Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 13 de 13
1.
Environ Res ; 243: 117875, 2024 Feb 15.
Article En | MEDLINE | ID: mdl-38072110

Arsenic (As) and lead (Pb) are toxins found in the natural surroundings, and the harmful health outcomes caused by the co-exposure of such toxins have become a considerable problem. However, the joint neurotoxicity of As and Pb to neurodevelopment and the underlying mechanisms remain unclear. Pluripotent stem cell-derived human brain organoids are emerging animal model alternatives for understanding neurological-related diseases. Therefore, we utilized brain organoids with optic vesicles (OVB-organoids) to systematically analyze the neurotoxicity of As and Pb. After 24 h of As and/or Pb exposure, hematoxylin-eosin staining revealed that As and Pb exposure could cause disorders in the structure of the ventricular zone and general cell disarrangement in OVB-organoids. Immunostaining displayed that OVB-organoids are more susceptible to As and Pb co-exposure than independent exposure in apoptosis, proliferation, and cell differentiation. Meanwhile, even though As and Pb could both hinder cell proliferation, contrary to Pb, As could induce an increasing proportion of mitotic (G2/M) cells. The proteome landscape of OVB-organoids illustrated that Pb synergized with As in G2/M arrest and the common role of As and Pb in carcinogenesis. Besides, proteomics analyses suggested the consequential role of autophagy and Wnt pathway in the neurotoxicity of As and Pb co-exposure. Overall, our findings provide penetrating insights into the cell cycle, carcinogenesis, autophagy, and Wnt pathway underlying the As and Pb binary exposure scenarios, which could enhance our understanding of the mixture neurotoxicity mechanisms.


Arsenic , Animals , Humans , Arsenic/toxicity , Arsenic/metabolism , Proteome/metabolism , Lead/toxicity , Lead/metabolism , Apoptosis , Cell Line, Tumor , G2 Phase Cell Cycle Checkpoints , Brain/metabolism , Organoids/metabolism , Carcinogenesis/metabolism
2.
Biosens Bioelectron ; 240: 115635, 2023 Nov 15.
Article En | MEDLINE | ID: mdl-37651948

Human cerebral organoids (COs), generated from stem cells, are emerging animal alternatives for understanding brain development and neurodegeneration diseases. Long-term growth of COs is currently hindered by the limitation of efficient oxygen infiltration and continuous nutrient supply, leading to general inner hypoxia and cell death at the core region of the organoids. Here, we developed a three-dimensional (3D) microfluidic platform with dynamic fluidic perturbation and oxygen supply. We demonstrated COs cultured in the 3D microfluidic system grew continuously for over 50 days without cell death at the core region. Increased cell proliferation and enhanced cell differentiation were also observed and verified with immunofluorescence staining, proteomics and metabolomics. Time-lapse proteomics from 7 consecutive acquisitions between day 4 and day 30 identified 546 proteins differently expressed accompanying COs growth, which were mainly relevant to nervous system development, in utero embryonic development, brain development and neuron migration. Our 3D microfluidic platform provides potential utility for culturing high-homogeneous human organoids.


Biosensing Techniques , Microfluidics , Animals , Female , Pregnancy , Humans , Cell Death , Organoids , Oxygen
3.
Environ Toxicol ; 37(7): 1790-1802, 2022 Jul.
Article En | MEDLINE | ID: mdl-35385208

Perfluorotetradecanoic acid (PFTeDA) is a long-chain perfluoroalkyl compound with increased applications. Its effect on Leydig cell function and its underlying mechanism remain unclear. Male Sprague-Dawley rats (60 days old) were gavaged with PFTeDA at doses of 0, 1, 5, and 10 mg/kg/day from 60 to 87 days after birth. PFTeDA significantly reduced serum testosterone levels at 1 mg/kg and higher doses, while markedly increasing serum luteinizing hormone level at 10 mg/kg and follicle-stimulating hormone at ≥1 mg/kg. PFTeDA significantly reduced the sperm number at the cauda of epididymis at ≥1 mg/kg. PFTeDA also reduced the number of CYP11A1-positive Leydig cells due to increased apoptosis shown by the terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) assay. PFTeDA significantly repressed the expression of Cyp17a1 and Star and their proteins at 1-10 mg/kg, while it increased the expression of Srd5a1 and its protein (an immature Leydig cell biomarker) at 10 mg/kg. PFTeDA markedly increased testicular malondialdehyde level, while inhibiting antioxidants (SOD1, SOD2, and CAT), triggering oxidative stress, thereby further inducing BAX and CASP3 while inhibiting BCL2, which led to cell apoptosis. PFTeDA also reduced DHH level secreted by Sertoli cells, which indirectly affected Leydig cell function. PFTeDA inhibited testosterone secretion in primary Leydig cells in vitro by increasing reactive oxygen species and inducing apoptosis at 50 µM. In conclusion, PFTeDA inhibits the function of Leydig cells by inducing oxidative stress and subsequently stimulating cell apoptosis.


Apoptosis , Fluorocarbons , Leydig Cells , Oxidative Stress , Animals , Fluorocarbons/adverse effects , Male , Rats , Rats, Sprague-Dawley , Testis , Testosterone/blood
4.
Environ Toxicol ; 37(5): 1160-1172, 2022 May.
Article En | MEDLINE | ID: mdl-35102696

Cypermethrin is a broad-spectrum pyrethroid insecticide that is widely used. It may induce adverse endocrine-disrupting effects on the male reproductive system. Whether cypermethrin can disrupt Leydig cell development and function in the late puberty remains elusive. The objective of this study was to explore the effect of cypermethrin exposure to male rats on the development and function of Leydig cells in late puberty and explore the underlying mechanism. Thirty-six male Sprague-Dawley rats (age of 35 days) were gavaged with cypermethrin (0, 12.5, 25, and 50 mg/kg/day) from postnatal day 35-49. Cypermethrin significantly lowered serum testosterone level while elevating serum luteinizing hormone level at a dose of 50 mg/kg, without altering serum follicle-stimulating hormone level. Cypermethrin markedly decreased CYP11A1-positive Leydig cell number at 50 mg/kg without affecting SOX9-positive Sertoli cell number. It significantly down-regulated the expression of Leydig cell genes, Lhcgr, Star, Cyp11a1, and Cyp17a1 and their proteins, while up-regulating the expression of Sertoli cell genes, Dhh and Amh, and their proteins, at doses of 12.5-50 mg/kg. In addition, cypermethrin significantly increased malondialdehyde level while lowering the expression of Sod1 and Sod2 and their proteins at 50 mg/kg. Cypermethrin markedly induced reactive oxidative species at a concentration of 200 µM and reduced mitochondrial membrane potential at 25 µM and higher concentrations after 24 h of treatment to primary Leydig cells in vitro. In conclusion, cypermethrin inhibits the development and function of Leydig cells in male rats in late puberty.


Leydig Cells , Pyrethrins , Animals , Male , Pyrethrins/toxicity , Rats , Rats, Sprague-Dawley , Sexual Maturation , Testis , Testosterone
5.
Toxicol Appl Pharmacol ; 439: 115903, 2022 03 15.
Article En | MEDLINE | ID: mdl-35143807

Perfluoroundecanoic acid (PFUnA), a perfluorinated compound, has environmental persistence, bioaccumulation, and potential toxicity. However, its effect on Leydig cell function remains unclear. Rats (age of 56 days) were gavaged with 0 (corn oil), 0.1, 0.5, 1, or 5 mg/kg/day PFUnA for 28 days. PFUnA significantly reduced serum testosterone levels as low as 0.5 mg/kg. PFUnA markedly decreased Leydig cell number as low as 0.1 mg/kg. PFUnA markedly reduced transcript levels of Star and Insl3 in the testes at 1 mg/kg after adjusting to Leydig cell number. It also reduced their protein levels. PFUnA significantly decreased the phosphorylation of AKT1 and mTOR as low as 0.1 mg/kg and the phosphorylation of ERK1/2 at 1 mg/kg and the phosphorylation of AKT1, AKT2, ERK1/2, and mTOR in Leydig cells at various concentrations (0.01-10 µM) after 24 h of in vitro treatment. In conclusion, PFUnA inhibits Leydig cell function possibly via AKT/ERK1/2/mTOR signaling pathways.


Fluorocarbons , Leydig Cells , Animals , Fatty Acids , Fluorocarbons/toxicity , Male , Rats , Rats, Sprague-Dawley , TOR Serine-Threonine Kinases/metabolism , Testis/metabolism , Testosterone
6.
Ecotoxicol Environ Saf ; 232: 113282, 2022 Mar 01.
Article En | MEDLINE | ID: mdl-35131586

Methyl tert-butyl ether (MTBE) is a widely used gasoline additive. It is considered an endocrine-disrupting chemical. Whether MTBE affects the development of Leydig cells in late puberty of males and its underlying mechanism remains unclear. Twenty-four male Sprague-Dawley rats (35 days old) were randomly allocated into four groups and were orally given MTBE (0, 300, 600, and 1200 mg/kg/day) from postnatal day (PND) 35-56. MTBE markedly reduced serum testosterone levels at 300 mg/kg and higher doses without altering the serum levels of luteinizing hormone and follicle-stimulating hormone. It mainly inhibited cell proliferation, induced mitochondrial autophagy and apoptosis, and indirectly stimulated Sertoli cells to secrete anti-Müllerian hormones, thereby significantly reducing the number of Leydig cells at 1200 mg/kg. MTBE also markedly down-regulated the expression of mature Leydig cell biomarker Cyp11a1 and Hsd3b1 and their proteins, while up-regulating the expression of immature Leydig cell biomarker Akr1c14 and its protein at 600 mg/kg and higher. MTBE significantly down-regulated the expression of cell cycle gene Ccnd1, antioxidant gene Gpx1, and anti-apoptotic gene Bcl2, while increasing pro-apoptotic gene Bax level at 1200 mg/kg. In vitro study further confirmed that MTBE can inhibit testosterone synthesis by inducing reactive oxygen species (ROS) generation, mitophagy, and apoptosis at 200 and 300 mM. In conclusion, exposure to MTBE compromises the development of Leydig cells in late puberty in male rats.


Leydig Cells , Testosterone , Animals , Apoptosis , Leydig Cells/metabolism , Male , Methyl Ethers , Mitophagy , Rats , Rats, Sprague-Dawley
7.
Chem Res Toxicol ; 34(6): 1542-1555, 2021 06 21.
Article En | MEDLINE | ID: mdl-34081457

Perfluorotridecanoic acid (PFTrDA) is a long-chain (C13) perfluoroalkyl carboxylic acid. Here, we report the influence of PFTrDA exposure on the maturation of rat Leydig cells in late puberty in vivo. Male Sprague-Dawley rats were administered PFTrDA by gavage of 0, 1, 5, and 10 mg/kg/day from 35 days to 56 days postpartum. PFTrDA had no effect on body weight, testis weight, and epididymis weight. It significantly decreased the serum testosterone level after 5 and 10 mg/kg exposure, while it did not alter the serum estradiol level. The serum luteinizing hormone level was markedly reduced after 10 mg/kg PFTrDA exposure, while the follicle-stimulating hormone level was unchanged. Star, Cyp11a1, Cyp17a1, Hsd3b1, and Insl3 transcript levels in the testis were markedly lowered in the 1-5 mg/kg PFTrDA group and the Lhb transcript level in the pituitary in the 10 mg/kg group. CYP11A1 and HSD11B1-positive Leydig cell numbers were markedly reduced after 10 mg/kg PFTrDA exposure. Testicular triglyceride and free fatty acid (palmitic acid, oleic acid, and linoleic acid) levels were significantly reduced by PFTrDA, while Mgll (up-regulation) and Scarb1 and Elovl5 (down-regulation) expression were altered. AKT1 and AMPK phosphorylation was stimulated after 10 PFTrDA mg/kg exposure. In conclusion, PFTrDA delays the maturation of Leydig cells in late puberty mainly by altering the free fatty acid profile.


Decanoic Acids/pharmacology , Fluorocarbons/pharmacology , Leydig Cells/drug effects , Lipids/analysis , Pituitary Gland/drug effects , Testis/drug effects , Administration, Oral , Animals , Decanoic Acids/administration & dosage , Dose-Response Relationship, Drug , Fluorocarbons/administration & dosage , Male , Pituitary Gland/pathology , Rats , Rats, Sprague-Dawley , Testis/pathology
8.
Food Chem Toxicol ; 153: 112248, 2021 Jul.
Article En | MEDLINE | ID: mdl-33940105

Bisphenol B (BPB) has been used as a substitute for bisphenol A (BPA) in plastic materials. Whether BPB disrupts the male reproductive system remains unknown. Here, we report the effect of BPB on Leydig cell maturation in late puberty. Male Sprague-Dawley (35 days old) rats were gavaged with BPB at 0, 10, 100, and 200 mg/kg/day for 21 days. BPB significantly reduced body and epididymis weight at 200 mg/kg. BPB markedly decreased serum testosterone levels at 100 and 200 mg/kg and serum luteinizing hormone and follicle-stimulating hormone levels at 200 mg/kg. BPB significantly increased Leydig cell number at 100 and 200 mg/kg, while down-regulating the expression of Leydig cell genes (Cyp11a1 and Hsd3b1) at ≥100 mg/kg and up-regulating the expression of Sertoli cell genes (Pdgfra, Fshr, Sox9) and cell cycle regulators (Pcna, Ccnb1, Cdk2, and Cdk4) at 10-200 mg/kg. BPB markedly increased the phosphorylation of AKT1, AKT2, and ERK1/2 at 200 mg/kg. BPB increased the proliferation of rat immature Leydig cells via promoting the S/M2 phase shift at 100 and 1000 nM after 24-h culture in vitro. In conclusion, BPB disrupts Leydig cell maturation in late puberty by increasing Leydig cell number while inhibiting its maturation.


Benzhydryl Compounds/toxicity , Cell Proliferation/drug effects , Estrogens, Non-Steroidal/toxicity , Leydig Cells/drug effects , Phenols/toxicity , Sexual Maturation/drug effects , Animals , Cell Cycle/drug effects , Cell Cycle/physiology , Down-Regulation , Follicle Stimulating Hormone/metabolism , Gonadal Steroid Hormones/genetics , Gonadal Steroid Hormones/metabolism , Luteinizing Hormone/metabolism , Male , Rats , Testosterone/metabolism
9.
Environ Toxicol ; 36(6): 1206-1216, 2021 Jun.
Article En | MEDLINE | ID: mdl-33683001

Perfluorotridecanoic acid (PFTrDA) is a long-chain perfluoroalkyl substance, and its effect on the differentiation of fetal Leydig cells remains unclear. The objective of this study is to explore the effect of in utero PFTrDA exposure on the differentiation of fetal Leydig cells and investigate its underlying mechanisms. Pregnant Sprague-Dawley female rats were daily administered by gavage of PFTrDA at doses of 0, 1, 5, and 10 mg/kg from gestational day 14 to 21. PFTrDA had no effect on the body weight of dams, but significantly reduced the body weight and anogenital distance of male pups at birth at a dose of 10 mg/kg. PFTrDA significantly decreased serum testosterone levels as low as 1 mg/kg. PFTrDA did not affect fetal Leydig cell number, but promoted abnormal aggregation of fetal Leydig cells at doses of 5 and 10 mg/kg. PFTrDA down-regulated the expression of Insl3, Lhcgr, Scarb1, Star, Hsd3b1, Cyp17a1, Nr5a1, and Dhh as well as their proteins. PFTrDA lowered the levels of antioxidants (SOD1, CAT, and GPX1), induced autophagy as shown by increased levels of LC3II and beclin1, and reduced the phosphorylation of mTOR. In conclusion, PFTrDA inhibits the differentiation of fetal Leydig cells in male pups after in utero exposure mainly through increasing oxidative stress and inducing autophagy.


Testis , Testosterone , Animals , Autophagy , Cell Differentiation , Female , Leydig Cells/metabolism , Male , Oxidative Stress , Pregnancy , Rats , Rats, Sprague-Dawley , Testis/metabolism , Testosterone/metabolism
10.
Toxicol Lett ; 342: 6-19, 2021 May 15.
Article En | MEDLINE | ID: mdl-33581290

2,2',4,4'-Tetrabromodiphenyl ether (BDE47), a flame retardant, is extensively distributed in the food chain. However, whether BDE47 affects Leydig cell development during prepuberty remains unclear. BDE47 was daily gavaged to 21-day-old Sprague-Dawley male rats with 0 (corn oil), 0.1, 0.2, and 0.4 mg/kg for 14 days. BDE47 did not affect the body weight or testis weight of rats. It significantly increased serum testosterone level at 0.4 mg/kg, but decreased luteinizing hormone (LH) level without affecting estradiol level. BDE47 induced Leydig cell hyperplasia (the number of CYP11A1-positive Leydig cells increased), and up-regulated the expression of Scarb1, Star, Hsd11b1, Pcna, and Ccnd1 in the testis. BDE47 significantly reduced p53 and p21 levels but increased CCND1 level. It also markedly increased the phosphorylation of AKT1, AKT2, ERK1/2, and CREB. BDE47 significantly up-regulated the expression of Scarb1, Star, and Hsd11b1 and stimulated androgen production by immature Leydig cells from rats under basal, LH, and 8Br-cAMP stimulated conditions at 100 nM in vitro. In conclusion, BDE47 increased Leydig cell number and up-regulated the expression of Scarb1 and Star, thereby leading to increased testosterone synthesis.


Halogenated Diphenyl Ethers/toxicity , Leydig Cells/physiology , Sexual Maturation/physiology , Stem Cells/drug effects , 11-beta-Hydroxysteroid Dehydrogenase Type 1/genetics , 11-beta-Hydroxysteroid Dehydrogenase Type 1/metabolism , Animals , Cells, Cultured , Cholesterol Side-Chain Cleavage Enzyme/genetics , Cholesterol Side-Chain Cleavage Enzyme/metabolism , Dose-Response Relationship, Drug , Follicle Stimulating Hormone , Gene Expression Regulation/drug effects , Halogenated Diphenyl Ethers/administration & dosage , Halogenated Diphenyl Ethers/chemistry , Luteinizing Hormone , Male , Molecular Structure , Proliferating Cell Nuclear Antigen/genetics , Proliferating Cell Nuclear Antigen/metabolism , Rats , Rats, Sprague-Dawley , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism
11.
Toxicol Lett ; 342: 38-49, 2021 May 15.
Article En | MEDLINE | ID: mdl-33582287

4-Bromodiphenyl ether (BDE3) is a photodegradation product of higher polybrominated diphenyl ether flame retardants and is known as an endocrine disruptor. However, it is unclear whether and how BDE3 affects the development of fetal testes. This study aimed to investigate the effect of in utero exposure to BDE3 on fetal testicular development in rats. From gestational day (GD) 12-21, BDE3 (0, 50, 100, and 200 mg/kg) was daily gavaged to female pregnant Sprague Dawley rats. BDE3 significantly reduced serum testosterone levels of male pups starting at 50 mg/kg. BDE3 reduced fetal Leydig cell number at a dose of 200 mg/kg without affecting fetal Leydig cell cluster frequency and Sertoli cell number. In addition, BDE3 down-regulated the expression of fetal Leydig cell genes (Cyp11a1, Hsd3b1, Cyp17a1, and Hsd17b3) and their proteins at 100 and/or 200 mg/kg. RNA-seq analysis showed that genes responsive to cAMP (Ass1, Gpd1, Rpl13a) were down-regulated and hypoxia-related genes (Egln3 and P4ha1) were up-regulated at 200 mg/kg. In utero exposure to BDE3 can promote autophagy and apoptosis of fetal Leydig cells via increasing the levels of Beclin1, LC3-II, BAX, and by decreasing the levels of p62 and BCL2. In conclusion, in utero exposure to BDE3 blocks the development of fetal rat testes.


Fetal Development/drug effects , Halogenated Diphenyl Ethers/toxicity , Testis/drug effects , Testis/embryology , Animals , Apoptosis/drug effects , Autophagy/drug effects , Dose-Response Relationship, Drug , Female , Fetus/drug effects , Gene Expression Regulation, Developmental/drug effects , Halogenated Diphenyl Ethers/administration & dosage , Male , Pregnancy , Rats , Rats, Sprague-Dawley , Sequence Analysis, RNA , Signal Transduction/drug effects , Transcriptome
12.
Toxicol Appl Pharmacol ; 415: 115440, 2021 03 15.
Article En | MEDLINE | ID: mdl-33549592

Perfluoroundecanoic acid (PFUnA) is one of long-chain perfluoroalkyl carboxylic acids. However, the effect of PFUnA on pubertal development of Leydig cells remains unclear. The goal of this study was to investigate the effect of PFUnA on Leydig cell development in pubertal male rats. We orally dosed male Sprague-Dawley rats (age 35 days) with PFUnA at doses of 0, 1, 5, and 10 mg/kg/day from postnatal day (PND) 35 to PND 56. Serum testosterone and luteinizing hormone levels were remarkably reduced by PFUnA at ≥1 mg/kg while serum follicle-stimulating hormone levels were lowered at 5 and 10 mg/kg. PFUnA down-regulated the expression of Lhcgr, Scarb1, Star, Cyp11a1, Hsd3b1, Cyp17a1, Hsd17b3, Hsd11b1, Insl3, Nr5a1, Fshr, Dhh, Sod1, and Sod2 and their proteins in the testis and the expression of Lhb and Fshb in the pituitary. PFUnA reduced Leydig cell number at 5 and 10 mg/kg. PFUnA induced oxidative stress and increased autophagy. These may result from the inhibition of phosphorylation of mTOR, AKT1, AKT2, and ERK1/2 in the testis. In conclusion, PFUnA exhibits inhibitory effects on pubertal Leydig cell development possibly via inducing oxidative stress and increasing autophagy.


Autophagy/drug effects , Fatty Acids/toxicity , Fluorocarbons/toxicity , Leydig Cells/drug effects , Oxidative Stress/drug effects , Age Factors , Animals , Autophagy-Related Proteins/metabolism , Follicle Stimulating Hormone/blood , Gene Expression Regulation, Enzymologic , Leydig Cells/metabolism , Leydig Cells/pathology , Luteinizing Hormone/blood , Male , Pituitary Gland/drug effects , Pituitary Gland/metabolism , Rats, Sprague-Dawley , Sexual Development , Signal Transduction , Sperm Count , Spermatozoa/drug effects , Spermatozoa/pathology , Testosterone/blood
13.
Toxicology ; 448: 152633, 2021 01 30.
Article En | MEDLINE | ID: mdl-33220336

Perfluoroheptanoic acid (PFHpA) is a short-chain alternative to long-chain perfluoroalkyl substances, which have been reported to possess reproductive toxicity. However, it is unclear whether PFHpA affects Leydig cell development during puberty. The 35-day-old Sprague Dawley male rats were exposed to PFHpA by gavage with 0 (corn oil), 10, 50, and 100 mg/kg/day for 21 days. PFHpA did not affect the body weight of rats, but it reduced testis weight, relative testis weight, and epididymis weight at 100 mg/kg. It significantly increased serum testosterone, luteinizing hormone, and follicle-stimulating hormone levels at a dose of 100 mg/kg without affecting serum estradiol levels. PFHpA suppressed sperm production at a dose of 100 mg/kg. PFHpA induced Leydig cell hyperplasia (increased number of CYP11A1-positive Leydig cells) at a dose of 100 mg/kg, but down-regulated the expression of Cyp11a1, Hsd3b1, and Cyp17a1 in individual Leydig cell pe se and up-regulated the expression of Fshr in the Sertoli cell pe se. PFHpA did not affect the number of HSD11B1 (a biomarker for more mature Leydig cells) positive Leydig cells and SOX9 positive Sertoli cells. PFHpA increased BCL2, and the phosphorylation of AKT1, AKT2, ERK1/2, and JNK, but decreased BAX levels. However, it had no effect on SIRT1 and PGC-1α levels. In conclusion, PFHpA induces Leydig cell hyperplasia due to the increase in the secretion of luteinizing hormone through negative feedback after down-regulating the expression of steroidogenic enzymes and inhibiting testosterone production in individual Leydig cells. This proliferation may be mediated by increasing BCL2 and phosphorylation of AKT, ERK1/2, and JNK, and decreasing BAX level.


Fluorocarbons/toxicity , Heptanoic Acids/toxicity , Leydig Cells/drug effects , Leydig Cells/pathology , Sexual Maturation/drug effects , Spermatogenesis/drug effects , Age Factors , Animals , Dose-Response Relationship, Drug , Hyperplasia/chemically induced , Hyperplasia/pathology , Male , Rats , Rats, Sprague-Dawley , Sexual Maturation/physiology , Spermatogenesis/physiology
...