Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 843
Filtrar
1.
Anal Chim Acta ; 1288: 342153, 2024 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-38220287

RESUMO

Transition metal carbonyl compound of CO releasing molecules (CORMs) are widely used to treat arthritis, tumor and immune. They play a physiological role by directly acting on target tissues to release CO for disease treatment without matrix metabolism after dissolution. It is important to track the level and diffusion process of CORMs in vivo to control CO dose and distribution, facilitating to understand the roles of CORMs in disease treatment. Herein, we designed two red ring Ir1/2 complexes with a large stokes shift. Both Ir1 and Ir2 complexes probes can sensitively and selectively respond to CORM-2. The probe Ir1 exhibits rapid reaction with CORM-2 in Phosphate Buffered Saline within 1 min, showing a detection limitation of 0.13 µM and manifesting a linear relationship with the CORM-2 concentration from 0 to 70 µM at λem = 618 nm. Due to low toxicity even after 12 h exposure and fluorescence stability, this probe has been successfully used for continuous tracking the diffusion process of CORM-2 in living cells for up to 60 min and visualizing CORM-2 distribution in zebrafish. Additionally, this probe showed a good capacity for deep penetration (126 µm), suggesting the potential in detecting CORM-2 in living tissues.


Assuntos
Neoplasias , Compostos Organometálicos , Animais , Peixe-Zebra , Irídio , Compostos Organometálicos/toxicidade
2.
Sci Rep ; 13(1): 23060, 2023 12 27.
Artigo em Inglês | MEDLINE | ID: mdl-38155222

RESUMO

Previous studies on copper pyrithione (CPT) and zinc pyrithione (ZPT) as antifouling agents have mainly focused on marine organisms. Even though CPT and ZPT pose a risk of human exposure, their neurotoxic effects remain to be elucidated. Therefore, in this study, the cytotoxicity and neurotoxicity of CPT and ZPT were evaluated after the exposure of human SH-SY5Y/astrocytic co-cultured cells to them. The results showed that, in a co-culture model, CPT and ZPT induced cytotoxicity in a dose-dependent manner (~ 400 nM). Exposure to CPT and ZPT suppressed all parameters in the neurite outgrowth assays, including neurite length. In particular, exposure led to neurotoxicity at concentrations with low or no cytotoxicity (~ 200 nM). It also downregulated the expression of genes involved in neurodevelopment and maturation and upregulated astrocyte markers. Moreover, CPT and ZPT induced mitochondrial dysfunction and promoted the generation of reactive oxygen species. Notably, N-acetylcysteine treatment showed neuroprotective effects against CPT- and ZPT-mediated toxicity. We concluded that oxidative stress was the major mechanism underlying CPT- and ZPT-induced toxicity in the co-cultured cells.


Assuntos
Neuroblastoma , Compostos Organometálicos , Humanos , Astrócitos/metabolismo , Técnicas de Cocultura , Estresse Oxidativo , Compostos Organometálicos/toxicidade , Compostos Organometálicos/metabolismo , Células Cultivadas
3.
Clin Nucl Med ; 47(7): 599-605, 2022 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-35426841

RESUMO

PURPOSE: Peptide receptor radionuclide therapy (PRRT) can cause dose-limiting toxicities (DLTs) of the bone marrow, liver, and kidneys. It is yet unknown whether women and men are equally at risk of these DLTs. METHODS: Neuroendocrine tumor patients treated with 177Lu-DOTATATE between 2000 and 2015 in our phase II trial with available laboratory data were included. For all DLTs, the highest Common Terminology Criteria for Adverse Events (version 4.03) grades that occurred from the start of PRRT until 3 months after the last cycle were scored. RESULTS: At baseline, women (n = 439) had a significantly lower body mass index, Karnofsky Performance Score, hemoglobin level, and creatinine clearance and a significantly higher platelet level than men (n = 534). Both groups received a median activity of 29.6 GBq (800 mCi). After the start of PRRT, women more frequently developed grade ≥2 thrombocytopenia compared with men (25% vs 18%, P = 0.004) due to a significant increase in grade ≥3 thrombocytopenia (11% vs 6%, P = 0.008). Furthermore, the incidence of grade ≥3 anemia was higher in women (7% vs 3%, P = 0.002). In the multivariable regression model, female sex (odds ratio, 2.50; 95% confidence interval, 1.67-3.74) was confirmed to be an independent risk factor for grade ≥2 thrombocytopenia, among baseline platelet count, bone metastases, uptake on 111In-DTPA-octreotide scan, Karnofsky Performance Score, alkaline phosphatase, lymphocytes, albumin, and renal function. CONCLUSIONS: Female neuroendocrine tumor patients more often experienced PRRT-induced toxicities of platelets and hemoglobin than males, but this did not lead to a lower cumulative activity.


Assuntos
Medula Óssea , Tumores Neuroendócrinos , Radioisótopos , Medula Óssea/patologia , Feminino , Humanos , Masculino , Tumores Neuroendócrinos/patologia , Tumores Neuroendócrinos/radioterapia , Octreotida/toxicidade , Compostos Organometálicos/toxicidade , Tomografia por Emissão de Pósitrons , Radioisótopos/toxicidade , Cintilografia , Receptores de Peptídeos , Distribuição por Sexo , Trombocitopenia
4.
Biol Trace Elem Res ; 200(5): 2205-2222, 2022 May.
Artigo em Inglês | MEDLINE | ID: mdl-34309801

RESUMO

The current study investigates the potential alleviating activity of bulbs (B) and leaves (L) of Allium triquetrum aqueous extract (ATE) on repro-toxicity induced by lead acetate (Pb) in male Wistar rats administrated orally for 3 consecutive weeks. Eighteen groups of rats were divided into the control, Pb (500 mg/kg body weight/day), positive controls of B and L (2 g, 3 g, 4 g, 6 g/kg body weight/day), in addition to four mixtures of each of Pb-B (Pb-B1, Pb-B2, Pb-B3, Pb-B4) and Pb-L (Pb-L1, Pb-L2, Pb-L3, Pb-L4). The two extracts were subjected to phytochemical screening and HPLC analysis. Sperm characteristics were evaluated by CASA system, as well as the serum testosterone, testicular and epididymal levels of glutathione (GSH), glutathione peroxidase (GPx), and malondialdehyde (MDA). The phytochemical screening proved that bulbs' and leaves' extracts were rich in various compounds and the HPLC showed that leaves contain more tannins. Results revealed a significant decrease in the testicular and in the epididymal weights, sperm concentration, motility, testosterone, velocity, vitality, round cells, GSH, and GPx levels in the Pb-intoxicated rats compared to the control, with the exception of MDA concentration that was significantly increased. However, the co-administration of garlic extracts (Pb-B and Pb-L) exhibited a significant increase in all mentioned markers, except for the MDA level which was reduced. Likewise, Pb caused histological injuries in the testicular seminiferous of rats, while the co-administration of wild garlic has reduced such effect, especially in the higher doses. Both extracts of Pb-B and Pb-L have attenuated Pb toxicity in a dose-dependent manner. In conclusion, aqueous extracts of A. triquetrum have the potential to reduce Pb testicular injuries by boosting sperm characteristics and ameliorating oxidative stress markers.


Assuntos
Alho , Chumbo , Compostos Organometálicos , Extratos Vegetais , Testículo , Animais , Antioxidantes/metabolismo , Peso Corporal , Alho/química , Glutationa/metabolismo , Glutationa Peroxidase/metabolismo , Chumbo/toxicidade , Masculino , Compostos Organometálicos/toxicidade , Estresse Oxidativo , Extratos Vegetais/farmacologia , Ratos , Ratos Wistar , Testículo/efeitos dos fármacos , Testosterona
5.
J Food Biochem ; 46(4): e13881, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-34346512

RESUMO

The purpose of this study was to investigate the potential side-effects of lead acetate (LA), which is toxic to the nerves, blood and muscles, in the rat brain. The neuroprotective effects of pomegranate juice (PJ) against LA exposure were also observed. The experiment involved 28 male Wistar albino rats aged 12 weeks. These were divided into four groups: Control, PJ, LA and LA+PJ. Stereological techniques were employed to determine hippocampal volume in each rat brain. Biochemical investigations and histopathological examinations were also performed. Analysis demonstrated a significant decrease in hippocampal volume in the LA group compared to the control group (p < .05). The stereology results also indicated that PJ has protective effects when compared with the LA and LA+PJ groups. A significant increase was also determined in malondialdehyde (MDA) levels and glutathione S-transferase (GST) activity in the LA group compared to the control group, in contrast to glutathione (GSH) levels and carboxylesterase (CaE) and acetylcholinesterase (AchE) activities. MDA and GST activity decreased significantly in the LA+PJ group compared to the LA group in contrast to GSH levels and CaE and AchE activities. Histopathological examination revealed a number of degenerative changes in the LA group. Exposure to LA adversely affects the hippocampus on the male rat brain. It might also be suggested that PJ may ameliorate these deleterious effects.


Assuntos
Sucos de Frutas e Vegetais , Chumbo , Síndromes Neurotóxicas , Compostos Organometálicos , Punica granatum , Acetilcolinesterase , Animais , Antioxidantes/farmacologia , Feminino , Glutationa , Chumbo/toxicidade , Masculino , Compostos Organometálicos/toxicidade , Ratos , Ratos Wistar
6.
Dev Neurosci ; 44(2): 59-66, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34942627

RESUMO

Lead acetate (lead ac.) is a widespread ecological toxicant that can cause marked neurotoxicity and decline in brain functions. This study aimed to evaluate the possible neuroprotective role of L-ascorbic acid (ASCR) and curcumin (CRCM) alone or together against lead ac.-induced neurotoxicity. Rats were injected with lead ac. then treated orally with ASCR and CRCM alone or in combination for 7 days. Lead ac. caused elevation in brain tumor necrosis factor-α, interleukin-6, caspase-3, and malondialdehyde levels, while superoxide dismutase, reduced glutathione as well as the expression of brain-derived neurotrophic factor, cAMP response element-binding, and Beclin1 were downregulated. Expressions of C/EBP homologous protein and mammalian Target of rapamycin kinase were upregulated in brain tissues matched with the control group. Histopathological examination supported the previously mentioned parameters, the administration of the antioxidants in question modulated all the altered previous parameters. The combination regimen achieved the superlative results in the antagonizing lead ac.-induced neurotoxicity via its antioxidant and antiapoptotic activities.


Assuntos
Curcumina , Compostos Organometálicos , Animais , Ácido Ascórbico/metabolismo , Ácido Ascórbico/farmacologia , Encéfalo , Curcumina/metabolismo , Curcumina/farmacologia , Mamíferos , Compostos Organometálicos/metabolismo , Compostos Organometálicos/toxicidade , Estresse Oxidativo , Ratos
7.
Chemistry ; 28(12): e202104049, 2022 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-34967066

RESUMO

The number of donor atoms available on peptides that can competitively coordinate to metal centers renders the site-selective generation of advanced metal-peptide conjugates in high purity a challenging venture. Herein, we present a transmetalation-based synthetic approach on solid support in which an imidazolium pro-ligand can be used to selectively anchor a range of transition metal half-sandwich complexes onto peptides in the presence of multiple coordinative motifs. Amenable to solid support, a range of N-terminus and/or lysine conjugated metal-peptide conjugates were obtained in high purity after cleavage from the resin. The metalated peptides were evaluated for their anticancer properties against human cancer cell lines. While no cytotoxic activity was observed, this platform has the potential to i) provide a pathway to site-selective peptide labelling, ii) be explored as a biorthogonal handle and/or iii) generate a new strategy for ligand design in transition metal catalysts.


Assuntos
Complexos de Coordenação , Compostos Organometálicos , Peptídeos , Complexos de Coordenação/toxicidade , Humanos , Ligantes , Compostos Organometálicos/toxicidade , Peptídeos/química , Elementos de Transição
8.
Arch Toxicol ; 95(8): 2643-2657, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34165617

RESUMO

N,N'-bis(2-mercaptoethyl)isophthalamide (NBMI) is a novel lipophilic heavy metal chelator and thiol redox antioxidant. This study was designed to investigate the neuroprotective activity of NBMI in U-87 MG cells exposed to lead acetate (PbAc). Cells were pretreated with NBMI for 24 h prior to a 48 h exposure to PbAc. Cell death (55%, p < 0.0001) and reduction of intracellular GSH levels (0.70-fold, p < 0.005) induced by 250 µM Pb were successfully attenuated by NBMI pretreatment at concentrations as low as 10 µM. A similar pretreatment with the FDA-approved Pb chelator dimercaptosuccinic acid (DMSA) proved ineffective, indicating a superior PKPD profile for NBMI. Pretreatment with NBMI successfully counteracted Pb-induced neuroinflammation by reducing IL-1ß (0.59-fold, p < 0.05) and GFAP expression levels. NBMI alone was also found to significantly increase ferroportin expression (1.97-fold, p < 0.05) thereby enhancing cellular ability to efflux heavy metals. While no response was observed on the apoptotic pathway, this study demonstrated for the first time that necrotic cell death induced by Pb in U-87 MG cells is successfully attenuated by NBMI. Collectively these data demonstrate NBMI to be a promising neuroprotective compound in the realm of Pb poisoning.


Assuntos
Derivados de Benzeno/farmacologia , Quelantes/farmacologia , Fármacos Neuroprotetores/farmacologia , Compostos Organometálicos/toxicidade , Compostos de Sulfidrila/farmacologia , Proteínas de Transporte de Cátions/metabolismo , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Glioblastoma/patologia , Humanos , Doenças Neuroinflamatórias/etiologia , Doenças Neuroinflamatórias/prevenção & controle , Succímero/farmacologia
9.
ChemistryOpen ; 10(4): 486-492, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33908707

RESUMO

The Cu(II)-diacetyl-bis (N4-methylthiosemicarbazone) complex (ATSM-Cu(II)) has been suggested as a promising positron emission tomography (PET) agent for hypoxia imaging. Several in-vivo studies have shown its potential to detect hypoxic tumors. However, its uptake mechanism and its specificity to various cancer cell lines have been less studied. Herein, we tested ATSM-Cu(II) toxicity, uptake, and reduction, using four different cell types: (1) mouse breast cancer cells (DA-3), (2) human embryonic kidney cells (HEK-293), (3) breast cancer cells (MCF-7), and (4) cervical cancer cells (Hela) under normoxic and hypoxic conditions. We showed that ATSM-Cu(II) is toxic to breast cancer cells under normoxic and hypoxic conditions; however, it is not toxic to normal HEK-293 non-cancer cells. We showed that the Cu(I) content in breast cancer cell after treatment with ATSM-Cu(II) under hypoxic conditions is higher than in normal cells, despite that the uptake of ATSM-Cu(II) is a bit higher in normal cells than in breast cancer cells. This study suggests that the redox potential of ATSM-Cu(II) is higher in breast cancer cells than in normal cells; thus, its toxicity to cancer cells is increased.


Assuntos
Hipóxia/metabolismo , Compostos Organometálicos/metabolismo , Tiossemicarbazonas/metabolismo , Animais , Linhagem Celular Tumoral , Complexos de Coordenação , Radioisótopos de Cobre/química , Radioisótopos de Cobre/metabolismo , Transportador de Cobre 1/metabolismo , Células HEK293 , Humanos , Camundongos , Compostos Organometálicos/química , Compostos Organometálicos/toxicidade , Oxirredução , Tiossemicarbazonas/química , Tiossemicarbazonas/toxicidade
10.
Mol Biol Rep ; 48(4): 3107-3115, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33856607

RESUMO

Metallothionein-3 (MT3) is an antioxidant protein that alters after exposure to heavy metals. In this study, we investigated the hepatic and renal expression of MT3 gene following exposure to lead acetate (PbAc) alone and PbAc plus CoQ10 as an adjuvant antioxidant. Twenty-four rats were allocated into three groups, including control, PbAc (free access to drinking water contaminated with PbAc at 1 g/100 ml), and PbAc plus CoQ10 (10 mg/kg/day Oral). After 28 consecutive days of treatment, the mRNA expression of MT3 and Cyt-c genes and MT3 protein levels were assessed using real-time PCR and immunosorbent assay. The serum lipid profile was also monitored in the three groups. PbAc exposure significantly reduced the hepatic and renal MT3 mRNA and protein expression compared to the control group. This reduction was significantly increased with addition of CoQ10 to levels near those of the control group. The hepatic and renal expression of Cyt-c mRNA increased after treatment with PbAc, while such effect was reversed after addition of CoQ10. Alteration in lipid profile including increased cholesterol and low-density lipoprotein levels were observed after PbAc exposure which were counteracted by CoQ10. Our results confirm the cytotoxic effects of acute lead exposure manifested as changes in the serum lipid profile and cellular levels of Cyt-c mRNA. These cytotoxic effects may have been caused by decreased MT3 gene expression and be reduced by the protective role of CoQ10.


Assuntos
Rim/patologia , Fígado/patologia , Metalotioneína 3/metabolismo , Compostos Organometálicos/toxicidade , Ubiquinona/análogos & derivados , Animais , Antioxidantes/metabolismo , Citocromos c/efeitos dos fármacos , Citocromos c/metabolismo , Rim/efeitos dos fármacos , Rim/metabolismo , Lipídeos/sangue , Fígado/efeitos dos fármacos , Fígado/metabolismo , Metalotioneína 3/efeitos dos fármacos , Metais Pesados/toxicidade , Ratos , Ubiquinona/farmacologia
11.
Eur J Nucl Med Mol Imaging ; 48(11): 3408-3421, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-33772332

RESUMO

PURPOSE: There is significant interest in the development of targeted alpha-particle therapies (TATs) for treatment of solid tumors. The metal chelator-peptide conjugate, DOTA-TATE, loaded with the ß-particle emitting radionuclide 177Lu ([177Lu]Lu-DOTA-TATE) is now standard care for neuroendocrine tumors that express the somatostatin receptor 2 (SSTR2) target. A recent clinical study demonstrated efficacy of the corresponding [225Ac]Ac-DOTA-TATE in patients that were refractory to [177Lu]Lu-DOTA-TATE. Herein, we report the radiosynthesis, toxicity, biodistribution (BD), radiation dosimetry (RD), and efficacy of [225Ac]Ac-DOTA-TATE in small animal models of lung neuroendocrine neoplasms (NENs). METHODS: [225Ac]Ac-DOTA-TATE was synthesized and characterized for radiochemical yield, purity and stability. Non-tumor-bearing BALB/c mice were tested for toxicity and BD. Efficacy was determined by single intravenous injection of [225Ac]Ac-DOTA-TATE into SCID mice-bearing human SSTR2 positive H727 and H69 lung NENs. RD was calculated using the BD data. RESULTS: [225Ac]Ac-DOTA-TATE was synthesized with 98% yield, 99.8% purity, and displayed 97% stability after 2 days incubation in human serum at 37 °C. All animals in the toxicity study appeared healthy 5 months post injection with no indications of toxicity, except that animals that received ≥111 kBq of [225Ac]Ac-DOTA-TATE had chronic progressive nephropathy. BD studies revealed that the primary route of elimination is by the renal route. RD calculations determined pharmacokinetics parameters and absorbed α-emission dosages from 225Ac and its daughters. For both tumor models, a significant tumor growth delay and time to experimental endpoint were observed following a single administration of [225Ac]Ac-DOTA-TATE relative to controls. CONCLUSIONS: These results suggest significant potential for the clinical translation of [225Ac]Ac-DOTA-TATE for lung NENs.


Assuntos
Neoplasias Pulmonares , Compostos Organometálicos , Animais , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Camundongos , Camundongos Endogâmicos BALB C , Camundongos SCID , Octreotida/uso terapêutico , Octreotida/toxicidade , Compostos Organometálicos/uso terapêutico , Compostos Organometálicos/toxicidade , Compostos Radiofarmacêuticos/uso terapêutico , Compostos Radiofarmacêuticos/toxicidade , Distribuição Tecidual
12.
Pharm Biol ; 59(1): 232-241, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33632062

RESUMO

CONTEXT: Andrographolide (Andro) has a neuroprotective effect and a potential for treating Alzheimer's disease (AD), but the mechanism has not been elucidated. OBJECTIVE: The efficacy of Andro on p62-mediated Kelch-like ECH-associated protein 1(Keap1)-Nuclear factor E2 related factor 2 (Nrf2) pathways in the aluminium maltolate (Al(mal)3)-induced neurotoxicity in PC12 cell was explored. MATERIALS AND METHODS: PC12 cells were induced by Al(mal)3 (700 µM) to establish a neurotoxicity model. Following Andro (1.25, 2.5, 5, 10, 20, 40 µM) co-treatment with Al(Mal)3, cell viability was detected with MTT, protein expression levels of ß-amyloid precursor protein (APP), ß-site APP cleaving enzyme 1 (BACE1), Tau, Nrf2, Keap1, p62 and LC3 were measured via western blotting or immunofluorescence analyses. Nrf2, Keap1, p62 and LC3 mRNA, were detected by reverse transcription-quantitative PCR. RESULTS: Compared with the 700 µM Al(mal)3 group, Andro (5, 10 µM) significantly increased Al(mal)3-induced cell viability from 67.4% to 91.9% and 91.2%, respectively, and decreased the expression of APP, BACE1 and Keap1 proteins and the ratio of P-Tau to Tau (from 2.75- fold to 1.94- and 1.70-fold, 2.12-fold to 1.77- and 1.56-fold, 0.68-fold to 0.51- and 0.55-fold, 1.45-fold to 0.82- and 0.91-fold, respectively), increased the protein expression of Nrf2, p62 and the ratio of LC3-II/LC3-I (from 0.67-fold to 0.93- and 0.94-fold, 0.64-fold to 0.88- and 0.87-fold, 0.51-fold to 0.63- and 0.79-fold, respectively), as well as the mRNA expression of Nrf2, p62 and LC3 (from 0.48-fold to 0.92-fold, 0.49-fold to 0.92-fold, 0.25-fold to 0.38-fold). Furthermore, Nrf2 and p62 nuclear translocation were increased and keap1 in the cytoplasm was decreased in the presence of Andro. Silencing p62 or Nrf2 can significantly reduce the protein and mRNA expression of Nrf2 and p62 under co-treatment with Andro and Al(mal)3. DISCUSSION AND CONCLUSIONS: Our results suggested that Andro could be a promising therapeutic lead against Al-induced neurotoxicity by regulating p62-mediated keap1-Nrf2 pathways.


Assuntos
Diterpenos/farmacologia , Fármacos Neuroprotetores/farmacologia , Compostos Organometálicos/toxicidade , Pironas/toxicidade , Animais , Diterpenos/administração & dosagem , Relação Dose-Resposta a Droga , Proteína 1 Associada a ECH Semelhante a Kelch/metabolismo , Fator 2 Relacionado a NF-E2/genética , Fator 2 Relacionado a NF-E2/metabolismo , Fármacos Neuroprotetores/administração & dosagem , Células PC12 , RNA Mensageiro/metabolismo , Ratos , Proteína Sequestossoma-1/genética , Proteína Sequestossoma-1/metabolismo
13.
Neuroendocrinology ; 111(4): 344-353, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-32259830

RESUMO

INTRODUCTION: The pituitary gland has a high expression of somatostatin receptors and is therefore a potential organ at risk for radiation-induced toxicity after 177Lu-DOTATATE treatment. OBJECTIVE: To study changes in pituitary function in patients with neuroendocrine tumors (NETs) treated with dosimetry-based 177Lu-DOTATATE to detect possible late toxicity. METHODS: 68 patients from a phase II clinical trial of dosimetry-based, individualized 177Lu-DOTATATE therapy were included in this analysis. Patients had received a median of 5 (range 3-9) treatment cycles of 7.4 GBq/cycle. Median follow-up was 30 months (range 11-89). The GH/IGF-1 axis, gonadotropins, and adrenal and thyroid axes were analyzed at baseline and on a yearly basis thereafter. Percent changes in hormonal levels over time were analyzed statistically using a linear mixed model and described graphically using box plots. The absorbed radiation dose to the pituitary was estimated based on post-therapeutic imaging, and the results analyzed versus percent change in IGF-1 levels over time. RESULTS: A statistically significant decrease in IGF-1 levels was found (p < 0.005), which correlated with the number of treatment cycles (p = 0.008) and the absorbed radiation dose (p = 0.03). A similar decrease, although non-significant, was seen in gonadotropins in postmenopausal women, while in men there was an increase during the first years after therapy, after which the levels returned to baseline. No change was observed in the adrenal or thyroid axes. CONCLUSIONS: No signs of severe endocrine disorders were detected, although a significant decrease in the GH/IGF-1 axis was found, where dosimetric analyses indicated radiation-induced damage to the pituitary gland as a probable cause.


Assuntos
Gonadotropinas/efeitos da radiação , Fator de Crescimento Insulin-Like I/efeitos da radiação , Tumores Neuroendócrinos/radioterapia , Octreotida/análogos & derivados , Compostos Organometálicos/administração & dosagem , Compostos Organometálicos/toxicidade , Hipófise/efeitos da radiação , Compostos Radiofarmacêuticos/administração & dosagem , Compostos Radiofarmacêuticos/toxicidade , Adulto , Idoso , Feminino , Seguimentos , Gonadotropinas/metabolismo , Humanos , Fator de Crescimento Insulin-Like I/metabolismo , Masculino , Pessoa de Meia-Idade , Tumores Neuroendócrinos/sangue , Octreotida/administração & dosagem , Octreotida/toxicidade , Avaliação de Resultados em Cuidados de Saúde , Hipófise/metabolismo , Pós-Menopausa/metabolismo , Fatores Sexuais
14.
Biochim Biophys Acta Mol Cell Res ; 1868(1): 118853, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32941941

RESUMO

Recently, oxidative stress is a common denominator in the pathogenesis of metal-induced neurotoxicity. Thus, antioxidant therapy is considered as a promising strategy for treating lead-related cognitive impairment. Here, we tested the hypothesis that astragaloside IV (AS-IV) ameliorates lead-associated cognitive deficits through Nrf2-dependent antioxidant mechanisms. Male Nrf2-KO and WT mice received drinking water with 2000 ppm lead and/or AS-IV by gavage for 8 weeks starting at 4 weeks of age. Morris water maze test and biochemical assays were employed to study cognition-enhancing and antioxidant effects of AS-IV. The signaling pathways involved were analyzed using RT-PCR and western blot technology. Significantly, AS-IV attenuated Morris water maze-based cognitive impairment in lead-intoxicated mice. Importantly, cognition-enhancing effect of AS-IV was lost in Nrf2-KO mice. In parallel, AS-IV suppressed lead acetate (PbAc)-induced oxidative stress, as measured by MDA. Mechanistically, AS-IV can up-regulate the expressions of the GCLc and HO-1 at the level of transcription and translation, but not SOD, TrxR activity, GCLm, Trx1, and NQO1 expression. Interestingly, AS-IV induced accumulation of Nrf2 in the nucleus, whereas Nrf2 mRNA levels were unchanged. Furthermore, AS-IV treatment resulted in elevated levels of phosphorylated Akt (active form) and phosphorylated GSK-3ß (inactive forms) but decreased level of phosphorylated Fyn. Collectively, our findings indicate that AS-IV may target Nrf2 to attenuate lead-triggered oxidative stress and subsequent cognitive impairments, suggesting that AS-IV is a potential candidate for the treatment of lead-associated cognitive diseases.


Assuntos
Disfunção Cognitiva/tratamento farmacológico , Heme Oxigenase-1/genética , Proteínas de Membrana/genética , Fator 2 Relacionado a NF-E2/genética , Compostos Organometálicos/toxicidade , Animais , Antioxidantes/farmacologia , Disfunção Cognitiva/induzido quimicamente , Disfunção Cognitiva/genética , Glicogênio Sintase Quinase 3 beta/genética , Camundongos , Estresse Oxidativo/efeitos dos fármacos , Células PC12 , Fosforilação/efeitos dos fármacos , Ratos , Espécies Reativas de Oxigênio/química , Saponinas/farmacologia , Transdução de Sinais/efeitos dos fármacos , Triterpenos/farmacologia
15.
Toxicol Mech Methods ; 31(1): 33-42, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-32900247

RESUMO

Aluminum (Al), a neurotoxic element, can induce Alzheimer's disease (AD) via triggering neuronal death. Ferroptosis is a new type of programmed cell death related to neurological diseases. Unfortunately, its role in aluminum-induced neuronal death remains completely unclear. This study aimed to investigate whether ferroptosis is involved in neuronal death in response to aluminum exposure as well as its underlying mechanism. In this study, rat adrenal pheochromocytoma (PC12) cells were treated with 200 µM aluminum maltolate (Al(mal)3) for 24 h, and related biochemical indicators were assessed to determine whether ferroptosis was induced by aluminum in neurons. Then, the potential mechanism was explored by detecting of these genes and proteins associated with ferroptosis after adding ferroptosis-specific agonist Erastin (5 µM) and antagonist Ferrostatin-1 (Fer-1) (5 µM). The experimental results demonstrated that aluminum exposure significantly increased the death of PC12 cells and caused specific mitochondrial pathological changes of ferroptosis in PC12 cells. Further research confirmed that ferroptosis was triggered by aluminum in PC12 cells by means of activating the oxidative damage signaling pathway, which was displayed as inhibition of the cysteine/glutamate antiporter system (system Xc-), causing the depletion of cellular glutathione (GSH) and inactivation of glutathione peroxidase (GSH-PX) eventually lead to accumulation of reactive oxygen species (ROS). Taken together, ferroptosis was a means of neuronal death induced by aluminum and oxidative damage may be its underlying mechanism, which also provided some new clues to potential target for the intervention and therapy of AD.


Assuntos
Ferroptose/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Compostos Organometálicos/toxicidade , Estresse Oxidativo/efeitos dos fármacos , Pironas/toxicidade , Animais , Mitocôndrias/metabolismo , Mitocôndrias/ultraestrutura , Neurônios/metabolismo , Neurônios/ultraestrutura , Células PC12 , Ratos , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais
16.
Neurotoxicology ; 81: 395-405, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33080273

RESUMO

Lead (Pb) is an environmental contaminant that primarily affects the central nervous system, particularly the developing brain. Recently, increasing evidence indicates the important roles of histone deacetylases (HDACs) in Pb-induced neurotoxicity. However, the precise molecular mechanisms involving HDAC4 remains unknown. The purpose of this study was to investigate the role of HDAC4 in Pb-induced neurotoxicity both in vivo and in vitro. In vitro study, PC12 cells were exposed to Pb (10 µM) for 24 h, then the mRNA and protein levels of HDAC4 were analyzed. In vivo study, pregnant rats and their female offspring were treated with lead (50 ppm) until postnatal day 30. Then the pups were sacrificed and the mRNA and protein levels of HDAC4 in the hippocampus were analyzed. The results showed that HDAC4 was significantly increased in both PC12 cells and rat hippocampus upon Pb exposure. Blockade of HDAC4 with either LMK-235 (an inhibitor of HDAC4) or shHDAC4 (HDAC4-knocking down plasmid) ameliorated the Pb-induced neurite outgrowth deficits. Interestingly, HDAC4 was aberrantly accumulated in the nucleus upon Pb exposure. By contrast, blocking the HDAC4 shuffling from the cytosol to the nucleus with ΔNLS2-HDAC4 (the cytosol-localized HDAC4 mutant) was able to rescue the neuronal impairment. In addition, Pb increased PP1 (protein phosphatase 1) expression which in turn influenced the subcellular localization of HDAC4 by dephosphorylation of specific serine/threonine residues. What's more, blockade of PP1 with PP1-knocking down construct (shPP1) ameliorated Pb-induced neurite outgrowth deficits. Taken together, nuclear accumulation of HDAC4 by PP1-mediated dephosphorylation involved in Pb-induced neurotoxicity. This study might provide a promising molecular target for medical intervention with environmental cues.


Assuntos
Hipocampo/efeitos dos fármacos , Histona Desacetilases/metabolismo , Intoxicação do Sistema Nervoso por Chumbo/enzimologia , Crescimento Neuronal/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Compostos Organometálicos/toxicidade , Proteína Fosfatase 1/metabolismo , Animais , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/enzimologia , Núcleo Celular/patologia , Feminino , Hipocampo/enzimologia , Hipocampo/patologia , Histona Desacetilases/genética , Intoxicação do Sistema Nervoso por Chumbo/genética , Intoxicação do Sistema Nervoso por Chumbo/patologia , Neurônios/enzimologia , Neurônios/patologia , Células PC12 , Fosforilação , Proteína Fosfatase 1/genética , Ratos , Ratos Sprague-Dawley
17.
J Toxicol Sci ; 45(9): 559-567, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32879255

RESUMO

Lead is a main threat to human health due to its neurotoxicity and the astrocyte is known to be a common deposit site of lead in vivo. However, the detailed mechanisms related to lead exposure in the astrocytes were unclear. In order to deeply investigate this issue, we used Sprague-Dawley (SD) rats and astrocytes isolated from the hippocampus of SD rats to establish the lead-exposed animal and cell models through treating with lead acetate. The expression levels of GFAP, LC3, and p62 in the rat hippocampus were detected by immunofluorescence and Western blot after lead exposure. The effects of autophagy on lead-exposed astrocytes were studied by further autophagy inhibitor 3-methyladenine (3-MA) induction. Transmission electron microscopy was used to observe autophagosomes in astrocytes after lead acetate treatment, followed by assessing related autophagy protein markers. In addition, some inflammatory cytokines and oxidative stress markers were also evaluated after lead exposure and 3-MA administration. We found that lead exposure induced activation of astrocytes, as evidenced by increased GFAP levels and GFAP-positive staining cells in the rat hippocampus. Moreover, lead exposure induced autophagy in astrocytes, as evidenced by increased LC3II and Beclin 1 protein levels and decreased p62 expression in both the rat hippocampus and astrocytes, and it was confirmed that this autophagy was activated through blocking the downstream Akt/target of the rapamycin (mTOR) pathway in astrocytes. Furthermore, it was shown that treatment of lead acetate increased the release of tumor necrosis factor-α (TNF-α) and interleukin-1ß (IL-1ß), and the accumulation of malondialdehyde (MDA) and myeloperoxidase (MPO) in astrocytes, which could be alleviated by further 3-MA induction. Therefore, we conclude that lead exposure can induce the autophagy of astrocytes via blocking the Akt/mTOR pathway, leading to accelerated release of inflammatory factors and oxidative stress indicators in astrocytes.


Assuntos
Astrócitos/metabolismo , Astrócitos/fisiologia , Autofagia/efeitos dos fármacos , Autofagia/genética , Compostos Organometálicos/toxicidade , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos , Serina-Treonina Quinases TOR/metabolismo , Animais , Células Cultivadas , Proteína Glial Fibrilar Ácida/genética , Proteína Glial Fibrilar Ácida/metabolismo , Hipocampo/citologia , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Masculino , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Estresse Oxidativo/genética , Ratos Sprague-Dawley , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
18.
Toxicology ; 443: 152557, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32791093

RESUMO

BACKGROUND: Lead induces endothelial dysfunction and hypertension in humans and animals. Seven-day exposure to a low dose in rats reduces vasocontractile responses and increases nitric oxide (NO) bioavailability. We hypothesized that this occurs by angiotensin II receptors (AT1/AT2) activation. MATERIALS AND RESULTS: Wistar rats were exposed to lead acetate (1 st dose 4 µg/100 g, subsequent dose 0.05 µg/100 g/day i.m., 7 days) or saline (control group). Lead acetate exposure reduced the phenylephrine vascular response. Pre-incubations with NO synthase inhibitor N-nitro-L-arginine methyl ester (L-NAME) or phosphatidylinositol 3-kinase (PI3K) inhibitor (wortmannin) increased the contractile response in aortas from lead-treated rats. Pre-incubation with AT2 antagonist (PD123319) restored normal vascular contraction, and both PD123319 or AT1 antagonist (losartan) impeded the potentiated effects of L-NAME and wortmannin. Reinforcing those findings, increased NO bioavailability was blunted by AT1 and AT2 antagonists without summative effect when co-incubated. Finally, to test whether activation of AT1 could upregulate AT2 to increase NO bioavailability rats were simultaneously exposed to lead acetate and treated with losartan (15 mg/kg/day, orally given). Losartan prevented changes on vascular reactivity and endothelial modulation in lead-exposed group. Moreover, incubation with PD123319 had no more effects in aortic from losartan-treated rats. CONCLUSION: Our results suggest that low-dose lead acetate exposure induces an increase of NO involving mainly AT2 receptor activation and the PI3K/Protein Kinase B (PI3K/Akt) pathway. Additionally, we suggest that AT1 activation plays a role in AT2 upregulation, probably as a protective mechanism. Altogether, these effects might contribute to preserving endothelial function against the harmful effects by lead in the vascular system.


Assuntos
Endotélio Vascular/efeitos dos fármacos , Óxido Nítrico/metabolismo , Compostos Organometálicos/toxicidade , Fosfatidilinositol 3-Quinase/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor Tipo 1 de Angiotensina/metabolismo , Animais , Aorta Torácica/efeitos dos fármacos , Aorta Torácica/metabolismo , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Endotélio Vascular/metabolismo , Masculino , Ratos Wistar , Transdução de Sinais/efeitos dos fármacos
19.
Aging (Albany NY) ; 12(14): 14189-14204, 2020 07 17.
Artigo em Inglês | MEDLINE | ID: mdl-32680977

RESUMO

Multi-walled carbon nanotubes (MWCNTs) have been widely used in sewage disposal, water purification, and disinfection. Co-exposure to MWCNTs and heavy metal ions is common during water disposal. However, the hepatotoxicity of co-exposure to MWCNTs and lead ions for nonalcoholic fatty liver disease (NAFLD) subjects has not been investigated. NAFLD mice were fed intragastrically with MWCNTs and lead acetate (PbAc). Combined administration of MWCNTs and PbAc significantly damaged the liver function, and aggravated the nonalcoholic steatohepatitis phenotype as well as the hepatic fibrosis and steatosis in NAFLD mice. Furthermore, MWCNTs and PbAc significantly induced apoptosis in primary hepatocytes isolated from NAFLD mice. Combined administration of MWCNTs and PbAc also resulted in hepatic lipid peroxidation by inducing antioxidant defense system dysfunction, and significantly enhanced the expression levels of inflammatory cytokines in NAFLD mice livers. Meanwhile, combined administration of MWCNTs and PbAc may exert its hepatotoxicity in the NAFLD via inhibiting the adenosine 5'-monophosphate activated protein kinase (AMPK)/peroxisome proliferator-activated receptors γ (PPARγ) pathway. Taken together, we conclude that co-exposure to MWCNTs and PbAc can remarkably aggravate the hepatotoxicity in NAFLD mice via inhibiting the AMPK/PPARγ pathway. This study may provide a biosafety evaluation for the application of nanomaterials in wastewater treatment.


Assuntos
Proteínas Quinases Ativadas por AMP/genética , Doença Hepática Induzida por Substâncias e Drogas/patologia , Nanotubos de Carbono/toxicidade , Hepatopatia Gordurosa não Alcoólica/patologia , Compostos Organometálicos/toxicidade , PPAR gama/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Animais , Antioxidantes/metabolismo , Apoptose/efeitos dos fármacos , Doença Hepática Induzida por Substâncias e Drogas/genética , Citocinas/biossíntese , Hepatócitos/efeitos dos fármacos , Metabolismo dos Lipídeos , Peroxidação de Lipídeos/efeitos dos fármacos , Fígado/efeitos dos fármacos , Fígado/metabolismo , Testes de Função Hepática , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Hepatopatia Gordurosa não Alcoólica/induzido quimicamente , Hepatopatia Gordurosa não Alcoólica/genética , Poluentes Químicos da Água/toxicidade
20.
Oxid Med Cell Longev ; 2020: 4981386, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32566085

RESUMO

Exposure to lead (Pb) causes multiorgan dysfunction including reproductive impairments. Here, we examined the protective effects of coenzyme Q10 (CoQ10) administration on testicular injury induced by lead acetate (PbAc) exposure in rats. This study employed four experimental groups (n = 7) that underwent seven days of treatment as follows: control group intraperitoneally (i.p.) treated with 0.1 ml of 0.9% NaCl containing 1% Tween 80 (v : v), CoQ10 group that was i.p. injected with 10 mg/kg CoQ10, PbAc group that was i.p. treated with PbAc (20 mg/kg), and PbAc+CoQ10 group that was i.p. injected with CoQ10 2 h after PbAc. PbAc injection resulted in increasing residual Pb levels in the testis and reducing testosterone, luteinizing hormone, and follicle-stimulating hormone levels. Additionally, PbAc exposure resulted in significant oxidative damage to the tissues on the testes. PbAc raised the levels of prooxidants (malondialdehyde and nitric oxide) and reduced the amount of endogenous antioxidative proteins (glutathione and its derivative enzymes, catalase, and superoxide dismutase) available in the cell. Moreover, PbAc induced the inflammatory response as evidenced by the upregulation of inflammatory mediators (tumor necrosis factor-alpha and interleukin-1 beta). Further, PbAc treatment induced apoptosis in the testicular cells, as indicated by an increase in Bax and caspase 3 expression, and reduced Bcl2 expression. CoQ10 supplementation improved testicular function by inhibiting Pb accumulation, oxidative stress, inflammation, cell death, and histopathological changes following PbAc exposure. Our findings suggest that CoQ10 can act as a natural therapeutic agent to protect against the reproductive impairments associated with PbAc exposure.


Assuntos
Compostos Organometálicos/toxicidade , Testículo/patologia , Ubiquinona/análogos & derivados , Animais , Caspase 3/genética , Caspase 3/metabolismo , Hormônio Foliculoestimulante/sangue , Glutationa/metabolismo , Interleucina-1beta/metabolismo , Peroxidação de Lipídeos/efeitos dos fármacos , Masculino , Óxido Nítrico/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos Wistar , Testículo/efeitos dos fármacos , Testosterona/sangue , Fator de Necrose Tumoral alfa/metabolismo , Ubiquinona/administração & dosagem , Ubiquinona/farmacologia , Proteína X Associada a bcl-2/genética , Proteína X Associada a bcl-2/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA