Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 249
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Int J Mol Sci ; 24(6)2023 Mar 10.
Article in English | MEDLINE | ID: mdl-36982431

ABSTRACT

Endocrine-disrupting chemicals (EDCs) have significant impacts on biological systems, and have been shown to interfere with physiological systems, especially by disrupting the hormone balance. During the last few decades, EDCs have been shown to affect reproductive, neurological, and metabolic development and function and even stimulate tumor growth. EDC exposure during development can disrupt normal development patterns and alter susceptibility to disease. Many chemicals have endocrine-disrupting properties, including bisphenol A, organochlorines, polybrominated flame retardants, alkylphenols, and phthalates. These compounds have gradually been elucidated as risk factors for many diseases, such as reproductive, neural, and metabolic diseases and cancers. Endocrine disruption has been spread to wildlife and species that are connected to the food chains. Dietary uptake represents an important source of EDC exposure. Although EDCs represent a significant public health concern, the relationship and specific mechanism between EDCs and diseases remain unclear. This review focuses on the disease-EDC relationship and the disease endpoints associated with endocrine disruption for a better understanding of the relationship between EDCs-disease and elucidates the development of new prevention/treatment opportunities and screening methods.


Subject(s)
Endocrine Disruptors , Animals , Endocrine Disruptors/toxicity , Endocrine Disruptors/chemistry , Reproduction , Animals, Wild , Endocrine System , Nervous System
2.
Int J Mol Sci ; 23(3)2022 Jan 29.
Article in English | MEDLINE | ID: mdl-35163501

ABSTRACT

There is growing concern regarding the health and safety issues of endocrine-disrupting chemicals (EDCs). Long-term exposure to EDCs has serious adverse health effects through both hormone-direct and hormone-indirect ways. Accordingly, some EDCs can be a pathogen and an inducer to the susceptibility of disease, even if they have a very low affinity on the estrogen receptor, or no estrogenic effect. Endoplasmic reticulum (ER) stress recently attracted attention in this research area. Because ER and ER stress could be key regulators of the EDC's adverse effects, such as the malfunction of the organ, as well as the death, apoptosis, and proliferation of a cell. In this review, we focused on finding evidence which shows that EDCs could be a trigger for ER stress and provide specific examples of EDCs, which are known to cause ER stress currently.


Subject(s)
Endocrine Disruptors/adverse effects , Endoplasmic Reticulum/drug effects , Animals , Apoptosis , Cell Proliferation/drug effects , Endoplasmic Reticulum Stress/drug effects , Humans , Unfolded Protein Response/drug effects
3.
Int J Mol Sci ; 23(8)2022 Apr 14.
Article in English | MEDLINE | ID: mdl-35457166

ABSTRACT

The localization and expression of amylin protein in the rodent brain and mouse neuroblastoma Neuro-2a (N2a) are less widely known. Thus, this study investigated the expression distribution of amylin in the rat brain and N2a treated with steroid hormones. Amylin protein was identified in the olfactory bulb, cerebral cortex, dentate gyrus, thalamus, hypothalamus, ventral tegmental area (VTA), cerebellum, and brain stem in the rat brain. Additionally, the amylin protein was localized with the mature neurons of the cerebral cortex and dopaminergic neurons of the VTA. Progesterone (P4) and dexamethasone (Dex) significantly decreased, and 17ß-estradiol (E2) increased the amylin protein level in the cerebral cortex. The P4 receptor antagonist RU486 significantly influenced the effects of P4 and Dex, and the E2 receptor antagonist ICI 182,780 slightly changed E2's effect. Amylin protein expression was significantly reduced in the VTA by P4 and Dex, and its expression was changed only following P4 plus RU486 treatment. It was confirmed for the first time that amylin protein is strongly expressed in the cytoplasm in N2a cells using immunofluorescent staining. P4 increased the levels of amylin, and RU486 treatment decreased them. Dex significantly increased the levels of amylin protein. RU486 treatment reversed the effects of Dex. Therefore, amylin protein is expressed in the cerebral cortex neurons and dopaminergic neurons of the VTA of the immature rat brain. P4 and Dex influence the expression of amylin protein in the rat brain and N2a cells.


Subject(s)
Islet Amyloid Polypeptide , Mifepristone , Animals , Brain/metabolism , Estradiol/pharmacology , Islet Amyloid Polypeptide/metabolism , Mice , Mifepristone/pharmacology , Progesterone/metabolism , Rats
4.
Int J Mol Sci ; 22(14)2021 Jul 20.
Article in English | MEDLINE | ID: mdl-34299375

ABSTRACT

A real-life environment during pregnancy involves multiple and simultaneous exposures to toxic chemicals. Perinatal exposures to toxic chemicals have been reported to exert an inhibitory effect on mouse neural development and behaviors. However, the effect of combined exposures of organophosphate and nicotine has not been previously reported. In this study, we investigated whether a combined exposure of diazinon and nicotine can have a synergistic effect. The effects of the combined chemical exposure on cell viability and neuronal differentiation were examined using mouse Sox1-GFP cells. Additionally, mice were maternally administered 0.18 mg/kg diazinon, a no adverse effect level (NOAEL) dose, combined with 0.4, 1, and 2 mg/kg nicotine. Mice offspring underwent behavior tests to assess locomotor, depressive, cognitive, and social behaviors. Morphological change in the brain was investigated with immunolocalization. We revealed that the combined exposure to diazinon and nicotine can have a synergistic adverse effect in vitro. In addition, the chemical-treated mouse offspring showed abnormalities in motor learning, compulsive-like behaviors, spatial learning, and social interaction patterns. Moreover, 0.18 mg/kg diazinon and 2 mg/kg nicotine co-exposure resulted in an increase in tyrosine hydroxylase (TH)-positive dopaminergic neurons. Thus, the findings suggest that perinatal co-exposure to nicotine and diazinon can result in abnormal neurodevelopment and behavior, even at low-level administration.


Subject(s)
Behavior, Animal/drug effects , Brain/drug effects , Diazinon/adverse effects , Nicotine/adverse effects , Animals , Cells, Cultured , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Female , Male , Maze Learning/drug effects , Mice , Mice, Inbred C57BL , Pregnancy , Prenatal Exposure Delayed Effects/metabolism , Social Behavior , Spatial Learning/drug effects , Tyrosine 3-Monooxygenase/metabolism
5.
Int J Mol Sci ; 22(23)2021 Nov 30.
Article in English | MEDLINE | ID: mdl-34884750

ABSTRACT

Cyclic siloxane octamethylcyclotetrasiloxane (D4) has raised concerns as an endocrine-disrupting chemical (EDC). D4 is widely used in detergent products, cosmetics, and personal care products. Recently, robust toxicological data for D4 has been reported, but the adverse effects of D4 on brain development are unknown. Here, pregnant mice on gestational day 9.5 were treated daily with D4 to postnatal day 28, and the offspring mice were studied. The prenatal D4-treated mice exhibited cognitive dysfunction, limited memory, and motor learning defect. Moreover, prenatal D4 exposure reduced the proliferation of neuronal progenitors in the offspring mouse brain. Next, the mechanisms through which D4 regulated the cell cycle were investigated. Aberrant gene expression, such as cyclin-dependent kinases CDK6 and cyclin-dependent kinase inhibitor p27, were found in the prenatal D4-treated mice. Furthermore, the estrogen receptors ERa and ERb were increased in the brain of prenatal D4-treated mice. Overall, these findings suggest that D4 exerts estrogen activity that affects the cell cycle progression of neuronal progenitor cells during neurodevelopment, which may be associated with cognitive deficits in offspring.


Subject(s)
Endocrine Disruptors/toxicity , Neural Stem Cells/drug effects , Prenatal Exposure Delayed Effects/pathology , Prenatal Exposure Delayed Effects/physiopathology , Siloxanes/toxicity , Animals , Behavior, Animal/drug effects , Brain/drug effects , Brain/growth & development , Brain/pathology , Cell Cycle Proteins/metabolism , Cell Line , Cell Proliferation , Cognition/drug effects , Endocrine Disruptors/administration & dosage , Female , Gene Knock-In Techniques , Green Fluorescent Proteins/genetics , Male , Mice , Mice, Inbred C57BL , Models, Neurological , Motor Activity/drug effects , Mouse Embryonic Stem Cells/drug effects , Mouse Embryonic Stem Cells/metabolism , Mouse Embryonic Stem Cells/pathology , Neural Stem Cells/cytology , Neural Stem Cells/physiology , Neurogenesis/drug effects , Pregnancy , Prenatal Exposure Delayed Effects/psychology , SOXB1 Transcription Factors/genetics , Siloxanes/administration & dosage , Social Behavior
6.
Int J Mol Sci ; 22(5)2021 Mar 05.
Article in English | MEDLINE | ID: mdl-33807999

ABSTRACT

Inflammatory bowel diseases (IBDs) comprises a range of chronic inflammatory conditions of the intestinal tract. The incidence and prevalence of IBDs are increasing worldwide, but the precise etiology of these diseases is not completely understood. Calcium signaling plays a regulatory role in cellular proliferation. Nckx3, a potassium-dependent Na+/Ca2+ exchanger, is not only expressed in the brain but also in the aortic, uterine, and intestinal tissues, which contain abundant smooth muscle cells. This study investigated the role of Nckx3 in intestinal inflammation. Microarray analyses revealed the upregulation of the innate immune response-associated genes in the duodenum of Nckx3 knockout (KO) mice. The Nckx3 KO mice also showed an increase in IBD- and tumorigenesis-related genes. Using dextran sodium sulfate (DSS)-induced experimental colitis mice models, the Nckx3 KO mice showed severe colitis. Furthermore, the pathways involving p53 and NF-κB signaling were significantly upregulated by the absence of Nckx3. Overall, Nckx3 plays a critical role in the innate immune and immune response and may be central to the pathogenesis of IBD.


Subject(s)
Colitis/metabolism , Inflammatory Bowel Diseases/metabolism , NF-kappa B/metabolism , Signal Transduction , Sodium-Calcium Exchanger/genetics , Tumor Suppressor Protein p53/metabolism , Animals , Colitis/chemically induced , Colitis/genetics , Dextran Sulfate/toxicity , Disease Models, Animal , Inflammatory Bowel Diseases/chemically induced , Inflammatory Bowel Diseases/genetics , Mice , Mice, Knockout , NF-kappa B/genetics , Sodium-Calcium Exchanger/metabolism , Tumor Suppressor Protein p53/genetics
7.
Cell Physiol Biochem ; 54(3): 438-456, 2020 May 02.
Article in English | MEDLINE | ID: mdl-32357291

ABSTRACT

BACKGROUND/AIMS: Calcium homeostasis plays a crucial role in neuronal development and disease. Calbindin-D9k (CaBP-9k) acts as calcium modulators and sensors in various tissues. However, the neurobiological functions of CaBP-9k are unknown. METHODS: We used CaBP-9k knockout (KO) mice to investigate the roles of these gene in neurodegenerative diseases, such as Alzheimer's and Parkinson's diseases. We used anatomical and biochemical approaches to characterize functional abnormalities of the brain in the CaBP-9k KO mice. RESULTS: We found that the brains of CaBP-9k KO mice have increased APP/ß-amyloid, Tau, and α-synuclein accumulation and endoplasmic reticulum (ER) stress-induced apoptosis. Neurons deficient for these CaBP-9k had abnormal intracellular calcium levels and responses. ER stress inhibitor TUDCA reduced ER stress-induced apoptosis and restored ER stress- and apoptosis-related proteins expression to wild-type levels in CaBP-9k KO mice. Furthermore, treatment with TUDCA rescued the abnormal memory and motor behaviors exhibited by older CaBP-9k KO mice. CONCLUSION: Our results suggest that a loss of CaBP-9k may contribute to the onset and progression of neurodegenerative diseases.


Subject(s)
Alzheimer Disease/genetics , Apoptosis/genetics , Calbindins/genetics , Endoplasmic Reticulum Stress/genetics , Parkinson Disease/genetics , Taurochenodeoxycholic Acid/therapeutic use , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Animals , Apoptosis/drug effects , Brain/metabolism , Brain/pathology , Calbindins/metabolism , Calcium/metabolism , Cell Proliferation/genetics , Cells, Cultured , Endoplasmic Reticulum Stress/drug effects , Maze Learning/drug effects , Memory Disorders/drug therapy , Memory Disorders/genetics , Mice , Mice, Knockout , Motor Activity/drug effects , Motor Activity/genetics , Neurons/metabolism , Neurons/pathology , Parkinson Disease/drug therapy , Parkinson Disease/metabolism , RNA, Small Interfering , Risk Factors , Taurochenodeoxycholic Acid/pharmacology , alpha-Synuclein/metabolism , tau Proteins/metabolism
8.
Environ Toxicol ; 35(1): 66-77, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31507073

ABSTRACT

The heart is the first organ formed in the developing fetus, and abnormal development of the heart is a major cause of fetal death. The adverse effects of cigarette smoke on the heart have been well established, but it is not well understood how cigarette smoke components regulate signaling molecules and cardiac specific functions during the early differentiation stage of the embryonic heart. In this study, we identified changes in the size of mouse embryoid bodies (mEBs) in response to treatment with cigarette smoke extract (CSE) via regulation of HDAC2, p53, p21, and cyclin D1 protein expression, which are cardiac differentiation and cell-cycle markers, respectively. In addition, exposure of mouse embryonic stem cells (mESCs) to cigarette smoke components inhibited myocardial differentiation and development through the expression of HDAC1, HDAC2, GATA4, NKX2-5, TBX5, HAND1, and Troponin I. Long-term exposure studies showed that CSE and nicotine may delay the development of mouse cardiomyocytes from mESCs and inhibit the contractibility, which is a fundamental function of the heart. Taken together, these findings suggest that cigarette smoke components, including nicotine, may affect abnormal myocardial differentiation and development.


Subject(s)
Cell Differentiation/drug effects , Mouse Embryonic Stem Cells/drug effects , Myocytes, Cardiac/drug effects , Nicotiana/toxicity , Smoke/adverse effects , Animals , Cell Cycle/drug effects , Cyclin D1/metabolism , Histone Deacetylase 2/metabolism , Mice , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Signal Transduction/drug effects , Smoking/adverse effects
9.
Int J Mol Sci ; 21(3)2020 Feb 05.
Article in English | MEDLINE | ID: mdl-32033337

ABSTRACT

This study investigated the effect of dexamethasone (DEX) on intracellular calcium levels and the expressions of transient receptor potential cation channel subcomponent V member 6 (TRPV6), sodium-calcium exchanger 1 (NCX1), and plasma membrane calcium ATPase 1 (PMCA1) in A549 cells. The intracellular calcium level, by using the calcium indicator pGP-CMV-GCaMP6f, increased following DEX treatment for 6, 12, and 24 h in A549 cells. In addition, Rhod-4 assay after DEX treatment for 24 h showed that DEX increased the level of intracellular calcium. The expression of the calcium influx TRPV6 gene significantly increased, whereas the expressions of the calcium outflow NCX1 and PMCA1 genes significantly decreased with DEX treatment. The mRNA levels of surfactant protein genes SFTPA1, SFTPB, SFTPC, and SFTPD and the secreted airway mucin genes MUC1 and MUC5AC were investigated by treating cells with DEX. The DEX treatment decreased the mRNA levels of SFTPA1 and SFTPB but increased the mRNA levels of SFTPC and SFTPD. The MUC1 mRNA level was increased by DEX treatment, whereas MUC5AC mRNA was significantly decreased. These results indicate that DEX influences the intracellular calcium level through TRPV6, and affects pulmonary surfactant genes and secreted airway mucin genes in A549 cells.


Subject(s)
Calcium Channels/metabolism , Calcium Signaling/drug effects , Calcium/analysis , Dexamethasone/pharmacology , Glucocorticoids/pharmacology , TRPV Cation Channels/metabolism , A549 Cells , Calcium Channels/genetics , Cell Line , Humans , Mucin 5AC/genetics , Mucin 5AC/metabolism , Mucin-1/genetics , Mucin-1/metabolism , Plasma Membrane Calcium-Transporting ATPases/genetics , Plasma Membrane Calcium-Transporting ATPases/metabolism , Pulmonary Surfactant-Associated Protein A/genetics , Pulmonary Surfactant-Associated Protein A/metabolism , Pulmonary Surfactant-Associated Protein C/genetics , Pulmonary Surfactant-Associated Protein C/metabolism , RNA, Messenger/genetics , Sodium-Calcium Exchanger/genetics , Sodium-Calcium Exchanger/metabolism , TRPV Cation Channels/genetics
10.
Int J Mol Sci ; 21(11)2020 Jun 03.
Article in English | MEDLINE | ID: mdl-32503345

ABSTRACT

Triclosan (TCS) is one of the most common endocrine-disrupting chemicals (EDCs) present in household and personal wash products. Recently, concerns have been raised about the association between abnormal behavior in children and exposure to EDC during gestation. We hypothesized that exposure to TCS during gestation could affect brain development. Cortical neurons of mice were exposed in vitro to TCS. In addition, we examined in vivo whether maternal TCS administration can affect neurobehavioral development in the offspring generation. We determined that TCS can impair dendrite and axon growth by reducing average length and numbers of axons and dendrites. Additionally, TCS inhibited the proliferation of and promoted apoptosis in neuronal progenitor cells. Detailed behavioral analyses showed impaired acquisition of spatial learning and reference memory in offspring derived from dams exposed to TCS. The TCS-treated groups also showed cognition dysfunction and impairments in sociability and social novelty preference. Furthermore, TCS-treated groups exhibited increased anxiety-like behavior, but there was no significant change in depression-like behaviors. In addition, TCS-treated groups exhibited deficits in nesting behavior. Taken together, our results indicate that perinatal exposure to TCS induces neurodevelopment disorder, resulting in abnormal social behaviors, cognitive impairment, and deficits in spatial learning and memory in offspring.


Subject(s)
Behavior, Animal/drug effects , Brain/drug effects , Maternal Exposure/adverse effects , Triclosan/adverse effects , Animals , Anti-Infective Agents, Local/adverse effects , Anxiety/drug therapy , Axons , Brain/physiopathology , Cell Death , Cell Proliferation , Cognition Disorders/chemically induced , Dendrites/metabolism , Female , Learning Disabilities/chemically induced , Male , Maze Learning , Memory , Memory Disorders/chemically induced , Mice , Mice, Inbred C57BL , Neurons/metabolism , Pregnancy , Pregnancy, Animal , Prenatal Exposure Delayed Effects , Social Behavior , Spatial Learning
11.
Carcinogenesis ; 40(8): 1031-1041, 2019 08 22.
Article in English | MEDLINE | ID: mdl-31168625

ABSTRACT

It is generally accepted that androgen receptors increase the risk of hepatocellular carcinoma (HCC), and that estrogen reduces risk of HCC. Many studies regarding this have involved males. We, therefore, have focused our attention on females, especially postmenopausal females, who typically have limited supplies of estrogen. By using sex hormone-binding globulin (SHBG) transgenic mice, we produced a humanoid environment, and facilitated deposition and modulation of sex hormones. After exposure to diethylnitrosamine to induce HCC and upon reaching the age of 40 weeks, mice were fed the fat-rich diet for 5 months. Fat-rich diet fed or ovariectomized (OVX) wild-type mice aged 62 weeks showed HCC progression, whereas fat-rich diet fed SHBG mice or OVX SHBG mice displayed fewer tumors. In the liver of fat-rich diet fed SHBG mice, estrogenic conditions including high levels of 17ß-estradiol and estrogen receptor alpha led to the induction of the lipogenesis inhibitor, phosphorylated acetyl-CoA carboxylase, and consequently suppressed fatty liver. The presence of plasma SHBG in HCC bearing mice suppressed the levels of steatosis and inflammation in a process mediated by estrogens and estrogen receptor alpha. Conversely, in the liver of OVX SHBG mice, lipogenic inhibition was also observed under conditions where the supply of estrogens is limited. Through in vitro experiment, it was confirmed SHBG suppresses lipogenesis via inhibition of acetyl-CoA carboxylase level. In conclusion, our results show that plasma SHBG might have a clinical impact on lipid-mediated hepatic diseases.


Subject(s)
Carcinogenesis/genetics , Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , Non-alcoholic Fatty Liver Disease/genetics , Sex Hormone-Binding Globulin/genetics , Acetyl-CoA Carboxylase/genetics , Animals , Carcinogenesis/metabolism , Carcinogenesis/pathology , Carcinoma, Hepatocellular/etiology , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Diet, High-Fat , Diethylnitrosamine/toxicity , Disease Models, Animal , Estradiol/metabolism , Estrogen Receptor alpha/genetics , Estrogens/metabolism , Female , Humans , Lipogenesis/genetics , Liver/metabolism , Liver/pathology , Liver Neoplasms/etiology , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Menopause/genetics , Menopause/metabolism , Mice , Mice, Transgenic , Non-alcoholic Fatty Liver Disease/chemically induced , Non-alcoholic Fatty Liver Disease/complications , Non-alcoholic Fatty Liver Disease/pathology , Receptors, Androgen/genetics
12.
Mol Reprod Dev ; 86(1): 109-117, 2019 01.
Article in English | MEDLINE | ID: mdl-30411422

ABSTRACT

Many steroid hormones such as estrogen (E2) bind to their receptors for the regulation of biological processes. Pregnenolone (P5) is the precursor form of almost all steroid hormones and is often used to treat skin disorders and neurological complications. However, the mechanism and physiological function of P5 in reproductive organs are not well established. In this study, we investigated the effects of P5 on activation and expression of E2 receptor (ER) in the uteri and ovaries. To study the mechanism of P5 directly, Ishikawa cells were transfected with E2 response element (ERE)-luciferase plasmid and isoforms of ER. ERE-luciferase activity induced by P5 was similar to that induced by E2, and P5 showed high activity for ERß without any relevance to P5-metabolizing hormones such as progesterone (P4) and E2. In an animal study, immature female rats treated with P5 showed upregulation of ERα and downregulation of ERß in the uteri, which is the main organ expressing ERα. In ERß-expressing organ ovaries, estrogen receptor 1, estrogen receptor 2, and P4 receptor were all downregulated by P5 and E2. Also, a decrease of ovarian cell proliferation and viability was observed in response to P5 relative to the control, suggesting that P5 may be a candidate for antiproliferative hormone of ovarian cancer. These findings suggest that P5 stimulates ERE promoter by ERß-mediated signaling in the uteri and ovaries. Activation of ERß by P5 may help in understanding the mechanism of ER-related female reproductive diseases such as endometriosis and ovarian cancer.


Subject(s)
Endometriosis/drug therapy , Estrogen Receptor beta/biosynthesis , Gene Expression Regulation, Neoplastic/drug effects , Hormone Replacement Therapy , Neoplasm Proteins/biosynthesis , Ovarian Neoplasms/drug therapy , Pregnenolone/therapeutic use , Animals , Endometriosis/metabolism , Endometriosis/pathology , Female , Hep G2 Cells , Humans , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Rats , Rats, Sprague-Dawley , Response Elements
13.
Mol Reprod Dev ; 86(11): 1705-1719, 2019 11.
Article in English | MEDLINE | ID: mdl-31490595

ABSTRACT

Phytosphingosine-1-phosphate (P1P) is a signaling sphingolipid that regulates various physiological activities. However, little is known about the effect of P1P in the context of reproduction. Thus, we aimed to investigate the influence of P1P on oocyte maturation during porcine in vitro maturation (IVM). Here, we report the expression of S1PR1-3 among P1P receptors (S1PR1-4) in cumulus cells and oocytes. When P1P was administered at concentrations of 10, 50, 100, and 1,000 nM during IVM, the metaphase II rate was significantly increased in the 1,000 nM (1 µM) P1P treatment group. Maturation rate improvement by P1P supplementation was observed only in the presence of epidermal growth factor (EGF). Oocytes under the influence of P1P showed decreased intracellular reactive oxygen species levels but no significant differences in glutathione levels. In our molecular studies, P1P treatment upregulated gene expression involved in cumulus expansion (Has2 and EGF), antioxidant enzymes (SOD3 and Cat), and developmental competence (Oct4) while activating extracellular signal-regulated kinase1/2 and Akt signaling. P1P treatment also influenced oocyte survival by shifting the ratio of Bcl-2 to Bax while inactivating JNK signaling. We further demonstrated that oocytes matured with P1P displayed significantly higher developmental competence (cleavage and blastocyst [BL] formation rate) and greater BL quality (total cell number and the ratio of apoptotic cells) when activated via parthenogenetic activation (PA) and in vitro fertilization. Despite the low levels of endogenous P1P found in animals, exogenous P1P influenced animal reproduction, as shown by increased porcine oocyte maturation as well as preimplantation embryo development. This study and its findings are potentially relevant for both human and animal-assisted reproduction.


Subject(s)
Apoptosis/drug effects , Gene Expression Regulation/drug effects , Oocytes/metabolism , Oxidative Stress/drug effects , Sphingosine/analogs & derivatives , Animals , Cells, Cultured , Oocytes/cytology , Sphingosine/pharmacology , Swine
14.
BMC Cancer ; 19(1): 6, 2019 Jan 03.
Article in English | MEDLINE | ID: mdl-30606143

ABSTRACT

BACKGROUND: Women have a lower risk of hepatocellular carcinoma (HCC) than men, and the decreased possibility of HCC in women is thought to depend on estrogen levels. As a soybean-isoflavone product, genistein has estrogenic activity in various reproductive tissues, because it mimics 17ß-estradiol and binds the estrogen receptor. Though genistein is a known liver cancer suppressor, its effects have not been studies in long-term experiment, where genistein is fed to a female animal model of HCC. METHODS: Mice were treated with diethylnitrosamine (DEN) to induce HCC at 2 weeks of age and fed with supplemental genistein for 5 months, from 40 to 62 weeks of age. RESULTS: The dietary intake of genistein decreased the incidence of HCC and suppressed HCC development. Genistein induced phospho-AMPK in total liver extracts, Hep3B cells, and Raw 264.7 cells, and phospho-AMPK promoted apoptosis in liver and Hep3B cells. Moreover, phospho-AMPK down-regulated pro-inflammatory responses and ameliorated liver damage. A suppressed pro-inflammatory response with increased mitochondrial respiration was concomitantly observed after genistein treatment. CONCLUSIONS: Genistein-mediated AMPK activation increases hepatocyte apoptosis through energy-dependent caspase pathways, suppresses the inflammatory response in resident liver macrophages by increased cellular respiration, and consequently inhibits the initiation and progression of HCC.


Subject(s)
Carcinoma, Hepatocellular/diet therapy , Genistein/administration & dosage , Liver Neoplasms/diet therapy , Protein Kinases/genetics , AMP-Activated Protein Kinase Kinases , Animals , Apoptosis/drug effects , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Liver/drug effects , Liver/pathology , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Male , Mice , RAW 264.7 Cells
15.
Environ Toxicol ; 34(6): 689-698, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30742351

ABSTRACT

Previous studies have reported that cigarette smoke and cigarette smoke extract (CSE) have negative effects on embryonic development. However, no studies have investigated the mechanism through which CSE affects the cellular signaling pathway leading to apoptosis and oxidative stress in embryonic cells, or how the two pathways are cross-linked. Thus, we studied the effects of CSE on apoptosis and oxidative stress in mouse embryonic stem cells (mESCs). Specifically, we measured changes in cell viability in response to CSEs (3R4F and two domestic cigarettes CSE 1 and 2) using a water soluble tetrazolium (WST) assay and a neutral red uptake (NRU) assay, which revealed that cell viability decreased in a concentration-dependent manner. Western blot analysis revealed that the expression of cyclin D1 and cyclin E1 was decreased and that of p21 and p27 was increased by CSE. Additionally, the number of terminal deoxynucleotidyl transferase (TUNEL)-stained cells was increased by CSE, while the levels of Bax and Caspase-3 increased and Bcl-2 decreased. Moreover, a 2',7'-dichlorofluorescin diacetate (DCF-DA) assay and reactive oxygen species (ROS)-Glo H2 O2 assay confirmed that ROS were generated in response to CSE and that they were associated with up-regulated Keaf-1 and CHOP. Overall, the results revealed that cigarette smoke extract (CSE) inhibited cell proliferation by regulating cell cycle-related protein expression and increased oxidative stress by regulating the expression of Kelch-like ECH-associated protein 1 (Keap-1) and CCAAT/enhancer-binding protein homologous protein (CHOP), resulting in apoptosis in mESCs.


Subject(s)
Apoptosis/drug effects , Mouse Embryonic Stem Cells/drug effects , Nicotiana/toxicity , Oxidative Stress/drug effects , Smoke/adverse effects , Animals , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Humans , In Situ Nick-End Labeling , Mice , Mouse Embryonic Stem Cells/metabolism , Mouse Embryonic Stem Cells/pathology , Reactive Oxygen Species/metabolism , Smoking/adverse effects , Tobacco Smoke Pollution/adverse effects
16.
Int J Mol Sci ; 20(21)2019 Oct 25.
Article in English | MEDLINE | ID: mdl-31731478

ABSTRACT

Intracellular calcium ion content is tightly regulated for the maintenance of cellular functions and cell survival. Calbindin-D9k (CaBP-9k) is responsible for regulating the distribution of cytosolic free-calcium ions. In this study, we aimed to investigate the effect of CaBP-9k on cell survival in pancreatic beta cells. Six-month-old wildtype CaBP-9k, CaBP-28k, and CaBP-9k/28k knockout (KO) mice were used to compare the pathological phenotypes of calcium-binding protein-deleted mice. Subsequently, the endoplasmic reticulum (ER) stress reducer tauroursodeoxycholic acid (TUDCA) was administered to wildtype and CaBP-9k KO mice. In vitro assessment of the role of CaBP-9k was performed following CaBP-9k overexpression and treatment with the ER stress inducer thapsigargin. Six-month-old CaBP-9k KO mice showed reduced islet volume and up-regulation of cell death markers resulting from ER stress, which led to pancreatic beta cell death. TUDCA treatment recovered islet volume, serum insulin level, and abdominal fat storage by CaBP-9k ablation. CaBP-9k overexpression elevated insulin secretion and recovered thapsigargin-induced ER stress in the INS-1E cell line. The results of this study show that CaBP-9k can protect pancreatic beta cell survival from ER stress and contribute to glucose homeostasis, which can reduce the risk of type 1 diabetes and provide the molecular basis for calcium supplementation to diabetic patients.


Subject(s)
Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/prevention & control , Endoplasmic Reticulum Stress , Insulin-Secreting Cells/metabolism , S100 Calcium Binding Protein G/metabolism , Animals , Cell Line , Cell Survival , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/pathology , Insulin-Secreting Cells/pathology , Mice , Mice, Knockout , S100 Calcium Binding Protein G/genetics , Taurochenodeoxycholic Acid/pharmacology , Thapsigargin/pharmacology
17.
Am J Physiol Lung Cell Mol Physiol ; 314(6): L956-L966, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29446320

ABSTRACT

Calcium is important for physiological functioning in many tissues and is essential in mucus secretion and muscle contraction. Intracellular concentrations of calcium are regulated by calcium-related proteins, such as transient receptor potential cation channel subfamily V member 4 (TRPV 4), TRPV6, Calbindin-D9k (CaBP-9k), sodium-calcium exchanger (NCX1), and plasma membrane Ca2+ ATPase 1 (PMCA1). In this study, the relationship between secretion of pulmonary mucus and calcium regulation was investigated. To confirm the effect of steroid hormones, immature mice were injected with estrogen (E2) or progesterone (P4), and mature mice were injected with dexamethasone (DEX). Subsequently, the location and expression of TRPV4, TRPV6, CaBP-9k, NCX1, and PMCA1 in lung tissue were examined. Periodic acid-Schiff staining was performed to investigate functional aspects of the protein expression. There were no significant differences in calcium-related gene expression in E2- and P4-treated mice, but TRPV4, NCX1, and PMCA1 were increased in DEX-treated mice and were recovered by RU486 treatment. DEX induces the expression of calcium-related proteins through the glucocorticoid receptor-mediated pathway and may involve decreased mucin secretion in the bronchiole. TRPV4, TRPV6, CaBP-9k, NCX1, and PMCA1 were specifically expressed in Clara and alveolar type 2 cells of mouse lung. CC10, a marker of Clara cells, was decreased by DEX. In addition, mucin secretion, which is a functional aspect of this cell, was also decreased by DEX treatment. Control of calcium-related gene expression may affect the control of mucus secretion in the lung. Such a control mechanism can form the basis of studies into diseases such as inflammation attributable to mucus secretion abnormalities, coughing, and respiratory disorders and distress.


Subject(s)
Calcium Channels/biosynthesis , Dexamethasone/pharmacology , Estrogens/pharmacology , Gene Expression Regulation/drug effects , Lung/metabolism , Mucins/biosynthesis , Plasma Membrane Calcium-Transporting ATPases/biosynthesis , Progesterone/pharmacology , Animals , Female , Lung/cytology , Male , Mice
18.
Int J Mol Sci ; 19(10)2018 Sep 26.
Article in English | MEDLINE | ID: mdl-30261648

ABSTRACT

We investigated whether combined fluid shear stress (FSS) and melatonin stimulated signal transduction in cilia-less MC3T3-E1 preosteoblast cells. MC3T3-E1 cells were treated with chloral hydrate or nocodazole, and mechanotransduction sensor primary cilia were removed. p-extracellular signal⁻regulated kinase (ERK) and p-Akt with/without melatonin increased with nocodazole treatment and decreased with chloral hydrate treatment, whereas p-ERK and p-Akt in FSS with/without melatonin increased in cilia-less groups compared to cilia groups. Furthermore, p-mammalian target of rapamycin (mTOR) with FSS-plus melatonin increased in cilia-less groups compared to only melatonin treatments in cilia groups. Expressions of Bcl-2, Cu/Zn-superoxide dismutase (SOD), and catalase proteins were higher in FSS with/without melatonin with cilia-less groups than only melatonin treatments in cilia groups. Bax protein expression was high in FSS-plus melatonin with chloral hydrate treatment. In chloral hydrate treatment with/without FSS, expressions of Cu/Zn-SOD, Mn-SOD, and catalase proteins were high compared to only-melatonin treatments. In nocodazole treatment, Mn-SOD protein expression without FSS was high, and catalase protein level with FSS was low, compared to only melatonin treatments. These data show that the combination with FSS and melatonin enhances ERK/Akt/mTOR signal in cilia-less MC3T3-E1, and the enhanced signaling in cilia-less MC3T3-E1 osteoblast cells may activate the anabolic effect for the preservation of cell structure and function.


Subject(s)
Melatonin/pharmacology , Osteoblasts/drug effects , Protein Kinases/metabolism , Signal Transduction/drug effects , Stress, Mechanical , Animals , Antioxidants/pharmacology , Cell Line , Chloral Hydrate/pharmacology , Cilia/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Hydrodynamics , Mice , Nocodazole/pharmacology , Osteoblasts/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Superoxide Dismutase-1/metabolism , TOR Serine-Threonine Kinases/metabolism , Tubulin Modulators/pharmacology
19.
J Reprod Dev ; 63(6): 581-590, 2017 Dec 15.
Article in English | MEDLINE | ID: mdl-28993559

ABSTRACT

Compared with the in vivo environment, porcine in vitro embryo-culture systems are suboptimal, as they induce oxidative stress via the accumulation of reactive oxygen species (ROS). High ROS levels during early embryonic development cause negative effects, such as apoptosis. In this study, we examined the effects of the antioxidant carboxyethylgermanium sesquioxide (Ge-132) during in vitro culture (IVC) on embryonic development in porcine in vitro fertilization (IVF) embryos. Zygotes were treated with different concentrations of Ge-132 (0, 100, 200 and 400 µg/ml). All of the Ge-132 treatment groups displayed greater total cell numbers after IVC (98.1, 98.5 and 103.4, respectively) compared with the control group (73.9). The 200 µg/ml Ge-132 treatment group exhibited significantly increased intracellular GSH levels compared with the control group, whereas the ROS generation levels decreased in Ge-132 dose-dependent manner (P < 0.05). The mRNA expression levels of the KEAP1 gene and proapoptotic genes BAX and CASPASE3 were lower in the Ge-132 treated blastocysts compared with the control group (P < 0.05). The percentages of apoptotic and necrotic cells in the Ge-132 treated embryos on day 2 (48 h) were significantly lower than the untreated embryos (9.1 vs. 17.1% and 0 vs. 2.7%, respectively). In the day 7 blastocysts, the percentages of apoptotic cells in 200 µg/ml Ge-132 treated group were lower compared to controls (1.6 vs. 2.5%). More KEAP1 protein was found to be localized in cytoplasm of the 200 µg/ml Ge-132 treated blastocysts, whereas KEAP1 protein was predominantly nuclei in the control blastocysts. These results indicate that the developmental competence of embryos cultured under Ge-132 treatment may be associated with KEAP1 signaling cascades involved in oxidative stress and apoptosis during porcine preimplantation embryo development.


Subject(s)
Apoptosis/drug effects , Embryo Culture Techniques , Embryo, Mammalian/drug effects , Organometallic Compounds/pharmacology , Oxidative Stress/drug effects , Animals , Embryo, Mammalian/metabolism , Germanium , Propionates , Swine
20.
Environ Toxicol ; 32(4): 1439-1454, 2017 Apr.
Article in English | MEDLINE | ID: mdl-27539251

ABSTRACT

Fludioxonil is an antifungal agent used in agricultural applications that is present at measurable amounts in fruits and vegetables. In this study, the effects of fludioxonil on cancer cell viability, epithelial-mesenchymal transition (EMT), and metastasis were examined in MCF-7 clonal variant breast cancer cell (MCF-7 CV cells) with estrogen receptors (ERs). MCF-7 CV cells were cultured with 0.1% DMSO (control), 17ß-estradiol (E2; 1 ×10-9 M, positive control), or fludioxonil (10-5 -10-8 M). MTT assay revealed that fludioxonil increased MCF-7 CV cell proliferation 1.2 to 1.5 times compared to the control, while E2 markedly increased the cell proliferation by about 3.5 times. When the samples were co-treated with ICI 182,780 (10-8 M), an ER antagonist, fludioxonil-induced cell proliferation was reversed to the level of the control. Protein levels of cyclin E1, cyclin D1, Snail, and N-cadherin increased in response to fludioxonil as the reaction to E2, but these increases were not observed when fludioxonil was administered with ICI 182,780. Moreover, the protein level of p21 and E-cadherin decreased in response to treatment with fludioxonil, but remained at the control level when co-treated with ICI 182,780. In xenografted mouse models transplanted with MCF-7 CV cells, fludioxonil significantly increased the tumor mass formation by about 2.5 times as E2 did when compared to vehicle (0.1% DMSO) during the experimental period (80 days). Immunohistochemistry revealed that the protein level of proliferating cell nuclear antigen (PCNA), Snail, and cathepsin D increased in response to fludioxonil as the reaction to E2. These results imply that fludioxonil may have a potential to induce growth or metastatic behaviors of breast cancer by regulation of the expression of cell cycle-, EMT-, and metastasis-related genes via the ER-dependent pathway. © 2016 Wiley Periodicals, Inc. Environ Toxicol 32: 1439-1454, 2017.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Dioxoles/pharmacology , Epithelial-Mesenchymal Transition/drug effects , Pyrroles/pharmacology , Receptors, Estrogen/metabolism , Animals , Antigens, CD , Antineoplastic Agents/therapeutic use , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cadherins/metabolism , Cathepsin D/metabolism , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Shape/drug effects , Cyclin D1/metabolism , Dioxoles/therapeutic use , Female , Humans , MCF-7 Cells , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Metastasis , Pyrroles/therapeutic use , Signal Transduction , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL