Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Harefuah ; 163(2): 114-119, 2024 Feb.
Article in Hebrew | MEDLINE | ID: mdl-38431861

ABSTRACT

INTRODUCTION: Israel is below the global average of cancer mortality thanks to early diagnosis plans and advanced treatment, yet every year about 30,000 patients are diagnosed with cancer and 11,000 die from it. Many patients are diagnosed at an advanced stage of malignancy in which curative surgery cannot be offered. Early detection and intervention have been proven to be of greatest importance in reducing cancer morbidity and mortality. However, despite the clinical use of a limited number of technologies, the means for detecting malignancy as early as possible, to the extent of predicting malignancy within a significant period of time before its clinical detection, some current efforts still exist only within the framework of development and clinical research. The main challenge remains - the development of a test with high sensitivity on the one hand, but with sufficient specificity to prevent unnecessary follow-up tests at the other hand.


Subject(s)
Early Detection of Cancer , Neoplasms , Humans , Neoplasms/diagnosis , Israel , Upper Extremity
2.
Cells ; 12(5)2023 02 27.
Article in English | MEDLINE | ID: mdl-36899898

ABSTRACT

The concept of chelation therapy as a valuable therapeutic approach in neurological disorders led us to develop multi-target, non-toxic, lipophilic, brain-permeable compounds with iron chelation and anti-apoptotic properties for neurodegenerative diseases, such as Parkinson's disease (PD), Alzheimer's disease (AD), age-related dementia and amyotrophic lateral sclerosis (ALS). Herein, we reviewed our two most effective such compounds, M30 and HLA20, based on a multimodal drug design paradigm. The compounds have been tested for their mechanisms of action using animal and cellular models such as APP/PS1 AD transgenic (Tg) mice, G93A-SOD1 mutant ALS Tg mice, C57BL/6 mice, Neuroblastoma × Spinal Cord-34 (NSC-34) hybrid cells, a battery of behavior tests, and various immunohistochemical and biochemical techniques. These novel iron chelators exhibit neuroprotective activities by attenuating relevant neurodegenerative pathology, promoting positive behavior changes, and up-regulating neuroprotective signaling pathways. Taken together, these results suggest that our multifunctional iron-chelating compounds can upregulate several neuroprotective-adaptive mechanisms and pro-survival signaling pathways in the brain and might function as ideal drugs for neurodegenerative disorders, such as PD, AD, ALS, and aging-related cognitive decline, in which oxidative stress and iron-mediated toxicity and dysregulation of iron homeostasis have been implicated.


Subject(s)
Alzheimer Disease , Amyotrophic Lateral Sclerosis , Hydroxyquinolines , Parkinson Disease , Mice , Animals , Alzheimer Disease/metabolism , Amyotrophic Lateral Sclerosis/drug therapy , Hydroxyquinolines/pharmacology , Hydroxyquinolines/therapeutic use , Mice, Inbred C57BL , Iron Chelating Agents/therapeutic use , Mice, Transgenic , Parkinson Disease/pathology , Aging , Iron/metabolism
3.
Nat Cancer ; 2(10): 1055-1070, 2021 10.
Article in English | MEDLINE | ID: mdl-35121883

ABSTRACT

Stochastic transition of cancer cells between drug-sensitive and drug-tolerant persister phenotypes has been proposed to play a key role in non-genetic resistance to therapy. Yet, we show here that cancer cells actually possess a highly stable inherited chance to persist (CTP) during therapy. This CTP is non-stochastic, determined pre-treatment and has a unimodal distribution ranging from 0 to almost 100%. Notably, CTP is drug specific. We found that differential serine/threonine phosphorylation of the insulin receptor substrate 1 (IRS1) protein determines the CTP of lung and of head and neck cancer cells under epidermal growth factor receptor inhibition, both in vitro and in vivo. Indeed, the first-in-class IRS1 inhibitor NT219 was highly synergistic with anti-epidermal growth factor receptor therapy across multiple in vitro and in vivo models. Elucidation of drug-specific mechanisms that determine the degree and stability of cellular CTP may establish a framework for the elimination of cancer persisters, using new rationally designed drug combinations.


Subject(s)
ErbB Receptors , Neoplasms , ErbB Receptors/genetics , Insulin Receptor Substrate Proteins/genetics , Phosphorylation , Probability
4.
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
5.
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
6.
Neuropharmacology ; 123: 359-367, 2017 Sep 01.
Article in English | MEDLINE | ID: mdl-28571715

ABSTRACT

In many of the neurodegenerative diseases, such as Alzheimer's disease (AD) and AD-related disorders, as well as in the regular ageing process, excessive generation of oxidative stress (OS) and accumulation of iron levels and deposition have been observed in specific affected-brain regions and thus, regarded as contributing factors to the pathogenesis of the diseases. In AD, iron promotes amyloid ß (Aß) neurotoxicity by producing free radical damage and OS in brain areas affected by neurodegeneration, presumably by facilitating the aggregation of Aß. In addition, it was shown that iron modulates intracellular levels of the holo amyloid precursor protein (APP) by iron-responsive elements (IRE) RNA stem loops in the 5' untranslated region (5'UTR) of the APP transcript. As a consequence of these observations, iron chelation is one of the major new therapeutic strategies for the treatment of AD. This review describes the benefits and importance of the multimodal brain permeable chimeric iron-chelating/propargylamine drug M30, concerning its neuroprotective/neurorestorative inter-related activities relevant of the pathological features ascribed to AD, with a special focus on the effect of the drug on APP regulation and processing.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/metabolism , Hydroxyquinolines/pharmacology , Iron Chelating Agents/pharmacology , Neuroprotective Agents/pharmacology , Animals , Humans
7.
Neurotox Res ; 29(2): 208-17, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26581376

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is the most common degenerative disease of the motoneuron system, involving various abnormalities, such as mitochondrial dysfunction, oxidative stress, transitional metal accumulation, neuroinflammation, glutamate excitotoxicity, apoptosis, decreased supply of trophic factors, cytoskeletal abnormalities, and extracellular superoxide dismutase (SOD)-1 toxicity. These multiple disease etiologies implicated in ALS gave rise to the perception that future therapeutic approaches for the disease should be aimed at targeting multiple pathological pathways. In line with this view, we have evaluated in the current study the therapeutic effects of low doses of the novel multifunctional monoamine oxidase (MAO) inhibitor/iron-chelating compound, M30 in combination with high Calorie Energy supplemented Diet (CED) in the SOD1-G93A transgenic mouse model of ALS. Our results demonstrated that the combined administration of M30 with CED produced additive neuroprotective effects on motor performance and increased survival of SOD1-G93A mice. We also found that both M30 and M30/CED regimens caused a significant inhibition of MAO-A and -B activities and decreased the turnover of dopamine in the brain of SOD1-G93A mice. In addition, M30/CED combined treatment resulted in a significant increase in mRNA expression levels of various mitochondrial biogenesis and metabolism regulators, such as peroxisome proliferator-activated receptor-γ (PPARγ)-co activator 1 alpha (PGC-1α), PPARγ, uncoupling protein 1, and insulin receptor in the gastrocnemius muscle of SOD1-G93A mice. These results suggest that a combination of drug/agents with different, but complementary mechanisms may be beneficial in the treatment of ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/prevention & control , Diet , Hydroxyquinolines/administration & dosage , Iron Chelating Agents/administration & dosage , Monoamine Oxidase Inhibitors/administration & dosage , Neuroprotective Agents/administration & dosage , 3,4-Dihydroxyphenylacetic Acid/metabolism , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/physiopathology , Animals , Biogenic Monoamines/metabolism , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Disease Models, Animal , Male , Mice , Mice, Transgenic , Monoamine Oxidase/metabolism , Motor Activity/drug effects , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Phenylacetates/metabolism , Superoxide Dismutase/genetics , Superoxide Dismutase-1 , Survival Analysis , Transcription Factors/metabolism
8.
Neurobiol Aging ; 36(3): 1529-42, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25499799

ABSTRACT

Recently, we have designed and synthesized a novel multipotent, brain-permeable iron-chelating drug, VAR10303 (VAR), possessing both propargyl and monoamine oxidase (MAO) inhibitory moieties. The present study was undertaken to determine the multiple pharmacological activities of VAR in neurodegenerative preclinical models. We demonstrate that VAR affords iron chelating/iron-induced lipid-peroxidation inhibitory potency and brain selective MAO-A and MAO-B inhibitory effects, with only limited tyramine-cardiovascular potentiation of blood pressure. The results show that in 6-hydroxydopamine rat (neuroprotection) and in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine mouse (neurorescue) Parkinson's disease models, VAR significantly attenuated the loss of striatal dopamine levels, markedly reduced dopamine turnover, and increased tyrosine-hydroxylase levels. Furthermore, chronic systemic treatment of aged rats with VAR improved cognitive behavior deficits and enhanced the expression levels of neurotrophic factors (e.g., brain-derived neurotrophic factor, glial cell-derived neurotrophic factor, and nerve growth factor), Bcl-2 family members and synaptic plasticity in the hippocampus. Our study indicates that the multitarget compound VAR exerted neuroprotective and neurorestorative effects in animal models of Parkinson's disease and aging, further suggesting that a drug that can regulate multiple brain targets could be an ideal treatment-strategy for age-associated neurodegenerative disorders.


Subject(s)
Hydroxyquinolines/pharmacology , Hydroxyquinolines/therapeutic use , Iron Chelating Agents/pharmacology , Iron Chelating Agents/therapeutic use , Monoamine Oxidase Inhibitors/pharmacology , Monoamine Oxidase Inhibitors/therapeutic use , Neuroprotective Agents , Parkinson Disease/drug therapy , Aging , Animals , Cognition , Disease Models, Animal , Dopamine/metabolism , Male , Mice, Inbred C57BL , Molecular Targeted Therapy , Monoamine Oxidase , Nerve Growth Factors/metabolism , Neuronal Plasticity/drug effects , Parkinson Disease/genetics , Parkinson Disease/metabolism , Parkinson Disease/psychology , Rats, Sprague-Dawley
9.
Parkinsonism Relat Disord ; 20 Suppl 1: S132-6, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24262165

ABSTRACT

The cascade of neurotoxic events involved in neuronal degeneration suggests that it is naive to think mono-target drugs can induce disease modification by slowing the process of neurodegeneration in Parkinson's disease (PD). Employing the pharmacophore of rasagiline (N-propargyl-1-R-aminoindan), we have developed a series of novel multi-target neuroprotective drugs, including: (A) drugs [ladostigil, TV-3326 (N-propargyl-3R-aminoindan-5yl)-ethyl methylcarbamate)] with both cholinesterase-butyrylesterase (Ch-BuE) and brain-selective monamine oxidase-AB (MAO-AB) inhibitory activities and (B) iron chelator-radical scavenging drugs (M30) possessing brain-selective MAO-AB inhibitor activity and the neuroprotective-neurorescue propargylamine moiety of rasagiline. This was considered to be valid since brain MAO and iron increase in PD and aging, which could lead to oxidative stress-dependent neurodegeneration. The multi-target iron chelator, M30, has all the properties of ladostigil, but is not an acetylcholinesterase (CHE) inhibitor. However, M30 has both neuroprotective and neurorestorative activities for nigrostriatal dopamine neurons in post-lesion MPTP, lactacystin and 6-hydroxydopamine animal models of PD. The neurorestorative activity has been identified as being related to the ability of the drug to activate hypoxia-inducible factor (HIF) by inhibiting prolyl-4-hydroxylase. M30 regulates cell cycle arrest and induces the neurotrophins brain-derived neurotrophic factor (BDNF), vascular endothelial growth factor (VEGF), erythropoietin (EPO), as well as glia-derived neurotrophic factor (GDNF). These unique multiple actions of M30 make it potentially useful as a disease modifying drug for the treatment of PD.


Subject(s)
Intercellular Signaling Peptides and Proteins/metabolism , Neuroprotective Agents/therapeutic use , Parkinson Disease/drug therapy , Animals , Humans
10.
Mech Ageing Dev ; 133(5): 267-74, 2012 May.
Article in English | MEDLINE | ID: mdl-22426424

ABSTRACT

Based on a multimodal drug design paradigm, we have synthesized a multifunctional non-toxic, brain permeable iron chelating compound, M30, possessing the neuroprotective N-propargyl moiety of the anti-Parkinsonian drug, monoamine oxidase (MAO)-B inhibitor, rasagiline and the antioxidant-iron chelator moiety of an 8-hydroxyquinoline derivative of the iron chelator, VK28. Here, we report that a chronic systemic treatment of aged mice with M30 (1 and 5mg/kg; 4 times weekly for 6 months), had a significant positive impact on neuropsychiatry functions and cognitive age-related impairment. M30 significantly reduced cerebral iron accumulation as demonstrated by Perl's staining, accompanied by a marked decrease in cerebral ß-amyloid plaques. In addition, our results demonstrate that M30 caused a significant inhibition of both MAO-A and -B activities in the cerebellum of aged mice, compared with vehicle-treated aged control mice. In summary, the present study indicates that the novel MAO inhibitor/iron chelating drug, M30, acting against multiple brain targets could reverse age-associated memory impairment and provide a potential treatment against the progression of neurodegeneration in ageing.


Subject(s)
Aging/drug effects , Chelating Agents/pharmacology , Hydroxyquinolines/pharmacology , Neuroprotective Agents/pharmacology , Amyloid beta-Peptides/analysis , Animals , Cerebrum/chemistry , Chelating Agents/chemical synthesis , Cognition Disorders/drug therapy , Hydroxyquinolines/chemical synthesis , Iron/analysis , Male , Mice , Mice, Inbred C57BL , Monoamine Oxidase Inhibitors/chemical synthesis , Monoamine Oxidase Inhibitors/pharmacology , Neuroprotective Agents/chemical synthesis , Neuropsychological Tests
11.
Antioxid Redox Signal ; 17(6): 860-77, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22360429

ABSTRACT

AIMS: The aim of the present study was to evaluate the therapeutic effect of the novel neuroprotective multi-target nontoxic, lipophilic, brain permeable monoamine oxidase inhibitor and iron chelating-radical scavenging drug, M30, on the neuropathology and deficits of spatial learning and memory in amyloid precursor protein (APP) and presenilin 1 (PS1) double-transgenic (Tg) Alzheimer's disease (AD) mice. RESULTS: Here, we report that systemic treatment of APP/PS1 Tg mice with M30 for 9 months, significantly attenuated cognitive impairments in a variety of tasks of spatial learning and memory retention, working memory, learning abilities, anxiety levels, and memory for novel food and nesting behavior. Furthermore, we found that M30 reduced cerebral iron accumulation accompanied by a marked decrease in several AD-like phenotypes, including cerebral APP levels, amyloid ß (Aß) levels and plaques, phospho-APP and phospho-tau. Signaling studies revealed that M30 markedly downregulated the levels of phosphorylated cyclin-dependent kinase 5 and increased protein kinase B and glycogen synthase kinase 3ß phosphorylation. INNOVATION: Accumulation and deposition of brain iron is central to various neuropathological processes in AD, including oxidative stress, amyloid deposition, and tau phosphorylation. Thus, the concept of iron chelation holds considerable promise as a therapeutic strategy for AD pathogenesis. Here, for the first time, we demonstrated that, when systemically administered to APP/PS1 Tg mice, our novel multifunctional iron chelating/radical scavenging compound, M30, effectively reduced Aß accumulation and tau phosphorylation, and attenuated memory deficits. CONCLUSIONS: These findings suggest that M30 is a potential therapeutic agent for the prevention and treatment of AD.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Hydroxyquinolines/therapeutic use , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Behavior, Animal , Blotting, Western , Cyclin-Dependent Kinase 5/genetics , Cyclin-Dependent Kinase 5/metabolism , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Immunohistochemistry , Male , Maze Learning/drug effects , Mice , Mice, Transgenic , Phosphorylation , Presenilin-1/genetics , Presenilin-1/metabolism , Real-Time Polymerase Chain Reaction
12.
Mol Neurobiol ; 46(1): 217-20, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22847630

ABSTRACT

Based on a multimodal drug design strategy for age-related neurodegenerative diseases, we have synthesized a multifunctional nontoxic, brain-permeable iron-chelating compound, M30, possessing the neuroprotective N-propargyl moiety of the anti-Parkinsonian drug, monoamine oxidase-B inhibitor, rasagiline and the antioxidant-iron chelator moiety of the 8-hydroxyquinoline derivative of the iron chelator, VK28. In the present short overview, we describe the neuroprotective and the neurorestorative activity of M30, acting against multiple brain targets, including regulation on amyloid ß, neurogenesis, and activation of hypoxia inducible factor signaling pathways. The diverse pharmacological properties and several pathological targets of M30 make this drug potential valuable for therapeutic strategy of Alzheimer's-like neuropathology and aging.


Subject(s)
Aging/pathology , Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Cognition Disorders/drug therapy , Cognition Disorders/pathology , Hydroxyquinolines/therapeutic use , Neuroprotective Agents/therapeutic use , Aging/drug effects , Amyloidosis/drug therapy , Amyloidosis/pathology , Animals , Brain/drug effects , Brain/pathology , Hydroxyquinolines/chemistry , Hydroxyquinolines/pharmacology , Mice , Neuroprotective Agents/pharmacology
13.
Antioxid Redox Signal ; 13(6): 919-49, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20095867

ABSTRACT

Accumulating evidence suggests that many cytotoxic signals occurring in the neurodegenerative brain can initiate neuronal death processes, including oxidative stress, inflammation, and accumulation of iron at the sites of the neuronal deterioration. Neuroprotection by iron chelators has been widely recognized with respect to their ability to prevent hydroxyl radical formation in the Fenton reaction by sequestering redox-active iron. An additional neuroprotective mechanism of iron chelators is associated with their ability to upregulate or stabilize the transcriptional activator, hypoxia-inducible factor-1alpha (HIF-1alpha). HIF-1alpha stability within the cells is under the control of a class of iron-dependent and oxygen-sensor enzymes, HIF prolyl-4-hydroxylases (PHDs) that target HIF-1alpha for degradation. Thus, an emerging novel target for neuroprotection is associated with the HIF system to promote stabilization of HIF-1alpha and increase transcription of HIF-1-related survival genes, which have been reported to be regulated in patient's brains afflicted with diverse neurodegenerative diseases. In accordance, a new potential therapeutic strategy for neurodegenerative diseases is explored, by which iron chelators would inhibit PHDs, target the HIF-1-signaling pathway and ultimately activate HIF-1-dependent neuroprotective genes. This review discusses two interrelated approaches concerning therapy targets in neurodegeneration, sharing in common the implementation of iron chelation activity: antioxidation and HIF-1-pathway activation.


Subject(s)
Iron Chelating Agents/therapeutic use , Neurodegenerative Diseases/drug therapy , Neuroprotective Agents/therapeutic use , Animals , Humans , Hypoxia-Inducible Factor 1/metabolism , Neurodegenerative Diseases/metabolism , Oxidation-Reduction , Oxidative Stress
14.
EPMA J ; 1(2): 343-61, 2010 Jun.
Article in English | MEDLINE | ID: mdl-23199069

ABSTRACT

Treatment of amyotrophic lateral sclerosis (ALS) has been fueled, in part, by frustration over the shortcomings of the symptomatic drugs available, since these do not impede the progression of this disease. Currently, over 150 different potential therapeutic agents or strategies have been tested in preclinical models of ALS. Unfortunately, therapeutic modifiers of murine ALS have failed to be successfully translated into strategies for patients, probably because of differences in pharmacokinetics of the therapeutic agents, route of delivery, inefficiency of the agents to affect the distinct pathologies of the disease or inherent limitations of the available animal models. Given the multiplicity of the pathological mechanisms implicated in ALS, new therapies should consider the simultaneous manipulation of multiple targets. Additionally, a better management of ALS therapy should include understanding the interactions between potential risk factors, biomarkers and heterogeneous clinical features of the patients, aiming to manage their adverse events or personalize the safety profile of these agents. This review will discuss novel pharmacological approaches concerning adjusted therapy for ALS patients: iron-binding brain permeable multimodal compounds, genetic manipulation and cell-based treatment.

15.
ACS Chem Biol ; 4(11): 910-4, 2009 Nov 20.
Article in English | MEDLINE | ID: mdl-19715343

ABSTRACT

Reactive oxygen species are heavily involved in the pathogenesis of diabetes mellitus (DM) because the insulin-producing beta cells are particularly vulnerable to free-radical-mediated cytotoxicity. Catalytic anti-oxidants have been successfully applied for attenuation of DM and its consequences, but most recent research revealed that preventing the nitration of vital proteins/enzymes might be an even more powerful strategy. We now report an unprecedented efficiency of manganese(III) corroles regarding the protection of rat pancreatic beta cells against intracellular nitration by peroxynitrite and subsequent cell death. A comparison between analogous corroles and porphyrin metal complexes reveals significant superiority of the former in all examined aspects. This is particularly true for the positively-charged manganese(III) corrole, which decomposes peroxynitrite fast enough and through a unique catalytic mechanism that is devoid of potentially nitrating reaction intermediates.


Subject(s)
Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Insulin/biosynthesis , Manganese/pharmacology , Nitrates/metabolism , Animals , Cell Death , Cell Line , Insulin-Secreting Cells/cytology , Intracellular Space/drug effects , Intracellular Space/metabolism , Rats
16.
J Neurochem ; 103(3): 962-71, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17680997

ABSTRACT

Activin is a member of the transforming growth factor-beta superfamily which comprises a growing list of multifunctional proteins that function as modulators of cell proliferation, differentiation, hormone secretion and neuronal survival. This study examined the neuroprotective effect of both Activin A and B in serum withdrawal and oxidative stress apoptotic cellular models and investigated the expression of pro- and anti-apoptotic proteins, which may account for the mechanism of Activin-induced neuroprotection. Here, we report that recombinant Activin A and B are neuroprotective against serum deprivation- and toxin- [either the parkinsonism-inducing neurotoxin, 6-hydroxydopamine (6-OHDA) or the peroxynitrite donor, 3-(4-morpholinyl) sydnonimine hydrochloride (SIN-1)] induced neuronal death in human SH-SY5Y neuroblastoma cells. Furthermore, we demonstrate for the first time that transient transfection with Activin betaA or betaB significantly protect SH-SY5Y and rat pheochromocytoma PC12 cells against serum withdrawal-induced apoptosis. This survival effect is mediated by the Bcl-2 family members and involves inhibition of caspase-3 activation; reduction of cleaved poly-ADP ribose polymerase and phosphorylated H2A.X protein levels and elevation of tyrosine hydroxylase expression. These results indicate that both Activin-A and -B share the potential to induce neuroprotective activity and thus may have positive impact on aging and neurodegenerative diseases to retard the accelerated rate of neuronal degeneration.


Subject(s)
Activins/pharmacology , Nerve Degeneration/drug therapy , Neurons/drug effects , Neuroprotective Agents/pharmacology , Activins/biosynthesis , Activins/genetics , Animals , Apoptosis/drug effects , Apoptosis/physiology , Apoptosis Regulatory Proteins/drug effects , Apoptosis Regulatory Proteins/metabolism , Brain/drug effects , Brain/metabolism , Brain/physiopathology , Cell Line, Tumor , Cell Survival/drug effects , Culture Media, Serum-Free/toxicity , Histones/drug effects , Histones/metabolism , Humans , Molsidomine/analogs & derivatives , Molsidomine/antagonists & inhibitors , Molsidomine/toxicity , Nerve Degeneration/physiopathology , Nerve Degeneration/prevention & control , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/prevention & control , Neurons/metabolism , Oxidative Stress/drug effects , Oxidative Stress/physiology , Oxidopamine/antagonists & inhibitors , Oxidopamine/toxicity , PC12 Cells , Poly(ADP-ribose) Polymerases/drug effects , Poly(ADP-ribose) Polymerases/metabolism , Rats , Transfection , Tyrosine 3-Monooxygenase/drug effects , Tyrosine 3-Monooxygenase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL