Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
J Cell Biochem ; 119(1): 347-357, 2018 01.
Article in English | MEDLINE | ID: mdl-28585712

ABSTRACT

The executioner caspase-3 has been proposed as a pharmacological intervention target to preserve degenerating dopaminergic (DA) neurons because apoptotic mechanisms involving caspase-3 contribute, at least in part, to the loss of DA neurons in patients and experimental models of Parkinson's disease (PD). Here, we determined that genetic intervention of caspase-3 was sufficient to prevent cell death against oxidative stress (OS), accompanied by unexpected severe mitochondrial dysfunction. Specifically, as we expected, caspase-3-deficient DA neuronal cells were very significantly resistant to OS-induced cell death, while the activation of the initiator caspase-9 by OS was preserved. Moreover, detailed phenotypic characterization of caspase-3-deficient DA cells revealed severe mitochondrial dysfunction, including an accumulation of damaged mitochondria with a characteristic swollen structure and broken cristae, reduced membrane potential, increased levels of reactive oxygen species (ROS), and deficits in mitochondrial oxidative phosphorylation (OXPHOS) enzymes. Of great interest, we found that mitochondrial biogenesis was dramatically decreased in caspase-3-deficient DA cells, whereas their capability of mitophagy was normal. In accordance with this observation, caspase-3 gene knock down (KD) resulted in dramatically decreased expression of the key transcriptional activators of mitochondrial biogenesis, such as Tfam and Nrf-1, implicating a non-apoptotic role of procaspase-3 in mitochondrial biogenesis. Therefore, a prolonged anti-apoptotic intervention targeting caspase-3 should be considered with caution due to the potential adverse effects in mitochondria dynamics resulting from a novel potential functional role of procaspase-3 in mitochondrial biogenesis via regulating the expression of mitochondrial biogenesis activators. J. Cell. Biochem. 119: 347-357, 2018. © 2017 Wiley Periodicals, Inc.


Subject(s)
Apoptosis , Caspase 3/metabolism , Mitochondria/metabolism , Neurons/metabolism , Caspase 3/genetics , Cell Line , Humans , Mitochondria/genetics , Parkinson Disease/genetics , Parkinson Disease/metabolism
2.
PLoS Genet ; 13(8): e1006975, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28827794

ABSTRACT

DJ-1 is one of the causative genes for early onset familiar Parkinson's disease (PD) and is also considered to influence the pathogenesis of sporadic PD. DJ-1 has various physiological functions which converge on controlling intracellular reactive oxygen species (ROS) levels. In RNA-sequencing analyses searching for novel anti-oxidant genes downstream of DJ-1, a gene encoding NADP+-dependent isocitrate dehydrogenase (IDH), which converts isocitrate into α-ketoglutarate, was detected. Loss of IDH induced hyper-sensitivity to oxidative stress accompanying age-dependent mitochondrial defects and dopaminergic (DA) neuron degeneration in Drosophila, indicating its critical roles in maintaining mitochondrial integrity and DA neuron survival. Further genetic analysis suggested that DJ-1 controls IDH gene expression through nuclear factor-E2-related factor2 (Nrf2). Using Drosophila and mammalian DA models, we found that IDH suppresses intracellular and mitochondrial ROS level and subsequent DA neuron loss downstream of DJ-1. Consistently, trimethyl isocitrate (TIC), a cell permeable isocitrate, protected mammalian DJ-1 null DA cells from oxidative stress in an IDH-dependent manner. These results suggest that isocitrate and its derivatives are novel treatments for PD associated with DJ-1 dysfunction.


Subject(s)
Drosophila Proteins/genetics , Isocitrate Dehydrogenase/genetics , Nerve Degeneration/genetics , Nerve Tissue Proteins/genetics , Parkinson Disease/genetics , Animals , Disease Models, Animal , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Drosophila melanogaster/genetics , Gene Expression Regulation , High-Throughput Nucleotide Sequencing , Humans , Isocitrates/metabolism , Mitochondria/genetics , Mitochondria/pathology , NADP/genetics , NF-E2 Transcription Factor/genetics , Nerve Degeneration/physiopathology , Oxidative Stress/genetics , Parkinson Disease/pathology
3.
Sci Rep ; 7(1): 6248, 2017 07 24.
Article in English | MEDLINE | ID: mdl-28740220

ABSTRACT

Defective hepatic autophagy is observed in obesity and diabetes, whereas autophagy is inhibited by insulin in hepatocytes. Insulin-induced anti-autophagy is mediated by non-canonical Gαi3 signaling via an unknown mechanism. Previously, we identified the anti-autophagic activity of Tnfaip8 via activation of mammalian target of rapamycin (mTOR) in the nervous system. Here, we demonstrate that insulin temporally induces Tnfaip8, which mediates the anti-autophagic action of insulin through formation of a novel ternary complex including Tnfaip8, phosphatidylethanolamine (PE) and Gαi3. Specifically, an X-ray crystallographic study of Tnfaip8 from Mus musculus (mTnfaip8) at 2.03 Å together with LC-MS analyses reveals PE in the hydrophobic cavity. However, an mTnfaip8 mutant lacking PE does not interact with Gαi3, indicating that the PE component is critical for the anti-autophagic action of mTnfaip8 via interaction with Gαi3. Therefore, the mTnfaip8-PE complex may act as an essential upstream effector via ternary complex formation most likely with active Gαi3 during insulin-induced anti-autophagy.


Subject(s)
Apoptosis Regulatory Proteins/chemistry , Apoptosis Regulatory Proteins/metabolism , Autophagy , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Insulin/metabolism , Phosphatidylethanolamines/metabolism , Amino Acid Sequence , Animals , Crystallography, X-Ray , GTP-Binding Protein alpha Subunits, Gi-Go/chemistry , Mice , Phosphatidylethanolamines/chemistry , Protein Binding , Protein Conformation , Sequence Homology
4.
J Biol Chem ; 290(16): 10325-35, 2015 Apr 17.
Article in English | MEDLINE | ID: mdl-25716315

ABSTRACT

The failure to trigger mitophagy is implicated in the pathogenesis of familial Parkinson disease that is caused by PINK1 or Parkin mutations. According to the prevailing PINK1-Parkin signaling model, mitophagy is promoted by the mitochondrial translocation of Parkin, an essential PINK1-dependent step that occurs via a previously unknown mechanism. Here we determined that critical concentrations of NO was sufficient to induce the mitochondrial translocation of Parkin even in PINK1 deficiency, with apparent increased interaction of full-length PINK1 accumulated during mitophagy, with neuronal nitric oxide synthase (nNOS). Specifically, optimum levels of NO enabled PINK1-null dopaminergic neuronal cells to regain the mitochondrial translocation of Parkin, which appeared to be significantly suppressed by nNOS-null mutation. Moreover, nNOS-null mutation resulted in the same mitochondrial electron transport chain (ETC) enzyme deficits as PINK1-null mutation. The involvement of mitochondrial nNOS activation in mitophagy was further confirmed by the greatly increased interactions of full-length PINK1 with nNOS, accompanied by mitochondrial accumulation of phospho-nNOS (Ser(1412)) during mitophagy. Of great interest is that the L347P PINK1 mutant failed to bind to nNOS. The loss of nNOS phosphorylation and Parkin accumulation on PINK1-deficient mitochondria could be reversed in a PINK1-dependent manner. Finally, non-toxic levels of NO treatment aided in the recovery of PINK1-null dopaminergic neuronal cells from mitochondrial ETC enzyme deficits. In summary, we demonstrated the full-length PINK1-dependent recruitment of nNOS, its activation in the induction of Parkin translocation, and the feasibility of NO-based pharmacotherapy for defective mitophagy and ETC enzyme deficits in Parkinson disease.


Subject(s)
Dopaminergic Neurons/metabolism , Mitochondria/metabolism , Mitophagy/genetics , Nitric Oxide Synthase Type I/genetics , Protein Kinases/genetics , Ubiquitin-Protein Ligases/genetics , Animals , Disease Models, Animal , Dopaminergic Neurons/pathology , Electron Transport , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Expression Regulation , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide/biosynthesis , Nitric Oxide Synthase Type I/deficiency , Parkinson Disease/genetics , Parkinson Disease/metabolism , Parkinson Disease/pathology , Primary Cell Culture , Protein Binding , Protein Kinases/deficiency , Protein Transport , Signal Transduction , Ubiquitin-Protein Ligases/metabolism
5.
Exp Neurobiol ; 23(4): 345-51, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25548534

ABSTRACT

Mitochondria are small organelles that produce the majority of cellular energy as ATP. Mitochondrial dysfunction has been implicated in the pathogenesis of Parkinson's disease (PD), and rare familial forms of PD provide valuable insight into the pathogenic mechanism underlying mitochondrial impairment, even though the majority of PD cases are sporadic. The regulation of mitochondria is crucial for the maintenance of energy-demanding neuronal functions in the brain. Mitochondrial biogenesis and mitophagic degradation are the major regulatory pathways that preserve optimal mitochondrial content, structure and function. In this mini-review, we provide an overview of the mitochondrial quality control mechanisms, emphasizing regulatory molecules in mitophagy and biogenesis that specifically interact with the protein products of three major recessive familial PD genes, PINK1, Parkin and DJ-1.

6.
Autophagy ; 10(11): 1906-20, 2014.
Article in English | MEDLINE | ID: mdl-25483962

ABSTRACT

CHDH (choline dehydrogenase) is an enzyme catalyzing the dehydrogenation of choline to betaine aldehyde in mitochondria. Apart from this well-known activity, we report here a pivotal role of CHDH in mitophagy. Knockdown of CHDH expression impairs CCCP-induced mitophagy and PARK2/parkin-mediated clearance of mitochondria in mammalian cells, including HeLa cells and SN4741 dopaminergic neuronal cells. Conversely, overexpression of CHDH accelerates PARK2-mediated mitophagy. CHDH is found on both the outer and inner membranes of mitochondria in resting cells. Interestingly, upon induction of mitophagy, CHDH accumulates on the outer membrane in a mitochondrial potential-dependent manner. We found that CHDH is not a substrate of PARK2 but interacts with SQSTM1 independently of PARK2 to recruit SQSTM1 into depolarized mitochondria. The FB1 domain of CHDH is exposed to the cytosol and is required for the interaction with SQSTM1, and overexpression of the FB1 domain only in cytosol reduces CCCP-induced mitochondrial degradation via competitive interaction with SQSTM1. In addition, CHDH, but not the CHDH FB1 deletion mutant, forms a ternary protein complex with SQSTM1 and MAP1LC3 (LC3), leading to loading of LC3 onto the damaged mitochondria via SQSTM1. Further, CHDH is crucial to the mitophagy induced by MPP+ in SN4741 cells. Overall, our results suggest that CHDH is required for PARK2-mediated mitophagy for the recruitment of SQSTM1 and LC3 onto the mitochondria for cargo recognition.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Choline Dehydrogenase/metabolism , Microtubule-Associated Proteins/metabolism , Mitophagy , Animals , Cell Line, Tumor , Chromatography, Liquid , Cytosol/metabolism , DNA, Mitochondrial/metabolism , Dopamine/chemistry , Endopeptidase K/metabolism , Flow Cytometry , Gene Deletion , Green Fluorescent Proteins/metabolism , HEK293 Cells , HeLa Cells , Humans , Mass Spectrometry , Mitochondria/metabolism , Neurons/metabolism , Protein Binding , RNA, Small Interfering/metabolism , Sequestosome-1 Protein , Ubiquitin-Protein Ligases/metabolism
7.
Int J Mol Sci ; 15(3): 4523-30, 2014 Mar 14.
Article in English | MEDLINE | ID: mdl-24637935

ABSTRACT

Tnfaip8/oxidative stress regulated gene-α (Oxi-α) is a novel protein expressed specifically in brain dopaminergic neurons and its over-expression has been reported to protect dopaminergic cells against OS-induced cell death. In this study, murine C165S mutant Tnfaip8/Oxi-α has been crystallized and X-ray data have been collected to 1.8 Å using synchrotron radiation. The crystal belonged to the primitive orthorhombic space group P21212, with unit-cell parameters a = 66.9, b = 72.3, c = 93.5 Å. A full structural determination is under way in order to provide insights into the structure-function relationships of this protein.


Subject(s)
Apoptosis Regulatory Proteins/chemistry , Apoptosis Regulatory Proteins/genetics , Mutation, Missense , Animals , Apoptosis Regulatory Proteins/metabolism , Crystallization , Crystallography, X-Ray , Electrophoresis, Polyacrylamide Gel , Mice , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutant Proteins/metabolism , Synchrotrons
8.
Neurosci Lett ; 561: 101-6, 2014 Feb 21.
Article in English | MEDLINE | ID: mdl-24397909

ABSTRACT

Abnormal autophagy is frequently observed during dopaminergic neurodegeneration in Parkinson's disease (PD). However, it is not yet firmly established whether active autophagy is beneficial or pathogenic with respect to dopaminergic cell loss. Staurosporine, a common inducer of apoptosis, is often used in mechanistic studies of dopaminergic cell death. Here we report that staurosporine activates both autophagy and mitophagy simultaneously during dopaminergic neuronal cell death, and evaluate the physiological significance of these processes during cell death. First, staurosporine treatment resulted in induction of autophagy in more than 75% of apoptotic cells. Pharmacological inhibition of autophagy by bafilomycin A1 decreased significantly cell viability. In addition, staurosporine treatment resulted in activation of the PINK1-Parkin mitophagy pathway, of which deficit underlies some familial cases of PD, in the dopaminergic neuronal cell line, SN4741. The genetic blockade of this pathway by PINK1 null mutation also dramatically increased staurosporine-induced cell death. Taken together, our data suggest that staurosporine induces both mitophagy and autophagy, and that these pathways exert a significant neuroprotective effect, rather than a contribution to autophagic cell death. This model system may therefore be useful for elucidating the mechanisms underlying crosstalk between autophagy, mitophagy, and cell death in dopaminergic neurons.


Subject(s)
Apoptosis/drug effects , Autophagy/drug effects , Dopaminergic Neurons/drug effects , Mitophagy/drug effects , Staurosporine/pharmacology , Animals , Cell Line , Dopaminergic Neurons/cytology , Mice , Protein Kinases/genetics
9.
J Neurochem ; 129(3): 527-38, 2014 May.
Article in English | MEDLINE | ID: mdl-24444419

ABSTRACT

Abnormal autophagy may contribute to neurodegeneration in Parkinson's disease (PD). However, it is largely unknown how autophagy is dysregulated by oxidative stress (OS), one of major pathogenic causes of PD. We recently discovered the potential autophagy regulator gene family including Tnfaip8/Oxi-α, which is a mammalian target of rapamycin (mTOR) activator down-regulated by OS in dopaminergic neurons (J. Neurochem., 112, 2010, 366). Here, we demonstrate that the OS-induced Tnfaip8 l1/Oxi-ß could increase autophagy by a unique mechanism that increases the stability of tuberous sclerosis complex 2 (TSC2), a critical negative regulator of mTOR. Tnfaip8 l1/Oxi-ß and Tnfaip8/Oxi-α are the novel regulators of mTOR acting in opposition in dopaminergic (DA) neurons. Specifically, 6-hydroxydopamine (6-OHDA) treatment up-regulated Tnfaip8 l1/Oxi-ß in DA neurons, thus inducing autophagy, while knockdown of Tnfaip8 l1/Oxi-ß prevented significantly activation of autophagic markers by 6-OHDA. FBXW5 was identified as a novel binding protein for Tnfaip8 l1/Oxi-ß. FBXW5 is a TSC2 binding receptor within CUL4 E3 ligase complex, and it promotes proteasomal degradation of TSC2. Thus, Tnfaip8 l1/Oxi-ß competes with TSC2 to bind FBXW5, increasing TSC2 stability by preventing its ubiquitination. Our data show that the OS-induced Tnfaip8 l1/Oxi-ß stabilizes TSC2 protein, decreases mTOR phosphorylation, and enhances autophagy. Therefore, altered regulation of Tnfaip8 l1/Oxi-ß may contribute significantly to dysregulated autophagy observed in dopaminergic neurons under pathogenic OS condition. Dysfunctional autophagy is frequently observed in post-mortem brains of patients and animal models of Parkinson's disease. In dopaminergic neurons of the 6-hydroxydopamine (6-OHDA) model, oxidative stress induces Tnfaip8 l1/Oxi-ß, which results in increased autophagy by its exclusive binding with FBXW5 to stabilize TSC2. Thus, altered regulation of Tnfaip8 l1/Oxi-ß may contribute to dysregulated autophagy in dopaminergic neurons under pathogenic oxidative stress, implicating both Oxi-ß and FBXW5 as potential intervention targets for dysfunctional autophagy in dopaminergic neurons under oxidative stress.


Subject(s)
Autophagy , Dopaminergic Neurons/metabolism , F-Box Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Parkinsonian Disorders/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Blotting, Western , Cell Line , Disease Models, Animal , Gene Knockdown Techniques , Humans , Immunoblotting , Immunohistochemistry , Immunoprecipitation , Microscopy, Confocal , Molecular Sequence Data , Oligonucleotide Array Sequence Analysis , Oxidative Stress/physiology , Protein Binding , Rats , Transfection , Tuberous Sclerosis Complex 2 Protein
10.
J Control Release ; 175: 10-6, 2014 Feb 10.
Article in English | MEDLINE | ID: mdl-24333627

ABSTRACT

Cell-based drug delivery systems (DDSs) have been increasingly exploited because cells can be utilized as a continuous drug delivering system to produce therapeutic molecules over a more extended period compared to the simple drug carriers. Although hydrogels have many advantages for this application, their mechanical properties are generally not desirable to structurally protect implanted cells. Here, we present a three-dimensional (3D) hybrid scaffold with a combination of a 3D framework and a hydrogel to enhance the mechanical properties without chemically altering the transport properties of the hydrogel. Based on the 3D Ormocomp scaffold (framework) fabricated by projection-based microstereolithography with defined parameters, we developed a 3D hybrid scaffold by injection of the mixture of cells and the alginate gel into the internal space of the framework. This hybrid scaffold showed the improved mechanical strength and the framework in the scaffold played the role of an adhesion site for the encapsulated cells during the culture period. Additionally, we confirmed its protection of exogenous human cells from acute immune rejection in a mouse model. Eventually, we demonstrated the feasibility of applying this hybrid scaffold to the treatment of Parkinson's disease as a cell-based DDS. Dopamine released from the 3D hybrid scaffolds encapsulating dopamine-secreting cells for 8weeks suggested its clinical applicability. Further study on its long-term efficacy is necessary for the clinical applicability of this 3D hybrid scaffold for the treatment of Parkinson's disease.


Subject(s)
Dopamine Agents/administration & dosage , Dopamine/administration & dosage , Drug Delivery Systems/methods , Hydrogel, Polyethylene Glycol Dimethacrylate/chemistry , Tissue Scaffolds/chemistry , Alginates/chemistry , Animals , Cells, Cultured , Female , Glucuronic Acid/chemistry , Hexuronic Acids/chemistry , Humans , Mice , Mice, Inbred C57BL , Parkinson Disease/drug therapy
11.
J Neurochem ; 2013 Dec 26.
Article in English | MEDLINE | ID: mdl-24372178

ABSTRACT

Abnormal autophagy may contribute to neurodegeneration in Parkinson's disease (PD). However, it is largely unknown how autophagy is dysregulated by oxidative stress (OS), one of major pathogenic causes of PD. We recently discovered the potential autophagy regulator gene family including Tnfaip8/Oxi-α, which is an mTOR activator downregulated by OS in dopaminergic neurons (Choi et al., 2010). Here we demonstrate that the OS-induced Tnfaip8l1/Oxi-ß could increase autophagy by a unique mechanism that increases the stability of TSC2, a critical negative regulator of mTOR. Tnfaip8l1/Oxi-ß and Tnfaip8/Oxi-α are the novel regulators of mTOR acting in opposition in DA neurons. Specifically, 6-hydroxydopamine (6-OHDA) treatment upregulated Tnfaip8l1/Oxi-ß in DA neurons, thus inducing autophagy, while knockdown of Tnfaip8l1/Oxi-ß prevented significantly activation of autophagic markers by 6-OHDA. FBXW5 was identified as a novel binding protein for Tnfaip8l1/Oxi-ß. FBXW5 is a TSC2 binding receptor within CUL4 E3 ligase complex, and it promotes proteasomal degradation of TSC2. Thus, Tnfaip8l1/Oxi-ß competes with TSC2 to bind FBXW5, increasing TSC2 stability by preventing its ubiquitination. Our data show that the OS-induced Tnfaip8l1/Oxi-ß stabilizes TSC2 protein, decreases mTOR phosphorylation and enhances autophagy. Therefore, altered regulation of Tnfaip8l1/Oxi-ß may contribute significantly to dysregulated autophagy observed in dopaminergic neurons under pathogenic OS condition. This article is protected by copyright. All rights reserved.

12.
Exp Neurobiol ; 22(4): 283-300, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24465144

ABSTRACT

Mitochondrial dysfunction in dopaminergic neurons of patients with idiopathic and familial Parkinson's disease (PD) is well known although the underlying mechanism is not clear. We established a homogeneous population of human adipose tissue-derived mesenchymal stromal cells (hAD-MSCs) from human adult patients with early-onset hereditary familial Parkin-defect PD as well as late-onset idiopathic PD by immortalizing cells with the hTERT gene to better understand the underlying mechanism of PD. The hAD-MSCs from patients with idiopathic PD were designated as "PD", from patients with Parkin-defect PD as "Parkin" and from patients with pituitary adenomas as "non-PD" in short. The pGRN145 plasmid containing hTERT was introduced to establish telomerase immortalized cells. The established hTERT-immortalized cell lines showed chromosomal aneuploidy sustained stably over two-years. The morphological study of mitochondria in the primary and immortalized hAD-MSCs showed that the mitochondria of the non-PD were normal; however, those of the PD and Parkin were gradually damaged. A striking decrease in mitochondrial complex I, II, and IV activities was observed in the hTERT-immortalized cells from the patients with idiopathic and Parkin-defect PD. Comparative Western blot analyses were performed to investigate the expressions of PD specific marker proteins in the hTERT-immortalized cell lines. This study suggests that the hTERT-immortalized hAD-MSC cell lines established from patients with idiopathic and familial Parkin-defect PD could be good cellular models to evaluate mitochondrial dysfunction to better understand the pathogenesis of PD and to develop early diagnostic markers and effective therapy targets for the treatment of PD.

13.
J Biol Chem ; 287(53): 44109-20, 2012 Dec 28.
Article in English | MEDLINE | ID: mdl-23144451

ABSTRACT

PINK1, linked to familial Parkinson's disease, is known to affect mitochondrial function. Here we identified a novel regulatory role of PINK1 in the maintenance of complex IV activity and characterized a novel mechanism by which NO signaling restored complex IV deficiency in PINK1 null dopaminergic neuronal cells. In PINK1 null cells, levels of specific chaperones, including Hsp60, leucine-rich pentatricopeptide repeat-containing (LRPPRC), and Hsp90, were severely decreased. LRPPRC and Hsp90 were found to act upstream of Hsp60 to regulate complex IV activity. Specifically, knockdown of Hsp60 resulted in a decrease in complex IV activity, whereas antagonistic inhibition of Hsp90 by 17-(allylamino) geldanamycin decreased both Hsp60 and complex IV activity. In contrast, overexpression of the PINK1-interacting factor LRPPRC augmented complex IV activity by up-regulating Hsp60. A similar recovery of complex IV activity was also induced by coexpression of Hsp90 and Hsp60. Drug screening identified ginsenoside Re as a compound capable of reversing the deficit in complex IV activity in PINK1 null cells through specific increases of LRPPRC, Hsp90, and Hsp60 levels. The pharmacological effects of ginsenoside Re could be reversed by treatment of the pan-NOS inhibitor L-NG-Nitroarginine Methyl Ester (L-NAME) and could also be reproduced by low-level NO treatment. These results suggest that PINK1 regulates complex IV activity via interactions with upstream regulators of Hsp60, such as LRPPRC and Hsp90. Furthermore, they demonstrate that treatment with ginsenoside Re enhances functioning of the defective PINK1-Hsp90/LRPPRC-Hsp60-complex IV signaling axis in PINK1 null neurons by restoring NO levels, providing potential for new therapeutics targeting mitochondrial dysfunction in Parkinson's disease.


Subject(s)
Electron Transport Complex IV/metabolism , Ginsenosides/pharmacology , Mitochondria/metabolism , Nitric Oxide/metabolism , Parkinson Disease/enzymology , Plant Extracts/pharmacology , Protein Kinases/deficiency , Signal Transduction , Animals , Chaperonin 60/genetics , Chaperonin 60/metabolism , HSP90 Heat-Shock Proteins/genetics , HSP90 Heat-Shock Proteins/metabolism , Humans , Mice , Mice, Transgenic , Mitochondria/drug effects , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Parkinson Disease/genetics , Parkinson Disease/metabolism , Protein Kinases/genetics , Signal Transduction/drug effects
14.
Exp Mol Med ; 44(2): 89-98, 2012 Feb 29.
Article in English | MEDLINE | ID: mdl-22257884

ABSTRACT

Autophagy is a dynamic cellular pathway involved in the turnover of proteins, protein complexes, and organelles through lysosomal degradation. The integrity of postmitotic neurons is heavily dependent on high basal autophagy compared to non-neuronal cells as misfolded proteins and damaged organelles cannot be diluted through cell division. Moreover, neurons contain the specialized structures for intercellular communication, such as axons, dendrites and synapses, which require the reciprocal transport of proteins, organelles and autophagosomes over significant distances from the soma. Defects in autophagy affect the intercellular communication and subsequently, contributing to neurodegeneration. The presence of abnormal autophagic activity is frequently observed in selective neuronal populations afflicted in common neurodegenerative diseases, such as Alzheimer's disease, Parkinson's disease, Huntington's disease and amyotrophic lateral sclerosis. These observations have provoked controversy regarding whether the increase in autophagosomes observed in the degenerating neurons play a protective role or instead contribute to pathogenic neuronal cell death. It is still unknown what factors may determine whether active autophagy is beneficial or pathogenic during neurodegeneration. In this review, we consider both the normal and pathophysiological roles of neuronal autophagy and its potential therapeutic implications for common neurodegenerative diseases.


Subject(s)
Autophagy/physiology , Neurodegenerative Diseases/pathology , Neurons/cytology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Animals , Humans , Huntington Disease/metabolism , Huntington Disease/pathology , Huntington Disease/physiopathology , Models, Biological , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/physiopathology , Parkinson Disease/metabolism , Parkinson Disease/pathology , Parkinson Disease/physiopathology
15.
Mitochondrion ; 11(5): 707-15, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21664494

ABSTRACT

The nature of mitochondrial dysfunction in dopaminergic neurons in familial Parkinson's disease (PD) is unknown. We characterized the pathophenotypes of dopaminergic neuronal cells that were deficient in PINK1 or DJ-1, genes with mutations linked to familial PD. Both PINK1- and DJ-1-deficient dopaminergic neurons had the increased production of ROS, severe mitochondrial structural damages and complex I deficits. A striking decrease in complex IV activity was also prominent by the PINK1-deficiency. The complex I deficits were relatively PD-specific and were significantly improved by an antioxidant Trolox. These data suggest that mitochondrial deficits are severe in dopaminergic neurons in familial PD and antioxidant-mediated functional recovery is feasible.


Subject(s)
Antioxidants/pharmacology , Chromans/pharmacology , Dopaminergic Neurons/drug effects , Intracellular Signaling Peptides and Proteins/deficiency , Oncogene Proteins/deficiency , Parkinsonian Disorders/drug therapy , Protein Kinases/deficiency , Adenosine Triphosphate/biosynthesis , Animals , Antioxidants/therapeutic use , Cells, Cultured , Chaperonin 60/metabolism , Chromans/therapeutic use , Citrate (si)-Synthase/metabolism , Cytochromes b5/metabolism , Dopaminergic Neurons/metabolism , Electron Transport Complex I/metabolism , Electron Transport Complex II/metabolism , Electron Transport Complex IV/metabolism , Enzyme Assays , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Knockout , Mitochondria/enzymology , Mitochondria/metabolism , Mitochondria/pathology , Mitochondrial Proteins/metabolism , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Oxidative Stress , Oxygen Consumption , Parkinsonian Disorders/metabolism , Parkinsonian Disorders/pathology , Protein Deglycase DJ-1 , Protein Kinases/genetics , Protein Kinases/metabolism
16.
Toxicol Lett ; 202(2): 85-92, 2011 Apr 25.
Article in English | MEDLINE | ID: mdl-21300143

ABSTRACT

There are two causes of Parkinson's disease (PD): environmental insults and genetic mutations of PD-associated genes. Environmental insults and genetic mutations lead to mitochondrial dysfunction, and a combination of mitochondrial dysfunction and increased oxidative stress in dopaminergic neurons is thought to contribute to the pathogenesis of PD. Among the PD-associated genes, DJ-1 acts as a redox sensor for oxidative stress and has been also proposed to maintain mitochondrial complex I activity. To understand molecular functions of DJ-1 in the cell, we have generated DJ-1 null cells from the DJ-1(-/-) mouse embryos. Using these null cells, we investigated the susceptibility to an environmental toxin, paraquat, which is known to inhibit mitochondrial complex I. Interestingly, we found that DJ-1 null cells showed a resistance to paraquat-induced apoptosis, including reduced poly (ADP-ribose) polymerase and procaspase-3. Also DJ-1 null cells generated less superoxide than SN4741 cells by paraquat treatment. Consistent with the reduced paraquat sensitivity, DJ-1 null cells showed reduced complex I activity, which was partially rescued by ectopic DJ-I expression. In summary, our results suggest that DJ-1 is critical to maintain mitochondrial complex I and complex I could be a key target in interaction of paraquat toxicity and DJ-1 for giving rise to PD.


Subject(s)
Apoptosis/drug effects , Dopamine/metabolism , Neurons/drug effects , Oncogene Proteins/metabolism , Oxidative Stress/physiology , Paraquat/toxicity , Parkinson Disease/metabolism , Animals , Blotting, Western , Caspase 3/metabolism , Cell Line , Flow Cytometry , Mice , Mice, Knockout , Microscopy, Fluorescence , Mitochondria/drug effects , Mitochondria/metabolism , Neurons/pathology , Oncogene Proteins/deficiency , Oncogene Proteins/genetics , Oxidative Stress/genetics , Parkinson Disease/genetics , Parkinson Disease/pathology , Peroxiredoxins , Poly(ADP-ribose) Polymerases/metabolism , Protein Deglycase DJ-1 , RNA/chemistry , RNA/genetics , Reverse Transcriptase Polymerase Chain Reaction , Superoxides/metabolism
17.
J Korean Med Sci ; 25(5): 798-803, 2010 May.
Article in English | MEDLINE | ID: mdl-20436722

ABSTRACT

A seventeen-year-old female patient was admitted with sudden-onset of headache and vomiting. Brain magnetic resonance imaging demonstrated a heterogeneously enhancing tumour in the left lateral ventricle. The tumour was removed and confirmed as a central neurocytoma (CN). For the residual tumour in the left lateral ventricle, gamma knife stereotactic radiosurgery was done at fifteen months after the initial surgery. Tumour recurred in the 4th ventricle at 5 yr after initial surgery. The tumour was removed and proved as a CN. In vitro primary culture was done with both tumours obtained from the left lateral ventricle and the 4th ventricle, respectively. Nestin, a neuronal stem cell marker was expressed in reverse Transcriptase-Polymerase Chain Reaction of both tumors. Both tumours showed different morphology and phenotypes of neuron and glia depending on the culture condition. When cultured in insulin, transferrin selenium and fibronectin media with basic fibroblast growth factors, tumour cells showed neuronal morphology and phenotypes. When cultured in the Dulbeco's Modified Essential Media with 20% fetal bovine serum, tumors cells showed glial morphology and phenotypes. It is suggested that CN has the characteristics of neuronal stem cells and potential to differentiate into mature neuron and glial cells depending on the environmental cue.


Subject(s)
Brain Neoplasms/pathology , Neurocytoma/pathology , Stem Cells/pathology , Cell Culture Techniques/methods , Cell Differentiation , Female , Humans , Young Adult
18.
Neurosci Lett ; 468(3): 272-6, 2010 Jan 14.
Article in English | MEDLINE | ID: mdl-19909785

ABSTRACT

Mutations in the PINK1 gene are known to cause early onset familial Parkinson's disease (PD). Genetic fruit fly model studies and rescue experiments with parkin overexpression suggest that PINK1 and parkin are associated via an unidentified mechanism. To gain additional insight into this interaction, we have investigated the impact of PINK1 deficiency on the biological function of parkin using actin filament dynamics. Actin is known to be associated with parkin and is a key regulator of eukaryotic cell death. PINK1 gene knockdown (KD) significantly increased actin aggregation and its binding with parkin. Known PD-related pathological conditions, such as oxidative stress and mitochondrial dysfunction, also increased actin aggregation and parkin binding. PINK1 KD resulted in the increased phosphorylation of cofilin, a protein important for the remodeling of actin filament and neurodegeneration. These results suggest that altered actin dynamics and increased association of parkin with actin filament might underlie the pathological conditions resulting from PINK1 deficiency.


Subject(s)
Actin Cytoskeleton/metabolism , Protein Kinases/genetics , Ubiquitin-Protein Ligases/metabolism , Animals , Cell Line , Dopamine/metabolism , Gene Knockdown Techniques , Mice , Neurons/metabolism , Protein Binding , Protein Kinases/biosynthesis
19.
J Neurochem ; 112(2): 366-76, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19878437

ABSTRACT

Our previous microarray analysis identified a neuroprotective protein Oxi-alpha, that was down-regulated during oxidative stress (OS)-induced cell death in dopamine neurons [Neurochem. Res. (2004) vol. 29, pp. 1223]. Here we find that the phylogenetically conserved Oxi-alpha protects against OS by a novel mechanism: activation of the mammalian target of rapamycin (mTOR) kinase and subsequent repression of autophagic vacuole accumulation and cell death. To the best of our knowledge, Oxi-alpha is the first molecule discovered in dopamine neurons, which activates mTOR kinase. Indeed, the down-regulation of Oxi-alpha by OS suppresses the activation of mTOR kinase. The pathogenic effect of down-regulated Oxi-alpha was confirmed by gene-specific knockdown experiment, which resulted in not only the repression of mTOR kinase and the subsequent phosphorylation of p70 S6 kinase and 4E-BP1, but also enhanced susceptibility to OS. In accordance with these observations, treatment with rapamycin, an mTOR inhibitor and autophagy inducer, potentiated OS-induced cell death, while similar treatment with an autophagy inhibitor, 3-methyladenine protected the dopamine cells. Our findings present evidence for the presence of a novel class of molecule involved in autophagic cell death triggered by OS in dopamine neurons.


Subject(s)
Autophagy/physiology , Dopamine/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Neurons/physiology , Oxidative Stress/physiology , Protein Serine-Threonine Kinases/metabolism , Adaptor Proteins, Signal Transducing , Animals , Autophagy/drug effects , Autophagy/genetics , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Cycle Proteins , Cell Line, Transformed , Cell Line, Tumor , Down-Regulation/drug effects , Down-Regulation/physiology , Eukaryotic Initiation Factors , Green Fluorescent Proteins/genetics , Hydrogen Peroxide/pharmacology , Mice , Microscopy, Confocal/methods , Neuroblastoma , Neurons/drug effects , Oxidative Stress/drug effects , Phosphoproteins/genetics , Phosphoproteins/metabolism , Phylogeny , Protein Kinases , Protein Serine-Threonine Kinases/genetics , RNA, Messenger/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/genetics , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Signal Transduction/drug effects , Sirolimus/pharmacology , TOR Serine-Threonine Kinases , Transfection/methods
20.
J Neurochem ; 94(4): 1040-53, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16092945

ABSTRACT

Septin 5, a parkin substrate, is a vesicle- and membrane-associated protein that plays a significant role in inhibiting exocytosis. The regulatory function of Septin 5 in dopaminergic (DAergic) neurons of substantia nigra (SN), maintained at relatively low levels, has not yet been delineated. As loss of function mutations of parkin are the principal cause of a familial Parkinson's disease, a prevailing hypothesis is that the loss of parkin activity results in accumulation of Septin 5 which confers neuron-specific toxicity in SN-DAergic neurons. In vitro and in vivo models were used to support this hypothesis. In our well-characterized DAergic SN4741 cell model, acute accumulation of elevated levels of Septin 5, but not synphilin-1 (another parkin substrate), resulted in cytotoxic cell death that was markedly reduced by parkin co-transfection. A transgenic mouse model expressing a dominant negative parkin mutant accumulated moderate levels of Septin 5 in SN-DAergic neurons. These mice acquired a progressive l-DOPA responsive motor dysfunction that developed despite a 25% higher than normal level of striatal dopamine (DA) and no apparent loss of DAergic neurons. The phenotype of this animal, increased striatal dopamine and reduced motor function, was similar to that observed in parkin knockout animals, suggesting a common DAergic alteration. These data suggest that a threshold level of Septin 5 accumulation is required for DAergic cell loss and that l-DOPA-responsive motor deficits can occur even in the presence of elevated DA.


Subject(s)
Cell Cycle Proteins/physiology , Dopamine/metabolism , Motor Activity/physiology , Neurons/metabolism , Neurotoxins/metabolism , Animals , Cell Line , Corpus Striatum/cytology , Corpus Striatum/metabolism , Dopamine Agents/pharmacology , Genes, Dominant , Immunoprecipitation , Levodopa/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Septins , Substantia Nigra/cytology , Substantia Nigra/metabolism , Tissue Distribution , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...