Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Biol Open ; 13(5)2024 May 15.
Article in English | MEDLINE | ID: mdl-38682287

ABSTRACT

Pex23 family proteins localize to the endoplasmic reticulum and play a role in peroxisome and lipid body formation. The yeast Hansenula polymorpha contains four members: Pex23, Pex24, Pex29 and Pex32. We previously showed that loss of Pex24 or Pex32 results in severe peroxisomal defects, caused by reduced peroxisome-endoplasmic reticulum contact sites. We now analyzed the effect of the absence of all four Pex23 family proteins on other cell organelles. Vacuoles were normal in all four deletion strains. The number of lipid droplets was reduced in pex23 and pex29, but not in pex24 and pex32 cells, indicating that peroxisome and lipid droplet formation require different Pex23 family proteins in H. polymorpha. In pex23 and pex29 cells mitochondria were fragmented and clustered accompanied by reduced levels of the fusion protein Fzo1. Deletion of DNM1 suppressed the morphological phenotype of pex23 and pex29 cells, suggesting that mitochondrial fusion is affected. pex23 and pex29 cells showed retarded growth and reduced mitochondrial activities. The growth defect was partially suppressed by DNM1 deletion as well as by an artificial mitochondrion-endoplasmic reticulum tether. Hence, the absence of Pex23 family proteins may influence mitochondrion-endoplasmic reticulum contact sites.


Subject(s)
Mitochondria , Peroxins , Pichia , Endoplasmic Reticulum/metabolism , Fungal Proteins/metabolism , Fungal Proteins/genetics , Gene Deletion , Mitochondria/pathology , Peroxins/metabolism , Peroxins/genetics , Peroxisomes/metabolism , Phenotype , Pichia/cytology , Pichia/genetics , Pichia/metabolism , Vacuoles/metabolism
2.
Environ Toxicol ; 38(5): 997-1010, 2023 May.
Article in English | MEDLINE | ID: mdl-36715143

ABSTRACT

Di-n-pentyl phthalate (DPeP) is an endocrine-disrupting phthalate plasticizer. The objective of this study was to investigate the effect of DPeP on adrenocortical function in adult male rats following in utero exposure. DPeP (0, 10, 50, 100, and 500 mg/kg/day) was administered by gavage to pregnant Sprague-Dawley rats from gestational day 14 to 21. The morphology and function of the adrenal cortex in 56-day-old male offspring were studied. DPeP at 100 and 500 mg/kg/day significantly reduced serum aldosterone levels and at 500 mg/kg/day markedly reduced corticosterone and adrenocorticotropic hormone levels. DPeP at 10-500 mg/kg markedly reduced the thickness of zona glomerulosa without affecting the thickness of zona fasciculata. DPeP significantly downregulated the expression of Agtr1a, Mc2r, Scarb1, Cyp11a1, Hsd3b1, Cyp21, Cyp11b1, Cyp11b2, Nr5a1, Nr4a2, and Bcl2 genes as well as their proteins. DPeP at 500 mg/kg/day significantly increased phosphorylated AMPK, while DPeP at 100 mg/kg/day and higher doses reduced phosphorylated AKT1 and total SIRT1 level. DPeP at 100 and 500 µM markedly induced reactive oxygen species and apoptosis in H295R cells after 24 h of culture. In conclusion, in utero exposure to DPeP disrupts adrenocortical function of the adult male offspring by (1) increasing AMPK phosphorylation and decreasing AKT1 phosphorylation and SIRT1 levels, (2) reducing adrenocorticotropic hormone levels, and (3) possibly inducing oxidative stress and apoptosis.


Subject(s)
AMP-Activated Protein Kinases , Adrenal Cortex , Pregnancy , Female , Rats , Animals , Male , Rats, Sprague-Dawley , AMP-Activated Protein Kinases/metabolism , Phosphorylation , Sirtuin 1/metabolism , Adrenal Cortex/metabolism , Adrenocorticotropic Hormone/metabolism
3.
Environ Toxicol ; 37(10): 2419-2433, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35762508

ABSTRACT

Phthalates may interfere with the biosynthesis of steroid hormones in the adrenal cortex. Bis (2-butoxyethyl) phthalate (BBOP) is a phthalate containing oxygen atoms in the alcohol moiety. In this study, 35-day-old male Sprague-Dawley rats were daily gavaged with BBOP (0, 10, 100, 250, and 500 mg/kg body weight) for 21 days. BBOP did not affect the weight of body and adrenal glands. BBOP significantly reduced serum corticosterone levels at 250 and 500 mg/kg, and lowered aldosterone level at 500 mg/kg without affecting adrenocorticotropic hormone. BBOP did not alter the thickness of the adrenal cortex. BBOP significantly down-regulated the expression of steroidogenesis-related genes (Scarb1, Star, Cyp11a1, Cyp21, Cyp11b1, Cyp11b2, Nr5a1, Nr4a1, and Nr4a2) and proteins, and antioxidant enzymes (Sod1, Sod2, Gpx1, and Cat) and their proteins, while up-regulating the expression of Mc2r and Agtr1a at various doses. BBOP reduced the phosphorylation of AKT1, AKT2, and ERK1/2, as well as the levels of SIRT1 and PGC1α without affecting the phosphorylation of AMPK. BBOP significantly induced the production of reactive oxygen species and apoptosis rate in H295R cells at 100 µM and higher after 24 h of treatment. In conclusion, male rats exposed to BBOP in puberty have significant reduction of steroid biosynthesis with a potential mechanism that is involved in the decrease in the phosphorylation of AKT1, AKT2, ERK1/2, as well as SIRT1 and PGC1α and increase in ROS.


Subject(s)
Sexual Maturation , Sirtuin 1 , Animals , Corticosterone/metabolism , Male , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Phthalic Acids , Rats , Rats, Sprague-Dawley , Sirtuin 1/metabolism , Steroidogenic Factor 1/metabolism , Steroids
4.
Toxicol Appl Pharmacol ; 439: 115903, 2022 03 15.
Article in English | MEDLINE | ID: mdl-35143807

ABSTRACT

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.


Subject(s)
Fluorocarbons , Leydig Cells , Animals , Fatty Acids , Fluorocarbons/toxicity , Male , Rats , Rats, Sprague-Dawley , TOR Serine-Threonine Kinases/metabolism , Testis/metabolism , Testosterone
5.
Ecotoxicol Environ Saf ; 232: 113282, 2022 Mar 01.
Article in English | MEDLINE | ID: mdl-35131586

ABSTRACT

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.


Subject(s)
Leydig Cells , Testosterone , Animals , Apoptosis , Leydig Cells/metabolism , Male , Methyl Ethers , Mitophagy , Rats , Rats, Sprague-Dawley
6.
Environ Toxicol ; 37(5): 1160-1172, 2022 May.
Article in English | MEDLINE | ID: mdl-35102696

ABSTRACT

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.


Subject(s)
Leydig Cells , Pyrethrins , Animals , Male , Pyrethrins/toxicity , Rats , Rats, Sprague-Dawley , Sexual Maturation , Testis , Testosterone
7.
Environ Pollut ; 298: 118825, 2022 Apr 01.
Article in English | MEDLINE | ID: mdl-35026324

ABSTRACT

Bisphenol A (BPA) is a ubiquitous environmental pollutant, mainly from the manufacture and use of plastics. The use of BPA is restricted, and its new analogs (including bisphenol AF, BPAF) are being produced to replace it. However, the effect of BPAF on the male reproductive system remains unclear. Here, we report the effect of BPAF on Leydig cell regeneration in rats. Leydig cells were eliminated by ethane dimethane sulfonate (EDS, i.p., 75 mg/kg) and the regeneration began 14 days after its treatment. We gavaged 0, 10, 100, and 200 mg/kg BPAF to rats on post-EDS day 7-28. BPAF significantly reduced serum testosterone and progesterone levels at ≧10 mg/kg. It markedly reduced serum levels of estradiol, luteinizing hormone, and follicle-stimulating hormone at 100 and 200 mg/kg. BPAF significantly reduced Leydig cell number at 200 mg/kg. BPAF significantly down-regulated the expression of Cyp17a1 at doses of 10 mg/kg and higher and the expression of Insl3, Star, Hsd17b3, Hsd11b1 in Leydig cells at 100 and 200 mg/kg, while it induced a significant up-regulation of Fshr, Dhh, and Sox9 in Sertoli cells at 200 mg/kg. BPAF induced oxidative stress and reduced the level of SOD2 at 200 mg/kg. It induced apoptosis and autophagy by increasing the levels of BAX, LC3B, and BECLIN1 and lowering the levels of BCL2 and p62 at 100 and 200 mg/kg. It induced autophagy possibly via decreasing the phosphorylation of AKT1 and mTOR. BPAF also significantly induced ROS production and apoptosis at a concentration of 10 µM, and reduced testosterone synthesis in rat R2C Leydig cells at a concentration of 10 µM in vitro, but did not affect cell viability after 24 h of treatment. In conclusion, BPAF is a novel endocrine disruptor, inhibiting the regeneration of Leydig cells.


Subject(s)
Leydig Cells , Testosterone , Animals , Benzhydryl Compounds/toxicity , Cell Proliferation , Fluorocarbons , Male , Phenols , Rats , Stem Cells
8.
Ecotoxicol Environ Saf ; 228: 112942, 2021 Oct 30.
Article in English | MEDLINE | ID: mdl-34737156

ABSTRACT

Triadimefon is a broad-spectrum fungicide widely applied in the agriculture. It is believed to be an endocrine disruptor. Whether triadimefon can inhibit the development of fetal Leydig cells and the underlying mechanisms are unknown. Thirty-two female pregnant Sprague-Dawley rats were randomly assigned into four groups and were dosed via gavage of triadimefon (0, 25, 50, and 100 mg/kg/day) for 9 days from gestational day (GD) 12-20. Triadimefon significantly reduced serum testosterone level in male fetuses at 100 mg/kg. The double immunofluorescence staining of proliferating cell nuclear antigen (PCNA) and cytochrome P450 cholesterol side-chain cleavage (a biomarker for fetal Leydig cells) was used to measure PCNA-labeling in fetal Leydig cells. It markedly increased fetal Leydig cell number primarily via increasing single cell population and elevated the PCNA-labeling of fetal Leydig cells in male fetuses at 100 mg/kg while it induced abnormal aggregation of fetal Leydig cells. The expression levels of fetal Leydig cell genes, Lhcgr, Scarb1, Star, Cyp11a1, Hsd3b1, Cyp17a1, Hsd17b3, Insl3 and Nr5a1, were determined to explore its effects on fetal Leydig cell development. We found that triadimefon markedly down-regulated the expression of Leydig cell genes, Hsd17b3, Insl3, and Nr5a1 as low as 25 mg/kg and Scarb1 and Cyp11a1 at 100 mg/kg. It did not affect Sertoli cell number but markedly down-regulated the expression of Sertoli cell gene Amh at 50 and 100 mg/kg. Triadimefon significantly down-regulated the expression of antioxidant genes Sod1, Gpx1, and Cat at 25-100 mg/kg, suggesting that it can induce oxidative stress in fetal testis, and it reduced the phosphorylation of ERK1/2 and AKT2 at 100 mg/kg, indicating that it can inhibit the development of fetal Leydig cells. In conclusion, gestational exposure to triadimefon inhibits the development of fetal Leydig cells in male fetuses by inhibiting its differentiation.

9.
Toxicol Lett ; 351: 65-77, 2021 Oct 15.
Article in English | MEDLINE | ID: mdl-34454012

ABSTRACT

Phthalates are plasticizers widely found in the environment. They are potential endocrine disruptors. Bis(2-butoxyethyl) phthalate (BBOP) is a unique phthalate that contains oxygen atoms in the carbon backbone. Little is known about its reproductive and developmental toxicity. The objective of this study was to determine the effect of BBOP on fetal Leydig cell development after in utero exposure to rats. Sprague Dawley pregnant dams were randomly allocated into 6 groups, and were gavaged with BBOP (0, 10, 100, 250, 500, and 1000 mg/kg body weight/day) from gestational day (GD) 14-21. Seven of the 8 dams in the 1000 mg/kg BBOP group died before giving birth. Twelve of the 20 dams in the 500 mg/kg BBOP group had whole litter loss. BBOP significantly reduced the body weight of dams and male offspring and serum testosterone level and anogenital distance of male fetus on GD 21 at 500 mg/kg. BBOP markedly increased fetal Leydig cell proliferation and number at 500 mg/kg while inducing their abnormal aggregation at 250 and 500 mg/kg. BBOP down-regulated the expression of Lhcgr, Scarb1, Star, Cyp11a1, Hsd3b1, Cyp17a1, Hsd17b3, Insl3, and Nr5a1 at various doses while up-regulating the expression of Sertoli cell gene Fshr and Sox9. The phosphorylation of AKT1, AKT2, and ERK1/2 was also markedly reduced by BBOP. In conclusion, BBOP in utero exposure can disrupt fetal Leydig cell development, possibly via the mechanism that may include inhibiting the phosphorylation of AKT1, AKT2, and ERK1/2.


Subject(s)
Fetal Development/drug effects , Leydig Cells/drug effects , Phthalic Acids/toxicity , Animals , Cholesterol Side-Chain Cleavage Enzyme/genetics , Cholesterol Side-Chain Cleavage Enzyme/metabolism , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Fetus , Gene Expression Regulation, Developmental/drug effects , Male , Molecular Structure , Pregnancy , Prenatal Exposure Delayed Effects , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Rats
10.
Toxicol Appl Pharmacol ; 419: 115514, 2021 05 15.
Article in English | MEDLINE | ID: mdl-33798595

ABSTRACT

Phthalates as plasticizers are widely used in many consumer products. Dipentyl phthalate (DPeP) is one of phthalates. However, there are currently few data on whether DPeP exposure affects rat Leydig cell development. In this study, we investigated the effects of in utero DPeP exposure on Leydig cell development in the testes of male newborn and adult rats. From gestational days 14 to 21, Sprague-Dawley pregnant rats were gavaged vehicle (corn oil, control) or DPeP (10, 50, 100, and 500 mg/kg body weight/day). Testosterone and the expression of Leydig cell genes and proteins in the testis at birth and at postnatal day 56 were examined. DPeP dose-dependently reduced serum testosterone levels of male offspring at birth and at postnatal day 56 at 100 and 500 mg/kg and lowered serum luteinizing hormone levels at adult males at ≥10 mg/kg when compared with the control. In addition, DPeP increased number of fetal Leydig cells by inducing their proliferation but down-regulated the expression of Lhcgr, Scarb1, Star, Cyp11a1, Hsd3b1, Cyp17a1, Hsd17b3, and Insl3 in fetal Leydig cells per se. DPeP reduced number of adult Leydig cells by inducing cell apoptosis and down-regulated the expression of Lhcgr and Star in adult Leydig cells at postnatal day 56. DPeP lowered SIRT1 and BCL2 levels in the testis of adult rats. In conclusion, DPeP adversely affects both fetal and adult Leydig cell development after in utero exposure.


Subject(s)
Cell Differentiation/drug effects , Cell Proliferation/drug effects , Leydig Cells/drug effects , Phthalic Acids/toxicity , Plasticizers/toxicity , Prenatal Exposure Delayed Effects , Testis/drug effects , Age Factors , Animals , Apoptosis/drug effects , Female , Gene Expression Regulation, Developmental , Gene Expression Regulation, Enzymologic , Gestational Age , Leydig Cells/metabolism , Leydig Cells/pathology , Luteinizing Hormone/blood , Male , Maternal Exposure , Pregnancy , Rats, Sprague-Dawley , Testis/embryology , Testis/metabolism , Testosterone/blood
11.
Front Pharmacol ; 11: 573475, 2020.
Article in English | MEDLINE | ID: mdl-33041816

ABSTRACT

Intestinal barrier dysfunction contributes to the development of intestinal diseases. Propionic acid (PA), a metabolite generated by anaerobic fermentation of dietary fiber in the intestinal cavity, has been proved to exert anti-inflammatory effects in a variety of diseases. However, the exact role of PA in LPS-induced intestinal barrier dysfunction is still unclear. Accordingly, we examined the latent mechanism of PA and its protective role in LPS-induced intestinal barrier dysfunction by both in vitro and in vivo experiments. In vitro, we identified that PA treatment could strongly promote cell migration, inhibit activation of NLRP3 inflammasome and maintain intestinal barrier function in LPS-induced IEC-6 cells, indicating the protective effect on the intestinal barrier function of PA. Further investigation of the mechanism involved revealed that PA could suppress the activation of TLR4/NF-κB pathway. In vivo, in a LPS-induced rat model, PA-induced protective effects in intestinal barrier dysfunction could be detected. In summary, our findings clarify the role of PA in intestinal barrier dysfunction and suggest that it is promising for the treatment of LPS-related intestinal diseases.

12.
Cancers (Basel) ; 11(10)2019 Oct 14.
Article in English | MEDLINE | ID: mdl-31615145

ABSTRACT

Successful cancer therapy requires drugs being precisely delivered to tumors. Nanosized drugs have attracted considerable recent attention, but their toxicity and high immunogenicity are important obstacles hampering their clinical translation. Here we report a novel "cocktail therapy" strategy based on excess natural killer cell-derived exosomes (NKEXOs) in combination with their biomimetic core-shell nanoparticles (NNs) for tumor-targeted therapy. The NNs were self- assembled with a dendrimer core loading therapeutic miRNA and a hydrophilic NKEXOs shell. Their successful fabrication was confirmed by transmission electron microscopy (TEM) and confocal laser scanning microscopy (CLSM). The resulting NN/NKEXO cocktail showed highly efficient targeting and therapeutic miRNA delivery to neuroblastoma cells in vivo, as demonstrated by two-photon excited scanning fluorescence imaging (TPEFI) and with an IVIS Spectrum in vivo imaging system (IVIS), leading to dual inhibition of tumor growth. With unique biocompatibility, we propose this NN/NKEXO cocktail as a new avenue for tumor therapy, with potential prospects for clinical applications.

SELECTION OF CITATIONS
SEARCH DETAIL
...