Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Lydia Reznichenko is active.

Publication


Featured researches published by Lydia Reznichenko.


Neurosignals | 2005

Multifunctional activities of green tea catechins in neuroprotection. Modulation of cell survival genes, iron-dependent oxidative stress and PKC signaling pathway.

Silvia Mandel; Yael Avramovich-Tirosh; Lydia Reznichenko; Hailin Zheng; Orly Weinreb; Tamar Amit; Moussa B. H. Youdim

Many lines of evidence suggest that oxidative stress resulting in reactive oxygen species (ROS) generation and inflammation play a pivotal role in the age-associated cognitive decline and neuronal loss in neurodegenerative diseases including Alzheimer’s (AD), Parkinson’s (PD) and Huntington’s diseases. One cardinal chemical pathology observed in these disorders is the accumulation of iron at sites where the neurons die. The buildup of an iron gradient in conjunction with ROS (superoxide, hydroxyl radical and nitric oxide) are thought to constitute a major trigger in neuronal toxicity and demise in all these diseases. Thus, promising future treatment of neurodegenerative diseases and aging depends on availability of effective brain permeable, iron-chelatable/radical scavenger neuroprotective drugs that would prevent the progression of neurodegeneration. Tea flavonoids (catechins) have been reported to possess potent iron- chelating, radical-scavenging and anti-inflammatory activities and to protect neuronal death in a wide array of cellular and animal models of neurological diseases. Recent studies have indicated that in addition to the known antioxidant activity of catechins, other mechanisms such as modulation of signal transduction pathways, cell survival/death genes and mitochondrial function, contribute significantly to the induction of cell viability. This review will focus on the multifunctional properties of green tea and its major component (–)-epigallocatechin-3-gallate (EGCG) and their ability to induce neuroprotection and neurorescue in vitro and in vivo. In particular, their transitional metal (iron and copper) chelating property and inhibition of oxidative stress.


CNS Neuroscience & Therapeutics | 2008

Simultaneous Manipulation of Multiple Brain Targets by Green Tea Catechins: A Potential Neuroprotective Strategy for Alzheimer and Parkinson Diseases

Silvia Mandel; Tamar Amit; Orly Weinreb; Lydia Reznichenko; Moussa B. H. Youdim

Current therapeutic approaches for Alzheimer and Parkinson disease (AD and PD, respectively) are merely symptomatic, intended for the treatment of symptoms, but offer only partial benefit, without any disease‐modifying activity. Novel promising strategies suggest the use of antiinflammatory drugs, antioxidants, iron‐complexing molecules, neurotrophic factor delivery, inhibitors of the amyloid precursor protein (APP)‐processing secretases, gamma and beta (that generate the amyloid‐beta peptides, Aβ), anti‐Aβ aggregation molecules, the interference with lipid cholesterol metabolism and naturally occurring plant flavonoids to potentially reverse the course of the diseases. Human epidemiological and new animal data suggest that tea drinking may decrease the incidence of dementia, AD, and PD. 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. In the current article, we review the literature on the impact of the multimodal activities of green tea polyphenols and their neuroprotective effect on AD and PD.


Journal of Neurochemistry | 2006

Reduction of iron‐regulated amyloid precursor protein and β‐amyloid peptide by (–)‐epigallocatechin‐3‐gallate in cell cultures: implications for iron chelation in Alzheimer's disease

Lydia Reznichenko; Tamar Amit; Hailin Zheng; Yael Avramovich-Tirosh; Moussa B. H. Youdim; Orly Weinreb; Silvia Mandel

Brain iron dysregulation and its association with amyloid precursor protein (APP) plaque formation are implicated in Alzheimers disease (AD) pathology and so iron chelation could be considered a rational therapeutic strategy for AD. Here we analyzed the effect of the main polyphenol constituent of green tea, (–)‐epigallocatechin‐3‐gallate (EGCG), which possesses metal‐chelating and radical‐scavenging properties, on the regulation of the iron metabolism‐related proteins APP and transferrin receptor (TfR). EGCG exhibited potent iron‐chelating activity comparable to that of the prototype iron chelator desferrioxamine, and dose dependently (1–10 µm) increased TfR protein and mRNA levels in human SH‐SY5Y neuroblastoma cells. Both the immature and full‐length cellular holo‐APP were significantly reduced by EGCG, as shown by two‐dimensional gel electrophoresis, without altering APP mRNA levels, suggesting a post‐transcriptional action. Indeed, EGCG suppressed the translation of a luciferase reporter gene fused to the APP mRNA 5′‐untranslated region, encompassing the APP iron‐responsive element. The finding that Fe2SO4 reversed the action of EGCG on APP and TfR proteins reinforces the likelihood that these effects are mediated through modulation of the intracellular iron pool. Furthermore, EGCG reduced toxic β‐amyloid peptide generation in Chinese hamster ovary cells overexpressing the APP ‘Swedish’ mutation. Thus, the natural non‐toxic brain‐permeable EGCG may provide a potential therapeutic approach for AD and other iron‐associated disorders.


Journal of Neurochemistry | 2005

Green tea polyphenol (–)-epigallocatechin-3-gallate induces neurorescue of long-term serum-deprived PC12 cells and promotes neurite outgrowth

Lydia Reznichenko; Tamar Amit; Moussa B. H. Youdim; Silvia Mandel

Our previous studies have shown that the green tea polyphenol (–)‐epigallocatechin‐3‐gallate (EGCG) prevents neuronal cell death caused by several neurotoxins. The present study sought to determine the neuroprotective effect of EGCG when it is administered after the induction of cell damage (‘neurorescue’). In an attempt to imitate a progressive mode of death, PC12 cells were initially subjected to serum‐starvation conditions for a period of 1 or 3 days before administration of EGCG (0.1–10 µm) for up to 3 days. In spite of the high percentage of cell death, single or repetitive administration of EGCG (1 µm) significantly attenuated cell death. The neurorescue effect of EGCG was abolished by pre‐treatment with the protein kinase C inhibitor GF109203X (2.5 µm), suggesting the involvement of the protein kinase C pathway in neurorescue by the drug. This is consistent with the rapid (15 min) translocation of the protein kinase C alpha isoform to the cell membrane in response to EGCG. The correlative neurite outgrowth activity of EGCG on PC12 cells may also contribute to its neurorescue effect. The present findings suggest that EGCG may have a positive impact on aging and neurodegenerative diseases to retard or perhaps even reverse the accelerated rate of neuronal degeneration.


Journal of Alzheimer's Disease | 2008

Cell Signaling Pathways and Iron Chelation in the Neurorestorative Activity of Green Tea Polyphenols: Special Reference to Epigallocatechin Gallate (EGCG)

Silvia Mandel; Tamar Amit; Limor Kalfon; Lydia Reznichenko; Orly Weinreb; Moussa B. H. Youdim

Although much progress has been made in understanding the pathogenesis of Alzheimers 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 Parkinsons 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.


Neurotoxicity Research | 2003

Green tea polyphenol (−)-epigallocatechin-3-gallate protects rat PC12 cells from apoptosis induced by serum withdrawal

Silvia Mandel; Lydia Reznichenko; Tamar Amit; Moussa B. H. Youdim

Our recent studies have demonstrated that green tea polyphenol (−)-epigallocatechin-3-gallate (EGCG) exerts neuroprotective/neurorescue effects against ß-amyloid toxicity and protects neuronal cells from 1-methyl-4-phenyl-1,2,3,6-tetrahydropy-ridinium ion (MPP+) and 6-hydroxydopaminein vitro, or fromN-methyl-4-phenyl-1,2,3,6-tetrahdropyridine-(MPTP-) induced nigral dopaminergic neuronal loss in mice. In the present study, we report that EGCG (0.1 and 1 μM) significantly protects rat pheochromocytoma PC12 cells from apoptosis induced by serum support withdrawal, suggesting that EGCG may play a role in the growth of PC12 cells, where it stimulates survival-promoting pathways.


Journal of Biological Chemistry | 2009

A Sporadic Parkinson Disease Model via Silencing of the Ubiquitin-Proteasome/E3 Ligase Component SKP1A

Tali Fishman-Jacob; Lydia Reznichenko; Moussa B. H. Youdim; Silvia Mandel

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 G0/G1 phase when induced to differentiate. Instead, the cells progressed through S phase, developing rounded aggregates with characteristics of aggresomes including immunoreactivity for γ-tubulin, α-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.


Neurodegenerative Diseases | 2010

Low Dosage of Rasagiline and Epigallocatechin Gallate Synergistically Restored the Nigrostriatal Axis in MPTP-Induced Parkinsonism

Lydia Reznichenko; Limor Kalfon; Tamar Amit; Moussa B. H. Youdim; Silvia Mandel

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 α-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.


Molecular Nutrition & Food Research | 2006

Green tea catechins as brain-permeable, natural iron chelators-antioxidants for the treatment of neurodegenerative disorders

Silvia Mandel; Tamar Amit; Lydia Reznichenko; Orly Weinreb; Moussa B. H. Youdim


Journal of Nutrition | 2008

Targeting Multiple Neurodegenerative Diseases Etiologies with Multimodal-Acting Green Tea Catechins

Silvia Mandel; Tamar Amit; Limor Kalfon; Lydia Reznichenko; Moussa B. H. Youdim

Collaboration


Dive into the Lydia Reznichenko's collaboration.

Top Co-Authors

Avatar

Moussa B. H. Youdim

Technion – Israel Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Silvia Mandel

Technion – Israel Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Tamar Amit

Technion – Israel Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Orly Weinreb

Technion – Israel Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Hailin Zheng

Technion – Israel Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Limor Kalfon

Technion – Israel Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Yael Avramovich-Tirosh

Technion – Israel Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Orly Weinreb

Technion – Israel Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Mati Fridkin

Weizmann Institute of Science

View shared research outputs
Top Co-Authors

Avatar

Tali Fishman-Jacob

Technion – Israel Institute of Technology

View shared research outputs
Researchain Logo
Decentralizing Knowledge