Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Kidney Int ; 97(4): 753-764, 2020 04.
Article in English | MEDLINE | ID: mdl-32059999

ABSTRACT

Zinc fingers and homeoboxes (ZHX) proteins are heterodimeric transcriptional factors largely expressed at the cell membrane in podocytes in vivo. We found ZHX2-based heterodimers in podocytes, with ZHX2-ZHX1 predominantly at the cell membrane of the podocyte cell body, and ZHX2-ZHX3 at the slit diaphragm. In addition to changes in overall ZHX2 expression, there was increased podocyte nuclear ZHX3 and ZHX2 in patients with focal segmental glomerulosclerosis, and increased podocyte nuclear ZHX1 in patients with minimal change disease. Zhx2 deficient mice had increased podocyte ZHX1 and ZHX3 expression. Zhx2 deficient mice and podocyte specific Zhx2 overexpressing transgenic rats develop worse experimental focal segmental glomerulosclerosis than controls, with increased nuclear ZHX3 and ZHX2, respectively. By contrast, podocyte specific Zhx2 overexpressing transgenic rats develop lesser proteinuria during experimental minimal change disease due to peripheral sequestration of ZHX1 by ZHX2. Using co-immunoprecipitation, the interaction of ZHX2 with aminopeptidase A in the podocyte body cell membrane, and EPHRIN B1 in the slit diaphragm were noted to be central to upstream events in animal models of minimal change disease and focal segmental glomerulosclerosis, respectively. Mice deficient in Enpep, the gene for aminopeptidase A, and Efnb1, the gene for ephrin B1 developed worse albuminuria in glomerular disease models. Targeting aminopeptidase A in Zhx2 deficient mice with monoclonal antibodies induced albuminuria and upregulation of the minimal change disease mediator angiopoietin-like 4 through nuclear entry of ZHX1. Thus, podocyte ZHX2 imbalance is a critical factor in human glomerular disease, with minimal change disease disparities mediated mostly through ZHX1, and focal segmental glomerulosclerosis deviations through ZHX3 and ZHX2.


Subject(s)
Glomerulosclerosis, Focal Segmental , Homeodomain Proteins , Podocytes , Transcription Factors , Animals , Genes, Homeobox , Glomerulosclerosis, Focal Segmental/genetics , Homeodomain Proteins/genetics , Humans , Mice , Podocytes/metabolism , Transcription Factors/genetics , Zinc Fingers
2.
Toxicol Appl Pharmacol ; 307: 91-101, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27473015

ABSTRACT

Point mutations in the AR ligand-binding domain (LBD) can result in altered AR structures leading to changes of ligand specificity and functions. AR mutations associated to prostate cancer (PCa) have been shown to result in receptor activation by non-androgenic substances and anti-androgenic drugs. Two AR mutations known to alter the function of anti-androgens are the ART877A mutation, which is frequently detected mutation in PCa tumors and the ARW741C that is rare and has been derived in vitro following exposure of cells to the anti-androgen bicalutamide. AR activation by non-androgenic environmental substances has been suggested to affect PCa progression. In the present study we investigated the effect of AR mutations (ARW741C and ART877A) on the transcriptional activation following exposure of cells to an androgenic brominated flame retardant, 1,2-dibromo-4-(1,2 dibromoethyl) cyclohexane (TBECH, also named DBE-DBCH). The AR mutations resulted in higher interaction energies and increased transcriptional activation in response to TBECH diastereomer exposures. The ART877A mutation rendered AR highly responsive to low levels of DHT and TBECH and led to increased AR nuclear translocation. Gene expression analysis showed a stronger induction of AR target genes in LNCaP cells (ART877A) compared to T-47D cells (ARWT) following TBECH exposure. Furthermore, AR knockdown experiments confirmed the AR dependency of these responses. The higher sensitivity of ART877A and ARW741C to low levels of TBECH suggests that cells with these AR mutations are more susceptible to androgenic endocrine disrupters.


Subject(s)
Androgens/pharmacology , Cyclohexanes/pharmacology , Endocrine Disruptors/pharmacology , Flame Retardants/pharmacology , Prostatic Neoplasms/genetics , Receptors, Androgen/genetics , Cell Line, Tumor , Humans , Male , Mutation , Receptors, Androgen/metabolism
3.
JCI Insight ; 8(10)2023 05 22.
Article in English | MEDLINE | ID: mdl-37040185

ABSTRACT

Viral illnesses like SARS-CoV-2 have pathologic effects on nonrespiratory organs in the absence of direct viral infection. We injected mice with cocktails of rodent equivalents of human cytokine storms resulting from SARS-CoV-2/COVID-19 or rhinovirus common cold infection. At low doses, COVID-19 cocktails induced glomerular injury and albuminuria in zinc fingers and homeoboxes 2 (Zhx2) hypomorph and Zhx2+/+ mice to mimic COVID-19-related proteinuria. Common Cold cocktail induced albuminuria selectively in Zhx2 hypomorph mice to model relapse of minimal change disease, which improved after depletion of TNF-α, soluble IL-4Rα, or IL-6. The Zhx2 hypomorph state increased cell membrane to nuclear migration of podocyte ZHX proteins in vivo (both cocktails) and lowered phosphorylated STAT6 activation (COVID-19 cocktail) in vitro. At higher doses, COVID-19 cocktails induced acute heart injury, myocarditis, pericarditis, acute liver injury, acute kidney injury, and high mortality in Zhx2+/+ mice, whereas Zhx2 hypomorph mice were relatively protected, due in part to early, asynchronous activation of STAT5 and STAT6 pathways in these organs. Dual depletion of cytokine combinations of TNF-α with IL-2, IL-13, or IL-4 in Zhx2+/+ mice reduced multiorgan injury and eliminated mortality. Using genome sequencing and CRISPR/Cas9, an insertion upstream of ZHX2 was identified as a cause of the human ZHX2 hypomorph state.


Subject(s)
COVID-19 , Common Cold , Humans , Mice , Animals , Homeodomain Proteins/genetics , Albuminuria , Tumor Necrosis Factor-alpha , Cytokine Release Syndrome , SARS-CoV-2/metabolism , Transcription Factors/genetics
4.
Environ Pollut ; 284: 117091, 2021 Sep 01.
Article in English | MEDLINE | ID: mdl-33901980

ABSTRACT

1,2-dibromo-4-(1,2-dibromoethyl)-cyclohexane (DBE-DBCH) is a brominated flame retardant used in commercial and industrial applications. The use of DBE-DBCH containing products has resulted in an increased release into the environment. However, limited information is available on the long-term effects of DBE-DBCH and its effects in aquatic invertebrates. Thus, the present study was aimed at determining how DBE-DBCH diastereomers (αß and γδ) affects aquatic invertebrates using Daphnia magna as a model organism. Survival, reproduction, feeding, swimming behavior and toxicogenomic responses to environmental relevant concentrations of DBE-DBCH were analyzed. Chronic exposure to DBE-DBCH resulted in decreased lifespan, and reduced fecundity. Expression of genes involved in reproductive processes, vtg1 and jhe, were also inhibited. DBE-DBCH also induced hypoxia by inhibiting the transcription of genes involved in heme biosynthesis and oxygen transport. Furthermore, DBE-DBCH also inhibited feeding resulting in emptiness of the alimentary canal. Increased expression of the stress response biomarkers was observed following DBE-DBCH exposure. In addition, DBE-DBCH diastereomers also altered the swimming behavior of Daphnia magna. The present study demonstrates that DBE-DBCH cause multiple deleterious effects on Daphnia magna, including effects on reproduction and hormonal systems. These endocrine disrupting effects are in agreement with effects observed on vertebrates. Furthermore, as is the case in vertebrates, DBE-DBCH γδ exerted stronger effects than DBE-DBCH αß on Daphnia magna. This indicate that DBE-DBCH γδ has properties making it more toxic to all so far studied animals than DBE-DBCH αß.


Subject(s)
Flame Retardants , Water Pollutants, Chemical , Animals , Daphnia/genetics , Endocrine System , Gene Expression , Reproduction , Water Pollutants, Chemical/toxicity
5.
Sci Rep ; 8(1): 4843, 2018 03 19.
Article in English | MEDLINE | ID: mdl-29556062

ABSTRACT

Endocrine disrupting compounds can interfere with androgen receptor (AR) signaling and disrupt steroidogenesis leading to reproductive failure. The brominated flame-retardant (BFR) 1, 2-dibromo-4-(1, 2-dibromoethyl) cyclohexane (TBECH), is an agonist to human, chicken and zebrafish AR. Recently another group of alternative BFRs, allyl 2, 4, 6-tribromophenyl ether (ATE), and 2, 3-dibromopropyl 2, 4, 6-tribromophenyl ether (DPTE) along with its metabolite 2-bromoallyl 2, 4, 6-tribromophenyl ether (BATE) were identified as potent human AR antagonists. These alternative BFRs are present in the environment. The aim of the present study was to determine the effect of mixed exposures to the AR agonist and the AR antagonists at environmentally relevant concentrations. In vitro reporter luciferase assay showed that the AR antagonists, when present at concentration higher than TBECH, were able to inhibit TBECH-mediated AR activity. These AR antagonists also promoted AR nuclear translocation. In vitro gene expression analysis in the non-tumorigenic human prostate epithelial cell RWPE1 showed that TBECH induced AR target genes whereas DPTE repressed these genes. Further analysis of steroidogenic genes showed that TBECH up-regulated most of the genes while DPTE down-regulated the same genes. The results indicate that when TBECH and DPTE are present together they will antagonize each other, thereby reducing their individual effects.


Subject(s)
Flame Retardants/pharmacology , Halogenation , Receptors, Androgen/metabolism , Active Transport, Cell Nucleus/drug effects , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Drug Interactions , Gene Expression Regulation/drug effects , HeLa Cells , Humans , RNA, Messenger/genetics , Receptors, Androgen/genetics , Steroids/biosynthesis
6.
Nat Commun ; 8(1): 1739, 2017 11 23.
Article in English | MEDLINE | ID: mdl-29170386

ABSTRACT

The glucocorticoid (GC) receptor (GR) suppresses inflammation by activating anti-inflammatory and repressing pro-inflammatory genes. GR-interacting protein-1 (GRIP1) is a GR corepressor in macrophages, however, whether GRIP1 mediates GR-activated transcription, and what dictates its coactivator versus corepressor properties is unknown. Here we report that GRIP1 loss in macrophages attenuates glucocorticoid induction of several anti-inflammatory targets, and that GC treatment of quiescent macrophages globally directs GRIP1 toward GR binding sites dominated by palindromic GC response elements (GRE), suggesting a non-redundant GRIP1 function as a GR coactivator. Interestingly, GRIP1 is phosphorylated at an N-terminal serine cluster by cyclin-dependent kinase-9 (CDK9), which is recruited into GC-induced GR:GRIP1:CDK9 hetero-complexes, producing distinct GRE-specific GRIP1 phospho-isoforms. Phosphorylation potentiates GRIP1 coactivator but, remarkably, not its corepressor properties. Consistently, phospho-GRIP1 and CDK9 are not detected at GR transrepression sites near pro-inflammatory genes. Thus, GR restricts actions of its own coregulator via CDK9-mediated phosphorylation to a subset of anti-inflammatory genes.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Carrier Proteins/metabolism , Cyclin-Dependent Kinase 9/metabolism , Glucocorticoids/metabolism , Macrophages/metabolism , Nerve Tissue Proteins/metabolism , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/genetics , Animals , Binding Sites/genetics , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Cell Line , Cells, Cultured , Dexamethasone/pharmacology , Gene Knockdown Techniques , Glucocorticoids/pharmacology , Humans , Inflammation/genetics , Inflammation/metabolism , Macrophages/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Phosphorylation , Receptors, Glucocorticoid/metabolism , Response Elements , Transcriptional Activation
7.
Environ Int ; 74: 60-70, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25454221

ABSTRACT

Brominated flame-retardants (BFRs) are used in industrial products to reduce the risk of fire. However, their continuous release into the environment is a concern as they are often persistent, bioaccumulating and toxic. Information on the impact these compounds have on human health and wildlife is limited and only a few of them have been identified to disrupt hormone receptor functions. In the present study we used in silico modeling to determine the interactions of selected BFRs with the human androgen receptor (AR). Three compounds were found to dock into the ligand-binding domain of the human AR and these were further tested using in vitro analysis. Allyl 2,4,6-tribromophenyl ether (ATE), 2-bromoallyl 2,4,6-tribromophenyl ether (BATE) and 2,3-dibromopropyl-2,4,6-tribromophenyl ether (DPTE) were observed to act as AR antagonists. These BFRs have recently been detected in the environment, in house dust and in aquatic animals. The compounds have been detected at high concentrations in both blubber and brain of seals and we therefore also assessed their impact on the expression of L-type amino acid transporter system (LAT) genes, that are needed for amino acid uptake across the blood-brain barrier, as disruption of LAT gene function has been implicated in several brain disorders. The three BFRs down-regulated the expression of AR target genes that encode for prostate specific antigen (PSA), 5α-reductases and ß-microseminoprotein. The potency of PSA inhibition was of the same magnitude as the common prostate cancer drugs, demonstrating that these compounds are strong AR antagonists. Western blot analysis of AR protein showed that ATE, BATE and DPTE decreased the 5α-dihydrotestosterone-induced AR protein levels, further confirming that these BFRs act as AR antagonists. The transcription of the LAT genes was altered by the three BFRs, indicating an effect on amino-acid uptake across cellular membranes and blood-brain barrier. This study demonstrated that ATE, BATE and DPTE are potent AR antagonists and the alterations in LAT gene transcription suggest that these compounds can affect neuronal functions and should be considered as potential neurotoxic and endocrine disrupting compounds.


Subject(s)
Androgen Receptor Antagonists/pharmacology , Flame Retardants/pharmacology , Hydrocarbons, Brominated/pharmacology , Amino Acid Transport System y+L/biosynthesis , Amino Acid Transport System y+L/genetics , Androgen Receptor Antagonists/chemistry , Androgen Receptor Antagonists/metabolism , Blood-Brain Barrier/metabolism , Cell Line, Tumor , Endocrine Disruptors/chemistry , Flame Retardants/metabolism , Humans , Hydrocarbons, Brominated/chemistry , Hydrocarbons, Brominated/metabolism , Male , Molecular Docking Simulation , Prostate-Specific Antigen/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Receptors, Androgen/chemistry , Receptors, Androgen/metabolism , Transcriptional Activation/drug effects
8.
Toxicol In Vitro ; 29(8): 1993-2000, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26318274

ABSTRACT

Increased exposure of birds to endocrine disrupting compounds has resulted in developmental and reproductive dysfunctions. We have recently identified the flame retardants, allyl-2,4,6-tribromophenyl ether (TBP-AE), 2-3-dibromopropyl-2,4,6-tribromophenyl ether (TBP-DBPE) and the TBP-DBPE metabolite 2-bromoallyl-2,4,6-tribromophenyl ether (TBP-BAE) as antagonists to both the human androgen receptor (AR) and the zebrafish AR. In the present study, we aimed at determining whether these compounds also interact with the chicken AR. In silico modeling studies showed that TBP-AE, TBP-BAE and TBP-DBPE were able to dock into to the chicken AR ligand-binding pocket. In vitro transfection assays revealed that all three brominated compounds acted as chicken AR antagonists, inhibiting testosterone induced AR activation. In addition, qRT-PCR studies confirmed that they act as AR antagonists and demonstrated that they also alter gene expression patterns of apoptotic, anti-apoptotic, drug metabolizing and amino acid transporter genes. These studies, using chicken LMH cells, suggest that TBP-AE, TBP-BAE and TBP-DBPE are potential endocrine disrupters in chicken.


Subject(s)
Androgen Receptor Antagonists/pharmacology , Chickens , Endocrine Disruptors/toxicity , Flame Retardants/toxicity , Hydrocarbons, Brominated/toxicity , Phenyl Ethers/toxicity , Receptors, Androgen/metabolism , Animals , Carcinoma, Hepatocellular , Cell Line, Tumor , Computer Simulation , Liver Neoplasms , Models, Chemical , Protein Binding
9.
Chem Biol Interact ; 233: 35-45, 2015 May 25.
Article in English | MEDLINE | ID: mdl-25818047

ABSTRACT

The brominated flame retardants (BFRs) 1,2-dibromo-4-(1,2-dibromoethyl)cyclohexane (TBECH or DBE-DCBH) and allyl 2,4,6-tribromophenyl ether (ATE or TBP-AE) are alternative BFRs that have been introduced to replace banned BFRs. TBECH is a potential endocrine disrupter in human, chicken and zebrafish and in a recent study we showed that ATE, along with the structurally similar BFR 2,3-dibromopropyl 2,4,6-tribromophenyl ether (DPTE or TBP-DBPE) and its metabolite 2-bromoallyl 2,4,6-tribromophenyl ether (BATE or TBP-BAE) are potential endocrine and neuronal disrupters in human. In this study we analyzed ATE, BATE and DPTE for zebrafish androgen receptor (zAR) modulating properties. In silico analysis with two softwares, Molecular Operating Environment (MOE) and Internal Coordinate Mechanics (ICM), showed that ATE, BATE and DPTE bind to zAR. In vitro AR activation assay revealed that these three BFRs down-regulate 11-ketotestosterone (KT) mediated zAR activation. Exposure to 10 µM DPTE resulted in reduced hatching success and like TBECH, BATE and DPTE at 10 µM also had teratogenic properties with 20% and 50% back-bone curvature respectively. Gene transcription analysis in zebrafish embryos as well as in juveniles showed down-regulation of the androgen receptor and androgen response genes, which further support that these BFRs are androgen antagonists and potential endocrine disrupting compounds. Genes involved in steroidogenesis were also down-regulated by these BFRs. In view of this, the impact of these BFRs on humans and wildlife needs further analysis.


Subject(s)
Androgen Antagonists/toxicity , Flame Retardants/toxicity , Hydrocarbons, Brominated/toxicity , Teratogens/toxicity , Zebrafish/embryology , Zebrafish/physiology , Androgen Antagonists/metabolism , Animals , Environmental Monitoring , Female , Flame Retardants/metabolism , Gene Expression Regulation, Developmental/drug effects , Halogenation , Hydrocarbons, Brominated/metabolism , Male , Molecular Docking Simulation , Receptors, Androgen/chemistry , Receptors, Androgen/metabolism , Teratogens/metabolism , Zebrafish/genetics
10.
Environ Toxicol Chem ; 33(4): 891-9, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24375616

ABSTRACT

The incorporation of brominated flame retardants into industrial and household appliances has increased their occurrence in the environment, resulting in deleterious effects on wildlife. With the increasing restraints on available compounds, there has been a shift to using brominated flame retardants that has seen the production of alternative brominated flame retardants such as 1,2-dibromo-4-(1,2 dibromoethyl) cyclohexane (TBECH), which has been detected in the environment. In previous in silico and in vitro studies the authors have shown that TBECH can activate both the human androgen receptor (hAR) and the zebrafish AR (zAR) suggesting that it is a potential endocrine disruptor. The present study was aimed at determining the interaction of TBECH with the chicken AR (cAR). In the present study, TBECH bound to cAR, but in vitro activation assay studies using the chicken LMH cell line showed it had a potency of only 15% compared with testosterone. Sequence difference between ARs from different species may contribute to the different responses to TBECH. Further quantitative reverse-transcriptase polymerase chain reaction (qRT-PCR) analysis showed that TBECH interacted with and altered the expression of both thyroid receptors and estrogen receptors. In addition, the qRT-PCR analysis showed that TBECH altered the transcription pattern of genes involved in inflammatory, apoptotic, proliferative, DNA methylation, and drug-metabolizing pathways. This demonstrates that TBECH, apart from activating cAR, can also influence multiple biological pathways in the chicken.


Subject(s)
Cyclohexanes/toxicity , Flame Retardants/toxicity , Receptors, Androgen/metabolism , Animals , Cell Line, Tumor , Chickens , Gene Expression Regulation/drug effects , Receptors, Androgen/genetics
11.
Toxicol Sci ; 141(1): 218-33, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24958932

ABSTRACT

The risk posed by complex chemical mixtures in the environment to wildlife and humans is increasingly debated, but has been rarely tested under environmentally relevant scenarios. To address this issue, two mixtures of 14 or 19 substances of concern (pesticides, pharmaceuticals, heavy metals, polyaromatic hydrocarbons, a surfactant, and a plasticizer), each present at its safety limit concentration imposed by the European legislation, were prepared and tested for their toxic effects. The effects of the mixtures were assessed in 35 bioassays, based on 11 organisms representing different trophic levels. A consortium of 16 laboratories was involved in performing the bioassays. The mixtures elicited quantifiable toxic effects on some of the test systems employed, including i) changes in marine microbial composition, ii) microalgae toxicity, iii) immobilization in the crustacean Daphnia magna, iv) fish embryo toxicity, v) impaired frog embryo development, and vi) increased expression on oxidative stress-linked reporter genes. Estrogenic activity close to regulatory safety limit concentrations was uncovered by receptor-binding assays. The results highlight the need of precautionary actions on the assessment of chemical mixtures even in cases where individual toxicants are present at seemingly harmless concentrations.


Subject(s)
Biological Assay/methods , Conservation of Natural Resources/legislation & jurisprudence , Environmental Monitoring , Toxicity Tests/methods , Water Pollutants, Chemical/toxicity , Animals , Environmental Monitoring/legislation & jurisprudence , Environmental Monitoring/methods , European Union , Government Regulation , Humans , Water Pollutants, Chemical/chemistry
12.
Aquat Toxicol ; 142-143: 63-72, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-23958786

ABSTRACT

Tetrabromoethylcyclohexane (TBECH) is a brominated flame retardant that has been shown to be a potent agonist to the human androgen receptor (AR). However, while it is present in the environment, it is not known if it interacts with AR from aquatic species. The present study was therefore aimed at improving our understanding of how TBECH affects aquatic animals using zebrafish as a model organism. In silico modeling demonstrated that TBECH diastereomers bind to the zebrafish androgen receptor (zAR) and in vitro and in vivo data showed that TBECH has androgenic properties. Deleterious effects of TBECH were studied on embryonic and juvenile zebrafish and qRT-PCR analysis in vitro and in vivo was performed to determine TBECH effects on gene regulation. TBECH was found to delay hatching at 1 µM and 10 µM doses while morphological abnormalities and juvenile mortality was observed at 10 µM. The qRT-PCR analysis showed alterations of multiple genes involved in chondrogenesis (cartilage development), metabolism and stress response. Thus, TBECH induces androgenic activity and has negative effects on zebrafish physiology and therefore its impact on the environment should be carefully monitored.


Subject(s)
Cyclohexanes/toxicity , Gene Expression Regulation/drug effects , Receptors, Androgen/genetics , Water Pollutants, Chemical/toxicity , Zebrafish/physiology , Animals , Cell Line , Embryo, Nonmammalian , Inhibitory Concentration 50 , Zebrafish/embryology
SELECTION OF CITATIONS
SEARCH DETAIL