Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Regul Toxicol Pharmacol ; 115: 104709, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32553751

RESUMO

Safety margin, a key aspect of any non-clinical toxicity studies, is calculated by dividing the systemic exposure (AUC) at NOAEL (No Adverse Effect Level) in toxicity studies by the clinical exposure. The validity of using total plasma concentration (Cp) to calculate AUC is often discussed, as it is the unbound plasma concentration (Cup) that elicits the pharmacological and toxicological effects. Data regarding plasma protein binding across species was collected for 114 MSD small molecule compounds which had been discontinued from development either due to non-clinical toxicity or due to clinical Adverse Effects. A >3-fold difference in unbound fraction in plasma (fup) was selected as a meaningful difference in plasma protein binding between non-clinical species and humans. In rats, dogs and non-human primates, approximately 3-5% of the compounds had a >3-fold difference in plasma protein binding than humans. Following assessment of toxicity profile of these compounds, it was concluded that calculation of safety margins after incorporating fup would have still led to the discontinuation of these compounds. Therefore, although fup can still be used for calculation of safety margin on a case by case basis, the routine use of fup for calculation of safety margins is not warranted.


Assuntos
Proteínas Sanguíneas/metabolismo , Animais , Área Sob a Curva , Cães , Avaliação Pré-Clínica de Medicamentos , Haplorrinos , Humanos , Farmacocinética , Ligação Proteica , Ratos , Especificidade da Espécie
2.
Mol Pharm ; 14(5): 1634-1645, 2017 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-28329443

RESUMO

The purpose of this research was to assess variability in pharmacokinetic profiles (PK variability) in preclinical species and identify the risk factors associated with the properties of a drug molecule that contribute to the variability. Exposure data in mouse, rat, dog, and monkey for a total of 16,592 research compounds studied between 1999 and 2013 were included in the analysis. Both in vivo study parameters and in silico/experimental physicochemical properties of the molecules were analyzed. Areas under the plasma concentration vs time curves (AUC) were used to assess PK variability. PK variability was calculated as the ratio of the highest AUC within a defined set of AUC values (AUCmax) over the lowest AUC within that set (AUCmin). Both intra- and inter-animal variability were analyzed, with intra-animal exposures found to be more variable than inter-animal exposures. While several routes of administration were initially studied, the analysis was focused on the oral route, which corresponds to the large majority of data points and displays higher variability than the subcutaneous, intraperitoneal, or intravenous routes. The association between inter-animal PK variability and physical properties was studied, and low solubility, high administered dose, high preclinical dose number (PDo), and pH-dependent solubility were found to be associated with high variability in exposures. Permeability-as assessed by the measured permeability coefficient in the LLC-PK1 cell line-was also considered but appeared to only have a weak association with variability. Consistent with these findings, BCS class I and III compounds were found to be less prone to PK variability than BCS class II and IV compounds. A modest association of PK variability with clearance was observed while the association with bioavailability, a higher PK variability for compounds with lower bioavailability, appeared to be more pronounced. Finally, two case studies that highlight PK variability issues are described, and successful mitigation strategies are presented.


Assuntos
Avaliação Pré-Clínica de Medicamentos/métodos , Animais , Área Sob a Curva , Líquidos Corporais/metabolismo , Cães , Humanos , Concentração de Íons de Hidrogênio , Absorção Intestinal/fisiologia , Células LLC-PK1 , Camundongos , Permeabilidade , Preparações Farmacêuticas/metabolismo , Farmacocinética , Ratos , Suínos
3.
Regul Toxicol Pharmacol ; 86: 181-192, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28279667

RESUMO

Suvorexant (Belsomra®) is a dual orexin receptor antagonist approved for the treatment of insomnia. Because of its pharmacology within the central nervous system, intended therapeutic indication, and first-in-class status, an assessment of suvorexant abuse liability potential was required prior to marketing approval. The nonclinical abuse liability potential studies for suvorexant included: 1) rat drug-dependence model to assess physical dependence following abrupt cessation; 2) rat drug-discrimination model to examine the potential similarity of the interoceptive or subjective effects of suvorexant to those elicited by zolpidem and morphine; 3) self-administration model to assess the relative reinforcing efficacy of suvorexant in rhesus monkeys conditioned to self-administer methohexital. No significant signs of spontaneous drug withdrawal or 'discontinuation syndrome' were observed in rats following abrupt discontinuation of suvorexant. Suvorexant did not elicit complete cross-generalization to either a zolpidem or morphine training/reference stimuli in rats, and suvorexant was devoid of behavioral evidence of positive reinforcing efficacy in monkeys. These nonclinical findings suggested that suvorexant will have low abuse potential in humans. In the final regulatory risk assessment, suvorexant was placed into Schedule IV, likely due to its first-in-class status, its sedative properties, and the outcome of the clinical abuse potential assessment.


Assuntos
Azepinas/farmacologia , Antagonistas dos Receptores de Orexina/farmacologia , Medicamentos Indutores do Sono/farmacologia , Transtornos Relacionados ao Uso de Substâncias/etiologia , Triazóis/farmacologia , Animais , Humanos , Hipnóticos e Sedativos/farmacologia , Macaca mulatta , Ratos , Distúrbios do Início e da Manutenção do Sono/tratamento farmacológico
4.
Indian J Dent Res ; 32(1): 44-50, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34269236

RESUMO

OBJECTIVES: (1) To assess the risk of Obstructive Sleep Apnea (OSA) using a pre-designed questionnaire; (2) To assess the periodontal parameters like pocket probing depth (PPD) and clinical attachment loss (CAL) in the study subjects and (3) To determine the association between the shared risk factors of OSA and periodontitis. BACKGROUND OF THE STUDY: Periodontitis is a microbially associated, host-mediated inflammatory disease that results in the loss of periodontal attachment. It has multifactorial etiology and has been linked to an array of systemic diseases. Though both periodontitis and obstructive sleep apnoea (OSA) share some common risk factors, the insight into the hypothetical speculative link remains vague. This study intended to probe into the association between periodontitis and OSA. METHODOLOGY: A cross-sectional study was conducted on 250 subjects. The subjects were explained the nature of the study and written informed consent was obtained for participation in the study. The patients were administered a STOP-BANG questionnaire following which the periodontal parameters were recorded. The data obtained was analyzed by descriptive and inferential statistics. RESULTS: A statistically significant increase in PPD and CAL scores were seen with the increase in OSA scores. A significant moderate positive correlation was found between OSA scores and PPD [r = 0.58, P < 0.001] and CAL [r = 0.55, P = 0.001]. Males were at increased risk for OSA and periodontitis. Age, hypertension and BMI which are the risk factors for periodontitis were also significantly higher in subjects at high risk for OSA. CONCLUSION: This study found a moderate positive association between periodontitis and OSA.


Assuntos
Periodontite , Apneia Obstrutiva do Sono , Estudos Transversais , Humanos , Masculino , Periodontite/complicações , Periodontite/epidemiologia , Fatores de Risco , Apneia Obstrutiva do Sono/complicações , Apneia Obstrutiva do Sono/epidemiologia , Inquéritos e Questionários
5.
Comp Med ; 71(2): 123-132, 2021 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-33789781

RESUMO

Alpha-1 acid glycoprotein (AGP) is a significant drug binding acute phase protein that is present in rats. AGP levels are known to increase during tissue injury, cancer and infection. Accordingly, when determining effective drug ranges and toxicity limits, consideration of drug binding to AGP is essential. However, AGP levels have not been well established during subclinical infections. The goal of this study was to establish a subclinical infection model in rats using AGP as a biomarker. This information could enhance health surveillance, aid in outlier identification, and provide more informed characterization of drug candidates. An initial study (n = 57) was conducted to evaluate AGP in response to various concentrations of Staphylococcus aureus (S. aureus) in Sprague-Dawley rats with or without implants of catheter material. A model validation study (n = 16) was then conducted using propranolol. Rats received vehicle control or S. aureus and when indicated, received oral propranolol (10 mg/kg). Health assessment and blood collection for measurement of plasma AGP or propranolol were performed over time (days). A dose response study showed that plasma AGP was elevated on day 2 in rats inoculated with S. aureus at 106, 107 or, 108 CFU regardless of implant status. Furthermore, AGP levels remained elevated on day 4 in rats inoculated with 107 or 108 CFUs of S. aureus. In contrast, significant increases in AGP were not detected in rats treated with vehicle or 10³ CFU S. aureus. In the validation study, robust elevations in plasma AGP were detected on days 2 and 4 in S. aureus infected rats with or without propranolol. The AUC levels for propranolol on days 2 and 4 were 493 ± 44 h × ng/mL and 334 ± 54 h × ng/mL, respectively), whereas in noninfected rats that received only propranolol, levels were 38 ± 11 h × ng/mL and 76 ± 16. h × ng/mL, respectively. The high correlation between plasma propranolol and AGP demonstrated a direct impact of AGP on drug pharmacokinetics and pharmacodynamics. The results indicate that AGP is a reliable biomarker in this model of subclinical infection and should be considered for accurate data interpretation.


Assuntos
Orosomucoide , Preparações Farmacêuticas , Animais , Biomarcadores , Orosomucoide/metabolismo , Ligação Proteica , Ratos , Ratos Sprague-Dawley , Staphylococcus aureus
6.
Appl Opt ; 48(7): 1382-8, 2009 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-19252640

RESUMO

The application of terahertz time-domain spectroscopy (THz-TDS) and imaging to differentiate flat and curved surfaces in reflection via the THz Gouy phase shift is demonstrated. For a THz-TDS system that is aligned for reflection from a flat surface, the presence of a curved surface displaces the image focal plane from the detector plane, resulting in a Gouy phase shift. The potential of utilizing this configuration for estimating the number and size of curved objects is discussed with particular emphasis on agriculture crop yield estimates.

7.
Toxicol Mech Methods ; 16(4): 173-80, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-20021043

RESUMO

In previous studies, the free radical generating toxin tertiary butylhydroperoxide (t-BuOOH) was found to induce significant cell death in human cortical neuronal cells (HCN2 cells). Pretreatment with the poly (ADP-ribose) polymerase (PARP) inhibitor nicotinamide was able to prevent HCN2 cell death. In this study it is observed that apoptosis is induced following the addition of t-BuOOH at 6 h as indicated by TUNEL-positive cells. When nicotinamide is added prior to t-BuOOH, it is able to prevent neuronal cell death and inhibit apoptosis. DNA microarray studies demonstrate that t-BuOOH administration causes an upregulation of proapoptotic genes like ICH-2 and BimL. On the other hand, nicotinamide-pretreated neurons have higher expression levels of inhibitors of apoptosis (IAP) genes. Therefore, it appears that one mechanism by which nicotinamide acts as neuroprotective agents is by elevating the gene expression levels of IAPs. Moreover, there is an upregulation of the glyceraldehydes-3-phosphate dehydrogenase gene in nicotinamide-pretreated HCN2 cells. Nicotinamide-pretreated cells also had higher expression levels of putative "death domain" genes like p75TNFR, TRAIL2, TNFR1, and HVEM-L. Thus, nicotinamide can regulate multiple apoptotic genes with seemingly opposite roles and through its action on these various genes prevent apoptosis of neuronal cells.

8.
Protein J ; 35(3): 237-46, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27272220

RESUMO

Combating Type-2 diabetes mellitus is a pivotal challenge in front of the present world. Several lines of therapy are in practice for resisting this deadly disease which often culminates with cardiovascular complexities, neuropathy and retinopathy. Among various therapies, administration of alpha glucosidase inhibitors is common and widely practiced. Sulfonylurea category of anti diabetic drug often suffers from cross reactivity with sulfamethoxazole (SMX), a common drug in use to treat a handful of microbial infections. However the specific cellular target generating postprandial hypoglycemia on SMX administration is till date unraveled. The present work has been initiated to elucidate the effects of a group of sulfonamide drugs inclusive of SMX for their amylase inhibitory role. SMX inhibits porcine pancreatic amylase (PPA) in a noncompetitive mode with an average IC50 value 0.94 mM respectively. Interaction of SMX with PPA is manifested with gradual quenching of tryptophan fluorescence with concomitant shift in lambda max value (λmax). Binding is governed by entropy driven factor (24.8 cal mol(-1) K(-1)) with unfavorable contribution from enthalpy change. SMX interferes with the activity of acarbose in a synergistic mode to reduce the effective dose of acarbose as evident from the in vitro PPA inhibition study. In summary, loss of PPA activity in presence of SMX is indicative of structural changes of PPA which is further augmented in the presence of acarbose as explained in the schematic model and docking study.


Assuntos
Anti-Infecciosos/farmacologia , Inibidores Enzimáticos/farmacologia , alfa-Amilases Pancreáticas/antagonistas & inibidores , Sulfametoxazol/farmacologia , Animais , Anti-Infecciosos/química , Inibidores Enzimáticos/química , Simulação de Acoplamento Molecular , alfa-Amilases Pancreáticas/química , alfa-Amilases Pancreáticas/metabolismo , Sulfametoxazol/química , Suínos
9.
Korean J Ophthalmol ; 29(2): 86-91, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25829824

RESUMO

PURPOSE: The purpose of the study was to differentiate ischemic central retinal vein occlusion (CRVO) from nonischemic CRVO during the early acute phase using plasma homocysteine as a biochemical marker. METHODS: Fasting plasma homocysteine, serum vitamin B12, and folate levels were measured in 108 consecutive unilateral elderly adult (age >50 years) ischemic CRVO patients in the absence of local and systemic disease and compared with a total of 144 age and sex matched nonischemic CRVO patients and 120 age and sex matched healthy control subjects. RESULTS: Homocysteine level was significantly increased in the patients with ischemic CRVO in comparison with nonischemic CRVO patients (p = 0.009) and also in comparison with control subjects (p < 0.001). Analysis also showed that hyperhomocysteinemia was associated with increased incidence of ischemic CRVO (odds ratio, 18) than that for nonischemic CRVO (odds ratio, 4.5). Serum vitamin B12 and folate levels were significantly lower (p < 0.001) in CRVO patients compared to the control but were not significantly different between nonischemic and ischemic CRVO patients (p > 0.1). CONCLUSIONS: Hyperhomocysteinemia can be regarded as useful in differentiating nonischemic and ischemic CRVO during the early acute phase in absence of local and systemic disease in the elderly adult (age >50 years) population.


Assuntos
Biomarcadores/sangue , Hiper-Homocisteinemia/complicações , Oclusão da Veia Retiniana/diagnóstico , Doença Aguda , Idoso , Estudos de Casos e Controles , Diagnóstico Diferencial , Feminino , Seguimentos , Humanos , Hiper-Homocisteinemia/sangue , Hiper-Homocisteinemia/diagnóstico , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos , Oclusão da Veia Retiniana/complicações , Fatores de Tempo
10.
Neurotoxicology ; 25(5): 885-91, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15288519

RESUMO

Genistein, a soy isoflavone, has been shown to mimic the pharmacological actions of the endogenous steroid estrogen with which it has structural similarities. There is now evidence that the genistein can prevent disorders-like heart diseases, cancer and diabetes as well. However, very few studies have looked at the effect of genistein on the central nervous system. Published studies also show conflicting conclusions regarding the effects of genistein in the brain. The current study was conducted in the human cortical cell lines HCN1-A and HCN2 in order to determine the neuroprotective efficacy of genistein. It was observed that pre-treatment with 50 or 10 microM genistein was able to protect HCN1-A and HCN2 cells from the cell death induced by 100 microM or 1 mM tertiary butylhydroperoxide (t-BuOOH; a free radical generating toxin). The morphological disruption caused by t-BuOOH was also prevented by genistein in HCN2 cells. Moreover, genistein was able to prevent the down-regulation of the anti-apoptotic protein bcl-2 that was caused by t-BuOOH treatment. These results indicate that genistein may have neuroprotective effect in cortical cells, which may be mediated by its regulation of the anti-apoptotic protein bcl-2.


Assuntos
Córtex Cerebral/patologia , Genisteína/farmacologia , Neurônios/patologia , Fármacos Neuroprotetores , Estresse Oxidativo/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Morte Celular/efeitos dos fármacos , Linhagem Celular , Córtex Cerebral/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Ensaio de Imunoadsorção Enzimática , Estradiol/farmacologia , Humanos , L-Lactato Desidrogenase/metabolismo , Neurônios/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , terc-Butil Hidroperóxido/toxicidade
11.
Neurotoxicology ; 24(3): 443-8, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12782109

RESUMO

It is well known that the generation of oxygen radicals can cause neuronal death by both apoptosis and necrosis, which may lead to the onset of neurodegenerative diseases. In previous in vivo studies, nicotinamide was found to prevent both DNA fragmentation and apoptosis that were induced by free radical generating toxins like tertiary butylhydroperoxide (t-BuOOH). Nicotinamide is a precursor for NAD and is an inhibitor of the enzyme poly(ADP-ribose) polymerase (PARP). However, the effect of nicotinamide on the regulation of pro- and anti-apoptotic proteins in neurons is not clear. In our study, the human cortical neuronal cell line HCN1-A has been used to determine the mechanism of action of nicotinamide at the cellular level. Cell viability studies showed that t-BuOOH treatment (both 100 microM and 1mM) caused significant cell death at 24 and 48h compared to control cells. Pretreatment with 1mM nicotinamide before t-BuOOH administration caused significant reduction in cell death. Moreover, the morphology of HCN1-A cells that were treated with both nicotinamide and t-BuOOH appeared to be closer to control cells when compared to HCN1-A cells treated with only t-BuOOH. Also, t-BuOOH treatment caused an elevation in the levels of the pro-apoptotic proteins p53 and p21/WAF-1 and a reduction in the levels of the anti-apoptotic protein bcl-2 compared to their levels in control HCN1-A cells, while pretreatment with nicotinamide reduced p53 and p21/WAF-1 levels even in the presence of t-BuOOH. However nicotinamide did not seem to alter bcl-2 levels. These results indicate that nicotinamide treatment can protect human neuronal cells from the toxic effects of t-BuOOH.


Assuntos
Córtex Cerebral/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Niacinamida/farmacologia , terc-Butil Hidroperóxido/farmacologia , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Córtex Cerebral/citologia , Córtex Cerebral/fisiologia , Humanos , Neurônios/citologia , Neurônios/fisiologia
12.
Environ Int ; 30(6): 811-4, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15120200

RESUMO

Environmental homogeneity is being continuously disturbed and affected by artificially introduced loads of chemical toxicants that also include heavy metals. The Tiljala wetlands of the eastern fringe of Calcutta, West Bengal (India) are a virtual sink for the deposition of urban and industrial wastes that get admixed with the aquatic environment. We have selected Lemna minor (duckweed), as a representative of the biota surviving therein for the present study. Concentrations of lead, cadmium, chromium, zinc, copper and mercury in the fronds of Lemna were measured to peep into the range of input of heavy metals in the duckweed subjects. Natural unexposed population of duckweed from a domestic pond in Batanagar area, 24 Parganas, West Bengal (India) was also found to accumulate similar concentrations of these metals when cultured in artificially contaminated water in the laboratory. The exposed individuals also exhibited polymorphism with respect to the loci of esterase, as compared to an unexposed control plants. Therefore, the present study suggests EST variations of L. minor to be a potential biomarker of heavy metal pollution.


Assuntos
Araceae/química , Araceae/enzimologia , Exposição Ambiental , Esterases/análise , Metais Pesados/farmacocinética , Poluentes da Água/farmacocinética , Biomarcadores/análise , Monitoramento Ambiental , Índia , Metais Pesados/análise , Poluentes da Água/análise
13.
Fungal Biol ; 116(8): 902-9, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22862918

RESUMO

Locoweed plants in the southwestern United States often harbour a slow-growing endophytic fungus, Undifilum oxytropis (Phylum: Ascomycota; Order: Pleosporales), which produces a toxic alkaloid, swainsonine. Consumption of U. oxytropis by grazing animals induces a neurological disorder called locoism for which the toxic alkaloid swainsonine has been reported to be the causal agent. Little is known about the biosynthetic pathway of swainsonine in endophytic fungi, but previous studies on non-endophytic ascomycetous fungi indicate that pipecolic acid and saccharopine are key intermediates. We have used degenerate primers, Rapid amplification of cDNA ends (RACE)-PCR and inverse PCR to identify the gene sequence of U. oxytropis saccharopine reductase. To investigate the role of this gene product in swainsonine metabolism, we have developed a gene deletion system for this slow-growing endophyte based on our recently established transformation protocol. A strain of U. oxytropis lacking saccharopine reductase had decreased levels of saccharopine and lysine along with increased accumulation of pipecolic acid and swainsonine. Thus, saccharopine reductase influences the accumulation of swainsonine and its precursor, pipecolic acid, in U. oxytropis.


Assuntos
Ascomicetos/enzimologia , Endófitos/enzimologia , Proteínas Fúngicas/metabolismo , Oxytropis/microbiologia , Sacaropina Desidrogenases/metabolismo , Swainsonina/metabolismo , Ascomicetos/genética , Ascomicetos/fisiologia , Vias Biossintéticas , Endófitos/genética , Endófitos/fisiologia , Proteínas Fúngicas/genética , Lisina/análogos & derivados , Lisina/metabolismo , Dados de Sequência Molecular , Mucoproteínas/metabolismo , Sacaropina Desidrogenases/genética
14.
J Microbiol Methods ; 81(2): 160-5, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20211666

RESUMO

Undifilum oxytropis (Phylum: Ascomycota; Family: Pleosporaceae) is a slow growing endophytic fungus that produces a toxic alkaloid, swainsonine. This endophyte resides in locoweeds, which are perennial flowering legumes. Consumption of this fungus by grazing animals induces a neurological disorder called locoism. The alkaloid swainsonine, an alpha-mannosidase inhibitor, is responsible for the field toxicity related to locoism. Little is known about the biosynthetic pathway of swainsonine in endophytic fungi. Genetic manipulation of endophytic fungi is important to better understand biochemical pathways involved in alkaloid synthesis, but no transformation system has been available for studying such enzymes in Undifilum. In this study we report the development of protoplast and transformation system for U. oxytropis. Fungal mycelia required for generating protoplasts were grown in liquid culture, then harvested and processed with various enzymes. Protoplasts were transformed with a fungal specific vector driving the expression of Enhanced Green Florescent Protein (EGFP). The quality of transformed protoplasts and transformation efficiency were monitored during the process. In all cases, resistance to antibiotic hygromycin B was maintained. Such manipulation will open avenues for future research to decipher fungal metabolic pathways.


Assuntos
Ascomicetos/genética , Técnicas de Transferência de Genes , Genética Microbiana/métodos , Transformação Genética , Ascomicetos/crescimento & desenvolvimento , Ascomicetos/metabolismo , Genes Reporter , Vetores Genéticos , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Micélio/genética , Micélio/crescimento & desenvolvimento , Protoplastos , Coloração e Rotulagem/métodos , Swainsonina/metabolismo
16.
Neurotox Res ; 4(7-8): 595-599, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12709297

RESUMO

In previous studies with mice the oxygen radical generating neurotoxin tertiary butylhydroperoxide (t-BuOOH) was used to mimic the oxidative injury that has been implicated in neurodegenerative diseases. In addition, previous studies have shown that the poly (ADP-ribose) polymerase (PARP) inhibitor nicotinamide is able to prevent DNA fragmentation and apoptosis that is induced by t-BuOOH in mouse brain. However, the molecular mechanism(s) by which nicotinamide is able to protect human brain cells at the cellular level is not clear. Therefore in this study a cell culture model system with human cortical neuronal cells (HCN2 cells) has been employed where the molecular mechanism(s) of nicotinamide action, both in the presence and absence of t-BuOOH has been studied. Human cortical neurons (HCN2 cells) have been shown to differentiate to a neuron-like morphology. In this study, exposure of HCN2 cells to t-BuOOH resulted in altered morphology and disruption of neuronal differentiation leading to cell death. However, in neurons, which were treated with nicotinamide before being exposed to t-BuOOH, neuronal differentiation was preserved; morphological disruption was prevented and cell death was reduced significantly. Moreover, our studies indicate that nicotinamide is able to prevent the up-regulation of the pro-apoptotic proteins p53 and p21/WAF-1, and the down-regulation of the anti-apoptotic protein bcl-2 that is induced by t-BuOOH in HCN2 cells. Thus this study indicates that nicotinamide protects human brain cells from the toxic effects of free radical generating toxins by regulating the levels of various pro- and anti-apoptotic proteins.

17.
Toxicol Appl Pharmacol ; 196(1): 29-36, 2004 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-15050405

RESUMO

Cytoskeleton disruption is a process by which oxidative stress disrupts cellular function. This study compares and contrasts the effect of oxidative stress on the three major cytoskeleton filaments, microfilaments (MFs), microtubule (MT), and vimentin in human cortical neuronal cell line (HCN2). HCN2 cells were treated with 100 microM tertiary butylhydroperoxide (t-BuOOH), a free radical generating neurotoxin for 1, 3, or 6 h. Cell viability studies demonstrated significant cell death although the morphology studies showed that there was a substantial loss in neurites of neurons treated with t-BuOOH for 6 h. Because the cytoskeleton plays a role in neurite outgrowth, the effect of oxidative stress on the cytoskeletal was studied. In neurons subjected to oxidative stress for 30 min or 1 h, there were no major changes in microfilament distribution though there was altered distribution of microtubule and vimentin filaments as compared to controls. However, loss and disruption of all the three cytoskeletal filaments was observed at later times (3 and 6 h), which was confirmed by Western Blot analysis. Further studies were done to measure the gene expression levels of actin, tubulin, and vimentin. Results indicated that the overall loss of the cytoskeletal proteins in neurons treated with free radical generating toxin might not be a direct result of the downregulation of the cytoskeletal genes. This study shows that free radical generation in human neurons leads to the disruption of the cytoskeleton, though there may be a difference in the susceptibility to oxidative stress among the individual components of the cytoskeletal filaments.


Assuntos
Córtex Cerebral/citologia , Proteínas do Citoesqueleto/metabolismo , Citoesqueleto/ultraestrutura , Neurônios/ultraestrutura , Estresse Oxidativo , Citoesqueleto de Actina/efeitos dos fármacos , Citoesqueleto de Actina/ultraestrutura , Actinas/genética , Actinas/metabolismo , Western Blotting , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Proteínas do Citoesqueleto/genética , Citoesqueleto/efeitos dos fármacos , Citoesqueleto/metabolismo , Imunofluorescência , Expressão Gênica/efeitos dos fármacos , Humanos , Microscopia de Fluorescência , Microtúbulos/efeitos dos fármacos , Microtúbulos/ultraestrutura , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Tubulina (Proteína)/genética , Tubulina (Proteína)/metabolismo , Vimentina/genética , Vimentina/metabolismo , terc-Butil Hidroperóxido/toxicidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA