Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Psychoneuroendocrinology ; 133: 105393, 2021 11.
Article in English | MEDLINE | ID: mdl-34481327

ABSTRACT

Maternal separation (MS) is a known chronic stressor in the postnatal period and when associated with another paradigm like the activity-based anorexia (ABA) rat model, causes different effects in the two sexes. In ABA females, the separation leads to increased hyperactivity and anxiety reduction, whereas, in males, the separation induces decreased locomotor activity without similar reduction of anxiety-like behaviors as observed in females. To understand the mechanisms altered by MS in synergy with the induction of the anorexic-like phenotype, we considered the reward system, which involves neurons synthesizing dopamine (DA) in the ventral tegmental area (VTA), substantia nigra pars compacta, and serotoninergic neurons in the dorsal raphe nucleus. Moreover, we analyzed the orexin circuit in the lateral hypothalamic area (LHA), which affects DA synthesis in the VTA and is also known to regulate food consumption and locomotor activity. Rats of both sexes were exposed to the two paradigms (MS and ABA), leading to four experimental groups for each sex: non-separated control (CON), non-separated ABA groups (ABA), MS control (MSCON), and MS plus ABA groups (MSABA). Immunohistochemistry analysis was performed to determine quantitative differences in the number of cells expressing DA, orexin, and serotonin (5-HT) among the experimental groups. The results showed that, in the DA system, the effect of MS was more evident in females than in males, with a substantial increase in DA cells in the VTA of MSABA. However, the analysis of the orexin system revealed a similar cellular increment in the LHA in the non-separated ABA groups of both sexes. Regarding 5-HT, there was an opposite effect in males and females of the MSABA groups, with only females showing a greater density of 5-HT cells. The changes in the reward system could partially explain the behavioral data: the hyperactivity, weight loss, and decreased anxiety levels of the MSABA females could be linked to an increase in DA and 5-HT cells, whereas in males, MS could mitigate the behavioral effects of the ABA protocol affecting the anxiety levels and locomotor activity through a lack of increased activation of the reward system.


Subject(s)
Anorexia , Maternal Deprivation , Reward , Animals , Anorexia/complications , Anxiety/complications , Disease Models, Animal , Dopamine , Dorsal Raphe Nucleus/cytology , Female , Male , Neurons , Orexins , Pars Compacta/cytology , Rats , Serotonin , Ventral Tegmental Area/cytology
2.
Nat Commun ; 12(1): 4409, 2021 07 20.
Article in English | MEDLINE | ID: mdl-34285209

ABSTRACT

Appetitive locomotion is essential for animals to approach rewards, such as food and prey. The neuronal circuitry controlling appetitive locomotion is unclear. In a goal-directed behavior-predatory hunting, we show an excitatory brain circuit from the superior colliculus (SC) to the substantia nigra pars compacta (SNc) to enhance appetitive locomotion in mice. This tectonigral pathway transmits locomotion-speed signals to dopamine neurons and triggers dopamine release in the dorsal striatum. Synaptic inactivation of this pathway impairs appetitive locomotion but not defensive locomotion. Conversely, activation of this pathway increases the speed and frequency of approach during predatory hunting, an effect that depends on the activities of SNc dopamine neurons. Together, these data reveal that the SC regulates locomotion-speed signals to SNc dopamine neurons to enhance appetitive locomotion in mice.


Subject(s)
Appetitive Behavior/physiology , Locomotion/physiology , Pars Compacta/physiology , Predatory Behavior/physiology , Superior Colliculi/physiology , Animals , Dopamine/metabolism , Dopaminergic Neurons/metabolism , Male , Mice , Mice, Transgenic , Models, Animal , Neural Pathways/physiology , Pars Compacta/cytology , Stereotaxic Techniques , Superior Colliculi/cytology , Synaptic Transmission/physiology
3.
N Engl J Med ; 382(20): 1926-1932, 2020 05 14.
Article in English | MEDLINE | ID: mdl-32402162

ABSTRACT

We report the implantation of patient-derived midbrain dopaminergic progenitor cells, differentiated in vitro from autologous induced pluripotent stem cells (iPSCs), in a patient with idiopathic Parkinson's disease. The patient-specific progenitor cells were produced under Good Manufacturing Practice conditions and characterized as having the phenotypic properties of substantia nigra pars compacta neurons; testing in a humanized mouse model (involving peripheral-blood mononuclear cells) indicated an absence of immunogenicity to these cells. The cells were implanted into the putamen (left hemisphere followed by right hemisphere, 6 months apart) of a patient with Parkinson's disease, without the need for immunosuppression. Positron-emission tomography with the use of fluorine-18-L-dihydroxyphenylalanine suggested graft survival. Clinical measures of symptoms of Parkinson's disease after surgery stabilized or improved at 18 to 24 months after implantation. (Funded by the National Institutes of Health and others.).


Subject(s)
Dopaminergic Neurons/cytology , Induced Pluripotent Stem Cells/transplantation , Parkinson Disease/therapy , Pars Compacta/cytology , Aged , Animals , Basal Ganglia/diagnostic imaging , Basal Ganglia/metabolism , Cell Differentiation , Disease Models, Animal , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/transplantation , Follow-Up Studies , Humans , Induced Pluripotent Stem Cells/immunology , Male , Mice , Mice, SCID , Parkinson Disease/diagnostic imaging , Positron-Emission Tomography , Putamen/diagnostic imaging , Tomography, X-Ray Computed , Transplantation, Autologous , Transplantation, Homologous
4.
PLoS Genet ; 15(8): e1008352, 2019 08.
Article in English | MEDLINE | ID: mdl-31449520

ABSTRACT

Parkinson's disease (PD) is a neurodegenerative disorder characterized by the loss of dopamine (DA) neurons in the substantia nigra pars compacta (SNc). Rare genetic mutations in genes such as Parkin, Pink1, DJ-1, α-synuclein, LRRK2 and GBA are found to be responsible for the disease in about 15% of the cases. A key unanswered question in PD pathophysiology is why would these mutations, impacting basic cellular processes such as mitochondrial function and neurotransmission, lead to selective degeneration of SNc DA neurons? We previously showed in vitro that SNc DA neurons have an extremely high rate of mitochondrial oxidative phosphorylation and ATP production, characteristics that appear to be the result of their highly complex axonal arborization. To test the hypothesis in vivo that axon arborization size is a key determinant of vulnerability, we selectively labeled SNc or VTA DA neurons using floxed YFP viral injections in DAT-cre mice and showed that SNc DA neurons have a much more arborized axon than those of the VTA. To further enhance this difference, which may represent a limiting factor in the basal vulnerability of these neurons, we selectively deleted in mice the DA D2 receptor (D2-cKO), a key negative regulator of the axonal arbour of DA neurons. In these mice, SNc DA neurons have a 2-fold larger axonal arborization, release less DA and are more vulnerable to a 6-OHDA lesion, but not to α-synuclein overexpression when compared to control SNc DA neurons. This work adds to the accumulating evidence that the axonal arborization size of SNc DA neurons plays a key role in their vulnerability in the context of PD.


Subject(s)
Dopaminergic Neurons/pathology , Neuronal Plasticity/genetics , Parkinson Disease/pathology , Pars Compacta/pathology , Receptors, Dopamine D2/genetics , Animals , Axons/pathology , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Knockout , Mitochondria/pathology , Oxidative Phosphorylation , Parkinson Disease/genetics , Pars Compacta/cytology , Receptors, Dopamine D2/metabolism
5.
Neuropharmacology ; 158: 107705, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31301335

ABSTRACT

Dopamine neurons in the substantia nigra zona compacta (SNC) are well known to express D2 receptors. When dopamine is released from somatodendritic sites, activation of D2 autoreceptors suppresses dopamine neuronal activity through activation of G protein-coupled K+ channels. AMP-activated protein kinase (AMPK) is a master enzyme that acts in somatic tissues to suppress energy expenditure and encourage energy production. We hypothesize that AMPK may also conserve energy in central neurons by reducing desensitization of D2 autoreceptors. We used whole-cell patch-clamp recordings to study the effects of AMPK activators and inhibitors on D2 autoreceptor-mediated current in SNC neurons in midbrain slices from rat pups (11-23 days post-natal). Slices were superfused with 100 µM dopamine or 30 µM quinpirole for 25 min, which evoked outward currents that decayed slowly over time. Although the AMPK activators A769662 and ZLN024 significantly slowed rundown of dopamine-evoked current, slowing of quinpirole-evoked current required the presence of a D1-like agonist (SKF38393). Moreover, the D1-like agonist also slowed the rundown of quinpirole-induced current even in the absence of an AMPK activator. Pharmacological antagonist experiments showed that the D1-like agonist effect required activation of either protein kinase A (PKA) or exchange protein directly activated by cAMP 2 (Epac2) pathways. In contrast, the effect of AMPK on rundown of current evoked by quinpirole plus SKF38393 required PKA but not Epac2. We conclude that AMPK slows D2 autoreceptor desensitization by augmenting the effect of D1-like receptors.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Autoreceptors/metabolism , Dopamine Agonists/pharmacology , Dopamine/pharmacology , Neurons/metabolism , Pars Compacta/cytology , Quinpirole/pharmacology , Receptors, Dopamine D2/metabolism , 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine/pharmacology , AMP-Activated Protein Kinases/drug effects , Animals , Autoreceptors/drug effects , Biphenyl Compounds , Cyclic AMP-Dependent Protein Kinases/metabolism , Enzyme Activators/pharmacology , Guanine Nucleotide Exchange Factors/metabolism , Neurons/drug effects , Patch-Clamp Techniques , Pyrimidines/pharmacology , Pyrones/pharmacology , Rats , Receptors, Dopamine D2/drug effects , Thiophenes/pharmacology
6.
JCI Insight ; 52019 05 23.
Article in English | MEDLINE | ID: mdl-31120439

ABSTRACT

Parkinson's is primarily a non-familial, age-related disorder caused by α-synuclein accumulation and the progressive loss of dopamine neurons in the substantia nigra pars compacta (SNc). G protein-coupled receptor (GPCR)-cAMP signaling has been linked to a reduction in human Parkinson's incidence and α-synuclein expression. Neuronal cAMP levels are controlled by GPCRs coupled to Gs or Gi/o, which increase or decrease cAMP, respectively. Regulator of G protein signaling 6 (RGS6) powerfully inhibits Gi/o signaling. Therefore, we hypothesized that RGS6 suppresses D2 autoreceptor- Gi/o signaling in SNc dopamine neurons promoting neuronal survival and reducing α-synuclein expression. Here we provide novel evidence that RGS6 critically suppresses late-age-onset SNc dopamine neuron loss and α-synuclein accumulation. RGS6 is restrictively expressed in human SNc dopamine neurons and, despite their loss in Parkinson's, all surviving neurons express RGS6. RGS6-/- mice exhibit hyperactive D2 autoreceptors with reduced cAMP signaling in SNc dopamine neurons. Importantly, RGS6-/- mice recapitulate key sporadic Parkinson's hallmarks, including: SNc dopamine neuron loss, reduced nigrostriatal dopamine, motor deficits, and α-synuclein accumulation. To our knowledge, Rgs6 is the only gene whose loss phenocopies these features of human Parkinson's. Therefore, RGS6 is a key regulator of D2R-Gi/o signaling in SNc dopamine neurons, protecting against Parkinson's neurodegeneration and α-synuclein accumulation.


Subject(s)
Dopaminergic Neurons/metabolism , Parkinson Disease/genetics , Pars Compacta/metabolism , RGS Proteins/genetics , RGS Proteins/metabolism , Receptors, Dopamine D2/metabolism , alpha-Synuclein/metabolism , Age Factors , Age of Onset , Animals , Dopamine/metabolism , Dopamine Agonists/pharmacology , Dopaminergic Neurons/pathology , Humans , Locomotion , Mice , Mice, Knockout , Parkinson Disease/metabolism , Parkinson Disease/pathology , Parkinson Disease/physiopathology , Pars Compacta/cytology , Pars Compacta/pathology , Quinpirole/pharmacology , Synaptic Transmission
7.
Int J Mol Sci ; 20(3)2019 Jan 29.
Article in English | MEDLINE | ID: mdl-30699944

ABSTRACT

Parkinson's disease (PD) is a neurodegenerative disorder defined by progressive deterioration of dopaminergic neurons in the substantia nigra pars compacta (SNpc). Dental pulp stem cells (DPSCs) have been proposed to replace the degenerated dopaminergic neurons due to its inherent neurogenic and regenerative potential. However, the effective delivery and homing of DPSCs within the lesioned brain has been one of the many obstacles faced in cell-based therapy of neurodegenerative disorders. We hypothesized that DPSCs, delivered intranasally, could circumvent these challenges. In the present study, we investigated the therapeutic efficacy of intranasally administered DPSCs in a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced mouse model of PD. Human deciduous DPSCs were cultured, pre-labelled with PKH 26, and intranasally delivered into PD mice following MPTP treatment. Behavioural analyses were performed to measure olfactory function and sensorimotor coordination, while tyrosine hydroxylase (TH) immunofluorescence was used to evaluate MPTP neurotoxicity in SNpc neurons. Upon intranasal delivery, degenerated TH-positive neurons were ameliorated, while deterioration in behavioural performances was significantly enhanced. Thus, the intranasal approach enriched cell delivery to the brain, optimizing its therapeutic potential through its efficacious delivery and protection against dopaminergic neuron degeneration.


Subject(s)
Dental Pulp/cytology , MPTP Poisoning/therapy , Parkinson Disease/therapy , Pars Compacta/cytology , Stem Cells/physiology , Animals , Behavior, Animal , Cell Differentiation/physiology , Cells, Cultured , Dopaminergic Neurons/metabolism , Humans , MPTP Poisoning/metabolism , Male , Mice , Nerve Degeneration/metabolism , Nerve Degeneration/therapy , Parkinson Disease/metabolism , Pars Compacta/metabolism , Tyrosine 3-Monooxygenase/metabolism
8.
Eur J Neurosci ; 46(11): 2746-2753, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29057540

ABSTRACT

ATP-sensitive K+ (K-ATP) channels play significant roles in regulating the excitability of dopamine neurons in the substantia nigra zona compacta (SNC). We showed previously that K-ATP channel function is up-regulated by AMP-activated protein kinase (AMPK). This study extended these studies to the neurons adjacent to the SNC in the ventral tegmental area (VTA). Using patch pipettes to record whole-cell currents in slices of rat midbrain, we found that the AMPK activator A769662 increased the amplitude of currents evoked by the K-ATP channel opener diazoxide in presumed dopamine-containing VTA neurons. However, current evoked by diazoxide with A769662 was significantly smaller in VTA neurons compared to SNC neurons. Moreover, a significantly lower proportion of VTA neurons responded to diazoxide with outward current. However, A769662 was able to increase the incidence of diazoxide-responsive neurons in the VTA. In contrast, A769662 did not potentiate diazoxide-evoked currents in presumed non-dopamine VTA neurons. These results show that AMPK activation augments K-ATP currents in presumed dopamine neurons in the VTA and SNC, although diazoxide-evoked currents remain less robust in the VTA. We conclude that K-ATP channels may play important physiological roles in VTA and SNC dopamine neurons.


Subject(s)
Adenylate Kinase/metabolism , KATP Channels/metabolism , Pars Compacta/cytology , Pars Compacta/metabolism , Ventral Tegmental Area/cytology , Ventral Tegmental Area/metabolism , Animals , Biphenyl Compounds , Diazoxide/pharmacology , Dopaminergic Neurons/physiology , Drug Synergism , Male , Membrane Potentials/drug effects , Membrane Potentials/physiology , Pars Compacta/drug effects , Pyrones/pharmacology , Rats , Thiophenes/pharmacology , Ventral Tegmental Area/drug effects
9.
Mol Pharmacol ; 92(6): 640-652, 2017 12.
Article in English | MEDLINE | ID: mdl-29025968

ABSTRACT

The autophagy-lysosome pathway (ALP) plays a critical role in the pathology of Parkinson's disease (PD). Clk1 (coq7) is a mitochondrial hydroxylase that is essential for coenzyme Q (ubiquinone) biosynthesis. We have reported previously that Clk1 regulates microglia activation via modulating microglia metabolic reprogramming, which contributes to dopaminergic neuronal survival. This study explores the direct effect of Clk1 on dopaminergic neuronal survival. We demonstrate that Clk1 deficiency inhibited dopaminergic neuronal autophagy in cultured MN9D dopaminergic neurons and in the substantia nigra pars compacta of Clk+/- mutant mice and consequently sensitized dopaminergic neuron damage and behavioral defects. These mechanistic studies indicate that Clk1 regulates the AMP-activated protein kinase (AMPK)/rapamycin complex 1 pathway, which in turn impairs the ALP and TFEB nuclear translocation. As a result, Clk1 deficiency promotes dopaminergic neuronal damage in vivo and in vitro, which ultimately contributes to sensitizing 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced dopaminergic neuronal death and behavioral impairments in Clk1-deficient mice. Moreover, we found that activation of autophagy by the AMPK activator metformin increases dopaminergic neuronal survival in vitro and in the MPTP-induced PD model in Clk1 mutant mice. These results reveal that Clk1 plays a direct role in dopaminergic neuronal survival via regulating ALPs that may contribute to the pathologic development of PD. Modulation of Clk1 activity may represent a potential therapeutic target for PD.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Autophagy , Dopaminergic Neurons/drug effects , Enzyme Activators/pharmacology , Mechanistic Target of Rapamycin Complex 1/metabolism , Membrane Proteins/metabolism , Metformin/pharmacology , Mitochondrial Proteins/metabolism , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Animals , Cell Survival , Cells, Cultured , Dopamine Agents/pharmacology , Dopaminergic Neurons/cytology , Dopaminergic Neurons/metabolism , Membrane Proteins/genetics , Mice , Mice, Mutant Strains , Mitochondrial Proteins/genetics , Mixed Function Oxygenases , Pars Compacta/cytology , Pars Compacta/drug effects
10.
Endocr Regul ; 51(2): 73-83, 2017 Apr 25.
Article in English | MEDLINE | ID: mdl-28609288

ABSTRACT

OBJECTIVE: Fos protein expression in catecholamine-synthesizing neurons of the substantia nigra (SN) pars compacta (SNC, A8), pars reticulata (SNR, A9), and pars lateralis (SNL), the ventral tegmental area (VTA, A10), the locus coeruleus (LC, A6) and subcoeruleus (sLC), the ventrolateral pons (PON-A5), the nucleus of the solitary tract (NTS-A2), the area postrema (AP), and the ventrolateral medulla (VLM-A1) was quantitatively evaluated aft er a single administration of asenapine (ASE) (designated for schizophrenia treatment) in male Wistar rats preconditioned with a chronic unpredictable variable mild stress (CMS) for 21 days. Th e aim of the present study was to reveal whether a single ASE treatment may 1) activate Fos expression in the brain areas selected; 2) activate tyrosine hydroxylase (TH)-synthesizing cells displaying Fos presence; and 3) be modulated by CMS preconditioning. METHODS: Control (CON), ASE, CMS, and CMS+ASE groups were used. CMS included restraint, social isolation, crowding, swimming, and cold. Th e ASE and CMS+ASE groups received a single dose of ASE (0.3 mg/kg, s.c.) and CON and CMS saline (300 µl/rat, s.c.). The animals were sacrificed 90 min aft er the treatments. Fos protein and TH-labeled immunoreactive perikarya were analyzed on double labeled histological sections and enumerated on captured pictures using combined light and fluorescence microscope illumination. RESULTS: Saline or CMS alone did not promote Fos expression in any of the structures investigated. ASE alone or in combination with CMS elicited Fos expression in two parts of the SN (SNC, SNR) and the VTA. Aside from some cells in the central gray tegmental nuclei adjacent to LC, where a small number of Fos profiles occurred, none or negligible Fos occurrence was detected in the other structures investigated including the LC and sLC, PON-A5, NTS-A2, AP, and VLM-A1. CMS preconditioning did not infl uence the level of Fos induction in the SN and VTA elicited by ASE administration. Similarly, the ratio between the amount of free Fos and Fos colocalized with TH was not aff ected by stress preconditioning in the SNC, SNR, and the VTA. CONCLUSIONS: Th e present study provides an anatomical/functional knowledge about the nature of the acute ASE treatment on the catecholamine-synthesizing neurons activity in certain brain structures and their missing interplay with the CMS preconditioning.


Subject(s)
Antipsychotic Agents/pharmacology , Brain/drug effects , Conditioning, Psychological , Heterocyclic Compounds, 4 or More Rings/pharmacology , Neurons/drug effects , Proto-Oncogene Proteins c-fos/drug effects , Stress, Psychological/metabolism , Tyrosine 3-Monooxygenase/drug effects , Animals , Area Postrema/cytology , Area Postrema/drug effects , Area Postrema/metabolism , Brain/cytology , Brain/metabolism , Catecholamines/biosynthesis , Dibenzocycloheptenes , Immunohistochemistry , Locus Coeruleus/cytology , Locus Coeruleus/drug effects , Locus Coeruleus/metabolism , Male , Medulla Oblongata/cytology , Medulla Oblongata/drug effects , Medulla Oblongata/metabolism , Microscopy, Fluorescence , Neurons/metabolism , Pars Compacta/cytology , Pars Compacta/drug effects , Pars Compacta/metabolism , Pars Reticulata/cytology , Pars Reticulata/drug effects , Pars Reticulata/metabolism , Pons/cytology , Pons/drug effects , Pons/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Rats , Rats, Wistar , Solitary Nucleus/cytology , Solitary Nucleus/drug effects , Solitary Nucleus/metabolism , Stress, Psychological/psychology , Tyrosine 3-Monooxygenase/metabolism , Ventral Tegmental Area/cytology , Ventral Tegmental Area/drug effects , Ventral Tegmental Area/metabolism
11.
Exp Neurol ; 287(Pt 1): 34-43, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27771354

ABSTRACT

Parkinson's disease (PD) is not only associated with degeneration of dopaminergic (DAergic) neurons in the Substantia Nigra, but also with profound loss of noradrenergic neurons in the Locus Coeruleus (LC). Remarkably, LC degeneration may exceed, or even precede the loss of nigral DAergic neurons, suggesting that LC neurons may be more susceptible to damage by various insults. Using a combination of electrophysiology, fluorescence imaging and electrochemistry, we directly compared the responses of LC, nigral DAergic and nigral non-dopaminergic (non-DAergic) neurons in rat brain slices to acute application of rotenone, a mitochondrial toxin used to create animal and in vitro models of PD. Rotenone (0.01-5.0µM) dose-dependently inhibited the firing of all three groups of neurons, primarily by activating KATP channels. The toxin also depolarised mitochondrial potential (Ψm) and released reactive oxygen species (H2O2). When KATP channels were blocked, rotenone (1µM) increased the firing of LC neurons by activating an inward current associated with dose-dependent increase of cytosolic free Ca2+ ([Ca2+]i). This effect was attenuated by blocking oxidative stress-sensitive TRPM2 channels, and by pre-treatment of slices with anti-oxidants. These results demonstrate that rotenone inhibits the activity of LC neurons mainly by activating KATP channels, and increases [Ca2+]ivia TRPM2 channels. Since the responses of LC neurons were smaller than those of nigral DAergic neurons, our study shows that LC neurons are paradoxically less sensitive to acute effects of this parkinsonian toxin.


Subject(s)
Action Potentials/drug effects , Insecticides/pharmacology , Locus Coeruleus/cytology , Neurons/drug effects , Pars Compacta/cytology , Rotenone/pharmacology , Animals , Animals, Newborn , Antihypertensive Agents/pharmacology , Calcium/metabolism , Diazoxide/pharmacology , Hydrogen Peroxide/metabolism , Hypoglycemic Agents/pharmacology , In Vitro Techniques , Membrane Potential, Mitochondrial/drug effects , Neurons/classification , Patch-Clamp Techniques , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , TRPM Cation Channels/metabolism , Tolbutamide/pharmacology
12.
PLoS One ; 11(10): e0164094, 2016.
Article in English | MEDLINE | ID: mdl-27788145

ABSTRACT

The neurotoxicity of paraquat dichloride (PQ) was assessed in two inbred strains of 9- or 16-week old male C57BL/6 mice housed in two different laboratories and compared to the effects of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). PQ was administered by intraperitoneal injections; either once (20 mg/kg) or twice (10 mg/kg) weekly for 3 weeks, while MPTP-HCl was injected 4 times on a single day (20 mg/kg/dose). Brains were collected 8, 16, 24, 48, 96 or 168 hours after the last PQ treatment, and 48 or 168 hours after MPTP treatment. Dopamine neurons in the substantia nigra pars compacta (SNpc) were identified by antibodies to tyrosine hydroxylase (TH+) and microglia were identified using Iba-1 immunoreactivity. The total number of TH+ neurons and the number of resting and activated microglia in the SNpc at 168 hours after the last dose were estimated using model- or design-based stereology, with investigators blinded to treatment. In a further analysis, a pathologist, also blinded to treatment, evaluated the SNpc and/or striatum for loss of TH+ neurons (SNpc) or terminals (striatum), cell death (as indicated by amino cupric silver uptake, TUNEL and/or caspase 3 staining) and neuroinflammation (as indicated by Iba-1 and/or GFAP staining). PQ, administered either once or twice weekly to 9- or 16-week old mice from two suppliers, had no effect on the number of TH+ neurons or microglia in the SNpc, as assessed by two groups, each blinded to treatment, using different stereological methods. PQ did not induce neuronal cell loss or degeneration in the SNpc or striatum. Additionally, there was no evidence of apoptosis, microgliosis or astrogliosis. In MPTP-treated mice, the number of TH+ neurons in the SNpc was significantly decreased and the number of activated microglia increased. Histopathological assessment found degenerating neurons/terminals in the SNpc and striatum but no evidence of apoptotic cell death. MPTP activated microglia in the SNpc and increased the number of astrocytes in the SNpc and striatum.


Subject(s)
Dopaminergic Neurons/drug effects , MPTP Poisoning/pathology , Microglia/drug effects , Paraquat/toxicity , Pars Compacta/cytology , Animals , Body Weight/drug effects , Cell Count , Dopaminergic Neurons/cytology , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Eating/drug effects , Male , Mice , Mice, Inbred C57BL , Microglia/cytology , Microglia/pathology , Pars Compacta/pathology , Survival Analysis , Tyrosine 3-Monooxygenase/metabolism
13.
Sci Rep ; 6: 30615, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27477243

ABSTRACT

The rodent ventral tegmental area (VTA) and substantia nigra pars compacta (SNC) contain dopamine neurons intermixed with glutamate neurons (expressing vesicular glutamate transporter 2; VGluT2), which play roles in reward and aversion. However, identifying the neuronal compositions of the VTA and SNC in higher mammals has remained challenging. Here, we revealed VGluT2 neurons within the VTA and SNC of nonhuman primates and humans by simultaneous detection of VGluT2 mRNA and tyrosine hydroxylase (TH; for identification of dopamine neurons). We found that several VTA subdivisions share similar cellular compositions in nonhuman primates and humans; their rostral linear nuclei have a high prevalence of VGluT2 neurons lacking TH; their paranigral and parabrachial pigmented nuclei have mostly TH neurons, and their parabrachial pigmented nuclei have dual VGluT2-TH neurons. Within nonhuman primates and humans SNC, the vast majority of neurons are TH neurons but VGluT2 neurons were detected in the pars lateralis subdivision. The demonstration that midbrain dopamine neurons are intermixed with glutamate or glutamate-dopamine neurons from rodents to humans offers new opportunities for translational studies towards analyzing the roles that each of these neurons play in human behavior and in midbrain-associated illnesses such as addiction, depression, schizophrenia, and Parkinson's disease.


Subject(s)
Dopamine/metabolism , Glutamic Acid/metabolism , Mesencephalon/cytology , Neurons/metabolism , Animals , Callithrix , Humans , Male , Mesencephalon/metabolism , Pars Compacta/cytology , Pars Compacta/metabolism , Tyrosine 3-Monooxygenase/genetics , Tyrosine 3-Monooxygenase/metabolism , Ventral Tegmental Area/cytology , Ventral Tegmental Area/metabolism , Vesicular Glutamate Transport Protein 2/genetics , Vesicular Glutamate Transport Protein 2/metabolism
14.
An Acad Bras Cienc ; 88(3): 1439-50, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27508995

ABSTRACT

The Portulaca oleracea L. (Portulacaceae) is a cosmopolitan species with a wide range of biological activities, including antioxidant and neuroprotective actions. We investigated the effects of P. oleracea extracts in a 6-hydroxydopamine rat model of Parkinson's disease, a debilitating disorder without effective treatments. Chemical profiles of aqueous and ethanolic extracts of whole plant were analyzed by thin layer chromatography and the antioxidant activity was assessed by 2,2-diphenyl-1-picrilhidrazila method. Male Wistar rats received intrastriatal 6-hydroxydopamine and were treated with vehicle or extracts (oral, 200 and 400 mg/kg) daily for two weeks. The behavioral open field test was conducted at days 1 and 15. Immunohistochemical analysis was performed 4 weeks after surgery to quantify tyrosine-hydroxylase cell counts in the substantia nigra pars compacta. Extracts presented antioxidant activity in concentrations above 300 µg/kg. The chromatographic analysis revealed the presence of Levodopa, alkaloids, flavonoids, saponins, tannins, terpenoids and polysaccharides. Both extracts improved motor recovery 15 days after lesion and protected from tyrosine-hydroxylase cell loss after 4 weeks, but these effects were more evident for the aqueous extract. Because the dopamine precursor is present, in addition to antioxidant compounds and neuroprotective effects, P. oleracea can be considered as potential strategy for treating Parkinson's disease.


Subject(s)
Dopaminergic Neurons/drug effects , Neuroprotective Agents/pharmacology , Oxidopamine/toxicity , Portulaca/chemistry , Sympatholytics/toxicity , Animals , Dopaminergic Neurons/enzymology , Male , Oxidopamine/antagonists & inhibitors , Parkinson Disease/drug therapy , Pars Compacta/cytology , Pars Compacta/enzymology , Rats , Rats, Wistar , Sympatholytics/antagonists & inhibitors , Tyrosine 3-Monooxygenase/analysis
15.
Cell Death Dis ; 7: e2217, 2016 05 05.
Article in English | MEDLINE | ID: mdl-27148690

ABSTRACT

Glial cell line-derived neurotrophic factor (GDNF) has strong neuroprotective and neurorestorative effects on dopaminergic (DA) neurons in the substantia nigra (SN); however, the underlying molecular mechanisms remain to be fully elucidated. In this study, we found that the expression level of transcription factor Six2 was increased in damaged DA neurons after GDNF rescue in vivo and in vitro. Knockdown of Six2 resulted in decreased cell viability and increased the apoptosis of damaged DA neurons after GDNF treatment in vitro. In contrast, Six2 overexpression increased cell viability and decreased cell apoptosis. Furthermore, genome-wide chromatin immunoprecipitation sequencing (ChIP-seq) indicated that Six2 directly bound to the promoter CAGCTG sequence of smad ubiquitylation regulatory factor 1 (Smurf1). ChIP-quantitative polymerase chain reaction (qPCR) analysis showed that Smurf1 expression was significantly upregulated after GDNF rescue. Moreover, knockdown of Six2 decreased Smurf1 expression, whereas overexpression of Six2 increased Smurf1 expression in damaged DA neurons after GDNF rescue. Meanwhile, knockdown and overexpression of Smurf1 increased and decreased p53 expression, respectively. Taken together, our results from in vitro and in vivo analysis indicate that Six2 mediates the protective effects of GDNF on damaged DA neurons by regulating Smurf1 expression, which could be useful in identifying potential drug targets for injured DA neurons.


Subject(s)
Dopaminergic Neurons/drug effects , Glial Cell Line-Derived Neurotrophic Factor/pharmacology , Homeodomain Proteins/genetics , Neuroprotective Agents/pharmacology , Pars Compacta/drug effects , Transcription Factors/genetics , Ubiquitin-Protein Ligases/genetics , Animals , Apoptosis/drug effects , Binding Sites , Cell Line , Cell Survival/drug effects , Dopaminergic Neurons/cytology , Dopaminergic Neurons/metabolism , Gene Expression Regulation , Homeodomain Proteins/metabolism , Humans , Nucleotide Motifs , Oxidopamine/antagonists & inhibitors , Oxidopamine/toxicity , Pars Compacta/cytology , Pars Compacta/metabolism , Promoter Regions, Genetic , Protein Binding , Rats , Rats, Sprague-Dawley , Signal Transduction , Stereotaxic Techniques , Transcription Factors/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Ubiquitin-Protein Ligases/metabolism
16.
Elife ; 52016 04 13.
Article in English | MEDLINE | ID: mdl-27074662

ABSTRACT

G protein gated inward rectifier K(+) (GIRK) channels open and thereby silence cellular electrical activity when inhibitory G protein coupled receptors (GPCRs) are stimulated. Here we describe an assay to measure neuronal GIRK2 activity as a function of membrane-anchored G protein concentration. Using this assay we show that four Gßγ subunits bind cooperatively to open GIRK2, and that intracellular Na(+) - which enters neurons during action potentials - further amplifies opening mostly by increasing Gßγ affinity. A Na(+) amplification function is characterized and used to estimate the concentration of Gßγ subunits that appear in the membrane of mouse dopamine neurons when GABAB receptors are stimulated. We conclude that GIRK2, through its dual responsiveness to Gßγ and Na(+), mediates a form of neuronal inhibition that is amplifiable in the setting of excess electrical activity.


Subject(s)
Dopaminergic Neurons/metabolism , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , GTP-Binding Protein beta Subunits/metabolism , GTP-Binding Proteins/metabolism , Protein Subunits/metabolism , Receptors, GABA-B/metabolism , Action Potentials/physiology , Animals , Biological Assay , Dopaminergic Neurons/cytology , G Protein-Coupled Inwardly-Rectifying Potassium Channels/genetics , GTP-Binding Protein beta Subunits/genetics , GTP-Binding Proteins/genetics , Gene Expression Regulation , Humans , Mice , Pars Compacta/cytology , Pars Compacta/metabolism , Patch-Clamp Techniques , Phosphatidylethanolamines/chemistry , Phosphatidylethanolamines/metabolism , Phosphatidylglycerols/chemistry , Phosphatidylglycerols/metabolism , Pichia/genetics , Pichia/metabolism , Primary Cell Culture , Protein Multimerization , Protein Subunits/genetics , Proteolipids/chemistry , Proteolipids/metabolism , Receptors, GABA-B/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction , Sodium/metabolism
17.
Neurobiol Aging ; 36(12): 3321-3333, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26433682

ABSTRACT

Age being a risk factor for Parkinson's disease, assessment of age-related changes in the human substantia nigra may elucidate its pathogenesis. Increase in Marinesco bodies, α-synuclein, free radicals and so forth in the aging nigral neurons are clear indicators of neurodegeneration. Here, we report the glial responses in aging human nigra. The glial numbers were determined on Nissl-stained sections. The expression of glial fibrillary acidic protein, S100ß, 2', 3'-cyclic nucleotide 3' phosphodiesterase, and Iba1 was assessed on cryosections of autopsied midbrains by immunohistochemistry and densitometry. The glial counts showed a biphasic increase, of which, the first prominent phase from fetal age to birth could be physiological gliogenesis whereas the second one after middle age may reflect mild age-related gliosis. Astrocytic morphology was altered, but glial fibrillary acidic protein expression increased only mildly. Presence of type-4 microglia suggests possibility of neuroinflammation. Mild reduction in 2', 3'-cyclic nucleotide 3' phosphodiesterase-labeled area denotes subtle demyelination. Stable age-related S100ß expression indicates absence of calcium overload. Against the expected prominent gliosis, subtle age-related morphological alterations in human nigral glia attribute them a participatory role in aging.


Subject(s)
Aging/pathology , Astrocytes/pathology , Microglia/pathology , Nerve Degeneration , Pars Compacta/cytology , Pars Compacta/pathology , 2',3'-Cyclic Nucleotide 3'-Phosphodiesterase/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Astrocytes/metabolism , Calcium/metabolism , Child , Child, Preschool , Female , Glial Fibrillary Acidic Protein/metabolism , Humans , Infant , Male , Microglia/metabolism , Middle Aged , Neurogenic Inflammation , Parkinson Disease , Risk Factors , S100 Calcium Binding Protein beta Subunit/metabolism , Young Adult
18.
Brain Res ; 1622: 240-51, 2015 Oct 05.
Article in English | MEDLINE | ID: mdl-26141374

ABSTRACT

A pathological hallmark of Parkinson׳s disease (PD) is progressive degeneration of nigrostriatal dopamine (NSDA) neurons, which underlies the motor symptoms of PD. While there is severe loss of midbrain NSDA neurons, tuberoinfundibular (TI) DA neurons in the mediobasal hypothalamus (MBH) remain intact. In the present study, confocal microscopic analysis revealed that mitochondrial content and numbers of mitophagosomes were lower in NSDA neuronal cell bodies in the substantia nigra pars compacta (SNpc) compared to TIDA neuronal cell bodies in the arcuate nucleus (ARC) of C57BL/6J male mice. Mitochondrial respiration, mass, membrane potential and morphology were determined using bioenergetic, flow cytometric and transmission electron microscopic analyses of synaptosomes isolated from discrete brain regions containing axon terminals of NSDA and TIDA neurons. Maximum and spare respiratory capacities, and mitochondrial mass were lower in synaptosomal mitochondria derived from the striatum (ST) as compared with the MBH, which correlated with lower numbers of mitochondria per synaptosome in these brain regions. In contrast, there was no regional difference in mitochondrial basal, maximum or spare respirations following inhibition of Complex I activity with rotenone. These results reveal that higher numbers of viable mitochondria are correlated with more extensive autophagic mitochondrial quality maintenance in TIDA neurons as compared with NSDA neurons.


Subject(s)
Arcuate Nucleus of Hypothalamus/metabolism , Autophagy/physiology , Corpus Striatum/metabolism , Dopaminergic Neurons/metabolism , Mitochondria/metabolism , Pars Compacta/metabolism , Animals , Arcuate Nucleus of Hypothalamus/cytology , Cell Respiration/physiology , Corpus Striatum/cytology , Dopaminergic Neurons/cytology , Electron Transport Complex I/antagonists & inhibitors , Electron Transport Complex I/metabolism , Male , Membrane Potentials/physiology , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/ultrastructure , Neural Pathways/cytology , Neural Pathways/metabolism , Pars Compacta/cytology , Rotenone/pharmacology , Synaptosomes/metabolism , Tyrosine 3-Monooxygenase/metabolism , Uncoupling Agents/pharmacology
19.
PLoS One ; 10(7): e0133957, 2015.
Article in English | MEDLINE | ID: mdl-26222442

ABSTRACT

Deep brain stimulation (DBS) is the most common neurosurgical treatment for Parkinson's disease (PD). Whereas the globus pallidus interna (GPi) has been less commonly targeted than the subthalamic nucleus (STN), a recent clinical trial suggests that GPi DBS may provide better outcomes for patients with psychiatric comorbidities. Several laboratories have demonstrated that DBS of the STN provides neuroprotection of substantia nigra pars compacta (SNpc) dopamine neurons in preclinical neurotoxin models of PD and increases brain-derived neurotrophic factor (BDNF). However, whether DBS of the entopeduncular nucleus (EP), the homologous structure to the GPi in the rat, has similar neuroprotective potential in preclinical models has not been investigated. We investigated the impact of EP DBS on forelimb use asymmetry and SNpc degeneration induced by 6-hydroxydopamine (6-OHDA) and on BDNF levels. EP DBS in male rats received unilateral, intrastriatal 6-OHDA and ACTIVE or INACTIVE stimulation continuously for two weeks. Outcome measures included quantification of contralateral forelimb use, stereological assessment of SNpc neurons and BDNF levels. EP DBS 1) did not ameliorate forelimb impairments induced by 6-OHDA, 2) did not provide neuroprotection for SNpc neurons and 3) did not significantly increase BDNF levels in any of the structures examined. These results are in sharp contrast to the functional improvement, neuroprotection and BDNF-enhancing effects of STN DBS under identical experimental parameters in the rat. The lack of functional response to EP DBS suggests that stimulation of the rat EP may not represent an accurate model of clinical GPi stimulation.


Subject(s)
Deep Brain Stimulation , Entopeduncular Nucleus/drug effects , Entopeduncular Nucleus/physiology , Neuroprotection , Oxidopamine/pharmacology , Animals , Behavior, Animal/drug effects , Brain-Derived Neurotrophic Factor/metabolism , Dopaminergic Neurons/cytology , Dopaminergic Neurons/drug effects , Entopeduncular Nucleus/cytology , Entopeduncular Nucleus/metabolism , Male , Neuroprotection/drug effects , Pars Compacta/cytology , Pars Compacta/drug effects , Pars Compacta/physiology , Rats , Rats, Sprague-Dawley
20.
Neuroscience ; 295: 23-38, 2015 Jun 04.
Article in English | MEDLINE | ID: mdl-25796140

ABSTRACT

Exercise reduces the risk of developing a number of neurological disorders and increases the efficiency of cellular energy production. However, overly strenuous exercise produces oxidative stress. Proper oxygenation is crucial for the health of all tissues, and tight regulation of cellular oxygen is critical to balance O2 levels and redox homeostasis in the brain. Hypoxia Inducible Factor (HIF)1α and HIF2α are transcription factors regulated by cellular oxygen concentration that initiate gene regulation of vascular development, redox homeostasis, and cell cycle control. HIF1α and HIF2α contribute to important adaptive mechanisms that occur when oxygen and ROS homeostasis become unbalanced. It has been shown that preconditioning by exposure to a stressor prior to a hypoxic event reduces damage that would otherwise occur. Previously we reported that 3 months of exercise protects SNpc dopaminergic (DA) neurons from toxicity caused by Complex I inhibition. Here, we identify the cells in the SNpc that express HIF1α and HIF2α and show that running exercise produces hypoxia in SNpc DA neurons, and alters the expression of HIF1α and HIF2α. In mice carrying a conditional knockout of Hif1α in postnatal neurons we observe that exercise alone produces SNpc TH+ DA neuron loss. Loss of HIF1α also abolishes exercise-induced neuroprotection. In mice lacking Hif2α in postnatal neurons, the number of TH+ DA neurons in the adult SNpc is diminished, but 3months of exercise rescues this loss. We conclude that HIF1α is necessary for exercise-induced neuroprotection and both HIF1α and HIF2α are necessary for the survival and function of adult SNpc DA neurons.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Dopaminergic Neurons/physiology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , MPTP Poisoning/rehabilitation , Pars Compacta/cytology , Physical Conditioning, Animal/methods , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cell Survival/genetics , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Glial Fibrillary Acidic Protein/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , MPTP Poisoning/chemically induced , MPTP Poisoning/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , RNA, Messenger/metabolism , RNA, Untranslated/genetics , RNA, Untranslated/metabolism , Tyrosine 3-Monooxygenase/metabolism , beta-Galactosidase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...