Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Toxicol Lett ; 398: 69-81, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38909920

ABSTRACT

Sodium para-aminosalicylic acid (PAS-Na) treatment for manganese (Mn) intoxication has shown efficacy in experimental and clinical studies, giving rise to additional studies on its efficacy for lead (Pb) neurotoxicity and its associated mechanisms of neuroprotection. The difference between PAS-Na and other metal complexing agents, such as edetate calcium sodium (CaNa2-EDTA), is firstly that PAS-Na can readily pass through the blood-brain barrier (BBB), and complex and facilitate the excretion of manganese and lead. Secondly, PAS-Na has anti-inflammatory effects. Recent studies have broadened the understanding on the mechanisms associated with efficacy of PAS-Na. The latter has been shown to modulate multifarious manganese- and lead- induced neurotoxicity, via its anti-apoptotic and anti-inflammatory effects, as well as its ability to inhibit pyroptosis, and regulate abnormal autophagic processes. These observations provide novel scientific bases and new concepts for the treatment of lead, mercury, copper, thallium, as well as other toxic encephalopathies, and implicate PAS-Na as a compound with greater prospects for clinical medical application.


Subject(s)
Aminosalicylic Acid , Lead Poisoning , Manganese Poisoning , Humans , Animals , Aminosalicylic Acid/therapeutic use , Manganese Poisoning/drug therapy , Lead Poisoning/drug therapy , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Anti-Inflammatory Agents/therapeutic use , Anti-Inflammatory Agents/pharmacology , Manganese/toxicity
2.
Biol Trace Elem Res ; 202(5): 2241-2252, 2024 May.
Article in English | MEDLINE | ID: mdl-37500820

ABSTRACT

The aim of study was to address the effects of manganese and iron, alone and in combination, on apoptosis of BV2 microglia cells, and to determine if combined exposure to these metals augments their individual toxicity. We used a murine microglial BV2 cell line. Cell cytotoxicity was analyzed by propidium iodide (PI) exclusion assay. Cell ROS production was analyzed by 2', 7'-dichlorofluorescin diacetate (DCFH-DA) probe staining. Pro-inflammatory cytokine production was monitored by ELISA. Cell apoptosis was analyzed by PE Annexin V/7-AAD staining. Mitochondrial membrane integrity was analyzed by flow cytometry. We used immunoblotting to analyze the effect of manganese, iron alone, or their combined exposure on the activation of caspase9, P53, Bax, and Bcl2 apoptosis signaling pathways. Caspase3 activity was determined using a Colorimetric. Manganese, iron, and their combined exposure for 24 h induced the activation of BV2 microglia cells and increased ROS production and the expression of the inflammatory cytokines, IL-1ß and TNF-α. And we also found that the apoptosis rate increased, mitochondrial membrane potential decreased, apoptosis-related proteins caspase9, P53, Bax, and Bcl2 expression increased, and caspase3 activity increased. Furthermore, we found that combined manganese-iron cytotoxicity was lower than that induced by manganese exposure alone. Manganese, iron alone, or their combination exposure can induce apoptosis in glial cells. Iron can reduce the toxicity of manganese, and there is an antagonistic effect between manganese and iron.


Subject(s)
Iron , Manganese , Mice , Animals , Manganese/toxicity , Manganese/metabolism , Reactive Oxygen Species/metabolism , Iron/metabolism , bcl-2-Associated X Protein/metabolism , Tumor Suppressor Protein p53/metabolism , Apoptosis , Apoptosis Regulatory Proteins/metabolism
4.
Toxicol Ind Health ; 39(8): 464-470, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37404003

ABSTRACT

Lead (Pb) is a corrosion-resistant, heavy, non-ferrous metal. Several metal chelators have been used for the treatment of Pb poisoning. However, the efficacy of sodium para-aminosalicylic acid (PAS-Na) in enhancing Pb excretion has yet to be fully characterized. Healthy male mice (90) were divided into six groups, the normal control group was intraperitoneally (i.p.) injected with saline and the remaining group of mice i.p. 120 mg/kg Pb acetate. Four hour later, mice were subcutaneously (back) injected (s.c.) with (80, 160, 240 mg/kg) PAS-Na or 240 mg/kg edetate calcium disodium (CaNa2EDTA) or an equivalent amount of saline, once per day for 6 days. After 24-h urine sample collections, the animals were anesthetized with 5% chloral hydrate and sacrificed in batches on the 2nd, 4th, or 6th day. Levels of Pb [including manganese (Mn) and copper (Cu)] in the urine, whole blood, and brain tissues were analyzed by graphite furnace atomic absorption spectrometry. The results showed that Pb exposure increased its levels in urine and blood, and PAS-Na treatment may afford antagonistic effect on Pb poisoning, suggesting that PAS-Na is a potentially effective treatment to promote excretion of Pb.


Subject(s)
Aminosalicylic Acid , Rats , Male , Mice , Animals , Aminosalicylic Acid/therapeutic use , Aminosalicylic Acid/pharmacology , Rats, Sprague-Dawley , Lead/toxicity , Sodium , Chelating Agents/pharmacology , Chelating Agents/therapeutic use
5.
Neurotox Res ; 41(1): 1-15, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36598679

ABSTRACT

Lead (Pb) is a common heavy metal contaminant in the environment, and it may perturb autophagy and cause neurodegeneration. Although sodium para-aminosalicylic (PAS-Na) has been shown to protect the brain from lead-induced toxicity, the mechanisms associated with its efficacy have yet to be fully understood. In this study, we evaluated the efficacy of PAS-Na in attenuating the neurotoxic effects of lead, as well as the specific mechanisms that mediate such protection. Lead exposure resulted in weight loss and injury to the liver and kidney, and PAS-Na had a protective effect against this damage. Both short-term and subchronic lead exposure impaired learning ability, and this effect was reversed by PAS-Na intervention. Lead exposure also perturbed autophagic processes through the modulation of autophagy-related factors. Short-term lead exposure downregulated LC3 and beclin1 and upregulated the expression of p62; subchronic lead exposure upregulated the expression of LC3, beclin1, and P62. It follows that PAS-Na had an antagonistic effect on the activation of the above autophagy-related factors. Overall, our novel findings suggest that PAS-Na can protect the rat cortex from lead-induced toxicity by regulating autophagic processes. (1) Short-term lead exposure inhibits autophagy, whereas subchronic lead exposure promotes autophagy. (2) PAS-NA ameliorated the abnormal process of lead-induced autophagy, which had a protective effect on the cerebral cortex.


Subject(s)
Aminosalicylic Acid , Autophagy , Cerebral Cortex , Animals , Rats , Aminosalicylic Acid/pharmacology , Autophagy/drug effects , Beclin-1 , Lead/toxicity , Rats, Sprague-Dawley , Sodium , Cerebral Cortex/pathology , Neurodegenerative Diseases/chemically induced , Neurodegenerative Diseases/pathology
7.
Biol Trace Elem Res ; 201(1): 282-293, 2023 Jan.
Article in English | MEDLINE | ID: mdl-35133610

ABSTRACT

Lead (Pb), a corrosion-resistant heavy non-ferrous metal, is one of the most common environmental neurotoxic metals. The effects of Pb on other essential metal elements are contradictory. Therefore, this in vivo study addressed the effects of sub-chronic Pb exposure on the distribution of other divalent metals, exploring the relationships between Pb levels in blood, teeth, bones, hair, and brain tissues. Thirty-two healthy male C57BL/6 mice received intragastric administration (i.g.) with 0, 12.5, 25, and 50 mg/kg Pb acetate, once a day for 8 weeks. Levels of Pb and other metal elements [including iron(Fe), zinc (Zn), magnesium (Mg), copper (Cu), and calcium(Ca)] in the whole blood, teeth, the right thighbone, hair, and brain tissues (including cortex, hippocampus, striatum, and hypothalamus) were detected with inductively coupled plasma-mass spectrometry (ICP-MS). Pb levels in all detected organs were increased after Pb-exposed for 8 weeks. The results of relationship analysis between Pb levels in the tissues and lifetime cumulative Pb exposure (LCPE) showed that Pb levels in the blood, bone, and hair could indirectly reflect the Pb accumulation in the murine brain. These measures might serve as valuable biomarkers for chronic Pb exposure reflective of the accumulation of Pb in the central nervous system (CNS). Sub-chronic Pb exposure for 8 weeks altered Ca, Cu, Fe, and Zn levels, but no effects were noted on Mg levels in any of the analyzed tissues. Pb decreased Ca in teeth, Cu in thighbone and teeth, Zn in whole blood and hair, and Fe in hair. In contrast, Pb increased Ca levels in corpus striatum and hypothalamus, Cu levels in striatum, Zn levels in teeth, and Fe levels in hippocampus, thighbone, and teeth. The Pb-induced changes in metal ratios in various tissues may serve as valuable biomarkers for chronic Pb exposure as they are closely related to the accumulations of Pb in the murine CNS. The results suggest that altered distribution of several essential metal elements may be involved in Pb-induced neurotoxicity. Additional studies should address the interaction between Pb and essential metal elements in the CNS and other organs.


Subject(s)
Lead , Trace Elements , Male , Mice , Animals , Lead/toxicity , Cadmium/analysis , Mice, Inbred C57BL , Copper/analysis , Zinc/analysis
8.
Neurochem Res ; 48(1): 238-249, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36063295

ABSTRACT

Lead (Pb) is considered to be a major environmental pollutant and occupational health hazard worldwide which may lead to neuroinflammation. However, an effective treatment for Pb-induced neuroinflammation remains elusive. The aim of this study was to investigate the mechanisms of Pb-induced neuroinflammation, and the therapeutic effect of sodium para-aminosalicylic acid (PAS-Na, a non-steroidal anti-inflammatory drug) in rat cerebral cortex. The results indicated that Pb exposure induced pathological damage in cerebral cortex, accompanied by increased levels of inflammatory factors tumor necrosis factor-alpha (TNF-α) and interleukin-1 beta (IL-1ß). Moreover, Pb decreased the expression of silencing information regulator 2 related enzyme 1 (SIRT1) and brain-derived neurotrophic factor (BDNF), and increased the levels of high mobile group box 1 (HMGB1) expression and p65 nuclear factor-κB (NF-κB) phosphorylation. PAS-Na treatment ameliorated Pb-induced histopathological changes in rat cerebral cortex. Moreover, PAS-Na reduced the Pb-induced increase of TNF-α and IL-1ß levels concomitant with a significant increase in SIRT1 and BDNF levels, and a decrease in HMGB1 and the phosphorylation of p65 NF-κB expression. Thus, PAS-Na may exert anti-inflammatory effects by mediating the SIRT1/HMGB1/NF-κB pathway and BDNF expression. In conclusion, in this novel study PAS-Na was shown to possess an anti-inflammatory effect on cortical neuroinflammation, establishing its efficacy as a potential treatment for Pb exposures.


Subject(s)
Aminosalicylic Acid , HMGB1 Protein , Rats , Animals , NF-kappa B/metabolism , Brain-Derived Neurotrophic Factor/metabolism , HMGB1 Protein/metabolism , Rats, Sprague-Dawley , Tumor Necrosis Factor-alpha/metabolism , Neuroinflammatory Diseases , Sodium , Sirtuin 1/metabolism , Lead/toxicity , Brain/metabolism , Cerebral Cortex/metabolism , Anti-Inflammatory Agents
9.
Toxicol Lett ; 375: 48-58, 2023 Feb 15.
Article in English | MEDLINE | ID: mdl-36586703

ABSTRACT

Lead (Pb) is a developmental neurotoxin that can disrupt brain development and damage the brain regions responsible for executive function, behavioral regulation and fine motor control. Sodium para-aminosalicylic acid (PAS-Na) is a non-steroidal anti-inflammatory drug that can cross the blood-brain barrier. The purpose of this study was to examine the effects of juvenile rat Pb exposure on behavioral changes and brain inflammation, and the efficacy of PAS-Na in ameliorating these effects. The results showed that Pb exposure during the juvenile period (from weaning to adult period) delayed rats' growth development and impaired their motor learning. Pb exposure not only increased Pb concentrations in several brain regions (including hippocampus, striatum and substantia nigra), but also disrupted metal-homeostasis in the brain, as higher levels of iron (Fe) and calcium (Ca) were observed in the substantia nigra. Moreover, Pb activated the MAPK pathway and increased levels of inflammatory factors such as IL-1ß, TNF-α and IL-6 in the hippocampus, striatum and substantia nigra. Furthermore, Pb increased the levels of alpha-synuclein (α-syn) in these brain sites. PAS-Na improved the motor deficits and brain inflammation in the Pb-exposed rats. Moreover, the elevated Pb, Fe and Ca concentrations in the brain were significantly reduced by PAS-Na, which contains amino, carboxyl and hydroxyl functional groups, suggesting that it may act as a chelator of brain metals. In addition, PAS-Na inhibited the Pb-induced MAPK pathway activation and α-syn accumulation in the same brain regions. Taken together, our novel study suggest that PAS-Na shows efficacy in improving the Pb-induced behavioral changes in rats by inhibiting MAPK-dependent inflammatory pathways and reducing α-syn accumulation.


Subject(s)
Aminosalicylic Acid , Encephalitis , Rats , Animals , Aminosalicylic Acid/pharmacology , Aminosalicylic Acid/therapeutic use , alpha-Synuclein , Lead/toxicity , Neuroinflammatory Diseases , Sodium , Brain , Encephalitis/chemically induced , Encephalitis/drug therapy , MAP Kinase Signaling System
10.
Biol Trace Elem Res ; 200(6): 2807-2815, 2022 Jun.
Article in English | MEDLINE | ID: mdl-34398420

ABSTRACT

Lead (Pb) is a toxic heavy metal and environmental pollutant that adversely affects the nervous system. However, effective therapeutic drugs for Pb-induced neurotoxicity have yet to be developed. In the present study, we investigated the ameliorative effect of sodium para-aminosalicylic acid (PAS-Na) on Pb-induced neurotoxicity. Male Sprague-Dawley rats were treated with (CH3COO)2 Pb•4H2O (6 mg/kg) for 4 weeks, followed by 3 weeks of PAS-Na (100, 200, and 300 mg/kg). The results showed that subacute Pb exposure significantly decreased rats body-weight gains and increased liver coefficient, and impaired spatial learning and memory. HE staining showed that Pb damaged the structure of the hippocampus. Moreover, Pb activated the ERK1/2-p90RSK/ NF-κB pathway concomitant with increased inflammatory cytokine IL-1ß levels in rat hippocampus. PAS-Na reversed the Pb-induced increase in the liver coefficient as well as the learning and memory deficits. In addition, PAS-Na reduced the phosphorylation of ERK1/2, p90RSK and NF-κB p65, decreasing IL-1ß levels in hippocampus. Our findings indicated that PAS-Na showed efficacy in reversing Pb-induced rats cognitive deficits and triggered an anti-inflammatory response. Thus, PAS-Na may be a promising therapy for treating Pb-induced neurotoxicity.


Subject(s)
Aminosalicylic Acid , Aminosalicylic Acid/pharmacology , Animals , Cognition , Lead/toxicity , MAP Kinase Signaling System , Male , Manganese/toxicity , NF-kappa B , Rats , Rats, Sprague-Dawley , Ribosomal Protein S6 Kinases, 90-kDa , Sodium , Spatial Learning
11.
J Trace Elem Med Biol ; 68: 126852, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34508950

ABSTRACT

BACKGROUND: Environmental lead (Pb) and cadmium (Cd) pollution has been considered a risk factor in the etiology of kidney stones. However, the association between Pb and Cd exposure and kidney stone incidence has yet to be determined. OBJECTIVES: This study aimed to determine a possible the association between kidney stones with Pb and Cd exposure (alone or combined) in a non-occupational population. METHODS: Pb and Cd contaminations in soil-plant system were determined by flame atomic absorption spectrophotometry. Health risk assessment of dietary Pb or Cd intake from rice and vegetables were calculated. Kidney stones were diagnosed with urinary tract ultrasonography. Urinary cadmium (UCd) and blood lead (BPb) levels were determined by graphite-furnace atomic absorption spectrometry. Multivariate logistic regression models were constructed. RESULTS: The hazard indexes (HI) of Pb and Cd were 7.91 and 7.31. The odds ratio (OR) was 2.83 (95 %CI:1.38-5.77) in males with high BPb (BPb ≥ 100 µg/L), compared with those with low BPb (BPb<100 µg/L). Compared to those with low BPb and low UCd (BPb<100 µg/L and UCd<2 µg/g creatinine), the ORs were 2.58 (95 % CI:1.17-5.70) and 3.43 (95 % CI:1.21-9.16) in females and males with high BPb and high UCd (BPb ≥100 µg/L and UCd ≥2 µg/g creatinine), respectively. The OR was 3.16 (95 % CI:1.26-7.88) in males with high BPb and low UCd (BPb ≥ 100 µg/L and UCd <2 µg/g creatinine), compared to those with low BPb and low UCd. CONCLUSIONS: Kidney stones incidence was increased by high Pb exposure in males, and by Pb and Cd co-exposure in males and females.


Subject(s)
Cadmium , Environmental Exposure , Kidney Calculi , Cadmium/toxicity , China , Creatinine , Environmental Exposure/adverse effects , Female , Humans , Kidney Calculi/chemically induced , Kidney Calculi/epidemiology , Lead , Male
12.
Biol Trace Elem Res ; 199(10): 3759-3771, 2021 Oct.
Article in English | MEDLINE | ID: mdl-33405079

ABSTRACT

Sodium p-aminosalicylic acid (PAS-Na) has been previously shown to protect the brain from manganese (Mn)-induced toxicity. However, the efficacy of PAS-Na in protecting other organs from Mn toxicity and the mechanisms associated with this protection have yet to be addressed. Therefore, here, we assessed pancreatic damage in response to Mn treatment and the efficacy of PAS-Na in limiting this effect, along with specific mechanisms that mediate PAS-Na's protection. Mn exposure led to increased blood Mn content in dose- and time-dependent manner. Furthermore, subchronic Mn exposure (20 mg/kg for 8 weeks) led to pancreatic damage in a dose-dependent manner. In addition, the elevated Mn levels increased iron and decreased zinc and magnesium content in the pancreas. These effects were noted even 8 weeks after Mn exposure cessation. Mn exposure also affected the levels of amylase, lipase, and inflammatory factors such as tumor necrosis factor (TNF-α) and interleukin-1 ß (IL-1ß). PAS-Na significantly inhibited the increase in Mn concentration in both blood and pancreas, restored Mn-induced pancreatic damage, reversed the Mn-induced alterations in metal levels, and restored amylase and lipase concentrations. Taken together, we conclude that in rats, PAS-Na shows pharmacological efficacy in protecting the pancreas from Mn-induced damage.


Subject(s)
Aminosalicylic Acid , Aminosalicylic Acid/pharmacology , Animals , Manganese/toxicity , Pancreas , Rats , Rats, Sprague-Dawley , Sodium
13.
Biol Trace Elem Res ; 199(12): 4688-4699, 2021 Dec.
Article in English | MEDLINE | ID: mdl-33447908

ABSTRACT

Exposure to high levels of manganese (Mn) leads to brain Mn accumulation, and a disease referred to as manganism. Activation of microglia plays an important role in Mn-induced neuroinflammation. Sodium p-aminosalicylic acid (PAS-Na) is a non-steroidal anti-inflammatory drug that inhibits Mn-induced neuroinflammation. The aim of the current study was to explore the role of NF-κB in the protective mechanism of PAS-Na on Mn-induced neuroinflammation in BV2 microglial experimental model. We treated BV2 microglia with 200 µM Mn for 24 h followed by 48 h treatment with graded concentrations of PAS-Na, using an NF-kB inhibitor, JSH-23, as a positive control. MTT results established that 200 and 400 µM PAS-Na treatment increased the Mn-induced cell viability reduction. NF-κB (P65) mRNA expression and the phosphorylation of p65 were increased in Mn-treated BV2 cell, and suppressed by PAS-Na, analogous to the effect of JSH-23 pretreatment. Furthermore, PAS-Na significantly reduced the contents of the inflammatory cytokine TNF-α and IL-1ß, both of which were increased by Mn treatment. The current results show that PAS-Na attenuated Mn-induced inflammation by abrogating the activation of the NF-κB signaling pathways and reduced the release of pro-inflammatory cytokines.


Subject(s)
Aminosalicylic Acid , Pharmaceutical Preparations , Aminosalicylic Acid/pharmacology , Lipopolysaccharides , Manganese/toxicity , Microglia , NF-kappa B , Sodium
14.
Biol Trace Elem Res ; 199(9): 3423-3432, 2021 Sep.
Article in English | MEDLINE | ID: mdl-33156491

ABSTRACT

BACKGROUND: Sodium p-aminosalicylic acid (PAS-Na) was reported to exhibit anti-inflammatory effect in the nervous system. However, the mechanism by which PAS-Na exhibits anti-inflammatory effects on manganese (Mn)-stimulated BV2 microglia cells remains unclear. Thus, this study investigated the role of PAS-Na in Mn-stimulated BV2 microglial cells. METHODS: Microglia-like BV2 were treated with MnCl2 with or without the non-steroidal anti-inflammatory drug PAS-Na for 12 or 24 h to examine cell viability using MTT; for 24 or 48 h to examine levels of NLRP3, CASP1, IL-1ß, and IL-18 mRNA using Real-Time quantitative PCR; for 48 h to examine levels of NLRP3 and CASP1 inflammasomes, measured by western blot analysis; and for 48 h to examine levels of inflammatory cytokines, measured by enzyme-linked immunosorbent assay. RESULTS: The MTT assay showed that PAS-Na produced significant neuroprotective effect by preventing Mn-induced inflammation in BV2 microglial cells. PAS-Na significantly concentration and time dependently inhibited Mn-induced production of NLRP3, CASP1, IL-1ß, and IL-18. CONCLUSION: Taken together, our results suggest that PAS-Na exerts anti-inflammatory effects in Mn-stimulated BV2 microglial cells via downregulation of NLRP3, CASP1, IL-1ß, and I L-18. Furthermore, a high concentration and prolonged PAS-Na treatment appear necessary for its therapeutic efficacy. Taken together, we conclude that PAS-Na affords therapeutic efficacy in mitigating neurological conditions associated with neuroinflammation.


Subject(s)
Aminosalicylic Acid , Pharmaceutical Preparations , Inflammasomes , Manganese/toxicity , Microglia , NLR Family, Pyrin Domain-Containing 3 Protein , Sodium
15.
Neurotoxicology ; 82: 177-185, 2021 01.
Article in English | MEDLINE | ID: mdl-33115663

ABSTRACT

BACKGROUND: Lead exposure is one of the most concerning public health problems worldwide, particularly among children. Yet the impact of chronic lead exposure on the thyroid status and related intelligence quotient performance among school-age children remained elusive. OBJECTIVE: The aim of this study was to evaluate the influence of lead exposure on the thyroid hormones, amino acid neurotransmitters balances, and intelligence quotient (IQ) among school-age children living nearby a lead-zinc mining site. Other factors such as rice lead levels, mothers' smoking behavior, and diet intake were also investigated. METHODS: A total of 255 children aged 7-12 years old were recruited in this study. Blood lead level (BLL), thyroid hormones including free triiodothyronine (FT3), free thyroxine (FT4) and thyroid stimulating hormone (TSH), and amino acid neurotransmitters such as glutamate (Glu), glutamine (Gln), and γ-aminobutyric acid (GABA) were measured using graphite furnace atomic absorption spectroscopy (GFAAS), chemiluminescence immunoassay, high performance liquid chromatography (HPLC). Raven's standard progressive matrices (SPM) and the questionnaire were used to determine IQ and collect related influence factors. RESULTS: The average BLL of children was 84.8 µg/L. The occurrence of lead intoxication (defined as the BLL ≥ 100 µg/L) was 31.8%. Serum TSH levels and IQ of lead-intoxicated children were significantly lower than those without lead toxicity. The GABA level of girls with the lead intoxication was higher than those with no lead-exposed group. Correlation analyses revealed that BLL were inversely associated with the serum TSH levels (R= -0.186, p < 0.05), but positively related with IQ grades (R = 0.147, p < 0.05). Moreover, BLL and Glu were inversely correlated with IQ. In addition, this study revealed four factors that may contribute to the incidence of lead intoxication among children, including the frequency of mother smoking (OR = 3.587, p < 0.05) and drinking un-boiled stagnant tap water (OR = 3.716, p < 0.05); eating fresh fruits and vegetables (OR = 0.323, p < 0.05) and soy products regularly (OR = 0.181, p < 0.05) may protect against lead intoxication. CONCLUSION: Lead exposure affects the serum TSH, GABA levels and IQ of school-aged children. Developing good living habits, improving environment, increasing the intake of high-quality protein and fresh vegetable and fruit may improve the condition of lead intoxication.


Subject(s)
Intelligence/drug effects , Lead Poisoning/complications , Lead , Mining , Thyroid Gland/drug effects , Zinc , Child , China/epidemiology , Diet, Healthy , Drinking Water/adverse effects , Female , Glutamic Acid/blood , Humans , Intelligence Tests , Lead/analysis , Lead/blood , Lead Poisoning/etiology , Male , Oryza/chemistry , Risk Factors , Thyroid Hormones/blood , Thyrotropin/blood , Tobacco Smoke Pollution/adverse effects , gamma-Aminobutyric Acid/blood
16.
J Neuroinflammation ; 17(1): 343, 2020 Nov 17.
Article in English | MEDLINE | ID: mdl-33203418

ABSTRACT

BACKGROUND: The activation of NOD-like receptor protein 3 (NLRP3) inflammasome-dependent pyroptosis has been shown to play a vital role in the pathology of manganese (Mn)-induced neurotoxicity. Sodium para-aminosalicylic acid (PAS-Na) has a positive effect on the treatment of manganism. However, the mechanism is still unclear. We hypothesized that PAS-Na might act through NLRP3. METHODS: The microglial cell line BV2 and male Sprague-Dawley rats were used to investigate the impacts of PAS-Na on Mn-induced NLRP3 inflammasome-dependent pyroptosis. The related protein of the NF-κB pathway and NLRP3-inflammasome-dependent pyroptosis was detected by western blot. The reactive oxygen species and mitochondrial membrane potential were detected by immunofluorescence staining and flow cytometry. The activation of microglia and the gasdermin D (GSDMD) were detected by immunofluorescence staining. RESULTS: Our results showed that Mn treatment induced oxidative stress and activated the NF-κB pathway by increasing the phosphorylation of p65 and IkB-α in BV2 cells and in the basal ganglia of rats. PAS-Na could alleviate Mn-induced oxidative stress damage by inhibiting ROS generation, increasing mitochondrial membrane potential and ATP levels, thereby reducing the phosphorylation of p65 and IkB-α. Besides, Mn treatment could activate the NLRP3 pathway and promote the secretion of IL-18 and IL-1ß, mediating pyroptosis in BV2 cells and in the basal ganglia and hippocampus of rats. But an inhibitor of NF-κb (JSH-23) treatment could significantly reduce LDH release, the expression of NLRP3 and Cleaved CASP1 protein and IL-1ß and IL-18 mRNA level in BV2 cells. Interestingly, the effect of PAS-Na treatment in Mn-treated BV2 cells is similar to those of JSH-23. Besides, immunofluorescence results showed that PAS-Na reduced the increase number of activated microglia, which stained positively for GSDMD. CONCLUSION: PAS-Na antagonized Mn-induced NLRP3 inflammasome dependent pyroptosis through inhibiting NF-κB pathway activation and oxidative stress.


Subject(s)
Aminosalicylic Acid/pharmacology , Manganese/toxicity , NF-kappa B/antagonists & inhibitors , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Oxidative Stress/drug effects , Pyroptosis/drug effects , Animals , Cell Line , Male , Mice , NF-kappa B/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Oxidative Stress/physiology , Pyroptosis/physiology , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Signal Transduction/physiology , Sodium/pharmacology
17.
J Trace Elem Med Biol ; 54: 134-141, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31109603

ABSTRACT

BACKGROUND: Sodium para-aminosalicylic acid (PAS-Na), an anti-tuberculosis drug, has been demonstrated its function in facilitating the Mn elimination in manganism patients and Mn-exposed models in vivo and improving the symptoms of Mn poisoning. But whether it can improve the growth retardation and inflammatory responses induced by Mn have not been reported. OBJECTIVES: This study was designed to investigate the preventive effects of PAS-Na on the development of retardation and inflammatory responses in Mn-exposed rats. METHODS: Male Sprague Dawley (SD) rats (8 weeks old, weighing 180 ± 20 g) were randomly divided into normal control group and Mn-exposed group in the 4 weeks experiment observation and normal control group, Mn-exposed group, PAS-Na preventive group and PAS-Na control group in the 8 weeks experiment observation. The Mn-exposed group received an intraperitoneal injection (i.p.) of 15 mg/kg MnCl2 and the normal control group i.p. physiological Saline in the same volume once a day for 4 or 8 weeks, 5 days per week. The PAS-Na preventive group i.p. 15 mg/kg MnCl2 along with back subcutaneous (s.c.) injection of 240 mg/kg PAS-Na once a day for 8 weeks, 5 days per week. PAS-Na control group received s.c. injection of 240 mg/kg PAS-Na along with i.p. injection of saline once daily. The body weight was determined once a week until the end of the experiment. The manganese contents in the blood were detected by graphite furnace atomic absorption spectrometry. The inflammatory factor levels (TNF-α, IL-1ß, IL-6, and PGE2) in the blood were detected by using enzyme-linked immunosorbent assay (Elisa) and each organ taking from rats were weighed and recorded. RESULTS: Mn exposure significantly suppressed the growth in rats and increased heart, liver, spleen and kidney coefficients as compared with the control group. The whole blood Mn level and serum levels of IL-1ß, IL-6, PGE2, and TNF-α in sub-chronic Mn-exposure group were markedly higher than those in the control group. However, preventive treatment with PAS-Na obviously reduced the whole blood Mn level, the spleen and liver coefficients of the Mn-exposed rats. And serum levels of IL-1ß and TNF-α were significantly reduced by 33.9% and 14.7% respectively in PAS-Na prevention group. CONCLUSIONS: PAS-Na could improve the growth retardation and alleviate inflammatory responses in Mn-exposed rats.


Subject(s)
Aminosalicylic Acid/therapeutic use , Manganese/adverse effects , Animals , Antitubercular Agents/therapeutic use , Dinoprostone/blood , Interleukin-1beta/blood , Interleukin-6/blood , Male , Manganese Poisoning/blood , Rats , Rats, Sprague-Dawley , Tumor Necrosis Factor-alpha/blood
18.
Genes Genet Syst ; 93(6): 221-228, 2019 Jan 19.
Article in English | MEDLINE | ID: mdl-30541985

ABSTRACT

Human lifespan is determined by genetic and environmental factors. Potential longevity genes are neither specific nor reproducible, and longevity-related genes are constantly confused with age-related genes. To distinguish specific age- and longevity-related genes, we analyzed a Gene Expression Omnibus (GEO) dataset established by the Leiden Longevity Study. The individuals were classified into longevity (mean age, 93.4 ± 3.0 years), longevity offspring (60.8 ± 6.1) and control (61.9 ± 6.9) groups. The series matrix files were downloaded, and average expression values were calculated. Differentially expressed genes (DEGs) between longevity and control groups and those between longevity and their offspring were identified by GEO2R online. A total of 507 longevity- and 755 age-related DEGs were visualized using a Venn diagram. Weighted gene co-expression network analysis (WGCNA) was performed on the longevity- and age-related DEGs. Age-related color modules and genes were identified. However, no longevity-related modules or genes were found. The green module, with 46 age-related DEGs, was the most biologically significant to age and aging. Gene Ontology, Kyoto Encyclopedia of Genes and Genomes, and protein-protein interaction pathway analyses were conducted on these 46 DEGs, which are mainly enriched in B cell activation and receptor signaling pathways. CR2, VPREB3, MS4A1 and CCR6 were considered the most crucial candidate genes for aging.


Subject(s)
Gene Regulatory Networks , Longevity/genetics , Transcriptome , Aged , Aged, 80 and over , Blood Cells/metabolism , Female , Gene Expression Profiling , Humans , Male , Middle Aged
19.
J Trace Elem Med Biol ; 50: 1-7, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30262264

ABSTRACT

BACKGROUND: Although manganese (Mn)-induced neurotoxicity effects are well known among occupational Mn exposure, few reports have investigated the effects on endocrine systems among welders and smelters. OBJECTIVE: To determine the effect of high level occupational manganese (Mn) exposure on neuropsychological parameters and hormonal status. METHODS: We used a cross-sectional design with 52 welders, 48 smelters and 43 age-matched office workers from the same factory in China. We analyzed serum endocrine hormones level and airborne Mn concentrations. Erythrocyte and urine Mn levels were quantified using inductively-coupled plasma atomic emission spectroscopy. RESULTS: The geometric mean of air Mn concentrations for the welders and smelters were 19.7 and 273.1 µg/m3, respectively. Mn concentrations in erythrocytes of smelters were markedly greater than those in controls and welders, but there was no difference between the erythrocytes Mn levels of Control and welders. We also found an increase of Mn levels in the urine of both welders and smelters vs. controls; Mn levels in urine of smelters were higher than in welders. Self-reported neurobehavioral symptoms were higher in welders and smelters than in controls. Finally, thyroid-stimulating hormone (TSH) levels of welders were significantly lower than in controls, whereas smelters had lower prolactin (PRL), testosterone (TST) and follicle-stimulating hormone (FSH) concentrations than either controls or welders. CONCLUSIONS: These results show that smelters have higher Mn exposure than do welders, and that Mn levels in erythrocytes or urine can be a marker for exposure. Moreover, high level occupational Mn exposure increases adverse neurobehavioral effects, and also may disrupt endocrine systems.


Subject(s)
Manganese/blood , Manganese/urine , China , Cross-Sectional Studies , Erythrocytes/metabolism , Female , Humans , Male , Manganese Poisoning/blood , Occupational Exposure , Prolactin/blood , Prolactin/urine , Spectrophotometry, Atomic , Testosterone/blood , Testosterone/urine , Thyrotropin/blood , Thyrotropin/urine , Welding
20.
J Toxicol Sci ; 41(5): 573-81, 2016.
Article in English | MEDLINE | ID: mdl-27665767

ABSTRACT

Sodium para-aminosalicylate (PAS-Na) was first applied successfully in clinical treatment of two manganism patients with good prognosis. However, the mechanism of how PAS-Na protects against Mn-induced neurotoxicity is still elusive. The current study was conducted to explore the effects of PAS-Na on Mn-induced basal ganglia astrocyte injury, and the involvement of amino acid neurotransmitter in vitro. Basal ganglia astrocytes were exposed to 500 µM manganese chloride (MnCl2) for 24 hr, following by 50, 150, or 450 µM PAS-Na treatment for another 24 hr. MnCl2 significantly decreased viability of astrocytes and induced DNA damages via increasing the percentage of tail DNA and Olive tail moment of DNA. Moreover, Mn interrupted amino acid neurotransmitters by decreasing Gln levels and increasing Glu, Gly levels. In contrast, PAS-Na treatment reversed the aforementioned Mn-induced toxic effects on basal ganglia astrocytes. Taken together, our results demonstrated that excessive Mn exposure may induce toxic effects on basal ganglia astrocytes, while PAS-Na could protect basal ganglia astrocytes from Mn-induced neurotoxicity.


Subject(s)
Aminosalicylic Acid/pharmacology , Astrocytes/drug effects , Basal Ganglia/drug effects , Chlorides/toxicity , DNA Damage/drug effects , Glutamic Acid/metabolism , Glutamine/metabolism , Glycine/metabolism , Manganese Poisoning/prevention & control , Protective Agents/pharmacology , Animals , Animals, Newborn , Astrocytes/metabolism , Astrocytes/pathology , Basal Ganglia/metabolism , Basal Ganglia/pathology , Cells, Cultured , Cytoprotection , Dose-Response Relationship, Drug , Manganese Compounds , Manganese Poisoning/genetics , Manganese Poisoning/metabolism , Manganese Poisoning/pathology , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL