Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters











Publication year range
2.
Antioxidants (Basel) ; 12(6)2023 Jun 15.
Article in English | MEDLINE | ID: mdl-37372013

ABSTRACT

Alzheimer's disease (AD) and Parkinson's disease (PD) are the two most common neurodegenerative diseases in the elderly. The key histopathological features of these diseases are the presence of abnormal protein aggregates and the progressive and irreversible loss of neurons in specific brain regions. The exact mechanisms underlying the etiopathogenesis of AD or PD remain unknown, but there is extensive evidence indicating that excessive generation of reactive oxygen species (ROS) and reactive nitrogen species (RNS), along with a depleted antioxidant system, mitochondrial dysfunction, and intracellular Ca2+ dyshomeostasis, plays a vital role in the pathophysiology of these neurological disorders. Due to an improvement in life expectancy, the incidence of age-related neurodegenerative diseases has significantly increased. However, there is no effective protective treatment or therapy available but rather only very limited palliative treatment. Therefore, there is an urgent need for the development of preventive strategies and disease-modifying therapies to treat AD/PD. Because dysregulated Ca2+ metabolism drives oxidative damage and neuropathology in these diseases, the identification or development of compounds capable of restoring Ca2+ homeostasis and signaling may provide a neuroprotective avenue for the treatment of neurodegenerative diseases. In addition, a set of strategies to control mitochondrial Ca2+ homeostasis and signaling has been reported, including decreased Ca2+ uptake through voltage-operated Ca2+ channels (VOCCs). In this article, we review the modulatory effects of several heterocyclic compounds on Ca2+ homeostasis and trafficking, as well as their ability to regulate compromised mitochondrial function and associated free-radical production during the onset and progression of AD or PD. This comprehensive review also describes the chemical synthesis of the heterocycles and summarizes the clinical trial outcomes.

3.
Exp Neurol ; 357: 114204, 2022 11.
Article in English | MEDLINE | ID: mdl-35973617

ABSTRACT

Traumatic brain injury (TBI) causes persistent cognitive impairment and neurodegeneration. Environmental enrichment (EE) refers to a housing condition that promotes sensory and social stimulation and improves cognition and motor performance but the underlying mechanisms responsible for such beneficial effects are not well defined. In this study, anesthetized adult rats received either a moderate-to-severe controlled cortical impact (CCI) or sham surgery and then were housed in either EE or standard conditions. The results showed a significant increase in protein nitration and oxidation of lipids, impaired cognition and motor performance, and augmented N-methyl-d-aspartate receptor subtype-1 (NMDAR1) levels. However, EE initiated 24 h after CCI resulted in reduced oxidative insult and microglial activation and significant improvement in beam-balance/walk performance and both spatial learning and memory. We hypothesize that following TBI there is an upstream activation of NMDAR that promotes oxidative insult and an inflammatory response, thereby resulting in impaired behavioral functioning but EE may exert a neuroprotective effect via sustained downregulation of NMDAR1.


Subject(s)
Brain Injuries, Traumatic , Psychomotor Performance , Animals , Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/psychology , Brain Injuries, Traumatic/therapy , Disease Models, Animal , Environment , Maze Learning/physiology , Phenotype , Rats , Rats, Sprague-Dawley
4.
Biomedicines ; 10(5)2022 May 17.
Article in English | MEDLINE | ID: mdl-35625890

ABSTRACT

The molecular basis of amyloid toxicity in Alzheimer's disease (AD) remains controversial. Amyloid ß (Aß) oligomers promote Ca2+ influx, mitochondrial Ca2+ overload and apoptosis in hippocampal neurons in vivo and in vitro, but the primary Ca2+ entry pathways are unclear. We studied Ca2+ entry pathways induced by Aß oligomers in rat hippocampal and cerebellar neurons. Aß oligomers induce Ca2+ entry in neurons. Ca2+ responses to Aß oligomers are large after synaptic networking and prevented by blockers of synaptic transmission. In contrast, in neurons devoid of synaptic connections, Ca2+ responses to Aß oligomers are small and prevented only by blockers of amyloid channels (NA7) and NMDA receptors (MK801). A combination of NA7 and MK801 nearly abolished Ca2+ responses. Non-neuronal cells bearing NMDA receptors showed Ca2+ responses to oligomers, whereas cells without NMDA receptors did not exhibit Ca2+ responses. The expression of subunits of the NMDA receptor NR1/ NR2A and NR1/NR2B in HEK293 cells lacking endogenous NMDA receptors restored Ca2+ responses to NMDA but not to Aß oligomers. We conclude that Aß oligomers promote Ca2+ entry via amyloid channels and NMDA receptors. This may recruit distant neurons intertwisted by synaptic connections, spreading excitation and recruiting further NMDA receptors and voltage-gated Ca2+ channels, leading to excitotoxicity and neuron degeneration in AD.

5.
IBRO Neurosci Rep ; 12: 217-227, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35321527

ABSTRACT

Exercise and consumption of plant-based foods rich in polyphenols are attractive therapeutic approaches for the prevention and treatment of Parkinson's disease (PD). Few studies, however, have examined the neuroprotective efficacy of combining these treatment modalities against PD. Therefore we investigated whether combining voluntary running and consumption of blueberry juice (BBJ) was more efficacious against 6-hydroxydopamine (6-OHDA) toxicity than either treatment alone. Four weeks of running before and after intrastriatal 6-OHDA reduced amphetamine-induced rotational behavior and loss of substantia nigra dopamine (DA) neurons. BBJ consumption alone had no ameliorative effects, but when combined with exercise, behavioral deficits and nigrostriatal DA neurodegeneration were reduced to a greater extent than exercise alone. The neuroprotection observed with exercise alone was associated with an increase in striatal glial cell-lined derived neurotrophic factor (GDNF), whereas combining exercise and BBJ was associated with an increase in nigral GDNF. These results suggest that polyphenols may potentiate the protective effects of exercise and that differential regulation of GDNF expression underlies protection observed with exercise alone versus combined treatment with consumption of BBJ.

6.
Nat Commun ; 13(1): 159, 2022 01 10.
Article in English | MEDLINE | ID: mdl-35013160

ABSTRACT

Abnormalities in brain glucose metabolism and accumulation of abnormal protein deposits called plaques and tangles are neuropathological hallmarks of Alzheimer's disease (AD), but their relationship to disease pathogenesis and to each other remains unclear. Here we show that succinylation, a metabolism-associated post-translational protein modification (PTM), provides a potential link between abnormal metabolism and AD pathology. We quantified the lysine succinylomes and proteomes from brains of individuals with AD, and healthy controls. In AD, succinylation of multiple mitochondrial proteins declined, and succinylation of small number of cytosolic proteins increased. The largest increases occurred at critical sites of amyloid precursor protein (APP) and microtubule-associated tau. We show that in vitro, succinylation of APP disrupted its normal proteolytic processing thereby promoting Aß accumulation and plaque formation and that succinylation of tau promoted its aggregation to tangles and impaired microtubule assembly. In transgenic mouse models of AD, elevated succinylation associated with soluble and insoluble APP derivatives and tau. These findings indicate that a metabolism-linked PTM may be associated with AD.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/metabolism , Plaque, Amyloid/metabolism , Protein Processing, Post-Translational , Succinic Acid/metabolism , tau Proteins/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amino Acid Sequence , Amyloid beta-Protein Precursor/genetics , Animals , Autopsy , Brain/metabolism , Brain/pathology , Case-Control Studies , Disease Models, Animal , Gene Expression Profiling , Humans , Mice , Mice, Transgenic , Mitochondria/genetics , Mitochondria/metabolism , Mitochondria/pathology , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Plaque, Amyloid/genetics , Plaque, Amyloid/pathology , Protein Aggregates , Proteolysis , Proteome/genetics , Proteome/metabolism , tau Proteins/genetics
7.
Acta Neuropathol Commun ; 8(1): 220, 2020 12 11.
Article in English | MEDLINE | ID: mdl-33308320

ABSTRACT

Lipid peroxidation is a key to a portfolio of neurodegenerative diseases and plays a central role in α-synuclein (α-syn) toxicity, mitochondrial dysfunction and neuronal death, all key processes in the pathogenesis of Parkinson's disease (PD). Polyunsaturated fatty acids (PUFAs) are important constituents of the synaptic and mitochondrial membranes and are often the first molecular targets attacked by reactive oxygen species (ROS). The rate-limiting step of the chain reaction of ROS-initiated PUFAs autoxidation involves hydrogen abstraction at bis-allylic sites, which can be slowed down if hydrogens are replaced with deuteriums. In this study, we show that targeted overexpression of human A53T α-syn using an AAV vector unilaterally in the rat substantia nigra reproduces some of pathological features seen in PD patients. Chronic dietary supplementation with deuterated PUFAs (D-PUFAs), specifically 0.8% D-linoleic and 0.3% H-linolenic, produced significant disease-modifying beneficial effects against α-syn-induced motor deficits, synaptic pathology, oxidative damage, mitochondrial dysfunction, disrupted trafficking along axons, inflammation and DA neuronal loss. These findings support the clinical evaluation of D-PUFAs as a neuroprotective therapy for PD.


Subject(s)
Brain/drug effects , Dopaminergic Neurons/drug effects , Exploratory Behavior/drug effects , Linoleic Acid/pharmacology , Mitochondria/drug effects , Parkinson Disease/physiopathology , Postural Balance/drug effects , alpha-Linolenic Acid/pharmacology , Animals , Axonal Transport/drug effects , Behavior, Animal/drug effects , Brain/pathology , Deuterium , Humans , Inflammation , Mitochondria/metabolism , Oxidative Stress/drug effects , Parkinson Disease/genetics , Parkinson Disease/pathology , Rats , Rats, Transgenic , Substantia Nigra , alpha-Synuclein/genetics
9.
Redox Biol ; 24: 101164, 2019 06.
Article in English | MEDLINE | ID: mdl-30925294

ABSTRACT

Impaired mitochondrial function has been associated with the etiopathogenesis of Parkinson's disease (PD). Sustained inhibition of complex I produces mitochondrial dysfunction, which is related to oxidative injury and nigrostriatal dopamine (DA) neurodegeneration. This study aimed to identify disease-modifying treatments for PD. Unsubstituted phenothiazine (PTZ) is a small and uncharged aromatic imine that readily crosses the blood-brain barrier. PTZ lacks significant DA receptor-binding activity and, in the nanomolar range, exhibits protective effects via its potent free radical scavenging and anti-inflammatory activities. Given that DAergic neurons are highly vulnerable to oxidative damage and inflammation, we hypothesized that administration of PTZ might confer neuroprotection in different experimental models of PD. Our findings showed that PTZ rescues rotenone (ROT) toxicity in primary ventral midbrain neuronal cultures by preserving neuronal integrity and reducing protein thiol oxidation. Long-term treatment with PTZ improved animal weight, survival rate, and behavioral deficits in ROT-lesioned rats. PTZ protected DA content and fiber density in the striatum and DA neurons in the SN against the deleterious effects of ROT. Mitochondrial dysfunction, axonal impairment, oxidative insult, and inflammatory response were attenuated with PTZ therapy. Furthermore, we have provided a new insight into the molecular mechanism underlying the neuroprotective effects of PTZ.


Subject(s)
Dopamine/metabolism , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , NAD/metabolism , Phenothiazines/pharmacology , Substantia Nigra/drug effects , Substantia Nigra/metabolism , Animals , Biomarkers , Cell Culture Techniques , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Dose-Response Relationship, Drug , Immunohistochemistry , Male , Models, Biological , Neuroprotection/drug effects , Neuroprotective Agents/pharmacology , Parkinson Disease/etiology , Parkinson Disease/metabolism , Rats , Rotenone/adverse effects
10.
Front Neurosci ; 13: 1372, 2019.
Article in English | MEDLINE | ID: mdl-31920522
11.
Hum Mol Genet ; 27(16): 2874-2892, 2018 08 15.
Article in English | MEDLINE | ID: mdl-29860433

ABSTRACT

Impaired glucose metabolism, decreased levels of thiamine and its phosphate esters, and reduced activity of thiamine-dependent enzymes, such as pyruvate dehydrogenase, alpha-ketoglutarate dehydrogenase and transketolase occur in Alzheimer's disease (AD). Thiamine deficiency exacerbates amyloid beta (Aß) deposition, tau hyperphosphorylation and oxidative stress. Benfotiamine (BFT) rescued cognitive deficits and reduced Aß burden in amyloid precursor protein (APP)/PS1 mice. In this study, we examined whether BFT confers neuroprotection against tau phosphorylation and the generation of neurofibrillary tangles (NFTs) in the P301S mouse model of tauopathy. Chronic dietary treatment with BFT increased lifespan, improved behavior, reduced glycated tau, decreased NFTs and prevented death of motor neurons. BFT administration significantly ameliorated mitochondrial dysfunction and attenuated oxidative damage and inflammation. We found that BFT and its metabolites (but not thiamine) trigger the expression of Nrf2/antioxidant response element (ARE)-dependent genes in mouse brain as well as in wild-type but not Nrf2-deficient fibroblasts. Active metabolites were more potent in activating the Nrf2 target genes than the parent molecule BFT. Docking studies showed that BFT and its metabolites (but not thiamine) bind to Keap1 with high affinity. These findings demonstrate that BFT activates the Nrf2/ARE pathway and is a promising therapeutic agent for the treatment of diseases with tau pathology, such as AD, frontotemporal dementia and progressive supranuclear palsy.


Subject(s)
Antioxidant Response Elements/genetics , NF-E2-Related Factor 2/genetics , Protein Aggregation, Pathological/drug therapy , Tauopathies/drug therapy , Thiamine/analogs & derivatives , Amyloid beta-Peptides/genetics , Animals , Brain/drug effects , Brain/metabolism , Brain/pathology , Disease Models, Animal , Humans , Kelch-Like ECH-Associated Protein 1/genetics , Mice , Mice, Transgenic , Neuroprotection/drug effects , Oxidative Stress/drug effects , Protein Aggregation, Pathological/genetics , Protein Aggregation, Pathological/pathology , Signal Transduction/drug effects , Tauopathies/genetics , Tauopathies/physiopathology , Thiamine/administration & dosage , tau Proteins/genetics
12.
Antioxid Redox Signal ; 28(1): 44-61, 2018 Jan 01.
Article in English | MEDLINE | ID: mdl-28816057

ABSTRACT

AIMS: This study was designed to explore the neuroprotective potential of inorganic nitrite as a new therapeutic avenue in Parkinson's disease (PD). RESULTS: Administration of inorganic nitrite ameliorates neuropathology in phylogenetically distinct animal models of PD. Beneficial effects are not confined to prophylactic treatment and also occur if nitrite is administered when the pathogenic cascade is already active. Mechanistically, the effect is mediated by both complex I S-nitrosation, which under nitrite administration is favored over formation of other forms of oxidation, and down-stream activation of the antioxidant Nrf2 pathway. Nitrite also rescues respiratory reserve capacity and increases proton leakage in LRRK2 PD patients' dermal fibroblasts. INNOVATION: The study proposes an unprecedented approach based on the administration of the nitrosonium donor nitrite to contrast complex I and redox anomalies in PD. Dysfunctional mitochondrial complex I propagates oxidative stress in PD, and treatments mitigating this defect may, therefore, limit disease progression. Therapeutic complex I targeting has been successfully achieved in ischemia/reperfusion by using nitrosonium donors such as nitrite to reversibly modify its subunits and protect from oxidative damage after reperfusion. This evidence led to the innovative hypothesis that nitrite could exert protective effects also in pathological conditions where complex I dysfunction occurs in normoxia, such as in PD. CONCLUSIONS: Overall, these results demonstrate that administration of inorganic nitrite improves mitochondrial function in PD, and it, therefore, represents an amenable intervention to hamper disease progression. Antioxid. Redox Signal. 28, 44-61.


Subject(s)
Electron Transport Complex I/metabolism , Energy Metabolism , Mitochondria/metabolism , Parkinson Disease/metabolism , Animals , Antioxidants/metabolism , Behavior, Animal , Cell Respiration/drug effects , Cell Survival , Cytoprotection , Disease Models, Animal , Energy Metabolism/drug effects , Fibroblasts , Humans , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Male , Mitochondria/drug effects , Motor Activity , Mutation , Neurons/metabolism , Nitrites/administration & dosage , Nitrites/metabolism , Oxidation-Reduction , Oxidative Stress , Parkinson Disease/genetics , Parkinson Disease/pathology , Protective Agents/administration & dosage , Rats , Tyrosine 3-Monooxygenase/metabolism
13.
J Nutr Biochem ; 54: 77-86, 2018 04.
Article in English | MEDLINE | ID: mdl-29268122

ABSTRACT

Age-related declines in motor function may be due, in part, to an increase in oxidative stress in the aging brain leading to dopamine (DA) neuronal cell death. In this study, we examined the neuroprotective effects of natural antioxidants resveratrol and pinostilbene against age-related DAergic cell death and motor dysfunction using SH-SY5Y neuroblastoma cells and young, middle-aged, and old male C57BL/6 mice. Resveratrol and pinostilbene protected SH-SY5Y cells from a DA-induced decrease in cell viability. Dietary supplementation with resveratrol and pinostilbene inhibited the decline of motor function observed with age. While DA and its metabolites (DOPAC and HVA), dopamine transporter, and tyrosine hydroxylase levels remain unchanged during aging or treatment, resveratrol and pinostilbene increased ERK1/2 activation in vitro and in vivo in an age-dependent manner. Inhibition of ERK1/2 in SH-SY5Y cells decreased the protective effects of both compounds. These data suggest that resveratrol and pinostilbene alleviate age-related motor decline via the promotion of DA neuronal survival and activation of the ERK1/2 pathways.


Subject(s)
Aging/drug effects , Neurons/drug effects , Neuroprotective Agents/pharmacology , Resveratrol/pharmacology , Stilbenes/pharmacology , Aging/physiology , Animals , Cell Line , Dopamine/metabolism , Dopamine/toxicity , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Humans , Male , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Motor Activity/drug effects , Neurons/metabolism , Substantia Nigra/drug effects , Substantia Nigra/metabolism
14.
Cell Mol Life Sci ; 74(15): 2851-2874, 2017 08.
Article in English | MEDLINE | ID: mdl-28534083

ABSTRACT

Intracellular accumulation of α-synuclein (α-syn) are hallmarks of synucleinopathies, including Parkinson's disease (PD). Exogenous addition of preformed α-syn fibrils (PFFs) into primary hippocampal neurons induced α-syn aggregation and accumulation. Likewise, intrastriatal inoculation of PFFs into mice and non-human primates generates Lewy bodies and Lewy neurites associated with PD-like neurodegeneration. Herein, we investigate the putative effects of synthetic human PFFs on cultured rat ventral midbrain dopamine (DA) neurons. A time- and dose-dependent accumulation of α-syn was observed following PFFs exposure that also underwent phosphorylation at serine 129. PFFs treatment decreased the expression levels of synaptic proteins, caused alterations in axonal transport-related proteins, and increased H2AX Ser139 phosphorylation. Mitochondrial impairment (including modulation of mitochondrial dynamics-associated protein content), enhanced oxidative stress, and an inflammatory response were also detected in our experimental paradigm. In attempt to unravel a potential molecular mechanism of PFFs neurotoxicity, the expression of inducible nitric oxide synthase was blocked; a significant decline in protein nitration levels and protection against PFFs-induced DA neuron death were observed. Combined exposure to PFFs and rotenone resulted in an additive toxicity. Strikingly, many of the harmful effects found were more prominent in DA rather than non-DA neurons, suggestive of higher susceptibility to degenerate. These findings provide new insights into the role of α-syn in the pathogenesis of PD and could represent a novel and valuable model to study DA-related neurodegeneration.


Subject(s)
Dopaminergic Neurons/pathology , Mitochondria/pathology , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide/metabolism , Protein Aggregation, Pathological/pathology , alpha-Synuclein/metabolism , Animals , Cell Survival , Cells, Cultured , Dopaminergic Neurons/cytology , Dopaminergic Neurons/metabolism , Humans , Inflammation/metabolism , Inflammation/pathology , Mesencephalon/metabolism , Mesencephalon/pathology , Mitochondria/metabolism , Oxidative Stress , Parkinson Disease/metabolism , Parkinson Disease/pathology , Protein Aggregation, Pathological/metabolism , Rats, Sprague-Dawley , alpha-Synuclein/analysis
15.
J Alzheimers Dis ; 56(3): 1075-1085, 2017.
Article in English | MEDLINE | ID: mdl-28106559

ABSTRACT

ATP-binding cassette transporter A1 (ABCA1) mediates cholesterol efflux to lipid-free apolipoproteins and regulates the generation of high density lipoproteins. Previously, we have shown that lack of Abca1 significantly increases amyloid deposition and cognitive deficits in Alzheimer's disease model mice expressing human amyloid-ß protein precursor (APP). The goal of this study was to determine if ABCA1 plays a role in memory deficits caused by amyloid-ß (Aß) oligomers and examine neurite architecture of pyramidal hippocampal neurons. Our results confirm previous findings that Abca1 deficiency significantly impairs spatial memory acquisition and retention in the Morris water maze and long-term memory in novel object recognition of APP transgenic mice at a stage of early amyloid pathology. Neither test demonstrated a significant difference between Abca1ko and wild-type (WT) mice. We also examined the effect of intra-hippocampal infused Aß oligomers on cognitive performance of Abca1ko mice, compared to control infusion of scrambled Aß peptide. Age-matched WT mice undergoing the same infusions were also used as controls. In this model system, we found a statistically significant difference between WT and Abca1ko mice infused with scrambled Aß, suggesting that Abca1ko mice are vulnerable to the effect of mild stresses. Moreover, examination of neurite architecture in the hippocampi revealed a significant decrease in neurite length, number of neurite segments, and branches in Abca1ko mice when compared to WT mice. We conclude that mice lacking ABCA1 have basal cognitive deficits that prevent them from coping with additional stressors, which is in part due to impairment of neurite morphology in the hippocampus.


Subject(s)
ATP Binding Cassette Transporter 1/deficiency , Cognition Disorders/metabolism , Cognition Disorders/pathology , Dendrites/metabolism , Dendrites/pathology , ATP Binding Cassette Transporter 1/genetics , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Cell Size , Female , Hippocampus/metabolism , Hippocampus/pathology , Humans , Male , Maze Learning/physiology , Mice, Inbred C57BL , Mice, Transgenic , Presenilin-1/genetics , Presenilin-1/metabolism , Recognition, Psychology/physiology , Spatial Memory/physiology
16.
PLoS One ; 11(5): e0156095, 2016.
Article in English | MEDLINE | ID: mdl-27227315

ABSTRACT

Nerve endings are often identified within solid tumors, but their impact on the tumor growth and progression remains poorly understood. Emerging data suggests that the central nervous system may affect cancer development and spreading via the hypothalamic-pituitary-adrenal axis and autonomous nervous system. However, the role of the afferent sensory neurons in tumor growth is unclear, except some reports on perineural invasion in prostate and pancreatic cancer and cancer-related pain syndrome. Here, we provide the results of primary testing of the concept that the interaction between melanoma cells and sensory neurons may induce the formation of tumor-supporting microenvironment via attraction of immune regulatory cells by the tumor-activated dorsal root ganglion (DRG) neurons. We report that despite DRG cells not directly up-regulating proliferation of melanoma cells in vitro, presence of DRG neurons allows tumors to grow significantly faster in vivo. This effect has been associated with increased production of chemokines by tumor-activated DRG neurons and attraction of myeloid-derived suppressor cells both in vitro and in vivo. These initial proof-of-concept results justify further investigations of the sensory (afferent) nervous system in the context of tumorigenesis and the local protumorigenic immunoenvironment.


Subject(s)
Apoptosis , Ganglia, Spinal/pathology , Melanoma/pathology , Sensory Receptor Cells/pathology , Animals , Blotting, Western , Cell Cycle , Cell Movement , Cell Proliferation , Flow Cytometry , Ganglia, Spinal/metabolism , Melanoma/metabolism , Mice , Mice, Inbred C57BL , Mice, Nude , Sensory Receptor Cells/metabolism , Tumor Cells, Cultured
17.
Brain ; 138(Pt 12): 3699-715, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26510953

ABSTRACT

UNLABELLED: ATP binding cassette transporter A1 (encoded by ABCA1) regulates cholesterol efflux from cells to apolipoproteins A-I and E (ApoA-I and APOE; encoded by APOA1 and APOE, respectively) and the generation of high density lipoproteins. In Abca1 knockout mice (Abca1(ko)), high density lipoproteins and ApoA-I are virtually lacking, and total APOE and APOE-containing lipoproteins in brain substantially decreased. As the ε4 allele of APOE is the major genetic risk factor for late-onset Alzheimer's disease, ABCA1 role as a modifier of APOE lipidation is of significance for this disease. Reportedly, Abca1 deficiency in mice expressing human APP accelerates amyloid deposition and behaviour deficits. We used APP/PS1dE9 mice crossed to Apoe and Apoa1 knockout mice to generate Apoe/Apoa1 double-knockout mice. We hypothesized that Apoe/Apoa1 double-knockout mice would mimic the phenotype of APP/Abca1(ko) mice in regards to amyloid plaques and cognitive deficits. Amyloid pathology, peripheral lipoprotein metabolism, cognitive deficits and dendritic morphology of Apoe/Apoa1 double-knockout mice were compared to APP/Abca1(ko), APP/PS1dE9, and single Apoa1 and Apoe knockouts. Contrary to our prediction, the results demonstrate that double deletion of Apoe and Apoa1 ameliorated the amyloid pathology, including amyloid plaques and soluble amyloid. In double knockout mice we show that (125)I-amyloid-ß microinjected into the central nervous system cleared at a rate twice faster compared to Abca1 knockout mice. We tested the effect of Apoe, Apoa1 or Abca1 deficiency on spreading of exogenous amyloid-ß seeds injected into the brain of young pre-depositing APP mice. The results show that lack of Abca1 augments dissemination of exogenous amyloid significantly more than the lack of Apoe. In the periphery, Apoe/Apoa1 double-knockout mice exhibited substantial atherosclerosis and very high levels of low density lipoproteins compared to APP/PS1dE9 and APP/Abca1(ko). Plasma level of amyloid-ß42 measured at several time points for each mouse was significantly higher in Apoe/Apoa1 double-knockout then in APP/Abca1(ko) mice. This result demonstrates that mice with the lowest level of plasma lipoproteins, APP/Abca1(ko), have the lowest level of peripheral amyloid-ß. Unexpectedly, and independent of amyloid pathology, the deletion of both apolipoproteins worsened behaviour deficits of double knockout mice and their performance was undistinguishable from those of Abca1 knockout mice. Finally we observed that the dendritic complexity in the CA1 region of hippocampus but not in CA2 is significantly impaired by Apoe/Apoa1 double deletion as well as by lack of ABCA1. IN CONCLUSION: (i) plasma lipoproteins may affect amyloid-ß clearance from the brain by the 'peripheral sink' mechanism; and (ii) deficiency of brain APOE-containing lipoproteins is of significance for dendritic complexity and cognition.


Subject(s)
Amyloid beta-Protein Precursor/genetics , Apolipoprotein A-I/deficiency , Apolipoproteins E/deficiency , Cognition Disorders/genetics , Cognition Disorders/psychology , Gene Deletion , Plaque, Amyloid/genetics , ATP Binding Cassette Transporter 1/genetics , Amyloid beta-Peptides/administration & dosage , Amyloid beta-Peptides/blood , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/pharmacokinetics , Amyloid beta-Protein Precursor/metabolism , Animals , Apolipoprotein A-I/genetics , Apolipoproteins E/genetics , Brain/metabolism , Brain/pathology , Cognition Disorders/pathology , Female , Hippocampus/metabolism , Lipoproteins/blood , Male , Mice , Mice, Knockout , Microinjections , Neurites/pathology , Peptide Fragments/administration & dosage , Peptide Fragments/blood , Peptide Fragments/metabolism , Peptide Fragments/pharmacokinetics , Plaque, Amyloid/pathology , Plaque, Amyloid/psychology
18.
J Clin Invest ; 125(7): 2721-35, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-26075822

ABSTRACT

Multiple convergent lines of evidence implicate both α-synuclein (encoded by SCNA) and mitochondrial dysfunction in the pathogenesis of sporadic Parkinson's disease (PD). Occupational exposure to the mitochondrial complex I inhibitor rotenone increases PD risk; rotenone-exposed rats show systemic mitochondrial defects but develop specific neuropathology, including α-synuclein aggregation and degeneration of substantia nigra dopaminergic neurons. Here, we inhibited expression of endogenous α-synuclein in the adult rat substantia nigra by adeno-associated virus-mediated delivery of a short hairpin RNA (shRNA) targeting the endogenous rat Snca transcript. Knockdown of α-synuclein by ~35% did not affect motor function or cause degeneration of nigral dopaminergic neurons in control rats. However, in rotenone-exposed rats, progressive motor deficits were substantially attenuated contralateral to α-synuclein knockdown. Correspondingly, rotenone-induced degeneration of nigral dopaminergic neurons, their dendrites, and their striatal terminals was decreased ipsilateral to α-synuclein knockdown. These data show that α-synuclein knockdown is neuroprotective in the rotenone model of PD and indicate that endogenous α-synuclein contributes to the specific vulnerability of dopaminergic neurons to systemic mitochondrial inhibition. Our findings are consistent with a model in which genetic variants influencing α-synuclein expression modulate cellular susceptibility to environmental exposures in PD patients. shRNA targeting the SNCA transcript should be further evaluated as a possible neuroprotective therapy in PD.


Subject(s)
Nerve Degeneration/prevention & control , Parkinsonian Disorders/therapy , RNA, Small Interfering/genetics , RNA, Small Interfering/therapeutic use , alpha-Synuclein/antagonists & inhibitors , alpha-Synuclein/genetics , Animals , Disease Models, Animal , Dopaminergic Neurons/pathology , Dopaminergic Neurons/physiology , Gene Knockdown Techniques , Male , Nerve Degeneration/pathology , Nerve Degeneration/physiopathology , Parkinsonian Disorders/pathology , Parkinsonian Disorders/physiopathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Inbred Lew , Rats, Transgenic , Rotenone/toxicity , Substantia Nigra/pathology , Substantia Nigra/physiopathology , alpha-Synuclein/physiology
19.
Neurobiol Aging ; 36(1): 505-18, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25174649

ABSTRACT

Mutations in leucine-rich repeated kinase 2 (LRRK2) cause autosomal dominant late-onset Parkinson's disease (PD), and the G2019S mutation in the kinase domain of LRRK2 is the most common genetic cause of familial PD. Enhanced kinase activity of G2019S LRRK2 is a suspected mechanism for carriers to develop PD but pathophysiological function of G2019S LRRK2 is not clear. The objective of the present study was to characterize a bacterial artificial chromosome rat expressing human G2019S LRRK2. Immunoblotting analysis showed that G2019S LRRK2 expression was approximately 5-8 times higher than wild-type rat LRRK2. At ages of 4, 8, and 12 months, our characterization showed that expression of G2019S LRRK2 induced oxidative stress in striatum and substantia nigra, increased inducible nitric oxide synthase expression in nigral dopamine neurons, and abnormal morphology of nigral dopaminergic neurons in transgenic rats compared with wild-type, without inducing overt neurodegeneration in nigrostriatal dopaminergic neurons. Thus, we conclude that although this model does not reproduce the key features of end-stage PD, important preclinical features of the disease are evident, which may be useful in studying the earliest stages of PD and for gene-environment interaction studies.


Subject(s)
Chromosomes, Artificial, Bacterial/genetics , Mutation , Parkinsonian Disorders/genetics , Protein Serine-Threonine Kinases/genetics , Animals , Disease Models, Animal , Dopaminergic Neurons/metabolism , Gene-Environment Interaction , Humans , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Nitric Oxide Synthase Type II/metabolism , Oxidative Stress/genetics , Rats, Sprague-Dawley , Rats, Transgenic
20.
Curr Protoc Cytom ; 68: 12.33.1-12.33.22, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24692056

ABSTRACT

Stereological methods for tissue cell counting, specifically for neuron quantification, decrease systematic error and sampling bias; however, they are tedious, labor intensive, and time consuming. Approaches for cell (neuron) quantification in vitro are not accurate, sensitive, or subsequently reproducible. Neuronal phenotype is related to alterations in cell morphology and neurite pattern. The techniques currently available for quantification of these features present several limitations. In this unit, we provide validated automated procedures for in vivo and in vitro quantification of cell number, morphological cell changes, and neurite morphometry in a fast, simple, and reliable manner. Our method counts up to 8 times as many neurons in less than 5% to 10% of the time required for stereological analysis (optical fractionator). In summary, this technology offers an unparalleled opportunity to examine features of cells at high resolution in a complex three-dimensional environment. These techniques provide an exceptional in vivo and in vitro system for neurotoxicity studies, disease modeling, and drug discovery.


Subject(s)
Imaging, Three-Dimensional , Neurites , Animals , Cell Count/instrumentation , Cell Count/methods , Humans
SELECTION OF CITATIONS
SEARCH DETAIL