Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 74
Filter
1.
Eur J Neurosci ; 60(4): 4393-4408, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38858171

ABSTRACT

Animal studies and clinical trials suggest that maintenance of gamma-aminobutyric acid (GABA)-ergic activity may be crucial in coping with stressful conditions, anxiety and mood disorders. Drugs highly efficient in promoting anxiolysis were shown to activate this system, particularly via the α2-subunit of type A receptors (GABAA α2). Given the high expression of GABAA α2 in the dentate gyrus (DG) sub-field of the hippocampus, we sought to examine whether manipulation of the α2 subunit in this area will evoke changes in emotional behaviour, memory and learning as well as in synaptic plasticity. We found that knockdown of GABAAα2 receptor specifically in the dorsal DG of rats caused increased anxiety without affecting locomotor activity. Spatial memory and learning in the Morris water maze were also impaired in GABAAα2 receptor knocked down rats, an effect accompanied by alterations in synaptic plasticity, as assessed by long-term potentiation in the DG. Our findings provide further support to the notion that emotional information processing in the hippocampus may be controlled, at least in part, via the inhibitory GABAA α2 receptor subunit, opening a potential avenue for early interventions from pre- puberty into adulthood, as a strategy for controlling anxiety-related psychopathology.


Subject(s)
Anxiety , Dentate Gyrus , Neuronal Plasticity , Receptors, GABA-A , Animals , Dentate Gyrus/metabolism , Receptors, GABA-A/metabolism , Receptors, GABA-A/genetics , Anxiety/metabolism , Neuronal Plasticity/physiology , Male , Rats , Memory Disorders/metabolism , Memory Disorders/genetics , Maze Learning/physiology , Gene Knockdown Techniques/methods , Long-Term Potentiation/physiology , Rats, Wistar
2.
J Neurochem ; 129(3): 434-47, 2014 May.
Article in English | MEDLINE | ID: mdl-24355073

ABSTRACT

DJ-1 is an oxidative stress sensor that localizes to the mitochondria when the cell is exposed to oxidative stress. DJ-1 mutations that result in gene deficiency are linked to increased risk of Parkinson's disease (PD). Activation of microglial stress conditions that are linked to PD may result in neuronal death. We postulated that DJ-1 deficiency may increase microglial neurotoxicity. We found that down-regulation of DJ-1 in microglia using an shRNA approach increased cell sensitivity to dopamine as measured by secreted pro-inflammatory cytokines such as IL-1ß and IL-6. Furthermore, we discovered that DJ-1-deficient microglia had increased monoamine oxidase activity that resulted in elevation of intracellular reactive oxygen species and nitric oxide leading to increased dopaminergic neurotoxicity. Rasagaline, a monoamine oxidase inhibitor approved for treatment of PD, reduced the microglial pro-inflammatory phenotype and significantly reduced neurotoxicity. Moreover, we discovered that DJ-1-deficient microglia have reduced expression of triggering receptor expressed on myeloid cells 2 (TREM2), previously suggested as a risk factor for pro-inflammation in neurodegenerative diseases. Further studies of DJ-1-mediated cellular pathways in microglia may contribute useful insights into the development of PD providing future avenues for therapeutic intervention


Subject(s)
Indans/pharmacology , Microglia/metabolism , Monoamine Oxidase Inhibitors/pharmacology , Neuroprotective Agents/pharmacology , Oncogene Proteins/deficiency , Animals , Blotting, Western , Cell Line , Cell Movement/drug effects , Cytokines/metabolism , Dopamine/toxicity , Enzyme-Linked Immunosorbent Assay , Inflammation/metabolism , Mice , Microglia/drug effects , Neurotransmitter Agents/toxicity , Peroxiredoxins , Phagocytosis/drug effects , Phenotype , Protein Deglycase DJ-1 , RNA, Small Interfering , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
3.
J Neural Transm (Vienna) ; 120(1): 37-48, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22446839

ABSTRACT

Iron accumulation and iron-related oxidative stress are involved in several pathological conditions and provide a rationale for the development of iron chelators as novel promising therapeutic strategies. Thus, we have recently synthesized multifunctional non-toxic, brain permeable iron chelating compounds, M30 and HLA20, possessing the neuroprotective N-propargyl moiety of the anti-Parkinsonian drug, monoamine oxidase (MAO)-B inhibitor, rasagiline and the antioxidant-iron chelating moiety of an 8-hydroxyquinoline derivative of the iron chelator, VK28. Here, we examined the hepatic regulatory effects of these novel compounds using two experimental approaches: chelation activity and glucose metabolism parameters. The present study demonstrated that M30 and HLA20 significantly decreased intracellular iron content and reduced ferritin expression levels in iron-loaded hepatoma Hep3B cells. In electron microscopy analysis, M30 was shown to reduce the electron-dense deposits of siderosomes by ~30 %, as well as down-regulate cytosolic ferritin particles observed in iron-overloaded cells. In vivo studies demonstrated that M30 administration (1 mg/kg, P.O. three times a week) reduced hepatic ferritin levels; increased hepatic insulin receptor and glucose transporter-1 levels and improved glucose tolerance in C57BL/6 mice and in a mouse model of type-2 diabetes, the ob/ob (leptin(-/-)). The results clearly indicate that the novel multifunctional drugs, especially M30, display significant capacity of chelating intracellular iron and regulating glucose metabolism parameters. Such effects can have therapeutic significance in conditions with abnormal local or systemic iron metabolism, including neurological diseases.


Subject(s)
Glucose/metabolism , Iron Chelating Agents/pharmacology , Iron/metabolism , Monoamine Oxidase Inhibitors/pharmacology , Neuroprotective Agents/pharmacology , Animals , Benzofurans , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Dose-Response Relationship, Drug , Ferric Compounds/pharmacology , Ferritins/metabolism , Glucose Tolerance Test , Humans , Hydroxyquinolines/chemistry , Hydroxyquinolines/pharmacology , Iron Chelating Agents/chemistry , Leptin/deficiency , Liver/drug effects , Liver/metabolism , Liver/ultrastructure , Male , Mice , Mice, Transgenic , Microscopy, Electron, Transmission , Neuroprotective Agents/chemistry , Piperazines/chemistry , Piperazines/pharmacology , Quaternary Ammonium Compounds/pharmacology , Quinolines
4.
Neurodegener Dis ; 10(1-4): 220-3, 2012.
Article in English | MEDLINE | ID: mdl-22205206

ABSTRACT

Microarray-derived transcriptomic studies in human substantia nigra pars compacta (SNpc) samples from sporadic Parkinson's disease (SPD) cases have opened an avenue to concentrate on potential gene intersections or cross-talks along the dopaminergic (DAergic) neurodegenerative cascade in SPD. One emerging gene candidate identified by our group was SKP1A (p19, S-phase kinase-associated protein 1A), found significantly decreased in the SNpc. It is part of the SCF (Skp1, Cullin 1, F-box protein) complex, the largest class of sophisticated ubiquitin-proteasome/E3 ligases, and can directly interact with Fbxo7, a gene defective in PARK15-linked PD. In vitro target validation by viral-mediated RNA interference revealed that the deficiency of Skp1 in a mouse SN-derived DAergic neuronal cell line potentiated the damage caused by exogenous insults implicated in PD pathology and caused the death of neurons undergoing differentiation, which developed Lewy body-like, α-synuclein-positive inclusions preceding cell death. Furthermore, recent animal studies show that site-directed intranigral stereotaxic injections of lentiviruses targeting SKP1A induce pathological and behavioral deficits in mice, supporting a significant role of Skp1 in SN DAergic neuronal survival in SPD. Thus, strategies aimed at increasing the activity or content of Skp1 may represent a novel therapeutic approach that has the potential to treat PD.


Subject(s)
Gene Expression Regulation, Enzymologic/physiology , Intracellular Signaling Peptides and Proteins/metabolism , Parkinson Disease , Protein Serine-Threonine Kinases/antagonists & inhibitors , Substantia Nigra/enzymology , Animals , Dopaminergic Neurons/physiology , Humans , Parkinson Disease/enzymology , Parkinson Disease/pathology , Parkinson Disease/physiopathology , Protein Serine-Threonine Kinases/metabolism , RNA Interference/physiology , Substantia Nigra/pathology
5.
Neurodegener Dis ; 10(1-4): 112-5, 2012.
Article in English | MEDLINE | ID: mdl-22156453

ABSTRACT

Our novel multimodal brain-permeable iron-chelating compounds M30 and HLA20 were demonstrated to possess neuroprotective/neurorescue activities in vitro and in vivo against several insults applicable to various neurodegenerative diseases, such as Alzheimer's disease, Parkinson's disease and amyotrophic lateral sclerosis. Neuroprotection by iron chelators has been widely recognized with respect to their ability to prevent reactive oxygen species generation in the Fenton reaction by sequestering redox-active iron. An additional neuroprotective mechanism of iron-chelating compounds is associated with their ability to regulate the transcriptional activator hypoxia-inducible factor 1 (HIF-1). HIF-1 is a 'master switch' being an important physiological response mechanism, likely enhancing neuroprotective compensatory pathways involved in many physiological processes within the brain. This mini-review will discuss the multifunctional mechanisms of action of the drugs, M30 and HLA20 in preclinical models of neurodegeneration with a specific emphasis on their ability to activate the HIF-1 signal transduction pathway.


Subject(s)
Hypoxia-Inducible Factor 1/metabolism , Iron Chelating Agents/therapeutic use , Neurodegenerative Diseases/drug therapy , Neuroprotective Agents/therapeutic use , Signal Transduction/drug effects , Animals , Humans , Iron Chelating Agents/pharmacology , Models, Biological
6.
J Neurochem ; 118(6): 939-57, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21138437

ABSTRACT

Alterations of iron levels in the brain has been observed and documented in a number of neurodegenerative disorders including Parkinson's disease (PD). The elevated nigral iron levels observed in PD may reflect a dysfunction of brain iron homeostasis. Under normal physiological conditions excess iron can be sequestrated in ferritin and neuromelanin. Alternatively, the excess iron may represent a component of brain iron deposition associated with ageing. The aetiology of idiopathic PD largely remains an enigma. However, intensive investigations have provided a host of putative mechanisms that might contribute to the pathogenesis underlying the characteristic degeneration of the dopaminergic neurons in the substantia nigra (SN). The mechanisms proposed include oxidative (and nitrative) stress, inflammation, excitotoxicity, mitochondrial dysfunction, altered proteolysis and finally apoptotic induced cell death. Iron-mediated cellular destruction is mediated primarily via reactive oxygen or/and nitrogen species induced oxidative stress. Furthermore, these pathogenic mechanisms appear to be closely interlinked to the cascade of events leading to cellular death. There are conflicting reports about the stage during disease progression at which nigral iron change occurs in PD. Some have found that there are no changes in iron content SN in asymptomatic incidental Lewy body disease, suggesting it may represent a secondary event in the cascade of neuronal degeneration. In contrast, others have found an elevation of iron in SN in pre-clinical stages. These discrepancies may be attributed to the occurrence of different sub-groups of the disease. This concurs with the notion that PD represents a group of related diseases with a number of potential pathogenic pathways.


Subject(s)
Iron/physiology , Parkinson Disease/pathology , Aging/physiology , Animals , Ferritins/metabolism , Homeostasis/physiology , Humans , Iron/metabolism , Lewy Bodies/physiology , Melanins/metabolism , Oxidative Stress/physiology , Parkinson Disease/metabolism
7.
J Neural Transm (Vienna) ; 118(3): 479-92, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21360301

ABSTRACT

It is for these authors a great privilege to dedicate this review article to Moussa Youdim, who is one of the most imperative pharmacologists and pioneer investigators in the search and development of novel therapeutics for neurodegenerative diseases. 40 years ago, Moussa Youdim has started studying brain iron, catecholamine receptor and monoamine oxidase (MAO)-A and -B functions. Although Moussa Youdim succeeded in exploring the novel anti-Parkinsonian, selective MAO-B inhibitor drug, rasagiline (Azilect, Teva Pharmaceutical Co.), he did not stop searching for superior therapeutic approaches for neurodegenerative disorders. To date, Moussa Youdim and his research group are designing and synthesizing pluripotential drug candidates possessing diverse pharmacological properties that can act on multiple targets and pathological features ascribed to Parkinson's disease, Alzheimer's disease (AD) and amyotrophic lateral sclerosis. One such example is the multimodal non-toxic, brain-permeable iron-chelating compound, M30 (5-[N-methyl-N-propargylaminomethyl]-8-hydroxyquinoline), which amalgamates the propargyl moiety of rasagiline with the backbone of the potent iron chelator, VK28. This review discusses the multiple effects of several leading compounds of this series, concerning their neuroprotective/neurorestorative molecular mechanisms in vivo and in vitro, with a special focus on the pathological features ascribed to AD, including antioxidant and iron chelating activities, regulation of amyloid precursor protein and amyloid ß peptide expression processing, activation of pro-survival signaling pathways and regulation of cell cycle and neurite outgrowth.


Subject(s)
Alzheimer Disease/drug therapy , Iron Chelating Agents/therapeutic use , Iron/metabolism , Monoamine Oxidase Inhibitors/therapeutic use , Alzheimer Disease/metabolism , History, 20th Century , Humans
8.
J Biol Chem ; 284(47): 32835-45, 2009 Nov 20.
Article in English | MEDLINE | ID: mdl-19748892

ABSTRACT

The aim of this study was to develop a new model of sporadic Parkinson disease (PD) based on silencing of the SKP1A gene, a component of the ubiquitin-proteasome/E3 ligase complex, Skp1, Cullin 1, F-box protein, which was found to be highly decreased in the substantia nigra of sporadic PD patients. Initially, an embryonic mouse substantia nigra-derived cell line (SN4741 cells) was infected with short hairpin RNA lentiviruses encoding the murine transcript of the SKP1A gene or with scrambled vector. SKP1A silencing resulted in increased susceptibility to neuronal damages induced by the parkinsonism-inducing neurotoxin 1-methyl-4-phenylpyridinium ion and serum starvation, in parallel with a decline in the expression of the dopaminergic markers, dopamine transporter and vesicular monoamine transporter-2. SKP1A-deficient cells presented a delay in completion of the cell cycle and the inability to arrest at the G(0)/G(1) phase when induced to differentiate. Instead, the cells progressed through S phase, developing rounded aggregates with characteristics of aggresomes including immunoreactivity for gamma-tubulin, alpha-synuclein, ubiquitin, tyrosine hydroxylase, Hsc-70 (70-kDa heat shock cognate protein), and proteasome subunit, and culminating in a lethal phenotype. Conversely, stably enforced expression of wild type SKP1A duplicated the survival index of naïve SN4741 cells under proteasomal inhibition injury, suggesting a new structural role of SKP1 in dopaminergic neuronal function, besides its E3 ligase activity. These results link, for the first time, SKP1 to dopamine neuronal function and survival, suggesting an essential role in sporadic PD. In summary, this new model has reproduced to a significant extent the molecular alterations described in sporadic PD at the cellular level, implicating Skp1 as a potential modifier in sporadic PD neurodegeneration.


Subject(s)
Gene Silencing , Parkinson Disease/genetics , Proteasome Endopeptidase Complex/chemistry , SKP Cullin F-Box Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitin/chemistry , Animals , Cell Line , Cell Survival , HSC70 Heat-Shock Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Neurodegenerative Diseases/metabolism , Neurons/metabolism , Neurotoxins/chemistry , Parkinson Disease/metabolism , Proteasome Endopeptidase Complex/metabolism , S-Phase Kinase-Associated Proteins , SKP Cullin F-Box Protein Ligases/physiology
9.
J Neurochem ; 113(2): 363-73, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20096090

ABSTRACT

Water-soluble iron, and manganese(III) complexes of corroles and porphyrins were examined with regard to their neuroprotective/neurorescue activities by using various neuronal cytotoxic models of oxidative and nitrative stress. The present study demonstrates that the metallocorroles significantly protect human neuroblastoma SH-SY5Y and mouse motor neuron-neuroblastoma fusion NSC-34 cell lines against neurotoxicity induced by either the peroxynitrite donor 3-morpholinosydnonimine or the parkinsonism-related neurotoxin 6-hydroxydopamine. The neuronal survival effect is further reflected by the prevention of 3-morpholinosydnonimine-induced protein nitration, inhibition of caspase 3 activation, as well as attenuation of 6-hydroxydopamine-mediated decrease in growth associated protein-43 levels. The iron(III) corrole, but not manganese (III) corrole, also significantly promotes neuronal survival of hydrogen peroxide (H(2)O(2))-impaired SH-SY5Y and NSC-34 cells. A substantial superiority of the metallocorroles relative to the corresponding porphyrin complexes is revealed in all examined aspects. These results highlight the large potential of corrole complexes as novel agents for therapeutic approaches in degenerative disorders of the central and peripheral nervous systems, where oxidative and nitrative stresses are involved.


Subject(s)
Metalloporphyrins/pharmacology , Motor Neurons/drug effects , Neuroprotective Agents/pharmacology , Nitrates/metabolism , Oxidative Stress/drug effects , Animals , Caspase 3/metabolism , Cell Count/methods , Cell Death/drug effects , Cell Line, Tumor , Cells, Cultured , Dose-Response Relationship, Drug , Drug Interactions , Embryo, Mammalian , Enzyme Inhibitors/pharmacology , Humans , Hydrogen Peroxide/pharmacology , In Situ Nick-End Labeling/methods , Mice , Molsidomine/analogs & derivatives , Molsidomine/pharmacology , Neuroblastoma/pathology , Oxidopamine/pharmacology , Porphyrins/pharmacology , Sympatholytics/pharmacology
10.
J Pharmacol Exp Ther ; 333(3): 874-82, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20237072

ABSTRACT

Increasing evidence suggests that oxidative stress (OS)-induced pancreatic beta-cell impairments is involved in diabetes and diabetic complications. Our group has recently synthesized two multifunctional nontoxic, lipophilic, iron-chelating drugs, 5-{N-methyl-N-propargylaminomethyl}-8-hydroxyquinoline (M30) and 5-{4-propargylpiperazin-1-ylmethyl}-8-hydroxyquinoline (HLA20), for the treatment of various OS-mediated pathogeneses. These compounds contain the N-propargylamine cytoprotective moiety of the antiparkinsonian drug rasagiline (Azilect) and the iron-complexing component 8-hydroxyquinoline. The aim of this research was to evaluate the protective effect of the multifunctional iron-chelating drugs on rat insulin-producing pancreatic beta-cells (INS-1E and RINm) against OS-induced cytotoxicity. We found that M30 and HLA20 markedly and dose-dependently inhibited H(2)O(2)-induced cytotoxicity, associated with decreased intracellular reactive oxygen species formation and increased catalase activity. In accordance, the catalase inhibitor 3-amino-1,2,4-triazol blocked the protective action of M30 against H(2)O(2)-induced damage. Both compounds significantly increased the levels of the iron-responsive protein transferrin receptor indicating their iron-chelating effect. Further mechanistic studies showed that M30 and HLA20 attenuated H(2)O(2)-induced mitochondrial membrane potential loss, decreased the release of cytochrome c into the cytoplasm, and inhibited the activation of caspase-3, suggesting that these drugs may produce cytoprotective effects via the preservation of mitochondrial function. These results indicate that the novel drugs, M30 and HLA20 display significant cytoprotective activity against OS-induced cytotoxicity in insulin producing beta-cells, which might be of therapeutic use in the treatment of diabetes mellitus.


Subject(s)
Antioxidants , Hydroxyquinolines/pharmacology , Insulin-Secreting Cells/drug effects , Iron Chelating Agents/pharmacology , Monoamine Oxidase Inhibitors/pharmacology , Oxidative Stress/drug effects , Piperazines/pharmacology , Animals , Apoptosis/drug effects , Blotting, Western , Caspase 3/metabolism , Catalase/metabolism , Cell Line , Cell Survival/drug effects , Coloring Agents , Cytochromes c/metabolism , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique , Hydrogen Peroxide/toxicity , Insulin-Secreting Cells/metabolism , Membrane Potentials/drug effects , Mitochondria/drug effects , Oxidants/toxicity , Rats , Signal Transduction/drug effects , Tetrazolium Salts , Thiazoles
11.
FASEB J ; 23(11): 3766-79, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19638399

ABSTRACT

Novel therapeutic approaches for the treatment of neurodegenerative disorders comprise drug candidates designed specifically to act on multiple central nervous system targets. We have recently synthesized multifunctional, nontoxic, brain-permeable iron-chelating drugs, M30 and HLA20, possessing the N-propargylamine neuroprotective moiety of rasagiline (Azilect) and the iron-chelating moiety of VK28. The present study demonstrates that M30 and HLA20 possess a wide range of pharmacological activities in mouse NSC-34 motor neuron cells, including neuroprotective effects against hydrogen peroxide- and 3-morpholinosydnonimine-induced neurotoxicity, induction of differentiation, and up-regulation of hypoxia-inducible factor (HIF)-1alpha and HIF-target genes (enolase1 and vascular endothelial growth factor). Both compounds induced NSC-34 neuritogenesis, accompanied by a marked increase in the expression of brain-derived neurotrophic factor and growth-associated protein-43, which was inhibited by PD98059 and GF109203X, indicating the involvement of mitogen-activated protein kinase and protein kinase C pathways. A major finding was the ability of M30 to significantly extend the survival of G93A-SOD1 amyotrophic lateral sclerosis mice and delay the onset of the disease. These properties of the novel multimodal iron-chelating drugs possessing neuroprotective/neuritogenic activities may offer future therapeutic possibilities for motor neurodegenerative diseases.


Subject(s)
Amyotrophic Lateral Sclerosis/drug therapy , Iron Chelating Agents/therapeutic use , Motor Neurons/drug effects , Neuroprotective Agents/therapeutic use , Animals , Apoptosis/drug effects , Brain-Derived Neurotrophic Factor/biosynthesis , Cell Differentiation/drug effects , Cell Line , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/metabolism , GAP-43 Protein/biosynthesis , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Hydrogen Peroxide/toxicity , Hydroxyquinolines/therapeutic use , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice , Mice, Transgenic , Molsidomine/analogs & derivatives , Molsidomine/toxicity , Motor Neurons/metabolism , Neurites/drug effects , Neurites/physiology , Phosphopyruvate Hydratase/biosynthesis , Piperazines/therapeutic use , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Transferrin/biosynthesis , Signal Transduction/drug effects , Superoxide Dismutase/toxicity , Superoxide Dismutase-1 , Vascular Endothelial Growth Factor A/biosynthesis
12.
Neurodegener Dis ; 7(1-3): 108-11, 2010.
Article in English | MEDLINE | ID: mdl-20173338

ABSTRACT

BACKGROUND: High-throughput gene-based platform studies in human postmortem substantia nigra (SN) from sporadic Parkinson's disease (PD) cases have revealed significant dysregulation of genes involved in biological processes linked to previously established neurodegenerative mechanisms in both sporadic and hereditary PD. OBJECTIVE: Our study aimed to develop a new genetic model of PD by modulating the expression of single genes that were found to be most significantly affected in SN of sporadic PD. METHODS: SN-derived cell line (SN4741 cells) was infected with short hairpin RNA lentiviruses carrying different gene-specific sequences. RESULTS: Silencing of the E3 ligase ubiquitin SKP1A resulted in a decline in the expression of dopaminergic phenotypic markers together with progression into an aberrant cell cycle and death. Furthermore, added knockout of the dopamine-metabolizing enzyme aldehyde dehydrogenase, found almost absent in sporadic PD SN pars compacta, exacerbated the vulnerability of SKP1A-silenced neurons to MPP(+) and neurotrophin deprivation. CONCLUSION: Future studies should focus on a careful consideration of crucial dopaminergic gene network interactions as emerged from human sporadic PD, which will serve as a basis for the development of a slowly progressive genetic animal model of sporadic PD, with the potential of evaluating drugs with 'disease-modifying activity'.


Subject(s)
Disease Models, Animal , Genetic Predisposition to Disease , Parkinson Disease/genetics , RNA Interference/physiology , Animals , Humans , Models, Biological , Parkinson Disease/etiology , Parkinson Disease/pathology , Silencer Elements, Transcriptional/genetics , Silencer Elements, Transcriptional/physiology , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
13.
Neurodegener Dis ; 7(4): 219-31, 2010.
Article in English | MEDLINE | ID: mdl-20197647

ABSTRACT

BACKGROUND: The anti-Parkinson monoamine oxidase B inhibitor rasagiline appears to be the first neuroprotective disease-modifying therapy in early-stage Parkinson's disease (PD). OBJECTIVE: Using a polypharmacy paradigm, we tested whether the distinct neuroprotective pharmacological profile of rasagiline would complement that of (-)-epigallocatechin-3-gallate (EGCG), the main antioxidant/iron chelator polyphenol constituent of green tea, and restore the neuronal loss and molecular targets damaged in animal parkinsonism. METHODS/RESULTS: We show by high-performance liquid chromatography, immunohistochemistry and Western blot analyses that the combination of rasagiline and EGCG, at subliminal doses which have no profound protective effect, acts synergistically to restore the nigrostriatal axis in N-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-treated mice. A detailed analysis revealed a complementary action of these drugs, differentially acting at MPTP-injured molecules/targets in the substantia nigra (SN): induction of brain-derived neurotrophic factor by rasagiline, increased membranal levels of the protein kinase C alpha-isoform by EGCG and a synergistic replenishment of their downstream effector, the serine/threonine kinase Akt/protein kinase B, suggesting that this kinase might represent one point of convergence of the distinct mechanisms of action of the drug cocktail. CONCLUSION: These results provide molecular evidence that activation of multiple brain targets by the combination of rasagiline and EGCG may synergistically contribute to the rescue of the dopamine neurons in the SN and replenishment of striatal dopamine. This may have important implications for rasagiline-treated PD patients who could further benefit from an adjunct administration of EGCG.


Subject(s)
Catechin/analogs & derivatives , Corpus Striatum/drug effects , Indans/pharmacology , Parkinsonian Disorders/drug therapy , Substantia Nigra/drug effects , Animals , Antioxidants/pharmacology , Antioxidants/therapeutic use , Catechin/pharmacology , Catechin/therapeutic use , Cell Line , Corpus Striatum/metabolism , Corpus Striatum/physiopathology , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Combinations , Drug Synergism , Indans/therapeutic use , Male , Mice , Mice, Inbred C57BL , Neural Pathways/drug effects , Neural Pathways/metabolism , Neural Pathways/physiopathology , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Parkinsonian Disorders/metabolism , Parkinsonian Disorders/physiopathology , Recovery of Function/drug effects , Recovery of Function/physiology , Substantia Nigra/metabolism , Substantia Nigra/physiopathology , Treatment Outcome
14.
iScience ; 23(8): 101375, 2020 Aug 21.
Article in English | MEDLINE | ID: mdl-32739834

ABSTRACT

Skp1, a component of the ubiquitin E3 ligases, was found to be decreased in the brains of sporadic Parkinson's disease (PD) patients, and its overexpression prevented death of murine neurons in culture. Here we expose the neuroprotective role of the Drosophila skp1 homolog, skpA, in the adult brain. Neuronal knockdown of skpA leads to accumulation of ubiquitinated protein aggregates and loss of dopaminergic neurons accompanied by motor dysfunction and reduced lifespan. Conversely, neuronal overexpression of skpA reduces aggregate load, improves age-related motor decline, and prolongs lifespan. Moreover, SkpA rescues neurodegeneration in a Drosophila model of PD. We also show that a Drosophila homolog of FBXO7, the F Box protein, Nutcracker (Ntc), works in the same pathway with SkpA. However, skpA overexpression rescues ntc knockdown phenotype, suggesting that SkpA interacts with additional F box proteins in the adult brain neurons. Collectively, our study discloses Skp1/SkpA as a potential therapeutic target in neurodegenerative diseases.

15.
FASEB J ; 22(5): 1296-305, 2008 May.
Article in English | MEDLINE | ID: mdl-18048580

ABSTRACT

Dysregulation of brain iron homeostasis is central to early neuropathological events in Alzheimer's disease (AD), including oxidative stress, inflammatory processes, amyloid deposition, tau phosphorylation, and neuronal cell cycle regulatory failure, leading to apoptosis. Also, there is a direct link between iron metabolism and AD pathogenesis, demonstrated by the presence of an iron-responsive element in the 5' UTR of the amyloid precursor protein transcript. As a consequence of these findings, a new paradigm is emerging that includes the development of iron-chelating neuroprotective-neurorescue drugs with multimodal functions, acting at various pathological brain targets. This concept is challenging the widely held assumption that "silver bullet" agents are superior to "dirty drugs" in drug therapy for neurodegenerative diseases. At best, the so-called magic bullets exhibit moderate symptomatic activity without modifying the course of disease progression. The present review elaborates on conventional and novel therapeutic targets of various multifunctional iron-chelating drugs (e.g., chemically designed compounds; natural polyphenols) that address multiple central nervous system etiologies in AD, aimed at preventing or slowing disease evolution. A similar approach in drug design is being investigated for treatment of cancer, AIDS, cardiovascular diseases, and depression.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/etiology , Iron Chelating Agents/therapeutic use , Neuroprotective Agents/therapeutic use , Amyloid beta-Protein Precursor/metabolism , Animals , Brain/metabolism , Catechin/analogs & derivatives , Catechin/therapeutic use , Cell Cycle/drug effects , Humans , Hydroxyquinolines/pharmacology , Iron/metabolism , Neurodegenerative Diseases/physiopathology , PC12 Cells , Piperazines/pharmacology , Rats
16.
Prog Neurobiol ; 82(6): 348-60, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17659826

ABSTRACT

Considering the multi-etiological character of Alzheimer's disease (AD), the current pharmacological approaches using drugs oriented towards a single molecular target possess limited ability to modify the course of the disease and thus, offer a partial benefit to the patient. In line with this concept, novel strategies include the use of a cocktail of several drugs and/or the development of a single molecule, possessing two or more active neuroprotective-neurorescue moieties that simultaneously manipulate multiple targets involved in AD pathology. A consistent observation in AD is a dysregulation of metal ions (Fe(2+), Cu(2+) and Zn(2+)) homeostasis and consequential induction of oxidative stress, associated with beta-amyloid aggregation and neurite plaque formation. In particular, iron has been demonstrated to modulate the Alzheimer's amyloid precursor holo-protein expression by a pathway similar to that of ferritin L-and H-mRNA translation through iron-responsive elements in their 5'UTRs. This review will discuss two separate scenarios concerning multiple therapy targets in AD, sharing in common the implementation of iron chelation activity: (i) novel multimodal brain-permeable iron chelating drugs, possessing neuroprotective-neurorescue and amyloid precursor protein-processing regulatory activities; (ii) natural plant polyphenols (flavonoids), such as green tea epigallocatechin gallate (EGCG) and curcumin, reported to have access to the brain and to possess multifunctional activities, such as metal chelation-radical scavenging, anti-inflammation and neuroprotection.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/metabolism , Iron Chelating Agents/administration & dosage , Iron Metabolism Disorders/drug therapy , Iron Metabolism Disorders/metabolism , Neuroprotective Agents/administration & dosage , Animals , Brain/drug effects , Brain/metabolism , Humans
17.
J Alzheimers Dis ; 15(2): 211-22, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18953110

ABSTRACT

Although much progress has been made in understanding the pathogenesis of Alzheimer's disease (AD), the current therapeutic approaches are merely symptomatic, intended for the treatment of cognitive symptoms, such as disturbances in memory and perception. Novel promising strategies suggest the use of anti-inflammatory drugs, antioxidants including natural occurring plant flavonoids, iron-complexing molecules, neurotrophic factor delivery, inhibitors of the amyloid-beta protein precursor processing secretases, gamma and beta, that generate amyloid-beta peptides and the interference with lipid and cholesterol metabolism. Human epidemiological and new animal data suggest that tea drinking may decrease the incidence of dementia, AD and Parkinson's disease. In particular, its main catechin polyphenol constituent (-)-epigallocatechin-3-gallate (EGCG) has been shown to exert neuroprotective/neurorescue activities in a wide array of cellular and animal models of neurological disorders. This review provides a detailed overview on the multimodal activities of green tea polyphenols with emphasis on their iron chelating, neurorescue/neuroregenerative and mitochondrial stabilization action.


Subject(s)
Catechin/analogs & derivatives , Flavonoids/pharmacology , Iron Chelating Agents , Nootropic Agents/pharmacology , Phenols/pharmacology , Signal Transduction/drug effects , Tea , Alzheimer Disease/pathology , Catechin/pharmacology , Humans , Neurodegenerative Diseases/pathology , Polyphenols
18.
J Nutr ; 138(8): 1578S-1583S, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18641210

ABSTRACT

Green tea is currently considered a source of dietary constituents endowed with biological and pharmacological activities relevant to human health. Human epidemiological and new animal data suggest that the pharmacological benefits of tea drinking may help to protect the brain as we age. Indeed, tea consumption is inversely correlated with the incidence of dementia and Alzheimer's and Parkinson's diseases. In particular, its main catechin polyphenol constituent (-)-epigallocatechin-3-gallate has been shown to exert neuroprotective/neurorescue activities in a wide array of cellular and animal models of neurological disorders. The intense efforts dedicated in recent years to shed light on the molecular mechanisms participating in the brain protective action of green tea indicate that in addition to the known antioxidant activity of catechins, the modulation of signal transduction pathways, cell survival/death genes, and mitochondrial function all contribute significantly to the induction of neuron viability. Because of the multietiological character of neurodegenerative disease pathology, these natural compounds are receiving significant attention as therapeutic cytoprotective agents that simultaneously manipulate multiple desired targets in the central nervous system. This article elaborates on the multimodal activities of green tea polyphenols with emphasis on their recently described neurorescue/neuroregenerative and mitochondrial stabilization actions.


Subject(s)
Catechin/therapeutic use , Neurodegenerative Diseases/prevention & control , Neuroprotective Agents/chemistry , Neuroprotective Agents/therapeutic use , Tea/chemistry , Catechin/pharmacology , Humans , Neurodegenerative Diseases/drug therapy , Neuroprotective Agents/pharmacology , Nutritional Physiological Phenomena
19.
Neurotox Res ; 33(3): 560-568, 2018 04.
Article in English | MEDLINE | ID: mdl-29218503

ABSTRACT

Parkinson's disease is the most common neurodegenerative disorder after Alzheimer's disease, with the majority of cases being sporadic or "idiopathic". The aetiology of the sporadic form is still unknown, but there is a broad consensus that Parkinson's disease involves multiple pathways. In previous human post-mortem studies investigating substantia nigra of parkinsonian subjects, gene expression alterations in various metabolic pathways including protein folding, trafficking, aggregation, ubiquitination and oxidative stress were found. These studies revealed transcriptomic dysregulation of various genes, amongst others Skp1A and PSMC4 (part of ubiquitin-proteasome system), HSC70 (belonging to the chaperone family) and ALDH1A1 (an enzyme involved in the catabolism of dopamine). To investigate whether these alterations are manifested at the protein level, we performed immunohistochemical analysis in the substantia nigra of Parkinson's disease and compared them to Alzheimer's disease and non-neurological post-mortem controls. We were able to confirm cell-specific reductions in the protein content of ALHD1A1 and Skp1A in the dopaminergic neurons of the substantia nigra of Parkinsonian patients compared to Alzheimer's and control subjects. Furthermore, we observed particular distribution for HSC70 and PSMC4 in the cytoplasm and accumulation within Lewy body in the dopaminergic neurons of the substantia nigra in Parkinson patients. These findings, together with previous evidence, suggest a malfunction of the ubiquitin-proteasome and possible autophagy systems as major players in protein misfolding and aggregation in Parkinson's disease. Nevertheless, this needs further proof, possibly with trajectory time line.


Subject(s)
ATPases Associated with Diverse Cellular Activities/metabolism , Aldehyde Dehydrogenase/metabolism , HSC70 Heat-Shock Proteins/metabolism , Parkinson Disease/pathology , Proteasome Endopeptidase Complex/metabolism , S-Phase Kinase-Associated Proteins/metabolism , Substantia Nigra/metabolism , Aged , Aged, 80 and over , Aldehyde Dehydrogenase 1 Family , Alzheimer Disease/pathology , Dopamine/metabolism , Female , Humans , Male , Middle Aged , Proteolysis , Retinal Dehydrogenase , Statistics, Nonparametric
20.
Curr Alzheimer Res ; 4(4): 403-11, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17908043

ABSTRACT

Accumulation of iron at sites where neurons degenerate in Parkinson's disease (PD) and Alzheimer's disease (AD) is thought to have a major role in oxidative stress induced process of neurodegeneration. The novel non-toxic lipophilic brain- permeable iron chelators, VK-28 (5- [4- (2- hydroxyethyl) piperazine-1-ylmethyl]- quinoline- 8- ol) and its multi-functional derivative, M-30 (5-[N-methyl-N-propargylaminomethyl]-8-hydroxyquinoline), as well as the main polyphenol constituent of green tea (-)-epigallocatechin-3-gallate (EGCG), which possesses iron metal chelating, radical scavenging and neuroprotective properties, offer potential therapeutic benefits for these diseases. M-30 and EGCG decreased apoptosis of human SH-SY5Y neuroblastoma cells in a neurorescue, serum deprivation model, via multiple protection mechanisms including: reduction of the pro-apoptotic proteins, Bad and Bax, reduction of apoptosis-associated Ser139 phosphorylated H2A.X and inhibition of the cleavage and activation of caspase-3. M-30 and EGCG also promoted morphological changes, resulting in axonal growth-associated protein-43 (GAP-43) implicating neuronal differentiation. Both compounds significantly reduced the levels of cellular holo-amyloid precursor protein (APP) in SH-SY5Y cells. The ability of theses novel iron chelators and EGCG to regulate APP are in line with the presence of an iron-responsive element (IRE) in the 5'-untranslated region (5'UTR) of APP. Also, EGCG reduced the levels of toxic amyloid-beta peptides in CHO cells over-expressing the APP "Swedish" mutation. The diverse molecular mechanisms and cell signaling pathways participating in the neuroprotective/neurorescue and APP regulation/processing actions of M-30 and EGCG, make these multifunctional compounds potential neuroprotective drugs for the treatment of neurodegenerative diseases, such as PD, AD, Huntington's disease and amyotrophic lateral sclerosis.


Subject(s)
Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Antioxidants/pharmacology , Apoptosis/drug effects , Catechin/analogs & derivatives , Hydroxyquinolines/pharmacology , Animals , Antioxidants/chemistry , Catechin/chemistry , Catechin/pharmacology , Cell Line, Tumor , Dose-Response Relationship, Drug , Gene Expression Regulation, Neoplastic/drug effects , Humans , Hydroxyquinolines/chemistry , Mice , Mice, Inbred C57BL , Models, Biological , Neuroblastoma
SELECTION OF CITATIONS
SEARCH DETAIL