delivery of CSPα rescues in vitro the impaired vesicle recycling in PC12 cells with α-synuclein aggregates and in vivo reduces synaptic α-synuclein aggregates increasing monomeric α-synuclein and restoring normal dopamine release in 1-120hαSyn mice. These novel findings reveal a mechanism by which α-synuclein aggregation alters CSPα at the synapse, and show that CSPα rescues α-synuclein aggregation-related phenotype in 1-120hαSyn mice similar to the effect of α-synuclein in CSPαKO mice. These results implicate CSPα as a potential therapeutic target for the treatment of early-stage Parkinson's disease.


Subject(s)
Corpus Striatum/metabolism , Dopamine/metabolism , HSP40 Heat-Shock Proteins/metabolism , Membrane Proteins/metabolism , Protein Aggregation, Pathological/metabolism , alpha-Synuclein/metabolism , Animals , Corpus Striatum/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Parkinson Disease/metabolism , Parkinson Disease/pathology , Protein Aggregation, Pathological/pathology , Synapses/metabolism , Synapses/pathology
2.
Hum Mol Genet ; 29(2): 216-227, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31813995

ABSTRACT

Huntington's disease (HD) is a neurodegenerative disorder caused by an expanded polyglutamine tract in the huntingtin (HTT) protein. Mutant HTT (mHTT) toxicity is caused by its aggregation/oligomerization. The striatum is the most vulnerable region, although all brain regions undergo neuronal degeneration in the disease. Here we show that the levels of Bim, a BH3-only protein, are significantly increased in HD human post-mortem and HD mouse striata, correlating with neuronal death. Bim reduction ameliorates mHTT neurotoxicity in HD cells. In the HD mouse model, heterozygous Bim knockout significantly mitigates mHTT accumulation and neuronal death, ameliorating disease-associated phenotypes and lifespan. Therefore, Bim could contribute to the progression of HD.


Subject(s)
Bcl-2-Like Protein 11/metabolism , Corpus Striatum/metabolism , Huntingtin Protein/genetics , Huntington Disease/metabolism , Neurons/pathology , Aged , Animals , Bcl-2-Like Protein 11/genetics , Corpus Striatum/pathology , Disease Models, Animal , Disease Progression , Female , Gene Knockout Techniques , Heterozygote , Humans , Huntingtin Protein/metabolism , Huntington Disease/genetics , Huntington Disease/mortality , Huntington Disease/pathology , Male , Mice , Middle Aged , Neurons/metabolism , Phenotype , Protein Aggregates/genetics , RNA, Small Interfering
3.
Acta Neuropathol ; 138(4): 575-595, 2019 10.
Article in English | MEDLINE | ID: mdl-31165254

ABSTRACT

Parkinson's disease (PD) is characterized by the presence of α-synuclein aggregates known as Lewy bodies and Lewy neurites, whose formation is linked to disease development. The causal relation between α-synuclein aggregates and PD is not well understood. We generated a new transgenic mouse line (MI2) expressing human, aggregation-prone truncated 1-120 α-synuclein under the control of the tyrosine hydroxylase promoter. MI2 mice exhibit progressive aggregation of α-synuclein in dopaminergic neurons of the substantia nigra pars compacta and their striatal terminals. This is associated with a progressive reduction of striatal dopamine release, reduced striatal innervation and significant nigral dopaminergic nerve cell death starting from 6 and 12 months of age, respectively. In the MI2 mice, alterations in gait impairment can be detected by the DigiGait test from 9 months of age, while gross motor deficit was detected by rotarod test at 20 months of age when 50% of dopaminergic neurons in the substantia nigra pars compacta are lost. These changes were associated with an increase in the number and density of 20-500 nm α-synuclein species as shown by dSTORM. Treatment with the oligomer modulator anle138b, from 9 to 12 months of age, restored striatal dopamine release, prevented dopaminergic cell death and gait impairment. These effects were associated with a reduction of the inner density of large α-synuclein aggregates and an increase in dispersed small α-synuclein species as revealed by dSTORM. The MI2 mouse model recapitulates the progressive dopaminergic deficit observed in PD, showing that early synaptic dysfunction is associated to fine behavioral motor alterations, precedes dopaminergic axonal loss and neuronal death that become associated with a more consistent motor deficit upon reaching a certain threshold. Our data also provide new mechanistic insight for the effect of anle138b's function in vivo supporting that targeting α-synuclein aggregation is a promising therapeutic approach for PD.


Subject(s)
Cell Death/physiology , Dopaminergic Neurons/pathology , Parkinson Disease/pathology , Protein Aggregation, Pathological/pathology , Substantia Nigra/pathology , alpha-Synuclein/metabolism , Animals , Disease Models, Animal , Gait/genetics , Mice , Mice, Transgenic , Motor Activity/genetics , Parkinson Disease/genetics , Parkinson Disease/metabolism , Protein Aggregation, Pathological/metabolism , Substantia Nigra/metabolism , Tyrosine 3-Monooxygenase/metabolism , alpha-Synuclein/genetics
4.
CNS Neurosci Ther ; 25(7): 825-836, 2019 07.
Article in English | MEDLINE | ID: mdl-30746905

ABSTRACT

The mitochondrion is a unique organelle with a diverse range of functions. Mitochondrial dysfunction is a key pathological process in several neurodegenerative diseases. Mitochondria are mostly important for energy production; however, they also have roles in Ca2+ homeostasis, ROS production, and apoptosis. There are two major systems in place, which regulate mitochondrial integrity, mitochondrial dynamics, and mitophagy. These two processes remove damaged mitochondria from cells and protect the functional mitochondrial population. These quality control systems often become dysfunctional during neurodegenerative diseases, such as Parkinson's and Alzheimer's disease, causing mitochondrial dysfunction and severe neurological symptoms.


Subject(s)
Mitochondria/physiology , Neurodegenerative Diseases/physiopathology , Animals , Humans
5.
Methods Mol Biol ; 1854: 1-12, 2019.
Article in English | MEDLINE | ID: mdl-29936691

ABSTRACT

Autophagy is an intracellular degradation process that mediates the clearance of cytoplasmic components. As well as being an important function for cellular homeostasis, autophagy also promotes the removal of aberrant protein accumulations, such as those seen in conditions like neurodegeneration. The dynamic nature of autophagy requires precise methods to examine the process at multiple stages. The protocols described herein enable the dissection of the complete autophagy process (the "autophagy flux"). These allow for the elucidation of which stages of autophagy may be altered in response to various diseases and treatments.


Subject(s)
Dementia/metabolism , Phagosomes/metabolism , Sequestosome-1 Protein/metabolism , Autophagy , Autopsy , Fluorescent Antibody Technique , HeLa Cells , Humans , Lysosomal Membrane Proteins/metabolism , Receptors, Scavenger/metabolism
6.
Sci Rep ; 8(1): 17394, 2018 11 26.
Article in English | MEDLINE | ID: mdl-30478411

ABSTRACT

Revealing the relationship between dysfunctional genes in blood and brain tissues from patients with Alzheimer's Disease (AD) will help us to understand the pathology of this disease. In this study, we conducted the first such large systematic analysis to identify differentially expressed genes (DEGs) in blood samples from 245 AD cases, 143 mild cognitive impairment (MCI) cases, and 182 healthy control subjects, and then compare these with DEGs in brain samples. We evaluated our findings using two independent AD blood datasets and performed a gene-based genome-wide association study to identify potential novel risk genes. We identified 789 and 998 DEGs common to both blood and brain of AD and MCI subjects respectively, over 77% of which had the same regulation directions across tissues and disease status, including the known ABCA7, and the novel TYK2 and TCIRG1. A machine learning classification model containing NDUFA1, MRPL51, and RPL36AL, implicating mitochondrial and ribosomal function, was discovered which discriminated between AD patients and controls with 85.9% of area under the curve and 78.1% accuracy (sensitivity = 77.6%, specificity = 78.9%). Moreover, our findings strongly suggest that mitochondrial dysfunction, NF-κB signalling and iNOS signalling are important dysregulated pathways in AD pathogenesis.


Subject(s)
Alzheimer Disease/genetics , Biomarkers/blood , Biomarkers/metabolism , Alzheimer Disease/blood , Alzheimer Disease/metabolism , Blood/metabolism , Brain/metabolism , Case-Control Studies , Cognitive Dysfunction/blood , Cognitive Dysfunction/genetics , Cognitive Dysfunction/metabolism , Genome-Wide Association Study , Humans , Machine Learning , Mitochondria/genetics , NF-kappa B/genetics , Nitric Oxide Synthase Type II/genetics , Ribosomes/genetics , Sensitivity and Specificity , Signal Transduction/genetics
7.
Glia ; 66(4): 813-827, 2018 04.
Article in English | MEDLINE | ID: mdl-29315804

ABSTRACT

Early white matter (WM) changes are common in dementia and may contribute to functional decline. We here examine this phenomenon in an induced dementia model for the first time. We report a novel and selective form of myelin injury as the first manifestation of tauopathy in the adult central nervous system. Myelin pathology rapidly followed the induction of a P301 tau mutation associated with fronto-temporal dementia in humans (rTG4510 line). Damage involved focal disruption of the ad-axonal myelin lamella and internal oligodendrocyte tongue process, followed by myelin remodeling with features of re-myelination that included myelin thinning and internodal shortening. The evolution of the re-myelinated phenotype was complete in the molecular layer of the dentate gyrus after 1 month and in the optic nerve (ON) after 9 months of transgene induction and proceeded in the absence of actual demyelination, reactive glial changes or inflammatory response. The initial rapid myelin pathology was associated with loss of WM function and performance decline in a novel recognition test and both these effects largely reversed during the myelin re-modeling phase. The initial phase of myelin injury was accompanied by disruption of the vesicle population present in the axoplasm of hippocampal and ON axons. Axoplasmic vesicle release is significant for the regulation of myelin plasticity and disruption of this pathway may underlie the myelin damage and remodeling evoked by tauopathy. WM dysfunction early in tauopathy will disorder neural circuits, the current findings suggest this event may make a significant contribution to early clinical deficit in dementia.


Subject(s)
Myelin Sheath/pathology , Myelin Sheath/physiology , Tauopathies/pathology , Tauopathies/physiopathology , White Matter/pathology , White Matter/physiopathology , Animals , Astrocytes/pathology , Astrocytes/physiology , Axons/pathology , Axons/physiology , Cytoplasmic Vesicles/metabolism , Cytoplasmic Vesicles/pathology , Disease Progression , Hippocampus/pathology , Hippocampus/physiopathology , Humans , Male , Memory Disorders/pathology , Memory Disorders/physiopathology , Mice, Inbred C57BL , Mice, Transgenic , Optic Nerve/pathology , Optic Nerve/physiopathology , Recognition, Psychology/physiology , Tauopathies/psychology , tau Proteins/genetics , tau Proteins/metabolism
8.
Brain Res ; 1681: 1-13, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29278715

ABSTRACT

Dementia with Lewy bodies (DLB) is the second most prevalent neurodegenerative dementia, where an accumulation of aggregated fibrillar alpha-synuclein in neurons of limbic and forebrain regions of the brain leads to visual hallucination, cognitive impairment of a fluctuating nature and extrapyramidal motor disturbances. Beta-synuclein counteracts aggregation of alpha-synuclein in vitro and in animal models, however it is not clear whether this effect occurs in human Lewy body dementia (LBD) diseases. Here we examine expression of alpha-, beta-synuclein and autophagy markers in the frontal cortex (BA9) and occipital cortex (BA18-19) of patients with neuropathologically confirmed DLB/LBD and age-matched controls. We provide evidence for neuronal upregulation of beta-synuclein within the frontal cortex and its decrease in occipital cortex of DLB patients. While beta-synuclein-containing neurons were consistently devoid of oligomeric alpha-synuclein in the frontal cortex, we did not observe an overall correlation between total beta-synuclein and 5G4 levels (marker of oligomeric alpha-synuclein). The autophagy markers LC3-II and p62 were increased in the areas of beta-synuclein upregulation in DLB brains, and we show attenuation of autophagy flux when beta-synuclein is overexpressed in vitro. Altogether, this data suggests that beta-synuclein changes in DLB may exacerbate neuronal dysfunction caused by accumulation of alpha-synuclein by influencing protein degradation pathways; this should be taken into consideration when designing therapeutic strategies aimed to decrease alpha-synuclein burden in Lewy body diseases.


Subject(s)
Autophagy , Frontal Lobe/metabolism , Lewy Body Disease/metabolism , Neurons/metabolism , Occipital Lobe/metabolism , beta-Synuclein/metabolism , Aged , Aged, 80 and over , Female , Frontal Lobe/pathology , Humans , Lewy Body Disease/pathology , Male , Neurons/pathology , Occipital Lobe/pathology , Up-Regulation , alpha-Synuclein/metabolism
9.
Exp Neurol ; 264: 8-13, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25450466

ABSTRACT

Accumulation and aggregation of alpha-synuclein in cortical and hippocampal areas is a pathological sign for dementia with Lewy bodies (DLB) and Parkinson's disease with dementia. However the mechanisms of alpha-synuclein triggered cellular dysfunction leading to the development of memory impairment is not clear. We have created a mouse model of DLB, where aggregation-prone human truncated (120 amino acid) alpha-synuclein is expressed in forebrain areas under the calcium/calmodulin-dependent protein kinase II alpha (CamKII-alpha) promoter. We have observed the presence of the transgenic protein in target forebrain areas, with small granular cytoplasmic accumulation of aggregated alpha-synuclein. This was associated with a progressive deficit in cortical-hippocampal memory tests including the Barnes maze and novel object recognition. This data suggests that low levels of aggregation prone alpha-synuclein are sufficient to induce memory deficits in mice and that forebrain regions associated with cognitive function may have an increased sensitivity to the truncated toxic form of alpha-synuclein.


Subject(s)
Lewy Body Disease/genetics , Lewy Body Disease/physiopathology , Peptide Fragments , alpha-Synuclein/chemistry , alpha-Synuclein/genetics , Age Factors , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Disease Models, Animal , Exploratory Behavior/physiology , Gene Expression Regulation/genetics , Humans , Lewy Body Disease/pathology , Male , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Prosencephalon/metabolism , Prosencephalon/pathology , RNA, Messenger/metabolism
10.
CNS Neurol Disord Drug Targets ; 12(8): 1094-100, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24040824

ABSTRACT

Pathological aggregation of alpha-synuclein as Lewy-bodies and neurites is a hallmark of a group of neurodegenerative disorders named alpha-synucleinopathies. It is becoming apparent that alpha-synuclein facilitates presynaptic neuronal function in the brain, and events leading to its aggregation produce marked disruption of neurotransmitter release mechanism. We discuss here the literature related to the function of alpha-synuclein at the neuronal synapse in synucleinopathies brains and corresponding animal models.


Subject(s)
Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/physiopathology , Synapses/pathology , Synapses/physiology , alpha-Synuclein/physiology , Animals , Humans , Neurodegenerative Diseases/diagnosis
11.
Exp Neurol ; 248: 541-5, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23933574

ABSTRACT

The presynaptic protein α-synuclein is central to the pathogenesis of α-synucleinopathies. We show that the presence of endogenous mouse α-synuclein leads to higher number of dopaminergic neurons in the substantia nigra of wild-type C57Bl/6J mice compared with C57Bl/6S mice with a spontaneous deletion of the α-synuclein gene or C57Bl/6J mice with a targeted deletion of the α-synuclein gene. This effect of α-synuclein on dopaminergic neuron occurs during development between E10.5 and E13.5 and persists in adult life supporting the involvement of α-synuclein in the development of a subset of dopaminergic neurons.


Subject(s)
Dopaminergic Neurons/cytology , Substantia Nigra/cytology , alpha-Synuclein/metabolism , Animals , Cell Count , Dopaminergic Neurons/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Substantia Nigra/metabolism , alpha-Synuclein/genetics
12.
Neurodegener Dis ; 8(6): 430-7, 2011.
Article in English | MEDLINE | ID: mdl-21576917

ABSTRACT

BACKGROUND: Recent clinical studies have demonstrated that dimebon, a drug originally designed and used as a non-selective antihistamine, ameliorates symptoms and delays progress of mild to moderate forms of Alzheimer's and Huntington's diseases. Although the mechanism of dimebon action on pathological processes in degenerating brain is elusive, results of studies carried out in cell cultures and animal models suggested that this drug might affect the process of pathological accumulation and aggregation of various proteins involved in the pathogenesis of proteinopathies. However, the effect of this drug on the pathology caused by overexpression and aggregation of alpha-synuclein, including Parkinson's disease (PD), has not been assessed. OBJECTIVE: To test if dimebon affected alpha-synuclein-induced pathology using a transgenic animal model. METHODS: We studied the effects of chronic dimebon treatment on transgenic mice expressing the C-terminally truncated (1-120) form of human alpha-synuclein in dopaminergic neurons, a mouse model that recapitulates several biochemical, histopathological and behavioral characteristics of the early stage of PD. RESULTS: Dimebon did not improve balance and coordination of aging transgenic animals or increase the level of striatal dopamine, nor did it prevent accumulation of alpha-synuclein in cell bodies of dopaminergic neurons. CONCLUSION: Our observations suggest that in the studied model of alpha-synucleinopathy dimebon has very limited effect on certain pathological alterations typical of PD and related diseases.


Subject(s)
Dopamine/physiology , Histamine/therapeutic use , Indoles/therapeutic use , Neurons/physiology , alpha-Synuclein/genetics , Animals , Behavior, Animal/drug effects , Biomarkers , Blotting, Western , Brain Chemistry/drug effects , Brain Chemistry/genetics , Cell Count , Chromatography, High Pressure Liquid , Corpus Striatum/drug effects , Corpus Striatum/pathology , Exploratory Behavior/drug effects , Gene Expression/drug effects , Humans , Immunohistochemistry , Mice , Mice, Transgenic , Motor Activity/drug effects , Neurons/pathology , Olfactory Bulb/drug effects , Olfactory Bulb/metabolism , Postural Balance/drug effects , RNA/biosynthesis , RNA/genetics , Ventral Tegmental Area/pathology , alpha-Synuclein/physiology
13.
Brain ; 133(Pt 7): 2032-44, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20534649

ABSTRACT

The pre-synaptic protein alpha-synuclein is the main component of Lewy bodies and Lewy neurites, the defining neuropathological characteristics of Parkinson's disease and dementia with Lewy bodies. Mutations in the alpha-synuclein gene cause familial forms of Parkinson's disease and dementia with Lewy bodies. We previously described a transgenic mouse line expressing truncated human alpha-synuclein(1-120) that develops alpha-synuclein aggregates, striatal dopamine deficiency and reduced locomotion, similar to Parkinson's disease. We now show that in the striatum of these mice, as in Parkinson's disease, synaptic accumulation of alpha-synuclein is accompanied by an age-dependent redistribution of the synaptic SNARE proteins SNAP-25, syntaxin-1 and synaptobrevin-2, as well as by an age-dependent reduction in dopamine release. Furthermore, the release of FM1-43 dye from PC12 cells expressing either human full-length alpha-synuclein(1-140) or truncated alpha-synuclein(1-120) was reduced. These findings reveal a novel gain of toxic function of alpha-synuclein at the synapse, which may be an early event in the pathogenesis of Parkinson's disease.


Subject(s)
Disease Models, Animal , Parkinson Disease/metabolism , Parkinson Disease/physiopathology , SNARE Proteins/metabolism , Synapses/metabolism , Aged , Animals , Exocytosis/genetics , Humans , Infant , Mice , Mice, Inbred C57BL , Mice, Transgenic , Middle Aged , Parkinson Disease/genetics , SNARE Proteins/analysis , SNARE Proteins/genetics , Synapses/genetics , Synapses/pathology , alpha-Synuclein/biosynthesis , alpha-Synuclein/genetics , alpha-Synuclein/physiology
14.
PLoS One ; 5(1): e8762, 2010 Jan 19.
Article in English | MEDLINE | ID: mdl-20098733

ABSTRACT

In patients with Parkinson's disease (PD), the associated pathology follows a characteristic pattern involving inter alia the enteric nervous system (ENS), the dorsal motor nucleus of the vagus (DMV), the intermediolateral nucleus of the spinal cord and the substantia nigra, providing the basis for the neuropathological staging of the disease. Here we report that intragastrically administered rotenone, a commonly used pesticide that inhibits Complex I of the mitochondrial respiratory chain, is able to reproduce PD pathological staging as found in patients. Our results show that low doses of chronically and intragastrically administered rotenone induce alpha-synuclein accumulation in all the above-mentioned nervous system structures of wild-type mice. Moreover, we also observed inflammation and alpha-synuclein phosphorylation in the ENS and DMV. HPLC analysis showed no rotenone levels in the systemic blood or the central nervous system (detection limit [rotenone]<20 nM) and mitochondrial Complex I measurements showed no systemic Complex I inhibition after 1.5 months of treatment. These alterations are sequential, appearing only in synaptically connected nervous structures, treatment time-dependent and accompanied by inflammatory signs and motor dysfunctions. These results strongly suggest that the local effect of pesticides on the ENS might be sufficient to induce PD-like progression and to reproduce the neuroanatomical and neurochemical features of PD staging. It provides new insight into how environmental factors could trigger PD and suggests a transsynaptic mechanism by which PD might spread throughout the central nervous system.


Subject(s)
Disease Models, Animal , Parkinson Disease/pathology , Rotenone/administration & dosage , Animals , Chromatography, High Pressure Liquid , Enteric Nervous System/drug effects , Mice , Mice, Inbred BALB C , Rotenone/pharmacology , Stomach
15.
J Neurochem ; 109(2): 403-15, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19222706

ABSTRACT

Serotonin (or 5-hydroxytryptamine; 5-HT) and monoamine oxidase (MAO) are involved in several physiological functions and pathological conditions. We show that the serotonergic system and its development in zebrafish are similar to those of other vertebrates rendering zebrafish a good model to study them. Development of MAO expression followed a similar time course as the 5-HT system. MAO expression and activity were located in or adjacent to serotonergic nuclei and their targets, especially in the ventral hypothalamus. MAO mRNA was detected in the brain from 24 h post-fertilization and histochemical enzyme activity from 42 h post-fertilization. Deprenyl (100 microM) decreased MAO activity 34-74% depending on the age. Inhibition of MAO by deprenyl strongly increased 5-HT but not dopamine and noradrenaline levels. Deprenyl decreased 5-HT-immunoreactivity in serotonergic neurons and induced novel ectopic 5-HT-immunoreactivity neurons in the diencephalon in a manner dependent on 5-HT uptake. Deprenyl administration decreased locomotion, altered vertical positioning and increased heart rate. Treatment with p-chlorophenylalanine normalized 5-HT levels and rescued the behavioral alteration, indicating that the symptoms were 5-HT dependent. These findings suggest that zebrafish MAO resembles mammalian MAO A more than MAO B, metabolizing mainly 5-HT. Applications of this model of hyperserotonergism include pharmacological and genetic screenings.


Subject(s)
Monoamine Oxidase Inhibitors/pharmacology , Monoamine Oxidase/metabolism , Phenotype , Serotonin/metabolism , Zebrafish Proteins/antagonists & inhibitors , Animals , Brain/drug effects , Brain/enzymology , Brain/growth & development , Larva , Motor Activity/drug effects , Motor Activity/physiology , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/metabolism , Selegiline/pharmacology , Zebrafish/growth & development , Zebrafish/metabolism , Zebrafish Proteins/chemistry
16.
J Neurosci ; 29(5): 1343-9, 2009 Feb 04.
Article in English | MEDLINE | ID: mdl-19193881

ABSTRACT

Huntington's disease (HD) is an autosomal dominant, neurodegenerative condition caused by a CAG trinucleotide repeat expansion that is translated into an abnormally long polyglutamine tract in the protein huntingtin. Genetic and transgenic studies suggest that the mutation causes disease predominantly via gain-of-function mechanisms. However, loss of normal huntingtin function resulting from the polyglutamine expansion might also contribute to the pathogenesis of HD. Here, we have studied the effects of huntingtin knockdown in zebrafish using morpholino antisense oligonucleotides, as its huntingtin orthologue has 70% amino acid identity with the human protein. Reduced huntingtin levels did not impact on gastrulation and early development, but caused massive apoptosis of neuronal cells by 24 hpf. This was accompanied by impaired neuronal development, resulting in small eyes and heads and enlargement of brain ventricles. Older huntingtin knockdown fish developed lower jaw abnormalities with most branchial arches missing. Molecular analysis revealed that BDNF expression was reduced by approximately 50%. Reduction of BDNF levels by injection of a BDNF morpholino resulted in phenotypes very similar to those seen in huntingtin knockdown zebrafish. The phenotypes of both huntingtin- and BDNF-knockdown zebrafish showed significant rescue when treated with exogenous BDNF protein. This underscores the physiological importance of huntingtin as a regulator of BDNF production and suggests that loss of BDNF is a major cause of the developmental abnormalities seen with huntingtin knockdown in zebrafish. Increasing BDNF expression may represent a useful strategy for Huntington's disease treatment.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Gene Knockdown Techniques , Nerve Tissue Proteins/genetics , Phenotype , Zebrafish Proteins/genetics , Zebrafish/embryology , Zebrafish/genetics , Animals , Brain-Derived Neurotrophic Factor/deficiency , Brain-Derived Neurotrophic Factor/genetics , Gene Knockdown Techniques/methods , Humans , Huntington Disease/genetics , Huntington Disease/metabolism , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/physiology , Zebrafish Proteins/deficiency , Zebrafish Proteins/physiology
17.
J Neurosci ; 28(33): 8199-207, 2008 Aug 13.
Article in English | MEDLINE | ID: mdl-18701682

ABSTRACT

Parkinson's disease (PD) is the second most prevalent neurodegenerative disorder in the Western world. PTEN (phosphatase/tensin homolog on chromosome 10)-induced putative kinase 1 (PINK1), a putative kinase that is mutated in autosomal recessive forms of PD, is also implicated in sporadic cases of the disease. Although the mutations appear to result in a loss of function, the roles of this protein and the pathways involved in PINK1 PD are poorly understood. Here, we generated a vertebrate model of PINK1 insufficiency using morpholino oligonucleotide knockdown in zebrafish (Danio rerio). PINK1 knockdown results in a severe developmental phenotype that is rescued by wild-type human PINK1 mRNA. Morphants display a moderate decrease in the numbers of central dopaminergic neurons and alterations of mitochondrial function, including increases in caspase-3 activity and reactive oxygen species (ROS) levels. When the morphants were exposed to several drugs with antioxidant properties, ROS levels were normalized and the associated phenotype improved. In addition, GSK3beta-related mechanisms can account for some of the effects of PINK1 knockdown, as morphant fish show elevated GSK3beta activity and their phenotype is partially abrogated by GSK3beta inhibitors, such as LiCl and SB216763 [3-(2,4-dichlorophenyl)-4-(1-methyl-1H-indol-3-yl)1H-pyrrole-2,5-dione]. This provides new insights into the biology of PINK1 and a possible therapeutic avenue for further investigation.


Subject(s)
Nerve Degeneration/enzymology , Nerve Degeneration/genetics , Protein Kinases/deficiency , Protein Kinases/genetics , Zebrafish Proteins/deficiency , Zebrafish Proteins/genetics , Animals , Axons/enzymology , Axons/pathology , Cell Death/genetics , Gene Expression Regulation, Developmental/genetics , Humans , Nerve Degeneration/embryology , Nerve Degeneration/pathology , Neurons/enzymology , Neurons/pathology , Phenotype , Protein Kinases/physiology , Zebrafish , Zebrafish Proteins/physiology
18.
Biochem Pharmacol ; 73(8): 1205-14, 2007 Apr 15.
Article in English | MEDLINE | ID: mdl-17266939

ABSTRACT

Neuronal histamine regulates several functions in the vertebrate brain. The zebrafish brain contains a widespread histaminergic system and H(3) receptor ligand binding has been reported. In this study we provide evidence for the existence of histamine H(1), H(2) and H(3) receptor genes in zebrafish. Single copies of putative histamine H(1), H(2) and H(3) receptors were identified and cloned from the zebrafish brain. Expression analysis suggested that they are expressed in the brain and a few other tissues. Widespread distribution of zebrafish H(2) receptor binding sites was detected with [(125)I]iodoaminopotentidine in brain sections. Zebrafish larvae were exposed to 1, 10 or 100 microM of the H(1) ligand pyrilamine, the H(2) ligand cimetidine and the H(3) ligands thioperamide and immepip for 5 days. Significant decreases in swimming distance were observed with the highest dose of all ligands, whereas cimetidine gave a significant decrease also with 1 and 10 microM doses. These results provide the first molecular biological evidence for the presence of histamine receptors in zebrafish. These histamine receptors resemble those of higher vertebrates and they provide a useful model for pharmacological and behavioral studies for characterizing the functions of histamine in more detail.


Subject(s)
Behavior, Animal/drug effects , Histamine Agonists/pharmacology , Histamine Antagonists/pharmacology , Receptors, Histamine H1/isolation & purification , Receptors, Histamine H2/isolation & purification , Receptors, Histamine H3/isolation & purification , Animals , Behavior, Animal/physiology , Cimetidine/pharmacology , Imidazoles/pharmacology , Piperidines/pharmacology , Zebrafish
19.
J Comp Neurol ; 498(5): 593-610, 2006 Oct 10.
Article in English | MEDLINE | ID: mdl-16917825

ABSTRACT

Monoamine oxidase (MAO) is a mitochondrial flavoprotein involved in the metabolism of, e.g., aminergic neurotransmitters and the parkinsonism-inducing neurotoxin 1-methyl-4-phenyl-1,2,5,6-tetrahydropyridine (MPTP). We have reported earlier MPTP-related alterations of brain catecholaminergic system in zebrafish (Danio rerio) brain. Here we describe the structural and functional properties of zebrafish MAO and the distribution of MAO mRNA and activity in zebrafish brain. The gene is located in chromosome 9 and consists of 15 exons. The amino acid composition of the active center resembles both human MAO-A and MAO-B. The enzyme displayed the highest substrate specificity for tyramine, followed by serotonin, phenylethylamine, MPTP, and dopamine; isoform-specific antagonists blocked the activity of the enzyme with equal potency. Zebrafish MAO mRNA, which was present in several tissues, and enzyme displayed differential distribution in the brain; dopaminergic cell clusters had low to moderate levels of MAO activity, whereas the highest levels of MAO activity were detected in noradrenergic and serotonergic cell groups and the habenulointerpeduncular pathway, including its caudal projection to the medial ventral rhombencephalon. The results of this study confirm the presence of functionally active MAO in zebrafish brain and other tissues and characterize the neural systems that express MAO and areas of intense activity in the brain. They also suggest that MPTP toxicity not related to MAO may affect the zebrafish brain.


Subject(s)
Brain/anatomy & histology , Brain/enzymology , Monoamine Oxidase/metabolism , Zebrafish/metabolism , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Analysis of Variance , Animals , Biogenic Monoamines/pharmacology , Brain/drug effects , Cloning, Molecular/methods , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Female , Gene Expression Regulation/drug effects , Immunohistochemistry/methods , In Situ Hybridization/methods , Male , Molecular Sequence Data , Monoamine Oxidase/chemistry , Monoamine Oxidase/genetics , Neurotoxins/pharmacology , Protein Conformation , RNA, Messenger/metabolism , Time Factors
20.
J Neurochem ; 88(2): 443-53, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14690532

ABSTRACT

Dopaminergic deficiency in the brain of zebrafish was produced by systemic administration of two catecholaminergic neurotoxins, 6-hydroxydopamine (6-OHDA) and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), and the neurochemical and behavioural changes were characterized. The levels of dopamine and noradrenaline decreased significantly after the injection of MPTP and 6-OHDA. Corresponding to these changes, fish exhibited characteristic changes in locomotor behaviour, i.e. the total distance moved and velocity decreased after both neurotoxins. Tyrosine hydroxylase and caspase 3 protein levels were not altered after MPTP or 6-OHDA injections, as studied by immunohistochemistry and western blotting. The catecholaminergic cell clusters suggested to correspond to the mammalian nigrostriatal cell group displayed normal tyrosine hydroxylase immunoreactivity after the toxin treatment and did not show signs of DNA fragmentation that would indicate activation of cascades that lead to cell death. The results show that single systemic injections of MPTP and 6-OHDA induce both biochemical and behavioural changes in zebrafish, albeit failing to produce any significant morphological alteration in catecholaminergic cell clusters at the tested doses. This approach may be used for the screening of chemicals affecting the dopaminergic system. The model may be especially useful for evaluation of the role of novel genes in neurotoxicity, as a large number of zebrafish mutants are becoming available.


Subject(s)
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Brain/drug effects , Motor Activity/drug effects , Oxidopamine/pharmacology , Zebrafish/metabolism , Animals , Brain/metabolism , Dopamine/metabolism , Female , Injections, Intramuscular , Male , Motor Activity/physiology , Norepinephrine/metabolism , Pilot Projects
SELECTION OF CITATIONS
SEARCH DETAIL