Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Food Chem Toxicol ; 136: 111079, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31891754

ABSTRACT

Cisplatin has dramatically improved the survival rate of cancer patients, but it has also increased the prevalence of hearing and neurological deficits in this population. Cisplatin induces ototoxicity, peripheral (most prevalent) and central (rare) neurotoxicity. This review addresses the ototoxicity and the neurotoxicity associated with cisplatin-based chemotherapy, providing an integrated view of the potential protective agents that have been evaluated in vitro, in vivo and in clinical trials, their targets and mechanisms of protection and their effects on the antitumor activity of cisplatin. So far, the findings are insufficient to support the use of any oto- or neuroprotective agent before, during or after cisplatin chemotherapy. Despite their promising effects in vitro and in animal studies, many agents have not been evaluated in clinical trials. Additionally, the clinical trials have limitations concerning the sample size, controls, measurement, heterogeneous groups, several arms of treatment, short follow-up or no blinding. Besides that, for most agents, the effects on the antitumor activity of cisplatin have not been evaluated in tumor-bearing animals, which discourages clinical trials. Further well-designed randomized controlled clinical trials are necessary to definitely demonstrate the effectiveness of the oto- or neuroprotective agents proposed by animal and in vitro studies.


Subject(s)
Antineoplastic Agents/adverse effects , Cisplatin/adverse effects , Neurotoxicity Syndromes/prevention & control , Ototoxicity/prevention & control , Protective Agents/administration & dosage , Animals , Antineoplastic Agents/therapeutic use , Cisplatin/therapeutic use , Humans , Neoplasms/drug therapy , Neurotoxicity Syndromes/etiology , Ototoxicity/etiology
2.
Neurotox Res ; 36(1): 175-192, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31016689

ABSTRACT

Peripheral sensory neuropathy (PSN) is a well-known side effect of cisplatin characterized by axonal damage. In the early stage of neurotoxicity, cisplatin affects proteins that modulate neurite outgrowth and neuroplasticity, without inducing mitochondrial damage or apoptosis. There are no preventive therapies for cisplatin-induced peripheral neuropathy; therefore, measures to improve axonal growth and connectivity would be beneficial. Caffeic acid phenethyl ester (CAPE) is a bioactive component of propolis with neurotrophic and neuroprotective activities. We have recently showed that CAPE protects against cisplatin-induced neurotoxicity by activating NGF high-affinity receptors (trkA) and inducing neuroplasticity. We have now assessed other potential early targets of cisplatin and additional mechanisms involved in the neuroprotection of CAPE. Cisplatin reduced axonal cytoskeletal proteins (F-actin and ß-III-tubulin) without inducing oxidative damage in PC12 cells. It also reduced energy-related proteins (AMPK α, p-AMPK α, and SIRT1) and glucose uptake. At this stage of neurotoxicity, glutamate excitotoxicity is not involved in the toxicity of cisplatin. CAPE attenuated the downregulation of the cytoskeleton and energy-related markers as well as SIRT1 and phosphorylated AMPK α. Moreover, the neuroprotective mechanism of CAPE also involves the activation of the neurotrophic signaling pathways MAPK/Erk and PI3k/Akt. The PI3K/Akt pathway is involved in the upregulation of SIRT1 induced by CAPE, but not in the upregulation of cytoskeletal proteins. Altogether, these findings suggest that the neuroprotective effect of CAPE against cisplatin-induced neurotoxicity involves both (a) a neurotrophic mechanism that mimics the mechanism triggered by the NGF itself and (b) a non-neurotrophic mechanism that upregulates the cytoskeletal proteins.


Subject(s)
Caffeic Acids/pharmacology , Cisplatin/toxicity , Neurons/drug effects , Neuroprotective Agents/pharmacology , Phenylethyl Alcohol/analogs & derivatives , Signal Transduction/drug effects , AMP-Activated Protein Kinases/metabolism , Animals , COS Cells , Cell Differentiation/drug effects , Chlorocebus aethiops , Cytoskeletal Proteins/metabolism , Glucose/metabolism , MAP Kinase Signaling System/drug effects , Neurons/metabolism , PC12 Cells , Phenylethyl Alcohol/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Reactive Oxygen Species/metabolism , Sirtuin 1/metabolism
3.
Peptides ; 104: 24-34, 2018 06.
Article in English | MEDLINE | ID: mdl-29684590

ABSTRACT

Venom small peptides that target neurotrophin receptors might be beneficial in neurodegeneration, including Parkinsons disease (PD). Their small size, ease of synthesis, structural stability and target selectivity make them important tools to overcome the limitations of endogenous neurotrophins as therapeutic agents. Additionally, they might be optimized to improve resistance to enzymatic degradation, bioavailability, potency and, mainly, lipophilicity, important to cross the blood brain barrier (BBB). Here, we evaluated the neuroprotective effects and mechanisms of the synthetic snake-venom-based peptide p-BTX-I (Glu-Val-Trp) in PC12 cells treated with MPP+ (1-methyl-4-phenylpyridinium), a dopaminergic neurotoxin that induces Parkinsonism in vivo. The peptide p-BTX-I induced neuritogenesis, which was reduced by (i) k252a, antagonist of the NGF-selective receptor, trkA (tropomyosin receptor kinase A); (ii) LY294002, inhibitor of the PI3 K/AKT pathway and (iii) U0126, inhibitor of the MAPK-ERK pathway. Besides that, p-BTX-I also increased the expression of GAP-43 and synapsin, which are molecular markers of axonal growth and synaptic communication. In addition, the peptide increased the viability and differentiation of cells exposed to MPP+, known to inhibit neuritogenesis. Altogether, our findings suggest that the synthetic peptide p-BTX-I protects PC12 cells from MPP+ toxicity by a mechanism that mimics the neurotrophic action of NGF. Therefore, the molecular structure of p-BTX-I might be relevant in the development of drugs aimed at restoring the axonal connectivity in neurodegenerative processes.


Subject(s)
1-Methyl-4-phenylpyridinium/toxicity , Oligopeptides/chemical synthesis , Oligopeptides/pharmacology , Signal Transduction/drug effects , Snake Venoms/chemistry , Animals , Cell Survival/drug effects , GAP-43 Protein/metabolism , Nerve Growth Factor/metabolism , Oligopeptides/chemistry , PC12 Cells , Rats , Receptor, trkB/metabolism , Synapsins/metabolism
4.
Neurotox Res ; 34(1): 32-46, 2018 07.
Article in English | MEDLINE | ID: mdl-29260495

ABSTRACT

Cisplatin is a highly effective chemotherapeutic drug that is toxic to the peripheral nervous system. Findings suggest that axons are early targets of the neurotoxicity of cisplatin. Although many compounds have been reported as neuroprotective, there is no effective treatment against the neurotoxicity of cisplatin. Caffeic acid phenethyl ester (CAPE) is a propolis component with neuroprotective potential mainly attributed to antioxidant and anti-inflammatory mechanisms. We have recently demonstrated the neurotrophic potential of CAPE in a cellular model of neurotoxicity related to Parkinson's disease. Now, we have assessed the neurotrophic and neuroprotective effects of CAPE against cisplatin-induced neurotoxicity in PC12 cells. CAPE (10 µM) attenuated the inhibition of neuritogenesis and the downregulation of markers of neuroplasticity (GAP-43, synapsin I, synaptophysin, and 200-kD neurofilament) induced by cisplatin (5 µM). This concentration of cisplatin does not affect cell viability, and it was used in order to assess the early neurotoxic events triggered by cisplatin. When a lethal dose of cisplatin was used (IC50 = 32 µM), CAPE (10 µM) increased cell viability. The neurotrophic effect of CAPE is not dependent on NGF nor is it additive to the effect of NGF, but it might involve the activation of the NGF-high-affinity receptors (trkA). The involvement of other neurotrophin receptors such as trkB and trkC is unlikely. This is the first study to demonstrate the protective potential of CAPE against the neurotoxicity of cisplatin and to suggest the involvement of trkA receptors in the neuroprotective mechanism of CAPE. Based on these findings, the beneficial effect of CAPE on cisplatin-induced peripheral neuropathy should be further investigated.


Subject(s)
Caffeic Acids/pharmacology , Cisplatin/pharmacology , Nerve Growth Factor/metabolism , Neuroprotective Agents/pharmacology , Neurotoxins/pharmacology , Phenylethyl Alcohol/analogs & derivatives , Signal Transduction/drug effects , Analysis of Variance , Animals , Cell Differentiation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , GAP-43 Protein/metabolism , Neuroblastoma/pathology , Neurofilament Proteins/metabolism , Neuronal Outgrowth/drug effects , PC12 Cells/drug effects , Phenylethyl Alcohol/pharmacology , Rats , Synapsins/metabolism , Synaptophysin/metabolism
5.
Toxicol In Vitro ; 39: 84-92, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27939611

ABSTRACT

Some organophosphorus compounds (OPs) induce a neurodegenerative disorder known as organophosphate-induced delayed neuropathy (OPIDN), which is related to irreversible inhibition of neuropathy target esterase (NTE) and impairment of neurite outgrowth. The present study addresses the effects of trichlorfon, mipafox (neuropathic model) and paraoxon (non-neuropathic model) on neurite outgrowth and neuroplasticity-related proteins in retinoic-acid-stimulated SH-SY5Y cells, a cellular model widely used to study the neurotoxicity of OPs. Mipafox (20µM) decreased cellular differentiation and the expression of neurofilament 200 (NF-200), growth associated- (GAP-43) and synaptic proteins (synapsin I and synaptophysin); whereas paraoxon (300µM) induced no effect on cellular differentiation, but significant decrease of NF-200, GAP-43, synapsin I and synaptophysin as compared to controls. However, the effects of paraoxon on these proteins were significantly lower than the effects of mipafox. In conclusion, axonal cytoskeletal proteins, as well as axonal plasticity-related proteins are more effectively affected by neuropathic (mipafox) than by non-neuropathic (paraoxon) OPs, suggesting that they might play a role in the mechanism of OPIDN. At high concentration (1mM), trichlorfon induced effects similar to those of the neuropathic OP, mipafox (20µM), but also caused high inhibition of AChE. Therefore, these effects are unlikely to occur in humans at non-lethal doses of trichlorfon.


Subject(s)
Axons/drug effects , Cholinesterase Inhibitors/toxicity , Insecticides/toxicity , Isoflurophate/analogs & derivatives , Paraoxon/toxicity , Trichlorfon/toxicity , Acetylcholinesterase/metabolism , Carboxylic Ester Hydrolases/metabolism , Cell Line, Tumor , Cytoskeleton/drug effects , GAP-43 Protein/metabolism , Humans , Isoflurophate/toxicity , L-Lactate Dehydrogenase/metabolism , Neuronal Plasticity , Synapsins/metabolism
6.
Chem Biol Interact ; 261: 86-95, 2017 Jan 05.
Article in English | MEDLINE | ID: mdl-27871898

ABSTRACT

Beta-caryophyllene (BCP) is a phytocannabinoid whose neuroprotective activity has been mainly associated with selective activation of cannabinoid-type-2 (CB2) receptors, inhibition of microglial activation and decrease of inflammation. Here, we addressed the potential of BCP to induce neuritogenesis in PC12 cells, a model system for primary neuronal cells that express trkA receptors, respond to NGF and do not express CB2 receptors. We demonstrated that BCP increases the survival and activates the NGF-specific receptor trkA in NGF-deprived PC12 cells, without increasing the expression of NGF itself. The neuritogenic effect of BCP in PC12 cells was abolished by k252a, an inhibitor of the NGF-specific receptor trkA. Accordingly, BCP did not induce neuritogenesis in SH-SY5Y neuroblastoma cells, a neuronal model that does not express trkA receptors and do not respond to NGF. Additionally, we demonstrated that BCP increases the expression of axonal-plasticity-associated proteins (GAP-43, synapsin and synaptophysin) in PC12 cells. It is known that these proteins are up-regulated by NGF in neurons and neuron-like cells, such as PC12 cells. Altogether, these findings suggest that BCP activates trka receptors and induces neuritogenesis by a mechanism independent of NGF or cannabinoid receptors. This is the first study to show such effects of BCP and their beneficial role in neurodegenerative processes should be further investigated.


Subject(s)
Cannabinoids/pharmacology , Neurites/metabolism , Neurogenesis/drug effects , Receptors, Cannabinoid/metabolism , Sesquiterpenes/pharmacology , Animals , Carbazoles/pharmacology , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Humans , Indole Alkaloids/pharmacology , Nerve Growth Factor/pharmacology , Nerve Tissue Proteins/metabolism , Neurites/drug effects , PC12 Cells , Polycyclic Sesquiterpenes , Rats , Receptor, trkA/antagonists & inhibitors , Receptor, trkA/metabolism
7.
Neurochem Res ; 41(11): 2993-3003, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27473385

ABSTRACT

Cisplatin is the most effective and neurotoxic platinum chemotherapeutic agent. It induces a peripheral neuropathy characterized by distal axonal degeneration that might progress to degeneration of cell bodies and apoptosis. Most symptoms occur nearby distal axonal branches and axonal degeneration might induce peripheral neuropathy regardless neuronal apoptosis. The toxic mechanism of cisplatin has been mainly associated with DNA damage, but cisplatin might also affect neurite outgrowth. Nevertheless, the neurotoxic mechanism of cisplatin remains unclear. We investigated the early effects of cisplatin on axonal plasticity by using non-cytotoxic concentrations of cisplatin and PC12 cells as a model of neurite outgrowth and differentiation. PC12 cells express NGF-receptors (trkA) and respond to NGF by forming neurites, branches and synaptic vesicles. For comparison, we used a neuronal model (SH-SY5Y cells) that does not express trkA nor responds to NGF. Cisplatin did not change NGF expression in PC12 cells and decreased neurite outgrowth in both models, suggesting a NGF/trkA independent mechanism. It also reduced axonal growth (GAP-43) and synaptic (synapsin I and synaptophysin) proteins in PC12 cells, without inducing mitochondrial damage or apoptosis. Therefore, cisplatin might affect axonal plasticity before DNA damage, NGF/trkA down-regulation, mitochondrial damage or neuronal apoptosis. This is the first study to show that neuroplasticity-related proteins might be early targets of the neurotoxic action of cisplatin and their role on cisplatin-induced peripheral neuropathy should be investigated in vivo.


Subject(s)
Cisplatin/pharmacology , Nerve Growth Factor/metabolism , Neuronal Outgrowth/drug effects , Neuronal Plasticity/drug effects , Animals , Axons/drug effects , Axons/metabolism , Cell Differentiation/drug effects , Down-Regulation/drug effects , GAP-43 Protein/metabolism , Neurites/drug effects , Neurites/physiology , PC12 Cells , Rats , Receptors, Nerve Growth Factor/metabolism
8.
Toxicol In Vitro ; 30(1 Pt B): 231-40, 2015 Dec 25.
Article in English | MEDLINE | ID: mdl-26556726

ABSTRACT

Cannabidiol (CBD) is a non-psychoactive constituent of Cannabis sativa with potential to treat neurodegenerative diseases. Its neuroprotection has been mainly associated with anti-inflammatory and antioxidant events; however, other mechanisms might be involved. We investigated the involvement of neuritogenesis, NGF receptors (trkA), NGF, and neuronal proteins in the mechanism of neuroprotection of CBD against MPP(+) toxicity in PC12 cells. CBD increased cell viability, differentiation, and the expression of axonal (GAP-43) and synaptic (synaptophysin and synapsin I) proteins. Its neuritogenic effect was not dependent or additive to NGF, but it was inhibited by K252a (trkA inhibitor). CBD did not increase the expression of NGF, but protected against its decrease induced by MPP(+), probably by an indirect mechanism. We also evaluated the neuritogenesis in SH-SY5Y cells, which do not express trkA receptors. CBD did not induce neuritogenesis in this cellular model, which supports the involvement of trkA receptors. This is the first study to report the involvement of neuronal proteins and trkA in the neuroprotection of CBD. Our findings suggest that CBD has a neurorestorative potential independent of NGF that might contribute to its neuroprotection against MPP(+), a neurotoxin relevant to Parkinson's disease.


Subject(s)
1-Methyl-4-phenylpyridinium/toxicity , Cannabidiol/pharmacology , Nerve Tissue Proteins/biosynthesis , Neurites/drug effects , Neuroprotective Agents/pharmacology , Parkinson Disease/prevention & control , Receptor, trkA/physiology , Animals , Axons/metabolism , Humans , Nerve Growth Factor/physiology , Neurites/physiology , Neuroblastoma/pathology , PC12 Cells , Rats , Synapses/metabolism , Up-Regulation
9.
Neurotoxicology ; 45: 131-8, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25454720

ABSTRACT

Neurite loss is an early event in neurodegenerative diseases; therefore, the regeneration of the network of neurites constitutes an interesting strategy of treatment for such disorders. Neurotrophic factors play a critical role in neuronal regeneration, but their clinical use is limited by their inability to cross the blood brain barrier. Oxidative and inflammatory events are implicated in neurodegeneration and antioxidant compounds have been suggested as potential neuroprotectors. The protective potential of CAPE (caffeic acid phenethyl ester) has been shown in different models of neurotoxicity (in vitro and in vivo) and it has been associated with immune-modulatory, antioxidant and anti-inflammatory properties; however, other mechanisms might be involved. The present study demonstrates that CAPE protects PC12 cells from the cellular death induced by the dopaminergic neurotoxin MPP(+) by increasing the network of neurites. Results showed that CAPE induced the formation, elongation and ramification of neurites in PC12 cells non-stimulated with NGF (nerve growth factor) and inhibited the shortage of neurites induced by the dopaminergic neurotoxin. These effects were associated with increased expression of neuron-typical proteins responsible for axonal growth (GAP-43) and synaptogenesis (synaptophysin and synapsin I). It is noteworthy that, unlike neurotrophins, CAPE would be able to cross the blood brain barrier and exert its neurotrophic effects in the brain. This study corroborates the therapeutic potential of CAPE in neurodegenerative diseases while proposes the involvement of neuroplasticity in the mechanism of neuroprotection.


Subject(s)
1-Methyl-4-phenylpyridinium/toxicity , Caffeic Acids/therapeutic use , Neurites/drug effects , Neuroprotective Agents/therapeutic use , Phenylethyl Alcohol/analogs & derivatives , Animals , Cell Death/drug effects , Cell Differentiation/drug effects , Dose-Response Relationship, Drug , GAP-43 Protein/metabolism , Nerve Growth Factor/metabolism , PC12 Cells , Phenylethyl Alcohol/therapeutic use , Rats , Synapsins/metabolism , Synaptophysin/metabolism
10.
Braz. j. pharm. sci ; 49(1): 149-154, Jan.-Mar. 2013. ilus, graf, tab
Article in English | LILACS | ID: lil-671410

ABSTRACT

Organophosphates (OPs) are widely used as pesticides, and its urinary metabolites as well as the blood cholinesterases (ChEs) activity have been reported as possible biomarkers for the assessment of this pesticide exposure. Moreover, the OPs can induce mutagenesis, and the bone marrow micronucleus test is an efficient way to assess this chromosomal damage. This paper reports a study carried out to verify the correlation among the disulfoton exposure, blood ChEs activity, urinary diethyl thiophosphate (DETP), and diethyl dithiophosphate (DEDTP), as well as micronucleated polychromatic erythrocytes (MNPCEs) frequency. Four groups of rats (n=12) were exposed to disulfoton at 0, 2.8, 4.7, and 6.6 mg kg-1 body weight. The blood ChEs activity, urinary DETP and DEDTP concentrations, and MNPCEs frequency were determined. It was observed that the plasmatic and erythrocytary ChEs activity decreased from 2.9% to 0.5% and from 35.9 to 3.3%, respectively, when the disulfoton dose was increased from 0 to 6.6 mg kg-1 (correlation of 0.99). Urinary DETP and DEDTP concentrations, as well as the MNPCEs frequency, increased from 0 to 6.58 µg mL-1, from 0 to 0.04 µg mL-1, and from 0 to 1.4%, respectively, when the disulfoton dose was increased from 0 to 6.58 mg kg-1 body weight.


Os organofosforados (OPs) são amplamente usados como praguicidas e a atividade da colinesterase sanguínea bem como os metabólitos urinários desses praguicidas têm sido reportados como biomarcadores eficazes para avaliar casos de exposição. Além disso, os OPs podem induzir mutagênese e o teste de micronúcleo de medula óssea é uma boa alternativa para avaliar os danos cromossômicos. Esse artigo reporta um estudo sobre a correlação entre a exposição a dissulfoton, a atividade da colinesterase sanguínea, a excreção urinária de dietil tiofosfato e dietil ditiofosfato e a frequência de micronúcleos em eritrócitos policromáticos. Quatro grupos de ratos (n=12) foram expostos a dissulfotom nas doses de 0, 2,8, 4,7, e 6,6 mg kg-1 de peso corpóreo. A atividade da colinesterase sanguínea as concentrações urinárias de dietil tiofosfato e dietil ditiofosfato e a frequência de micronúcleos foram determinadas. Os resultados demonstraram que as atividades da colinesterase plasmática e eritrocitária diminuíram de 2,9 para 0,5% e de 35,9 para 3,3% , respectivamente, quando a dose de dissulfoton foi aumentada de 0 para 6,6 mg kg-1 (correlação de 0,99). As concentrações urinárias de dietil tiofosfato e dietil ditiofosfato bem como a frequência de micronúcleos aumentaram de 0 a 6,56 µg mL-1, 0 a 0.04 µg mL-1 e de 0 a 1.4%, respectivamente, quando a dose de dissulfotom foi aumentada de 0 a 6,58 mg kg-1.


Subject(s)
Rats , Organophosphorus Compounds/pharmacokinetics , Cholinesterases/analysis , Erythrocytes/classification , Pesticide Exposure , Disulfoton/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL
...