Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters










Publication year range
1.
Toxicol Rep ; 9: 806-813, 2022.
Article in English | MEDLINE | ID: mdl-36518412

ABSTRACT

The pathophysiology underlying the loss of dopaminergic neurons in Parkinson's disease (PD) is unclear. A gap of knowledge in the molecular and cellular events leading to degeneration of the nigrostriatal DA system is a major barrier to the development of effective therapies for PD. 1-methyl-4-phenylpyridinium (MPP+) is used as a reliable in vitro model of PD in dopaminergic neurons; however, the molecular mechanisms that lead to cell death with this model are not fully understood. Additionally, there is a lack of translational in vitro models to fully understand progressive dopaminergic neurotoxicity. Here, we propose cultures of primary human dopaminergic neuronal precursor cells (HDNPCs) as a model to study progressive dopaminergic toxicity and neuronal damage in PD. We evaluated the concentration-response of MPP+ (0-10 mM) at 24 h, using cell viability and mitochondrial activity assays (LDH, XTT, Live/Dead staining, and MitoTracker). Based on concentration-response data, we chose two concentrations (1.0 and 2.5 mM) of MPP+ to evaluate markers of autophagy and dopaminergic status [tyrosine hydroxylase (TH)] after a 24-h exposure. Exposure to MPP+ induced cytotoxicity, reduced cell viability, and decreased mitochondrial activity. MPP+ at 1.0 and 2.5 mM also induced expression of lysosome-associated membrane protein 1 (LAMP-1) and increased the ratio of light chain 3 (LC3), LC3BII/LC3BI. The expression of TH also decreased. Furthermore, α-synuclein (α-SYN) and parkin were evaluated by immunofluorescence (IF) at 1.0 and 2.5 mM MPP+ after 24 h. A qualitative analysis revealed decreased parkin expression while α-SYN aggregation was observed in the cytoplasm and the nucleus. These data suggest that in HDNPCs MPP+ can cause cytotoxicity and neuronal damage. This damage may be mediated by autophagy, dopamine synthesis, and protein aggregation. The combination of HDNPCs and MPP+ may serve as valuable in vitro model of progressive dopaminergic neurotoxicity for research into potential treatments for PD.

2.
Metab Brain Dis ; 34(5): 1365-1374, 2019 10.
Article in English | MEDLINE | ID: mdl-31267346

ABSTRACT

The amyloid ß-peptide (Aß) is transported across the blood-brain barrier (BBB) by binding with the receptor for advanced glycation end products (RAGE). Previously, we demonstrated that the Aß fraction 25-35 (Aß25-35) increases RAGE expression in the rat hippocampus, likely contributing to its neurotoxic effects. However, it is still debated if the interaction of Aß with RAGE compromises the BBB function in Alzheimer' disease (AD). Here, we evaluated the effects of Aß25-35 in an established in vitro model of the BBB. Rat brain microvascular endothelial cells (rBMVECs) were treated with 20 µM active Aß25-35 or the inactive Aß35-25 (control), for 24 h. Exposure to Aß25-35 significantly decreased cell viability, increased cellular necrosis, and increased the production of reactive oxygen species (ROS), which triggered a decrease in the enzyme glutathione peroxidase when compared to the control condition. Aß25-35 also increased BBB permeability by altering the expression of tight junction proteins (decreasing zonula occludens-1 and increasing occludin). Aß25-35 induced monolayer disruption and cellular disarrangement of the BBB, with RAGE being highly expressed in the zones of disarrangement. Together, these data suggest that Aß25-35-induces toxicity by compromising the functionality and integrity of the BBB in vitro. Graphical abstract Aß25-35 induces BBB dysfunction in vitro, wich is likely mediated by OS and ultimately leads to disruption of BBB integrity and cell death.


Subject(s)
Amyloid beta-Peptides/pharmacology , Blood-Brain Barrier/drug effects , Endothelial Cells/drug effects , Oxidative Stress/drug effects , Peptide Fragments/pharmacology , Animals , Blood-Brain Barrier/metabolism , Cell Survival/drug effects , Endothelial Cells/metabolism , Glutathione Peroxidase/metabolism , Male , Mice , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Receptor for Advanced Glycation End Products/metabolism , Tight Junction Proteins/metabolism
3.
J Appl Toxicol ; 39(7): 966-973, 2019 07.
Article in English | MEDLINE | ID: mdl-30784107

ABSTRACT

Graphene-based nanomaterials hold the potential to be used in a wide variety of applications, including biomedical devices. Pristine graphene (PG) is an un-functionalized, defect-free type of graphene that could be used as a material for neural interfacing. However, the neurotoxic effects of PG, particularly to the blood-brain barrier (BBB), have not been fully studied. The BBB separates the brain tissue from the circulating substances in the blood and is essential to maintain the brain homeostasis. The principal components of the BBB are brain microvascular endothelial cells (BMVECs), which maintain a protectively low permeability due to the expression of tight junction proteins. Here we analyzed the effects of PG on BMVECs in an in vitro model of the BBB. BMVECs were treated with PG at 0, 10, 50 and 100 µg/mL for 24 hours and viability and functional analyses of BBB integrity were performed. PG increased lactate dehydrogenase release at 50 and 100 µg/mL, suggesting the induction of necrosis. Surprisingly, 2,3,-bis(2-methoxy-4-nitro-5-sulfophenyl)-5-[(phenylamino)-carbonyl]-2H-tetrazolium (XTT) conversion was increased at 10 and 50 µg/mL. In contrast, XTT conversion was decreased at 100 µg/mL, suggesting the induction of cell death. In addition, 100 µg/mL PG increased DNA fragmentation, suggesting induction of apoptosis. At the same time, 50 and 100 µg/mL of PG increased the endothelial permeability, which corresponded with a decrease in the expression of the tight junction protein occludin at 100 µg/mL. In conclusion, these results suggest that PG negatively affects the viability and function of the BBB endothelial cells in vitro.


Subject(s)
Apoptosis/drug effects , Blood-Brain Barrier/drug effects , Endothelial Cells/drug effects , Graphite/toxicity , Microvessels/drug effects , Animals , Apoptosis/genetics , Blood-Brain Barrier/enzymology , Blood-Brain Barrier/pathology , Brain/blood supply , Capillary Permeability/drug effects , Cell Survival/drug effects , Cell Survival/genetics , DNA Fragmentation/drug effects , Dose-Response Relationship, Drug , Endothelial Cells/enzymology , Endothelial Cells/pathology , Graphite/pharmacokinetics , L-Lactate Dehydrogenase/metabolism , Microvessels/enzymology , Microvessels/pathology , Rats
4.
Exp Biol Med (Maywood) ; 243(13): 1037-1045, 2018 09.
Article in English | MEDLINE | ID: mdl-30253665

ABSTRACT

IMPACT STATEMENT: Attrition in drug discovery and development remains a major challenge. Safety/toxicity is the most prevalent reason for failure with cardiovascular and CNS toxicities predominating. Non-invasive biomarkers of neurotoxicity would provide significant advantage by allowing earlier prediction of likely neurotoxicity in preclinical studies as well as facilitating clinical trials of new therapies for neurodegenerative conditions such as Parkinson's disease (PD) and multiple sclerosis (MS).


Subject(s)
Biomarkers/analysis , Brain Injuries/drug therapy , Drug Discovery , Neurodegenerative Diseases/drug therapy , Neurotoxicity Syndromes/drug therapy , Animals , Disease Models, Animal , Humans
5.
Neurosci Lett ; 672: 123-129, 2018 04 13.
Article in English | MEDLINE | ID: mdl-29458086

ABSTRACT

Traumatic brain injury (TBI) occurs when external mechanical forces induce brain damage as result of impact, penetration or rapid acceleration/deceleration that causes deformation of brain tissue. Depending on its severity, TBI can be classified as mild, moderate or severe and can lead to blood-brain barrier (BBB) dysfunction. In the present study, we evaluated the effects of uniaxial high-speed stretch (HSS) at 0, 5, 10 and 15% on a pure culture of primary rat brain endothelial cells as an in vitro model of TBI to the BBB. LDH release, viability and apoptosis analysis, expression of tight junction proteins and endothelial permeability were evaluated 24 h after a single stretch episode. HSS slightly increased cell death and apoptosis at 10 and 15%, while LDH release was increased only at 15% stretch. Occludin expression was increased at 10% stretch, while claudin-5 expression was increased at 5% stretch, which also decreased the endothelial permeability. In summary, 15% HSS induced low levels of cell death, consistent with mild TBI and very low percentages of HSS (5%) enhanced the BBB properties, promoting the formation of a stronger barrier. These data support the use of 15% HSS as valuable tool in the study of mild TBI to the BBB in vitro.


Subject(s)
Blood-Brain Barrier/metabolism , Brain Concussion/metabolism , Endothelial Cells/metabolism , Animals , Biological Transport , Cells, Cultured , Claudin-5/metabolism , Occludin/metabolism , Permeability , Rats , Tight Junction Proteins/metabolism , Tight Junctions/metabolism
6.
Mol Neurobiol ; 55(1): 258-266, 2018 01.
Article in English | MEDLINE | ID: mdl-28842857

ABSTRACT

Traumatic brain injury (TBI) is one of the major causes of disability in the USA. It occurs when external mechanical forces induce brain damage that causes deformation of brain tissue. TBI is also associated with alterations of the blood-brain barrier (BBB). Using primary rat brain microvascular endothelial cells as an in vitro BBB model, the effects of biaxial stretch were characterized at 5, 10, 15, 25, and 50% deformation using a commercially available system. The results were compared to the effects of mild and moderate TBI in vivo, induced by the weight-drop method in mice. In vitro, live/dead cells, lactate dehydrogenase (LDH) release, caspase 3/7 staining, and tight junction (TJ) protein expression were evaluated 24 h after a single stretch episode. In vivo, Evans blue extravasation, serum levels of S100ß, and TJ protein expression were evaluated. Stretch induced a deformation-dependent increase in LDH release, cell death, and activation of caspase 3/7, suggesting the induction of apoptosis. Interestingly, low magnitudes of deformation increased the expression of TJ proteins, likely in an attempt to compensate for stretch damage. High magnitudes of deformation decreased the expression of TJ proteins, suggesting that the damage was too severe to counteract. In vivo, mild TBI did not affect BBB permeability or the expression of TJ proteins. However, moderate TBI significantly increased BBB permeability and decreased the expression of these proteins, similar to the results obtained with a high magnitude deformation. These data support the use biaxial stretch as valuable tool in the study of TBI in vitro.


Subject(s)
Blood-Brain Barrier/metabolism , Brain Injuries, Traumatic/metabolism , Capillary Permeability/physiology , Disease Models, Animal , Endothelium, Vascular/metabolism , Tight Junctions/metabolism , Animals , Blood-Brain Barrier/pathology , Brain Injuries, Traumatic/pathology , Endothelium, Vascular/pathology , Rats , Rats, Sprague-Dawley , Tight Junctions/pathology
7.
Exp Biol Med (Maywood) ; 243(3): 228-236, 2018 02.
Article in English | MEDLINE | ID: mdl-29105512

ABSTRACT

Neurotoxicity has been linked with exposure to a number of common drugs and chemicals, yet efficient, accurate, and minimally invasive methods to detect it are lacking. Fluid-based biomarkers such as those found in serum, plasma, urine, and cerebrospinal fluid have great potential due to the relative ease of sampling but at present, data on their expression and translation are lacking or inconsistent. In this pilot study using a trimethyl tin rat model of central nervous system toxicity, we have applied state-of-the-art assessment techniques to identify potential individual biomarkers and patterns of biomarkers in serum, plasma, urine or cerebral spinal fluid that may be indicative of nerve cell damage and degeneration. Overall changes in metabolites and microRNAs were observed in biological fluids that were associated with neurotoxic damage induced by trimethyl tin. Behavioral changes and magnetic resonance imaging T2 relaxation and ventricle volume changes served to identify animals that responded to the adverse effects of trimethyl tin. Impact statement These data will help design follow-on studies with other known neurotoxicants to be used to assess the broad applicability of the present findings. Together this approach represents an effort to begin to develop and qualify a set of translational biochemical markers of neurotoxicity that will be readily accessible in humans. Such biomarkers could prove invaluable for drug development research ranging from preclinical studies to clinical trials and may prove to assist with monitoring of the severity and life cycle of brain lesions.


Subject(s)
Biomarkers , Body Fluids/chemistry , Central Nervous System/pathology , MicroRNAs/analysis , Neurons/pathology , Neurotoxicity Syndromes/diagnosis , Trimethyltin Compounds/toxicity , Amino Acids/analysis , Animals , Behavior, Animal/drug effects , Biomarkers/blood , Biomarkers/cerebrospinal fluid , Biomarkers/urine , Humans , Magnetic Resonance Imaging , Male , Metabolome/physiology , MicroRNAs/genetics , Pilot Projects , Rats , Rats, Sprague-Dawley
8.
Neurosci Lett ; 655: 76-81, 2017 Aug 10.
Article in English | MEDLINE | ID: mdl-28684237

ABSTRACT

Bath salts, or synthetic cathinones, have cocaine-like or amphetamine-like properties and induce psychoactive effects via their capacity to modulate serotonin (5-HT) and dopamine (DA). Structurally distinct synthetic cathinones are continuously being generated to skirt existing drug laws. One example of these modified compounds is cathinone phthalimide (CP), which has already appeared on the global market. The lack of toxicological studies on the effects of CP on monoaminergic systems led to the development of the present study in order to generate an acute toxicity profile for CP, and to clarify whether it primarily affects both dopamine and serotonin, like the synthetic cathinones mephedrone and methylone, or primarily affects dopamine, like 3, 4-methylenedioxypyrovalerone (MDPV). For the first time, the toxicity profile of CP (10µM-1000µM) is reported. In pheochromocytoma cells, exposure to CP induced cell death, and altered mitochondrial function, as well as intracellular DA and 5-HT levels; at the same time, reduced glutathione (GSH) levels remained unaffected. This seems to indicate that CP functions like mephedrone or methylone. The role of CP metabolites, the effect of CP induced hyperthermia on neurotoxicity, and its ability to traverse the blood-brain barrier warrant further consideration.


Subject(s)
Central Nervous System Stimulants/toxicity , Dopamine/metabolism , Phthalimides/toxicity , Propiophenones/toxicity , Serotonin/metabolism , Animals , Cell Death/drug effects , Glutathione/metabolism , Mitochondria/drug effects , Mitochondria/physiology , PC12 Cells , Rats
9.
Int J Mol Sci ; 17(9)2016 Sep 20.
Article in English | MEDLINE | ID: mdl-27657053

ABSTRACT

Currently, the lack of new drug candidates for the treatment of major neurological disorders such as Parkinson's disease has intensified the search for drugs that can be repurposed or repositioned for such treatment. Typically, the search focuses on drugs that have been approved and are used clinically for other indications. Kinase inhibitors represent a family of popular molecules for the treatment and prevention of various cancers, and have emerged as strong candidates for such repurposing because numerous serine/threonine and tyrosine kinases have been implicated in the pathobiology of Parkinson's disease. This review focuses on various kinase-dependent pathways associated with the expression of Parkinson's disease pathology, and evaluates how inhibitors of these pathways might play a major role as effective therapeutic molecules.

10.
Mol Neurobiol ; 52(2): 913-26, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26099304

ABSTRACT

Various iron-oxide nanoparticles have been in use for a long time as therapeutic and imaging agents and for supplemental delivery in cases of iron-deficiency. While all of these products have a specified size range of ∼ 40 nm and above, efforts are underway to produce smaller particles, down to ∼ 1 nm. Here, we show that after a 24-h exposure of SHSY-5Y human neuroblastoma cells to 10 µg/ml of 10 and 30 nm ferric oxide nanoparticles (Fe-NPs), cellular dopamine content was depleted by 68 and 52 %, respectively. Increases in activated tyrosine kinase c-Abl, a molecular switch induced by oxidative stress, and neuronal α-synuclein expression, a protein marker associated with neuronal injury, were also observed (55 and 38 % percent increases, respectively). Inhibition of cell-proliferation, significant reductions in the number of active mitochondria, and a dose-dependent increase in reactive oxygen species (ROS) were observed in neuronal cells. Additionally, using a rat in vitro blood-brain barrier (BBB) model, a dose-dependent increase in ROS accompanied by increased fluorescein efflux demonstrated compromised BBB integrity. To assess translational implications, in vivo Fe-NP-induced neurotoxicity was determined using in vivo MRI and post-mortem neurochemical and neuropathological correlates in adult male rats after exposure to 50 mg/kg of 10 nm Fe-NPs. Significant decrease in T 2 values was observed. Dynamic observations suggested transfer and retention of Fe-NPs from brain vasculature into brain ventricles. A significant decrease in striatal dopamine and its metabolites was also observed, and neuropathological correlates provided additional evidence of significant nerve cell body and dopaminergic terminal damage as well as damage to neuronal vasculature after exposure to 10 nm Fe-NPs. These data demonstrate a neurotoxic potential of very small size iron nanoparticles and suggest that use of these ferric oxide nanoparticles may result in neurotoxicity, thereby limiting their clinical application.


Subject(s)
Dopaminergic Neurons/drug effects , Magnetite Nanoparticles/toxicity , Animals , Apoptosis/drug effects , Blood-Brain Barrier/drug effects , Caspases/metabolism , Catecholamines/analysis , Cell Division/drug effects , Cell Line, Tumor , Corpus Striatum/chemistry , Corpus Striatum/drug effects , Dopaminergic Neurons/chemistry , Dopaminergic Neurons/ultrastructure , Enzyme Activation/drug effects , Humans , Magnetic Resonance Imaging , Magnetic Resonance Spectroscopy , Male , Mitochondria/drug effects , Mitochondria/metabolism , Nanospheres , Neuroblastoma/pathology , Oxidative Stress , Particle Size , Permeability/drug effects , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/analysis , Spectrometry, X-Ray Emission
11.
PLoS One ; 8(5): e65129, 2013.
Article in English | MEDLINE | ID: mdl-23741470

ABSTRACT

Experimental evidence suggests that oxidative and nitrative mechanisms account for much of the dopaminergic neuronal injury in Parkinson's disease (PD). The ubiquitously expressed non-receptor tyrosine kinase c-Abl is activated by oxidative stress and thus, may play a role in redox-mediated neurodegeneration. Recently, we reported that c-Abl is activated in PD and that a c-Abl inhibitor mitigated neuronal damage in a PD animal model, suggesting a novel neuroprotective therapeutic approach. In the studies presented here, we evaluated the efficacy of a potent and clinically relevant second-generation irreversible Abl kinase inhibitor, INNO-406, as a therapeutic agent for PD. Our studies reveal that INNO-406 is capable of preventing the progression of dopaminergic neuronal damage in a toxin-induced C57 mouse model of PD. Using bovine brain microvessel endothelium as an in vitro blood-brain barrier (BBB) model, we detected rapid and significant transfer of INNO-406. Additionally, pharmacokinetic analyses demonstrated significant nanomolar concentrations of INNO-406 in brain in the presence or absence of MPTP administration, however, INNO-406 did not alter the brain levels of MPP+ in MPTP-treated mice. Finally, we showed that 10 mg/kg of INNO-406 given to C57 mice for one week before MPTP treatment (4×20 mg/kg i.p., every 2 h) and then for one week after MPTP treatment decreased the loss of dopamine in the striatum by 45% and the loss of TH+ neurons in substantia nigra pars compacts by 40%. This treatment regimen also abrogated activation of c-Abl, tyrosine phosphorylation of the Abl substrate and E3-ubiquitin ligase parkin, and accumulation of the toxic parkin substrate AIMP2. We propose that compounds of the INNO-406 class of Abl inhibitors will be useful new neuroprotective drugs for the treatment of PD-like pathology in preclinical systems that should be easily translated to the clinic.


Subject(s)
Brain/metabolism , Neuroprotective Agents/pharmacokinetics , Parkinson Disease/metabolism , Protein Kinase Inhibitors/pharmacokinetics , Proto-Oncogene Proteins c-abl/antagonists & inhibitors , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/adverse effects , Animals , Blood-Brain Barrier/metabolism , Brain/drug effects , Brain/pathology , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Corpus Striatum/pathology , Disease Models, Animal , Dopaminergic Neurons/drug effects , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Male , Mice , Neuroprotective Agents/administration & dosage , Parkinson Disease/drug therapy , Permeability , Phosphorylation/drug effects , Protein Kinase Inhibitors/administration & dosage , Proto-Oncogene Proteins c-abl/metabolism , Pyrimidines/administration & dosage , Pyrimidines/pharmacology , Ubiquitin-Protein Ligases/metabolism
12.
Neurosci Lett ; 541: 233-7, 2013 Apr 29.
Article in English | MEDLINE | ID: mdl-23499956

ABSTRACT

Rotenone, a widely used pesticide, causes a syndrome in rats that replicates, both pathologically and behaviorally, the symptoms of Parkinson's disease (PD). In the present study, we sought to determine if a chronic exposure to rotenone, resulting in dopaminergic loss, could also lead to peripheral neuronal damage related to motor dysfunction. Adult male Sprague-Dawley rats (n=14) were treated with rotenone (1 or 2mg/kg, s.c., once daily) on days 1, 3, 6, 8, 10, 13, 15, 17, 21, 22, and 27 to minimize mortality. Control rats received vehicle (DMSO) injections. Animals were weighed on the days of injection and monitored daily. A mortality of 21% was observed in rotenone treated rats. The motor nerve conduction velocity (MCV) was assessed using action potentials detected from the tail muscle through surface receiver electrodes installed around the distal portion of the tail. Rats exposed to rotenone often developed hind limb paresis with a significant decrease in MCV as detected in tail nerves (p<0.05). Animals were then sacrificed, either 24h after rotenone exposure on day 6 or 24h after the last dose of rotenone on day 27. The striatum and sciatic nerves were dissected on dry ice and flash-frozen and kept at -80°C until further analysis. Striatal dopamine (DA) was analyzed using HPLC-ECD and sciatic nerve pathology was analyzed for neurodegeneration. A time-dependent rotenone-induced striatal depletion of DA (60% after 7 days and 80% after 27 days) was observed. Furthermore, Neurofilament-neurofilament B, Flouro-Jade C and myelin basic protein analyses suggested a time-dependent rotenone-induced neurodegeneration in sciatic nerves. These data, for the first time, indicate an association between dopaminergic damage and peripheral motor nerve degeneration in an animal model of dopaminergic toxicity. Peripheral motor nerve dysfunction in rats following a chronic exposure to rotenone may serve not only as a relevant experimental model of motor neuropathy but also as a peripheral marker of dopaminergic neuronal damage to the central nervous system.


Subject(s)
Dopaminergic Neurons/pathology , Peripheral Nervous System Diseases/pathology , Pesticides/toxicity , Rotenone/toxicity , Action Potentials , Animals , Corpus Striatum/metabolism , Disease Models, Animal , Dopamine/metabolism , Dopaminergic Neurons/metabolism , Male , Nerve Degeneration/chemically induced , Nerve Degeneration/pathology , Nerve Degeneration/physiopathology , Neural Conduction , Peripheral Nervous System Diseases/chemically induced , Peripheral Nervous System Diseases/physiopathology , Rats , Rats, Sprague-Dawley , Sciatic Nerve/drug effects , Sciatic Nerve/pathology , Tail/innervation
13.
Neurosci Lett ; 535: 24-9, 2013 Feb 22.
Article in English | MEDLINE | ID: mdl-23295906

ABSTRACT

Although neurotrophic factors have long been recognized as potent agents for protecting against neuronal degeneration, clinical success in treating Parkinson's disease and other neurodegenerative disorders has been hindered by difficulties in delivery of trophic factors across the blood brain barrier (BBB). Bone marrow hematopoietic stem cell-based gene therapy is emerging as a promising tool for overcoming drug delivery problems, as myeloid cells can cross the BBB and are recruited in large numbers to sites of neurodegeneration, where they become activated microglia that can secrete trophic factors. We tested the efficacy of bone marrow-derived microglial delivery of neurturin (NTN) in protecting dopaminergic neurons against neurotoxin-induced death in mice. Bone marrow cells were transduced ex vivo with lentivirus expressing the NTN gene driven by a synthetic macrophage-specific promoter. Infected bone marrow cells were then collected and transplanted into recipient animals. Eight weeks after transplantation, the mice were injected with the neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropuridine (MPTP) for seven days to induce dopaminergic neurodegeneration. Microglia-mediated NTN delivery dramatically ameliorated MPTP-induced degeneration of tyrosine hydroxylase (TH)-positive neurons of the substantia nigra and their terminals in the striatum. Microglia-mediated NTN delivery also induced significant recovery of synaptic marker staining in the striatum of MPTP-treated animals. Functionally, NTN treatment restored MPTP-induced decline in general activity, rearing behavior, and food intake. Thus, bone marrow-derived microglia can serve as cellular vehicles for sustained delivery of neurotrophic factors capable of mitigating dopaminergic injury.


Subject(s)
Bone Marrow Cells/metabolism , Brain/pathology , Dopaminergic Neurons/pathology , Microglia/metabolism , Nerve Degeneration/prevention & control , Neurturin/metabolism , Parkinson Disease/prevention & control , Animals , Bone Marrow Transplantation , Brain/metabolism , Genetic Therapy , Lentivirus/genetics , Male , Maze Learning , Mice , Mice, Inbred C57BL , Microglia/transplantation , Motor Activity , Nerve Degeneration/genetics , Nerve Degeneration/physiopathology , Neurturin/genetics , Parkinson Disease/genetics , Parkinson Disease/physiopathology
14.
J Neurosci ; 31(1): 157-63, 2011 Jan 05.
Article in English | MEDLINE | ID: mdl-21209200

ABSTRACT

Mutations in parkin, an E3 ubiquitin ligase, are the most common cause of autosomal-recessive Parkinson's disease (PD). Here, we show that the stress-signaling non-receptor tyrosine kinase c-Abl links parkin to sporadic forms of PD via tyrosine phosphorylation. Under oxidative and dopaminergic stress, c-Abl was activated in cultured neuronal cells and in striatum of adult C57BL/6 mice. Activated c-Abl was found in the striatum of PD patients. Concomitantly, parkin was tyrosine-phosphorylated, causing loss of its ubiquitin ligase and cytoprotective activities, and the accumulation of parkin substrates, AIMP2 (aminoacyl tRNA synthetase complex-interacting multifunctional protein 2) (p38/JTV-1) and FBP-1.STI-571, a selective c-Abl inhibitor, prevented tyrosine phosphorylation of parkin and restored its E3 ligase activity and cytoprotective function both in vitro and in vivo. Our results suggest that tyrosine phosphorylation of parkin by c-Abl is a major post-translational modification that leads to loss of parkin function and disease progression in sporadic PD. Moreover, inhibition of c-Abl offers new therapeutic opportunities for blocking PD progression.


Subject(s)
Gene Expression Regulation/physiology , MPTP Poisoning/metabolism , Proto-Oncogene Proteins c-abl/metabolism , Tyrosine/metabolism , Ubiquitin-Protein Ligases/metabolism , Acetylcysteine/pharmacology , Animals , Benzamides , Brain/drug effects , Brain/metabolism , Brain/pathology , Case-Control Studies , Cell Line , Disease Models, Animal , Dopamine/pharmacology , Drug Administration Schedule , Free Radical Scavengers/pharmacology , Gene Expression Regulation/drug effects , Green Fluorescent Proteins/genetics , Humans , Imatinib Mesylate , Immunoprecipitation/methods , MPTP Poisoning/chemically induced , MPTP Poisoning/drug therapy , MPTP Poisoning/pathology , Male , Metalloporphyrins/pharmacology , Mice , Mice, Inbred C57BL , Oxidative Stress/drug effects , Oxidative Stress/physiology , Peptide Elongation Factors/genetics , Peptide Elongation Factors/metabolism , Phosphorylation/drug effects , Phosphorylation/physiology , Piperazines/toxicity , Polyethylene Glycols/pharmacology , Protein Kinase Inhibitors/toxicity , Proto-Oncogene Proteins c-abl/genetics , Pyrimidines/toxicity , RNA, Small Interfering/pharmacology , Statistics, Nonparametric , Transfection/methods , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitination/drug effects
15.
Int J Environ Res Public Health ; 8(12): 4702-13, 2011 12.
Article in English | MEDLINE | ID: mdl-22408597

ABSTRACT

Parkinson's disease (PD) is a common neurodegenerative movement disorder that is characterized pathologically by a progressive loss of midbrain dopaminergic neurons and by protein inclusions, designated Lewy bodies and Lewy neurites. PD is one of the most common neurodegenerative diseases, affecting almost 1% of the population over 60 years old. Although the symptoms and neuropathology of PD have been well characterized, the underlying mechanisms and causes of the disease are still not clear. Genetic mutations can provide important clues to disease mechanism, but most PD cases are sporadic rather than familial; environmental factors have long been suspected to contribute to the disease. Although more than 90% of PD cases occur sporadically and are thought to be due, in part, to oxidative stress and mitochondrial dysfunction, the study of genetic mutations has provided great insight into the molecular mechanisms of PD. Furthermore, rotenone, a widely used pesticide, and paraquat and maneb cause a syndrome in rats and mice that mimics, both behaviorally and neurologically, the symptoms of PD. In the current review, we will discuss various aspects of gene-environment interaction that lead to progressive dopaminergic neurodegenration, mainly focusing on our current finding based on stress-mediated parkin dysfunction.


Subject(s)
Dopamine/metabolism , Gene-Environment Interaction , Parkinson Disease/genetics , Ubiquitin-Protein Ligases/physiology , Animals , Humans , Rats , Ubiquitin-Protein Ligases/genetics
16.
Mol Biol Cell ; 21(22): 4042-56, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-20861316

ABSTRACT

Nonmuscle myosin light chain kinase (nmMLCK), a multi-functional cytoskeletal protein critical to vascular homeostasis, is highly regulated by tyrosine phosphorylation. We identified multiple novel c-Abl-mediated nmMLCK phosphorylation sites by mass spectroscopy analysis (including Y231, Y464, Y556, Y846) and examined their influence on nmMLCK function and human lung endothelial cell (EC) barrier regulation. Tyrosine phosphorylation of nmMLCK increased kinase activity, reversed nmMLCK-mediated inhibition of Arp2/3-mediated actin polymerization, and enhanced binding to the critical actin-binding phosphotyrosine protein, cortactin. EC challenge with sphingosine 1-phosphate (S1P), a potent barrier-enhancing agonist, resulted in c-Abl and phosphorylated nmMLCK recruitment into caveolin-enriched microdomains, rapid increases in Abl kinase activity, and spatial targeting of c-Abl to barrier-promoting cortical actin structures. Conversely, reduced c-Abl expression in EC (siRNA) markedly attenuated S1P-mediated cortical actin formation, reduced the EC modulus of elasticity (assessed by atomic force microscopy), reduced nmMLCK and cortactin tyrosine phosphorylation, and attenuated S1P-mediated barrier enhancement. These studies indicate an essential role for Abl kinase in vascular barrier regulation via posttranslational modification of nmMLCK and strongly support c-Abl-cortactin-nmMLCK interaction as a novel determinant of cortical actin-based cytoskeletal rearrangement critical to S1P-mediated EC barrier enhancement.


Subject(s)
Endothelial Cells/metabolism , Myosin-Light-Chain Kinase/metabolism , Proto-Oncogene Proteins c-abl/metabolism , Tyrosine/metabolism , Actins/metabolism , Amino Acid Sequence , Binding Sites/genetics , Blotting, Western , Capillary Permeability/drug effects , Caveolins/metabolism , Cell Line , Cytoskeleton/drug effects , Cytoskeleton/metabolism , Endothelial Cells/cytology , Endothelial Cells/drug effects , Enzyme Activation , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Lysophospholipids/pharmacology , Mass Spectrometry , Membrane Microdomains/drug effects , Membrane Microdomains/metabolism , Microscopy, Atomic Force , Microscopy, Confocal , Molecular Sequence Data , Myosin-Light-Chain Kinase/genetics , Phosphorylation , Protein Binding , Proto-Oncogene Proteins c-abl/genetics , RNA Interference , Sphingosine/analogs & derivatives , Sphingosine/pharmacology , Tyrosine/genetics
17.
Mol Ther ; 18(8): 1536-44, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20531393

ABSTRACT

Glial cell line-derived neurotrophic factor (GDNF) has emerged as the most potent neuroprotective agent tested in experimental models for the treatment of Parkinson's disease (PD). However, its use is hindered by difficulties in delivery to the brain due to the presence of the blood-brain barrier (BBB). In order to circumvent this problem, we took advantage of the fact that bone marrow stem cell-derived macrophages are able to pass the BBB and home to sites of neuronal degeneration. Here, we report the development of a method for brain delivery of GDNF by genetically modified macrophages. Bone marrow stem cells were transduced ex vivo with lentivirus expressing a GDNF gene driven by a synthetic macrophage-specific promoter and then transplanted into recipient mice. Eight weeks after transplantation, the mice were injected with the neurotoxin, MPTP, for 7 days to induce dopaminergic neurodegeneration. Macrophage-mediated GDNF treatment dramatically ameliorated MPTP-induced degeneration of tyrosine hydroxylase (TH)-positive neurons of the substantia nigra and TH(+) terminals in the striatum, stimulated axon regeneration, and reversed hypoactivity in the open field test. These results indicate that macrophage-mediated GDNF delivery is a promising strategy for developing a neuroprotective therapy for PD.


Subject(s)
Dopamine/metabolism , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Macrophages/metabolism , Nerve Degeneration/therapy , Parkinson Disease/therapy , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Animals , Body Weight/drug effects , Cells, Cultured , Chromatography, High Pressure Liquid , Eating/drug effects , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Glial Cell Line-Derived Neurotrophic Factor/genetics , Macrophages/drug effects , Male , Mice , Mice, Inbred C57BL , Nerve Degeneration/chemically induced , Neurotoxins/pharmacology , Parkinson Disease/metabolism , Substantia Nigra/drug effects , Substantia Nigra/metabolism
18.
Neurochem Res ; 33(6): 1113-6, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18320306

ABSTRACT

One of the tuberous sclerosis complex (TSC) gene products, tuberin is assumed to be the functional component, being involved in a wide variety of cellular processes. Here, we report for the first time that tuberin dysfunction may represent a mechanism for neuronal damage in Alzheimer's disease (AD), Parkinson's disease with dementia (PD/DLB), and a mouse model of PD. Tuberin was hyperphosphorylated at Thr1462 in post-mortem frontal cortex tissue of both AD and PD/DLB patients and in mice treated with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine hydrochloride (MPTP). Both PTEN and Akt phosphoactivation corresponded to the hyperphosphorylation patterns of tuberin suggesting that the PTEN-Akt pathway might be the mechanism of tuberin phosphorylation. Our data provide new information regarding the possible role of tuberin dysfunction in major neurodegenerative disorders, such as AD and PD, whereby inhibition of tuberin function may trigger an onset of neuronal cell death.


Subject(s)
Nerve Degeneration/metabolism , Neurons/metabolism , Neurons/pathology , Tumor Suppressor Proteins/metabolism , Animals , Disease Models, Animal , Humans , Male , Mice , Mice, Inbred C57BL , Nerve Degeneration/pathology , Neurons/cytology , PTEN Phosphohydrolase/metabolism , Parkinson Disease/pathology , Parkinson Disease/physiopathology , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Threonine/metabolism , Tuberous Sclerosis/genetics , Tuberous Sclerosis/pathology , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins/genetics
19.
J Biol Chem ; 283(11): 6979-87, 2008 Mar 14.
Article in English | MEDLINE | ID: mdl-18195004

ABSTRACT

Parkinson disease (PD) belongs to a heterogeneous group of neurodegenerative disorders with movement alterations, cognitive impairment, and alpha-synuclein accumulation in cortical and subcortical regions. Jointly, these disorders are denominated Lewy body disease. Mutations in the parkin gene are the most common cause of familial parkinsonism, and a growing number of studies have shown that stress factors associated with sporadic PD promote parkin accumulation in the insoluble fraction. alpha-Synuclein and parkin accumulation and mutations in these genes have been associated with familial PD. To investigate whether alpha-synuclein accumulation might be involved in the pathogenesis of these disorders by interfering with parkin solubility, synuclein-transfected neuronal cells were transduced with lentiviral vectors expressing parkin. Challenging neurons with proteasome inhibitors or amyloid-beta resulted in accumulation of insoluble parkin and, to a lesser extent, alpha-tubulin. Similarly to neurons in the brains of patients with Lewy body disease, in co-transduced cells alpha-synuclein and parkin colocalized and co-immunoprecipitated. These effects resulted in decreased parkin and alpha-tubulin ubiquitination, accumulation of insoluble parkin, and cytoskeletal alterations with reduced neurite outgrowth. Taken together, accumulation of alpha-synuclein might contribute to the pathogenesis of PD and other Lewy body diseases by promoting alterations in parkin and tubulin solubility, which in turn might compromise neural function by damaging the neuronal cytoskeleton. These studies provide a new perspective on the potential nature of pathogenic alpha-synuclein and parkin interactions in Parkinson disease.


Subject(s)
Gene Expression Regulation , Parkinson Disease/metabolism , Tubulin/physiology , Ubiquitin-Protein Ligases/physiology , alpha-Synuclein/physiology , Aged , Aged, 80 and over , Animals , Cerebral Cortex/metabolism , Female , Humans , Male , Models, Biological , Neurons/metabolism , Rats , alpha-Synuclein/metabolism , beta-Synuclein/metabolism
20.
Nucleic Acids Res ; 35(15): 4941-51, 2007.
Article in English | MEDLINE | ID: mdl-17626041

ABSTRACT

The Cockayne Syndrome group B (CSB) protein plays important roles in transcription, transcription-coupled nucleotide excision repair and base excision DNA repair. c-Abl kinase also plays a role in DNA repair as a regulator/coordinator of the DNA damage response. This study presents evidence that the N-terminal region of CSB interacts with the SH3 domain of c-Abl in vitro and in vivo. In addition, c-Abl kinase phosphorylates CSB at Tyr932. The subcellular localization of CSB to the nucleus and nucleolus is altered after phosphorylation by c-Abl. c-Abl-dependent phosphorylation of CSB increased in cells treated with hydrogen peroxide and decreased in cells pre-treated with STI-571, a c-Abl-specific protein kinase inhibitor. Activation of the c-Abl kinase in response to oxidative damage is not observed in CSB null cells. These results suggest that c-Abl and CSB may regulate each other in a reciprocal manner in response to oxidative stress.


Subject(s)
DNA Helicases/metabolism , DNA Repair Enzymes/metabolism , Oxidative Stress , Proto-Oncogene Proteins c-abl/metabolism , Animals , Cell Line , Cells, Cultured , DNA Helicases/analysis , DNA Helicases/chemistry , DNA Repair Enzymes/analysis , DNA Repair Enzymes/chemistry , Humans , Mice , Phosphorylation , Poly-ADP-Ribose Binding Proteins , Proto-Oncogene Proteins c-abl/analysis , Tyrosine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...