Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Behav Brain Res ; 363: 161-172, 2019 05 02.
Article in English | MEDLINE | ID: mdl-30735759

ABSTRACT

Parkinson's disease (PD) is characterized by motor impairments and several non-motor features, including frequent depression and anxiety. Stress-induced deficits of adult hippocampal neurogenesis (AHN) have been linked with abnormal affective behavior in animals. It has been speculated that AHN defects may contribute to affective symptoms in PD, but this hypothesis remains insufficiently tested in animal models. Mice that lack the PD-linked kinase PINK1 show impaired differentiation of adult-born neurons in the hippocampus. Here, we examined the relationship between AHN deficits and affective behavior in PINK1-/- mice under basal (no stress) conditions and after exposure to chronic stress. PINK1 loss and corticosterone negatively and jointly affected AHN, leading to lower numbers of neural stem cells and newborn neurons in the dentate gyrus of corticosterone-treated PINK1-/- mice. Despite increased basal AHN deficits, PINK1-deficient mice showed normal affective behavior. However, lack of PINK1 sensitized mice to corticosterone-induced behavioral despair in the tail suspension test at a dose where wildtype mice were unaffected. Moreover, after two weeks of chronic restraint stress male PINK1-/- mice displayed increased immobility in the forced swim test, and protein expression of the glucocorticoid receptor in the hippocampus was reduced. Thus, while impaired AHN as such is insufficient to cause affective dysfunction in this PD model, PINK1 deficiency may lower the threshold for chronic stress-induced depression in PD. Finally, PINK1-deficient mice displayed reduced basal voluntary wheel running but normal rotarod performance, a finding whose mechanisms remain to be determined.


Subject(s)
Depression/physiopathology , Neurogenesis/physiology , Protein Kinases/physiology , Animals , Anxiety/physiopathology , Anxiety Disorders/physiopathology , Behavior, Animal , Cell Differentiation , Cell Proliferation , Corticosterone/metabolism , Dentate Gyrus/metabolism , Depression/drug therapy , Depression/metabolism , Depressive Disorder/physiopathology , Disease Models, Animal , Hippocampus/metabolism , Hippocampus/physiology , Hypothalamo-Hypophyseal System , Male , Mice , Mice, Inbred C57BL , Motor Activity , Neurons/metabolism , Parkinson Disease/physiopathology , Pituitary-Adrenal System , Protein Kinases/genetics , Receptors, Glucocorticoid/metabolism , Stress, Psychological/metabolism , Stress, Psychological/physiopathology , Swimming , Temporal Lobe/physiopathology
2.
Curr Neuropharmacol ; 17(6): 550-562, 2019.
Article in English | MEDLINE | ID: mdl-29512466

ABSTRACT

BACKGROUND: Depression is a widespread phenomenon with varying degrees of pathology in different patients. Various hypotheses have been proposed for the cause and continuance of depression. Some of these include, but not limited to, the monoamine hypothesis, the neuroendocrine hypothesis, and the more recent epigenetic and inflammatory hypotheses. OBJECTIVE: In this article, we review all the above hypotheses with a focus on the role of mitochondria as the connecting link. Oxidative stress, respiratory activity, mitochondrial dynamics and metabolism are some of the mitochondria-dependent factors which are affected during depression. We also propose exogenous ATP as a contributing factor to depression. RESULT: Literature review shows that pro-inflammatory markers are elevated in depressive individuals. The cause for elevated levels of cytokines in depression is not completely understood. We propose exogenous ATP activates purinergic receptors which in turn increase the levels of various proinflammatory factors in the pathophysiology of depression. CONCLUSION: Mitochondria are integral to the function of neurons and undergo dysfunction in major depressive disorder patients. This dysfunction is reflected in all the various hypotheses that have been proposed for depression. Among the newer targets identified, which also involve mitochondria, includes the role of exogenous ATP. The diversity of purinergic receptors, and their differential expression among various individuals in the population, due to genetic and environmental (prenatal) influences, may influence the susceptibility and severity of depression. Identifying specific receptors involved and using patient-specific purinergic receptor antagonist may be an appropriate therapeutic course in the future.


Subject(s)
Depressive Disorder, Major/etiology , Mitochondria/metabolism , Animals , Brain/metabolism , Brain/pathology , Depressive Disorder, Major/genetics , Depressive Disorder, Major/metabolism , Depressive Disorder, Major/pathology , Epigenesis, Genetic , Humans , Hypothalamo-Hypophyseal System/metabolism , Inflammation/metabolism , Inflammation/pathology , Mitochondria/genetics , Mitochondria/pathology , Monoamine Oxidase/metabolism , Nerve Growth Factors/metabolism , Receptors, Glutamate/metabolism , Receptors, Purinergic/metabolism
3.
Neurodegener Dis ; 12(3): 136-49, 2013.
Article in English | MEDLINE | ID: mdl-23295771

ABSTRACT

BACKGROUND: Mutations in PTEN-induced kinase 1 (PINK1) cause early-onset recessive parkinsonism. PINK1 and Parkin regulate mitochondrial quality control. However, PINK1 ablation in Drosophila and cultured mammalian cell lines affected mitochondrial function/dynamics in opposite ways, confounding the elucidation of the role of PINK1 in these processes. OBJECTIVE: We recently generated PINK1-deficient (PINK1-/-) mice and reasoned that primary cells from these mice provide a more physiological substrate to study the role of PINK1 in mammals and to investigate metabolic adaptations and neuron-specific vulnerability in PINK1 deficiency. METHODS AND RESULTS: Using real-time measurement of oxygen consumption and extracellular acidification, we show that basal mitochondrial respiration is increased, while maximum respiration and spare respiratory capacity are decreased in PINK1-/- mouse embryonic fibroblasts (MEF), as is the membrane potential. In addition, a Warburg-like effect in PINK1-/- MEF promotes survival that is abrogated by inhibition of glycolysis. Expression of uncoupling protein-2 is decreased in PINK1-/- MEF and the striatum of PINK1-/- mice, possibly increasing the sensitivity to oxidative stress. Mitochondria accumulate in large foci in PINK1-/- MEF, indicative of abnormal mitochondrial dynamics and/or transport. Like in PINK1-/- Drosophila, enlarged/swollen mitochondria accumulate in three different cell types from PINK1-/- mice (MEF, primary cortical neurons and embryonic stem cells). However, mitochondrial enlargement is greatest and most prominent in primary cortical neurons that also develop cristae fragmentation and disintegration. CONCLUSION: Our results reveal mechanisms of PINK1-related parkinsonism, show that the function of PINK1 is conserved between Drosophila and mammals when studied in primary cells, and demonstrate that the same PINK1 mutation can affect mitochondrial morphology/degeneration in a cell type-specific manner, suggesting that tissue-/cell-specific metabolic capacity and adaptations determine phenotypes and cellular vulnerability in PINK1-/- mice and cells.


Subject(s)
Mitochondria/metabolism , Mitochondria/ultrastructure , Neurons/metabolism , Protein Kinases/genetics , Adaptation, Physiological , Animals , Cells, Cultured , Mice , Mice, Knockout , Primary Cell Culture
4.
Neurobiol Dis ; 45(1): 469-78, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21945539

ABSTRACT

Mutations in the PARK6 gene coding for PTEN-induced kinase 1 (PINK1) cause recessive early-onset Parkinsonism. Although PINK1 and Parkin promote the degradation of depolarized mitochondria in cultured cells, little is known about changes in signaling pathways that may additionally contribute to dopamine neuron loss in recessive Parkinsonism. Accumulating evidence implicates impaired Akt cell survival signaling in sporadic and familial PD (PD). IGF-1/Akt signaling inhibits dopamine neuron loss in several animal models of PD and both IGF-1 and insulin are neuroprotective in various settings. Here, we tested whether PINK1 is required for insulin-like growth factor 1 (IGF-1) and insulin dependent phosphorylation of Akt and the regulation of downstream Akt target proteins. Our results show that embryonic fibroblasts from PINK1-deficient mice display significantly reduced Akt phosphorylation in response to both IGF-1 and insulin. Moreover, phosphorylation of glycogen synthase kinase-3ß (GSK-3ß) and nuclear exclusion of FoxO1 are decreased in IGF-1 treated PINK1-deficient cells. In addition, phosphorylation of ribosomal protein S6 is reduced indicating decreased activity of mitochondrial target of rapamycin (mTOR) in IGF-1 treated PINK1(-/-) cells. Importantly, the protection afforded by IGF-1 against staurosporine-induced metabolic dysfunction and apoptosis is abrogated in PINK1-deficient cells. Moreover, IGF-1-induced Akt phosphorylation is impaired in primary cortical neurons from PINK1-deficient mice. Inhibition of cellular Ser/Thr phosphatases did not increase the amount of phosphorylated Akt in PINK1(-/-) cells, suggesting that components upstream of Akt phosphorylation are compromised in PINK1-deficient cells. Our studies show that PINK1 is required for optimal IGF-1 and insulin dependent Akt signal transduction, and raise the possibility that impaired IGF-1/Akt signaling is involved in PINK1-related Parkinsonism by increasing the vulnerability of dopaminergic neurons to stress-induced cell death.


Subject(s)
Apoptosis/physiology , Insulin-Like Growth Factor I/metabolism , Protein Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/physiology , Animals , Apoptosis/drug effects , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Forkhead Box Protein O1 , Forkhead Transcription Factors/metabolism , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Insulin/metabolism , Insulin/pharmacology , Insulin-Like Growth Factor I/pharmacology , Mice , Mice, Knockout , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Phosphorylation/physiology , Protein Kinases/genetics , Ribosomal Protein S6/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism
5.
PLoS One ; 6(1): e16038, 2011 Jan 13.
Article in English | MEDLINE | ID: mdl-21249202

ABSTRACT

BACKGROUND: PTEN-induced kinase 1 (PINK1) is linked to recessive Parkinsonism (EOPD). Pink1 deletion results in impaired dopamine (DA) release and decreased mitochondrial respiration in the striatum of mice. To reveal additional mechanisms of Pink1-related dopaminergic dysfunction, we studied Ca²+ vulnerability of purified brain mitochondria, DA levels and metabolism and whether signaling pathways implicated in Parkinson's disease (PD) display altered activity in the nigrostriatal system of Pink1⁻/⁻ mice. METHODS AND FINDINGS: Purified brain mitochondria of Pink1⁻/⁻ mice showed impaired Ca²+ storage capacity, resulting in increased Ca²+ induced mitochondrial permeability transition (mPT) that was rescued by cyclosporine A. A subpopulation of neurons in the substantia nigra of Pink1⁻/⁻ mice accumulated phospho-c-Jun, showing that Jun N-terminal kinase (JNK) activity is increased. Pink1⁻/⁻ mice 6 months and older displayed reduced DA levels associated with increased DA turnover. Moreover, Pink1⁻/⁻ mice had increased levels of IL-1ß, IL-12 and IL-10 in the striatum after peripheral challenge with lipopolysaccharide (LPS), and Pink1⁻/⁻ embryonic fibroblasts showed decreased basal and inflammatory cytokine-induced nuclear factor kappa-ß (NF-κB) activity. Quantitative transcriptional profiling in the striatum revealed that Pink1⁻/⁻ mice differentially express genes that (i) are upregulated in animals with experimentally induced dopaminergic lesions, (ii) regulate innate immune responses and/or apoptosis and (iii) promote axonal regeneration and sprouting. CONCLUSIONS: Increased mitochondrial Ca²+ sensitivity and JNK activity are early defects in Pink1⁻/⁻ mice that precede reduced DA levels and abnormal DA homeostasis and may contribute to neuronal dysfunction in familial PD. Differential gene expression in the nigrostriatal system of Pink1⁻/⁻ mice supports early dopaminergic dysfunction and shows that Pink1 deletion causes aberrant expression of genes that regulate innate immune responses. While some differentially expressed genes may mitigate neurodegeneration, increased LPS-induced brain cytokine expression and impaired cytokine-induced NF-κB activation may predispose neurons of Pink1⁻/⁻ mice to inflammation and injury-induced cell death.


Subject(s)
Calcium/metabolism , Dopamine/deficiency , Gene Expression Regulation , Immunity, Innate/genetics , Mitochondria/metabolism , Protein Kinases/metabolism , Animals , Brain/metabolism , Brain/ultrastructure , Corpus Striatum , Inflammation/etiology , JNK Mitogen-Activated Protein Kinases/metabolism , Mice , Mice, Knockout , Mitochondrial Membrane Transport Proteins , Mitochondrial Permeability Transition Pore , NF-kappa B/metabolism , Neurons/pathology , Parkinson Disease/genetics , Parkinson Disease/immunology , Parkinson Disease/pathology , Protein Kinases/deficiency
6.
J Neuroimmunol ; 174(1-2): 39-51, 2006 May.
Article in English | MEDLINE | ID: mdl-16529823

ABSTRACT

Inhibition of neuronal cyclooxygenase-2 (COX-2) and hence prostaglandin E2 (PGE2) synthesis by non-steroidal anti-inflammatory drugs has been suggested to protect neuronal cells in a variety of pathophysiological situations including Alzheimer's disease and ischemic stroke. Ascorbic acid (vitamin C) has also been shown to protect cerebral tissue in a variety of experimental conditions, which has been attributed to its antioxidant capacity. In the present study, we show that ascorbic acid dose-dependently inhibited interleukin-1beta (IL-1beta)-mediated PGE2 synthesis in the human neuronal cell line, SK-N-SH. Furthermore, in combination with aspirin, ascorbic acid augmented the inhibitory effect of aspirin on PGE2 synthesis. However, ascorbic acid had no synergistic effect along with other COX inhibitors (SC-58125 and indomethacin). The inhibition of IL-1beta-mediated PGE2 synthesis by ascorbic acid was not due to the inhibition of the expression of COX-2 or microsomal prostaglandin E synthase (mPGES-1). Rather, ascorbic acid dose-dependently (0.1-100 microM) produced a significant reduction in IL-1beta-mediated production of 8-iso-prostaglandin F2alpha (8-iso-PGF2alpha), a reliable indicator of free radical formation, suggesting that the effects of ascorbic acid on COX-2-mediated PGE2 biosynthesis may be the result of the maintenance of the neuronal redox status since COX activity is known to be enhanced by oxidative stress. Our results provide in vitro evidence that the neuroprotective effects of ascorbic acid may depend, at least in part, on its ability to reduce neuronal COX-2 activity and PGE2 synthesis, owing to its antioxidant properties. Further, these experiments suggest that a combination of aspirin with ascorbic acid constitutes a novel approach to render COX-2 more sensitive to inhibition by aspirin, allowing an anti-inflammatory therapy with lower doses of aspirin, thereby avoiding the side effects of the usually high dose aspirin treatment.


Subject(s)
Antioxidants/pharmacology , Ascorbic Acid/pharmacology , Aspirin/pharmacology , Cyclooxygenase 2/metabolism , Dinoprostone/metabolism , Neurons/drug effects , Blotting, Western/methods , Cell Line, Tumor , Dinoprost/analogs & derivatives , Dinoprost/metabolism , Dinoprostone/antagonists & inhibitors , Dose-Response Relationship, Drug , Drug Interactions , Gene Expression/drug effects , Humans , Interleukin-1/pharmacology , Neural Inhibition/drug effects , Neuroblastoma
7.
Neurochem Int ; 46(6): 501-12, 2005 May.
Article in English | MEDLINE | ID: mdl-15769552

ABSTRACT

Adenosine binds to a class of G-protein coupled receptors, which are further distinguished as A(1), A(2a), A(2b) and A(3) adenosine receptors. As we have shown earlier, the stable adenosine analogue NECA (N6-(R)-phenylisopropyladenosine) stimulates IL-6 expression in the human astrocytoma cell line U373 MG via the A(2b) receptor. The mechanism by which NECA promotes astrocytic IL-6 expression has not been identified. By using various inhibitors of signal transduction, we found that p38 mitogen-activated protein kinases (MAPK) activation (inhibitor SB202190), but not extracellular signal-regulated kinase (ERK) (PD98059) and c-jun N-terminal kinase (JNK)(SP600125), is essential in the NECA-induced signalling cascade that leads to the increase in IL-6 synthesis in U373 MG cells. Results obtained with protein kinase C (PKC) inhibitors that have different substrate specificities, indicated that the PKC delta and epsilon isoforms are also involved in adenosine receptor A(2b) dependent upregulation of IL-6 expression. This is supported by the fact that NECA induced the activation of PKC delta and epsilon in U373 MG cells.


Subject(s)
Adenosine/metabolism , Astrocytoma/metabolism , Interleukin-6/metabolism , Protein Kinase C/metabolism , Receptor, Adenosine A2B/metabolism , Signal Transduction/physiology , p38 Mitogen-Activated Protein Kinases/metabolism , Adenosine A2 Receptor Agonists , Cell Line, Tumor , Enzyme Inhibitors/pharmacology , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , JNK Mitogen-Activated Protein Kinases/genetics , JNK Mitogen-Activated Protein Kinases/metabolism , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/genetics , Protein Kinase C-delta , Protein Kinase C-epsilon , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Signal Transduction/drug effects , Up-Regulation/drug effects , Up-Regulation/physiology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...