Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 79
Filtrar
1.
Drug Chem Toxicol ; 45(4): 1449-1460, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33106064

RESUMEN

The current study has focused on the effects of enniatin B (ENN B, a major mycotoxin produced by Fusarium fungi) on early embryonic development. In in vitro analysis, mouse blastocysts were incubated in medium with ENN B (0-40 µM) or 0.5% DMSO (control group) for 24 hours. In an animal study, blastocysts were collected from mice which were intravenously injected with ENN B (1, 3, 5, and 7mg/kg body weight/day) for 4 days in order to analyze apoptosis and necrosis via Annexin V/PI staining assay; and proliferation using dual differential staining. Exposure to low ENN B concentration (10 µM in vitro and 3 mg/kg/day in vivo) promoted Reactive Oxygen Species (ROS) generation and apoptosis in the Inner Cell Mass (ICM), the mass of cells inside the blastocyst, impairing post-implantation development alone. On the other hand, exposure to a higher ENN B concentration (40 µM in vitro and 7 mg/kg/day in vivo) induced ROS generation and decreased in intracellular ATP which encouraged necrotic processes in both trophectoderm (TE) and ICM of blastocysts leading to impaired implantation and post-implantation development. Moreover, 5 and 7 mg/kg/day ENN B intraperitoneal injection to female mice for 4 days has caused downregulation of CXCL1, IL-1ß and IL-8 expressions and increased ROS generation in the liver of newborn mice. Over all, ENN B can induce apoptosis and/or necrosis depending on the treatment dosage in mouse blastocysts. ENN B-induced necrosis in blastocysts may exert long-term harmful effects on next-generation newborns.


Asunto(s)
Apoptosis , Blastocisto , Depsipéptidos , Desarrollo Embrionario , Necrosis , Animales , Blastocisto/efectos de los fármacos , Depsipéptidos/toxicidad , Desarrollo Embrionario/efectos de los fármacos , Femenino , Ratones , Necrosis/inducido químicamente , Embarazo , Especies Reactivas de Oxígeno/metabolismo
2.
Environ Toxicol ; 34(9): 1001-1012, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31112002

RESUMEN

Ginsenoside Rb1 (GRb1), the major saponin component of ginseng root, has a wide range of therapeutic applications for various diseases. Previously, our group showed that GRb1 triggers ROS-mediated apoptotic cascades in mouse blastocysts, leading to decreased cell viability and impairment of pre- and postimplantation embryonic development, both in vitro and in vivo. In this study, we further found that GRb1 exerted dose-dependent effects on oocyte maturation and sequent development in vitro. Oocytes preincubated with 25 µg/mL GRB1 displayed significantly enhanced maturation and in vitro fertilization (IVF) rates, along with progression of subsequent embryonic development. In contrast, treatment with 50 and 100 µg/mL GRB1 led to impairment of mouse oocyte maturation, decreased IVF rates, and injurious effects on subsequent embryonic development. In vivo, intravenous injection of 1 mg/kg body weight GRb1 significantly promoted mouse oocyte maturation, IVF, and early-stage embryo development after fertilization while administration of 5 mg/kg body weight GRb1 led to a marked decrease in oocyte maturation and IVF rates concomitant with impairment of early embryonic development in our animal model. In terms of the mechanisms underlying the regulatory effects of GRb1 demonstrated increased intracellular reactive oxygen species (ROS) production and apoptosis in the 100 µg/mL GRb1 treatment group. However, we observed a significant decrease in total intracellular ROS content and inhibition of apoptosis events in the 25 µg/mL GRb1 treatment group, signifying that the intracellular ROS content serves as a key upstream regulator of GRb1 that influences its dose-dependent beneficial or deleterious effects on oocyte maturation and sequent embryonic development. For further clarification of the mechanisms underlying GRb1-triggered injurious effects, oocytes were pretreated with Ac-DEVD-CHO, a caspase-3-specific inhibitor, which effectively blocked injury to oocyte maturation, fertilization, and sequent development. In sum, study findings highlight the potential involvement of p53-, p21-, and caspase-3-dependent regulatory signaling cascades in GRb1-mediated apoptotic processes.


Asunto(s)
Desarrollo Fetal/efectos de los fármacos , Ginsenósidos , Oocitos/efectos de los fármacos , Oogénesis/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Blastocisto/efectos de los fármacos , Blastocisto/metabolismo , Relación Dosis-Respuesta a Droga , Desarrollo Embrionario/efectos de los fármacos , Femenino , Fertilización In Vitro , Ginsenósidos/administración & dosificación , Ginsenósidos/farmacología , Ginsenósidos/toxicidad , Técnicas In Vitro , Ratones , Ratones Endogámicos ICR , Oocitos/metabolismo , Embarazo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal
3.
Environ Toxicol ; 34(5): 573-584, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30698892

RESUMEN

Ochratoxin A (OTA), a mycotoxin constituent of a range of food commodities, including coffee, wine, beer, grains, and spices, exerts toxicological and pathological effects in vivo, such as nephrotoxicity, hepatotoxicity, and immunotoxicity. In a previous report, we highlighted the potential of OTA to induce apoptosis via reactive oxygen species (ROS) generation in mouse blastocysts that led to impaired preimplantation and postimplantation embryo development in vitro and in vivo. Here, we have shown that liquiritigenin (LQ), a type of flavonoid isolated from Glycyrrhiza radix, effectively protects against OTA-mediated apoptosis and inhibition of cell proliferation in mouse blastocysts. Preincubation of blastocysts with LQ clearly prevented OTA-triggered impairment of preimplantation and postimplantation embryonic development and fetal weight loss, both in vitro and in vivo. Detailed investigation of regulatory mechanisms revealed that OTA mediated apoptosis and embryotoxicity through ROS generation, loss of mitochondrial membrane potential (MMP), and activation of caspase-9 and caspase-3, which were effectively prevented by LQ. The embryotoxic effects of OTA were further validated in an animal model in vivo. Intravenous injection of dams with OTA (3 mg/kg/day) led to apoptosis of blastocysts, impairment of embryonic development from zygote to blastocyst stage and decrease in day 18 fetal weight. Notably, preinjection of dams with LQ (5 mg/kg/day) effectively prevented OTA-induced apoptosis and toxic effects on embryo development. Our collective results clearly demonstrate that OTA exposure via injection has the potential to damage preimplantation and postimplantation embryonic development against which LQ has a protective effect.


Asunto(s)
Apoptosis/efectos de los fármacos , Blastocisto/efectos de los fármacos , Desarrollo Embrionario/efectos de los fármacos , Flavanonas/farmacología , Ocratoxinas/toxicidad , Estrés Oxidativo/efectos de los fármacos , Sustancias Protectoras/farmacología , Animales , Blastocisto/metabolismo , Blastocisto/patología , Proliferación Celular/efectos de los fármacos , Femenino , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Embarazo
4.
Environ Toxicol ; 34(1): 48-59, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30259633

RESUMEN

Enniatins are mycotoxins of Fusarium fungi that naturally exist as mixtures of cyclic depsipeptides. Previous reports have documented hazardous effects of enniatins on cells, such as apoptosis. However, their effects on pre- and post-implantation embryonic development require further clarification. Here, we showed for the first time that enniatin B1 (ENN B1) exerts cytotoxic effects on mouse blastocyst-stage embryos and induces intracellular oxidative stress and immunotoxicity in mouse fetuses. Co-incubation of blastocysts with ENN B1 triggered significant apoptosis and led to a decrease in total cell number predominantly through loss of inner cell mass. In addition, ENN B1 appeared to exert hazardous effects on pre and postimplantation embryo development potential in an in vitro development assay. Treatment of blastocysts with 1-10 µM ENN B1 led to increased resorption of post-implantation embryos and decreased fetal weight in the embryo transfer assay in a dose-dependent manner. Importantly, in an in vivo model, intravenous injection with ENN B1 (1, 3, and 5 mg/kg body weight/d) for 4 days resulted in apoptosis of blastocyst-stage embryos and impairment of embryonic development from the zygote to blastocyst stage, subsequent degradation of embryos, and further decrease in fetal weight. Intravenous injection with 5 mg/kg body weight/d ENN B1 additionally induced a significant increase in total reactive oxygen species (ROS) content and transcription levels of genes encoding antioxidant proteins in mouse fetal liver. Moreover, ENN B1 triggered apoptosis through ROS generation and strategies to prevent apoptotic processes effectively rescued ENN B1-mediated hazardous effects on embryonic development. Transcription levels of CXCL1, IL-1ß, and IL-8 related to innate immunity were downregulated after intravenous injection of ENN B1. These results collectively highlight the potential of ENN B1 to exert cytotoxic effects on embryos as well as oxidative stress and immunotoxicity during mouse embryo development.


Asunto(s)
Blastocisto/efectos de los fármacos , Blastocisto/inmunología , Citotoxicidad Inmunológica/efectos de los fármacos , Depsipéptidos/toxicidad , Desarrollo Embrionario/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Blastocisto/citología , Depsipéptidos/farmacología , Embrión de Mamíferos , Desarrollo Embrionario/inmunología , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Micotoxinas/farmacología , Micotoxinas/toxicidad , Embarazo , Especies Reactivas de Oxígeno/metabolismo
5.
Environ Toxicol ; 33(10): 1039-1049, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29964317

RESUMEN

Silver nanoparticles (AgNPs) are antibacterial materials widely used in numerous products and medical supplies. Previously, we showed that AgNPs trigger apoptotic processes in mouse blastocysts, leading to a decrease in cell viability and impairment of preimplantation and postimplantation embryonic development in vitro and in vivo. In the present study, we further investigated the hazardous effects of AgNPs on mouse oocyte maturation, in vitro fertilization (IVF), and subsequent preimplantation and postimplantation development in vitro and in vivo. Data from in vitro experiments revealed that AgNPs impair mouse oocyte maturation, decrease IVF rates, and induce injury effects on subsequent embryonic development to a significant extent. In an animal model, intravenous injection of AgNPs (5 mg/kg body weight) led to a significant decrease in mouse oocyte maturation and IVF concomitant with impairment of early embryonic development in vivo. Importantly, pretreatment with N-acetylcysteine effectively prevented AgNP-triggered reactive oxygen species (ROS) production and apoptosis, clearly suggesting a critical role of ROS as an upstream initiator or key regulator of AgNP-induced hazardous effects on oocyte maturation and sequent embryonic development. Furthermore, preincubation of oocytes with Ac-DEVD-cho, a caspase-3-specific inhibitor, effectively prevented hazardous effects, highlighting the potential involvement of caspase-dependent apoptotic signaling cascades in AgNP-mediated events. Expression levels of p53 and p21 of blastocysts were upregulated upon preincubation of mouse oocytes with AgNPs. Our collective results imply that cell apoptosis in mouse blastocysts derived from the AgNP-pretreated oocytes via intracellular ROS generation, which is further mediated through p53-, p21-, and caspase-3-dependent regulatory mechanisms.


Asunto(s)
Apoptosis/efectos de los fármacos , Desarrollo Fetal/efectos de los fármacos , Nanopartículas del Metal/química , Oocitos/efectos de los fármacos , Plata/química , Animales , Blastocisto/efectos de los fármacos , Blastocisto/metabolismo , Caspasa 3/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/antagonistas & inhibidores , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Desarrollo Embrionario/efectos de los fármacos , Femenino , Fertilización In Vitro/efectos de los fármacos , Nanopartículas del Metal/toxicidad , Ratones , Ratones Endogámicos ICR , Embarazo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Proteína p53 Supresora de Tumor/antagonistas & inhibidores , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Regulación hacia Arriba/efectos de los fármacos
6.
Environ Toxicol ; 33(3): 280-294, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29168595

RESUMEN

Berberine, an isoquinoline alkaloid isolated from several traditional Chinese herbal medicines, has been shown to suppress growth and induce apoptosis in some tumor cell lines. However, berberine has also been reported to attenuate H2 O2 -induced oxidative injury and apoptosis. The basis for these ambiguous effects of berberine-triggering or preventing apoptosis-has not been well characterized to date. In the current investigation, we examined whether berberine exerts cytotoxic effects on mouse embryos at the blastocyst stage and affects subsequent embryonic development in vitro and in vivo. Treatment of blastocysts with berberine (2.5-10 µM) induced a significant increase in apoptosis and a corresponding decrease in trophectoderm cell number. Moreover, the implantation success rate of blastocysts pretreated with berberine was lower than that of their control counterparts. Pretreatment with berberine was also associated with increased resorption of postimplantation embryos and decreased fetal weight. In an animal model, intravenous injection of berberine (2, 4, or 6 mg/kg body weight/d) for 4 days resulted in apoptosis of blastocyst cells and early embryonic developmental injury. Berberine-induced injury of mouse blastocysts appeared to be attributable to oxidative stress-triggered intrinsic apoptotic signaling processes that impaired preimplantation and postimplantation embryonic development. Taken together, our results clearly demonstrate that berberine induces apoptosis and retards early preimplantation and postimplantation development of mouse embryos, both in vitro and in vivo.


Asunto(s)
Apoptosis/efectos de los fármacos , Berberina/toxicidad , Desarrollo Embrionario/efectos de los fármacos , Estrés Oxidativo , Animales , Blastocisto/citología , Blastocisto/efectos de los fármacos , Recuento de Células , Implantación del Embrión/efectos de los fármacos , Femenino , Ratones , Ratones Endogámicos ICR , Embarazo , Transducción de Señal
7.
Environ Toxicol ; 32(7): 1937-1951, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28371286

RESUMEN

Ginsenoside Rg1, which is the most abundant compound found in Asian ginseng (Panax ginseng), has demonstrated various pharmacological actions, including neuroprotective, immune-stimulatory, and antidiabetic effects. Pregnant women, especially in the Asian community, consume ginseng as a nutritive supplement. Thus, the effects of ginsenoside-Rg1 on embryonic development need to be investigated, such as in a mouse model. As previous investigations have found that ginsenoside Rg1 appears to either trigger or prevent apoptosis in different cell lines, the effects of this agent on apoptosis remain to be clarified. In this study, we investigated whether ginsenoside Rg1 exerts a hazardous effect on mouse blastocysts and/or affects subsequent embryonic development in vitro and in vivo. Blastocysts treated with 25-100 µM ginsenoside Rg1 exhibited significant induction of apoptosis and a corresponding decrease in the inner cell mass (ICM) cell number. Importantly, the implantation rate was lower among ginsenoside Rg1-treated blastocysts compared to untreated controls. Moreover, embryo transfer assays revealed that blastocysts treated with 100 µM ginsenoside Rg1 exhibited increased resorption of postimplantation embryos and decreased weight among surviving fetuses. In vivo, intravenous injection of mice with ginsenoside Rg1 (2, 4, or 6 mg/kg body weight/day) for 4 days was associated with increased apoptosis of blastocyst-stage embryos and negatively impacted early embryonic development. Further experiments revealed that these effects may reflect the ability of ginsenoside Rg1 to trigger oxidative stress-mediated intrinsic apoptotic signaling. Our in vitro results indicate that ginsenoside Rg1 treatment increases intracellular oxidative stress, decreases mitochondrial membrane potential, increases the Bax/Bcl-2 ratio, and activates caspase-9 and caspase-3, but not caspase-8. Taken together, our study results strongly suggest that ginsenoside Rg1 induces apoptosis and impairs the early preimplantation and postimplantation development of mouse embryos, both in vitro and in vivo.


Asunto(s)
Apoptosis/efectos de los fármacos , Medicamentos Herbarios Chinos/toxicidad , Desarrollo Embrionario/efectos de los fármacos , Ginsenósidos/toxicidad , Animales , Blastocisto/citología , Blastocisto/efectos de los fármacos , Caspasa 3/metabolismo , Caspasa 8/metabolismo , Caspasa 9/metabolismo , Recuento de Células , Activación Enzimática , Femenino , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones Endogámicos ICR , Estrés Oxidativo/efectos de los fármacos , Embarazo , Transducción de Señal/efectos de los fármacos
8.
Environ Toxicol ; 32(8): 1990-2003, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27640876

RESUMEN

Ginsenoside Rb1, the major saponin component of ginseng root, has a wide range of therapeutic application. Previous studies have established that ginsenoside Rb1 inhibits the cell cycle and induces apoptosis. However, its side-effects, particularly those on embryonic development, have not been well characterized to date. In the current study, we examined whether ginsenoside Rb1 exerts a cytotoxic effect on mouse embryos at the blastocyst stage, and affects subsequent embryonic development in vitro and in vivo. Blastocysts treated with 25-100 µg mL-1 ginsenoside Rb1 exhibited significantly increased apoptosis and a corresponding decrease in total cell number. Notably, the implantation success rate of blastocysts pretreated with ginsenoside Rb1 was lower than that of their control counterparts. Moreover, in vitro treatment with 25-100 µg mL-1 ginsenoside Rb1 was associated with increased resorption of post-implantation embryos and decreased fetal weight. In an in vivo model, intravenous injection with ginsenoside Rb1 (1, 3, 5 mg kg-1 body weight/day) for 4 days resulted in apoptosis of blastocyst stage embryos and early embryonic developmental injury. In addition, ginsenoside Rb1 appeared to induce injury in mouse blastocysts through oxidative stresses-triggered intrinsic apoptotic signaling processes to impair sequent embryonic development. The collective results strongly indicate that ginsenoside Rb1 induces apoptosis and retards early pre- and post-implantation development of mouse embryos, both in vitro and in vivo. © 2016 Wiley Periodicals, Inc. Environ Toxicol 32: 1990-2003, 2017.


Asunto(s)
Apoptosis/efectos de los fármacos , Desarrollo Embrionario/efectos de los fármacos , Ginsenósidos/toxicidad , Estrés Oxidativo/efectos de los fármacos , Animales , Blastocisto/efectos de los fármacos , Implantación del Embrión/efectos de los fármacos , Femenino , Masculino , Ratones Endogámicos ICR , Embarazo , Transducción de Señal/efectos de los fármacos
9.
Int J Mol Sci ; 18(9)2017 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-28930172

RESUMEN

Rhein, a glucoside chemical compound found in a traditional Chinese medicine derived from the roots of rhubarb, induces cell apoptosis and is considered to have high potential as an antitumor drug. Several previous studies showed that rhein can inhibit cell proliferation and trigger mitochondria-related or endoplasmic reticulum (ER) stress-dependent apoptotic processes. However, the side effects of rhein on pre- and post-implantation embryonic development remain unclear. Here, we show that rhein has cytotoxic effects on blastocyst-stage mouse embryos and induces oxidative stress and immunotoxicity in mouse fetuses. Blastocysts incubated with 5-20 µM rhein showed significant cell apoptosis, as well as decreases in their inner cell mass cell numbers and total cell numbers. An in vitro development assay showed that rhein affected the developmental potentials of both pre- and post-implantation embryos. Incubation of blastocysts with 5-20 µM rhein was associated with increased resorption of post-implantation embryos and decreased fetal weight in an embryo transfer assay. Importantly, in an in vivo model, intravenous injection of dams with rhein (1, 3, and 5 mg/kg body weight/day) for four days resulted in apoptosis of blastocyst-stage embryos, early embryonic developmental injury, and decreased fetal weight. Intravenous injection of dams with 5 mg/kg body weight/day rhein significantly increased the total reactive oxygen species (ROS) content of fetuses and the transcription levels of antioxidant proteins in fetal livers. Additional work showed that rhein induced apoptosis through ROS generation, and that prevention of apoptotic processes effectively rescued the rhein-induced injury effects on embryonic development. Finally, the transcription levels of the innate-immunity related genes, CXCL1, IL-1ß and IL-8, were down-regulated in the fetuses of dams that received intravenous injections of rhein. These results collectively show that rhein has the potential to induce embryonic cytotoxicity and induce oxidative stress and immunotoxicity during the development of mouse embryos.


Asunto(s)
Antraquinonas/farmacología , Apoptosis/efectos de los fármacos , Blastocisto/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Desarrollo Embrionario/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Animales , Relación Dosis-Respuesta a Droga , Implantación del Embrión/efectos de los fármacos , Embrión de Mamíferos/efectos de los fármacos , Embrión de Mamíferos/embriología , Embrión de Mamíferos/metabolismo , Inhibidores Enzimáticos/farmacología , Femenino , Ratones Endogámicos ICR , Embarazo , Especies Reactivas de Oxígeno/metabolismo
10.
Int J Mol Sci ; 18(12)2017 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-29186930

RESUMEN

The mycotoxin citrinin (CTN), a natural contaminant in foodstuffs and animal feeds, exerts cytotoxic and genotoxic effects on various mammalian cells and embryos. A previous investigation by our group revealed potentially hazardous effects of CTN on mouse oocyte maturation and pre- and post-implantation embryo development via the induction of apoptosis. The present study showed that CTN induces apoptosis and inhibits cell proliferation in the inner cell mass of mouse blastocysts. Notably, we observed for the first time that both these effects are suppressed by liquiritigenin (LQ). LQ is a type of flavonoid isolated from Glycyrrhiza radix with several biochemical and pharmacological activities, including antioxidant and anti-inflammatory properties. The preincubation of blastocysts with LQ clearly prevented CTN-induced disruption of pre- and post-implantation embryonic development and fetal weight loss, both in vitro and in vivo. CTN-induced damage processes directly promoted reactive oxygen species (ROS) generation, loss of mitochondrial membrane potential (MMP) and activation of caspase-9 and caspase-3, which were effectively blocked by LQ. Moreover, in an animal model, intravenous injection of dams with CTN (3 mg/kg/day) triggered apoptosis of blastocysts, disruption of embryonic development from the zygote to the blastocyst stage and a decrease in fetal weight. Pre-injection with LQ (5 mg/kg/day) effectively reduced apoptosis and impaired the cytotoxic effects of CTN on development. Our in vivo findings further confirm that CTN exposure via injection has the potential to impair pre- and post-implantation development, leading to apoptosis and the suppression of sequent embryonic development, which can be effectively prevented by LQ.


Asunto(s)
Apoptosis , Blastocisto/efectos de los fármacos , Desarrollo Embrionario , Flavanonas/farmacología , Estrés Oxidativo , Animales , Blastocisto/metabolismo , Citrinina/toxicidad , Citoprotección , Femenino , Ratones , Ratones Endogámicos ICR
11.
Drug Chem Toxicol ; 39(4): 380-7, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26712108

RESUMEN

Previously we identified puerarin, an isoflavone compound, as a risk factor for normal embryonic development that triggers apoptotic processes in the inner cell mass of mouse blastocysts, leading to retardation of embryonic development and cell viability. In the current study, we investigated whether puerarin exerts deleterious effects on mouse oocyte maturation, in vitro fertilization (IVF) and subsequent pre- and post-implantation development, both in vitro and in vivo. Notably, puerarin caused significant impairment of these processes in vitro. Pre-incubation of oocytes with puerarin during in vitro maturation led to increased post-implantation embryo resorption and decreased mouse fetal weight. In an in vivo animal model, intravenous injection with or without puerarin (1, 3 and 5 mg/kg body weight/day) for 4 days caused a decrease in oocyte maturation and IVF, and led to deleterious effects on early embryonic development. Importantly, pre-incubation of oocytes with a caspase-3-specific inhibitor effectively blocked puerarin-triggered deleterious effects, clearly implying that embryonic injury induced by puerarin is mediated by a caspase-dependent apoptotic mechanism. These results clearly demonstrate that puerarin has deleterious effects on mouse oocyte maturation, fertilization and subsequent embryonic development in vitro and in vivo.


Asunto(s)
Apoptosis/efectos de los fármacos , Fertilización/efectos de los fármacos , Desarrollo Fetal/efectos de los fármacos , Técnicas de Maduración In Vitro de los Oocitos , Isoflavonas/toxicidad , Oocitos/efectos de los fármacos , Vasodilatadores/toxicidad , Animales , Desarrollo Embrionario/efectos de los fármacos , Femenino , Ratones Endogámicos ICR , Embarazo
12.
Environ Toxicol ; 31(6): 724-35, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25504763

RESUMEN

We previously reported that ochratoxin A (OTA), a mycotoxin found in many foods worldwide, causes nephrotoxicity, hepatotoxicity, and immunotoxicity, and is a risk factor for abnormal embryonic development. More specifically, OTA triggers apoptotic processes in the inner cell mass of mouse blastocysts, decreasing cell viability and embryonic development. In the current study, we investigated the deleterious effects of OTA on mouse oocyte maturation, in vitro fertilization (IVF), and subsequent pre- and postimplantation development both in vitro and in vivo. Notably, OTA significantly impaired mouse oocyte maturation, decreased IVF rates, and inhibited subsequent embryonic development in vitro. Preincubation of oocytes with OTA during in vitro maturation increased postimplantation embryonic resorption and decreased mouse fetal weight. In an in vivo animal model, provision of 1-10 µM OTA in the drinking water or intravenous injection of 1 or 2 mg/kg body weight of OTA decreased oocyte maturation and IVF, and had deleterious effects on early embryonic development. Importantly, preincubation of oocytes with a caspase-3-specific inhibitor effectively blocked these OTA-triggered deleterious effects, suggesting that the embryonic injury induced by OTA is mediated via a caspase-dependent apoptotic mechanism. Furthermore, OTA upregulated the levels of p53 and p21 in blastocyst cells derived from OTA-pretreated oocytes, indicating that such cells undergo apoptosis via p53-, p21-, and caspase-3-dependent regulatory mechanisms. This could have deleterious effects on embryonic implantation and fetal survival rates, as seen in our animal models. © 2014 Wiley Periodicals, Inc. Environ Toxicol 31: 724-735, 2016.


Asunto(s)
Apoptosis/efectos de los fármacos , Carcinógenos/toxicidad , Fertilización/efectos de los fármacos , Ocratoxinas/toxicidad , Oocitos/efectos de los fármacos , Animales , Desarrollo Embrionario/efectos de los fármacos , Femenino , Fertilización In Vitro , Ratones , Ratones Endogámicos ICR , Oocitos/citología
13.
Drug Chem Toxicol ; 38(4): 469-76, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25721892

RESUMEN

Previously, we reported that dillapiole, a phenylpropanoid with antileishmanial, anti-inflammatory, antifungal and acaricidal activities, is a risk factor for normal embryonic development that triggers apoptotic processes in the inner cell mass of mouse blastocysts, leading to impaired embryonic development and cell viability. In the current study, we investigated the deleterious effects of dillapiole on mouse oocyte maturation, in vitro fertilization (IVF) and subsequent pre- and post-implantation development, both in vitro and in vivo. Notably, dillapiole induced significant impairment of mouse oocyte maturation, decrease in the IVF rate and inhibition of subsequent embryonic development in vitro. Pre-incubation of oocytes with dillapiole during in vitro maturation led to an increase in post-implantation embryo resorption and decrease in mouse fetal weight. In an in vivo animal model, 2.5, 5 or 10 µM dillapiole provided in drinking water caused a decrease in oocyte maturation and IVF, and led to deleterious effects on early embryonic development. Importantly, pre-incubation of oocytes with a caspase-3-specific inhibitor effectively blocked dillapiole-triggered deleterious effects, clearly implying that embryonic injury induced by dillapiole is mediated via a caspase-dependent apoptotic mechanism. To the best of our knowledge, this is the first study to establish the impact of dillapiole on maturation of mouse oocytes, fertilization and sequential embryonic development.


Asunto(s)
Compuestos Alílicos/toxicidad , Apoptosis/efectos de los fármacos , Dioxoles/toxicidad , Fertilización In Vitro/efectos de los fármacos , Oocitos/efectos de los fármacos , Compuestos Alílicos/administración & dosificación , Animales , Blastocisto/efectos de los fármacos , Blastocisto/metabolismo , Caspasa 3/metabolismo , Dioxoles/administración & dosificación , Relación Dosis-Respuesta a Droga , Desarrollo Embrionario/efectos de los fármacos , Femenino , Desarrollo Fetal/efectos de los fármacos , Peso Fetal/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos ICR , Oocitos/metabolismo , Embarazo
14.
Environ Toxicol ; 30(8): 946-55, 2015 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-24677673

RESUMEN

Previously, we reported that sanguinarine, a phytoalexin with antimicrobial, anti-oxidant, anti-inflammatory and pro-apoptotic effects, is a risk factor for normal embryonic development that triggers apoptotic processes in the inner cell mass of mouse blastocysts, causing decreased embryonic development and cell viability. In the current study, we investigated the deleterious effects of sanguinarine on mouse oocyte maturation, in vitro fertilization (IVF), and subsequent pre- and postimplantation development both in vitro and in vivo. Notably, sanguinarine significantly impaired mouse oocyte maturation, decreased IVF rates, and inhibited subsequent embryonic development in vitro. Preincubation of oocytes with sanguinarine during in vitro maturation induced an increase in postimplantation embryo resorption and a decrease in mouse fetal weight. In an in vivo animal model, 1 to 5 µM sanguinarine, provided in drinking water, caused a decrease in oocyte maturation and IVF, and led to deleterious effects on early embryonic development. Importantly, preincubation of oocytes with a caspase-3-specific inhibitor effectively blocked sanguinarine-triggered deleterious effects, clearly implying that embryonic injury induced by sanguinarine is mediated by a caspase-dependent apoptotic mechanism.


Asunto(s)
Apoptosis/efectos de los fármacos , Benzofenantridinas/toxicidad , Fertilización In Vitro/efectos de los fármacos , Desarrollo Fetal/efectos de los fármacos , Isoquinolinas/toxicidad , Oocitos/efectos de los fármacos , Animales , Blastocisto/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Caspasa 3/metabolismo , Agua Potable , Desarrollo Embrionario/efectos de los fármacos , Femenino , Ratones , Ratones Endogámicos ICR , Oogénesis/efectos de los fármacos , Embarazo
15.
Drug Chem Toxicol ; 37(3): 247-54, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24164432

RESUMEN

Catechins, a family of polyphenols found in tea, evoke various responses, including cell death. However, the side effects of these compounds, particularly those on embryonic development, have not been characterized in detail. A previous study by our group showed that (-)-epigallocatechin-3-gallate (EGCG), a catechin highly abundant in green tea, induces different cell-death modes in MCF-7 cells, depending on the treatment dosage. In the current study, we examined the effects of EGCG on mouse embryos at the blastocyst stage, subsequent embryonic attachment and outgrowth in vitro and in vivo implantation by embryo transfer. Blastocysts treated with 25-50 µM of EGCG exhibited a significant increase in apoptosis and a corresponding decrease in total cell number. Notably, the implantation success rate of blastocysts pretreated with EGCG was lower than that of their control counterparts. Moreover, in vitro treatment with 25-50 µM of EGCG led to increased resorption of postimplantation embryos and decreased fetal weight. EGCG appeared to induce injury in mouse blastocysts through intrinsic apoptotic signaling processes to impair sequent embryonic development. These results collectively highlight the potential of EGCG to induce embryonic cytotoxicity.


Asunto(s)
Apoptosis/efectos de los fármacos , Blastocisto/efectos de los fármacos , Catequina/análogos & derivados , Desarrollo Embrionario/efectos de los fármacos , Animales , Catequina/administración & dosificación , Catequina/toxicidad , Relación Dosis-Respuesta a Droga , Implantación del Embrión/efectos de los fármacos , Transferencia de Embrión , Femenino , Humanos , Células MCF-7 , Ratones , Ratones Endogámicos ICR , Embarazo , Transducción de Señal/efectos de los fármacos
16.
Int J Mol Sci ; 15(6): 10751-65, 2014 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-24933639

RESUMEN

We examined the cytotoxic effects of dillapiole, a phenylpropanoid with antileishmanial, anti-inflammatory, antifungal, and acaricidal activities, on the blastocyst stage of mouse embryos, subsequent embryonic attachment and outgrowth in vitro, and in vivo implantation via embryo transfer. Blastocysts treated with 2.5-10 µM dillapiole exhibited a significant increase in apoptosis and corresponding decrease in total cell number. Notably, the implantation success rates of blastocysts pretreated with dillapiole were lower than those of their control counterparts. Moreover, in vitro treatment with 2.5-10 µM dillapiole was associated with increased resorption of post-implantation embryos and decreased fetal weight. Our results collectively indicate that dillapiole induces apoptosis and retards early post-implantation development, both in vitro and in vivo. However, the extent to which this organic compound exerts teratogenic effects on early human development is not known at present. Further studies are required to establish effective protection strategies against the cytotoxic effects of dillapiole.


Asunto(s)
Compuestos Alílicos/toxicidad , Blastocisto/citología , Dioxoles/toxicidad , Desarrollo Embrionario/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Blastocisto/metabolismo , Proliferación Celular/efectos de los fármacos , Transferencia de Embrión , Embrión de Mamíferos/citología , Embrión de Mamíferos/efectos de los fármacos , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR
17.
Toxicol Res (Camb) ; 13(1): tfad122, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38162594

RESUMEN

Methylmercury (MeHg) is a potent toxin that exerts deleterious effects on human health via environmental contamination. Significant effects of MeHg on neuronal development in embryogenesis have been reported. Recently, our group demonstrated that MeHg exerts toxic effects on pre- and post-implantation embryonic development processes from zygote to blastocyst stage. Our results showed that MeHg impairs embryo development by induction of apoptosis through reactive oxygen species (ROS) generation that triggers caspase-3 cleavage and activation, which, in turn, stimulates p21-activated kinase 2 (PAK2) activity. Importantly, ROS were identified as a key upstream regulator of apoptotic events in MeHg-treated blastocysts. Data from the current study further confirmed that MeHg exerts hazardous effects on cell proliferation, apoptosis, implantation, and pre- and post-implantation embryo development. Notably, MeHg-induced injury was markedly prevented by co-culture with adipose-derived mesenchymal stem cells (ADMSCs) in vitro. Furthermore, ADMSC injection significantly reduced MeHg-mediated deleterious effects on embryo, placenta, and fetal development in vivo. Further investigation of the regulatory mechanisms by which co-cultured ADMSCs could prevent MeHg-induced impairment of embryo development revealed that ADMSCs effectively reduced ROS generation and its subsequent downstream apoptotic events, including loss of mitochondrial membrane potential and activation of caspase-3 and PAK2. The collective findings indicate that co-culture with mesenchymal stem cells (MSCs) or utilization of MSC-derived cell-conditioned medium offers an effective potential therapeutic strategy to prevent impairment of embryo development by MeHg.

18.
Environ Toxicol ; 28(8): 431-41, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21793156

RESUMEN

Methylglyoxal (MG) is a glucose metabolite. Diabetic patients have increased serum levels of MG, and MG is also implicated in tissue injury during embryonic development. In the present work, we show that MG induces apoptosis in the inner cell mass of mouse blastocysts and inhibits cell proliferation. Both effects are suppressed by resveratrol, a grape-derived phytoalexin with known antioxidant and anti-inflammatory properties. MG-treated blastocysts displayed lower levels of implantation (compared to controls) when plated on culture dishes in vitro and a reduced ability to proceed to later stages of embryonic development. Pretreatment with resveratrol prevented MG-induced disruption of embryonic development, both in vitro and in vivo. Further investigation of these processes revealed that MG directly promotes reactive oxygen species (ROS) generation, loss of mitochondrial membrane potential (MMP), and activation of caspase-3, whereas resveratrol effectively blocks MG-induced ROS production and the accompanying apoptotic biochemical changes. Our results collectively imply that MG triggers the mitochondrion-dependent apoptotic pathway via ROS generation, and the antioxidant activity of resveratrol prevents MG-induced toxicity.


Asunto(s)
Antioxidantes/farmacología , Apoptosis/efectos de los fármacos , Blastocisto/efectos de los fármacos , Piruvaldehído/metabolismo , Estilbenos/farmacología , Animales , Blastocisto/citología , Blastocisto/fisiología , Caspasa 3/metabolismo , Proliferación Celular/efectos de los fármacos , Desarrollo Embrionario/efectos de los fármacos , Activación Enzimática , Femenino , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Ratones Endogámicos ICR , Embarazo , Piruvaldehído/toxicidad , Especies Reactivas de Oxígeno/metabolismo , Resveratrol
19.
Environ Toxicol ; 28(4): 190-200, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21626648

RESUMEN

Although all-trans retinoic acid (RA), the oxidative metabolite of vitamin A, is essential for normal development, high levels are teratogenic in many species. RA results in immediate effects on the preimplantation embryo and on blastocyst development in vitro and in vivo. To further elucidate the cellular mechanisms of early postimplantation embryo development induced by RA, we present an embryonic cell line, B5, as a candidate system for the investigation of these processes. We used undifferentiated ES cells as the model, which is from the undifferentiated status to differentiated status [embryoid body (EB) formation] mimicking postimplantation embryo development (egg-cylinder stage of embryo formation) to clarify the cellular mechanism of action of RA in the implanted blastocysts and cell apoptosis following the series of exposures to differing RA concentrations. Using an in vitro model, we identified the impact of RA on undifferentiated embryonic stem (ES) cells, including inhibition of cell proliferation and induction of cell apoptosis. JNK, P-38 and caspase activation were shown in the nature of RA-triggered apoptotic signaling in ES cells. The carry-on influences of RA on the ES cell were shown in the formation of EB from the pretreated ES cells. RA resulted in apparent impact on undifferentiated ES cells in vitro, with increased numbers of apoptotic cells initially and inhibited cell proliferation, which led to decreased size of EB. The process of EB formation (mimicking the early postimplantation embryo development) is regulated by RA-induced apoptosis through the activation of caspase and P38 MAPK/JNK pathway.


Asunto(s)
Apoptosis/efectos de los fármacos , Caspasas/biosíntesis , Cuerpos Embrioides/efectos de los fármacos , Células Madre Embrionarias/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Tretinoina/toxicidad , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología , Animales , Diferenciación Celular/efectos de los fármacos , Línea Celular , Proliferación Celular/efectos de los fármacos , Cuerpos Embrioides/citología , Cuerpos Embrioides/metabolismo , Desarrollo Embrionario/efectos de los fármacos , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Inducción Enzimática , Femenino , Ratones , Embarazo
20.
Environ Toxicol ; 28(2): 87-97, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21462292

RESUMEN

α-Lipoic acid (LA) is a thiol with antioxidant properties that protects against oxidative stress-induced apoptosis. LA is absorbed from the diet, taken up by cells and tissues, and subsequently reduced to dihydrolipoic acid (DHLA). Recently, DHLA has been used as the hydrophilic nanomaterial preparations, and therefore, determination of its bio-safety profile is essential. In this article, we show that DHLA (50-100 µM) induces apoptotic processes in mouse embryonic stem cells (ESC-B5), but exerts no injury effects at treatment dosages below 50 µM. Higher concentrations of DHLA (50-100 µM) directly increased the reactive oxygen species (ROS) content in ESC-B5 cells, along with a significant increase in cytoplasmic free calcium and nitric oxide (NO) levels, loss of mitochondrial membrane potential (MMP), activation of caspases-9 and -3, and cell death. Pretreatment with NO scavengers suppressed the apoptotic biochemical changes induced by 100 µM DHLA and promoted the gene expression levels of p53 and p21 involved in apoptotic signaling. Our results collectively indicate that DHLA at concentrations of 50-100 µM triggers apoptosis of ESC-B5 cells, which involves both ROS and NO. Importantly, at doses of less than 50 µM (0-25 µM), DHLA does not exert hazardous effects on ESC-B5 cell properties, including viability, development and differentiation. These results provide important information in terms of dosage safety and biocompatibility of DHLA to facilitate its further use as a precursor for biomaterial preparation.


Asunto(s)
Apoptosis/efectos de los fármacos , Células Madre Embrionarias/efectos de los fármacos , Ácido Tióctico/análogos & derivados , Animales , Antioxidantes/metabolismo , Calcio/metabolismo , Caspasa 3/metabolismo , Caspasa 9/metabolismo , Células Madre Embrionarias/metabolismo , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Óxido Nítrico/metabolismo , Estrés Oxidativo/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Ácido Tióctico/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA