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










Publication year range
1.
Article in English | MEDLINE | ID: mdl-38985660

ABSTRACT

Significance: Redox stress underlies numerous vascular disease mechanisms. Metabolic adaptability is essential for vascular cells to preserve energy and redox homeostasis. Recent Advances: Single-cell technologies and multiomic studies demonstrate significant metabolic heterogeneity among vascular cells in health and disease. Increasing evidence shows that reductive or oxidative stress can induce metabolic reprogramming of vascular cells. A recent example is intracellular L-2-hydroxyglutarate accumulation in response to hypoxic reductive stress, which attenuates the glucose flux through glycolysis and mitochondrial respiration in pulmonary vascular cells and provides protection against further reductive stress. Critical Issues: Regulation of cellular redox homeostasis is highly compartmentalized and complex. Vascular cells rely on multiple metabolic pathways, but the precise connectivity among these pathways and their regulatory mechanisms is only partially defined. There is also a critical need to understand better the cross-regulatory mechanisms between the redox system and metabolic pathways as perturbations in either systems or their cross talk can be detrimental. Future Directions: Future studies are needed to define further how multiple metabolic pathways are wired in vascular cells individually and as a network of closely intertwined processes given that a perturbation in one metabolic compartment often affects others. There also needs to be a comprehensive understanding of how different types of redox perturbations are sensed by and regulate different cellular metabolic pathways with specific attention to subcellular compartmentalization. Lastly, integration of dynamic changes occurring in multiple metabolic pathways and their cross talk with the redox system is an important goal in this multiomics era.

3.
bioRxiv ; 2024 May 30.
Article in English | MEDLINE | ID: mdl-38853866

ABSTRACT

Hypoxia-inducible factor 1α (HIF1α) is a master regulator of numerous biological processes under low oxygen tensions. Yet, the mechanisms and biological consequences of aerobic HIF1α activation by intrinsic factors, particularly in primary cells remain elusive. Here, we show that HIF1α signaling is activated in several human primary vascular cells under ambient oxygen tensions, and in vascular smooth muscle cells (VSMCs) of normal human lung tissue, which contributed to a relative resistance to further enhancement of glycolytic activity in hypoxia. Mechanistically, aerobic HIFα activation is mediated by paracrine secretion of three branched chain α-ketoacids (BCKAs), which suppress prolyl hydroxylase domain-containing protein 2 (PHD2) activity via direct inhibition and via lactate dehydrogenase A (LDHA)-mediated generation of L-2-hydroxyglutarate (L2HG). Metabolic dysfunction induced by BCKAs was observed in the lungs of rats with pulmonary arterial hypertension (PAH) and in pulmonary artery smooth muscle cells (PASMCs) from idiopathic PAH patients. BCKA supplementation stimulated glycolytic activity and promoted a phenotypic switch to the synthetic phenotype in PASMCs of normal and PAH subjects. In summary, we identify BCKAs as novel signaling metabolites that activate HIF1α signaling in normoxia and that the BCKA-HIF1α pathway modulates VSMC function and may be relevant to pulmonary vascular pathobiology.

4.
Food Chem Toxicol ; 188: 114686, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38663762

ABSTRACT

Dibutyl phthalate (DBP) and di(2-ethylhexyl) phthalate (DEHP), two common types of phthalates, are known to cause reproductive and developmental toxicity in animals and humans. The reference doses (RfD) of DBP and DEHP should be determined by sensitive endpoints. We here aimed to identify sensitive endpoints for DBP- and DEHP-induced such toxicity using published literatures. By examining the impacts of maternal exposure to DBP or DEHP on anogenital distance (AGD) and semen quality of offspring, we discovered that DBP or DEHP caused AGD decline in boys but increase in girls with DBP being more potent and the first 14weeks of pregnancy being more susceptible, suggesting a chemical- and time-dependent phenomenon. We also identified AGD shortening and total sperm count reduction as two sensitive endpoints for DBP- or DEHP-induced reproductive and developmental toxicity, respectively. Based upon these two endpoints and the employment of the Bayesian benchmark dose approach with an uncertainty factor of 3,000, we estimated the RfD values of DBP and DEHP were 15 µg/kg/day and 36 µg/kg/day, respectively. Thus, we uncover previously unrecognized phenomena of DBP- or DEHP-induced reproductive and developmental toxicity and establish new and comparable or more conservative RfDs for the risk assessment of phthalates exposure in humans.


Subject(s)
Dibutyl Phthalate , Reproduction , Male , Humans , Reproduction/drug effects , Female , Animals , Dibutyl Phthalate/toxicity , Pregnancy , Diethylhexyl Phthalate/toxicity , Phthalic Acids/toxicity , Maternal Exposure/adverse effects
5.
Sci Total Environ ; 904: 166955, 2023 Dec 15.
Article in English | MEDLINE | ID: mdl-37704144

ABSTRACT

Cadmium (Cd) is a commonly found environmental pollutant and is known to damage multiple organs with kidneys being the most common one. N-methyl-d-aspartate receptor 1 (NMDAR1) is a ligand-gated ion channel that is highly permeable to calcium ion (Ca2+). Because Cd2+ and Ca2+ have structural and physicochemical similarities, whether and how Cd could interfere NMDAR1 function to cause renal epithelial cells dysfunction remains unknown. In this study, we investigated the role of NMDAR1 in Cd-induced renal damage and found that Cd treatment upregulated NMDAR1 expression and promoted epithelial-mesenchymal transition (EMT) in mouse kidneys in vivo and human proximal tubular epithelial HK-2 cells in vitro, which were accompanied with activation of the inositol-requiring enzyme 1 (IRE-1α) / spliced X box binding protein-1 (XBP-1s) pathway, an indicative of endoplasmic reticulum (ER) stress. Mechanistically, NMDAR1 upregulation by Cd promoted Ca2+ channel opening and Ca2+ influx, resulting in ER stress and subsequently EMT in HK-2 cells. Inhibition of NMDAR1 by pharmacological antagonist MK-801 significantly attenuated Cd-induced Ca2+ influx, ER stress, and EMT. Pretreatment with the IRE-1α/XBP-1s pathway inhibitor STF-083010 also restored the epithelial phenotype of Cd-treated HK-2 cells. Therefore, our findings suggest that NMDAR1 activation mediates Cd-induced EMT in proximal epithelial cells likely through the IRE-1α/XBP-1s pathway, supporting the idea that NMDAR1 could be a potential therapeutic target for Cd-induced renal damage.


Subject(s)
Cadmium , Epithelial-Mesenchymal Transition , Mice , Animals , Humans , Cadmium/toxicity , Cadmium/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Cell Line , Epithelial Cells
6.
Circ Res ; 131(7): 562-579, 2022 09 16.
Article in English | MEDLINE | ID: mdl-36043417

ABSTRACT

BACKGROUND: L-2-hydroxyglutarate (L2HG) couples mitochondrial and cytoplasmic energy metabolism to support cellular redox homeostasis. Under oxygen-limiting conditions, mammalian cells generate L2HG to counteract the adverse effects of reductive stress induced by hypoxia. Very little is known, however, about whether and how L2HG provides tissue protection from redox stress during low-flow ischemia (LFI) and ischemia-reperfusion injury. We examined the cardioprotective effects of L2HG accumulation against LFI and ischemia-reperfusion injury and its underlying mechanism using genetic mouse models. METHODS AND RESULTS: L2HG accumulation was induced by homozygous (L2HGDH [L-2-hydroxyglutarate dehydrogenase]-/-) or heterozygous (L2HGDH+/-) deletion of the L2HGDH gene in mice. Hearts isolated from these mice and their wild-type littermates (L2HGDH+/+) were subjected to baseline perfusion and 90-minute LFI or 30-minute no-flow ischemia followed by 60- or 120-minute reperfusion. Using [13C]- and [31P]-NMR (nuclear magnetic resonance) spectroscopy, high-performance liquid chromatography, reverse transcription quantitative reverse transcription polymerase chain reaction, ELISA, triphenyltetrazolium staining, colorimetric/fluorometric spectroscopy, and echocardiography, we found that L2HGDH deletion induces L2HG accumulation at baseline and under stress conditions with significant functional consequences. In response to LFI or ischemia-reperfusion, L2HG accumulation shifts glucose flux from glycolysis towards the pentose phosphate pathway. These key metabolic changes were accompanied by enhanced cellular reducing potential, increased elimination of reactive oxygen species, attenuated oxidative injury and myocardial infarction, preserved cellular energy state, and improved cardiac function in both L2HGDH-/- and L2HGDH+/- hearts compared with L2HGDH+/+ hearts under ischemic stress conditions. CONCLUSION: L2HGDH deletion-induced L2HG accumulation protects against myocardial injury during LFI and ischemia-reperfusion through a metabolic shift of glucose flux from glycolysis towards the pentose phosphate pathway. L2HG offers a novel mechanism for eliminating reactive oxygen species from myocardial tissue, mitigating redox stress, reducing myocardial infarct size, and preserving high-energy phosphates and cardiac function. Targeting L2HG levels through L2HGDH activity may serve as a new therapeutic strategy for cardiovascular diseases related to oxidative injury.


Subject(s)
Myocardial Infarction , Myocardial Reperfusion Injury , Animals , Glucose/pharmacology , Glutarates , Mammals , Mice , Myocardial Infarction/metabolism , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/prevention & control , Oxidative Stress , Oxygen , Phosphates/pharmacology , Reactive Oxygen Species/metabolism
7.
Arterioscler Thromb Vasc Biol ; 42(9): 1169-1185, 2022 09.
Article in English | MEDLINE | ID: mdl-35924558

ABSTRACT

BACKGROUND: Endothelial dysfunction is a critical component in the pathogenesis of cardiovascular diseases and is closely associated with nitric oxide (NO) levels and oxidative stress. Here, we report on novel findings linking endothelial expression of CD70 (also known as CD27 ligand) with alterations in NO and reactive oxygen species. METHODS: CD70 expression was genetically manipulated in human aortic and pulmonary artery endothelial cells. Intracellular NO and hydrogen peroxide (H2O2) were measured using genetically encoded biosensors, and cellular phenotypes were assessed. RESULTS: An unbiased phenome-wide association study demonstrated that polymorphisms in CD70 associate with vascular phenotypes. Endothelial cells treated with CD70-directed short-interfering RNA demonstrated impaired wound closure, decreased agonist-stimulated NO levels, and reduced eNOS (endothelial nitric oxide synthase) protein. These changes were accompanied by reduced NO bioactivity, increased 3-nitrotyrosine levels, and a decrease in the eNOS binding partner heat shock protein 90. Following treatment with the thioredoxin inhibitor auranofin or with agonist histamine, intracellular H2O2 levels increased up to 80% in the cytosol, plasmalemmal caveolae, and mitochondria. There was increased expression of NADPH oxidase 1 complex and gp91phox; expression of copper/zinc and manganese superoxide dismutases was also elevated. CD70 knockdown reduced levels of the H2O2 scavenger catalase; by contrast, glutathione peroxidase 1 expression and activity were increased. CD70 overexpression enhanced endothelial wound closure, increased NO levels, and attenuated the reduction in eNOS mRNA induced by TNFα. CONCLUSIONS: Taken together, these data establish CD70 as a novel regulatory protein in endothelial NO and reactive oxygen species homeostasis, with implications for human vascular disease.


Subject(s)
CD27 Ligand , Endothelial Cells , Nitric Oxide , CD27 Ligand/metabolism , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Humans , Hydrogen Peroxide/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/metabolism , Oxidation-Reduction , Oxidative Stress , Reactive Oxygen Species/metabolism
9.
J Biol Chem ; 295(20): 6946-6957, 2020 05 15.
Article in English | MEDLINE | ID: mdl-32265301

ABSTRACT

The incidence of pancreatic cancer increases with age, suggesting that chronological aging is a significant risk factor for this disease. Fibroblasts are the major nonmalignant cell type in the stroma of human pancreatic ductal adenocarcinoma (PDAC). In this study, we investigated whether the chronological aging of normal human fibroblasts (NHFs), a previously underappreciated area in pancreatic cancer research, influences the progression and therapeutic outcomes of PDAC. Results from experiments with murine xenografts and 2D and 3D co-cultures of NHFs and PDAC cells revealed that older NHFs stimulate proliferation of and confer resistance to radiation therapy of PDAC. MS-based metabolite analysis indicated that older NHFs have significantly increased arachidonic acid 12-lipoxygenase (ALOX12) expression and elevated levels of its mitogenic metabolite, 12-(S)-hydroxy-5,8,10,14-eicosatetraenoic acid (12-(S)-HETE) compared with their younger counterparts. In co-cultures with older rather than with younger NHFs, PDAC cells exhibited increases in mitogen-activated protein kinase signaling and cellular metabolism, as well as a lower oxidation state that correlated with their enhanced proliferation and resistance to radiation therapy. Expression of ALOX12 was found to be significantly lower in PDAC cell lines and tumor biopsies, suggesting that PDAC cells rely on a stromal supply of mitogens for their proliferative needs. Pharmacological (hydroxytyrosol) and molecular (siRNA) interventions of ALOX12 in older NHFs suppressed their ability to stimulate proliferation of PDAC cells. We conclude that chronological aging of NHFs contributes to PDAC progression and that ALOX12 and 12-(S)-HETE may be potential stromal targets for interventions that seek to halt progression and improve therapy outcomes.


Subject(s)
Arachidonate 12-Lipoxygenase/metabolism , Carcinoma, Pancreatic Ductal/metabolism , Cellular Senescence , Hydroxyeicosatetraenoic Acids/metabolism , Neoplasm Proteins/metabolism , Pancreatic Neoplasms/metabolism , Animals , Arachidonate 12-Lipoxygenase/genetics , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Mice , Neoplasm Proteins/genetics , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Stromal Cells/metabolism , Stromal Cells/pathology
10.
Antioxid Redox Signal ; 32(18): 1330-1347, 2020 06.
Article in English | MEDLINE | ID: mdl-31218894

ABSTRACT

Significance: Reducing equivalents (NAD(P)H and glutathione [GSH]) are essential for maintaining cellular redox homeostasis and for modulating cellular metabolism. Reductive stress induced by excessive levels of reduced NAD+ (NADH), reduced NADP+ (NADPH), and GSH is as harmful as oxidative stress and is implicated in many pathological processes. Recent Advances: Reductive stress broadens our view of the importance of cellular redox homeostasis and the influences of an imbalanced redox niche on biological functions, including cell metabolism. Critical Issues: The distribution of cellular NAD(H), NADP(H), and GSH/GSH disulfide is highly compartmentalized. Understanding how cells coordinate different pools of redox couples under unstressed and stressed conditions is critical for a comprehensive view of redox homeostasis and stress. It is also critical to explore the underlying mechanisms of reductive stress and its biological consequences, including effects on energy metabolism. Future Directions: Future studies are needed to investigate how reductive stress affects cell metabolism and how cells adapt their metabolism to reductive stress. Whether or not NADH shuttles and mitochondrial nicotinamide nucleotide transhydrogenase enzyme can regulate hypoxia-induced reductive stress is also a worthy pursuit. Developing strategies (e.g., antireductant approaches) to counteract reductive stress and its related adverse biological consequences also requires extensive future efforts.


Subject(s)
Glutathione/metabolism , NADP/metabolism , Animals , Homeostasis , Humans , Oxidation-Reduction , Oxidative Stress
11.
Antioxid Redox Signal ; 28(3): 251-272, 2018 Jan 20.
Article in English | MEDLINE | ID: mdl-28648096

ABSTRACT

SIGNIFICANCE: The nicotinamide adenine dinucleotide (NAD+)/reduced NAD+ (NADH) and NADP+/reduced NADP+ (NADPH) redox couples are essential for maintaining cellular redox homeostasis and for modulating numerous biological events, including cellular metabolism. Deficiency or imbalance of these two redox couples has been associated with many pathological disorders. Recent Advances: Newly identified biosynthetic enzymes and newly developed genetically encoded biosensors enable us to understand better how cells maintain compartmentalized NAD(H) and NADP(H) pools. The concept of redox stress (oxidative and reductive stress) reflected by changes in NAD(H)/NADP(H) has increasingly gained attention. The emerging roles of NAD+-consuming proteins in regulating cellular redox and metabolic homeostasis are active research topics. CRITICAL ISSUES: The biosynthesis and distribution of cellular NAD(H) and NADP(H) are highly compartmentalized. It is critical to understand how cells maintain the steady levels of these redox couple pools to ensure their normal functions and simultaneously avoid inducing redox stress. In addition, it is essential to understand how NAD(H)- and NADP(H)-utilizing enzymes interact with other signaling pathways, such as those regulated by hypoxia-inducible factor, to maintain cellular redox homeostasis and energy metabolism. FUTURE DIRECTIONS: Additional studies are needed to investigate the inter-relationships among compartmentalized NAD(H)/NADP(H) pools and how these two dinucleotide redox couples collaboratively regulate cellular redox states and cellular metabolism under normal and pathological conditions. Furthermore, recent studies suggest the utility of using pharmacological interventions or nutrient-based bioactive NAD+ precursors as therapeutic interventions for metabolic diseases. Thus, a better understanding of the cellular functions of NAD(H) and NADP(H) may facilitate efforts to address a host of pathological disorders effectively. Antioxid. Redox Signal. 28, 251-272.


Subject(s)
Energy Metabolism , NADP/metabolism , NAD/metabolism , Oxidation-Reduction , Animals , Extracellular Space , Gene Expression Regulation, Enzymologic , Homeostasis , Humans , Intracellular Space , Metabolic Networks and Pathways , Oxidative Stress , Signal Transduction
12.
Arch Toxicol ; 90(2): 319-32, 2016 Feb.
Article in English | MEDLINE | ID: mdl-25417049

ABSTRACT

Polychlorinated biphenyls (PCBs) and their metabolites are environmental pollutants that are known to have adverse health effects. 1-(4-Chlorophenyl)-benzo-2,5-quinone (4-ClBQ), a quinone metabolite of 4-monochlorobiphenyl (PCB3, present in the environment and human blood) is toxic to human skin keratinocytes, and breast and prostate epithelial cells. This study investigates the hypothesis that 4-ClBQ-induced metabolic oxidative stress regulates toxicity in human keratinocytes. Results from Seahorse XF96 Analyzer showed that the 4-ClBQ treatment increased extracellular acidification rate, proton production rate, oxygen consumption rate and ATP content, indicative of metabolic oxidative stress. Results from a q-RT-PCR assay showed significant increases in the mRNA levels of hexokinase 2 (hk2), pyruvate kinase M2 (pkm2) and glucose-6-phosphate dehydrogenase (g6pd), and decreases in the mRNA levels of succinate dehydrogenase (complex II) subunit C and D (sdhc and sdhd). Pharmacological inhibition of G6PD-activity enhanced the toxicity of 4-ClBQ, suggesting that the protective function of the pentose phosphate pathway is functional in 4-ClBQ-treated cells. The decrease in sdhc and sdhd expression was associated with a significant decrease in complex II activity and increase in mitochondrial levels of ROS. Overexpression of sdhc and sdhd suppressed 4-ClBQ-induced inhibition of complex II activity, increase in mitochondrial levels of ROS, and toxicity. These results suggest that the 4-ClBQ treatment induces metabolic oxidative stress in HaCaT cells, and while the protective function of the pentose phosphate pathway is active, inhibition of complex II activity sensitizes HaCaT cells to 4-ClBQ-induced toxicity.


Subject(s)
Benzoquinones/toxicity , Keratinocytes/drug effects , Oxidative Stress/drug effects , Succinate Dehydrogenase/metabolism , Adenosine Triphosphate/metabolism , Biphenyl Compounds/toxicity , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line , Hexokinase/metabolism , Humans , Keratinocytes/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Pentose Phosphate Pathway/drug effects , Succinate Dehydrogenase/genetics , Thyroid Hormones/genetics , Thyroid Hormones/metabolism , Thyroid Hormone-Binding Proteins
13.
Toxicol In Vitro ; 29(7): 1332-8, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26004620

ABSTRACT

Peroxisome proliferator activated receptor γ coactivator 1α (PGC-1α) is a transcriptional coactivator that is known to regulate oxidative stress response by enhancing the expression of antioxidant genes. We have shown previously that 1-(4-Chlorophenyl)-benzo-2,5-quinone (4-ClBQ), a quinone-metabolite of 4-monochlorobiphenyl (PCB3) induces oxidative stress and toxicity in human skin keratinocytes, and breast and prostate epithelial cells. In this study, we investigate whether PGC-1α regulates oxidative stress and toxicity in 4-ClBQ treated HaCaT human keratinocytes. Results showed significant down-regulation in the expression of PGC-1α and catalase in 4-ClBQ treated HaCaT cells. Down-regulation of PGC-1α expression was associated with 4-ClBQ induced increase in the steady-state levels of cellular reactive oxygen species (ROS) and toxicity. Overexpression of pgc-1α enhanced the expression of catalase and suppressed 4-ClBQ induced increase in cellular ROS levels and toxicity. These results suggest that pgc-1α mediates 4-ClBQ induced oxidative stress and toxicity in HaCaT cells presumably by regulating catalase expression.


Subject(s)
Benzoquinones/toxicity , Keratinocytes/drug effects , Transcription Factors/metabolism , Catalase/metabolism , Cell Line , Down-Regulation , Humans , Keratinocytes/metabolism , Oxidative Stress , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Transcription Factors/genetics
14.
Toxicol Lett ; 233(3): 258-66, 2015 Mar 18.
Article in English | MEDLINE | ID: mdl-25668756

ABSTRACT

Aryl hydrocarbon receptor (AhR) is a ligand-dependent transcription factor that plays a critical role in metabolism, cell proliferation, development, carcinogenesis, and xenobiotic response. In general, dioxin-like polychlorinated biphenyls (PCBs) exhibit a ligand-dependent activation of AhR-signaling. Results from this study show that a quinone-derivative (1-(4-Chlorophenyl)-benzo-2,5-quinone; 4-ClBQ) of a non-dioxin like PCB (PCB3) also activates AhR-signaling. Treatments of HaCaT human keratinocytes with 4-ClBQ and dioxin-like PCB126 significantly increased AhR-target gene expression, CYP1A1 mRNA and protein levels. 4-ClBQ-induced increase CYP1A1 expression was associated with an increase in the nuclear translocation of AhR protein as well as an increase in the luciferase-reporter activity of a human CYP1A1 xenobiotic response element (XRE). 6,2',4'-Trimethoxyflavone (TMF), a well-characterized AhR-ligand antagonist significantly suppressed PCB126-induced increase in CYP1A1 expression, while the same treatment did not suppress 4-ClBQ-induced increase in CYP1A1 expression. However, siRNA-mediated down-regulation of AhR significantly inhibited 4-ClBQ-induced increase in CYP1A1 expression, suggesting that AhR mediates 4-ClBQ-induced increase in CYP1A1 expression. Interestingly, treatment with the antioxidant N-acetyl-l-cysteine significantly suppressed 4-ClBQ-induced increase in CYP1A1 expression. Furthermore, CYP1A1 expression also increased in cells treated with hydrogen peroxide. These results demonstrate that a ligand-independent and oxidative stress dependent pathway activates AhR-signaling in 4-ClBQ treated HaCaT cells. Because AhR signaling is believed to mediate xenobiotics response, our results may provide a mechanistic rationale for the use of antioxidants as effective countermeasure to environmental pollutant-induced adverse health effects.


Subject(s)
Benzoquinones/toxicity , Keratinocytes/drug effects , Receptors, Aryl Hydrocarbon/physiology , Signal Transduction/physiology , Active Transport, Cell Nucleus , Biphenyl Compounds/toxicity , Cells, Cultured , Cytochrome P-450 CYP1A1/genetics , Flavones/pharmacology , Humans , Keratinocytes/metabolism , Ligands , Oxidative Stress , Polychlorinated Biphenyls/toxicity
15.
Int J Radiat Oncol Biol Phys ; 87(3): 619-25, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-24074935

ABSTRACT

PURPOSE: Radiation is a common mode of cancer therapy whose outcome is often limited because of normal tissue toxicity. We have shown previously that the accumulation of radiation-induced late reactive oxygen species (ROS) precedes cell death, suggesting that metabolic oxidative stress could regulate cellular radiation response. The purpose of this study was to investigate whether selenoprotein P (SEPP1), a major supplier of selenium to tissues and an antioxidant, regulates late ROS accumulation and toxicity in irradiated normal human fibroblasts (NHFs). METHODS AND MATERIALS: Flow cytometry analysis of cell viability, cell cycle phase distribution, and dihydroethidium oxidation, along with clonogenic assays, were used to measure oxidative stress and toxicity. Human antioxidant mechanisms array and quantitative real-time polymerase chain reaction assays were used to measure gene expression during late ROS accumulation in irradiated NHFs. Sodium selenite addition and SEPP1 overexpression were used to determine the causality of SEPP1 regulating late ROS accumulation and toxicity in irradiated NHFs. RESULTS: Irradiated NHFs showed late ROS accumulation (4.5-fold increase from control; P<.05) that occurs after activation of the cell cycle checkpoint pathways and precedes cell death. The mRNA levels of CuZn- and Mn-superoxide dismutase, catalase, peroxiredoxin 3, and thioredoxin reductase 1 increased approximately 2- to 3-fold, whereas mRNA levels of cold shock domain containing E1 and SEPP1 increased more than 6-fold (P<.05). The addition of sodium selenite before the radiation treatment suppressed toxicity (45%; P<.05). SEPP1 overexpression suppressed radiation-induced late ROS accumulation (35%; P<.05) and protected NHFs from radiation-induced toxicity (58%; P<.05). CONCLUSION: SEPP1 mitigates radiation-induced late ROS accumulation and normal cell injury.


Subject(s)
Radiation Injuries/prevention & control , Reactive Oxygen Species/metabolism , Selenoprotein P/physiology , Cell Cycle Checkpoints/physiology , Cell Cycle Checkpoints/radiation effects , Cell Death , Cell Survival , Dose-Response Relationship, Radiation , Ethidium/analogs & derivatives , Ethidium/metabolism , Fibroblasts/metabolism , Fibroblasts/radiation effects , Genes, vif , Humans , Oxidative Stress/genetics , Real-Time Polymerase Chain Reaction , Selenoprotein P/genetics , Selenoprotein P/metabolism , Sodium Selenite/pharmacology
16.
Free Radic Biol Med ; 65: 70-77, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23770201

ABSTRACT

Polychlorinated biphenyls and their metabolites are environmental pollutants that are believed to have adverse health effects presumably by inducing oxidative stress. To determine if 1-(4-Chlorophenyl)-benzo-2,5-quinone (4-ClBQ; metabolite of 4-monochlorobiphenyl, PCB3)-induced oxidative stress is associated with changes in the expression of specific antioxidant genes, mRNA levels of 92 oxidative stress-response genes were analyzed using TaqMan Array Human Antioxidant Mechanisms (Life Technologies), and results were verified by performing quantitative RT-PCR assays. The expression of selenoprotein P (sepp1) was significantly downregulated (8- to 10-fold) in 4-ClBQ-treated HaCaT human skin keratinocytes, which correlated with a significant increase in MitoSOX oxidation. Overexpression of Mn-superoxide dismutase or catalase or treatment with N-acetyl-l-cysteine suppressed 4-ClBQ-induced toxicity. Sodium selenite supplementation also suppressed 4-ClBQ-induced decrease in sepp1 expression, which was associated with a significant inhibition in cell death. Furthermore, HaCaT cells overexpressing sepp1 were resistant to 4-ClBQ-induced oxidative stress and toxicity. These results demonstrate that SEPP1 represents a previously unrecognized regulator of PCB-induced biological effects. These results support the speculation that selenoproteins can be an attractive countermeasure for PCB-induced adverse biological effects.


Subject(s)
Benzoquinones/toxicity , Environmental Pollutants/toxicity , Keratinocytes/metabolism , Oxidative Stress/physiology , Selenoprotein P/metabolism , Cell Line , Flow Cytometry , Humans , Immunoblotting , Keratinocytes/drug effects , Oligonucleotide Array Sequence Analysis , Real-Time Polymerase Chain Reaction
17.
Environ Toxicol ; 26(6): 657-68, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21374784

ABSTRACT

In this study, we investigated the time-course changes of hormone levels and sperm numbers in male Sprague-Dawley (SD) rats after neonatal exposure to 3,3',4,4',5,5'-hexachlorobiphenyl (PCB169). Neonatal rats were given (through oral gavages) doses of 0, 0.025, 0.25, or 0.5 mg/kg-day of PCB169 in corn oil from postnatal day 1 (PND1) to PND7. The rats were sacrificed at PND8, PND21, and PND90. PCB169 exposure was confirmed by the marked induction of liver CYP1A1 mRNA expression at these three time points. The testicular daily sperm production and the sperm counts of the epididymis cauda significantly decreased at PND90 compared to that of control. Although reductions in serum thyroxine and triiodothyronine levels occurred at all these three time points and at both PND21 and PND90, respectively, the mRNA expression of testicular thyroid hormone receptor α1 was suppressed significantly only at PND8. The serum and testicular testosterone (T) levels declined markedly at PND90 compared to the controls, but there was no effect at PND21. The mRNA expression of testicular steroidogenic factor 1 was inhibited markedly at the three time points, whereas those of StAR, P450c17, P450scc, and 3ß-HSD were suppressed significantly only at PND90 relative to the controls. PCB169 treatment had no effects on pituitary follicle-stimulating hormone and luteinizing hormone levels and on their receptors' expression in the testes. These results indicate that neonatal exposure to PCB169 damages hormone levels and testicular function in the long-term, resulting in persistent hypothyroidism and decreases in adult T levels and sperm counts.


Subject(s)
Environmental Pollutants/toxicity , Hormones/metabolism , Polychlorinated Biphenyls/toxicity , Testis/drug effects , Animals , Cytochrome P-450 CYP1A1/metabolism , Follicle Stimulating Hormone/blood , Follicle Stimulating Hormone/metabolism , Follicle Stimulating Hormone/pharmacology , Hypothyroidism/metabolism , Liver/metabolism , Luteinizing Hormone/metabolism , Male , Rats , Rats, Sprague-Dawley , Sperm Count , Testis/metabolism , Testis/physiology , Testosterone/metabolism , Triiodothyronine/metabolism
18.
Article in Chinese | MEDLINE | ID: mdl-21114030

ABSTRACT

OBJECTIVE: To determine the long-term testicular effect after neonatal exposure to 2,2', 4,4',5,5'-hexa-chlorobiphenyl (PCB153). METHODS: On birth day (Postnatal day 0, PNDO), the Sprague-Dawley (SD) male rats were mixed together and divided into 12 pups/litter. At PND1, the rats were grouped randomly into control and treatment groups according to different litters, 24 pups/group. They were treated by oral gavage with PCB153 in corn oil at doses of 0, 0.025, 0.250 and 2.500 mg/kg BW-day from PNDI to PND7. The rats were sacrificed at PND8 and PND90 by anesthesia. The testes were collected and weighed for histological examination and daily sperm production at PND8 or/and PND90. The epididymidis and the epididymidis cauda also were collected and weighed for determination the sperm counts at PND90. RESULTS: The body weight of 2.500 mg/kg dose group was decreased significantly from PND3 to PND8 compared with that of control (P < 0.05). At PND8, the loose structure in seminiferous cord and the spermatogonia with enlarged volume and detached from the cord were observed in 2.500 mg/kg dose group by light microscope and electronic microscopy. With the increase of exposure doses, the testicular daily sperm production (DSP) and the sperm counts of epididymidis cauda were decreased in dose-dependent manner at PND90. The DSP in 0.250 mg/kg [30 x 10(6)/testis(g)] and 2.500 mg/kg [18 x l0(6)/testis(g)] dose groups were significantly reduced compared with that of control [36 x 10(6)/testis(g)] (P < 0.05). And there was a significant reduction in the sperm counts of epididymidis cauda in 0.250 mg/kg [42 x 10(7)/epididymidis cauda (g)] and 2.500 mg/kg [18 x 10(7)/epididymidis cauda (g)] dose groups compared with that of control [51 x 10(7)/epididymidis cauda (g)] (P < 0.05). CONCLUSIONS: The spermatogenesis of adult testis is disturbed, which causes the decrease in the testicular DSP and the sperm counts of epididymidis cauda after neonatal exposure to PCB153. The long-term damage in male reproductive function is caused by neonatal exposure to chemicals.


Subject(s)
Environmental Exposure , Polychlorinated Biphenyls/toxicity , Spermatogenesis/drug effects , Testis/drug effects , Animals , Animals, Newborn , Male , Rats , Rats, Sprague-Dawley , Sperm Count , Testis/pathology , Testis/ultrastructure
19.
Article in English | MEDLINE | ID: mdl-20025066

ABSTRACT

BACKGROUND: The aim of the present study is to determine the long-term testicular effects of neonatal exposure to PCB153. METHODS: Sprague-Dawley (SD) rats were treated by oral gavage with PCB153 in corn oil at doses of 0, 0.025, and 2.5 mg/kg per day from postnatal day 3 (PND 3) to PND7. The rats were sacrificed on PND 8 and PND 77. TUNEL in situ detection for testis apoptosis, immunohistochemical staining of thyroid gland for thyroxine (T4), semi-quantitative RT-PCR for mRNA expression, and radioimmunoassay (RIA) for serum hormone levels were performed. RESULTS: Neonatal treatment with PCB153 at both doses had no obvious effects on body weight, testis weight, testis histology, and germ cell apoptosis, but decreased T4 staining in thyroid gland was observed on PND 8. On PND 77, neonatal treatment with 2.5 mg/kg per day of PCB153 significantly reduced daily sperm product (DSP). Serum levels of thyroxine (T4) and free thyroxine (FT4) decreased, but there were no differences in thyroid-stimulating hormone (TSH) level between the control and exposed groups. Gap junction connexin43 (CX43) and cyclin-dependent kinase inhibitor (CDKI) P27kip1 mRNA expression, which was associated with Sertoli cell differentiation, was significantly reduced after PCB153 treatment on PND 8 but not on PND 77. Androgen-binding protein (ABP) and androgen receptor (AR) mRNA expression, which indicates Sertoli cell maturation, was suppressed on PND 77 after neonatal PCB153 exposure. CONCLUSIONS: The findings in this study suggest that neonatal exposure to PCB153 induces persistent T4 reduction, which disturbs Sertoli cell function, and subsequently results in alterations in adult spermatogenesis. Birth Defects Res (Part B) 89:18-25, 2010. (c) 2009 Wiley-Liss, Inc.


Subject(s)
Biphenyl Compounds/toxicity , Polychlorinated Biphenyls/toxicity , Spermatogenesis/drug effects , Thyroxine/chemistry , Administration, Oral , Animals , Apoptosis , Female , Immunohistochemistry/methods , In Situ Nick-End Labeling , Male , Maternal Exposure , Rats , Rats, Sprague-Dawley , Thyroid Hormones , Time Factors
20.
Article in Chinese | MEDLINE | ID: mdl-20137407

ABSTRACT

OBJECTIVE: To investigate the effects of neonatal exposure of DNA methylation inhibitor, Cadmium and PCB153 on DNA methylation, apoptosis and spermatogenesis in SD rats. METHODS: Neonatal SD rats were randomly divided into 10 groups and received oral administrations of PCB153 (0.025, 0. 250, 2.500 mg/kg), or Cadmium (1, 2, 4 mg/kg), or positive control 5-Aza-CdR (0.025, 0.250 mg/kg), or vehicle control for five days from PND3. Half of the rats were killed 24 h after the last administration. The remains were fed until 12 weeks. Sperm numbers, apoptosis and DNA methylation levels in testis were investigated. RESULTS: The daily sperm production was significantly decreased in each neonatal exposed group (P < 0.05). Neonatal rats exposed to 5-Aza-CdR and Cadmium reduced the global DNA methylation level, increased apoptosis, while PCB153 exposure did not significantly change DNA methylation and apoptosis. CONCLUSION: Neonatal rats exposed to chemicals could reduce spermatogenesis via multiple pathways. Lower DNA methylation and increased neonatal apoptosis were suggested as one of the causes.


Subject(s)
Cadmium/toxicity , DNA Methylation/drug effects , Polychlorinated Biphenyls/toxicity , Spermatogenesis/drug effects , Animals , Animals, Newborn , Apoptosis/drug effects , Male , Rats , Rats, Sprague-Dawley , Testis/drug effects , Testis/metabolism , Testis/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...