[go: up one dir, main page]

 
 
Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (96)

Search Parameters:
Keywords = pregnane X receptor

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
12 pages, 2364 KiB  
Communication
Pregnenolone 16-Alpha Carbonitrile, an Agonist of Rodent Pregnane X Receptor, Regulates Testosterone Biosynthesis in Rodent Leydig Cells
by Julia M. Salamat, Elizabeth M. Ayala, Chen-Che J. Huang, Frank S. Wilbanks, Rachel C. Knight, Benson T. Akingbemi and Satyanarayana R. Pondugula
J. Xenobiot. 2024, 14(3), 1256-1267; https://doi.org/10.3390/jox14030071 - 16 Sep 2024
Viewed by 673
Abstract
Leydig cells (LCs) in the testes produce the male sex hormone testosterone (T). Several xenobiotics, including clinical drugs, supplements, and environmental chemicals, are known to disrupt T homeostasis. Notably, some of these xenobiotics are known to activate the pregnane X receptor (PXR), a [...] Read more.
Leydig cells (LCs) in the testes produce the male sex hormone testosterone (T). Several xenobiotics, including clinical drugs, supplements, and environmental chemicals, are known to disrupt T homeostasis. Notably, some of these xenobiotics are known to activate the pregnane X receptor (PXR), a ligand-dependent nuclear receptor. However, it is currently unknown whether PXR is expressed in LCs and whether PXR activation alters T synthesis in rodent LCs. Therefore, in this study, we sought to determine whether PXR is expressed in rodent LCs and whether pregnenolone 16-alpha carbonitrile (PCN), the prototype agonist of rodent PXR, regulates T biosynthesis in rodent LCs. Hormonal as well as protein and gene expression analyses were conducted in rat primary LCs and MA-10 mouse Leydig cells. Results showed that PXR was expressed at the mRNA and protein level in both rat primary LCs and MA-10 cells. Incubation of rat primary LCs with PCN resulted in a significant decrease in T secretion. This PCN-induced decrease in T secretion was associated with decreased protein expression of key steroidogenic enzymes such as 3β-HSD and CYP17A1. RNA-seq results from MA-10 cells showed that PCN down-regulated the transcripts of steroidogenic enzymes and proteins involved in the T synthesis pathway. Together, these results suggest that PCN, an agonist of rodent PXR, can regulate T biosynthesis in rodent LCs by down-regulating the expression of the steroidogenic enzymes involved in T biosynthesis. Our results are significant as they provide a potential novel mechanism for disruption of testosterone homeostasis by a variety of xenobiotics. Full article
Show Figures

Figure 1

Figure 1
<p><b>PXR protein expression in rodent LCs.</b> (<b>A</b>). Protein expression of rPXR in rat primary LCs was determined by Western blotting analysis (<span class="html-italic">n</span> = 2). Image shown is from a representative experiment. Lane 1, dog liver; Lane 2, COS-7 cells transfected with pcDNA; Lane 3, COS-7 cells transfected with FLAG-pcDNA; Lane 4, COS-7 cells transfected with human PXR (hPXR); Lane 5, COS-7 cells transfected with FLAG-hPXR; Lane 6, COS-7 cells transfected with 3XFLAG-hPXR; and Lanes 7 to 9, rat primary LCs. Marker molecular weights represent KDa. (<b>B</b>). Protein expression of mPXR in mouse MA-10 cells was determined by Western blotting analysis (<span class="html-italic">n</span> = 2). Image shown is from a representative experiment. All Lanes (1 to 7), MA-10 cells. Marker molecular weights represent KDa.</p>
Full article ">Figure 2
<p><b>Effect of pharmacological activation of rPXR on testosterone secretion by rat primary LCs.</b> (<b>A</b>). LCs were isolated from 35-day-old male rats. Pooled LCs were incubated in the culture media and treated with either DMSO (<span class="html-italic">n</span> = 6) or 10 µM PCN (<span class="html-italic">n</span> = 6) for 24 h. The media was aspirated and collected after 24 h treatments to measure the T levels using RIA. Data represent mean ± SD from six independent experiments. Statistical significance was determined using an unpaired Students <span class="html-italic">t</span> test. (<b>B</b>). Both DMSO and PCN-treated rat primary LCs were incubated in the culture medium without (basal) (<span class="html-italic">n</span> = 3) or with 100 ng/mL LH (LH) (<span class="html-italic">n</span> = 3) for 3 h. The media aliquots were collected to determine T production using RIA. Results are shown as the mean ± S.D. Determined by ANOVA and a Tukey’s multiple comparisons test.</p>
Full article ">Figure 3
<p><b>Effect of pharmacological activation of rPXR on protein expression of key enzymes involved in testosterone biosynthesis in rat primary LCs</b>. Western blots (<span class="html-italic">n</span> = 2) showing the protein expression of 3β-HSD, CYP17A1, CYP19, StAR, and β-actin in rat primary LCs treated by DMSO or 10 µM PCN for 24 h. Data shown are from representative experiments.</p>
Full article ">Figure 4
<p><b>RNA-seq analysis identifies 1552 DEGs in MA-10 cells.</b> (<b>A</b>) Principal component analysis and (<b>B</b>) dendrogram showing the clustering pattern of four RNA-seq samples. (<b>C</b>) MA plot and (<b>D</b>) volcano plot provide an overview of PCN (10 µM)-induced DEGs in an MA-10 cell transcriptome. (<b>E</b>) Gene ontology (GO) analysis shows top GO terms linked to up- and down-regulated DEGs.</p>
Full article ">Figure 5
<p><b>Heatmaps of genes associated with (A) steroidogenesis and (B) cholesterol metabolism in MA-10 cells.</b> Heatmaps show diverse expression levels (log<sub>2</sub> fold-change in FPKM value). To reduce the influence of low-expression genes, 10 was added to each FPKM value before calculating the fold change. C, Control (DMSO); P, PCN (10 µM).</p>
Full article ">
15 pages, 1520 KiB  
Article
Nuclear Receptors and Stress Response Pathways Associated with the Development of Oral Mucositis Induced by Antineoplastic Agents
by Moena Kagaya and Yoshihiro Uesawa
Pharmaceuticals 2024, 17(8), 1086; https://doi.org/10.3390/ph17081086 - 20 Aug 2024
Viewed by 803
Abstract
Oral mucositis (OM) is one of the common adverse events associated with cancer treatment that decreases the quality of life and affects treatment outcomes. However, the medications used to manage OM are generally only palliative, and our knowledge of the syndrome is limited. [...] Read more.
Oral mucositis (OM) is one of the common adverse events associated with cancer treatment that decreases the quality of life and affects treatment outcomes. However, the medications used to manage OM are generally only palliative, and our knowledge of the syndrome is limited. The etiology of the syndrome is thought to be complex and multifactorial. We investigated the trends and characteristics of OM and estimated molecular initiating events (MIEs) associated with the development of the syndrome using the FDA Adverse Event Reporting System. The study of trends and characteristics suggested that OM is significantly more likely to occur in females and nonelderly patients and is likely to be induced by protein kinase inhibitors such as afatinib and everolimus. Next, we used Toxicity Predictor, an in-house quantitative structure–activity relationship system, to estimate OM-associated MIEs. The results revealed that the agonist activity of the human pregnane X receptor, thyroid-stimulating hormone-releasing hormone receptor, and androgen receptor may be associated with OM development. Our study findings are expected to help avoid the risk of OM induction during the drug discovery process and clinical use of antineoplastic agents. Full article
(This article belongs to the Special Issue Data-Driven Biomarker and Drug Discovery for Complex Disease)
Show Figures

Figure 1

Figure 1
<p>Flowchart of the creation of a data table for analysis. DRUG: drug information, REAC: adverse reaction information, and DEMO: patient demographic information. Duplicate data in the DRUG, REAC, and DEMO tables were deleted and combined using primary IDs.</p>
Full article ">Figure 2
<p>The number of reports with 17 preferred terms related to stomatitis, defined using high-level terms of MedDRA.</p>
Full article ">Figure 3
<p>Correlation between antineoplastic agents and stomatitis. The <span class="html-italic">x</span>-axis shows lnROR; the <span class="html-italic">y</span>-axis shows the ordinary logarithm of the reciprocal <span class="html-italic">p</span>-value of Fisher’s exact test (−log [<span class="html-italic">p</span>-Value]). The reporting odds ratio (ROR) was calculated from cross-tabulation tables. The dotted line on the <span class="html-italic">x</span>-axis indicates lnROR = 0; the dotted line on the <span class="html-italic">y</span>-axis indicates <span class="html-italic">p</span> = 0.5. The colors in the plots indicate the ordinary logarithm of the total number of adverse events reported for each drug.</p>
Full article ">Figure 4
<p>MIEs associated with OM. The vertical axis shows lnROR and the horizontal axis shows MIE activity or not: MIE activity values &lt; 0.5 indicate no activity with a predictive label of “inactive”, whereas MIE activity values &gt; 0.5 indicate activity with a predictive label of “active”. The green line shows the mean value of lnROR and its 95% confidence interval. Each plot represents the antineoplastic agent under analysis. PXR, human pregnane X receptor; TRHR, thyrotropin-releasing hormone receptor; ARlbd, androgen receptor ligand-binding domain; ARant, androgen receptor with antagonist.</p>
Full article ">
18 pages, 3252 KiB  
Review
5β-Dihydrosteroids: Formation and Properties
by Trevor M. Penning and Douglas F. Covey
Int. J. Mol. Sci. 2024, 25(16), 8857; https://doi.org/10.3390/ijms25168857 - 14 Aug 2024
Viewed by 601
Abstract
5β-Dihydrosteroids are produced by the reduction of Δ4-3-ketosteroids catalyzed by steroid 5β-reductase (AKR1D1). By analogy with steroid 5α-reductase, genetic deficiency exists in AKR1D1 which leads to errors in newborn metabolism and in this case to bile acid deficiency. Also, like the [...] Read more.
5β-Dihydrosteroids are produced by the reduction of Δ4-3-ketosteroids catalyzed by steroid 5β-reductase (AKR1D1). By analogy with steroid 5α-reductase, genetic deficiency exists in AKR1D1 which leads to errors in newborn metabolism and in this case to bile acid deficiency. Also, like the 5α-dihydrosteroids (e.g., 5α-dihydrotestosterone), the 5β-dihydrosteroids produced by AKR1D1 are not inactive but regulate ligand access to nuclear receptors, can act as ligands for nuclear and membrane-bound receptors, and regulate ion-channel opening. For example, 5β-reduction of cortisol and cortisone yields the corresponding 5β-dihydroglucocorticoids which are inactive on the glucocorticoid receptor (GR) and provides an additional mechanism of pre-receptor regulation of ligands for the GR in liver cells. By contrast, 5β-pregnanes can act as neuroactive steroids at the GABAA and NMDA receptors and at low-voltage-activated calcium channels, act as tocolytic agents, have analgesic activity and act as ligands for PXR, while bile acids act as ligands for FXR and thereby control cholesterol homeostasis. The 5β-androstanes also have potent vasodilatory properties and work through blockade of Ca2+ channels. Thus, a preference for 5β-dihydrosteroids to work at the membrane level exists via a variety of mechanisms. This article reviews the field and identifies gaps in knowledge to be addressed in future research. Full article
(This article belongs to the Special Issue Molecular Insights in Steroid Biosynthesis and Metabolism)
Show Figures

Figure 1

Figure 1
<p>Bent steroid configuration seen in 5β-dihydrosteroids.</p>
Full article ">Figure 2
<p>Metabolism of Δ<sup>4</sup>-3-ketosteroids to tetrahydrosteroids. The sequential role of aldo-keto reductases is illustrated. Reproduced with permission from Endocrine Society [<a href="#B5-ijms-25-08857" class="html-bibr">5</a>].</p>
Full article ">Figure 3
<p>AKR1D1 splice variants. Reproduced with permission from <span class="html-italic">Steroids</span> [<a href="#B14-ijms-25-08857" class="html-bibr">14</a>].</p>
Full article ">Figure 4
<p>Control of ligand access to the glucocorticoid receptor in liver cells.</p>
Full article ">Figure 5
<p>Biosynthesis of 5β-pregnanes from progesterone.</p>
Full article ">Figure 6
<p>Bioactive 5β-dihydrosteroids.</p>
Full article ">Figure 7
<p>Allopregnanolone, <span class="html-italic">ent</span>-allopregnanolone and pregnanolone, <span class="html-italic">ent</span>-pregnanolone. The plane of the page is the mirror plane with allopregnanolone and pregnanolone behind the plane of the page and the <span class="html-italic">ent</span>-allopregnanolone and <span class="html-italic">ent</span>-pregnanolone in front of the plane of the page. Overlay of the respective enantiomer pairs would superimpose the A and C rings as well as the 18 and 19 methyl groups in each enantiomer pair.</p>
Full article ">Figure 8
<p>Properties of <span class="html-italic">ent</span>-steroids. The figure summarizes various effects where the enantiomers of AlloP (allopregnanolone) have been compared, including effects with enantioselectivity (<span class="html-italic">nat &gt; ent</span>), and effects where the enantiomers are equivalent (<span class="html-italic">nat = ent</span>). Reproduced with permission from <span class="html-italic">Neuroscience Biohav. Res</span> [<a href="#B60-ijms-25-08857" class="html-bibr">60</a>].</p>
Full article ">
30 pages, 9017 KiB  
Article
Large-Scale Screening of Per- and Polyfluoroalkyl Substance Binding Interactions and Their Mixtures with Nuclear Receptors
by Saptarshi Roy, James Moran, Keerthana Danasekaran, Kate O’Brien and Sivanesan Dakshanamurthy
Int. J. Mol. Sci. 2024, 25(15), 8241; https://doi.org/10.3390/ijms25158241 - 28 Jul 2024
Viewed by 976
Abstract
Despite their significant impact, comprehensive screenings and detailed analyses of per- and polyfluoroalkyl substance (PFAS) binding strengths at the orthosteric and allosteric sites of NRs are currently lacking. This study addresses this gap by focusing on the binding interaction analysis of both common [...] Read more.
Despite their significant impact, comprehensive screenings and detailed analyses of per- and polyfluoroalkyl substance (PFAS) binding strengths at the orthosteric and allosteric sites of NRs are currently lacking. This study addresses this gap by focusing on the binding interaction analysis of both common and uncommon PFAS with the nuclear receptors (NRs) vitamin D receptor (VDR), peroxisome proliferator-activated receptor gamma (PPARγ), pregnane X receptor (PXR), and estrogen receptor alpha (ERα). Advanced docking simulations were used to screen 9507 PFAS chemicals at the orthosteric and allosteric sites of PPARγ, PXR, VDR, and ERα. All receptors exhibited strong binding interactions at the orthosteric and allosteric site with a significant number of PFAS. We verified the accuracy of the docking protocol through multiple docking controls and validations. A mixture modeling analysis indicates that PFAS can bind in various combinations with themselves and endogenous ligands simultaneously, to disrupt the endocrine system and cause carcinogenic responses. These findings reveal that PFAS can interfere with nuclear receptor activity by displacing endogenous or native ligands by binding to the orthosteric and allosteric sites. The purpose of this study is to explore the mechanisms through which PFAS exert their endocrine-disrupting effects, potentially leading to more targeted therapeutic strategies. Importantly, this study is the first to explore the binding of PFAS at allosteric sites and to model PFAS mixtures at nuclear receptors. Given the high concentration and persistence of PFAS in humans, this study further emphasizes the urgent need for further research into the carcinogenic mechanisms of PFAS and the development of therapeutic strategies that target nuclear receptors. Full article
(This article belongs to the Section Biochemistry)
Show Figures

Figure 1

Figure 1
<p>(<b>a</b>) The redocked calcitriol (pink) and native co-crystalized structure of calcitriol (green) (PDB ID: 1IE9); The blue dotted lines are the hydrogen bonds. (<b>b</b>) The redocked lithocholic acid (pink) and native co-crystalline structure of lithocholic acid (green) are displayed (PDB ID: 4Q0A). The blue dotted lines are the hydrogen bonds.</p>
Full article ">Figure 2
<p>(<b>a</b>) Superimposed co-crystal structure in blue (PDB ID: 3ET3) and docked binding pose of ET1 in red. Amino acid residues within 5 Å of co-crystal structure are shown in stick form. (<b>b</b>) Superimposed co-crystal structure in blue (PDB ID: 5GTO) and docked binding pose of T35 in red. Amino acid residues within 5 Å of co-crystal structure are shown in stick form.</p>
Full article ">Figure 3
<p>(<b>a</b>) Superimposed conformation of redocked 4WH ligand onto co-crystallized 4WH ligand on PXR protein. The orange ligand represents the redocked ligand. Amino acid residues within distance of 5 Å are labeled. (<b>b</b>) Superimposed conformation of redocked glycerol ligand onto co-crystallized allosteric site on PXR protein. Light blue ligand represents redocked ligand. Amino acid residues within distance of 5 Å are labeled.</p>
Full article ">Figure 4
<p>(<b>a</b>) The redocked estradiol is displayed at the orthosteric site. (<b>b</b>) The redocked SRC-1 is displayed at the allosteric site. The yellow dotted lines are the hydrogen bonds. The amino acid residues involved with hydrogen bonding are displayed.</p>
Full article ">Figure 5
<p>(<b>1a</b>) The top PFAS chemical (blue) ranked by the docking score at the orthosteric site is displayed, Perfluoroperhydrobenzyl tetralin, under the classification of Polycyclic Perfluoroalkane. (<b>1b</b>) Perfluoroperhydrobenzyl tetralin and the redocked calcitriol (purple) are displayed. (<b>2a</b>) The second top PFAS chemical (blue) by docking score at the orthosteric site is displayed, DTXSID60881337, under the classification of Polycyclic Perfluoroalkane. (<b>2b</b>) DTXSID60881337 and the redocked calcitriol (purple) are displayed. (<b>3a</b>) The third top PFAS chemical (blue) by docking score at the orthosteric site is displayed, DTXSID101023399, under the classification of Cyclic Perfluoroalkane. (<b>3b</b>) DTXSID101023399 and the redocked calcitriol (purple) are displayed. (<b>4a</b>) The fourth top PFAS chemical (blue) by docking score at the orthosteric site is displayed, DTXSID701036930, under the classification of Perfluoroalkyl Sulfonic Acids. (<b>4b</b>) DTXSID701036930 and the redocked calcitriol (purple) are displayed. (<b>5a</b>) The fourth top PFAS chemical (blue) by docking score at the orthosteric site is displayed, Hexatetracontafluorodocosane, under the classification of Perfluoroalkane. (<b>5b</b>) Hexatetracontafluorodocosane and the redocked calcitriol (purple) are displayed. The hydrogen bonds are displayed.</p>
Full article ">Figure 6
<p>(<b>1a</b>) The top PFAS chemical (blue) by docking score at the allosteric site is displayed, DTXSID70597457, under the classification of Polycyclic Perfluoroalkane. (<b>1b</b>) DTXSID70597457 and lithocholic acid (purple) are displayed. (<b>2a</b>) The second top PFAS chemical (blue) by docking score at the allosteric site is displayed, DTXSID60881337, under the classification of Polycyclic Perfluoroalkane. (<b>2b</b>) DTXSID60881337 and lithocholic acid (purple) are displayed. (<b>3a</b>) The third top PFAS chemical (blue) by docking score at the allosteric site is displayed, Perfluoroperhydrobenzyl tetralin, under the classification of Polycyclic Perfluoroalkane. (<b>3b</b>) Perfluoroperhydrobenzyl tetralin and lithocholic acid (purple) are displayed. (<b>4a</b>) The fourth top PFAS chemical (blue) by docking score at the allosteric site is displayed, DTXSID90984683, under the classification of Polycyclic Perfluoroalkane. (<b>4b</b>) DTXSID90984683 and lithocholic acid (purple) are displayed. (<b>5a</b>) The fifth top PFAS chemical (blue) by docking score at the allosteric site is DTXSID501041016 under the classification of Polycyclic Perfluoroalkane. (<b>5b</b>) DTXSID501041016 and lithocholic acid (purple) are displayed. The hydrogen bonds are displayed.</p>
Full article ">Figure 7
<p>(<b>a</b>) The co-crystal structure of PPARy (PBD ID: 3ET3) and docked binding pose of the PFAS compound (DTXSID201033555) in the orthosteric binding site with the second-lowest docking score (−12.1). Amino acid residues within 5 Å of the co-crystal structure are shown in stick form. (<b>b</b>) The superimposed co-crystal structure of PPARy with ET1 in red (PDB ID: 3ET3) and the docked binding pose of the PFAS compound (DTXSID201033555) in blue bound to the orthosteric binding site with a docking score of −12.1. Amino acid residues within 5 Å of the co-crystal structure are shown in stick form.</p>
Full article ">Figure 8
<p>(<b>a</b>) The co-crystal structure of PPARy (PDB ID: 3ET3) and docked binding pose of the top PFAS compound (DTXSID20816403) in the orthosteric binding site with a docking score of −12.2. Amino acid residues within 5 Å of the co-crystal structure are shown in stick form. (<b>b</b>) The superimposed co-crystal structure of PPARy with ET1 in red (PDB ID: 3ET3) and the docked binding pose of the PFAS compound (DTXSID20816403) in blue bound to the orthosteric binding site with a docking score of −12.2. Amino acid residues within 5 Å of the co-crystal structure are shown in stick form.</p>
Full article ">Figure 9
<p>(<b>a</b>) The co-crystal structure of PPARy (PDB ID: 5GTO) and docked binding pose of the PFAS compound (DTXSID90984683) in the alternative binding site with the lowest docking score (−13.5). Amino acid residues within 5 Å of the co-crystal structure are shown in stick form. (<b>b</b>) The superimposed co-crystal structure of PPARy with T35 in red (PDB ID: 5GTO) and the docked binding pose of the PFAS compound (DTXSID90984683) in blue bound to the alternative binding site with a docking score of −13.5. Amino acid residues within 5 Å of the co-crystal structure are shown in stick form.</p>
Full article ">Figure 10
<p>The 11 highest-binding-affinity PFAS superimposed conformations at the orthosteric site of the PXR are displayed along with the reference ligand, 4WH. (<b>a</b>) DTXSID501041016 (salmon) with a docking score of −13.5. (<b>b</b>) DTXSID401026885 (yellow) with a docking score of −13.1. (<b>c</b>) DTXSID60881337 (salmon) with a docking score of −12.6. (<b>d</b>) DTXSID701026989 (salmon) with a docking score of −12.4. (<b>e</b>) DTXSID70896735 (yellow) with a docking score of −12.1. (<b>f</b>) DTXSID10896198 (green) with a docking score of −12.1. (<b>g</b>) DTXSID90984683 (salmon)with a docking score of −12. (<b>h</b>) DTXSID70597457 (green) with a docking score of −12. (<b>i</b>) DTXSID801026875 (green) with a docking score of −12. (<b>j</b>) DTXSID60896264 (green)with a docking score of −12. (<b>k</b>) DTXSID40881335 (beige) with a docking score of −12. Amino acid residues within a distance of 5 Å are labeled.</p>
Full article ">Figure 11
<p>The distributions of PFAS binding affinities measured in kcal/mol are displayed at the orthosteric site of (<b>a</b>) PXR, (<b>b</b>) PPARγ, (<b>c</b>) ERα, and (<b>d</b>) VDR.</p>
Full article ">Figure 12
<p>The distributions of PFAS binding affinities measured in kcal/mol are displayed at the allosteric site of (<b>a</b>) PXR, (<b>b</b>) PPARγ, (<b>c</b>) ERα, and (<b>d</b>) VDR.</p>
Full article ">Figure 13
<p>(<b>a</b>) The top PFAS by docking score at the allosteric site is displayed, Hexatriacontafluorotetracosahydrocoronene, under the classification of Polycyclic Perfluoroalkane. (<b>b</b>) The top PFAS by docking score at the orthosteric site is displayed, Perfluoroperhydrobenzyl tetralin, under the classification of Polycyclic Perfluoroalkane. (<b>c</b>) The top PFAS at both the orthosteric and allosteric sites are displayed, Perfluoroperhydrobenzyl tetralin and Hexatriacontafluorotetracosahydrocoronene, respectively, and both are under the classification of Polycyclic Perfluoroalkane. (<b>d</b>) Perfluorooctanoic acid is displayed at the orthosteric and allosteric site of the VDR, and the PFAS class is perfluorinated alkyl acids, and more specifically carboxylic acids. (<b>e</b>) Perfluorooctane sulfonic acid is displayed at the orthosteric and allosteric site of the VDR, and the PFAS class is perfluorinated alkyl acids, and more specifically sulfonic acids. (<b>f</b>) Perfluorooctanoic acid is displayed at the orthosteric and perfluorooctane sulfonic acid is displayed at the allosteric site of the VDR. (<b>g</b>) Perfluorooctane sulfonic acid is displayed at the orthosteric and perfluorooctanoic acid is displayed at the allosteric site of the VDR.</p>
Full article ">
22 pages, 3217 KiB  
Article
Safety Implications of Modulating Nuclear Receptors: A Comprehensive Analysis from Non-Clinical and Clinical Perspectives
by Mohan Rao, Eric McDuffie, Sanjay Srivastava, Warren Plaisted and Clifford Sachs
Pharmaceuticals 2024, 17(7), 875; https://doi.org/10.3390/ph17070875 - 3 Jul 2024
Viewed by 1716
Abstract
The unintended modulation of nuclear receptor (NR) activity by drugs can lead to toxicities amongst the endocrine, gastrointestinal, hepatic cardiovascular, and central nervous systems. While secondary pharmacology screening assays include NRs, safety risks due to unintended interactions of small molecule drugs with NRs [...] Read more.
The unintended modulation of nuclear receptor (NR) activity by drugs can lead to toxicities amongst the endocrine, gastrointestinal, hepatic cardiovascular, and central nervous systems. While secondary pharmacology screening assays include NRs, safety risks due to unintended interactions of small molecule drugs with NRs remain poorly understood. To identify potential nonclinical and clinical safety effects resulting from functional interactions with 44 of the 48 human-expressed NRs, we conducted a systematic narrative review of the scientific literature, tissue expression data, and used curated databases (OFF-X™) (Off-X, Clarivate) to organize reported toxicities linked to the functional modulation of NRs in a tabular and machine-readable format. The top five NRs associated with the highest number of safety alerts from peer-reviewed journals, regulatory agency communications, congresses/conferences, clinical trial registries, and company communications were the Glucocorticoid Receptor (GR, 18,328), Androgen Receptor (AR, 18,219), Estrogen Receptor (ER, 12,028), Retinoic acid receptors (RAR, 10,450), and Pregnane X receptor (PXR, 8044). Toxicities associated with NR modulation include hepatotoxicity, cardiotoxicity, endocrine disruption, carcinogenicity, metabolic disorders, and neurotoxicity. These toxicities often arise from the dysregulation of receptors like Peroxisome proliferator-activated receptors (PPARα, PPARγ), the ER, PXR, AR, and GR. This dysregulation leads to various health issues, including liver enlargement, hepatocellular carcinoma, heart-related problems, hormonal imbalances, tumor growth, metabolic syndromes, and brain function impairment. Gene expression analysis using heatmaps for human and rat tissues complemented the functional modulation of NRs associated with the reported toxicities. Interestingly, certain NRs showed ubiquitous expression in tissues not previously linked to toxicities, suggesting the potential utilization of organ-specific NR interactions for therapeutic purposes. Full article
(This article belongs to the Section Pharmacology)
Show Figures

Figure 1

Figure 1
<p>Pie chart showing the approximate number of reported targets in key target families. The number within each pie indicates the number of targets for the respective target class.</p>
Full article ">Figure 2
<p>Number of alerts associated with each of the 44 NRs. Green indicates agonists, red indicates antagonists, magenta indicates degraders, and blue indicates modulators (either agonist, antagonist, or partial agonist). The X-axis represents the name of the NR, and the Y-axis shows the number of alerts.</p>
Full article ">Figure 3
<p>Heatmap of human mRNA expression of NRs. The color gradient from red to blue indicates high to low expression levels. The X-axis represents the NR name, and the Y-axis shows the tissues where the expression is noted. The heatmap is colored using log<sub>2</sub>(FPKM + 0.1) values by tissues, ranging from −9.5 (red) to 9.5 (blue).</p>
Full article ">Figure 4
<p>Heatmap of Rat mRNA expression of NRs. The color gradient from red to blue indicates high to low expression levels. The X-axis represents the NR name, and the Y-axis shows the tissues where the expression is noted. The heatmap is colored using log<sub>2</sub>(FPKM + 0.1) values by tissues, ranging from −9.5 (red) to 9.5 (blue).</p>
Full article ">Figure 5
<p>Computed mean brain fold expression of each NR relative to the median expression across all tissues. A higher number indicates relatively higher mean expression in the brain. Red to blue represents high to low mean expression in the brain.</p>
Full article ">Figure 6
<p>Venn diagram of published safety screening panels by pharmaceutical companies and the FDA. The names of each panel are shown as A, B, C, D, and E. Corresponding companies or the FDA are labeled separately.</p>
Full article ">Figure 7
<p>Pie chart showing the number of annotated CNS alerts (blue), CV alerts (red), and Hepatic alerts (beige).</p>
Full article ">
16 pages, 2117 KiB  
Article
Characterization of NR1J1 Paralog Responses of Marine Mussels: Insights from Toxins and Natural Activators
by Antonio Casas-Rodríguez, Concepción Medrano-Padial, Angeles Jos, Ana M. Cameán, Alexandre Campos and Elza Fonseca
Int. J. Mol. Sci. 2024, 25(12), 6287; https://doi.org/10.3390/ijms25126287 - 7 Jun 2024
Viewed by 774
Abstract
The pregnane X receptor (PXR) is a nuclear hormone receptor that plays a pivotal role in regulating gene expression in response to various ligands, particularly xenobiotics. In this context, the aim of this study was to shed light on the ligand affinity and [...] Read more.
The pregnane X receptor (PXR) is a nuclear hormone receptor that plays a pivotal role in regulating gene expression in response to various ligands, particularly xenobiotics. In this context, the aim of this study was to shed light on the ligand affinity and functions of four NR1J1 paralogs identified in the marine mussel Mytilus galloprovincialis, employing a dual-luciferase reporter assay. To achieve this, the activation patterns of these paralogs in response to various toxins, including freshwater cyanotoxins (Anatoxin-a, Cylindrospermopsin, and Microcystin-LR, -RR, and -YR) and marine algal toxins (Nodularin, Saxitoxin, and Tetrodotoxin), alongside natural compounds (Saint John’s Wort, Ursolic Acid, and 8-Methoxypsoralene) and microalgal extracts (Tetraselmis, Isochrysis, LEGE 95046, and LEGE 91351 extracts), were studied. The investigation revealed nuanced differences in paralog response patterns, highlighting the remarkable sensitivity of MgaNR1J1γ and MgaNR1J1δ paralogs to several toxins. In conclusion, this study sheds light on the intricate mechanisms of xenobiotic metabolism and detoxification, particularly focusing on the role of marine mussel NR1J1 in responding to a diverse array of compounds. Furthermore, comparative analysis with human PXR revealed potential species-specific adaptations in detoxification mechanisms, suggesting evolutionary implications. These findings deepen our understanding of PXR-mediated metabolism mechanisms, offering insights into environmental monitoring and evolutionary biology research. Full article
(This article belongs to the Special Issue Recent Developments in Metabolism of Algal Toxins in Animals)
Show Figures

Figure 1

Figure 1
<p>Firefly luciferase transactivation activity mediated by marine mussel NR1J1 paralogs with the freshwater cyanotoxins (<b>A</b>) Anatoxin-A and (<b>B</b>) Cylindrospermopsin. Human PXR was used as a control assay and OA as a positive control. Values are expressed as mean ± SEM of three replicates. Distinct lowercase letters indicate values significantly different at <span class="html-italic">p</span> &lt; 0.01 according to one-way analysis of variance (ANOVA) per gene and Tukey’s multiple range test (<span class="html-italic">n</span> = 3).</p>
Full article ">Figure 2
<p>Firefly luciferase transactivation activity mediated by marine mussel NR1J1 paralogs with the freshwater cyanotoxins (<b>A</b>) Microcystin-LR, (<b>B</b>) Microcystin-RR, and (<b>C</b>) Microcystin-YR. Human PXR was used as a control assay and OA as a positive control. Values are expressed as mean ± SEM of three replicates. Distinct lowercase letters indicate values significantly different at <span class="html-italic">p</span> &lt; 0.01 according to one-way analysis of variance (ANOVA) per gene and Tukey’s multiple range test (<span class="html-italic">n</span> = 3).</p>
Full article ">Figure 3
<p>Firefly luciferase transactivation activity mediated by marine mussel NR1J1 paralogs with 3 marine algal toxins: (<b>A</b>) Nodularin, (<b>B</b>) Saxitoxin, and (<b>C</b>) Tetrodotoxin. Human PXR was used as a control assay and OA as a positive control. Values are expressed as mean ± SEM of three replicates. Distinct lowercase letters indicate values significantly different at <span class="html-italic">p</span> &lt; 0.01 according to one-way analysis of variance (ANOVA) per gene and Tukey’s multiple range test (<span class="html-italic">n</span> = 3).</p>
Full article ">Figure 4
<p>Firefly luciferase transactivation activity mediated by marine mussel NR1J1 paralogs with 3 natural non-toxic compounds: (<b>A</b>) Saint John’s Wort, (<b>B</b>) Ursolic Acid, and (<b>C</b>) 8-Methoxypsoralene. Human PXR was used as a control assay and OA as a positive control. Values are expressed as mean ± SEM of three replicates. Distinct lowercase letters indicate values significantly different at <span class="html-italic">p</span> &lt; 0.01 according to one-way analysis of variance (ANOVA) per gene and Tukey’s multiple range test (<span class="html-italic">n</span> = 3).</p>
Full article ">Figure 5
<p>Firefly luciferase transactivation activity mediated by marine mussel NR1J1 α paralog with 4 microalgae extracts: (<b>A</b>) <span class="html-italic">Tetraselmis</span>, (<b>B</b>) <span class="html-italic">Isochrysis</span>, (<b>C</b>) LEGE CC 95046, and (<b>D</b>) LEGE CC 91351. Human PXR was used as a control assay and OA as a positive control. Values are expressed as mean ± SEM of three replicates. Distinct lowercase letters indicate values significantly different at <span class="html-italic">p</span> &lt; 0.01 according to one-way analysis of variance (ANOVA) per gene and Tukey’s multiple range test (<span class="html-italic">n</span> = 3).</p>
Full article ">
13 pages, 2019 KiB  
Article
Synergistic Steatosis Induction in Mice: Exploring the Interactions and Underlying Mechanisms between PFOA and Tributyltin
by Yannick Dauwe, Lucile Mary, Fabiana Oliviero, Louise Dubois, Elodie Rousseau-Bacquie, Jelskey Gomez, Véronique Gayrard and Laïla Mselli-Lakhal
Cells 2024, 13(11), 940; https://doi.org/10.3390/cells13110940 - 30 May 2024
Viewed by 1078
Abstract
This study explores the impact of environmental pollutants on nuclear receptors (CAR, PXR, PPARα, PPARγ, FXR, and LXR) and their heterodimerization partner, the Retinoid X Receptor (RXR). Such interaction may contribute to the onset of non-alcoholic fatty liver disease (NAFLD), which is initially [...] Read more.
This study explores the impact of environmental pollutants on nuclear receptors (CAR, PXR, PPARα, PPARγ, FXR, and LXR) and their heterodimerization partner, the Retinoid X Receptor (RXR). Such interaction may contribute to the onset of non-alcoholic fatty liver disease (NAFLD), which is initially characterized by steatosis and potentially progresses to steatohepatitis and fibrosis. Epidemiological studies have linked NAFLD occurrence to the exposure to environmental contaminants like PFAS. This study aims to assess the simultaneous activation of nuclear receptors via perfluorooctanoic acid (PFOA) and RXR coactivation via Tributyltin (TBT), examining their combined effects on steatogenic mechanisms. Mice were exposed to PFOA (10 mg/kg/day), TBT (5 mg/kg/day) or a combination of them for three days. Mechanisms underlying hepatic steatosis were explored by measuring nuclear receptor target gene and lipid metabolism key gene expressions, by quantifying plasma lipids and hepatic damage markers. This study elucidated the involvement of the Liver X Receptor (LXR) in the combined effect on steatosis and highlighted the permissive nature of the LXR/RXR heterodimer. Antagonistic effects of TBT on the PFOA-induced activation of the Pregnane X Receptor (PXR) and Peroxisome Proliferator-Activated Receptor Gamma (PPARγ) were also observed. Overall, this study revealed complex interactions between PFOA and TBT, shedding light on their combined impact on liver health. Full article
(This article belongs to the Collection Functions of Nuclear Receptors)
Show Figures

Figure 1

Figure 1
<p>Histologic hepatic steatosis assessment. Histology was conducted on frozen liver sections stained with Harris Haematoxylin and red oil stain. (<b>A</b>) An accumulation of multiple small red droplets in the cytoplasm allows to assess steatosis. Zoom focus: 63×. (<b>B</b>) Optic quantification and morphological assessment of steatosis. Area of lipid droplets and droplet diameters are presented as mean ± standard error of the mean. The <span class="html-italic">p</span>-values indicate the level of significance, with * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 and **** <span class="html-italic">p</span> &lt; 0.0001 being calculated via the one-way ANOVA test followed by Tukey’s multiple comparisons test.</p>
Full article ">Figure 2
<p>Nuclear receptor activation in the liver. RT-qPCR analysis was performed on (<b>A</b>) CAR prototypical target genes (<span class="html-italic">Cyp2b10</span> and <span class="html-italic">Cyp2c29</span>); (<b>B</b>) PPARα target genes <span class="html-italic">Cyp4a10</span> and <span class="html-italic">Cyp4a14</span>; (<b>C</b>) PXR prototypical target gene <span class="html-italic">Cyp3a11</span>; (<b>D</b>) <span class="html-italic">Pparγ</span> expression; (<b>E</b>) LXR target gene <span class="html-italic">Cyp7a1</span>; (<b>F</b>) FXR target gene <span class="html-italic">Abcb11</span>. The results are presented as a graph, showing the expression levels in fold changes of the DMSO group. The data are presented as mean ± standard error of the means. The statistical analysis used was a one-way ANOVA test followed by Tukey’s multiple comparisons test. The <span class="html-italic">p</span>-values indicate the level of significance, with * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, and **** <span class="html-italic">p</span> &lt; 0.0001 indicating significant differences.</p>
Full article ">Figure 3
<p>RT-qPCR of genes involved in lipid metabolism and transport. RT-qPCR of genes involved in (<b>A</b>) lipid synthesis and lipid droplet formation, (<b>B</b>) in fatty acid β-oxidation, (<b>C</b>) in fatty acid and triglyceride transport, and (<b>D</b>) in cholesteryl ester uptake. The results are expressed as fold changes of the DMSO group. Data are presented as mean ± standard error of the mean. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001. <span class="html-italic">p</span>-values represent significant differences between each treatment group according to one-way ANOVA test followed by Tukey’s multiple comparisons test. # for <span class="html-italic">p</span> &lt; 0.05 represents a significant difference, with additivity calculated using a one-sample t-test.</p>
Full article ">
15 pages, 2087 KiB  
Review
Pregnane X Receptor Signaling Pathway and Vitamin K: Molecular Mechanisms and Clinical Relevance in Human Health
by Jeff L. Staudinger, Avina Mahroke, Gauri Patel, Cole Dattel and Sahana Reddy
Cells 2024, 13(8), 681; https://doi.org/10.3390/cells13080681 - 14 Apr 2024
Cited by 1 | Viewed by 2353
Abstract
This review explores the likely clinical impact of Pregnane X Receptor (PXR) activation by vitamin K on human health. PXR, initially recognized as a master regulator of xenobiotic metabolism in liver, emerges as a key regulator influencing intestinal homeostasis, inflammation, oxidative stress, and [...] Read more.
This review explores the likely clinical impact of Pregnane X Receptor (PXR) activation by vitamin K on human health. PXR, initially recognized as a master regulator of xenobiotic metabolism in liver, emerges as a key regulator influencing intestinal homeostasis, inflammation, oxidative stress, and autophagy. The activation of PXR by vitamin K highlights its role as a potent endogenous and local agonist with diverse clinical implications. Recent research suggests that the vitamin K-mediated activation of PXR highlights this vitamin’s potential in addressing pathophysiological conditions by promoting hepatic detoxification, fortifying gut barrier integrity, and controlling pro-inflammatory and apoptotic pathways. PXR activation by vitamin K provides an intricate association with cancer cell survival, particularly in colorectal and liver cancers, to provide new insights into potential novel therapeutic strategies. Understanding the clinical implications of PXR activation by vitamin K bridges molecular mechanisms with health outcomes, further offering personalized therapeutic approaches for complex diseases. Full article
Show Figures

Figure 1

Figure 1
<p>The scheme illustrates a likely key role of vitamin K2 (MK-4) and PXR activation in human health. MK-4-mediated activation of PXR plays a pivotal role in detoxification of blood, liver, bone, and gut. PXR activation accomplishes these biological functions through regulating the expression and activity of genes involved in autophagy, oxidative stress, and the inflammatory response.</p>
Full article ">Figure 2
<p>The scheme illustrates the pathway used to accomplish the gamma-glutamyl carboxylation of proteins using vitamin K as a cofactor. Vitamin K plays a crucial role in the synthesis of Gla proteins by serving as a cofactor for the GGCX enzyme, which catalyzes the conversion of glutamic acid residues to gamma-carboxyglutamic acid residues. The GGCX enzyme can utilize both vitamin K1 and vitamin K2 in the gamma-carboxylation process, and the choice of cofactor depends on the specific Gla protein being modified and its location within the body. One significant biological consequence of gamma-glutamyl carboxylation of clotting factors II, VII, IX, and X is the ability to bind the released Calcium (Ca<sup>2+</sup>) ions and form the tenase complex at the site of blood vessel injury.</p>
Full article ">Figure 3
<p>The scheme illustrates the specific Gla protein name and the categories of proteins to include clotting factors, bone proteins, regulatory proteins, other Gla proteins, as well as less well-characterized Gla proteins. Their respective biological function is also listed in this scheme.</p>
Full article ">Figure 4
<p>The scheme illustrates the structure, key forms, and sources of vitamin K. Vitamin K1 (phylloquinone) is primarily found in green leafy vegetables like kale, spinach, broccoli, and Brussels sprouts. On the other hand, vitamin K2 (menaquinones), which includes specific forms like menaquinone-4 (MK-4) and menaquinone-7 (MK-7), is present in animal products such as meat, eggs, and dairy, as well as fermented foods like natto—a traditional Japanese dish made from fermented soybeans known for its unique taste, slimy texture, and strong aroma. Gut bacteria play a role in converting K1 into K2 through menaquinone biosynthesis, a process that involves modifying the side chain of the vitamin K molecule. This conversion results in various forms of vitamin K2, including MK-4. MK-4 is not only produced by gut bacteria but can also be found in certain animal products, particularly in meats like chicken and pork, organ meats, and dairy products such as eggs.</p>
Full article ">Figure 5
<p>The scheme illustrates the likely pathway of local biosynthesis of MK-4 in extrahepatic tissues by UBIAD1. MK-4 stands out as the primary form of vitamin K in vertebrate animals.</p>
Full article ">
20 pages, 3918 KiB  
Article
Exploring Microbial Metabolite Receptors in Inflammatory Bowel Disease: An In Silico Analysis of Their Potential Role in Inflammation and Fibrosis
by Michail Spathakis, Nikolas Dovrolis, Eirini Filidou, Leonidas Kandilogiannakis, Gesthimani Tarapatzi, Vassilis Valatas, Ioannis Drygiannakis, Vasilis Paspaliaris, Konstantinos Arvanitidis, Vangelis G. Manolopoulos, George Kolios and Stergios Vradelis
Pharmaceuticals 2024, 17(4), 492; https://doi.org/10.3390/ph17040492 - 12 Apr 2024
Viewed by 1342
Abstract
Metabolites produced by dysbiotic intestinal microbiota can influence disease pathophysiology by participating in ligand–receptor interactions. Our aim was to investigate the differential expression of metabolite receptor (MR) genes between inflammatory bowel disease (IBD), healthy individuals (HIs), and disease controls in order to identify [...] Read more.
Metabolites produced by dysbiotic intestinal microbiota can influence disease pathophysiology by participating in ligand–receptor interactions. Our aim was to investigate the differential expression of metabolite receptor (MR) genes between inflammatory bowel disease (IBD), healthy individuals (HIs), and disease controls in order to identify possible interactions with inflammatory and fibrotic pathways in the intestine. RNA-sequencing datasets containing 643 Crohn’s disease (CD) patients, 467 ulcerative colitis (UC) patients and 295 HIs, and 4 Campylobacter jejuni-infected individuals were retrieved from the Sequence Read Archive, and differential expression was performed using the RaNA-seq online platform. The identified differentially expressed MR genes were used for correlation analysis with up- and downregulated genes in IBD, as well as functional enrichment analysis using a R based pipeline. Overall, 15 MR genes exhibited dysregulated expression in IBD. In inflamed CD, the hydroxycarboxylic acid receptors 2 and 3 (HCAR2, HCAR3) were upregulated and were associated with the recruitment of innate immune cells, while, in the non-inflamed CD ileum, the cannabinoid receptor 1 (CNR1) and the sphingosine-1-phospate receptor 4 (S1PR4) were downregulated and were involved in the regulation of B-cell activation. In inflamed UC, the upregulated receptors HCAR2 and HCAR3 were more closely associated with the process of TH-17 cell differentiation, while the pregnane X receptor (NR1I2) and the transient receptor potential vanilloid 1 (TRPV1) were downregulated and were involved in epithelial barrier maintenance. Our results elucidate the landscape of metabolite receptor expression in IBD, highlighting associations with disease-related functions that could guide the development of new targeted therapies. Full article
(This article belongs to the Special Issue Gut Microbiota Metabolites in Intestinal Inflammation and Fibrosis)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Heat map of metabolite receptor gene expression in (<b>A</b>) UC as compared to HI and (<b>B</b>) CD as compared to HI. Each row represents a single analysis, including either all samples found in the datasets or subgroups of data from a single location, with further subcategorization regarding inflammation status (inflamed and non-inflamed regions). Each column represents the expression of a single receptor gene in each analysis performed. Criteria for significant up- or downregulation were set as follows: fold regulation between −1.5 and 1.5 and adjusted <span class="html-italic">p</span> value &lt; 0.05 (−1.5 &lt; FR &gt; 1.5 + padj &lt; 0.05), and significantly up- or downregulated genes are noted with *.</p>
Full article ">Figure 2
<p>Heat map of metabolite receptor gene expression in (<b>A</b>) inflamed compared to non-inflamed intestinal regions and (<b>B</b>) <span class="html-italic">C. jejuni</span> infection compared to HI. Each row represents a single analysis, including subgroups of data from a single location. Each column represents the expression of a single receptor gene in each analysis performed. Criteria for significant up- or downregulation were set as follows: fold regulation between −1.5 and 1.5 and adjusted <span class="html-italic">p</span> value &lt; 0.05 (−1.5 &lt; FR &gt; 1.5 + padj &lt; 0.05), and significantly up- or downregulated genes are noted with *.</p>
Full article ">Figure 3
<p>Heat map of metabolite receptor gene expression in the ileum compared to the colon. Each row represents a single analysis, including either all samples found in the datasets or subgroups of data from subcategorization regarding inflammation status (inflamed and non-inflamed regions). Each column represents the expression of a single receptor gene in each analysis performed. Criteria for significant up- or downregulation were set as fold regulation between −1.5 and 1.5 and adjusted <span class="html-italic">p</span> value &lt; 0.05 (−1.5 &lt; FR &gt; 1.5 + padj &lt; 0.05), and significantly up- or downregulated genes are noted with *.</p>
Full article ">Figure 4
<p>Expression correlation networks of MRs with up- and downregulated genes in CD. Each tetrad represents data from a single analysis and consists of four networks. In each tetrad, MR connections with up- (first column) and downregulated (second column) genes in patients with CD are shown on the upper row, and HIs are shown on the lower row. Specifically, (<b>A</b>) Crohn’s Disease versus Healthy Individuals regardless of inflammation status or disease location (<b>B</b>) Crohn’s Disease versus Healthy Individuals in the inflamed terminal ileum (<b>C</b>) Crohn’s Disease versus Healthy Individuals in the inflamed colon (<b>D</b>) Crohn’s Disease versus Healthy Individuals in the non-inflamed terminal ileum. Red circular nodes represent upregulated genes, green circular nodes represent downregulated genes, and blue rhomboidal nodes represent MR genes. Only connections with strongly correlated genes are shown (r &gt; 0.8).</p>
Full article ">Figure 5
<p>Expression correlation networks of MRs with up- and downregulated genes in UC. Each tetrad represents data from a single analysis and consists of four networks. In each tetrad, MR connections with up- (first column) and downregulated (second column) genes in patients with UC are shown on the upper row, and HIs are shown on the lower row. Specifically, (<b>A</b>) Ulcerative Colitis versus Healthy Individuals regardless of inflammation status or disease location (<b>B</b>) Ulcerative Colitis versus Healthy Individuals in the inflamed colon (<b>C</b>) Ulcerative Colitis versus Healthy Individuals in the non-inflamed colon (<b>D</b>) Ulcerative Colitis versus Healthy Individuals in the non-inflamed terminal ileum. Red circular nodes represent upregulated genes, green circular nodes represent downregulated genes, and blue rhomboidal nodes represent MR genes. Only connections with strongly correlated genes are shown (r &gt; 0.8).</p>
Full article ">Figure 6
<p>Flowchart depicting the steps followed and the tools used for this study.</p>
Full article ">
28 pages, 3801 KiB  
Review
A Review of P-Glycoprotein Function and Regulation in Fish
by Christina U. Johnston and Christopher J. Kennedy
Fishes 2024, 9(2), 51; https://doi.org/10.3390/fishes9020051 - 27 Jan 2024
Viewed by 1922
Abstract
The teleost ATP Binding Cassette (ABC) transporter P-glycoprotein (P-gp) is an active transmembrane transporter that plays a pivotal role in facilitating the movement of both endogenous and xenobiotic substrates (moderately hydrophobic and amphipathic compounds) across cell membranes. P-gp exhibits substrate specificity often shared [...] Read more.
The teleost ATP Binding Cassette (ABC) transporter P-glycoprotein (P-gp) is an active transmembrane transporter that plays a pivotal role in facilitating the movement of both endogenous and xenobiotic substrates (moderately hydrophobic and amphipathic compounds) across cell membranes. P-gp exhibits substrate specificity often shared with other ABC transporters and solute carrier proteins, thereby ensuring the maintenance of chemical homeostasis within cells. These transporters are integral to chemical defense systems in fish, as they actively expel a wide range of substrates, primarily unmodified compounds, from cells. This transport process assists in preventing chemical absorption (e.g., intestine), safeguarding sensitive tissues (e.g., brain and gonads), and effectively excreting substances (e.g., liver and kidney). Upregulated P-gp export activity in aquatic animals results in the multi-xenobiotic resistance (MXR) phenotype that plays an essential protective role in survival in contaminated environments. Pollutants inhibiting P-gp are termed chemosensitizers and heighten fish sensitivity to toxic P-gp substrates. While the known intrinsic functions of P-gp in fish encompass steroid hormone and bile acid processing, relatively little attention has been given to endogenous substrates and inhibitors. Fish P-glycoprotein regulation is orchestrated by pivotal nuclear transcription factors, including pregnane X receptor (PXR) and nuclear factor erythroid 2-related factor 2 (Nrf2). This comprehensive review provides profound insights into P-gp’s significance across diverse fish species, contributing to an enhanced understanding of fish physiology, evolution, and toxicology, and provides information with potential applications, such as environmental monitoring. Full article
Show Figures

Figure 1

Figure 1
<p>A schematic representation of the relationships between transmembrane transporters and biotransformation enzymes in epithelial cells. The apical membrane faces the lumen of the vessel, which is in contact with blood in blood–tissue barriers (e.g., blood–brain, blood–eye, blood–gonad), and is in contact with excretory fluid (urine, bile, feces) in absorptive (e.g., intestine) and excretory (e.g., liver, kidney) tissues. Substrate entry on the basolateral membrane is represented by membrane diffusion, as well as importer proteins: the SLC21 family (organic anion transporter proteins [OATP]) and the SLC22 family (organic cation transporters [OCT], organic anion transporters [OAT]). Chemical biotransformation occurs inside the cell by phase I functionalization enzymes (represented by cytochrome monooxygenases [CYP]), and phase II conjugation enzymes (represented by glutathione-S-transferase [GST]). Glutathione conjugates are further processed through the mercapturic acid pathway, and exported as mercapturate conjugates. Substrate efflux on the apical membrane is performed in phase 0 (unmodified substrates) by the ABCB family (p-glycoprotein [P-gp]), and in phase III (biotransformed substrates) by the ABCC family (multidrug resistance proteins [MRP]) and the ABCG family (breast cancer resistance protein [BCRP]). The SLC47 family (multidrug and toxicant extrusion proteins [MATE]) exports substrates, but its specificity for the transport of unmodified or biotransformed substrates (and therefore its phase) is unknown in fish.</p>
Full article ">Figure 2
<p>Structure of fish P-glycoprotein. The protein is composed of two transmembrane domains (TMDs) and two nucleotide-binding domains (NBDs). Each transmembrane domain contains six transmembrane helices [<a href="#B41-fishes-09-00051" class="html-bibr">41</a>,<a href="#B69-fishes-09-00051" class="html-bibr">69</a>].</p>
Full article ">Figure 3
<p>The P-glycoprotein transport cycle. 1. P-gp has an inward-open conformation, with no substrate or ATP bound. 2. A substrate binds in the drug-binding pocket between the transmembrane domains (TMDs), and one ATP molecule binds to each of the two nucleotide binding domains (NBDs). The NBDs dimerize with the two ATP molecules sandwiched between them. 3. One of the ATP molecules is hydrolyzed. The P-gp molecule transitions to an outward-facing conformation and releases the substrate into the extracellular space. 4. The second ATP molecule is hydrolyzed, allowing P-gp to return to an inward-facing conformation [<a href="#B112-fishes-09-00051" class="html-bibr">112</a>].</p>
Full article ">
18 pages, 1725 KiB  
Review
AhR, PXR and CAR: From Xenobiotic Receptors to Metabolic Sensors
by Leonida Rakateli, Rosanna Huchzermeier and Emiel P. C. van der Vorst
Cells 2023, 12(23), 2752; https://doi.org/10.3390/cells12232752 - 30 Nov 2023
Cited by 7 | Viewed by 2576
Abstract
Traditionally, xenobiotic receptors are known for their role in chemical sensing and detoxification, as receptor activation regulates the expression of various key enzymes and receptors. However, recent studies have highlighted that xenobiotic receptors also play a key role in the regulation of lipid [...] Read more.
Traditionally, xenobiotic receptors are known for their role in chemical sensing and detoxification, as receptor activation regulates the expression of various key enzymes and receptors. However, recent studies have highlighted that xenobiotic receptors also play a key role in the regulation of lipid metabolism and therefore function also as metabolic sensors. Since dyslipidemia is a major risk factor for various cardiometabolic diseases, like atherosclerosis and non-alcoholic fatty liver disease, it is of major importance to understand the molecular mechanisms that are regulated by xenobiotic receptors. In this review, three major xenobiotic receptors will be discussed, being the aryl hydrocarbon receptor (AhR), pregnane X receptor (PXR) and the constitutive androstane receptor (CAR). Specifically, this review will focus on recent insights into the metabolic functions of these receptors, especially in the field of lipid metabolism and the associated dyslipidemia. Full article
Show Figures

Figure 1

Figure 1
<p><b>Main pathways of lipid metabolism.</b> The exogenous pathway begins with the transport of ingested dietary lipids from the intestine to, for example, muscles and adipose tissue via chylomicrons, and the resulting chylomicron remnants transport the remaining lipids to the liver. The endogenous pathway starts in the liver with the synthesis of VLDL by conveying triglycerides and cholesterol esters in the endoplasmic reticulum to newly synthesized apoB-100 to form the core of VLDL. As VLDL circulates through the bloodstream, triglycerides are removed from VLDL, making it denser and more cholesterol-rich, eventually transforming into LDL. LDL primarily carries cholesterol to peripheral tissues, such as muscle, heart or adipose tissue, while HDL is responsible for reverse cholesterol transport, where excess cholesterol is transported from peripheral tissues back to the liver. ApoA-I: apolipoprotein A-I; ApoB-100: apolipoprotein B-100; CD36: cluster of differentiation 36; CE: cholesterol esters; CETP: cholesteryl ester transfer protein; HDL: high-density lipoprotein; IDL: intermediate-density lipoprotein; LCAT: lecithin cholesterol acyl transferase; LDL: low-density lipoprotein; LDLR: low-density lipoprotein receptor; LPL: lipoprotein lipase; SR-BI: scavenger receptor B-I; TG: triglyceride; VLDL: very low-density lipoprotein. Figure created with <a href="http://BioRender.com" target="_blank">BioRender.com</a>.</p>
Full article ">Figure 2
<p><b>Mechanism of xenobiotic receptor activation.</b> (<b>Left panel</b>): In the cytoplasm, AhR is bound by Hsp90, AIP, the co-chaperone p23 and protein kinase SRC. After ligand binding, with either exogenous ligands or endogenous ligands such as tryptophane metabolites, the AhR complex releases AIP and translocates to the nucleus, where the rest of the cofactors dissociate from AhR, enabling ARNT to heterodimerize with AhR. The AhR–ARNT complex then binds to the DNA and regulates the gene expression of several genes, such as CYP1A1, CYP1B1 or AHRR. AHRR can negatively regulate AhR by inhibiting the AhR–ARNT complex. (<b>Middle panel</b>): Mechanism of PXR activation. In the cytoplasm, PXR forms a complex with Hsp90 and CCRP, while non-activated PXR in the nucleus is bound to NcoR1 and SMRT. Upon receptor activation, PXR dissociates from its complex and translocates into the nucleus, where it binds to SRC-1 and GRIP1. Together with these coactivators, PXR heterodimerizes with RXR and binds to a PXR response element to induce target gene expression. (<b>Right panel</b>): CAR signaling. CAR is located in the cytoplasm, where it is bound to CCR and HSP90 in its inactive state. In the direct activation pathway, ligand binding activates PP2A, which dephosphorylates CAR, resulting in the dissociation of its co-chaperones. The activated CAR translocates into the nucleus, where it heterodimerizes with RXR and binds to the PBREM to induce target gene expression. In the indirect pathway (highlighted with green and red arrows), specific ligands of the Car can inhibit the binding of EGF to the EGFR. This results in the dephosphorylation of p-ERK and simultaneously the dephosphorylation of RACK. RACK activates PP2A and the dephosphorylated ERK dissociates from the CAR complex, enabling its activation by PP2A. AhR: aryl hydrocarbon receptor; AHRR: aryl hydrocarbon receptor repressor; AIP: AhR interacting protein; ARNT: aryl hydrocarbon receptor nuclear translocator; CAR: constitutive androstane receptor; CCRP: CAR cytoplasmic retention protein; CYP1A1: cytochrome P450 family 1 subfamily A member 1; CYP1B1: cytochrome P450 family 1 subfamily B member 1; EGF: epidermal growth factor; EGFR: EGF receptor; ERK: extracellular signal-regulated kinase, p-ERK: phosphorylated extracellular signal-regulated kinase; GRIP1: glucocorticoid receptor interacting protein 1; Hsp90: heat shock protein 90; NcoR1: nuclear receptor corepressor 1; P: phosphate; PBREM: phenobarbital responsive enhancer module; PP2A: protein phosphatase 2A; PXR: pregnane X receptor; PXRE: PXR-responsive element; RACK: receptor for activated C kinase 1; p-RACK: phosphorylated receptor for activated C kinase 1; RXR: retinoid X receptor; SMRT: silencing mediator of retinoid and thyroid receptors; SRC-1: steroid receptor coactivator 1; XRE: xenobiotic response element. Figure created with <a href="http://BioRender.com" target="_blank">BioRender.com</a>.</p>
Full article ">Figure 3
<p><b>Effects of xenobiotic receptor activation on lipid metabolism.</b> Activation of AhR (<b>left</b>) by ligands such as halogenated aromatic hydrocarbons leads to changes in gene transcription, which affects the lipid metabolism in various ways. PXR (<b>middle</b>) and CAR (<b>right</b>) share many ligands; however, their signaling pathways occur based on different cofactors. Nevertheless, both pathways result in similarly altered gene transcription of, for example, phase I and II enzymes. The activation of both receptors impacts lipid metabolism, targeting partly similar but also specific parts of lipid metabolism. Disruptions in lipid signaling can contribute to the development of CMDs, assigning the xenobiotic receptors a more metabolic sensing role. AhR: aryl hydrocarbon receptor; AHRR: aryl hydrocarbon receptor repressor; ARNT: aryl hydrocarbon receptor nuclear translocator; CAR: constitutive androstane receptor; CYP1A1: cytochrome P450 family 1 subfamily A member 1; CYP1B1: cytochrome P450 family 1 subfamily B member 1; GRIP1: glucocorticoid receptor interacting protein 1; NAFLD: non-alcoholic fatty liver disease; PBREM: phenobarbital-responsive enhancer module; PXR: pregnane X receptor; PXRE: PXR-responsive element; RXR: retinoid X receptor; SRC1: steroid receptor coactivator 1; TCDD: 2,3,7,8-tetrachlorodibenzo-p-dioxin; XRE: xenobiotic response element. Figure created with <a href="http://BioRender.com" target="_blank">BioRender.com</a>.</p>
Full article ">
13 pages, 3221 KiB  
Article
The Role of Adopted Orphan Nuclear Receptors in the Regulation of an Organic Anion Transporting Polypeptide 1B1 (OATP1B1) under the Action of Sex Hormones
by Aleksey V. Shchulkin, Yulia V. Abalenikhina, Aleksandr A. Slepnev, Egor D. Rokunov and Elena N. Yakusheva
Curr. Issues Mol. Biol. 2023, 45(12), 9593-9605; https://doi.org/10.3390/cimb45120600 - 29 Nov 2023
Cited by 1 | Viewed by 1096
Abstract
Organic anion transporting polypeptide 1B1 (OATP1B1) is an influx transporter protein of the SLC superfamily, expressed mainly in the liver and some tumor cells. The mechanisms of its regulation are being actively studied. In the present study, the effect of sex hormones (estradiol, [...] Read more.
Organic anion transporting polypeptide 1B1 (OATP1B1) is an influx transporter protein of the SLC superfamily, expressed mainly in the liver and some tumor cells. The mechanisms of its regulation are being actively studied. In the present study, the effect of sex hormones (estradiol, progesterone and testosterone) on OATP1B1 expression in HepG2 cells was examined. The role of adopted orphan receptors, farnasoid X receptor (FXR), constitutive androstane receptor (CAR), pregnane X receptor (PXR) and liver X receptor subtype alpha (LXRa), was also evaluated. Hormones were used in concentrations of 1, 10 and 100 μM, with incubation for 24 h. The protein expression of OATP1B1, FXR, CAR, PXR and LXRa was analyzed by Western blot. It was shown that estradiol (10 and 100 μM) increased the expression of OATP1B1, acting through CAR. Testosterone (1, 10 and 100 μM) increased the expression of OATP1B1, acting through FXR, PXR and LXRa. Progesterone (10 and 100 μM) decreased the expression of OATP1B1 (10 and 100 μM) and adopted orphan receptors are not involved in this process. The obtained results have important practical significance and determine ways for targeted regulation of the transporter, in particular in cancer. Full article
(This article belongs to the Section Biochemistry, Molecular and Cellular Biology)
Show Figures

Figure 1

Figure 1
<p>Effect of estradiol (1, 10, 100 µM, incubation for 24 h) on OATP1B1 expression in HepG2 cells. ** <span class="html-italic">p</span> &lt; 0.01—statistically significant differences with the control, ANOVA, post hoc Dunnet’s test. Es—estradiol, Cntr—control.</p>
Full article ">Figure 2
<p>Effect of testosterone (1, 10, 100 µM, incubation for 24 h) on OATP1B1 expression in HepG2 cells. **** <span class="html-italic">p</span> &lt; 0.001—statistically significant differences with the control, ANOVA, post hoc Dunnet’s test. Ts—testosterone, Cntr—control.</p>
Full article ">Figure 3
<p>Effect of progesterone (1, 10, 100 µM, incubation for 24 h) on OATP1B1 expression in HepG2 cells. ** <span class="html-italic">p</span> &lt; 0.01—statistically significant differences with the control, ANOVA, post hoc Dunnet’s test. Pg—progesterone, Cntr—control.</p>
Full article ">Figure 4
<p>Effect of sex hormones (1, 10, 100 µM, incubation for 24 h) on the FXR, CAR, PXR and LXR<span class="html-italic">a</span> expression. (<b>a</b>) Results of Western blotting; (<b>b</b>) densitometric analysis of Western blotting of FRX; (<b>c</b>) densitometric analysis of Western blotting of CAR; (<b>d</b>) densitometric analysis of Western blotting of PXR; (<b>e</b>) densitometric analysis of Western blotting of LXR<span class="html-italic">a</span>. * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001; **** <span class="html-italic">p</span> &lt; 0.0001—statistically significant differences with the control, ANOVA, post hoc Dunnet’s test. Es—estradiol, Ts—testosterone, Pg—progesterone, Cntr—control.</p>
Full article ">Figure 4 Cont.
<p>Effect of sex hormones (1, 10, 100 µM, incubation for 24 h) on the FXR, CAR, PXR and LXR<span class="html-italic">a</span> expression. (<b>a</b>) Results of Western blotting; (<b>b</b>) densitometric analysis of Western blotting of FRX; (<b>c</b>) densitometric analysis of Western blotting of CAR; (<b>d</b>) densitometric analysis of Western blotting of PXR; (<b>e</b>) densitometric analysis of Western blotting of LXR<span class="html-italic">a</span>. * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001; **** <span class="html-italic">p</span> &lt; 0.0001—statistically significant differences with the control, ANOVA, post hoc Dunnet’s test. Es—estradiol, Ts—testosterone, Pg—progesterone, Cntr—control.</p>
Full article ">Figure 5
<p>The role of the adopted orphan nuclear receptors in the induction of OATP1B1 under the action of estradiol at concentrations of 10 and 100 µM. (<b>a</b>) Results of Western blotting of OATP1B1 under the action of 10 µM estradiol + inhibitors of orphan receptors; (<b>b</b>) densitometric analysis of Western blotting of OATP1B1 under the action of 10 µM estradiol + inhibitors of orphan receptors; (<b>c</b>) results of Western blotting of OATP1B1 under the action of 100 µM estradiol + inhibitors of orphan receptors; (<b>d</b>) densitometric analysis of Western blotting of OATP1B1 under the action of 100 µM estradiol + inhibitors of orphan receptors. ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.01—statistically significant differences with the control, ANOVA, post hoc Dunnet’s test. Es—estradiol, Cntr—control, β-TA—FXR inhibitor, CINPA1—CAR inhibitor.</p>
Full article ">Figure 6
<p>The role of orphan nuclear receptors in the induction of OATP1B1 under the action of 10 and 100 µM testosterone. (<b>a</b>) Results of Western blotting of OATP1B1 under the action of testosterone 10 µM + inhibitors of orphan receptors; (<b>b</b>) densitometric analysis of Western blotting of OATP1B1 under the action of 10 µM testosterone + inhibitors of orphan receptors; (<b>c</b>) Results of Western blotting of OATP1B1 under the action of 100 µM testosterone + inhibitors of orphan receptors; (<b>d</b>) densitometric analysis of Western blotting of OATP1B1 under the action of 100 µM testosterone + inhibitors of orphan receptors. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001—statistically significant differences with the control, ANOVA, post hoc Dunnet’s test. Ts—testosterone, Cntr—control, β-TA—FXR inhibitor, CINPA1—CAR inhibitor, KTZ (ketoconazole)—PXR inhibitor, TFCA—LXR<span class="html-italic">a</span> inhibitor.</p>
Full article ">Figure 7
<p>Effect of testosterone (10, 100 µM, incubation for 24 h) on OATP1B1 expression with simultaneous inhibition of FXR, PXR and LXR<span class="html-italic">a.</span> Ts—testosterone, Cntr—control, β-TA—FXR inhibitor, KTZ (ketoconazole)—PXR inhibitor, TFCA—LXR<span class="html-italic">a</span> inhibitor.</p>
Full article ">Figure 8
<p>The role of orphan nuclear receptors in the inhibition of OATP1B1 under the action of 10 and 100 µM progesterone. (<b>a</b>) Results of Western blotting of OATP1B1 under the action of 10 µM progesterone + inhibitors of orphan receptors; (<b>b</b>) densitometric analysis of Western blotting of OATP1B1 under the action of 10 µM progesterone + inhibitors of orphan receptors; (<b>c</b>) results of Western blotting of OATP1B1 under the action of 100 µM progesterone + inhibitors of orphan receptors; (<b>d</b>) densitometric analysis of Western blotting of OATP1B1 under the action of 100 µM progesterone + inhibitors of orphan receptors. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, ANOVA, post hoc Dunnet’s test. Pg—progesterone, Cntr—control, β-TA—FXR inhibitor, KTZ (ketoconazole)—PXR inhibitor, TFCA—LXR<span class="html-italic">a</span> inhibitor.</p>
Full article ">
14 pages, 1352 KiB  
Case Report
Importance of Pharmacogenetics and Drug–Drug Interactions in a Kidney Transplanted Patient
by Julia Concha, Estela Sangüesa, Ana M. Saez-Benito, Ignacio Aznar, Nuria Berenguer, Loreto Saez-Benito, M. Pilar Ribate and Cristina B. García
Life 2023, 13(8), 1627; https://doi.org/10.3390/life13081627 - 26 Jul 2023
Cited by 1 | Viewed by 1464
Abstract
Tacrolimus (TAC) is a narrow-therapeutic-range immunosuppressant drug used after organ transplantation. A therapeutic failure is possible if drug levels are not within the therapeutic range after the first year of treatment. Pharmacogenetic variants and drug–drug interactions (DDIs) are involved. We describe a patient [...] Read more.
Tacrolimus (TAC) is a narrow-therapeutic-range immunosuppressant drug used after organ transplantation. A therapeutic failure is possible if drug levels are not within the therapeutic range after the first year of treatment. Pharmacogenetic variants and drug–drug interactions (DDIs) are involved. We describe a patient case of a young man (16 years old) with a renal transplant receiving therapy including TAC, mycophenolic acid (MFA), prednisone and omeprazole for prophylaxis of gastric and duodenal ulceration. The patient showed great fluctuation in TAC blood concentration/oral dose ratio, as well as pharmacotherapy adverse effects (AEs) and frequent diarrhea episodes. Additionally, decreased kidney function was found. A pharmacotherapeutic follow-up, including pharmacogenetic analysis, was carried out. The selection of the genes studied was based on the previous literature (CYP3A5, CYP3A4, POR, ABCB1, PXR and CYP2C19). A drug interaction with omeprazole was reported and the nephrologist switched to rabeprazole. A lower TAC concentration/dose ratio was achieved, and the patient’s condition improved. In addition, the TTT haplotype of ATP Binding Cassette Subfamily B member 1 (ABCB1) and Pregnane X Receptor (PXR) gene variants seemed to affect TAC pharmacotherapy in the studied patient and could explain the occurrence of long-term adverse effects post-transplantation. These findings suggest that polymorphic variants and co-treatments must be considered in order to achieve the effectiveness of the immunosuppressive therapy with TAC, especially when polymedicated patients are involved. Moreover, pharmacogenetics could influence the drug concentration at the cellular level, both in lymphocyte and in renal tissue, and should be explored in future studies. Full article
(This article belongs to the Section Genetics and Genomics)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Timeline of tacrolimus (TAC) trough concentration (ng/mL), serum creatinine levels (mg/dL) and TAC oral dose (mg/day).</p>
Full article ">Figure 2
<p>Comparation of TAC concentration/dose ratio with omeprazole and rabeprazole. Omep = omeprazole; Rabep = rabeprazole.</p>
Full article ">Figure 3
<p>Chromosome 7, <span class="html-italic">ABCB1</span> haplotype location, and p-gp structure. (<b>a</b>) Chromosome 7 structure and <span class="html-italic">ABCB1</span> gene location; (<b>b</b>) c.1236C &gt; T, c.2677G/A &gt; T and c.3435C &gt; T single-nucleotide polymorphisms (SNPs) location in unfolded p-gp; (<b>c</b>) folded p-gp structure and two nucleotide-binding domains (NBDs). Source: Adapted figure from (<b>a</b>) National Library of Medicine (NLM), (<b>b</b>) Adapted with permission from Ref. [<a href="#B46-life-13-01627" class="html-bibr">46</a>] Copyright 2023, Dove Press. (<b>c</b>) own source.</p>
Full article ">Figure 4
<p>Influence of genotype on drug pharmacokinetics: (<b>A</b>) Wildtype genotype producing regular tacrolimus (TAC) active ejection; (<b>B</b>) TTT haplotype and Pregnane X Receptor (PXR) 69789A &gt; G plus the presence of omeprazole inhibit TAC ejection. Omeprazole (OMEP); adenosine triphosphate (ATP); adenosine diphosphate (ADP). Source: own elaboration.</p>
Full article ">
20 pages, 1033 KiB  
Review
Molecular Factors and Pathways of Hepatotoxicity Associated with HIV/SARS-CoV-2 Protease Inhibitors
by Cheng Ji
Int. J. Mol. Sci. 2023, 24(9), 7938; https://doi.org/10.3390/ijms24097938 - 27 Apr 2023
Cited by 1 | Viewed by 2853
Abstract
Antiviral protease inhibitors are peptidomimetic molecules that block the active catalytic center of viral proteases and, thereby, prevent the cleavage of viral polyprotein precursors into maturation. They continue to be a key class of antiviral drugs that can be used either as boosters [...] Read more.
Antiviral protease inhibitors are peptidomimetic molecules that block the active catalytic center of viral proteases and, thereby, prevent the cleavage of viral polyprotein precursors into maturation. They continue to be a key class of antiviral drugs that can be used either as boosters for other classes of antivirals or as major components of current regimens in therapies for the treatment of infections with human immunodeficiency virus (HIV) and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). However, sustained/lifelong treatment with the drugs or drugs combined with other substance(s) often leads to severe hepatic side effects such as lipid abnormalities, insulin resistance, and hepatotoxicity. The underlying pathogenic mechanisms are not fully known and are under continuous investigation. This review focuses on the general as well as specific molecular mechanisms of the protease inhibitor-induced hepatotoxicity involving transporter proteins, apolipoprotein B, cytochrome P450 isozymes, insulin-receptor substrate 1, Akt/PKB signaling, lipogenic factors, UDP-glucuronosyltransferase, pregnane X receptor, hepatocyte nuclear factor 4α, reactive oxygen species, inflammatory cytokines, off-target proteases, and small GTPase Rab proteins related to ER-Golgi trafficking, organelle stress, and liver injury. Potential pharmaceutical/therapeutic solutions to antiviral drug-induced hepatic side effects are also discussed. Full article
(This article belongs to the Special Issue Cell and Molecular Biology of Hepatocytes)
Show Figures

Figure 1

Figure 1
<p>Adverse effects of antiviral protease inhibitors (PIs) on the liver. ApoB, apolipoprotein B; ACC, acetyl-CoA carboxylase; ATF 4 and 6, activating transcription factor 4 and 6; Bcl2, B-cell lymphoma-2; Casp3, caspase 3; C/EBP α and β, CCAAT/enhancer-binding protein α and β; CHOP, C/EBP homology protein 10; CREBP3, CAMP responsive element binding protein 3; CYPs, cytochrome P450 enzymes; eNOS, endothelial nitric oxide synthase 3; FAS, fatty acid synthase; FXR, farnesoid X receptor; GLUT2&amp;4, glucose transporter 2 and 4; GST, glutathione-S-transferase; HNF4α, hepatocyte nuclear factor 4α; HO1, heme oxygenase-1; IL-1β, 6, and 10, interleukin 1β, 6, and 10; INSTIs, integrase strand transfer inhibitors; IRS1&amp;2, insulin receptor substrate 1 and 2; IRE1, inositol requiring enzyme 1; JNK, c-JUN N-terminal kinase; MRP2/ABCC2, canalicular efflux transporter multidrug resistance-associated protein 2; NFκB, nuclear factor κ B; Nrf2, nuclear erythroid-derived factor 2; OATP1, organic anion transporting polypeptide 1; OCT1, human organic cation transporter 1; PERK, PKR-like ER- localized eIF2α kinase; P-gp, P-glycoprotein; PKA, protein kinase B; PPARγ, peroxisome proliferator- activating receptor γ; PXR, pregnane X receptor; RDV, remdesivir; ROS, reactive oxygen species; SCD1, stearoyl-CoA desaturase 1; SERCA, sarco/endoplamic reticulum Ca2+-ATPase; SREBP, sterol regulatory element binding protein; TFE3, transcription factor for immunoglobulin heavy-chain enhancer 3; TNFα, tumor necrosis factor α; UCP2, mitochondrial uncoupling protein 2; UDPGT, uridine-diphosphoglucuronic glucuronosyltransferase; XBP1, X-box binding protein 1.</p>
Full article ">Figure 2
<p>Mechanisms of antiviral protease inhibitor-induced organelle stress and liver injury. ARF1/4, ADP-ribosylation factor 1/4; COP I/II, coat protein complex I/II; GAPs, GTPase-activating proteins; GCP60, Golgi resident protein 60; GM130, Golgi matrix protein; GSR, Golgi stress response; HSP47, heat shock protein 47; Rab, small GTPase binding protein; RCE1, the Ras converting CaaX endopeptidase 1; SAR1, secretion-associated RAS-related; SNAREs, tail-anchored membrane fusion proteins; STE24, ER-associated CaaX protease; UPR, unfolded protein response.</p>
Full article ">
15 pages, 2790 KiB  
Article
Expanding the Library of 1,2,4-Oxadiazole Derivatives: Discovery of New Farnesoid X Receptor (FXR) Antagonists/Pregnane X Receptor (PXR) Agonists
by Claudia Finamore, Carmen Festa, Bianca Fiorillo, Francesco Saverio Di Leva, Rosalinda Roselli, Silvia Marchianò, Michele Biagioli, Lucio Spinelli, Stefano Fiorucci, Vittorio Limongelli, Angela Zampella and Simona De Marino
Molecules 2023, 28(6), 2840; https://doi.org/10.3390/molecules28062840 - 21 Mar 2023
Cited by 2 | Viewed by 1859
Abstract
Compounds featuring a 1,2,4-oxadiazole core have been recently identified as a new chemotype of farnesoid X receptor (FXR) antagonists. With the aim to expand this class of compounds and to understand the building blocks necessary to maintain the antagonistic activity, we describe herein [...] Read more.
Compounds featuring a 1,2,4-oxadiazole core have been recently identified as a new chemotype of farnesoid X receptor (FXR) antagonists. With the aim to expand this class of compounds and to understand the building blocks necessary to maintain the antagonistic activity, we describe herein the synthesis, the pharmacological evaluation, and the in vitro pharmacokinetic properties of a novel series of 1,2,4-oxadiazole derivatives decorated on the nitrogen of the piperidine ring with different N-alkyl and N-aryl side chains. In vitro pharmacological evaluation showed compounds 5 and 11 as the first examples of nonsteroidal dual FXR/Pregnane X receptor (PXR) modulators. In HepG2 cells, these compounds modulated PXR- and FXR-regulated genes, resulting in interesting leads in the treatment of inflammatory disorders. Moreover, molecular docking studies supported the experimental results, disclosing the ligand binding mode and allowing rationalization of the activities of compounds 5 and 11. Full article
(This article belongs to the Section Medicinal Chemistry)
Show Figures

Figure 1

Figure 1
<p>Library of 1,2,4-oxadiazole derivatives generated in this study.</p>
Full article ">Figure 2
<p>Panels (<b>A</b>–<b>C</b>) Effects of compounds <b>5</b> and <b>11</b> on the relative mRNA expression of FXR genes target in HepG2 cells left untreated, treated with 6-ECDCA (10 μM), compounds <b>5</b> or <b>11</b> (10 μM), or a combination of 6-ECDCA plus <b>5</b> or <b>11</b>. Results are expressed as mean ± SEM. (<b>D</b>–<b>F</b>) Effects of compounds <b>5</b> and <b>11</b> on the relative expression of PXR and inflammatory cytokines in Human-CaCo2 cells left untreated, treated with TNFα (100 ng/mL) alone, or in combination with <b>5</b> or <b>11</b> (10 μM). Results are expressed as mean ± SEM. * <span class="html-italic">p</span> &lt; 0.05 versus not treated cells (NT).</p>
Full article ">Figure 3
<p>Binding modes of (<b>A</b>) compound <b>5</b> (in yellow sticks) in the X-ray structure of PXR-LBD (PDB ID 7AXE) and (<b>B</b>) compound <b>11</b> (in light green sticks) were observed in the X-ray structure of PXR-LBD (PDB ID 3HVL) [<a href="#B34-molecules-28-02840" class="html-bibr">34</a>]. PXR is shown as tan cartoon, while the interacting residues of the receptors are shown in tan sticks. Hydrogens are omitted for clarity. Hydrogen bonds are shown as dashed black lines.</p>
Full article ">Figure 4
<p>Binding modes of (<b>A</b>) compound <b>5</b> (in yellow sticks) and (<b>B</b>) compound <b>11</b> (in light green sticks) in the X-ray structure of FXR-LBD (PDB ID 4OIV) [<a href="#B35-molecules-28-02840" class="html-bibr">35</a>]. FXR is shown as a gray cartoon, while the interacting residues of the receptors are shown as gray sticks. Hydrogens are omitted for clarity. Hydrogen bonds are shown as dashed black lines. The bridging water molecules are reported as red sticks with explicit hydrogens.</p>
Full article ">Scheme 1
<p>Reagents and conditions: (<b>a</b>) NH<sub>2</sub>OH HCl, K<sub>2</sub>CO<sub>3</sub> in CH<sub>3</sub>OH, reflux; (<b>b</b>) N-Boc-Inp-OH, DIPEA, HBTU in DMF dry, 80 °C, 80% yield; (<b>c</b>) TFA:CH<sub>2</sub>Cl<sub>2</sub> 1:1, 2 h, quantitative yield; (<b>d</b>) bromoethane, 1-bromopropane, 1-bromobutane, 1-bromopentane, 1-bromohexane, 1-bromoheptane, 2-bromopropane, 2-bromobutane, methyl 3-(bromomethyl)benzoate or methyl 4-(bromomethyl)benzoate, DIPEA, in CH<sub>3</sub>CN dry, 60 °C, overnight, the yield of each compound is: 77% for <b>2</b>, 87% for 3, 72% for <b>4</b> and <b>5</b>, 63% for <b>6</b>, 82% for <b>7</b>, 50% for <b>8</b>, 72% for <b>9</b> and 80% for <b>12</b> and <b>13</b> (<b>e</b>) DIBAL-H (1 M in toluene) in THF dry, 0 °C, 48 h, and 75 and 92% yields for compounds <b>10</b> and <b>11</b>.</p>
Full article ">
Back to TopTop