Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 476
Filtrar
1.
Ecotoxicol Environ Saf ; 274: 116195, 2024 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-38479315

RESUMO

Fluoride is known to induce nephrotoxicity; however, the underlying mechanisms remain incompletely understood. Therefore, this study aims to explore the roles and mechanisms of lysosomal membrane permeabilization (LMP) and the GSDME/HMGB1 axis in fluoride-induced nephrotoxicity and the protective effects of rutin. Rutin, a naturally occurring flavonoid compound known for its antioxidative and anti-inflammatory properties, is primarily mediated by inhibiting oxidative stress and reducing proinflammatory markers. To that end, we established in vivo and in vitro models. In the in vivo study, rats were exposed to sodium fluoride (NaF) throughout pregnancy and up until 2 months after birth. In parallel, we employed in vitro models using HK-2 cells treated with NaF, n-acetyl-L-cysteine (NAC), or rutin. We assessed lysosomal permeability through immunofluorescence and analyzed relevant protein expression via western blotting. Our findings showed that NaF exposure increased ROS levels, resulting in enhanced LMP and increased cathepsin B (CTSB) and D (CTSD) expression. Furthermore, the exposure to NaF resulted in the upregulation of cleaved PARP1, cleaved caspase-3, GSDME-N, and HMGB1 expressions, indicating cell death and inflammation-induced renal damage. Rutin mitigates fluoride-induced nephrotoxicity by suppressing ROS-mediated LMP and the GSDME/HMGB1 axis, ultimately preventing fluoride-induced renal toxicity occurrence and development. In conclusion, our findings suggest that NaF induces renal damage through ROS-mediated activation of LMP and the GSDME/HMGB1 axis, leading to pyroptosis and inflammation. Rutin, a natural antioxidative and anti-inflammatory dietary supplement, offers a novel approach to prevent and treat fluoride-induced nephrotoxicity.


Assuntos
Fluoretos , Proteína HMGB1 , Nefropatias , Rutina , Animais , Ratos , Anti-Inflamatórios/farmacologia , Antioxidantes/metabolismo , Caspase 3/metabolismo , Fluoretos/metabolismo , Fluoretos/toxicidade , Proteína HMGB1/efeitos dos fármacos , Proteína HMGB1/metabolismo , Inflamação/metabolismo , Lisossomos/efeitos dos fármacos , Piroptose/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Espécies Reativas de Oxigênio/toxicidade , Rutina/farmacologia , Fluoreto de Sódio/toxicidade , Nefropatias/induzido quimicamente , Nefropatias/tratamento farmacológico , Gasderminas/efeitos dos fármacos , Gasderminas/metabolismo
2.
Environ Sci Pollut Res Int ; 31(13): 19844-19855, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38367109

RESUMO

It is widespread of endemic fluorosis in China, and the exposure of excessive fluoride will cause nervous system disease and activate microglia. However, the mechanism of the damage is not clear. It is well-known that NLRP3/Caspase-1/GSDMD pathway, a classic pyroptosis pathway, is widely involved in the occurrence and development of nervous system-related diseases, infectious diseases, and atherosclerotic diseases. This research aimed to explore the molecular mechanism of sodium fluoride on inflammation and pyroptosis in BV2 microglia based on the NLRP3/Caspase-1/GSDMD signaling pathway. BV2 microglia was treated with sodium fluoride at the dose of 0.25, 1, and 2 mmol/L for 24, 48, and 72 h, respectively. Cell viability, cell morphology, lactate dehydrogenase content, and related proteins and genes were examined to investigate if sodium fluoride caused damage to BV2 microglia through the pyroptosis pathway. Dithiolam (5 µmol/L), a pyroptosis inhibitor, was added for further verification. NaF could induced BV2 cells injury in a dose-dependent fashion through disrupting the integrity of cell membranes and increasing IL-1ß via upregulating NLRP3, Caspase-1, and its downstream protein GSDMD. Disulfiram could improve these changes caused by NaF. In conclusion, our results suggested that NLRP3/Caspase-1/GSDMD-mediated classical pyroptosis pathway was involved in fluoride-induced BV2 microglia damage.


Assuntos
Fluoretos , Microglia , Fluoreto de Sódio , Caspase 1/efeitos dos fármacos , Caspase 1/metabolismo , Fluoretos/toxicidade , Microglia/efeitos dos fármacos , Microglia/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/efeitos dos fármacos , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Fluoreto de Sódio/toxicidade , Gasderminas/efeitos dos fármacos , Gasderminas/metabolismo , Animais , Camundongos
3.
Environ Toxicol Pharmacol ; 106: 104360, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38176602

RESUMO

Anthropogenic activities are increasing fluoride concentration in watercourses. The present study focuses on the sublethal toxicity of sodium fluoride during sub-chronic and chronic time periods in the freshwater fish Anabas testudineus. The 96-hour LC50 value for fluoride was found to be 616.50 mg/L. Excessive mucous production and hyper excitability, followed by loss of balance, were seen in fish under acute fluoride exposure. Significant reduction in yield and specific growth rate of fish were assessed at 15, 30 and 45-days exposure intervals. Different bio-indicators like Hepatosomatic-index, Gonadosomatic-index and fecundity were reduced significantly in fish exposed to 10% (61.6 mg/L) and 20% (123.2 mg/L) of 96 h of LC50 values of fluoride in comparison to control. Toxicant concentrations directly correlated with parameter lowering. Fluoride exposure increased plasma glucose, creatinine, AST, and ALT and reduced total RBC, haemoglobin content, Hct (%), plasma protein, and cholesterol. Moreover, fluoride exposure significantly reduces the mitochondrial membrane potential in liver. This may result in metabolic depression, haematological, biochemical, and enzymological stress. The in-silico structural analysis predicts that fluoride may impede cytochrome c oxidase of the electron transport system, hence inhibiting mitochondrial functionality. These findings collectively highlight the urgent need for stringent regulation and monitoring of fluoride levels in freshwater ecosystems, as the subchronic and chronic effects observed in A. testudineus may have broader implications for aquatic ecosystems.


Assuntos
Doenças Mitocondriais , Percas , Animais , Fluoreto de Sódio/toxicidade , Fluoretos/toxicidade , Ecossistema , Fígado
4.
Ecotoxicol Environ Saf ; 271: 115947, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38215664

RESUMO

Fluoride induced reprotoxicity through oxidative stress-mediated reproductive cell death. Hence, the current study evaluated the importance of the MST/Nrf2/MAPK/NQO-HO1 signaling pathway in fluorosis-induced reproductive toxicity. For this purpose, the reproductive toxicity of sodium fluoride (NaF) at physiological, biochemical, and intracellular levels was evaluated. In-vivo, NaF at 100 mg/L instigated physiological dysfunction, morphological, stereological, and structural injuries in the gut-gonadal axis of fluorosis mice through weakening the antioxidant signaling, Nrf2/HO-1/NQO1signaling pathway, causing the gut-gonadal barrier disintegrated via oxidative stress-induced inflammation, mitochondrial damage, apoptosis, and autophagy. Similar trends were also observed in-vitro in the isolated Leydig cells (LCs) challenging with 20 mg/L NaF. Henceforth, activating the cellular antioxidant signaling pathway, Nrf2/HO-1/NQO1, inactivating autophagy and apoptosis, or attenuating lipopolysaccharide (LPS) can be the theoretical basis and valuable therapeutic targets for coping with NaF-induced reproductive toxicity.


Assuntos
Antioxidantes , Fator 2 Relacionado a NF-E2 , Masculino , Camundongos , Animais , Antioxidantes/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Proteína 1 Associada a ECH Semelhante a Kelch/metabolismo , Transdução de Sinais , Estresse Oxidativo , Fluoreto de Sódio/toxicidade , Apoptose
5.
Toxicology ; 501: 153688, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-38036095

RESUMO

Fluorosis poses a significant threat to human and animal health and is an urgent public safety concern in various countries. Subchronic exposure to fluoride has the potential to result in pathological damage to the heart, but its potential mechanism requires further investigation. This study investigated the effects of long-term exposure to sodium fluoride (0, 500, 1000, and 2000 mg/kg) on the hearts of chickens were investigated. The results showed that an elevated exposure dose of sodium fluoride led to congested cardiac tissue and disrupted myofiber organisation. Sodium fluoride exposure activated the ERS pathways of PERK, IRE1, and ATF6, increasing HSP60 and HSP70 and decreasing HSP90. The NF-κB pathway and the activation of TNF-α and iNOS elicited an inflammatory response. BAX, cytc, and cleaved-caspase3 were increased, triggering apoptosis and leading to cardiac injury. The abnormal expression of HSP90 and HSP70 affected the stability and function of RIPK1, RIPK3, and MLKL, which are crucial necroptosis markers. HSPs inhibited TNF-α-mediated necroptosis and apoptosis of the death receptor pathway. Sodium fluoride resulted in heart injury in chickens because of the ERS and variations in HSPs, inducing inflammation and apoptosis. Cardiac-adapted HSPs impeded the activation of necroptosis. This paper may provide a reference for examining the potential cardiotoxic effects of sodium fluoride.


Assuntos
Fluoretos , Proteínas de Choque Térmico , Animais , Humanos , Proteínas de Choque Térmico/metabolismo , Proteínas de Choque Térmico/farmacologia , Fluoretos/toxicidade , Galinhas/metabolismo , Fluoreto de Sódio/toxicidade , Cardiotoxicidade , Fator de Necrose Tumoral alfa , Proteínas de Choque Térmico HSP70 , Apoptose , Proteínas de Choque Térmico HSP90 , Fator 6 Ativador da Transcrição/metabolismo , Fator 6 Ativador da Transcrição/farmacologia
6.
Biomed Pharmacother ; 170: 116080, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38147737

RESUMO

The current study aimed to explore the possible prophylactic and therapeutic effect of Nigella sativa L. oil (NSO) against disruption of endocrine signals and injuries in the thyroid gland, ovary, and uterine tissues induced by sodium fluoride (NaF). Twenty-eight mature female Wistar rats were randomly allocated into four experimental groups (n = 7/group) as follows: control group; NaF group, orally received NaF (20 mg/kg b.wt.) daily; NSO/NaF, orally received NSO (300 mg/kg b.wt.) two weeks before being given NaF and continued throughout the experiment; and NSO+NaF group orally received NSO concurrently with NaF. Our results indicated that NSO restored hormonal balance and suppressed oxidative damage and inflammation. Moreover, the levels of triiodothyronine, thyroxine, thyroid peroxidase, estrogen (E2), progesterone, follicle-stimulating hormone, and luteinizing hormone were elevated, while prostaglandins F2-α and cortisol levels were decreased in NSO treated groups compared to NaF-intoxicated rats. As well, NSO significantly boosted levels of antioxidant molecules, and lowered lipid peroxidation of examined tissues, unlike NaF-treated group. NSO also up-regulated antioxidant enzymes, anti-apoptotic protein, zona pellucida sperm-binding protein, bone morphogenetic protein, and thyroid stimulating hormone, conversely down-regulated inflammatory cytokines, apoptotic proteins, estrogen receptor-α, estrogen receptor-ß, and thyroid stimulating hormone receptors compared to NaF-intoxicated group. Additionally, NSO ameliorated tissue damage of the thyroid gland, ovary, and uterus induced by NaF. -Overall, the prophylactic group (NSO/NaF) performed better antioxidant and anti-inflammatory activities than the treated group almost in all examined tissues, which is reflected by the improvement in the structure of the thyroid, ovarian, and uterine tissues.


Assuntos
Nigella sativa , Glândula Tireoide , Ratos , Feminino , Masculino , Animais , Ratos Wistar , Antioxidantes/farmacologia , Antioxidantes/metabolismo , Ovário , Fluoreto de Sódio/toxicidade , Fluoreto de Sódio/metabolismo , Óleos de Plantas/farmacologia , Estresse Oxidativo , Útero/metabolismo , Receptores de Estrogênio/metabolismo , Sementes
7.
J Trace Elem Med Biol ; 80: 127293, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37677921

RESUMO

BACKGROUND: Out of all measure systemic exposure to fluorides can cause defect of skeletal and dental fluorosis. Endoplasmic reticulum (ER) stress is caused by fluorine-induced oxidative stress and importance of vitamin D in its prevention is not known enough in bone cells. This study was carried out to investigate fluorine-induced oxidative stress, ER stress, and death pathways and the effect of vitamin D on them. METHODS: MC3T3-E1 mouse osteoblast cell line was used as the material of the study. The NaF and vitamin D concentrations were determined by the MTT assay. NaF treatments and vitamin D supplementation (pre-add, co-add, and post-add) was administered in the cell line at 24th and 48th hours. The expression of the genes in oxidative stress, ER stress, and death pathways was determined using RT-qPCR and Western blotting techniques. RESULTS: Vitamin D significantly reduced mRNA expression levels of SOD2, CYGB, ATF6, PERK, IRE1, ATG5 and BECN1 whereas caused an increase in levels GPX1, SOD1, NOS2 and Caspase-3 in MC3T3-E1 mouse osteoblast cell line of NaF-induced. In addition, GPX1, SOD1, ATF6, PERK, IRE1, BECN1, Caspase-3 and RIPK1 protein levels were examined by Western blot analysis, and it was determined that vitamin D decreased IRE1 and PERK protein levels, but increased GPX1, SOD1, ATF6 and Caspase-3 protein levels. CONCLUSION: The findings of the study suggest that vitamin D has protective potential against NaF-induced cytotoxicity reasonably through the attenuation of oxidative stress, ER stress, ATG5, IRE1 and by increasesing caspase-3 in vitro conditions.


Assuntos
Fluoreto de Sódio , Vitamina D , Camundongos , Animais , Fluoreto de Sódio/toxicidade , Vitamina D/metabolismo , Caspase 3/metabolismo , Flúor , Superóxido Dismutase-1/metabolismo , Linhagem Celular , Estresse do Retículo Endoplasmático , Osteoblastos/metabolismo , Estresse Oxidativo , Apoptose
8.
P R Health Sci J ; 42(2): 132-138, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37352535

RESUMO

OBJECTIVE: To evaluate the morphological changes of cardiomyocytes exposed to different sodium fluoride (NaF) concentrations, as well as to evaluate the behavior of the mitochondria. METHODS: Rat H9c2 cardiomyocytes were exposed to NaF at concentrations of 0.5 to 5 mmol/L. The morphology and number of mitochondria in these cells were monitored, and the calcium ion (Ca2+) concentration was determined. RESULTS: Morphological changes were evident in the cells treated with different NaF concentrations, and both the number of mitochondria and the Ca2+ concentration decreased in a dose-dependent manner. CONCLUSION: Sodium fluoride induced morphological damage in cardiomyocytes, decreases the Ca2+ concentration and mitochondrial number.


Assuntos
Fluoretos , Fluoreto de Sódio , Ratos , Animais , Fluoretos/toxicidade , Fluoreto de Sódio/toxicidade , Miócitos Cardíacos , Cálcio , Células Cultivadas
9.
Neuroreport ; 34(9): 449-456, 2023 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-37161984

RESUMO

Fluorosis is widespread in several areas of the world and including India leading to dental and skeletal fluorosis as well as neurological manifestations. With a limited number of treatment options available, we have tried to address the issue with a nutraceutical such as naringin which is an alkaloid derived from the citrus fruit. Naringin is a potent antioxidant and has neuroprotective action which can counteract the redox imbalance induced by sodium fluoride ingestion. Neurological effects of fluorosis were evaluated in Wistar rats by open field test (OFT) and novel object recognition test (NORT) along with lipid peroxidation (LPO) and glutathione estimation in brain homogenate and cresyl violet staining of CA3 neurons in the hippocampus. Animals were divided into groups namely, normal, vehicle, fluoride, naringin 100 mg/kg bd.wt group and fluoride with naringin (FLU-NAR) group. Fluorosis was induced by providing 100 ppm of sodium fluoride ad libitum in drinking water for 30 days and prophylactic treatment of naringin for 15 days per oral. OFT, NORT and forced swim test showed significant (P ≤ 0.05) changes in the FLU-NAR group as compared to the fluoride group indicating behavioral changes in the fluoride group and positive changes in the FLU-NAR group with attenuation of stress, fear, hyperactivity and memory impairment. The decrease in LPO and increase in glutathione levels in the treatment group compared to the fluoride group were supported by histological improvement as compared to the fluoride group. Prophylactic treatment of naringin showed its possible neuroprotective effect, thus giving an alternative treatment strategy to deal with neurological manifestations of fluorosis.


Assuntos
Fármacos Neuroprotetores , Ratos , Animais , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Fluoretos/toxicidade , Ratos Wistar , Fluoreto de Sódio/toxicidade , Glutationa
10.
Ecotoxicol Environ Saf ; 255: 114772, 2023 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-36924562

RESUMO

Chronic fluoride exposure can cause developmental neurotoxicity, however the precise mechanisms remain unclear. To explore the mechanism of mitophagy in fluoride-induced developmental neurotoxicity, specifically focusing on PRKAA1 in regulating the PINK1/Parkin pathway, we established a Sprage Dawley rat model with continuous sodium fluoride (NaF) exposure and an NaF-treated SH-SY5Y cell model. We found that NaF exposure increased the levels of LC3-Ⅱ and p62, impaired autophagic degradation, and subsequently blocked autophagic flux. Additionally, NaF exposure increased the expression of PINK1, Parkin, TOMM-20, and Cyt C and cleaved PARP in vivo and in vitro, indicating NaF promotes mitophagy and neuronal apoptosis. Meanwhile, phosphoproteomics and western blot analysis showed that NaF treatment enhanced PRKAA1 phosphorylation. Remarkably, the application of both 3-methyladenosine (3-MA; autophagy inhibitor) and dorsomorphin (DM; AMPK inhibitor) suppressed NaF-induced neuronal apoptosis by restoring aberrant mitophagy. In addition, 3-MA attenuated an increase in p62 protein levels and NaF-induced autophagic degradation. Collectively, our findings indicated that NaF causes aberrant mitophagy via PRKAA1 in a PINK1/Parkin-dependent manner, which triggers neuronal apoptosis. Thus, regulating PRKAA1-activated PINK1/Parkin-dependent mitophagy may be a potential treatment for NaF-induced developmental neurotoxicity.


Assuntos
Neuroblastoma , Síndromes Neurotóxicas , Ratos , Humanos , Animais , Mitofagia/fisiologia , Fluoretos/metabolismo , Proteínas Quinases/genética , Proteínas Quinases/metabolismo , Mitocôndrias/metabolismo , Neuroblastoma/metabolismo , Síndromes Neurotóxicas/etiologia , Síndromes Neurotóxicas/metabolismo , Fluoreto de Sódio/toxicidade , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo , Proteínas Quinases Ativadas por AMP/metabolismo
11.
Biol Trace Elem Res ; 201(12): 5734-5746, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36884125

RESUMO

Hepatotoxicity induced by excessive fluoride (F) exposure has been extensively studied in both humans and animals. Chronic fluorosis can result in liver apoptosis. Meanwhile, moderate exercise alleviates apoptosis caused by pathological factors. However, the effect of moderate exercise on F-induced liver apoptosis remains unclear. In this research, sixty-four three-week-old Institute of Cancer Research (ICR) mice, half male and half female, were randomly divided into four groups: control group (distilled water); exercise group (distilled water and treadmill exercise); F group [100 mg/L sodium fluoride (NaF)]; and exercise plus F group (100 mg/L NaF and treadmill exercise). The liver tissues of mice were taken at 3 months and 6 months, respectively. Hematoxylin-eosin (HE) staining and situ terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) results showed that nuclear condensation and apoptotic hepatocytes occurred in the F group. However, this phenomenon could be reversed with the intervention of treadmill exercise. The results of QRT-PCR and western blot displayed NaF- induced apoptosis via tumor necrosis factor recpter 1 (TNFR1) signaling pathway, while treadmill exercise could restore the molecular changes caused by excessive NaF exposure.


Assuntos
Fluoretos , Fígado , Humanos , Camundongos , Masculino , Feminino , Animais , Fluoretos/toxicidade , Fluoretos/metabolismo , Fígado/metabolismo , Apoptose , Fluoreto de Sódio/toxicidade , Água/metabolismo
12.
Ecotoxicol Environ Saf ; 252: 114646, 2023 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-36791501

RESUMO

Mitophagy has distinct functions, which can lead to either protection or damage of tissues. Though current evidence indicated that NaF triggers mitophagy, the role and regulation of mitophagy in sodium fluoride (NaF)-induced liver injury still remain unclear. Therefore, we exployed the cell and mouse models and confirmed that NaF treatment activates mitophagy. Knocking down PTEN-induced putative kinase protein 1 (PINK1) expression attenuated mitophagy and increased the degree of mitochondrial impairment, oxidative stress, and apoptosis in NaF-treated HepG2 cells. In vivo experiments indicated that PINK1 deficiency weakened NaF-induced mitophagy. Moreover, PINK1-deficient mices aggravated NaF-induced hepatic mitochondrial injury, oxidative stress, and inflammation in livers, evidenced by the increased number of abnormal mitochondria, decreased adenosine triphosphate (ATP) and glutathione (GSH) levels, elevated reactive oxygen species (ROS) and malondialdehyde (MDA) content, enhanced hepatic macrophage infiltration and inflammatory cytokine levels. Notably, NaF exposure activated Nrf2 signaling both in vitro and in vivo. Nrf2 siRNA transfection blocked the upregulation of PINK1 expression and the induction of mitophagy in NaF-treated HepG2 cells. Also, ML385 (Nrf2 inhibitor) partially blocked the upregulation of PINK1 expression caused by NaF in mice livers. To sum up, the present study provided the demonstration that Nrf2/PINK1-mediated mitophagy activation offers a hepatoprotective effect by inhibiting NaF-induced mitochondrial dysfunction, oxidative stress, and inflammation.


Assuntos
Mitofagia , Fluoreto de Sódio , Camundongos , Animais , Fluoreto de Sódio/toxicidade , Fator 2 Relacionado a NF-E2/genética , Fator 2 Relacionado a NF-E2/metabolismo , Proteínas Quinases/genética , Proteínas Quinases/metabolismo , Mitocôndrias , Estresse Oxidativo , Espécies Reativas de Oxigênio/metabolismo , Inflamação/induzido quimicamente , Inflamação/metabolismo , Fígado/metabolismo , Glutationa/metabolismo
13.
Ecotoxicol Environ Saf ; 253: 114674, 2023 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-36827899

RESUMO

Excessive fluoride exposure can cause liver injury, but the specific mechanisms need further investigation. We aimed to explore the role of impaired lysosomal biogenesis and defective autophagy in fluoride-induced hepatotoxicity and its potential mechanisms, focusing on the role of transcription factor E3 (TFE3) in regulating hepatocyte lysosomal biogenesis. To this end, we established a Sprague-Dawley (SD) rat model exposed to sodium fluoride (NaF) and a rat liver cell line (BRL3A) model exposed to NaF. The results showed that NaF exposure diminished liver function and led to apoptosis as well as autophagosome accumulation and impaired autophagic degradation. In addition, NaF exposure caused compromised lysosome biogenesis and decreased lysosomal degradation, and inhibited TFE3 nuclear translocation. Notably, the mTOR inhibitors rapamycin (RAPA) and Ad-TFE3 promoted lysosomal biogenesis and enhanced lysosomal degradation function. Furthermore, RAPA and Ad-TFE3 reduced NaF-induced apoptosis by alleviating impaired autophagic degradation. In conclusion, NaF impairs lysosomal biogenesis by inhibiting TFE3 nuclear translocation, decreasing lysosomal degradation function, resulting in impaired autophagic degradation, and ultimately inducing apoptosis. Therefore, TFE3 may be a promising therapeutic target for fluoride-induced hepatotoxicity.


Assuntos
Doença Hepática Induzida por Substâncias e Drogas , Fluoretos , Ratos , Animais , Fluoretos/toxicidade , Fluoretos/metabolismo , Ratos Sprague-Dawley , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/genética , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Autofagia , Fluoreto de Sódio/toxicidade , Lisossomos/metabolismo , Doença Hepática Induzida por Substâncias e Drogas/etiologia , Doença Hepática Induzida por Substâncias e Drogas/metabolismo
14.
Chem Biodivers ; 20(3): e202200982, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36808882

RESUMO

The ameliorative effects of hesperidin (HES) on the toxicities created by sodium fluoride (NaF) in the testes tissue of rats were studied via oxidative stress, apoptosis and endoplasmic reticulum (ER) stress pathways. The animals were divided into five distinct groups (7 rats in each group). Group 1 was control group, group 2 received NaF-only (600 ppm), group 3 received HES-only (200 mg/kg bw); group 4 received NaF (600 ppm)+HES (100 mg/kg bw) and group 5 received NaF (600 ppm)+HES (200 mg/kg bw) for 14 days. NaF-induced testes tissue damage by reducing activities of superoxide dismutase (SOD), catalase (CAT) and glutathione peroxidase (GPx) and levels of glutathione (GSH), and increasing lipid peroxidation levels. NaF treatment significantly downregulated the mRNA levels of SOD1, CAT and GPx. NaF supplementation caused apoptosis in the testes by upregulating p53, NFkB, caspase-3, caspase-6, caspase-9, and Bax and downregulating Bcl-2. Furthermore, NaF caused ER stress via increasing mRNA transcript levels of PERK, IRE1, ATF-6 and GRP78. NaF treatment led to autophagy via upregulation of Beclin1, LC3A, LC3B and AKT2. In testes tissue, however, co-treatment with HES at doses of 100 and 200 mg/kg significantly reduced oxidative stress, apoptosis, autophagy and ER stress. Overall, the findings of this study suggest that HES may help to reduce testes damage caused by NaF toxicity.


Assuntos
Hesperidina , Fluoreto de Sódio , Masculino , Ratos , Animais , Fluoreto de Sódio/toxicidade , Testículo , Hesperidina/farmacologia , Estresse Oxidativo , Apoptose , Estresse do Retículo Endoplasmático , Autofagia , RNA Mensageiro , Antioxidantes/farmacologia
15.
Ecotoxicol Environ Saf ; 250: 114511, 2023 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-36608573

RESUMO

Fluoride is capable of inducing developmental neurotoxicity; regrettably, the mechanism is obscure. We aimed to probe the role of lysosomal biogenesis disorder in developmental fluoride neurotoxicity-specifically, the regulating effect of the transient receptor potential mucolipin 1 (TRPML1)/transcription factor EB (TFEB) signaling pathway on lysosomal biogenesis. Sprague-Dawley rats were given fluoridated water freely, during pregnancy to the parental rats to 2 months after delivery to the offspring. In addition, neuroblastoma SH-SY5Y cells were treated with sodium fluoride (NaF), with or without mucolipin synthetic agonist 1 (ML-SA1) or adenovirus TFEB (Ad-TFEB) intervention. Our findings revealed that NaF impaired learning and memory as well as memory retention capacities in rat offspring, induced lysosomal biogenesis disorder, and decreased lysosomal degradation capacity, autophagosome accumulation, autophagic flux blockade, apoptosis, and pyroptosis. These changes were evidenced by the decreased expression of TRPML1, nuclear TFEB, LAMP2, CTSB, and CTSD, as well as increased expression of LC3-II, p62, cleaved PARP, NLRP3, Caspase1, and IL-1ß. Furthermore, TRPML1 activation and TFEB overexpression both restored TFEB nuclear protein expression and promoted lysosomal biogenesis while enhancing lysosomal degradation capacity, recovering autophagic flux, and attenuating NaF-induced apoptosis and pyroptosis. Taken together, these results show that NaF promotes the progression of developmental fluoride neurotoxicity by inhibiting TRPML1/TFEB expression and impeding lysosomal biogenesis. Notably, the activation of TRPML1/TFEB alleviated NaF-induced developmental neurotoxicity. Therefore, TRPML1/TFEB may be promising markers of developmental fluoride neurotoxicity.


Assuntos
Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos , Fluoretos , Neuroblastoma , Síndromes Neurotóxicas , Canais de Potencial de Receptor Transitório , Animais , Humanos , Ratos , Autofagia , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Fluoretos/toxicidade , Lisossomos , Neuroblastoma/metabolismo , Síndromes Neurotóxicas/etiologia , Síndromes Neurotóxicas/metabolismo , Ratos Sprague-Dawley , Fluoreto de Sódio/toxicidade , Canais de Potencial de Receptor Transitório/metabolismo
16.
Biol Trace Elem Res ; 201(1): 295-305, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-35226278

RESUMO

It is critical to determine the mechanism underlying fluoride (F)-induced damage of the testes to develop appropriate strategies for monitoring and intervention. In the present study, exposure to 50 mg/L sodium fluoride (NaF) for 90 days damaged the normal structure of the testes and quality of the sperm, particularly the spermatocytes, and triggered overexpression of human antigen R (Elavl1/HuR) according to western blotting and immunofluorescence. Furthermore, 0.5 mM NaF exposure for 24 h exposure increased the proportion of apoptosis and expression of caspase-3 and caspase-9 in mouse spermatocytes (GC-2spd cell line), whereas inhibition of HuR reduced apoptosis and the expression of caspase-3 and caspase-9. Additionally, inhibition of HuR alleviated F-induced autophagy based on observation of the autophagy bodies, detection of autophagy activity, and analysis of the expression of the LC3II/LC3I and p62 proteins. These results reveal that excessive F can lead to overexpression of HuR, resulting in high levels of apoptosis and autophagy in spermatocytes. These findings improve the understanding of the mechanisms underlying F-induced male reproductive toxicity, and HuR may be explored as a treatment target for certain conditions. Excessive fluoride can induce overexpression of HuR in testis and result in excessive apoptosis and autophagy in spermatocytes as well as male reproductive damage, such as a decreased sperm count, decreased sperm motility, and increased deformity rate.


Assuntos
Fluoretos , Espermatócitos , Masculino , Camundongos , Humanos , Animais , Fluoretos/metabolismo , Espermatócitos/metabolismo , Caspase 3/metabolismo , Caspase 9/metabolismo , Sêmen/metabolismo , Motilidade dos Espermatozoides , Fluoreto de Sódio/toxicidade , Apoptose , Autofagia
17.
Biol Trace Elem Res ; 201(2): 698-705, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-35267138

RESUMO

This study was planned to evaluate the effect of vitamin D administration on cytotoxicity due to fluoride exposure in vitro. NaF (IC50) and vitamin D (proliferative) were applied to human osteoblast (hFOB 1.19) cells. The major genes of apoptotic, autophagic, and necrotic pathways were determined by RT-PCR. 2-∆∆Ct formulation was used for expression analysis. In the NaF group, caspase 3, Bax, Bad, Bak, Bclx, Atg3, Atg5, Atg6, pG2, LC3-I, LC3-II, RIP1, and RIP3 genes were increased (2.6-15 times). It was observed that the expressions of these genes approached the control when vitamin D was given together with NaF. The Bcl2 gene increased significantly (sixfold) with the effect of NaF, and was down-regulated to some extent with additional vitamin D administration, but still more than in the control. As a result, it was determined that apoptotic, necrotic, and autophagic pathways were activated as the molecular basis of the damage in the bone tissue, which was most affected by fluorine, and these genes were down-regulated and approached the control group with the addition of vitamin D. It was concluded that this is an important data to explain the molecular basis of the protective and therapeutic effect of vitamin D against fluorine toxicity.


Assuntos
Fluoreto de Sódio , Vitamina D , Humanos , Fluoreto de Sódio/toxicidade , Vitamina D/farmacologia , Vitamina D/metabolismo , Flúor/farmacologia , Fluoretos/farmacologia , Osteoblastos , Apoptose
18.
Biol Trace Elem Res ; 201(1): 229-241, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-35023047

RESUMO

The study was designed to determine the fluoride distribution after its oral exposure in drinking water and its associated impact on biochemical, antioxidant markers and histology in the liver, kidney, and heart of male Wistar rats. On 100 ppm exposure, the highest accretion of fluoride occurred in the liver followed by the kidney and heart. Fluoride exposure significantly (p˂0.05) increased the plasma levels of dehydrogenase, aminotransferases, kidney injury molecule-1 (KIM-1), and other plasma renal biomarkers but decreased the levels of total plasma proteins and albumin in a dose-dependent manner. Reduction (p˂0.05) in the activities of antioxidant enzymes viz. acetylcholinesterase, arylesterase, superoxide dismutase, catalase, glutathione peroxidase, and reductase with increased levels of protein and lipid peroxidation was recorded in the liver, kidney, and heart of fluoride-administered rats. Fluoride exposure (100 ppm) induced lipid peroxidation was highest in kidney (4.4 times) followed by liver (2.6 times) and heart (2.5 times) and as compared to their respective control. The percent rise in protein oxidation at 30% was almost equal in the kidney and liver but was 21.5% in the heart as compared to control. The histopathological alterations observed included congestion and hemorrhage along with degeneration and necrosis of parenchymal cells in hepato-renal tissues and myocardium, severity of which varied in a dose-dependent manner. Taken together, fluoride distribution in the liver, heart, and kidney after chronic fluoride intake correlated well with fluoride-induced hepatic and cardio-renal toxicity in a concentration-dependent manner. These results draw attention that chronic fluoride intake pose a significant health risk for human and animal residents of fluoride endemic areas.


Assuntos
Antioxidantes , Fluoreto de Sódio , Humanos , Ratos , Masculino , Animais , Antioxidantes/metabolismo , Ratos Wistar , Fluoreto de Sódio/toxicidade , Fluoretos/toxicidade , Fluoretos/metabolismo , Acetilcolinesterase/metabolismo , Fígado/metabolismo , Catalase/metabolismo , Rim/metabolismo , Peroxidação de Lipídeos , Superóxido Dismutase/metabolismo , Estresse Oxidativo
19.
Biol Trace Elem Res ; 201(8): 3994-4007, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-36255553

RESUMO

Fluoride is a persistent environmental pollutant, and its excessive intake causes skeletal and dental fluorosis. However, few studies focused on the effects of fluoride on osteocytes, making up over 95% of all bone cells. This study aimed to investigate the effect of fluoride on osteocytes in vitro, as well as explore the underlying mechanisms. CCK-8, LDH assay, fluorescent probes, flow cytometry, and western blotting were performed to examine cell viability, apoptosis, mitochondria changes, reactive oxygen species (ROS) and mitochondrial ROS (mtROS), and protein expressions. Results showed that sodium fluoride (NaF) exposure (4, 8 mmol/L) for 24 h inhibited the cell viability of osteocytes MLO-Y4 and promoted G0/G1 phase arrest and increased cell apoptosis. NaF treatment remarkably caused mitochondria damage, loss of MMP, ATP decrease, Cyto c release, and Bax/Bcl-2 ratio increase and elevated the activity of caspase-9 and caspase-3. Furthermore, NaF significantly upregulated the expressions of LC-3II, PINK1, and Parkin and increased autophagy flux and the accumulation of acidic vacuoles, while the p62 level was downregulated. In addition, NaF exposure triggered the production of intracellular ROS and mtROS and increased malondialdehyde (MDA); but superoxide dismutase (SOD) activity and glutathione (GSH) content were decreased. The scavenger N-acetyl-L-cysteine (NAC) significantly reversed NaF-induced apoptosis and mitophagy, suggesting that ROS is responsible for the mitochondrial-mediated apoptosis and mitophagy induced by NaF exposure. These findings provide in vitro evidence that apoptosis and mitophagy are cellular mechanisms for the toxic effect of fluoride on osteocytes, thereby suggesting the potential role of osteocytes in skeletal and dental fluorosis.


Assuntos
Fluoretos , Fluorose Dentária , Humanos , Fluoretos/toxicidade , Fluoretos/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Mitofagia , Fluorose Dentária/metabolismo , Osteócitos/metabolismo , Apoptose , Fluoreto de Sódio/toxicidade , Mitocôndrias/metabolismo
20.
Environ Toxicol ; 38(1): 193-204, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36190517

RESUMO

Excessive fluoride intake can cause dental fluorosis during teeth development and growth. However, the mechanisms underlying fluoride-induced enamel damage are still not fully elucidated. Previously, we observed fluoride-induced autophagy in ameloblasts, but the effects of fluoride on autophagy flux in ameloblasts remain unclear. Hence, this study aimed to clarify the effects of fluoride and rapamycin, an autophagy activator, on autophagy flux in ameloblasts. This in vitro study used the murine ameloblast-derived cell line LS8. Cells were treated with different concentrations of sodium fluoride (NaF) to evaluate NaF-induced cytotoxicity. Using transmission electron microscopy, we observed an increase in the number of autophagosomes with increasing fluoride concentrations. Western blot analyses showed increases in microtubule-associated protein 1 light chain 3 (LC3) and SQSTM1 (p62) expression after NaF treatment and an increase in LC3II expression after bafilomycin A1 administration. Together with changes in RFP-GFP-LC3 lentivirus expression, this demonstrated that fluoride impaired autophagy flux. Furthermore, we evaluated whether rapamycin can alleviate fluoride-induced cytotoxicity by restoring autophagy flux. Compared to the NaF-treated group, LS8 cells cotreated with NaF and rapamycin grew considerably better and had significantly decreased p62 expression. Taken together, these data suggest that fluoride-induced impaired autophagosome degradation may damage ameloblasts. This provides experimental in vitro evidence and an explanation for the observed NaF-induced toxicity of ameloblasts. Rapamycin probably alleviates this impairment by decreasing the expression of p62, thereby preventing autophagy defects.


Assuntos
Ameloblastos , Fluoretos , Camundongos , Animais , Ameloblastos/metabolismo , Fluoretos/toxicidade , Sirolimo/farmacologia , Autofagia , Fluoreto de Sódio/toxicidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...