Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Naunyn Schmiedebergs Arch Pharmacol ; 396(8): 1787-1796, 2023 08.
Article in English | MEDLINE | ID: mdl-36843128

ABSTRACT

Doxycycline (DOX) is a widely used antibiotic that is able to cross the blood-brain barrier. Several studies have shown its neuroprotective effect against neurodegeneration and have associated it with antioxidant, anti-apoptotic, and anti-inflammatory mechanisms. We have recently demonstrated that DOX mimics nerve growth factor (NGF) signaling in PC12 cells. However, the involvement of this mechanism in the neuroprotective effect of DOX is unknown. Axonal degeneration and synaptic loss are key events at the early stages of neurodegeneration, and precede the neuronal death in neurodegenerative diseases, including Parkinson's disease (PD). Therefore, the regeneration of the axonal and synaptic network might be beneficial in PD. The effect of DOX in PC12 cells treated with the Parkinsonian neurotoxin 1-methyl-4-phenylpyridinium (MPP+) was addressed. Doxycycline reduced the inhibition of neuritogenesis induced by MPP+, even in cells deprived of NGF. The mechanism involved the upregulation of GAP-43, synapsin I, ß-III-tubulin, F-actin, and neurofilament-200, proteins that are associated with axonal and synaptic plasticity. Considering the role of axonal degeneration and synaptic loss at the initial stages of PD, the recent advances in early diagnosis of neurodegeneration, and the advantages of drug repurposing, doxycycline is a promising candidate to treat PD.


Subject(s)
Neuroprotective Agents , Parkinson Disease , Rats , Animals , Humans , Up-Regulation , Doxycycline/pharmacology , Doxycycline/therapeutic use , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Nerve Growth Factor/metabolism , Nerve Growth Factor/therapeutic use , Proteins/metabolism , Parkinson Disease/drug therapy , PC12 Cells , Tubulin/metabolism , 1-Methyl-4-phenylpyridinium/toxicity , 1-Methyl-4-phenylpyridinium/therapeutic use
2.
Naunyn Schmiedebergs Arch Pharmacol ; 395(6): 659-672, 2022 06.
Article in English | MEDLINE | ID: mdl-35246694

ABSTRACT

Neurodegenerative diseases are characterized by progressive loss of the structure and function of specific neuronal populations, and have been associated with reduced neurotrophic support. Neurotrophins, like NGF (nerve growth factor), are endogenous proteins that induce neuritogenesis and modulate axonal growth, branching, and synapsis; however, their therapeutic application is limited mainly by low stability, short half-life, and inability to cross the blood-brain barrier (BBB). Small neurotrophic molecules that have suitable pharmacokinetics and are able to cross the BBB are potential candidates for neuroprotection. Baccharin is a bioactive small molecule isolated from Brazilian green propolis. In the present study, we investigated the neurotrophic and neuroprotective potential of baccharin in the PC12 cell neuronal model. We used pharmacological inhibitors (K252a, LY294002, and U0126), and ELISA (phospho-trkA, phospho-Akt, and phospho-MEK) to investigate the involvement of trkA receptor, PI3k/Akt pathway, and MAPK/Erk pathway, respectively. Additionally, we evaluated the expression of axonal (GAP-43) and synaptic (synapsin I) proteins by western blot. The results showed that baccharin induces neuritogenesis in NGF-deprived PC12 cells, through activation of trkA receptor and the downstream signaling cascades (PI3K/Akt and MAPK/ERK), which is the same neurotrophic pathway activated by NGF in PC12 cells and neurons. Baccharin also induced the expression of GAP-43 and synapsin I, which mediate axonal and synaptic plasticity, respectively. Additionally, in silico predictions of baccharin showed favorable physicochemical properties, pharmacokinetics, drug-likeness, and medicinal chemistry friendliness. Altogether, these findings suggest that baccharin is a promising neurotrophic agent whose therapeutic application in neurodegeneration should be further investigated.


Subject(s)
Nerve Growth Factor , Propolis , Animals , Brazil , GAP-43 Protein/metabolism , Nerve Growth Factor/metabolism , Nerve Growth Factor/pharmacology , PC12 Cells , Phosphatidylinositol 3-Kinases/metabolism , Propolis/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Rats , Receptor, trkA/metabolism , Signal Transduction , Synapsins/metabolism , Trichothecenes
3.
Neurotox Res ; 39(3): 886-896, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33666886

ABSTRACT

Carvacrol (CARV) is a phytochemical widely used as flavoring, preservative, and fragrance in food and cosmetic industries. CARV is able to cross the blood-brain barrier (BBB) and has demonstrated protective potential against neurodegenerative diseases by several mechanisms, including antioxidant, anti-inflammatory, anticholinesterase, and antiapoptotic effects. However, it is not known whether CARV is able to modulate axonal and synaptic plasticity, crucial events in cognition, memory, and learning. Abnormalities in axonal and synaptic plasticity, low levels of neurotrophins, and bioenergetic failure have been associated with the pathogenesis of neurodegenerative diseases, including Parkinson's (PD) and Alzheimer's diseases (ADs). Small lipophilic molecules with neurotrophic activity might be able to restore the axonal and synaptic networks that are lost in neurodegenerative processes. Therefore, this study investigated the neurotrophic potential of CARV in PC12 cell-based neuronal model. Carvacrol induced neurite outgrowth by activating the NGF high-affinity trkA receptor and the downstream PI3K-AKT and MAPK-ERK pathways, without depending on NGF. In addition, CARV increased the expression of proteins involved in neuronal plasticity (ß-tubulin III, F-actin, 200-kDa neurofilament, GAP-43 and synapsin-I) and improved bioenergetics (AMPKα, p-AMPKα, and ATP). Our study showed, for the first time, a promising neurotrophic mechanism of CARV that could be beneficial in neurodegenerative and neurological diseases.


Subject(s)
Axons/drug effects , Cymenes/pharmacology , Nerve Growth Factors/pharmacology , Nerve Regeneration/drug effects , Synapses/drug effects , Animals , Axons/physiology , Cell Survival/drug effects , Cell Survival/physiology , Dose-Response Relationship, Drug , Nerve Growth Factor/pharmacology , Nerve Regeneration/physiology , PC12 Cells , Rats , Synapses/physiology
4.
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
5.
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
6.
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
7.
Toxicol In Vitro ; 29(5): 1079-87, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25910916

ABSTRACT

Organophosphorus-induced delayed neuropathy (OPIDN) is a central-peripheral distal axonopathy that develops 8-14 days after poisoning by a neuropathic organophosphorus compound (OP). Several OPs that caused OPIDN were withdrawn from the agricultural market due to induction of serious delayed effects. Therefore, the development of in vitro screenings able to differentiate neuropathic from non-neuropathic OPs is of crucial importance. Thus, the aim of this study was to evaluate the differences in the neurotoxic effects of mipafox (neuropathic OP) and paraoxon (non-neuropathic OP) in SH-SY5Y human neuroblastoma cells, using the inhibition and aging of neuropathy target esterase (NTE), inhibition of acetylcholinesterase (AChE), activation of calpain, neurite outgrowth, cytotoxicity and intracellular calcium as indicators. Additionally, the potential of fenamiphos and profenofos to cause acute and/or delayed effects was also evaluated. Mipafox had the lowest IC50 and induced the highest percentage of aging of NTE among the OPs evaluated. Only mipafox was able to cause calpain activation after 24 h of incubation. Concentrations of mipafox and fenamiphos which inhibited at least 70% of NTE were also able to reduce neurite outgrowth. Cytotoxicity was higher in non-neuropathic than in neuropathic OPs while the intracellular calcium levels were higher in neuropathic than in non-neuropathic OPs. In conclusion, the SH-SY5Y cellular model was selective to differentiate neuropathic from non-neuropathic OPs; fenamiphos, but not profenofos presented results compatible with the induction of OPIDN.


Subject(s)
Cholinesterase Inhibitors/toxicity , Insecticides/toxicity , Organophosphorus Compounds/toxicity , Acetylcholinesterase/metabolism , Calcium/metabolism , Calpain/metabolism , Carboxylic Ester Hydrolases/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Humans , Neurites/drug effects , Neurotoxicity Syndromes
8.
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
9.
Arch Toxicol ; 86(8): 1233-50, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22382776

ABSTRACT

Cisplatin is a highly effective antitumor agent whose clinical application is limited by the inherent nephrotoxicity. The current measures of nephroprotection used in patients receiving cisplatin are not satisfactory, and studies have focused on the investigation of new possible protective strategies. Many pathways involved in cisplatin nephrotoxicity have been delineated and proposed as targets for nephroprotection, and many new potentially protective agents have been reported. The multiple pathways which lead to renal damage and renal cell death have points of convergence and share some common modulators. The most frequent event among all the described pathways is the oxidative stress that acts as both a trigger and a result. The most exploited pathways, the proposed protective strategies, the achievements obtained so far as well as conflicting data are summarized and discussed in this review, providing a general view of the knowledge accumulated with past and recent research on this subject.


Subject(s)
Antineoplastic Agents/toxicity , Cisplatin/toxicity , Kidney Diseases/chemically induced , Kidney Diseases/prevention & control , Kidney/drug effects , Protective Agents/therapeutic use , Animals , Cell Death/drug effects , Cytoprotection , Humans , Kidney/metabolism , Kidney/pathology , Kidney Diseases/metabolism , Kidney Diseases/pathology , Signal Transduction/drug effects
10.
Chem Biol Interact ; 179(2-3): 402-6, 2009 May 15.
Article in English | MEDLINE | ID: mdl-19330886

ABSTRACT

PDT has been used in the treatment of malignant brain tumors for the last 2 decades. It is based on the interaction of a photosensitizer (PS) and light of an appropriate wavelength, with generation of oxygen species, mainly singlet oxygen. Brain is particularly susceptible to oxidative stress; therefore the study of PDT effects on cerebral mitochondria might provide mechanistic insights into the action of the therapy, contributing to its optimization. In the present study, we addressed the mitochondrial toxicity of the second generation PS, zinc phthalocyanine tetrasulfonate (ZnPcS4), on rat brain isolated mitochondria, by investigating both intrinsic toxicity and photodynamic action. At higher concentrations (15 and 25 microM/mg protein) ZnPcS4 caused (a) inhibition of state-3 respiration and (b) decrease of RCR and ADP/O. Electrochemical potential, state-4 respiration and Ca2+ retention capacity were not affected. Cytochrome c release was not observed. Coupled with 600 or 1800 mJ/cm2 laser irradiation, ZnPcS4 (5 microM/mg protein) caused more intense effects on state 3, RCR and ADP/O; moreover state-4 respiration and membrane potential were affected. Besides that, Ca2+ and cytochrome c release were induced. Cyclosporine A (CsA) decreased Ca2+ release and ameliorated the electrochemical potential, suggesting that membrane permeability transition (MPT) might be involved in the photodynamic effect. The low intrinsic toxicity and the high photodynamic effect on rat brain mitochondria induced by ZnPcS4, allied to its improved photophysical properties, might indicate its potential for the treatment of malignant brain tumors.


Subject(s)
Brain/cytology , Indoles/pharmacology , Mitochondria/drug effects , Organometallic Compounds/pharmacology , Photochemotherapy , Animals , Calcium/metabolism , Calcium/radiation effects , Cytochromes c/drug effects , Cytochromes c/metabolism , Cytochromes c/radiation effects , Lasers , Mitochondria/metabolism , Mitochondria/radiation effects , Mitochondrial Membranes/drug effects , Mitochondrial Membranes/radiation effects , Rats , Time Factors
11.
Chem Biol Interact ; 170(3): 177-86, 2007 Dec 15.
Article in English | MEDLINE | ID: mdl-17850778

ABSTRACT

Cisplatin is one of the most effective chemotherapeutic agents. However, at higher doses liver injury may occur. The purpose of this study was to explore whether the hydroxyl radical scavenger dimethylthiourea (DMTU) protects against cisplatin-induced oxidative damage in vivo and to define the mitochondrial pathways involved in cytoprotection. Adult male Wistar rats (200-220 g) were divided into four groups of eight animals each. The control group was treated only with an intraperitoneal (i.p.) injection of saline solution (1 ml/100 g body weight). The DMTU group was given only DMTU (500 mg/kg body weight, i.p), followed by 125 mg/kg body weight, i.p. (twice a day) until sacrifice. The cisplatin group was given a single injection of cisplatin (10 mg/kg body weight, i.p.). The DMTU+cisplatin group was given DMTU (500 mg/kg body weight, i.p.), just before the cisplatin injection (10 mg/kg body weight, i.p.), followed by injections of DMTU (125 mg/kg body weight, i.p.) twice a day until sacrifice (72 h after the treatment). DMTU did not present any direct effect on mitochondria and substantially inhibited cisplatin-induced mitochondrial damage in liver, therefore preventing elevation of AST and ALT serum levels. DMTU protected against (a) decreased hepatic ATP levels; (b) lipid peroxidation; (c) cardiolipin oxidation; (d) sulfhydryl protein oxidation; (e) mitochondrial membrane rigidification; (f) GSH oxidation; (g) NADPH oxidation; (h) apoptosis. Results suggest that antioxidants, particularly hydroxyl radical scavengers, protect liver mitochondria against cisplatin-induced oxidative damage. Several mitochondrial changes were delineated and proposed as interesting targets for cytoprotective strategy.


Subject(s)
Cisplatin/toxicity , Liver/drug effects , Mitochondria/drug effects , Oxidative Stress/drug effects , Thiourea/analogs & derivatives , Adenosine Triphosphate/metabolism , Animals , Apoptosis/drug effects , Chemical and Drug Induced Liver Injury , Cytoprotection/drug effects , Liver/cytology , Liver/metabolism , Liver Diseases/metabolism , Liver Diseases/pathology , Liver Diseases/physiopathology , Male , Mitochondria/metabolism , Mitochondrial Membranes/drug effects , Oxidation-Reduction/drug effects , Rats , Rats, Wistar , Thiourea/pharmacology
12.
J Pharm Biomed Anal ; 40(2): 389-96, 2006 Feb 13.
Article in English | MEDLINE | ID: mdl-16243469

ABSTRACT

A LC/MS/MS method for the quantitative determination of lorazepam in human plasma and urine samples was developed and validated. The enantioselective assay allowed to separate the enantiomers and to verify the stereochemical instability of lorazepam. The linearity assessed for lorazepam unchanged was 0.2-20 ng of each enantiomer/ml plasma and 0.2-15 ng of each enantiomer/ml urine. The linearity assessed for total lorazepam (after enzymatic hydrolysis) was 1-30 ng of each enantiomer/ml plasma and 10-150 ng of each enantiomer/ml urine. The coefficients of variation obtained for the intra- and interassay precision were less than 15%. The method was applied to the investigation of the kinetic disposition and metabolism of racemic lorazepam administered as a single oral dose of 2 mg to a parturient. The occurrence of racemization required the calculation of the pharmacokinetic parameters as enantiomeric mixtures of lorazepam (t(1/2a) 3.5h; K(a) 0.198 ngh(-1); t(1/2) 11.5h; beta 0.060 h(-1); AUC(0-infinity) 192.1ngh/ml; CLt/f 2.41ml/minkg; Vd/f 173.5l; Fel 0.41%, and Cl(R) 0.0099 ml/minkg) and its metabolite lorazepam-glucuronide (t(1/2f) 1.2h; K(f) 0.578 h(-1); t(1/2) 16.6h; beta 0.042 h(-1); AUC(0-infinity) 207.6 ngh/ml; Fel 51.80%, and Cl(R) 98.32 ml/minkg). However, the determined confidence limits make the method suitable for application to clinical pharmacokinetic studies, even if the quantification of both the enantiomers is required.


Subject(s)
Anti-Anxiety Agents/pharmacokinetics , Chromatography, Liquid/methods , Lorazepam/analogs & derivatives , Acetonitriles , Anti-Anxiety Agents/blood , Anti-Anxiety Agents/urine , Drug Stability , Female , Humans , Lorazepam/blood , Lorazepam/pharmacokinetics , Lorazepam/urine , Parturition/metabolism , Pregnancy , Reproducibility of Results , Spectrometry, Mass, Electrospray Ionization
13.
Article in English | MEDLINE | ID: mdl-14581082

ABSTRACT

We describe here the first method for the enantioselective analysis of the calcium antagonist lercanidipine in human plasma by high performance liquid chromatography (HPLC) employing tandem mass spectrometric (MS) detection. Routine determination of lercanidipine enantiomers in human plasma in the working range of 0.025-50.0 ng ml(-1) plasma for each enantiomer with an accuracy and precision less than 15% was possible. Application of the method to a stereospecific study of the pharmacokinetics showed that plasma levels after an oral dose of rac-lercanidipine administered to a healthy volunteer were found to be higher for the (S)-enantiomer.


Subject(s)
Antihypertensive Agents/blood , Calcium Channel Blockers/blood , Chromatography, High Pressure Liquid/methods , Dihydropyridines/blood , Antihypertensive Agents/pharmacokinetics , Calcium Channel Blockers/pharmacokinetics , Dihydropyridines/pharmacokinetics , Humans , Reproducibility of Results , Sensitivity and Specificity , Stereoisomerism
14.
Eur J Clin Pharmacol ; 58(9): 607-14, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12483453

ABSTRACT

OBJECTIVE: Nisoldipine (N) is a dihydropyridine calcium antagonist marketed as a racemic mixture and used for the treatment of hypertension. In the present study, we investigated the influence of type-2 diabetes mellitus (DM) on the enantioselective pharmacokinetic and dynamic parameters of N. METHODS: Seventeen hypertensive patients, nine of them with DM, were investigated in a cross-over study with administration of rac-N as coat-core tablets (20 mg day(-1)) or placebo for 15 days each. Serial blood samples (0-24 h) were collected on the 15th day, and 24-h ambulatory blood pressure (BP) monitoring was simultaneously evaluated. N enantiomers in plasma samples were analysed using chiral high-performance liquid chromatography combined with gas chromatography/mass spectrometry. The enantiomeric ratios differing from one were evaluated using the Wilcoxon test, and the results are reported as means with the 95% confidence intervals. A lidocaine (L) test was carried out as an in vivo marker of CYP3A4 (and CYP1A2) activities. RESULTS: The following differences were observed between the (+)-N and (-)-N enantiomers, respectively, in the patients presenting with DM (means and ranges): C(max) 3.9 (1.7-6.1) ng ml(-1) versus 0.7 (0.4-1.0) ng ml(-1), AUC(0-24) 51.5 (29.0-74.0) ng ml(-1) h versus 9.4 (5.9-12.8) ng ml(-1) h, and Cl/f 3.6 (1.9-5.4) l h(-1) kg(-1) versus 18.7 (11.7-25.7) l h(-1) kg(-1). The Cl/f value of (+)-N was lower (Mann-Whitney test) in patients with DM: 6.0 (4.3-7.5) l h(-1) kg(-1) versus 3.6 (1.9-5.4) l h(-1) kg(-1). The same observation was made for the (-)-N, with Cl/f reaching 38.8 (26.8-51.0) l h(-1) kg(-1) and 18.7 (11.7-25.7) l h(-1) kg(-1) for the non-diabetic and DM groups, respectively. The L test resulted in higher ratios (P < 0.05) of plasma L/MEGX concentrations (30 min after i.v. L) for DM (11.1 vs 18.6). N significantly reduced systolic and diastolic BP (P < 0.05, Wilcoxon test) in all patients investigated relative to placebo. No differences in BP reduction were observed between diabetic and non-diabetic patients. N significantly increased noradrenaline concentrations in plasma of both patient groups. The data also demonstrated that the plasma concentrations of noradrenaline 30 min after N administration were lower (P < 0.05) in diabetic (mean 2.86 pmol ml(-1)) than in non-diabetic patients (4.80 pmol ml(-1)). CONCLUSIONS: The present data permit us to infer that type-2 diabetes mellitus alters the kinetic disposition of the (+)-N eutomer and (-)-N distomer, presumably due to a lower activity of CYP3A4, although it does not modify the clinical effect brought about by the reduction in BP.


Subject(s)
Antihypertensive Agents/pharmacology , Antihypertensive Agents/pharmacokinetics , Diabetes Mellitus, Type 2/metabolism , Hypertension/metabolism , Nisoldipine/pharmacology , Nisoldipine/pharmacokinetics , Adult , Antihypertensive Agents/therapeutic use , Area Under Curve , Biomarkers , Blood Glucose/analysis , Blood Pressure/drug effects , Cross-Over Studies , Cytochrome P-450 CYP1A2/metabolism , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme System/metabolism , Delayed-Action Preparations , Diabetes Mellitus, Type 2/complications , Female , Heart Rate/drug effects , Humans , Hypertension/complications , Hypertension/drug therapy , Lidocaine , Male , Middle Aged , Nisoldipine/therapeutic use , Norepinephrine/blood , Stereoisomerism
SELECTION OF CITATIONS
SEARCH DETAIL
...