Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 615
Filter
Add more filters

Publication year range
1.
Ecotoxicol Environ Saf ; 283: 116859, 2024 Sep 15.
Article in English | MEDLINE | ID: mdl-39137466

ABSTRACT

The developmental toxicity and human health risks of triazole fungicides (TFs) have attracted worldwide attention due to the ability to enter the human body in a variety of ways. Nevertheless, the specific mechanism by which TFs exert remains incompletely understood. Given that retinoic acid (RA) signaling pathway are closely related to development, this study aimed to screen and identify developmentally disabled chemicals in commonly used TFs and to reveal the potential effects of TFs on developmental retardation through the RA signaling pathway in mouse embryonic stem cells (mESCs). Specifically, six typical TFs (myclobutanil, tebuconazole, hexaconazole, propiconazole, difenoconazole, and flusilazole) were exposed through the construction of an embryoid bodies (EBs)-based in vitro global differentiation models. Our results clarified that various TFs disturbed lineage commitment during early embryonic development. Crucially, the activation of RA signaling pathway, which alters the expression of key genes and interferes the transport and metabolism of retinol, may be responsible for this effect. Furthermore, molecular docking, molecular dynamics simulations, and experiments using a retinoic acid receptor α inhibitor provide evidence supporting the potential modulatory role of the retinoic acid signaling pathway in developmental injury. The current study offers new insights into the TFs involved in the RA signaling pathway that interfere with the differentiation process of mESCs, which is crucial for understanding the impact of TFs on pregnancy and early development.


Subject(s)
Cell Differentiation , Fungicides, Industrial , Signal Transduction , Tretinoin , Triazoles , Triazoles/toxicity , Fungicides, Industrial/toxicity , Cell Differentiation/drug effects , Tretinoin/toxicity , Animals , Mice , Signal Transduction/drug effects , Mouse Embryonic Stem Cells/drug effects , Molecular Docking Simulation , Dioxolanes/toxicity , Embryonic Stem Cells/drug effects , Nitriles , Silanes
2.
Arch Toxicol ; 97(8): 2143-2153, 2023 08.
Article in English | MEDLINE | ID: mdl-37278767

ABSTRACT

While exposure to high levels of all-trans retinoic acid (atRA) during pregnancy is known to suppress murine embryonic palate mesenchymal (MEPM) cells proliferation and to result in cleft palate (CP) development, the underlying mechanisms are poorly understood. Accordingly, this study was designed with the goal of clarifying the etiological basis for atRA-induced CP. A murine model of CP was established via the oral administration of atRA to pregnant mice on gestational day (GD) 10.5, after which transcriptomic and metabolomic analyses were performed with the goal of clarifying the critical genes and metabolites associated with CP development through an integrated multi-omics approach. MEPM cells proliferation was altered by atRA exposure as expected, contributing to CP incidence. In total, 110 genes were differentially expressed in the atRA treatment groups, suggesting that atRA may influence key biological processes including stimulus, adhesion, and signaling-related activities. In addition, 133 differentially abundant metabolites were identified including molecules associated with ABC transporters, protein digestion and absorption, mTOR signaling pathway, and the TCA cycle, suggesting a link between these mechanisms and CP. Overall, combined analyses of these transcriptomic and metabolomic results suggested that the MAPK, calcium, PI3K-Akt, Wnt, and mTOR signaling pathways are particularly important pathways enriched in the palatal cleft under conditions of atRA exposure. Together, these integrated transcriptomic and metabolomic approaches provided new evidence with respect to the mechanisms underlying altered MEPM cells proliferation and signal transduction associated with atRA-induced CP, revealing a possible link between oxidative stress and these pathological changes.


Subject(s)
Cleft Palate , Pregnancy , Female , Animals , Mice , Cleft Palate/chemically induced , Cleft Palate/genetics , Cleft Palate/pathology , Transcriptome , Phosphatidylinositol 3-Kinases/metabolism , Tretinoin/toxicity , Cell Proliferation , TOR Serine-Threonine Kinases/metabolism , Mice, Inbred C57BL
3.
Pharmacogenomics J ; 21(6): 638-648, 2021 12.
Article in English | MEDLINE | ID: mdl-34145402

ABSTRACT

Retinoids are widely used in diseases spanning from dermatological lesions to cancer, but exhibit severe adverse effects. A novel all-trans-Retinoic Acid (atRA)-spermine conjugate (termed RASP) has shown previously optimal in vitro and in vivo anti-inflammatory and anticancer efficacy, with undetectable teratogenic and toxic side-effects. To get insights, we treated HaCaT cells which resemble human epidermis with IC50 concentration of RASP and analyzed their miRNA expression profile. Gene ontology analysis of their predicted targets indicated dynamic networks involved in cell proliferation, signal transduction and apoptosis. Furthermore, DNA microarrays analysis verified that RASP affects the expression of the same categories of genes. A protein-protein interaction map produced using the most significant common genes, revealed hub genes of nodal functions. We conclude that RASP is a synthetic retinoid derivative with improved properties, which possess the beneficial effects of retinoids without exhibiting side-effects and with potential beneficial effects against skin diseases including skin cancer.


Subject(s)
Keratinocytes/drug effects , MicroRNAs/metabolism , Spermine/analogs & derivatives , Transcriptome , Tretinoin/analogs & derivatives , Apoptosis/drug effects , Apoptosis/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , Dose-Response Relationship, Drug , Gene Regulatory Networks , HaCaT Cells , Humans , Inhibitory Concentration 50 , Keratinocytes/metabolism , Keratinocytes/pathology , MicroRNAs/genetics , Protein Interaction Maps , Signal Transduction/drug effects , Signal Transduction/genetics , Spermine/pharmacology , Spermine/toxicity , Tretinoin/pharmacology , Tretinoin/toxicity
4.
Toxicol Appl Pharmacol ; 433: 115792, 2021 12 15.
Article in English | MEDLINE | ID: mdl-34742744

ABSTRACT

Concurrent with the '3R' principle, the embryonic stem cell test (EST) using mouse embryonic stem cells, developed in 2000, remains the solely accepted in vitro method for embryotoxicity testing. However, the scope and implementation of EST for embryotoxicity screening, compliant with regulatory requirements, are limited. This is due to its technical complexity, long testing period, labor-intensive methodology, and limited endpoint data, leading to misclassification of embryotoxic potential. In this study, we used human induced pluripotent stem cell (hiPSC)-derived embryoid bodies (EB) as an in vitro model to investigate the embryotoxic effects of a carefully selected set of pharmacological compounds. Morphology, viability, and differentiation potential were investigated after exposing EBs to folic acid, all-trans-retinoic acid, dexamethasone, and valproic acid for 15 days. The results showed that the compounds differentially repressed cell growth, compromised morphology, and triggered apoptosis in the EBs. Further, transcriptomics was employed to compare subtle temporal changes between treated and untreated cultures. Gene ontology and pathway analysis revealed that dysregulation of a large number of genes strongly correlated with impaired neuroectoderm and cardiac mesoderm formation. This aberrant gene expression pattern was associated with several disorders of the brain like mental retardation, multiple sclerosis, stroke and of the heart like dilated cardiomyopathy, ventricular tachycardia, and ventricular arrhythmia. Lastly, these in vitro findings were validated using in ovo chick embryo model. Taken together, pharmacological compound or drug-induced defective EB development from hiPSCs could potentially be used as a suitable in vitro platform for embryotoxicity screening.


Subject(s)
Cell Differentiation/drug effects , Embryoid Bodies/drug effects , Gene Expression Profiling , Induced Pluripotent Stem Cells/drug effects , Myocytes, Cardiac/drug effects , Neural Stem Cells/drug effects , Teratogens/toxicity , Toxicity Tests , Transcriptome/drug effects , Animals , Apoptosis/drug effects , Cell Line , Cell Lineage , Chick Embryo , Dexamethasone/toxicity , Dose-Response Relationship, Drug , Embryoid Bodies/metabolism , Embryoid Bodies/pathology , Gene Expression Regulation, Developmental/drug effects , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology , Inhibitory Concentration 50 , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Neural Stem Cells/metabolism , Neural Stem Cells/pathology , Neurogenesis/drug effects , Risk Assessment , Tretinoin/toxicity , Valproic Acid/toxicity
5.
Exp Cell Res ; 386(2): 111734, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31770533

ABSTRACT

BACKGROUND: The molecular mechanisms of abnormal palatogenesis were investigated in this study. A key regulator, miR-106a-5p, and its target pathway were analyzed. OBJECTIVES: This research is trying to clarify the underlying mechanism of the modulation of miRNA transcription during the formation of cleft palate by 7T and 9.4T NMR metabolomic platforms. METHOD: Differentially expressed miRNAs and mRNAs were analyzed by microarray analysis and verified by qRT-PCR. The protein expression in TGFß signaling pathways were analyzed by Western Blotting. The relationship between miR-106a-5p and TGFß were analyzed by luciferase reporter assay. Cell apoptosis were analyzed by flow cytometer. And finally, the metabonomics were analyzed by NMR and multivariate data analysis models (MVDA). RESULTS: The expression of miR-106a-5p increased in cleft palatal tissue and negatively correlated with the protein level of Tgfbr2. The luciferase assay further proved that the tgfbr2 was a direct target of miR-106a-5p. In another aspect, miR-106a-5p increased apoptosis level in palatal mesenchymal cells, possibly because its inhibition of TGFß signaling pathway. Moreover, low cholesterol and choline levels with high citric acid and lipid levels were observed by 7T and 9.4T NMR metabonomic analysis, which inferred the disorder of cell membrane synthesis in cleft palate formation. Furthermore, transformation from choline to phosphatidylcholine regulated by miR-106a-5p was also disrupted, resulting in phosphatidic choline synthesis disorder and reduced cell membrane synthesis. CONCLUSIONS: The regulatory mechanism of cleft palate was studied at transcriptional and metabolomics levels, which may provide important information in understanding the primary cause of this abnormality.


Subject(s)
Cleft Palate/genetics , Mesenchymal Stem Cells/metabolism , MicroRNAs/genetics , Palate/drug effects , Smad2 Protein/genetics , Transforming Growth Factor beta/genetics , Animals , Apoptosis/drug effects , Apoptosis/genetics , Cell Membrane/chemistry , Cell Membrane/drug effects , Cell Membrane/metabolism , Citric Acid/metabolism , Cleft Palate/chemically induced , Cleft Palate/metabolism , Cleft Palate/pathology , Disease Models, Animal , Embryo, Mammalian , Female , Gene Expression Regulation, Developmental , Humans , Male , Membrane Lipids/chemistry , Membrane Lipids/metabolism , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/pathology , Metabolome/genetics , Mice , MicroRNAs/classification , MicroRNAs/metabolism , Palate/growth & development , Palate/metabolism , Palate/pathology , Receptor, Transforming Growth Factor-beta Type II/genetics , Receptor, Transforming Growth Factor-beta Type II/metabolism , Signal Transduction , Smad2 Protein/metabolism , Smad3 Protein/genetics , Smad3 Protein/metabolism , Transcriptome , Transforming Growth Factor beta/metabolism , Tretinoin/toxicity
6.
Int J Mol Sci ; 22(15)2021 Jul 29.
Article in English | MEDLINE | ID: mdl-34360880

ABSTRACT

To prevent congenital defects arising from maternal exposure, safety regulations require pre-market developmental toxicity screens for industrial chemicals and pharmaceuticals. Traditional embryotoxicity approaches depend heavily on the use of low-throughput animal models which may not adequately predict human risk. The validated embryonic stem cell test (EST) developed in murine embryonic stem cells addressed the former problem over 15 years ago. Here, we present a proof-of-concept study to address the latter challenge by updating all three endpoints of the classic mouse EST with endpoints derived from human induced pluripotent stem cells (hiPSCs) and human fibroblasts. Exposure of hiPSCs to selected test chemicals inhibited differentiation at lower concentrations than observed in the mouse EST. The hiPSC-EST also discerned adverse developmental outcomes driven by novel environmental toxicants. Evaluation of the early cardiac gene TBX5 yielded similar toxicity patterns as the full-length hiPSC-EST. Together, these findings support the further development of hiPSCs and early molecular endpoints as a biologically relevant embryotoxicity screening approach for individual chemicals and mixtures.


Subject(s)
Cell Differentiation/drug effects , Fluorouracil/toxicity , Induced Pluripotent Stem Cells/cytology , Myocytes, Cardiac/cytology , Penicillin G/pharmacology , Teratogens/pharmacology , Toxicity Tests/methods , Tretinoin/toxicity , Animals , Cell Survival/drug effects , Cells, Cultured , Congenital Abnormalities/prevention & control , Embryonic Development/drug effects , Fibroblasts/cytology , Humans , Mice , Mouse Embryonic Stem Cells/cytology , Myocytes, Cardiac/drug effects , T-Box Domain Proteins
7.
Wei Sheng Yan Jiu ; 50(6): 1000-1005, 2021 Nov.
Article in Zh | MEDLINE | ID: mdl-34949330

ABSTRACT

OBJECTIVE: To investigate the expression of key genes and proteins of retinoic acid signaling pathway in procymidone-induced uterine injury in adolescent mice, and analyze the relationship between the signaling pathway and female reproductive damage. METHODS: The 3-week age ICR mice were randomly divided into low, medium, and high-dose groups and one control group with 8 mice in each group by weight. The low, medium and high dose groups were respectively given 50, 100 and 200 mg/(kg·d) procymidone orally for 21 days continuously, while the control group was given equal volume of soybean oil. After the mice were sacrificed, the uterus was taken from both sides for observing the histological changes in the cross-sectional slices of the uterus, the detection of the expression abundance of genes which related to the retinoic acid signaling pathway by the real-time fluorescent quantitative PCR, and the measurement of ALDH2 and CYP26 a1 proteins expression by Western blot. RESULTS: The body weight of mice in low-dose, medium-dose and high-dose groups were(27.50±1.49) g, (27.72±1.40) g and(26.89±1.19) g, respectively, which were lower than those in control group(31.48±1.14) g(P<0.05). The density of uterine lining monolayer columnar epithelium and lamina propria tubular uterine glands gradually decreases, at the same time the uterine folds become less with the dose of procymidone increases. adh1, ad/2, aldh1a1 in each experimental group were higher than those in the control group(P<0.05); the expression levels of aldh1a2 and aldh1a3 genes in the middle and high dose groups were higher than those in the control group(P<0.05); the expression levels of retinoic acid nuclear receptor rarα, rarγ, rxrα and rxrß genes in the high-dose group were higher than those in the control(P<0.05); yet the expression levels of cyp26a2 and cyp26a3 in the high-dose group were lower than those in the control group(P<0.05); the jnk family in medium and high dose groups were higher than the control(P<0.05). The expression of ALDH2 in each experimental group was higher than that in the control group, and increased with the increase of the dose(P<0.05); the expression of CYP26 a1 in each experimental group was not significantly different from that of the control group. CONCLUSION: The retinoic acid signal pathway is activated in procymidone-induced uterine injury in mice, then regulates the increase of the expression of jnk family, leading to the damage.


Subject(s)
Signal Transduction , Tretinoin , Animals , Bridged Bicyclo Compounds , Cross-Sectional Studies , Female , Mice , Mice, Inbred ICR , Tretinoin/toxicity , Uterus
8.
Mol Hum Reprod ; 26(2): 111-116, 2020 02 29.
Article in English | MEDLINE | ID: mdl-31943113

ABSTRACT

An adverse outcome pathway (AOP) is a simplified description of the sequence of mechanistic events that lead to a particular toxicological effect, from initial trigger to adverse outcome. Although designed to inform regulatory risk assessors, the AOP framework also provides a platform for innovative collaborations between experts from relevant research fields and the regulatory community. The underpinning for any AOP is basic knowledge about molecular and developmental processes; such knowledge can only be attained by solid bioscientific research. Starting with this fundamental knowledge, the objective is to devise novel testing strategies that focus on key events in a causative pathway. It is anticipated that such a knowledge-based approach will ultimately alleviate many of the burdens associated with classical chemical testing strategies that typically involve large-scale animal toxicity regimens. This hails from the notion that a solid understanding of the underlying mechanisms will allow the development and use of alternative test methods, including both in vitro and in silico approaches. This review is specifically targeted at professionals working in bioscientific fields, such as developmental and reproductive biology, and aims to (i) inform on the existence of the AOP framework and (ii) encourage new cross-disciplinary collaborations. It is hoped that fundamental biological knowledge can thus be better exploited for applied purposes: firstly, an improved understanding of how our perpetual exposure to environmental chemicals is causing human reproductive disease and, secondly, new approaches to screen for harmful chemicals more efficiently. This is not an instructional manual on how to create AOPs; rather, we discuss how to harness fundamental knowledge from the biosciences to assist regulatory toxicologists in their efforts to protect humans against chemicals that harm human reproductive development and function.


Subject(s)
Adverse Outcome Pathways , Developmental Biology/methods , Noxae/adverse effects , Reproduction/drug effects , Reproductive Medicine/methods , Toxicology/methods , Anal Canal/embryology , Androgens/physiology , Animals , Endocrine Disruptors/toxicity , Genitalia/embryology , Humans , Interdisciplinary Communication , Internet , Models, Animal , Nipples/embryology , Noxae/toxicity , Reproduction/physiology , Tretinoin/toxicity
9.
Contact Dermatitis ; 81(3): 184-193, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31006867

ABSTRACT

BACKGROUND: Retinoic acid (RA)-induced dermatitis is the most frequent side-effect limiting its widespread use. However, the exact mechanisms triggering dermatitis are not fully understood, including the role of skin mast cells. The newly discovered Mas-related G-protein-coupled receptor-X2 (MRGPRX2) in mast cells mediates pseudoallergic drug reactions in several types of dermatitis. A possible contribution of MRGPRX2 to contact dermatitis induced by RA has hitherto not been examined. OBJECTIVES: To investigate whether all-trans-RA (ATRA) activates mast cells via MRGPRX2/MrgprB2 (the mouse orthologue), contributing to the pathogenesis of retinoid-induced dermatitis. METHODS: Wild-type (WT) and MrgprB2-/- mice were treated with topical ATRA to observe local inflammation and mast cell degranulation in vivo by the use of haematoxylin and eosin and immunofluorescence staining. Release of histamine and release of ß-hexosaminidase were measured and calcium influx was detected in Laboratory of Allergic Disease 2 (LAD2) cells with specific knockdown targeting MRGPRX2 by small interfering RNA (siRNA) and in primary cells from MrgprB2-/- mice. RESULTS: As compared with WT mice, MrgprB2-/- mice showed resistance to ATRA-triggered contact dermatitis and local inflammatory reactions in the paws. ATRA activated mast cells via the MrgprB2 pathway in murine cells, and via the MRGPRX2 pathway in human mast cells. CONCLUSIONS: ATRA-induced dermatitis could be achieved by activating mast cells via MRGPRX2/MrgprB2, which may provide a potential therapy target to reduce the side-effect.


Subject(s)
Cell Degranulation/drug effects , Dermatitis, Contact/etiology , Mast Cells/physiology , Receptors, G-Protein-Coupled/genetics , Tretinoin/pharmacology , Animals , Calcium/metabolism , Cell Line , Dermatitis , Dermatitis, Contact/genetics , Gene Knockdown Techniques , Histamine/metabolism , Humans , Male , Mast Cells/metabolism , Mice , Nerve Tissue Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, Neuropeptide/metabolism , Tretinoin/toxicity , beta-N-Acetylhexosaminidases/metabolism
10.
Ecotoxicol Environ Saf ; 169: 573-582, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30476819

ABSTRACT

Ampullariidae are unique among gastropods in that females normally show a primordium of the copulatory apparatus (CApp). The aims of this study were (a) to quantitatively evaluate the development and growth of the female CApp with age; (b) to compare the effects of RXR and PPARγ agonists in adult females of known age and (c) to explore the effect of masculinizing RXR agonists on the expression of RXR in the CApp. It was found that the CApp grows and develops with age. A significant increase in penile sheath length (PsL) and also in a developmental index (DI) was observed in 7-8 months old females, as compared with 4-5 months old ones. A reported endogenous agonist of RXR, 9-cis retinoic acid (9cis-RA), as well as two organotin compounds, tributyltin (TBT) and triphenyltin (TPT) which have been also reported to bind to RXR, were injected and its masculinizing effects were measured. Also, the effect of a PPARγ agonist, rosiglitazone, was studied. All studied RXR agonists, but not the PPARγ agonist, were effective in increasing PsL, penile length (PL) and DI. Finally, the expression of the RXR in the CApp was studied (Western blot) in control, TBT, TPT, and 9cis-RA treated females. A significantly increased expression of RXR was only observed after 9cis-RA treatment. It is concluded that (a) development and growth of the CApp is significantly affected by female age; (b) reported RXR agonists, but not a PPARγ agonist, cause female masculinization of young females. An appraisal of previous studies of female masculinization in the Ampullariidae has also been made and it is emphasized that the masculinizing effect of aging should be considered, particularly when interpreting field data.


Subject(s)
Aging/drug effects , Endocrine Disruptors/toxicity , Gastropoda/drug effects , Penis/drug effects , Retinoid X Receptors/agonists , Sex Characteristics , Aging/metabolism , Animals , Female , Gastropoda/growth & development , Gastropoda/metabolism , Male , Organotin Compounds/toxicity , Penis/anatomy & histology , Penis/growth & development , Tretinoin/toxicity , Trialkyltin Compounds/toxicity
11.
J Cell Biochem ; 119(12): 9967-9973, 2018 12.
Article in English | MEDLINE | ID: mdl-30155966

ABSTRACT

Epithelial-mesenchymal transformation of the medial edge epithelium is the most crucial process in embryonic palatal fusion. This study aimed to explore the relationship and potential mechanism between enhancer DNA methylation and mRNA expression of histone deacetylase 4 (HDAC4) during palatal fusion induced by maternal exposure to all-trans retinoic acid (ATRA). Pregnant mice were administered ATRA (70 mg/kg) by gavage at embryonic gestation day 10.5 (E10.5) to establish a cleft palate (CP) model in C57BL/6J mice. Control groups were given an equivalent volume of corn oil. Pregnant mice were dissected at E14.5 (n = 6) to obtain embryonic palates. HDAC4 enhancer DNA methylation data were obtained from a previous MethylRAD-seq. Methylation-specific polymerase chain reaction (MSP) and real-time quantitative PCR were used to quantify enhancer methylation and the mRNA expression level of HDAC4. Enhancer DNA methylation at a non-CpG site within the HDAC4 gene was hyper-methylated at E14.5 (P: 0.011, log2 FC:1.67). The MSP results indicated a similar trend, in agreement with the MethylRAD-seq results. The change in the HDAC4 expression level was negatively correlated with its enhancer DNA methylation level, at the non-CpG site, during palatal fusion induced by ATRA. Enhancer DNA methylation of HDAC4 might play an important regulatory role during palatogenesis, especially in embryonic palatal fusion at E 14.5, and may facilitate the development of novel epigenetic biomarkers in the treatment of CP.


Subject(s)
Cleft Palate/genetics , DNA Methylation , Epithelial-Mesenchymal Transition , Histone Deacetylases/genetics , Tretinoin/toxicity , Animals , Cleft Palate/chemically induced , Cleft Palate/metabolism , Disease Models, Animal , Enhancer Elements, Genetic , Epigenesis, Genetic , Female , Gene Expression Regulation, Developmental , Maternal Exposure , Mice , Mice, Inbred C57BL , Palate , Pregnancy , RNA, Messenger/genetics , Tretinoin/administration & dosage
12.
Theor Biol Med Model ; 15(1): 16, 2018 10 16.
Article in English | MEDLINE | ID: mdl-30322383

ABSTRACT

BACKGROUND: In the classical pathway of retinoic acid (RA) mediated gene transcription, RA binds to a nuclear hormone receptor dimer composed of retinoic acid receptor (RAR) and retinoid X receptor (RXR), to induce the expression of its downstream target genes. In addition to nuclear receptors, there are other intracellular RA binding proteins such as cellular retinoic acid binding proteins (CRABP1 and CRABP2) and cytochrome P450 (CYP) enzymes, whose contributions to the RA signaling pathway have not been fully understood. The objective of this study was to compare the significance of various RA binding receptors, i.e. CRABP1, CRABP2, CYP and RAR in the RA signaling pathway. In this regard, we developed a mathematical model of the RA pathway, which is one of the few models, if not the only one, that includes all main intracellular RA binding receptors. We then performed a global sensitivity analysis (GSA) to investigate the contribution of the RA receptors to RA-induced mRNA production, when the cells were treated with a wide range of RA levels, from physiological to pharmacological concentrations. RESULTS: Our results show that CRABP2 and RAR are the most and the least important proteins, respectively, in controlling the model performance at physiological concentrations of RA (1-10 nM). However, at higher concentrations of RA, CYP and RAR are the most sensitive parameters of the system. Furthermore, we found that depending on the concentrations of all RA binding proteins, the rate of metabolism of RA can either change or remain constant following RA therapy. The cellular levels of CRABP1 are more important than that of CRABP2 in controlling RA metabolite formation at pharmacological conditions (RA = 0.1-1 µM). Finally, our results indicate a significant negative correlation between total mRNA production and total RA metabolite formation at pharmacological levels of RA. CONCLUSIONS: Our simulations indicate that the significance of the RA binding proteins in the RA pathway of gene expression strongly depends on intracellular concentration of RA. This study not only can explain why various cell types respond to RA therapy differently, but also can potentially help develop pharmacological methods to increase the efficacy of the drug.


Subject(s)
Proteins/metabolism , Signal Transduction , Tretinoin/metabolism , Gene Expression Regulation/drug effects , Metabolome/drug effects , Metabolome/genetics , Models, Biological , Proteins/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Transcription, Genetic/drug effects , Tretinoin/pharmacology , Tretinoin/toxicity
13.
Dev Biol ; 415(2): 314-325, 2016 07 15.
Article in English | MEDLINE | ID: mdl-26777098

ABSTRACT

Prenatal exposure to ethanol results in fetal alcohol spectrum disorder (FASD), a syndrome characterised by a broad range of clinical manifestations including craniofacial dysmorphologies and neurological defects. The characterisation of the mechanisms by which ethanol exerts its teratogenic effects is difficult due to the pleiotropic nature of its actions. Different experimental model systems have been employed to investigate the aetiology of FASD. Here, I will review studies using these different model organisms that have helped to elucidate how ethanol causes the craniofacial abnormalities characteristic of FASD. In these studies, ethanol was found to impair the prechordal plate-an important embryonic signalling centre-during gastrulation and to negatively affect the induction, migration and survival of the neural crest, a cell population that generates the cartilage and most of the bones of the skull. At the cellular level, ethanol appears to inhibit Sonic hedgehog signalling, alter levels of retionoic acid activity, trigger a Ca(2+)-CamKII-dependent pathway that antagonises WNT signalling, affect cytoskeletal dynamics and increase oxidative stress. Embryos of the domestic chick Gallus gallus domesticus have played a central role in developing a working model for the effects of ethanol on craniofacial development because they are easily accessible and because key steps in craniofacial development are particularly well established in the avian embryo. I will finish this review by highlighting some potential future avenues of fetal alcohol research.


Subject(s)
Abnormalities, Drug-Induced/embryology , Chick Embryo/drug effects , Craniofacial Abnormalities/chemically induced , Disease Models, Animal , Ethanol/toxicity , Face/embryology , Fetal Alcohol Spectrum Disorders/physiopathology , Maxillofacial Development/drug effects , Skull/embryology , Animals , Calcium Signaling/drug effects , Craniofacial Abnormalities/embryology , Embryo, Mammalian/drug effects , Embryo, Nonmammalian/drug effects , Endoderm/drug effects , Face/abnormalities , Fetal Alcohol Spectrum Disorders/pathology , Gastrula/drug effects , Genetic Predisposition to Disease , Hedgehog Proteins/physiology , Holoprosencephaly/chemically induced , Holoprosencephaly/embryology , Humans , Maxillofacial Development/physiology , Neural Crest/drug effects , Neural Crest/pathology , Signal Transduction/drug effects , Skull/abnormalities , Species Specificity , Tretinoin/physiology , Tretinoin/toxicity , Wnt Signaling Pathway/drug effects
14.
Cell Biochem Funct ; 35(7): 433-440, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28961318

ABSTRACT

Progress in the understanding of the molecular mechanism for acute myeloid leukaemia is of great significance to generate new potential targets for treatment. Recent studies showed that HOXA9, a homeodomain-containing transcription factor, is commonly deregulated in acute leukaemia. In this study, we elucidated the direct correlation between HoxA9 expression and progression of leukaemia using 2 different types of leukaemia cells HL-60 and MOLT-3. The HoxA9 expression level was decreased in leukaemia cells with the treatment of all-trans retinoic acid or arsenic trioxide (As2 O3 ). Downregulation of HoxA9 could impair the proliferation and promote the leukaemia cell death. HoxA9 silencing also potentiated the differentiation of leukaemia cells, and in vivo studies demonstrated that HoxA9 downregulation could interfere the tumour growth. Interestingly, HoxA9 silencing also led to the alteration in miRNA expression, mediating the promoting effect on the leukaemia cell differentiation. Therefore, this work provided a promising and potentially efficient target to leukaemia treatment, indicating that HoxA9 is likely to be an ideal candidate in the gene therapy against acute myeloid leukaemia. In this study, we elucidated the critical role of HoxA9 in the proliferation and differentiation of leukaemia cells both in vitro and in vivo. The effect of HoxA9 modulation was correlated with the clinical effect of all-trans retinoic acid and As2 O3 . Furthermore, HoxA9 also regulated the miRNA expression, controlling the leukaemia cell differentiation. Therefore, this work provided new insights into molecular mechanism underlying the leukaemia treatment, potentially putting forward a brand new target to the gene therapy against leukaemia.


Subject(s)
Cell Differentiation , Cell Proliferation , Homeodomain Proteins/metabolism , Animals , Apoptosis/drug effects , Arsenic Trioxide , Arsenicals , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Down-Regulation/drug effects , HL-60 Cells , Homeodomain Proteins/antagonists & inhibitors , Homeodomain Proteins/genetics , Humans , Leukemia/metabolism , Leukemia/pathology , Male , Mice , Mice, Nude , MicroRNAs/metabolism , Oxides/toxicity , RNA Interference , RNA, Messenger/metabolism , Transplantation, Heterologous , Tretinoin/toxicity
15.
Ecotoxicol Environ Saf ; 138: 32-38, 2017 Apr.
Article in English | MEDLINE | ID: mdl-27992848

ABSTRACT

Teratogenic effects, which were remarkably similar to those induced by retinoic acids, have been seen in wild frogs indicating possible source of retinoids in the environment. Recent studies indicate that some cyanobacterial species can contain teratogenic retinoic acids (RAs) and their analogues. Retinoids are known to regulate important processes such as differentiation, development, and embryogenesis. The study investigated the effects of exudates (extracellular compounds) of two cyanobacteria species with retinoic-like activity and one algae species on embryonic development of amphibians. The retinoid-like activity determined by in vitro reporter gene assay reached 528ng retinoid equivalents (REQ)/L and 1000ng REQ/L in exudates of Cylindrospermopsis raciborskii and Microcystis aeruginosa, respectively, while algal exudates showed no detectable activity. Total mean of retinoid-like copounds into exudate was 35.6ng ATRA/mil.cells for M.aeruginosa and 6.71ng ATRA/mil.cells for C.raciborskii, respectively. Toxicity tests with amphibian embryos up to 96h of development were carried out according to the standard guide for the Frog Embryo Teratogenesis Assay Xenopus. Lowest observed effect concentrations (LOEC) of malformations (2.5-2.6µg/L REQ) were two times lower than LOEC for ATRA (5µg/L). The exudates of both cyanobacteria were indeed provoking diverse teratogenic effects (e.g. tail, gut and eyes deformation) and interference with growth in frogs embryos, while such effects were not observed for the algae. Xenopus embryos were also exposed to all-trans retinoic acid (ATRA) in concentration range (1-40µg/L) equivalent to the REQs detected in cyanobacterial exudates. ATRA (10µg/L) caused similar teratogenic phenotypes at corresponding REQs as cyanobacterial exudates. The study confirms the ability of some species of cyanobacteria to produce retinoids naturally and excrete them directly into the environment at concentrations which might have adverse influence on the development of amphibians.


Subject(s)
Cyanobacteria/metabolism , Embryonic Development/drug effects , Phytoplankton/metabolism , Teratogens/toxicity , Tretinoin/toxicity , Water Pollutants, Chemical/toxicity , Xenopus laevis/embryology , Animals , Biological Assay , Genes, Reporter/drug effects , Microcystis/drug effects , Tretinoin/metabolism
16.
Cleft Palate Craniofac J ; 54(4): 391-399, 2017 07.
Article in English | MEDLINE | ID: mdl-27487015

ABSTRACT

OBJECTIVE: To characterize the prenatal and postnatal craniofacial bone development in mouse model of all-trans retinoic acid (ATRA) exposure at different ages by a quantitative and morphological analysis of skull morphology. METHODS: Pregnant mice were exposed to ATRA at embryonic day 10 (E10) and 13 (E13) by oral gavage. Skulls of mice embryos at E19.5 and adult mice at postnatal day 35 (P35) were collected for high-resolution microcomputed tomography (microCT) imaging scanning and section HE staining. Reconstruction and measurement of mouse skulls were performed for prenatal and postnatal analysis of the control and ATRA-exposed mice. RESULTS: Craniofacial malformations in mouse models caused by ATRA exposure were age dependent. ATRA exposure at E10 induced cleft palate in 81.8% of the fetuses, whereas the palatine bone of E13-exposed mice was intact. Inhibitions of maxilla and mandible development with craniofacial asymmetry induced were observed at E19.5 and P35. Compared with control and E13-exposed mice, the palatine bones of E10-exposed mice were not elevated and were smaller in dimension. Some E10-exposed mice exhibited other craniofacial abnormalities, including premature fusion of mandibular symphysis with a missing mandibular incisor and a smaller mandible. Severe deviated snouts and amorphous craniofacial suture were detected in E13-exposed mice at P35. CONCLUSION: These morphological variations in E10- and E13-exposed mice suggested that ATRA was teratogenic in craniofacial bone development in mice and the effect was age dependent.


Subject(s)
Craniofacial Abnormalities/chemically induced , Tretinoin/toxicity , Age Factors , Animals , Animals, Newborn , Craniofacial Abnormalities/diagnostic imaging , Disease Models, Animal , Female , Mice , Pregnancy , Radiographic Image Interpretation, Computer-Assisted , X-Ray Microtomography
17.
Hippocampus ; 26(7): 911-23, 2016 07.
Article in English | MEDLINE | ID: mdl-26860546

ABSTRACT

Clinical studies have highlighted an association between retinoid treatment and depressive symptoms. As we had shown before that chronic application of all-trans retinoic acid (RA) potently activated the hypothalamus-pituitary-adrenal (HPA) stress axis, we here questioned whether RA also induced changes in adult hippocampal neurogenesis, a form of structural plasticity sensitive to stress and implicated in aspects of depression and hippocampal function. RA was applied intracerebroventricularly (i.c.v.) to adult rats for 19 days after which animals were subjected to tests for depressive-like behavior (sucrose preference) and spatial learning and memory (water maze) performance. On day 27, adult hippocampal neurogenesis and astrogliosis was quantified using BrdU (newborn cell survival), PCNA (proliferation), doublecortin (DCX; neuronal differentiation), and GFAP (astrocytes) as markers. RA was found to increase retinoic acid receptor-α (RAR-α) protein expression in the hippocampus, suggesting an activation of RA-induced signaling mechanisms. RA further potently suppressed cell proliferation, newborn cell survival as well as neurogenesis, but not astrogliosis. These structural plasticity changes were significantly correlated with scores for anhedonia, a core symptom of depression, but not with water maze performance. Our results suggest that RA-induced impairments in hippocampal neurogenesis correlate with depression-like symptoms but not with spatial learning and memory in this design. Thus, manipulations aimed to enhance neurogenesis may help ameliorate emotional aspects of RA-associated mood disorders. © 2016 Wiley Periodicals, Inc.


Subject(s)
Depression/chemically induced , Hippocampus/drug effects , Neurogenesis/drug effects , Neurons/drug effects , Tretinoin/toxicity , Anhedonia/drug effects , Anhedonia/physiology , Animals , Astrocytes/drug effects , Astrocytes/pathology , Astrocytes/physiology , Blotting, Western , Cell Survival/drug effects , Cell Survival/physiology , Depression/pathology , Depression/physiopathology , Dietary Sucrose , Doublecortin Protein , Gliosis/pathology , Gliosis/physiopathology , Hippocampus/pathology , Hippocampus/physiopathology , Immunohistochemistry , Male , Maze Learning/drug effects , Maze Learning/physiology , Neurogenesis/physiology , Neurons/pathology , Neurons/physiology , Random Allocation , Rats, Sprague-Dawley , Spatial Memory/drug effects , Spatial Memory/physiology , Taste Perception/drug effects , Taste Perception/physiology
18.
Biochem Biophys Res Commun ; 477(3): 322-8, 2016 08 26.
Article in English | MEDLINE | ID: mdl-27343556

ABSTRACT

Administration of all-trans retinoic acid (atRA) on E12.0 (embryonic day 12.0) leads to failure of medial edge epithelium (MEE) disappearance and cleft palate. However, the molecular mechanism underlying the relationship between atRA and MEE remains to be identified. In this study, atRA (200 mg/kg) administered by gavage induced a 75% incidence of cleft palate in C57BL/6 mice. Notch1 was up-regulated in MEE cells in the atRA-treated group compared with the controls at E15.0, together with reduced apoptosis and elevated proliferation. Next, we investigated the mechanisms underlying atRA, Notch1 and MEE degradation in palate organ culture. Our results revealed that down-regulation of Notch1 partially rescued the inhibition of atRA-induced palate fusion. Molecular analysis indicated that atRA increased the expression of Notch1 and Rbpj and decreased the expression of P21. In addition, depletion of Notch1 expression decreased the expression of Rbpj and increased the expression of P21. Moreover, inhibition of Rbpj expression partially reversed atRA-induced MEE persistence and increased P21 expression. These findings demonstrate that atRA inhibits MEE degradation, which in turn induces a cleft palate, possibly through the Notch1/RBPjk/P21 signaling pathway.


Subject(s)
Apoptosis , Cleft Palate/pathology , Receptor, Notch1/metabolism , Teratogens/toxicity , Tretinoin/toxicity , Animals , Cleft Palate/chemically induced , Epithelium/pathology , Female , Gene Knockdown Techniques , Mice , Mice, Inbred C57BL , Pregnancy , Receptor, Notch1/genetics
19.
J Exp Zool B Mol Dev Evol ; 326(8): 464-473, 2016 12.
Article in English | MEDLINE | ID: mdl-28097823

ABSTRACT

In marine aquaculture fish, excessive supplement of vitamin A (VA) to zooplanktons for larval culture and experimental exposure of larvae to retinoic acid (RA: active form of VA) have been known to cause vertebral deformity. However, the tissues in the developing vertebral column that are affected by RA and the progression of vertebral deformity remain undetermined. To examine these questions, we histologically traced the progress of vertebral deformity induced by RA in Japanese flounder (Paralichthys olivaceus). Larvae were exposed to RA for 3 days at mid-metamorphosis (G-stage), a critical stage for vertebral deformity. Intervertebral ligament, which is known to form intervertebral joints in cooperation with the notochord, was severely degenerated by RA, leading to fusion of centra. During further development to adult, growth of centra was severely suppressed in an anterior-posterior direction in RA-treated fish and the notochord tissue was lost from fused centra, resulting in complete loss of intervertebral joints and fusion of centra. We conclude that RA initially damages the intervertebral ligaments, and these defects lead to fusion, narrowing of centra, and loss of intervertebral joints in the vertebral column. The cumulative effect of these modifications is a truncated body form.


Subject(s)
Fish Diseases/chemically induced , Flounder , Ligaments/drug effects , Spine/drug effects , Tretinoin/toxicity , Animal Feed , Animals , Bone Development/drug effects , Dose-Response Relationship, Drug , Gene Expression Regulation, Developmental/drug effects , Larva/drug effects , Spine/pathology , Tretinoin/administration & dosage
20.
Pediatr Res ; 79(4): 614-20, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26672737

ABSTRACT

BACKGROUND: Low pulmonary retinol levels and disrupted retinoid signaling pathway (RSP) have been implicated in the pathogenesis of congenital diaphragmatic hernia (CDH) and associated pulmonary hypoplasia (PH). It has been demonstrated that nitrofen disturbs the main retinol-binding protein (RBP)-dependent trophoblastic retinol transport. Several studies have demonstrated that prenatal treatment with retinoic acid (RA) can reverse PH in the nitrofen-induced CDH model. We hypothesized that maternal administration of RA can increase trophoblastic RBP-dependent retinol transport in a nitrofen model of CDH. METHODS: Pregnant rats were treated with nitrofen or vehicle on gestational day 9 (D9) and sacrificed on D21. RA was given i.p. on D18, D19, and D20. Retinol and RA levels were measured using high-performance liquid chromatography. Immunohistochemistry was performed to evaluate trophoblastic expression of RBP. Expression levels of the primary RSP genes were determined using quantitative real-time PCR and immunohistochemistry. RESULTS: Markedly increased trophoblastic RBP immunoreactivity was observed in CDH+RA compared to CDH. Significantly increased serum and pulmonary retinol and RA levels were detected in CDH+RA compared to CDH. Pulmonary expression of RSP genes and proteins were increased in CDH+RA compared to CDH. CONCLUSION: Increased trophoblastic RBP expression and retinol transport after antenatal administration of RA suggest that retinol-triggered RSP activation may attenuate CDH-associated PH by elevating serum and pulmonary retinol levels.


Subject(s)
Disease Models, Animal , Hernias, Diaphragmatic, Congenital/metabolism , Lung/metabolism , Phenyl Ethers/toxicity , Retinol-Binding Proteins/metabolism , Tretinoin/toxicity , Trophoblasts/drug effects , Vitamin A/metabolism , Animals , Biological Transport , Female , Fetal Weight , Pregnancy , Rats , Trophoblasts/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL