Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Int J Mol Sci ; 10(5): 2122-2135, 2009 May 14.
Article in English | MEDLINE | ID: mdl-19564943

ABSTRACT

Quantum dots (QDs) are useful novel luminescent markers, but their embryonic toxicity is yet to be fully established, particularly in oocyte maturation and sperm fertilization. Earlier experiments by our group show that CdSe-core QDs have cytotoxic effects on mouse blastocysts and are associated with defects in subsequent development. Here, we further investigate the influence of CdSe-core QDs on oocyte maturation, fertilization, and subsequent pre- and postimplantation development. CdSe-core QDs induced a significant reduction in the rates of oocyte maturation, fertilization, and in vitro embryo development, but not ZnS-coated CdSe QDs. Treatment of oocytes with 500 nM CdSe-core QDs during in vitro maturation (IVM) led to increased resorption of postimplantation embryos and decreased placental and fetal weights. To our knowledge, this is the first study to report the negative impact of CdSe-core QDs on mouse oocyte development. Moreover, surface modification of CdSe-core QDs with ZnS effectively prevented this cytotoxicity.


Subject(s)
Apoptosis/drug effects , Cadmium Compounds/metabolism , Embryonic Development/drug effects , Oocytes/drug effects , Quantum Dots/toxicity , Selenium Compounds/metabolism , Animals , Blastocyst/drug effects , Blastocyst/metabolism , Cadmium Compounds/chemistry , Cell Proliferation/drug effects , Female , Fertilization/drug effects , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Oocytes/growth & development , Oocytes/metabolism , Placenta/physiology , Pregnancy , Quantum Dots/chemistry , Quantum Dots/metabolism , Selenium Compounds/chemistry , Zinc Sulfate/chemistry
2.
Toxicol Lett ; 188(1): 63-9, 2009 Jul 10.
Article in English | MEDLINE | ID: mdl-19433271

ABSTRACT

Ginkgolide B (GKB), the major active component of Ginkgo biloba extracts, exerts both stimulatory and inhibitory effects on apoptotic signaling. Previous studies by our group demonstrated that ginkgolide treatment of mouse blastocysts induces apoptosis, decreases cell number, hinders early postimplantation blastocyst development, and increases early-stage blastocyst death. Here, we further investigate the effects of GKB on oocyte maturation, and subsequent pre- and postimplantation development in vitro and in vivo. In our experiments, GKB induced a significant reduction in the rate of oocyte maturation, fertilization, and in vitro embryonic development. Treatment of oocytes with 1-6 microM GKB during in vitro maturation (IVM) led to increased resorption of postimplantation embryos and decreased placental and fetal weights. Data obtained using an in vivo mouse model further disclosed that consumption of drinking water containing 3-6 microM GKB led to decreased oocyte maturation and in vitro fertilization, as well as early embryo developmental injury, specifically, inhibition of development to the blastocyst stage in vivo. To our knowledge, this is the first study to investigate the impact of GKB on maturation of mouse oocytes, fertilization, and sequential embryonic development.


Subject(s)
Blastocyst/drug effects , Drugs, Chinese Herbal/toxicity , Fertilization/drug effects , Ginkgolides/toxicity , Lactones/toxicity , Oocytes/drug effects , Oogenesis/drug effects , Administration, Oral , Animals , Apoptosis/drug effects , Blastocyst/pathology , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drugs, Chinese Herbal/administration & dosage , Embryo Culture Techniques , Embryo Transfer , Embryonic Development/drug effects , Female , Fertilization in Vitro , Ginkgolides/administration & dosage , Lactones/administration & dosage , Mice , Mice, Inbred ICR , Models, Animal , Oocytes/pathology
3.
Acta Pharmacol Sin ; 29(2): 259-66, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18215357

ABSTRACT

AIM: The aim of this study was to examine the cytotoxic effect of quantum dots (QD), a novel luminescent material, on early post-implantation embryonic development. METHODS: Mouse blastocysts were incubated in medium with or without CdSe-core QD (250 or 500 nmol/L) for 24 h. Cell apoptosis was analyzed by terminal deoxynucleotidyl transferase-mediated digoxigenin-dUTP nick-end labeling assay and Annexin V/propidium iodide staining, and proliferation was investigated by dual differential staining. Pre-implantation and post-implantation development was assessed by in vitro and in vivo analyses, respectively. RESULTS: The apoptotic staining analysis showed that CdSe-core QD induced apoptosis in mouse blastocysts in a dose-dependent manner. Pretreatment of blastocysts with CdSe-core QD inhibited cell proliferation, primarily in the inner cell mass. CdSe-core QD also inhibited post-implantation embryonic development; fewer CdSe-core QD-pretreated blastocysts reached the later stages of development compared to the controls. The pre-implantation development of morulas into blastocysts was also inhibited by CdSe-core QD. Furthermore, CdSe-core QD at 500 nmol/L were associated with resorption of post-implantation blastocysts and a decrease in fetal weight. The cytotoxicity of CdSe QD in embryonic development was significantly reduced by the addition of a ZnS coating. CONCLUSION: Our results show that CdSe-core QD induce apoptosis in mouse blastocysts, inhibit cell proliferation, retard early post-implantation blastocyst development, and increase early-stage blastocyst death in vitro and in vivo.


Subject(s)
Cadmium Compounds/pharmacology , Cell Survival/drug effects , Embryonic Development/drug effects , Quantum Dots , Selenium Compounds/pharmacology , Animals , Apoptosis/drug effects , Blastocyst/cytology , Blastocyst/drug effects , Cell Count , Cell Proliferation/drug effects , Embryo Transfer , Female , Mice , Mice, Inbred ICR , Pregnancy
4.
Toxicology ; 242(1-3): 109-22, 2007 Dec 05.
Article in English | MEDLINE | ID: mdl-17950979

ABSTRACT

Previous studies have established that ethanol induces apoptosis, but the precise molecular mechanisms are currently unclear. Here, we show that 0.3-1.0% (w/v) ethanol induces apoptosis in mouse blastocysts and that resveratrol, a grape-derived phytoalexin with known antioxidant and anti-inflammatory properties, prevents ethanol-induced apoptosis and inhibition of cell proliferation. Moreover, ethanol-treated blastocysts show normal levels of implantation on culture dishes in vitro but a reduced ability to reach the later stages of embryonic development. Pretreatment with resveratrol prevented ethanol-induced disruption of embryonic development in vitro and in vivo. In an in vitro cell-based assay, we further found that ethanol increases the production of reactive oxygen species in ESC-B5 embryonic stem cells, leading to an increase in the intracellular concentrations of cytoplasmic free Ca(2+) and NO, loss of mitochondrial membrane potential, mitochondrial release of cytochrome c, activation of caspase-9 and -3, and apoptosis. These changes were blocked by pretreatment with resveratrol. Based on these results, we propose a model for the protective effect of resveratrol on ethanol-induced cell injury in blastocysts and ESC-B5 cells.


Subject(s)
Antioxidants/pharmacology , Apoptosis/drug effects , Blastocyst/drug effects , Embryonic Stem Cells/drug effects , Ethanol/toxicity , Stilbenes/pharmacology , Animals , Blastocyst/metabolism , Blastocyst/pathology , Calcium/metabolism , Caspase 3/metabolism , Caspase 9/metabolism , Cell Line , Cell Proliferation/drug effects , Cell Survival/drug effects , Cytochromes c/metabolism , Dose-Response Relationship, Drug , Embryo Culture Techniques , Embryonic Development/drug effects , Embryonic Stem Cells/enzymology , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/pathology , Enzyme Activation , Female , Male , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Inbred ICR , Mitochondria/drug effects , Mitochondria/metabolism , Nitric Oxide/metabolism , Pregnancy , Reactive Oxygen Species/metabolism , Resveratrol , Time Factors , Up-Regulation , p21-Activated Kinases/antagonists & inhibitors , p21-Activated Kinases/metabolism
5.
Reprod Toxicol ; 24(1): 120-5, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17572064

ABSTRACT

Citrinin (CTN), a mycotoxin that is often found as a natural contaminant in foodstuffs and animal feeds, has been demonstrated to have cytotoxic and genotoxic effects on various mammalian cells. In this study, we examined the cytotoxic effects of CTN on mouse blastocysts and subsequent early development in vitro and in vivo. Blastocysts treated with 15 or 30 microM CTN showed significant increases in apoptosis and significant decreases in total cell number. In addition, CTN-pretreated blastocysts showed a significantly lower implantation success rate. Treatment with 30 microM CTN was associated with increased resorption of postimplantation embryos and decreased fetal weight. Our results collectively indicate that CTN-induced apoptosis in the mouse blastocyst reduced cell number and retarded early postimplantation development. The extent to which CTN may have teratogenic potential in early human development is not known.


Subject(s)
Blastocyst/drug effects , Citrinin/toxicity , Embryonic Development/drug effects , Mycotoxins/toxicity , Animals , Apoptosis/drug effects , Blastocyst/pathology , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Embryo Culture Techniques , Embryo Implantation/drug effects , Embryo Loss , Embryo Transfer , Female , Fetal Weight/drug effects , Mice , Mice, Inbred ICR
6.
Acta Pharmacol Sin ; 28(2): 238-45, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17241527

ABSTRACT

AIM: To examine the cytotoxic effects of genistein, an isoflavone compound, on early postimplantation embryonic development in vitro. METHODS: Mouse blastocysts were incubated in medium with or without genistein (25 or 50 micromol/L) or daidzein (50 micromol/L) for 24 h. Cell proliferation and growth was investigated by dual differential staining, apoptosis was analyzed by terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL) assay, and apoptotic or necrotic cells were visualized by Annexin-V and propidium iodide (PI) staining. Implantation and postimplantation development of embryos were measured by in vitro development analysis. RESULTS: TUNEL staining and Annexin-V/PI staining showed that genistein dose-dependently increased apoptosis in mouse blastocysts, while daidzein, another soy isoflavone, had no such effect. The pretreatment of the blastocysts with genistein caused fewer cells than the control group and this effect was primary in the inner cell mass. The genistein-pretreated blastocysts showed normal levels of implantation on culture dishes in vitro, but significantly fewer genistein-pretreated embryos reached the later stages of embryonic development versus the controls, with many of the former embryos dying at relatively early stages of development. In addition, genistein treatment decreased the development of morulas into blastocysts, and dietary genistein was found to induce cell apoptosis and decrease cell proliferation in an animal assay model of embryogenesis. CONCLUSIONS: Our results collectively indicate that genistein treatment of mouse blastocysts induces apoptosis, decreases cell numbers, retards early postimplantation blastocyst development, and increases early-stage blastocyst death in vitro, while dietary genistein appears to negatively affect mouse embryonic development in vivo by inducing cell apoptosis and inhibiting cell proliferation. These novel findings provide important new insights into the effect of genistein on mouse blastocysts.


Subject(s)
Blastocyst/drug effects , Embryonic Development/drug effects , Genistein/pharmacology , Animals , Apoptosis/drug effects , Blastocyst/physiology , Chorionic Gonadotropin/biosynthesis , Female , Mice , Mice, Inbred ICR , Receptors, Estrogen/drug effects
7.
J Cell Biochem ; 100(4): 1056-69, 2007 Mar 01.
Article in English | MEDLINE | ID: mdl-17131386

ABSTRACT

Methylglyoxal (MG) is a reactive dicarbonyl compound endogenously produced mainly from glycolytic intermediates. MG is cytotoxic through induction of cell death, and elevated MG levels in diabetes patients are believed to contribute to diabetic complications. In this report, we show for the first time that MG treatment triggers apoptosis in human osteoblasts. We further show that MG-induced apoptosis of osteoblasts involves specific apoptotic biochemical changes, including oxidative stress, c-Jun N-terminal kinase (JNK) activation, mitochondrial membrane potential changes, cytochrome C release, increased Bax/Bcl-2 protein ratios, and activation of caspases (caspase-9, caspase-3) and p21-activated protein kinase 2 (PAK2). Treatment of osteoblasts with SP600125, a JNK-specific inhibitor, led to a reduction in MG-induced apoptosis and decreased activation of caspase-3 and PAK2, indicating that JNK activity is upstream of these events. Experiments using anti-sense oligonucleotides against PAK2 further showed that PAK2 activation is required for MG-induced apoptosis in osteoblasts. Interestingly, we also found that MG treatment triggered nuclear translocation of NF-kappaB, although the precise regulatory role of NF-kappaB activation in MG-induced apoptosis remains unclear. Lastly, we examined the effect of MG on osteoblasts in vivo, and found that exposure of rats to dietary water containing 100-200 microM MG caused bone mineral density (BMD) loss. Collectively, these results reveal for the first time that MG treatment triggers apoptosis in osteoblasts via specific apoptotic signaling, and causes BMD loss in vivo.


Subject(s)
Apoptosis/drug effects , Caspase 3/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Osteoblasts/drug effects , Pyruvaldehyde/pharmacology , Animals , Bone Density/drug effects , Caspase 9/metabolism , Cells, Cultured , Enzyme Activation/drug effects , Female , Humans , Immunoblotting , NF-kappa B/metabolism , Oligonucleotides, Antisense/genetics , Osteoblasts/cytology , Osteoblasts/metabolism , Oxidative Stress/drug effects , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Transfection , p21-Activated Kinases
8.
Toxicol Lett ; 167(3): 191-200, 2006 Dec 15.
Article in English | MEDLINE | ID: mdl-17049762

ABSTRACT

Quantum dots (QDs) may be useful as novel luminescent markers, but their cytotoxicity has not been fully investigated. In this report, we demonstrate that CdSe-core QDs can induce apoptotic biochemical changes, including JNK activation, loss of mitochondrial membrane potential, mitochondrial release of cytochrome c and activation of caspase-9 and caspase-3 in the IMR-32 human neuroblastoma cell line. Importantly, treatment of IMR-32 cells with CdSe-core QD triggered an increase in reactive oxygen species (ROS) and inhibited survival-related signaling events, such as decreased Ras and Raf-1 protein expression and decreased ERK activation. These apoptotic biochemical changes were not detected in cells treated with ZnS-coated CdSe QDs. Collectively, these results demonstrate that CdSe-core QD treatment of IMR-32 cells induced JNK activation and mitochondrial-dependent apoptotic processes while inhibiting Ras-->ERK survival signaling and that a ZnS coating could effectively reduce QD cytotoxicity.


Subject(s)
Apoptosis/drug effects , Cadmium Compounds/toxicity , Mitochondria/physiology , Neuroblastoma/pathology , Quantum Dots , Selenium Compounds/toxicity , Caspase 3/physiology , Caspase 9/physiology , Cell Line, Tumor , Cell Survival/drug effects , HSP90 Heat-Shock Proteins/biosynthesis , Humans , JNK Mitogen-Activated Protein Kinases/physiology , Reactive Oxygen Species , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...