Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Inflammopharmacology ; 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39136812

ABSTRACT

Parkinson's disease (PD) is an age-related chronic neurological condition characterized by progressive degeneration of dopaminergic neurons and the presence of Lewy bodies, primarily composed of alpha-synuclein and ubiquitin. The pathophysiology of PD encompasses alpha-synuclein aggregation, oxidative stress, neuroinflammation, mitochondrial dysfunction, and impaired autophagy and ubiquitin-proteasome systems. Among these, the Keap1-Nrf2 pathway is a key regulator of antioxidant defense mechanisms. Nrf2 has emerged as a crucial factor in managing oxidative stress and inflammation, and it also influences ubiquitination through p62 expression. Keap1 negatively regulates Nrf2 by targeting it for degradation via the ubiquitin-proteasome system. Disruption of the Nrf2-Keap1 pathway in PD affects cellular responses to oxidative stress and inflammation, thereby playing a critical role in disease progression. In addition, the role of neuroinflammation in PD has gained significant attention, highlighting the interplay between immune responses and neurodegeneration. This review discusses the various mechanisms responsible for neuronal degeneration in PD, with a special emphasis on the neuroprotective role of the Nrf2-Keap1 pathway. Furthermore, it explores the implications of inflammopharmacology in modulating these pathways to provide therapeutic insights for PD.

2.
Nat Commun ; 15(1): 6143, 2024 Jul 21.
Article in English | MEDLINE | ID: mdl-39034309

ABSTRACT

Wolfram syndrome is a rare genetic disease caused by mutations in the WFS1 or CISD2 gene. A primary defect in Wolfram syndrome involves poor ER Ca2+ handling, but how this disturbance leads to the disease is not known. The current study, performed in primary neurons, the most affected and disease-relevant cells, involving both Wolfram syndrome genes, explains how the disturbed ER Ca2+ handling compromises mitochondrial function and affects neuronal health. Loss of ER Ca2+ content and impaired ER-mitochondrial contact sites in the WFS1- or CISD2-deficient neurons is associated with lower IP3R-mediated Ca2+ transfer from ER to mitochondria and decreased mitochondrial Ca2+ uptake. In turn, reduced mitochondrial Ca2+ content inhibits mitochondrial ATP production leading to an increased NADH/NAD+ ratio. The resulting bioenergetic deficit and reductive stress compromise the health of the neurons. Our work also identifies pharmacological targets and compounds that restore Ca2+ homeostasis, enhance mitochondrial function and improve neuronal health.


Subject(s)
Calcium , Endoplasmic Reticulum , Membrane Proteins , Mitochondria , Neurons , Wolfram Syndrome , Wolfram Syndrome/metabolism , Wolfram Syndrome/genetics , Calcium/metabolism , Mitochondria/metabolism , Endoplasmic Reticulum/metabolism , Animals , Neurons/metabolism , Membrane Proteins/metabolism , Membrane Proteins/genetics , Mice , Humans , Adenosine Triphosphate/metabolism , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Inositol 1,4,5-Trisphosphate Receptors/genetics , Mice, Knockout , NAD/metabolism , Calcium Signaling
3.
Brain Res ; 1826: 148742, 2024 03 01.
Article in English | MEDLINE | ID: mdl-38159591

ABSTRACT

The Endoplasmic reticulum (ER), a critical cellular organelle, maintains cellular homeostasis by regulating calcium levels and orchestrating essential functions such as protein synthesis, folding, and lipid production. A pivotal aspect of ER function is its role in protein quality control. When misfolded proteins accumulate within the ER due to factors like protein folding chaperone dysfunction, toxicity, oxidative stress, or inflammation, it triggers the Unfolded protein response (UPR). The UPR involves the activation of chaperones like calnexin, calreticulin, glucose-regulating protein 78 (GRP78), and Glucose-regulating protein 94 (GRP94), along with oxidoreductases like protein disulphide isomerases (PDIs). Cells employ the Endoplasmic reticulum-associated degradation (ERAD) mechanism to counteract protein misfolding. ERAD disruption causes the detachment of GRP78 from transmembrane proteins, initiating a cascade involving Inositol-requiring kinase/endoribonuclease 1 (IRE1), Activating transcription factor 6 (ATF6), and Protein kinase RNA-like endoplasmic reticulum kinase (PERK) pathways. The accumulation and deposition of misfolded proteins within the cell are hallmarks of numerous neurodegenerative diseases. These aberrant proteins disrupt normal neuronal signalling and contribute to impaired cellular homeostasis, including oxidative stress and compromised protein degradation pathways. In essence, ER stress is defined as the cellular response to the accumulation of misfolded proteins in the endoplasmic reticulum, encompassing a series of signalling pathways and molecular events that aim to restore cellular homeostasis. This comprehensive review explores ER stress and its profound implications for the pathogenesis and progression of neurodegenerative diseases.


Subject(s)
Neurodegenerative Diseases , Humans , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum-Associated Degradation , Endoplasmic Reticulum Stress , Unfolded Protein Response , Molecular Chaperones , Glucose
4.
Autophagy ; 18(9): 2249-2251, 2022 09.
Article in English | MEDLINE | ID: mdl-35090371

ABSTRACT

If cellular reactive oxygen species (ROS) production surpasses the intracellular antioxidant capacity, thus altering the ROS homeostasis, the cell needs to eradicate faulty mitochondria responsible for these excessive ROS. We have shown that even moderate ROS production breaks the KEAP1-PGAM5 complex, inhibiting the proteasomal removal of PGAM5. This leads to an accumulation of PGAM5 interfering with PINK1 processing that sensitizes mitochondria to autophagic removal. We propose that such a negative feedback system maintains cell ROS homeostasis.


Subject(s)
Mitochondrial Proteins , Mitophagy , Autophagy , Feedback , Homeostasis , Kelch-Like ECH-Associated Protein 1 , Mitochondrial Proteins/metabolism , NF-E2-Related Factor 2 , Phosphoprotein Phosphatases/metabolism , Reactive Oxygen Species/metabolism
5.
Redox Biol ; 48: 102186, 2021 Nov 11.
Article in English | MEDLINE | ID: mdl-34801863

ABSTRACT

When ROS production exceeds the cellular antioxidant capacity, the cell needs to eliminate the defective mitochondria responsible for excessive ROS production. It has been proposed that the removal of these defective mitochondria involves mitophagy, but the mechanism of this regulation remains unclear. Here, we demonstrate that moderate mitochondrial superoxide and hydrogen peroxide production oxidates KEAP1, thus breaking the interaction between this protein and PGAM5, leading to the inhibition of its proteasomal degradation. Accumulated PGAM5 interferes with the processing of the PINK1 in the mitochondria leading to the accumulation of PINK1 on the outer mitochondrial membrane. In turn, PINK1 promotes Parkin recruitment to mitochondria and sensitizes mitochondria for autophagic removal. We also demonstrate that inhibitors of the KEAP1-PGAM5 protein-protein interaction (including CPUY192018) mimic the effect of mitochondrial ROS and sensitize mitophagy machinery, suggesting that these inhibitors could be used as pharmacological regulators of mitophagy. Together, our results show that KEAP1/PGAM5 complex senses mitochondrially generated superoxide/hydrogen peroxide to induce mitophagy.

6.
Cells ; 11(1)2021 12 23.
Article in English | MEDLINE | ID: mdl-35011599

ABSTRACT

Mitochondria in the cell are the center for energy production, essential biomolecule synthesis, and cell fate determination. Moreover, the mitochondrial functional versatility enables cells to adapt to the changes in cellular environment and various stresses. In the process of discharging its cellular duties, mitochondria face multiple types of challenges, such as oxidative stress, protein-related challenges (import, folding, and degradation) and mitochondrial DNA damage. They mitigate all these challenges with robust quality control mechanisms which include antioxidant defenses, proteostasis systems (chaperones and proteases) and mitochondrial biogenesis. Failure of these quality control mechanisms leaves mitochondria as terminally damaged, which then have to be promptly cleared from the cells before they become a threat to cell survival. Such damaged mitochondria are degraded by a selective form of autophagy called mitophagy. Rigorous research in the field has identified multiple types of mitophagy processes based on targeting signals on damaged or superfluous mitochondria. In this review, we provide an in-depth overview of mammalian mitophagy and its importance in human health and diseases. We also attempted to highlight the future area of investigation in the field of mitophagy.


Subject(s)
Mammals/metabolism , Animals , Humans , Mitophagy/genetics , Models, Biological , Organelle Biogenesis , Receptors, Cell Surface/metabolism , Ubiquitin/metabolism
9.
Autophagy ; 15(5): 930-931, 2019 05.
Article in English | MEDLINE | ID: mdl-30806158

ABSTRACT

The Parkinson disease-associated proteins PINK1 and PRKN coordinate the ubiquitination of mitochondrial outer membrane proteins to tag them either for degradation or for autophagic clearance of the mitochondrion. The proteins include the mitochondrial trafficking proteins RHOT1 and RHOT2, the removal of which may be required for immobilization of mitochondria prior to mitophagy. Here, we demonstrate that RHOT1 and RHOT2 are not only substrates for PINK1-PRKN-dependent degradation but that they also play an active role in the process of mitophagy. RHOT1, and likely also RHOT2, may act as a docking site for inactive PRKN prior to mitochondrial damage, thus keeping PRKN in close proximity to its potential substrates and thereby facilitating mitophagy. We also show that RHOT1 functions as a calcium-sensing docking site for PRKN, and we suggest that calcium binding to RHOT is a key step in the calcium-dependent activation of mitophagy machinery.


Subject(s)
Autophagy , Mitophagy , Carrier Proteins , Mitochondria , Mitochondrial Proteins , Protein Kinases , Ubiquitin-Protein Ligases
10.
EMBO J ; 38(2)2019 01 15.
Article in English | MEDLINE | ID: mdl-30504269

ABSTRACT

The Parkinson's disease-associated protein kinase PINK1 and ubiquitin ligase Parkin coordinate the ubiquitination of mitochondrial proteins, which marks mitochondria for degradation. Miro1, an atypical GTPase involved in mitochondrial trafficking, is one of the substrates tagged by Parkin after mitochondrial damage. Here, we demonstrate that a small pool of Parkin interacts with Miro1 before mitochondrial damage occurs. This interaction does not require PINK1, does not involve ubiquitination of Miro1 and also does not disturb Miro1 function. However, following mitochondrial damage and PINK1 accumulation, this initial pool of Parkin becomes activated, leading to the ubiquitination and degradation of Miro1. Knockdown of Miro proteins reduces Parkin translocation to mitochondria and suppresses mitophagic removal of mitochondria. Moreover, we demonstrate that Miro1 EF-hand domains control Miro1's ubiquitination and Parkin recruitment to damaged mitochondria, and they protect neurons from glutamate-induced mitophagy. Together, our results suggest that Miro1 functions as a calcium-sensitive docking site for Parkin on mitochondria.


Subject(s)
Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Protein Kinases/metabolism , Ubiquitin-Protein Ligases/metabolism , rho GTP-Binding Proteins/metabolism , Animals , Calcium/metabolism , Cell Line , Gene Knockdown Techniques , HEK293 Cells , Humans , Mitochondrial Proteins/chemistry , Mitochondrial Proteins/genetics , Mitophagy , Protein Domains , Protein Transport , Proteolysis , Rats , Ubiquitination , rho GTP-Binding Proteins/chemistry , rho GTP-Binding Proteins/genetics
11.
PLoS Biol ; 14(7): e1002511, 2016 07.
Article in English | MEDLINE | ID: mdl-27434582

ABSTRACT

Deficiency of the protein Wolfram syndrome 1 (WFS1) is associated with multiple neurological and psychiatric abnormalities similar to those observed in pathologies showing alterations in mitochondrial dynamics. The aim of this study was to examine the hypothesis that WFS1 deficiency affects neuronal function via mitochondrial abnormalities. We show that down-regulation of WFS1 in neurons leads to dramatic changes in mitochondrial dynamics (inhibited mitochondrial fusion, altered mitochondrial trafficking, and augmented mitophagy), delaying neuronal development. WFS1 deficiency induces endoplasmic reticulum (ER) stress, leading to inositol 1,4,5-trisphosphate receptor (IP3R) dysfunction and disturbed cytosolic Ca2+ homeostasis, which, in turn, alters mitochondrial dynamics. Importantly, ER stress, impaired Ca2+ homeostasis, altered mitochondrial dynamics, and delayed neuronal development are causatively related events because interventions at all these levels improved the downstream processes. Our data shed light on the mechanisms of neuronal abnormalities in Wolfram syndrome and point out potential therapeutic targets. This work may have broader implications for understanding the role of mitochondrial dynamics in neuropsychiatric diseases.


Subject(s)
Mitochondria/metabolism , Mitochondrial Dynamics , Neurogenesis , Neurons/metabolism , Animals , Animals, Newborn , Brain/cytology , Brain/metabolism , Calcium/metabolism , Cells, Cultured , Endoplasmic Reticulum Stress/genetics , Fluorescence Resonance Energy Transfer , Homeostasis , Humans , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Membrane Potential, Mitochondrial/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice, Knockout , Microscopy, Confocal , Mitochondria/genetics , Mitophagy/genetics , Neurons/cytology , PC12 Cells , RNA Interference , Rats , Rats, Wistar , Time-Lapse Imaging/methods , Wolfram Syndrome/genetics , Wolfram Syndrome/metabolism
12.
Development ; 143(11): 1981-92, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27122166

ABSTRACT

During early development, neurons undergo complex morphological rearrangements to assemble into neuronal circuits and propagate signals. Rapid growth requires a large quantity of building materials, efficient intracellular transport and also a considerable amount of energy. To produce this energy, the neuron should first generate new mitochondria because the pre-existing mitochondria are unlikely to provide a sufficient acceleration in ATP production. Here, we demonstrate that mitochondrial biogenesis and ATP production are required for axonal growth and neuronal development in cultured rat cortical neurons. We also demonstrate that growth signals activating the CaMKKß, LKB1-STRAD or TAK1 pathways also co-activate the AMPK-PGC-1α-NRF1 axis leading to the generation of new mitochondria to ensure energy for upcoming growth. In conclusion, our results suggest that neurons are capable of signalling for upcoming energy requirements. Earlier activation of mitochondrial biogenesis through these pathways will accelerate the generation of new mitochondria, thereby ensuring energy-producing capability for when other factors for axonal growth are synthesized.


Subject(s)
Axons/metabolism , Organelle Biogenesis , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Adenylate Kinase/metabolism , Animals , Animals, Newborn , Calcium-Calmodulin-Dependent Protein Kinase Kinase/metabolism , Cell Proliferation , Cells, Cultured , Cerebral Cortex/cytology , Energy Metabolism , MAP Kinase Kinase Kinases/metabolism , Mitochondria/metabolism , Models, Biological , Neurogenesis , Nuclear Respiratory Factor 1/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Rats, Wistar , Transforming Growth Factor beta/metabolism
13.
J Biol Chem ; 290(47): 28540-28558, 2015 Nov 20.
Article in English | MEDLINE | ID: mdl-26420483

ABSTRACT

Neurogenesis involves generation of new neurons through finely tuned multistep processes, such as neural stem cell (NSC) proliferation, migration, differentiation, and integration into existing neuronal circuitry in the dentate gyrus of the hippocampus and subventricular zone. Adult hippocampal neurogenesis is involved in cognitive functions and altered in various neurodegenerative disorders, including Alzheimer disease (AD). Ethosuximide (ETH), an anticonvulsant drug is used for the treatment of epileptic seizures. However, the effects of ETH on adult hippocampal neurogenesis and the underlying cellular and molecular mechanism(s) are yet unexplored. Herein, we studied the effects of ETH on rat multipotent NSC proliferation and neuronal differentiation and adult hippocampal neurogenesis in an amyloid ß (Aß) toxin-induced rat model of AD-like phenotypes. ETH potently induced NSC proliferation and neuronal differentiation in the hippocampus-derived NSC in vitro. ETH enhanced NSC proliferation and neuronal differentiation and reduced Aß toxin-mediated toxicity and neurodegeneration, leading to behavioral recovery in the rat AD model. ETH inhibited Aß-mediated suppression of neurogenic and Akt/Wnt/ß-catenin pathway gene expression in the hippocampus. ETH activated the PI3K·Akt and Wnt·ß-catenin transduction pathways that are known to be involved in the regulation of neurogenesis. Inhibition of the PI3K·Akt and Wnt·ß-catenin pathways effectively blocked the mitogenic and neurogenic effects of ETH. In silico molecular target prediction docking studies suggest that ETH interacts with Akt, Dkk-1, and GSK-3ß. Our findings suggest that ETH stimulates NSC proliferation and differentiation in vitro and adult hippocampal neurogenesis via the PI3K·Akt and Wnt·ß-catenin signaling.


Subject(s)
Alzheimer Disease/chemically induced , Amyloid beta-Peptides/toxicity , Ethosuximide/pharmacology , Hippocampus/drug effects , Neurogenesis/drug effects , Alzheimer Disease/enzymology , Alzheimer Disease/metabolism , Animals , Cell Differentiation , Cell Proliferation , Cognition Disorders/chemically induced , Cognition Disorders/enzymology , Cognition Disorders/metabolism , Disease Models, Animal , Hippocampus/enzymology , Hippocampus/metabolism , Hippocampus/pathology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Wnt Proteins/metabolism , beta Catenin/metabolism
14.
J Biol Chem ; 290(34): 21163-21184, 2015 Aug 21.
Article in English | MEDLINE | ID: mdl-26139607

ABSTRACT

The human health hazards related to persisting use of bisphenol-A (BPA) are well documented. BPA-induced neurotoxicity occurs with the generation of oxidative stress, neurodegeneration, and cognitive dysfunctions. However, the cellular and molecular mechanism(s) of the effects of BPA on autophagy and association with oxidative stress and apoptosis are still elusive. We observed that BPA exposure during the early postnatal period enhanced the expression and the levels of autophagy genes/proteins. BPA treatment in the presence of bafilomycin A1 increased the levels of LC3-II and SQSTM1 and also potentiated GFP-LC3 puncta index in GFP-LC3-transfected hippocampal neural stem cell-derived neurons. BPA-induced generation of reactive oxygen species and apoptosis were mitigated by a pharmacological activator of autophagy (rapamycin). Pharmacological (wortmannin and bafilomycin A1) and genetic (beclin siRNA) inhibition of autophagy aggravated BPA neurotoxicity. Activation of autophagy against BPA resulted in intracellular energy sensor AMP kinase (AMPK) activation, increased phosphorylation of raptor and acetyl-CoA carboxylase, and decreased phosphorylation of ULK1 (Ser-757), and silencing of AMPK exacerbated BPA neurotoxicity. Conversely, BPA exposure down-regulated the mammalian target of rapamycin (mTOR) pathway by phosphorylation of raptor as a transient cell's compensatory mechanism to preserve cellular energy pool. Moreover, silencing of mTOR enhanced autophagy, which further alleviated BPA-induced reactive oxygen species generation and apoptosis. BPA-mediated neurotoxicity also resulted in mitochondrial loss, bioenergetic deficits, and increased PARKIN mitochondrial translocation, suggesting enhanced mitophagy. These results suggest implication of autophagy against BPA-mediated neurodegeneration through involvement of AMPK and mTOR pathways. Hence, autophagy, which arbitrates cell survival and demise during stress conditions, requires further assessment to be established as a biomarker of xenoestrogen exposure.


Subject(s)
Autophagy/drug effects , Benzhydryl Compounds/toxicity , Environmental Pollutants/toxicity , Hippocampus/drug effects , Neurons/drug effects , Phenols/toxicity , Protein Kinases/metabolism , TOR Serine-Threonine Kinases/metabolism , AMP-Activated Protein Kinase Kinases , Androstadienes/pharmacology , Animals , Animals, Newborn , Apoptosis Regulatory Proteins/antagonists & inhibitors , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Autophagy/genetics , Beclin-1 , Benzhydryl Compounds/antagonists & inhibitors , Environmental Pollutants/antagonists & inhibitors , Gene Expression Regulation , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Hippocampus/metabolism , Hippocampus/pathology , Humans , Macrolides/pharmacology , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Neurons/metabolism , Neurons/pathology , Oxidative Stress , Phenols/antagonists & inhibitors , Primary Cell Culture , Protein Kinases/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Sequestosome-1 Protein , Signal Transduction , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/genetics , Wortmannin
15.
Autophagy ; 10(6): 1105-19, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24879156

ABSTRACT

The autophagy protein BECN1/Beclin 1 is known to play a central role in autophagosome formation and maturation. The results presented here demonstrate that BECN1 interacts with the Parkinson disease-related protein PARK2. This interaction does not require PARK2 translocation to mitochondria and occurs mostly in cytosol. However, our results suggest that BECN1 is involved in PARK2 translocation to mitochondria because loss of BECN1 inhibits CCCP- or PINK1 overexpression-induced PARK2 translocation. Our results also demonstrate that the observed PARK2-BECN1 interaction is functionally important. Measurements of the level of MFN2 (mitofusin 2), a PARK2 substrate, demonstrate that depletion of BECN1 prevents PARK2 translocation-induced MFN2 ubiquitination and loss. BECN1 depletion also rescues the MFN2 loss-induced suppression of mitochondrial fusion. In sum, our results demonstrate that BECN1 interacts with PARK2 and regulates PARK2 translocation to mitochondria as well as PARK2-induced mitophagy prior to autophagosome formation.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Mitophagy/physiology , Ubiquitin-Protein Ligases/metabolism , Animals , Apoptosis Regulatory Proteins/antagonists & inhibitors , Apoptosis Regulatory Proteins/genetics , Autophagy , Beclin-1 , Biological Transport, Active , Cells, Cultured , GTP Phosphohydrolases , Gene Knockdown Techniques , HEK293 Cells , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mitochondria/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Mutant Proteins/genetics , Mutant Proteins/metabolism , PC12 Cells , Protein Kinases/genetics , Protein Kinases/metabolism , RNA, Small Interfering/genetics , Rats , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitination
16.
J Cell Sci ; 126(Pt 10): 2187-97, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23525002

ABSTRACT

Mitochondrial fusion-fission dynamics play a crucial role in many important cell processes. These dynamics control mitochondrial morphology, which in turn influences several important mitochondrial properties including mitochondrial bioenergetics and quality control, and they appear to be affected in several neurodegenerative diseases. However, an integrated and quantitative understanding of how fusion-fission dynamics control mitochondrial morphology has not yet been described. Here, we took advantage of modern visualisation techniques to provide a clear explanation of how fusion and fission correlate with mitochondrial length and motility in neurons. Our main findings demonstrate that: (1) the probability of a single mitochondrion splitting is determined by its length; (2) the probability of a single mitochondrion fusing is determined primarily by its motility; (3) the fusion and fission cycle is driven by changes in mitochondrial length and deviations from this cycle serves as a corrective mechanism to avoid extreme mitochondrial length; (4) impaired mitochondrial motility in neurons overexpressing 120Q Htt or Tau suppresses mitochondrial fusion and leads to mitochondrial shortening whereas stimulation of mitochondrial motility by overexpressing Miro-1 restores mitochondrial fusion rates and sizes. Taken together, our results provide a novel insight into the complex crosstalk between different processes involved in mitochondrial dynamics. This knowledge will increase understanding of the dynamic mitochondrial functions in cells and in particular, the pathogenesis of mitochondrial-related neurodegenerative diseases.


Subject(s)
Mitochondria/metabolism , Mitochondrial Dynamics , Mitochondrial Proteins/metabolism , Neurodegenerative Diseases/metabolism , Neurons/ultrastructure , rho GTP-Binding Proteins/metabolism , Animals , Humans , Huntingtin Protein , Mitochondria/ultrastructure , Mitochondrial Proteins/genetics , Mitochondrial Size/genetics , Mutation/genetics , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , PC12 Cells , Rats , Rats, Wistar , Transgenes/genetics , rho GTP-Binding Proteins/genetics , tau Proteins/genetics , tau Proteins/metabolism
17.
J Biol Chem ; 286(12): 10814-24, 2011 Mar 25.
Article in English | MEDLINE | ID: mdl-21252228

ABSTRACT

Parkinson disease is characterized by the accumulation of aggregated α-synuclein as the major component of the Lewy bodies. α-Synuclein accumulation in turn leads to compensatory effects that may include the up-regulation of autophagy. Another common feature of Parkinson disease (PD) is mitochondrial dysfunction. Here, we provide evidence that the overactivation of autophagy may be a link that connects the intracellular accumulation of α-synuclein with mitochondrial dysfunction. We found that the activation of macroautophagy in primary cortical neurons that overexpress mutant A53T α-synuclein leads to massive mitochondrial destruction and loss, which is associated with a bioenergetic deficit and neuronal degeneration. No mitochondrial removal or net loss was observed when we suppressed the targeting of mitochondria to autophagosomes by silencing Parkin, overexpressing wild-type Mitofusin 2 and dominant negative Dynamin-related protein 1 or blocking autophagy by silencing autophagy-related genes. The inhibition of targeting mitochondria to autophagosomes or autophagy was also partially protective against mutant A53T α-synuclein-induced neuronal cell death. These data suggest that overactivated mitochondrial removal could be one of the contributing factors that leads to the mitochondrial loss observed in PD models.


Subject(s)
Autophagy , Mitochondria/metabolism , Mutation, Missense , Neurons/metabolism , Parkinson Disease/metabolism , alpha-Synuclein/metabolism , Amino Acid Substitution , Animals , Disease Models, Animal , GTP Phosphohydrolases , Gene Silencing , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mitochondria/genetics , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , PC12 Cells , Parkinson Disease/genetics , Rats , Rats, Wistar , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , alpha-Synuclein/genetics
18.
J Biol Chem ; 284(32): 21379-85, 2009 Aug 07.
Article in English | MEDLINE | ID: mdl-19542216

ABSTRACT

Recent studies indicate that regulation of cellular oxidative capacity through enhancing mitochondrial biogenesis may be beneficial for neuronal recovery and survival in human neurodegenerative disorders. The peroxisome proliferator-activated receptor gamma coactivator-1alpha (PGC-1alpha) has been shown to be a master regulator of mitochondrial biogenesis and cellular energy metabolism in muscle and liver. The aim of our study was to establish whether PGC-1alpha and PGC-1beta control mitochondrial density also in neurons and if these coactivators could be up-regulated by deacetylation. The results demonstrate that PGC-1alpha and PGC-1beta control mitochondrial capacity in an additive and independent manner. This effect was observed in all studied subtypes of neurons, in cortical, midbrain, and cerebellar granule neurons. We also observed that endogenous neuronal PGC-1alpha but not PGC-1beta could be activated through its repressor domain by suppressing it. Results demonstrate also that overexpression of SIRT1 deacetylase or suppression of GCN5 acetyltransferase activates transcriptional activity of PGC-1alpha in neurons and increases mitochondrial density. These effects were mediated exclusively via PGC-1alpha, since overexpression of SIRT1 or suppression of GCN5 was ineffective where PGC-1alpha was suppressed by short hairpin RNA. Moreover, the results demonstrate that overexpression of PGC-1beta or PGC-1alpha or activation of the latter by SIRT1 protected neurons from mutant alpha-synuclein- or mutant huntingtin-induced mitochondrial loss. These evidences demonstrate that activation or overexpression of the PGC-1 family of coactivators could be used to compensate for neuronal mitochondrial loss and suggest that therapeutic agents activating PGC-1 would be valuable for treating neurodegenerative diseases in which mitochondrial dysfunction and oxidative damage play an important pathogenic role.


Subject(s)
Gene Expression Regulation , Mitochondria/metabolism , Neurons/metabolism , RNA-Binding Proteins/physiology , Transcription Factors/physiology , Adenosine Triphosphate/metabolism , Animals , Animals, Newborn , Autophagy , Humans , Oxygen/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , RNA-Binding Proteins/metabolism , Rats , Rats, Wistar , Sirtuin 1 , Sirtuins/biosynthesis , Transcription Factors/metabolism , p300-CBP Transcription Factors/biosynthesis
19.
J Biol Chem ; 283(21): 14391-401, 2008 May 23.
Article in English | MEDLINE | ID: mdl-18375387

ABSTRACT

We have investigated the mechanism of antiapoptotic and cell renewal effects of lansoprazole, a proton pump inhibitor, to protect and heal gastric mucosal injury in vivo induced by indomethacin, a non-steroidal anti-inflammatory drug (NSAID). Lansoprazole prevents indomethacin-induced gastric damage by blocking activation of mitochondrial and Fas pathways of apoptosis. Lansoprazole prevents indomethacin-induced up-regulation of proapoptotic Bax and Bak and down-regulation of antiapoptotic Bcl-2 and Bcl(xL) to maintain the normal proapoptotic/antiapoptotic ratio and thereby arrests indomethacin-induced mitochondrial translocation of Bax and collapse of mitochondrial membrane potential followed by cytochrome c release and caspase-9 activation. Lansoprazole also inhibits indomethacin-induced Fas-mediated mucosal cell death by down-regulating Fas or FasL expression and inhibiting caspase-8 activation. Lansoprazole favors mucosal cell renewal simultaneously by stimulating gene expression of prosurvival proliferating cell nuclear antigen, survivin, epidermal growth factor, and basic fibroblast growth factor. The up-regulation of Flt-1 further indicates that lansoprazole activates vascular epidermal growth factor-mediated controlled angiogenesis to repair gastric mucosa. Lansoprazole also stimulates the healing of already formed ulcers induced by indomethacin. Time course study of healing indicates that it switches off the mitochondrial death pathway completely but not the Fas pathway. However, lansoprazole heals mucosal lesions almost completely after overcoming the persisting Fas pathway, probably by favoring the prosurvival genes expression. This study thus provides the detailed mechanism of antiapoptotic and prosurvival effects of lansoprazole for offering gastroprotection against indomethacin-induced gastropathy.


Subject(s)
2-Pyridinylmethylsulfinylbenzimidazoles/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cytoprotection/drug effects , Gastric Mucosa/drug effects , Mitochondria/drug effects , Stomach Diseases/pathology , fas Receptor/metabolism , Animals , Caspases/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Gastric Mucosa/injuries , Gastric Mucosa/metabolism , Gastric Mucosa/pathology , Indomethacin/pharmacology , Lansoprazole , Microscopy, Electron, Transmission , Mitochondria/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction , Stomach Diseases/chemically induced , Stomach Diseases/metabolism , Wound Healing/drug effects
20.
Antimicrob Agents Chemother ; 52(2): 705-15, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18025110

ABSTRACT

A series of [(aryl)arylsufanylmethyl]pyridines (AASMP) have been synthesized. These compounds inhibited hemozoin formation, formed complexes (K(D) = 12 to 20 muM) with free heme (ferriprotoporphyrin IX) at a pH close to the pH of the parasite food vacuole, and exhibited antimalarial activity in vitro. The inhibition of hemozoin formation may develop oxidative stress in Plasmodium falciparum due to the accumulation of free heme. Interestingly, AASMP developed oxidative stress in the parasite, as evident from the decreased level of glutathione and increased formation of lipid peroxide, H(2)O(2), and hydroxyl radical (.OH) in P. falciparum. AASMP also caused mitochondrial dysfunction by decreasing mitochondrial potential (DeltaPsim) in malaria parasite, as measured by both flow cytometry and fluorescence microscopy. Furthermore, the generation of .OH may be mainly responsible for the antimalarial effect of AASMP since .OH scavengers such as mannitol, as well as spin trap alpha-phenyl-n-tertbutylnitrone, significantly protected P. falciparum from AASMP-mediated growth inhibition. Cytotoxicity testing of the active compounds showed selective activity against malaria parasite with selectivity indices greater than 100. AASMP also exhibited profound antimalarial activity in vivo against chloroquine resistant P. yoelii. Thus, AASMP represents a novel class of antimalarial.


Subject(s)
Antimalarials/pharmacology , Malaria/drug therapy , Plasmodium falciparum/drug effects , Plasmodium yoelii/drug effects , Pyridines/chemistry , Pyridines/pharmacology , Animals , Antimalarials/chemical synthesis , Antimalarials/chemistry , Antimalarials/therapeutic use , Benzoin/metabolism , Erythrocytes/drug effects , Erythrocytes/parasitology , Hemin/metabolism , Humans , Malaria/parasitology , Mice , Mice, Inbred BALB C , Oxidative Stress , Parasitic Sensitivity Tests , Pyridines/chemical synthesis , Pyridines/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL