Network


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

Hotspot


Dive into the research topics where Silvia Mandel is active.

Publication


Featured researches published by Silvia Mandel.


Journal of Neurochemistry | 2001

Green tea polyphenol (-)-epigallocatechin-3-gallate prevents N-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced dopaminergic neurodegeneration

Yona Levites; Orly Weinreb; Gila Maor; Moussa B. H. Youdim; Silvia Mandel

In the present study we demonstrate neuroprotective property of green tea extract and (–)‐epigallocatechin‐3‐gallate in N‐methyl‐4‐phenyl‐1,2,3,6‐tetrahydropyridine mice model of Parkinsons disease. N‐Methyl‐4‐phenyl‐1,2,3,6‐tetrahydropyridine neurotoxin caused dopamine neuron loss in substantia nigra concomitant with a depletion in striatal dopamine and tyrosine hydroxylase protein levels. Pretreatment of mice with either green tea extract (0.5 and 1 mg/kg) or (–)‐epigallocatechin‐3‐gallate (2 and 10 mg/kg) prevented these effects. In addition, the neurotoxin caused an elevation in striatal antioxidant enzymes superoxide dismutase (240%) and catalase (165%) activities, both effects being prevented by (–)‐epigallocatechin‐3‐gallate. (–)‐Epigallocatechin‐3‐gallate itself also increased the activities of both enzymes in the brain. The neuroprotective effects are not likely to be caused by inhibition of N‐methyl‐4‐phenyl‐1,2,3,6‐tetrahydropyridine conversion to its active metabolite 1‐methyl‐4‐phenylpyridinium by monoamine oxidase‐B, as both green tea and (–)‐epigallocatechin‐3‐gallate are very poor inhibitors of this enzyme in vitro (770 µg/mL and 660 µM, respectively). Brain penetrating property of polyphenols, as well as their antioxidant and iron‐chelating properties may make such compounds an important class of drugs to be developed for treatment of neurodegenerative diseases where oxidative stress has been implicated.


The FASEB Journal | 2003

Neuroprotection and neurorescue against Aβ toxicity and PKC-dependent release of nonamyloidogenic soluble precursor protein by green tea polyphenol (-)-epigallocatechin-3-gallate

Yona Levites; Tamar Amit; Silvia Mandel; Moussa B. H. Youdim

Green tea extract and its main polyphenol constituent (‐)‐epigallocatechin‐3‐gallate (EGCG) possess potent neuroprotective activity in cell culture and mice model of Parkinsons disease. The central hypothesis guiding this study is that EGCG may play an important role in amyloid precursor protein (APP) secretion and protection against toxicity induced by β‐amyloid (Aβ). The present study shows that EGCG enhances (~6‐fold) the release of the non‐amyloidogenic soluble form of the amyloid precursor protein (sAPPα) into the conditioned media of human SH‐SY5Y neuroblastoma and rat pheochromocytoma PC12 cells. sAPPα release was blocked by the hydroxamic acid‐based metalloprotease inhibitor Ro31–9790, which indicated mediation via α‐secretase activity. Inhibition of protein kinase C (PKC) with the inhibitor GF109203X, or by down‐regulation of PKC, blocked the EGCG‐induced sAPPα secretion, suggesting the involvement of PKC. Indeed, EGCG induced the phosphorylation of PKC, thus identifying a novel PKC‐dependent mechanism of EGCG action by activation of the non‐amyloidogenic pathway. EGCG is not only able to protect, but it can rescue PC12 cells against the β‐amyloid (Aβ) toxicity in a dose‐dependent manner. In addition, administration of EGCG (2 mg/kg) to mice for 7 or 14 days significantly decreased membrane‐bound holoprotein APP levels, with a concomitant increase in sAPPα levels in the hippocampus. Consistently, EGCG markedly increased PKCα and PKCε in the membrane and the cytosolic fractions of mice hippocampus. Thus, EGCG has protective effects against Aβ‐induced neurotoxicity and regulates secretory processing of non‐amyloidogenic APP via PKC pathway.


Journal of Neurochemistry | 2004

Cell signaling pathways in the neuroprotective actions of the green tea polyphenol (‐)‐epigallocatechin‐3‐gallate: implications for neurodegenerative diseases

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

Accumulating evidence supports the hypothesis that brain iron misregulation and oxidative stress (OS), resulting in reactive oxygen species (ROS) generation from H2O2 and inflammatory processes, trigger a cascade of events leading to apoptotic/necrotic cell death in neurodegenerative disorders, such as Parkinsons (PD), Alzheimers (AD) and Huntingtons diseases, and amyotrophic lateral sclerosis (ALS). Thus, novel therapeutic approaches aimed at neutralization of OS-induced neurotoxicity, support the application of ROS scavengers, transition metals (e.g. iron and copper) chelators and non-vitamin natural antioxidant polyphenols, in monotherapy, or as part of antioxidant cocktail formulation for these diseases. Both experimental and epidemiological evidence demonstrate that flavonoid polyphenols, particularly from green tea and blueberries, improve age-related cognitive decline and are neuroprotective in models of PD, AD and cerebral ischemia/reperfusion injuries. However, recent studies indicate that the radical scavenger property of green tea polyphenols is unlikely to be the sole explanation for their neuroprotective capacity and in fact, a wide spectrum of cellular signaling events may well account for their biological actions. In this article, the currently established mechanisms involved in the beneficial health action and emerging studies concerning the putative novel molecular neuroprotective activity of green tea and its major polyphenol (-)-epigallocatechin-3-gallate (EGCG), will be reviewed and discussed.


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.


Journal of Neural Transmission | 2004

Gene expression profiling of parkinsonian substantia nigra pars compacta; alterations in ubiquitin-proteasome, heat shock protein, iron and oxidative stress regulated proteins, cell adhesion/cellular matrix and vesicle trafficking genes

Edna Grünblatt; Silvia Mandel; J. Jacob-Hirsch; S. Zeligson; N. Amariglo; Gideon Rechavi; J. Li; Rivka Ravid; Wolfgang Roggendorf; Peter Riederer; Moussa B. H. Youdim

Summary.Gene expression profiling of human substantia nigra pars compacta (SNpc) from Parkinson’s disease (PD) patients, was examined employing high density microarrays. We identified alterations in the expression of 137 genes, with 68 down regulated and 69 up regulated. The down regulated genes belong to signal transduction, protein degradation (e.g. ubiquitin-proteasome subunits), dopaminergic transmission/metabolism, ion transport, protein modification/phosphorylation and energy pathways/glycolysis functional classes. Up-regulated genes, clustered mainly in biological processes involving cell adhesion/cytoskeleton, extracellular matrix components, cell cycle, protein modification/phosphorylation, protein metabolism, transcription and inflammation/stress (e.g. key iron and oxygen sensor EGLN1). One major finding in the present study is the particular decreased expression of SKP1A, a member of the SCF (E3) ligase complex specifically in the substantia nigra (SN) of sporadic parkinsonian patients, which may lead to a wide impairment in the function of an entire repertoire of proteins subjected to regulatory ubiquitination. These findings reveal novel players in the neurodegenerative scenario and provide potential targets for the development of novel drug compounds.


Biochemical Pharmacology | 2002

Attenuation of 6-hydroxydopamine (6-OHDA)-induced nuclear factor-kappaB (NF-kappaB) activation and cell death by tea extracts in neuronal cultures

Yona Levites; Moussa B. H. Youdim; Gila Maor; Silvia Mandel

Antioxidant and anti-inflammatory therapy approaches have been in the focus of attention in the treatment of neurodegenerative Parkinsons and Alzheimers diseases where oxidative stress has been implicated. Tea extracts have been previously reported to possess radical scavenger, iron chelating and anti-inflammatory properties in a variety of tissues. The purpose of this study was to investigate potential neuroprotective effects of tea extracts and possible signal pathway involved in a neuronal cell model of Parkinsons disease. We demonstrated highly potent antioxidant-radical scavenging activities of green tea (GT) and black tea (BT) extracts on brain mitochondrial membrane fraction, against iron (2.5 microM)-induced lipid peroxidation. Both extracts (0.6-3 microM total polyphenols) were shown to attenuate the neurotoxic action of 6-hydroxydopamine (6-OHDA)-induced neuronal death. 6-OHDA (350 and 50 microM) activated the iron dependent inflammatory redox sensitive nuclear factor-kappaB (NF-kappaB) in rat pheochromocytoma (PC12) and human neuroblastoma (NB) SH-SY5Y cells, respectively. Immunofluorescence and electromobility shift assays showed increased nuclear translocation and binding activity of NF-kappaB after exposure to 6-OHDA in NB SH-SY5Y cells, with a concomitant disappearance from the cytoplasm. Introduction of GT extract (0.6, 3 microM total polyphenols) before 6-OHDA inhibited both NF-kappaB nuclear translocation and binding activity induced by this toxin in NB SH-SY5Y cells. Neuroprotection was attributed to the potent antioxidant and iron chelating actions of the polyphenolic constituents of tea extracts, preventing nuclear translocation and activation of cell death promoting NF-kappaB. Brain penetrating property of polyphenols may make such compounds an important class of drugs for treatment of neurodegenerative diseases.


Journal of Neurochemistry | 2001

Gene expression analysis in N-methyl-4-phenyl-1,2,3,6-tetrahydropyridine mice model of Parkinson's disease using cDNA microarray: effect of R-apomorphine.

Edna Grünblatt; Silvia Mandel; Gila Maor; Moussa B. H. Youdim

To establish the possible roles of oxidative stress, inflammatory processes and other unknown mechanisms in neurodegeneration, we investigated brain gene alterations in N‐methyl‐4‐phenyl‐1,2,3,6‐tetrahydropyridine (MPTP) mice model of Parkinsons disease using Atlas mouse cDNA expression array membrane. The expression of 51 different genes involved in oxidative stress, inflammation, glutamate and neurotrophic factors pathways as well as in still undefined processes, such as cell cycle regulators and signal transduction molecules, was differentially affected by the treatment. The present study indicates the involvement of an additional cascade of events that might act in parallel to oxidative stress and inflammation to converge eventually into a common pathway leading to neurodegeneration. The attenuation of these gene changes by R‐apomorphine, an iron chelator‐radical scavenger drug, supports our previous findings in vivo where R‐apomorphine was neuroprotective.


CNS Drugs | 2003

Neuroprotective strategies in Parkinson's disease: An update on progress

Silvia Mandel; Edna Grünblatt; Peter Riederer; Manfred Gerlach; Yona Levites; Moussa B. H. Youdim

In spite of the extensive studies performed on postmortem substantia nigra from Parkinson’s disease patients, the aetiology of the disease has not yet been established. Nevertheless, these studies have demonstrated that, at the time of death, a cascade of events had been initiated that may contribute to the demise of the melanin-containing nigro-striatal dopamine neurons. These events include increased levels of iron and monoamine oxidase (MAO)-B activity, oxidative stress, inflammatory processes, glutamatergic excitotoxicity, nitric oxide synthesis, abnormal protein folding and aggregation, reduced expression of trophic factors, depletion of endogenous antioxidants such as reduced glutathione, and altered calcium homeostasis. To a large extent, the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) and 6-hydroxydopamine (6-OHDA) animal models of Parkinson’s disease confirm these findings. Furthermore, neuroprotection can be afforded in these models with iron chelators, radical scavenger antioxidants, MAO-B inhibitors, glutamate antagonists, nitric oxide synthase inhibitors, calcium channel antagonists and trophic factors.Despite the success obtained with animal models, clinical neuroprotection is much more difficult to accomplish. Although the negative studies obtained with the MAO-B inhibitor selegiline (deprenyl) and the antioxidant tocopherol (vitamin E) may have resulted from an inappropriate choice of drug (selegiline) or an inadequate dose (tocopherol), the niggling problem that still remains is why these drugs, and others, do work in animals while they fail in the clinic. One reason for this may be related to the fact that in normal human brains the number of dopaminergic neurons falls by around 3–5% every decade, while in Parkinson’s disease this decline is greater. Brain autopsy studies have shown that by the time the disease is identified, some 70–75% of the dopamine-containing neurons have been lost. More sensitive reliable methods and clinical correlative markers are required to discern between confoundable symptomatic effects versus a possible neuroprotective action of drugs, namely, the ability to delay or forestall disease progression by protecting or rescuing the remaining dopamine neurons or even restoring those that have been lost.A number of other possibilities for the clinical failure of potential neuroprotectants also exist. First, the animal models of Parkinson’s disease may not be totally reflective of the disease and, therefore, the chemical pathologies established in the animal models may not cause, or contribute to, the progression of the disease clinically. Second, because of the series of events occurring in neurode-generation and our ignorance about which of these factors constitutes the primary event in the pathogenic process, a single drug may not be adequate to induce neuroprotection and, as a consequence, use of a cocktail of drugs may be more appropriate. The latter concept receives support from recent complementary DNA (cDNA) microarray gene expression studies, which show the existence of a gene cascade of events occurring in the nigrostriatal pathway of MPTP, 6-OHDA and methamphetamine animal models of Parkinson’s disease.Even with the advent of powerful new tools such as genomics, proteomics, brain imaging, gene replacement therapy and knockout animal models, the desired end result of neuroprotection is still beyond our current capability.


Journal of Neurochemistry | 2004

Cell signaling pathways in the neuroprotective actions of the green tea polyphenol (-)-epigallocatechin-3-gallate: implications for neurodegenerative diseases: Signaling pathways in EGCG neuroprotection

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

Accumulating evidence supports the hypothesis that brain iron misregulation and oxidative stress (OS), resulting in reactive oxygen species (ROS) generation from H2O2 and inflammatory processes, trigger a cascade of events leading to apoptotic/necrotic cell death in neurodegenerative disorders, such as Parkinsons (PD), Alzheimers (AD) and Huntingtons diseases, and amyotrophic lateral sclerosis (ALS). Thus, novel therapeutic approaches aimed at neutralization of OS‐induced neurotoxicity, support the application of ROS scavengers, transition metals (e.g. iron and copper) chelators and non‐vitamin natural antioxidant polyphenols, in monotherapy, or as part of antioxidant cocktail formulation for these diseases. Both experimental and epidemiological evidence demonstrate that flavonoid polyphenols, particularly from green tea and blueberries, improve age‐related cognitive decline and are neuroprotective in models of PD, AD and cerebral ischemia/reperfusion injuries. However, recent studies indicate that the radical scavenger property of green tea polyphenols is unlikely to be the sole explanation for their neuroprotective capacity and in fact, a wide spectrum of cellular signaling events may well account for their biological actions. In this article, the currently established mechanisms involved in the beneficial health action and emerging studies concerning the putative novel molecular neuroprotective activity of green tea and its major polyphenol (‐)‐epigallocatechin‐3‐gallate (EGCG), will be reviewed and discussed.


Genes and Nutrition | 2009

Neuroprotective molecular mechanisms of (−)-epigallocatechin-3-gallate: a reflective outcome of its antioxidant, iron chelating and neuritogenic properties

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

Tea, the major source of dietary flavonoids, particularly the epicatechins, signifies the second most frequently consumed beverage worldwide, which varies its status from a simple ancient cultural drink to a nutrient component, endowed possible beneficial neuro-pharmacological actions. Accumulating evidence suggests that oxidative stress, resulting in reactive oxygen species generation, plays a pivotal role in neurodegenerative diseases, supporting the implementation of radical scavengers and metal chelating agents, such as natural tea polyphenols, for therapy. Vast epidemiology data indicate a correlation between occurrence of neurodegenerative disorders, such as Parkinson’s and Alzheimer’s diseases, and green tea consumption. In particular, recent literature strengthens the perception that diverse molecular signaling pathways, participating in the neuroprotective activity of the major green tea polyphenol, (−)-epigallocatechin-3-gallate (EGCG), renders this natural compound as potential agent to reduce the risk of various neurodegenerative diseases. In the current review, we discuss the studies concerning the mechanisms of action implicated in EGCG-induced neuroprotection and discuss the vision to translate these findings into a lifestyle arena.

Collaboration


Dive into the Silvia Mandel's collaboration.

Top Co-Authors

Avatar

Moussa B. H. Youdim

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
Top Co-Authors

Avatar

Orly Weinreb

Technion – Israel Institute of Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Orit Bar-Am

Technion – Israel Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Lydia Reznichenko

Technion – Israel Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Moussa B. H. Youdim

Technion – Israel Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Gila Maor

Technion – Israel Institute of Technology

View shared research outputs
Researchain Logo
Decentralizing Knowledge