[go: up one dir, main page]
More Web Proxy on the site http://driver.im/
Next Issue
Volume 4, March
Previous Issue
Volume 3, September
You seem to have javascript disabled. Please note that many of the page functionalities won't work as expected without javascript enabled.
 
 

Receptors, Volume 3, Issue 4 (December 2024) – 9 articles

  • Issues are regarded as officially published after their release is announced to the table of contents alert mailing list.
  • You may sign up for e-mail alerts to receive table of contents of newly released issues.
  • PDF is the official format for papers published in both, html and pdf forms. To view the papers in pdf format, click on the "PDF Full-text" link, and use the free Adobe Reader to open them.
Order results
Result details
Select all
Export citation of selected articles as:
19 pages, 8887 KiB  
Article
LPA3: Pharmacodynamic Differences Between Lysophosphatidic Acid and Oleoyl-Methoxy Glycerophosphothionate: Biased Agonism, Two Sites
by K. Helivier Solís, M. Teresa Romero-Ávila, Ruth Rincón-Heredia, Juan Carlos Martínez-Morales and J. Adolfo García-Sáinz
Receptors 2024, 3(4), 555-573; https://doi.org/10.3390/receptors3040029 - 20 Dec 2024
Viewed by 376
Abstract
Background: Lysophosphatidic acid (LPA) receptor 3 (LPA3) is involved in many physiological and pathophysiological actions of this bioactive lipid, particularly in cancer. The actions of LPA and oleoyl-methoxy glycerophosphothionate (OMPT) were compared in LPA3-transfected HEK 293 cells. Methods: [...] Read more.
Background: Lysophosphatidic acid (LPA) receptor 3 (LPA3) is involved in many physiological and pathophysiological actions of this bioactive lipid, particularly in cancer. The actions of LPA and oleoyl-methoxy glycerophosphothionate (OMPT) were compared in LPA3-transfected HEK 293 cells. Methods: Receptor phosphorylation, ERK 1/2 activation, LPA3-β-arrestin 2 interaction, and changes in intracellular calcium were analyzed. Results: Our data indicate that LPA and OMPT increased LPA3 phosphorylation, OMPT being considerably more potent than LPA. OMPT was also more potent than LPA to activate ERK 1/2. In contrast, OMPT was less effective in increasing intracellular calcium than LPA. The LPA-induced LPA3-β-arrestin 2 interaction was fast and robust, whereas that induced by OMPT was only detected at 60 min of incubation. LPA- and OMPT-induced receptor internalization was fast, but that induced by OMPT was more marked. LPA-induced internalization was blocked by Pitstop 2, whereas OMPT-induced receptor internalization was partially inhibited by Pitstop 2 and Filipin and entirely by the combination of both. When LPA-stimulated cells were rechallenged with 1 µM LPA, hardly any response was detected, i.e., a “refractory” state was induced. However, a conspicuous and robust response was observed if OMPT was used as the second stimulus. Conclusions: The differences in these agents’ actions suggest that OMPT is a biased agonist. These findings suggest that two binding sites for these agonists might exist in the LPA3 receptor, one showing a very high affinity for OMPT and another likely shared by LPA and OMPT (structural analogs) with lower affinity. Full article
Show Figures

Figure 1

Figure 1
<p>Concentration–response curves for LPA- and OMPT-induced LPA<sub>3</sub> receptor phosphorylation. Cells were incubated with the indicated concentrations of the agonists for 15 min. Receptor phosphorylation is expressed as the percentage of the baseline value. The means are plotted, and vertical lines indicate the SEM of 10 experiments performed on different days. Representative autoradiographs (<sup>32</sup>P) and Western blots (WBs) are presented above the graph.</p>
Full article ">Figure 2
<p>Concentration–response curves for LPA- and OMPT-induced ERK 1/2 phosphorylation. Cells were incubated with the indicated concentrations of the agonists for 2 min. ERK 1/2 phosphorylation is expressed as the percentage of the baseline value. The means are plotted, and vertical lines indicate the SEM of 6 experiments performed on different days. Representative Western blots for phosphorylated (pERK) and total (ERK) kinase are presented above the graph.</p>
Full article ">Figure 3
<p>Time course of LPA- and OMPT-induced ERK 1/2 phosphorylation. Cells were incubated for the times indicated with 1 µM of each agonist. ERK 1/2 phosphorylation is expressed as the percentage of the baseline value. The means are plotted, and vertical lines indicate the SEM of 6 experiments performed on different days. Representative Western blots for phosphorylated (pERK) and total (ERK) kinase are presented above the graph. <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 LPA vs. OMPT.</p>
Full article ">Figure 4
<p>Time-course of LPA- and OMPT-induced LPA<sub>3</sub>-β-arrestin interaction (FRET). Cells were incubated for the times indicated with 1 µM LPA (black symbols and line) or 1 µM OMPT (red symbols and line). The baseline WT FRET index was considered as 100%. The means are plotted, and vertical lines indicate the SEM of 9–10 experiments performed on different days; 10–14 cells were analyzed for each experimental condition in all the experiments. Representative FRET index images are presented above the graph. Bars, 10 µm. *** <span class="html-italic">p</span> &lt; 0.001 vs. baseline, ** <span class="html-italic">p</span> &lt; 0.005 vs. baseline, * <span class="html-italic">p</span> &lt; 0.05 vs. baseline (color coded).</p>
Full article ">Figure 5
<p>Time course of 1µM LPA- and 1µM OMPT-induced changes in intracellular (panel (<b>A</b>)) and plasma membrane (panel (<b>B</b>)) fluorescence. In both cases, data are presented as the percentage of the baseline values. The means are plotted, and vertical lines indicate the SEM of 4–5 experiments in which 10–14 images were taken for each condition. Representative images (fluorescence, confocal microscopy) are presented above the graph. Bars, 10 µm. *** <span class="html-italic">p</span> &lt; 0.001 vs. baseline, ** <span class="html-italic">p</span> &lt; 0.005 vs. baseline, * <span class="html-italic">p</span> &lt; 0.05 vs. baseline, <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 LPA vs. OMPT.</p>
Full article ">Figure 6
<p>Effect of Pitstop 2 on LPA and OMPT-induced internalization. Cells were preincubated for 15 min without (gray or pale red symbols and lines) or with Pitstop 2 (PIT) (black or bright red symbols and lines) before being stimulated with 1 µM LPA (panel (<b>A</b>)) or 1 µM OMPT (panel (<b>B</b>)). The means are plotted, and vertical lines indicate the SEM of 4–5 experiments in which 10–14 images were taken for each condition. Representative images (fluorescence, confocal microscopy) are presented above the graphs. <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 LPA vs. OMPT.</p>
Full article ">Figure 7
<p>Effects of Pitstop 2 and Filipin on LPA-, OMPT-, and PMA-induced internalization. Cells were preincubated without any internalization inhibitor or with Pitstop 2 (PIT, 15 min, blue columns), Filipin (FIL, 60 min, yellow columns), or both agents (PIT + FIL, purple columns). After the preincubation, the cells were challenged with the agent and for the time indicated: vehicle (B, baseline, 5 min), 1 µM LPA (5 min), 1 µM OMPT (30 min), and 1 µM PMA (30 min). The baseline intracellular fluorescence was considered as 100%. The means are plotted, and vertical lines indicate the SEM of 5 experiments in which 10–14 images were taken for each condition. Representative images (fluorescence, confocal microscopy) are presented above the graphs. *** <span class="html-italic">p</span> &lt; 0.001 vs. baseline, ** <span class="html-italic">p</span> &lt; 0.01 vs. baseline; <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001, indicated conditions.</p>
Full article ">Figure 8
<p>Cell proliferation as reflected by the MTT and crystal violet staining assays. Proliferation was studied without any agent (none) or with the following stimuli: 10% serum, 1 µM LPA, 1 µM PMA, 1 µM OMPT, or 100 ng/mL EGF. ** <span class="html-italic">p</span> &lt; 0.01 vs. none, *** <span class="html-italic">p</span> &lt; 0.001 vs. none; <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001, comparing indicated conditions.</p>
Full article ">Figure 9
<p>Increases in intracellular calcium in response to LPA and OMPT. Representative calcium tracings of cells incubated with distinct concentrations (color coded) of LPA (panel (<b>A</b>)) or OMPT (panel (<b>B</b>)). The concentration–response curves for LPA- and OMPT-induced intracellular calcium increases are presented in panel (<b>C</b>). The means are plotted, and vertical lines indicate the SEM of 5–8 distinct curves.</p>
Full article ">Figure 10
<p>Response to a second stimulation without or with an intermediate washing step. In the first two columns, cells were incubated with the vehicle, followed by a challenge with LPA or OMPT (control responses). In the second group of columns, cells were stimulated with the agonist indicated (first), and when the response vanished, the second stimulus was applied. In the third group of columns, after the cells were stimulated with the first agonist, they were extensively washed to eliminate the agent and rechallenged with the second stimulus. The concentration of LPA and OMPT was 1 µM in all cases. The means are plotted, and vertical lines indicate the SEM of 8–10 determination with cells from distinct cultures. <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 vs. vehicle+LPA, <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01 vs. vehicle+OMPT, <sup>#</sup> <span class="html-italic">p</span> &lt; 0.001 vs. vehicle+OMPT. Agonist stimulation was for 100 s (sec = seconds). Cell washing procedure took approximately 10 min and cells were challenged after washing.</p>
Full article ">Figure 11
<p>Representative calcium tracings of data that are presented in <a href="#receptors-03-00029-f010" class="html-fig">Figure 10</a>. Agonist stimulation was for 100 s (sec = seconds). Panels (<b>A</b>–<b>F</b>), continuous tracings without washing. Panels (<b>G</b>–<b>J</b>), cells were washed and the response to the second stimulus is shown. Cell washing procedure took approximately 10 min and cells were challenged after washing.</p>
Full article ">
14 pages, 2460 KiB  
Article
The New Nitric Oxide Donor, FOR 911B, Induces Relaxation in Isolated Rat Aorta Involving the NO/sGC/cGMP Pathway and K+ Channels
by Mirelly Cunha da Silva, Florêncio Sousa Gouveia Júnior and Thyago Moreira de Queiroz
Receptors 2024, 3(4), 541-554; https://doi.org/10.3390/receptors3040028 - 10 Dec 2024
Viewed by 435
Abstract
Background: Nitric oxide (NO) is a gaseous molecule considered to be a protagonist in the dilation of blood vessels, and its property and/or bioavailability are reduced in pathophysiological conditions such as cardiovascular diseases. Therefore, its exogenous administration becomes attractive, and new classes of [...] Read more.
Background: Nitric oxide (NO) is a gaseous molecule considered to be a protagonist in the dilation of blood vessels, and its property and/or bioavailability are reduced in pathophysiological conditions such as cardiovascular diseases. Therefore, its exogenous administration becomes attractive, and new classes of compounds able to induce NO release have emerged to minimize the adverse effects found by existing NO donor drugs. Objective: Our aim was to investigate the vasorelaxant effect and mechanism of action induced by the ruthenium complex, which contains nitric oxide in its structure, [Ru(phen)2(TU)NO](PF6)3 (FOR 911B), in isolated rat aorta. Methods: The animals were euthanized, and the aorta artery was identified, removed, and immediately placed in modified Krebs–Henseleit solution. To verify tissue viability, a contraction was obtained with phenylephrine (Phe) (0.1 μM), and to assess endothelial integrity, acetylcholine (ACh) (1 μM) was added. Results: In the present study, we demonstrated, for the first time, that FOR 911B promotes vasorelaxation in a concentration-dependent manner in isolated rat aortic artery rings. After the removal of the vascular endothelium, the potency and efficacy of the relaxation were not altered. With pre-incubation with hydroxocobalamin, the relaxing response was abolished, and with the use of ODQ, the main NO receptor blocker, the vasorelaxant effect was attenuated with a shift of the curve to the right. To investigate the participation of K+ channels, the solution concentration was changed to KCl (20 and 60 mM), and it was pre-incubated with the non-selective K+ channels blocker (TEA). Under these conditions, relaxation was altered, demonstrating that K+ channels are activated by FOR 911B. By selectively blocking the different subtypes of K+ channels with specific blockers, we demonstrated that the subtypes KV, KIR, SKCa, and BKCa are involved in the vasodilator effect induced by FOR 911B. Conclusions: The results obtained demonstrated that FOR 911B promotes vascular relaxation in aortic artery rings in a concentration-dependent manner and independent of the vascular endothelium through the participation of the NO/sGC/cGMP pathway, as well as with the involvement of different K+ channels. Full article
Show Figures

Figure 1

Figure 1
<p>FOR 911B. (<b>A</b>) Planar structure of FOR911B, (<b>B</b>) 3D-structure depiction of the coordination complex, and (<b>C</b>) NO detection assay using a chemiluminescent NO detector.</p>
Full article ">Figure 2
<p>Concentration–response curve for the vasorelaxant effect induced by FOR911B in isolated rat aortic rings, with and without endothelium, pre-contracted with Phe (0.1 μM) (<b>A</b>) and without endothelium, pre-contracted with Phe and KCl (60 mM) (<b>B</b>). Values are expressed as mean ± S.E.M. (n = 6). * <span class="html-italic">p</span> &lt; 0.05 vs. Control, Student’s <span class="html-italic">t</span>-test.</p>
Full article ">Figure 3
<p>Concentration–response curve for the vasorelaxant effect induced by FOR911B in isolated endothelium-denuded rat aortic rings pre-contracted with Phe (0.1 μM) in the presence of L-NAME (100 μM) (<b>A</b>), L- NMMA (100 μM) (<b>B</b>), HDX (30 μM (<b>C</b>), and ODQ (1 μM) (<b>D</b>). Values are expressed as mean ± S.E.M. (n = 6). * <span class="html-italic">p</span> &lt; 0.05 vs. Control, Student’s <span class="html-italic">t</span>-test.</p>
Full article ">Figure 4
<p>Concentration–response curve for the vasorelaxant effect induced by FOR911B in isolated endothelium-denuded rat aortic rings pre-contracted with Phe in the presence of 20 mM KCl (<b>A</b>) and 3 mM TEA (<b>B</b>). Values are expressed as mean ± S.E.M. (n = 6). * <span class="html-italic">p</span> &lt; 0.05 vs. Control, Student’s <span class="html-italic">t</span>-test.</p>
Full article ">Figure 5
<p>Concentration–response curve for the vasorelaxant effect induced by FOR911B in isolated endothelium-denuded rat aortic rings pre-contracted with Phe in the presence of Glibenclamide (10 μM) (<b>A</b>), 4-AP (0.3 mM) (<b>B</b>), Apamine (100 nM) (<b>C</b>), Barium chloride (100 μM) (<b>D</b>), TEA (1 mM) (<b>E</b>), and Iberotoxin (20 nM) (<b>F</b>). Values are expressed as mean ± S.E.M. (n = 6). * <span class="html-italic">p</span> &lt; 0.05 vs. Control, Student’s <span class="html-italic">t</span>-test.</p>
Full article ">
3 pages, 331 KiB  
Commentary
Intracellular ATP Levels: Challenge to the Current Consensus and Its Implications for Intracellular Signaling
by Ferenc A. Antoni
Receptors 2024, 3(4), 538-540; https://doi.org/10.3390/receptors3040027 - 26 Nov 2024
Viewed by 408
Abstract
Work with novel indicators that report intracellular ATP concentrations with improved spatial and temporal resolution have challenged the current consensus that under physiological conditions, intracellular ATP concentrations are not rate-limiting to enzymatic reactions. Recent data from cardiac myocytes and cultured neurons show marked [...] Read more.
Work with novel indicators that report intracellular ATP concentrations with improved spatial and temporal resolution have challenged the current consensus that under physiological conditions, intracellular ATP concentrations are not rate-limiting to enzymatic reactions. Recent data from cardiac myocytes and cultured neurons show marked fluctuations of intracellular ATP levels, as well as evidence for compartmentalization. It is likely that the availability of these genetically encoded indicators will produce rapid progress in the mapping of the dynamics of intracellular ATP concentrations in various types of cells. Here, a brief account of the most recent indicators is provided as well as a review of how natural evolution appears to have obviated the potential shortage of the ATP supply to one of key enzymes of the cyclic AMP signaling cascade, adenylyl cyclase 9. Full article
Show Figures

Figure 1

Figure 1
<p><b>The auto-regulatory motif of AC9 is unique to vertebrates species and highly conserved.</b> The single amino acid codes of the primary sequences of the auto-regulatory motif in the isoform-specfic carboxyl-teminal domain of AC9 are shown. In human AC9 these correspond to positions 1263–1278 (data from Genbank and Ensembl databases).</p>
Full article ">
25 pages, 1429 KiB  
Review
Mechanistic Insights into the Interaction Between Kinin Receptors and Histamine H2 Receptor Pathways in Oxidative Stress
by Marcos Fernandes Gregnani, Leonardo Martins and Wieslawa Agnieszka Fogel
Receptors 2024, 3(4), 513-537; https://doi.org/10.3390/receptors3040026 - 14 Nov 2024
Viewed by 513
Abstract
Reactive oxygen species (ROS) encompass various molecular oxygen derivatives naturally produced during aerobic metabolism, including superoxide anions, hydrogen peroxide, and hydroxyl radicals. Excessive ROS production leads to oxidative distress, causing cellular damage and contributing to various pathologies, often alongside inflammation. Endogenous sources of [...] Read more.
Reactive oxygen species (ROS) encompass various molecular oxygen derivatives naturally produced during aerobic metabolism, including superoxide anions, hydrogen peroxide, and hydroxyl radicals. Excessive ROS production leads to oxidative distress, causing cellular damage and contributing to various pathologies, often alongside inflammation. Endogenous sources of ROS include mitochondrial activity and NADPH oxidases. The antioxidant system, comprising enzymes such as superoxide dismutase, peroxiredoxin, and catalase, mitigates ROS-induced damage. This review explores the regulation of ROS by membrane receptors, focusing on B1 and B2 kinin receptors and histamine H2 receptors, which are implicated in vasodilation, angiogenesis, inflammation, and gastric acid secretion. Understanding these interactions provides insights into ROS modulation and its role in disease mechanisms. Full article
Show Figures

Figure 1

Figure 1
<p>This flowchart illustrates the search strategy applied to find possible interactions between kinin receptors (B1R and B2R) and histamine H2 receptors. Horizontally oriented boxes represent the number of manuscripts reviewed at each stage.</p>
Full article ">Figure 2
<p>Overview of the role of the B1 and B2 kinin receptors and the H2 histamine receptor in modulating the production of reactive oxide species (ROS) in different organs and tissues. Bradykinin B1 Receptor (B1R), bradykinin B2 receptor (B2R), histamine H2 receptor (H2R), reactive oxygen species (ROS), nicotinamide adenine dinucleotide phosphate oxidase (NADPH oxidase), superoxide dismutase (SOD), glutathione (reduced form) (GSH), glutathione disulfide (oxidized form) (GSSG), catalase (CAT), malondialdehyde (MDA), thiobarbituric acid reactive substances (TBARSs), glutathione peroxidase (GPx), hydrogen peroxide (H<sub>2</sub>O<sub>2</sub>), and myeloperoxidase (MPO).</p>
Full article ">
19 pages, 1554 KiB  
Review
Molecular Perspectives on Prostate Cancer: The Role of microRNAs in Androgen Receptor Regulation
by Asbiel Felipe Garibaldi-Ríos, Alicia Rivera-Cameras, Luis E. Figuera, Guillermo Moisés Zúñiga-González, Belinda Claudia Gómez-Meda, José Elías García-Ortíz and Martha Patricia Gallegos-Arreola
Receptors 2024, 3(4), 494-512; https://doi.org/10.3390/receptors3040025 - 12 Nov 2024
Viewed by 642
Abstract
Prostate cancer (PCa) is the most prevalent cancer among men globally. In addition to environmental risk factors, genetic factors play a crucial role in its development and progression, highlighting the regulation of key genes as an essential aspect. The androgen receptor gene ( [...] Read more.
Prostate cancer (PCa) is the most prevalent cancer among men globally. In addition to environmental risk factors, genetic factors play a crucial role in its development and progression, highlighting the regulation of key genes as an essential aspect. The androgen receptor gene (AR) plays a pivotal role in this disease, so its post-transcriptional regulation must be meticulously coordinated. In this review, we explore the role of microRNAs (miRNAs) in the regulation of AR in PCa, a field not yet fully investigated. We note that the AR, due to its extensive 3′UTR region, is targeted by numerous miRNAs, and that this regulation can occur at different levels: directly, indirectly, and through mutual regulation, thus amplifying the influence of these molecules on AR regulation. Full article
Show Figures

Figure 1

Figure 1
<p>AR pathway. Free androgens in the bloodstream can diffuse freely across the plasma membrane of cells. Within the cell cytoplasm, testosterone is converted to dihydrotestosterone (DHT) by the action of the enzyme 5α-reductase. This conversion is essential because DHT has an increased affinity for AR. Within the cell cytoplasm, the AR remains inactive and bound to chaperone proteins, such as HSPs. However, when the androgen binds to the AR, it undergoes conformational changes that allow dissociation from chaperone proteins and subsequent activation. Once active, the androgen-bound AR forms a homodimer with another complex. This complex then translocates to the cell nucleus, where it binds to the AREs, located in the promoters of various target genes. The binding of the AR to the AREs allows the recruitment of coactivator proteins that modulate the transcription of genes involved in cell proliferation, differentiation, and apoptosis [<a href="#B14-receptors-03-00025" class="html-bibr">14</a>,<a href="#B15-receptors-03-00025" class="html-bibr">15</a>,<a href="#B16-receptors-03-00025" class="html-bibr">16</a>,<a href="#B26-receptors-03-00025" class="html-bibr">26</a>]; created with Biorender.com.</p>
Full article ">Figure 2
<p>Indirect regulatory mechanisms of miRNAs on the <span class="html-italic">AR</span> gene. (<b>A</b>) Some miRNAs regulate <span class="html-italic">AR</span> regulators, such as <span class="html-italic">HSP</span> and <span class="html-italic">MYC</span>, which act as transcription factors for <span class="html-italic">AR</span> expression, or <span class="html-italic">DMT1</span>, which is responsible for methylating the <span class="html-italic">AR</span> promoter, thereby regulating its expression. (<b>B</b>) Furthermore, it has been observed that <span class="html-italic">AR</span> can be regulated by miRNAs that control master genes in <span class="html-italic">AR</span> splicing, such as hnRNPH1, or genes that act as cofactors (<span class="html-italic">MYC</span>) or transporters (<span class="html-italic">FNDC1</span>) (<b>C</b>), thus affecting the overall activity of <span class="html-italic">AR</span>; created with Biorender.com.</p>
Full article ">Figure 3
<p>AR–miRNAs mutual regulation. In positive feedback, <span class="html-italic">AR</span> facilitates the transcription of hsa-miR-21, which in turn promotes <span class="html-italic">AR</span> expression by regulating <span class="html-italic">PTEN,</span> creating a reinforcing cycle. In negative feedback, AR increases the expression of hsa-miR-31 and hsa-miR-421, which inhibit <span class="html-italic">AR</span> expression, establishing a control mechanism that prevents <span class="html-italic">AR</span> overexpression. Finally, mutual feedback shows how <span class="html-italic">AR</span> represses hsa-miR-190a, and this miRNA, in turn, represses <span class="html-italic">AR</span> expression by regulating the activity of <span class="html-italic">YB1</span>, a coactivator of <span class="html-italic">AR</span>, forming a reciprocal suppression cycle; created with Biorender.com.</p>
Full article ">
20 pages, 589 KiB  
Review
The Regulation and Function of the Amino Acid Transporters LAT1, ASCT2, xCT in Urological Cancers
by Xue Zhao, Shinichi Sakamoto, Shinpei Saito, Sangjon Pae, Yasutaka Yamada, Sanji Kanaoka, Jiaxing Wei, Yusuke Goto, Tomokazu Sazuka, Yusuke Imamura, Naohiko Anzai and Tomohiko Ichikawa
Receptors 2024, 3(4), 474-493; https://doi.org/10.3390/receptors3040024 - 31 Oct 2024
Viewed by 696
Abstract
Amino acid transporters play pivotal roles in cancer biology, including in urological cancers. Among them, L-type amino acid transporter 1 (LAT1), alanine-serine-cysteine transporter 2 (ASCT2), and cystine-glutamate transporter (xCT) have garnered significant attention due to their involvement in various aspects of tumor progression [...] Read more.
Amino acid transporters play pivotal roles in cancer biology, including in urological cancers. Among them, L-type amino acid transporter 1 (LAT1), alanine-serine-cysteine transporter 2 (ASCT2), and cystine-glutamate transporter (xCT) have garnered significant attention due to their involvement in various aspects of tumor progression and response to therapy. This review focuses on elucidating the regulation and functions of these amino acid transporters in urological cancers, including prostate, bladder, and renal cancers. Understanding the intricate regulatory mechanisms governing these amino acid transporters is essential for developing effective therapeutic strategies. Furthermore, exploring their interactions with signaling pathways and microenvironmental cues in the context of urological cancers may uncover novel therapeutic vulnerabilities. This comprehensive overview highlights the importance of amino acid transporters, particularly LAT1, ASCT2, and xCT, in urological cancers and underscores the potential of their inhibitors as therapeutic targets for improving patient outcomes. Full article
Show Figures

Figure 1

Figure 1
<p>LAT1, ASCT2, and xCT as key transporters responsible for substance transport. They supply branched-chain amino acids (BCAAs) to the mammalian target of rapamycin complex 1 (mTORC1), fueling the tricarboxylic acid (TCA) cycle for energy production and fatty acid metabolism, thereby enhancing cell proliferation. Conversely, the nuclear expression of LAT1, ASCT2, and xCT is modulated by mTORC1 downstream factors. Gln = Glutamine; cMyc = cellular Myc; ATF4 = Activating Transcription Factor 4; NRF2 = Nuclear Factor Erythroid 2-Related Factor 2; SIRT4 = Sirtuin 4; GDH = glutamate dehydrogenase; Leu = Leucine; GSH = Glutathione. Upregulation: red upward arrow; Downregulation: green downward arrow; Inhibition: red “×” symbol; Inverse correlation between the expression of the former and the latter: green curve.</p>
Full article ">
17 pages, 6649 KiB  
Article
Glycosyl Mobile Radical Structures of Folic Acid Receptors Impact the Internalization of Functionalized Folate Amphiphilic Alternating Copolymer in Cancer Cells
by Emilyn B. Aucoin, Elizabeth Skapinker, Abdulrahman M. Yaish, Yunfan Li, Haley L. Kombargi, Daniel Jeyaraj, Pankaj Garg, Nicole Mendonza, Cecile Malardier-Jugroot and Myron R. Szewczuk
Receptors 2024, 3(4), 457-473; https://doi.org/10.3390/receptors3040023 - 21 Oct 2024
Viewed by 1094
Abstract
Folate receptor alpha (FRα) is a glycosylphosphatidylinositol (GPI) membrane-anchored protein containing three N-glycosylated residues at the N47, N139, and N179 termini. These glycosylation sites have been reported to be crucial for the receptor’s structural integrity and its ability to bind and internalize FA. [...] Read more.
Folate receptor alpha (FRα) is a glycosylphosphatidylinositol (GPI) membrane-anchored protein containing three N-glycosylated residues at the N47, N139, and N179 termini. These glycosylation sites have been reported to be crucial for the receptor’s structural integrity and its ability to bind and internalize FA. Here, we investigated the role of FRα glycosylation in the binding and internalization efficacy of FA–DABA–SMA in pancreatic PANC-1 cancer cells. There is a strong association of the FA copolymer with FRα with a Pearson coefficient R-value of 0.7179. PANC-1 cancer cells were pretreated with maackia amurensis lectin II (MAL-2), sambucus Nigra lectin (SNA-1), peanut agglutinin (PNA), and wheat germ agglutinin lectin (WGA) at different doses followed by 20 kDa and 350 kDa FA–DABA–SMA loaded with coumarin 153 (C153). Increasing the dosage of MAL2, SNA-1, PNA, and WGA concomitantly and significantly increased the internalization of C153-loaded FA–DABA–SMA in the cells. The half maximal effective lectin concentrations (EC50) to induce cellular internalization into the cytoplasm of the lectins for MAL-2 were 35.88 µg/mL, 3.051 µg/mL for SNA-1, 7.883 µg/mL for PNA, and 0.898 µg/mL for WGA. Live cell imaging of the internalization of 20 kDa and 350 kDa FA copolymers indicated an aggregation of 350 kDa copolymer with FRα in the cytoplasm. In contrast, the 20 kDa FA copolymer remained in the membrane. The data indicate for the first time that the mobile positions of the glycosyl radical groups and the receptor tilt in generating steric hindrance impacted the individual FRα receptors in the binding and internalization of 350 kDa FA–DABA–SMA in cancer cells. Full article
Show Figures

Figure 1

Figure 1
<p>Concentration dependence of (<b>A</b>) MAL-2, SNA-1, PNA, WGA, and their half maximal effective concentration (EC50) of lectins (<b>B</b>,<b>C</b>) MAL-2, (<b>D</b>,<b>E</b>) SNA-1, (<b>F</b>,<b>G</b>) PNA, and (<b>H</b>,<b>I</b>) WGA to induce C153-loaded FA–DABA–SMA internalization into the cytoplasm of PANC-1 cells. Cells were plated in a 24-well plate and incubated overnight. Each lectin was added to wells at designated concentrations, and cells were incubated for 35 min and washed with 1× PBS. Coumarin 153 (C153; C16H14F3NO2) belongs to the class of 7-aminocoumarins and functions as a green fluorochrome. Cells were incubated in 150 µg/mL of CellMask<sup>TM</sup> Deep Red plasma membrane stain diluted 1:1000 in 1× PBS for 7 min and washed three times in 1× PBS. C153-loaded FA–DABA–SMA was added to wells, incubated for 1 h, and washed three more times with 1× PBS. Samples were mounted onto glass slides with Fluoroshield Mounting Medium and imaged using a ZEISS Axio Imager M2 fluorescent microscope (Carl Zeiss AG, Oberkochen, Germany) at 40× magnification. (<b>B</b>–<b>I</b>) The results were analyzed with Corel Photo-Paint X8 for background means, green C-153 stain image means, and pixel measurements. The results are depicted as scatter plots for visualization using dots to represent fluorescent density (n = 7–11). The mean fluorescent density corrected for background (bkg) + SEM is indicated for each concentration of lectin. The mean fluorescent density at each concentration of lectin was compared to the mean of the untreated cells by ANOVA using a Fisher’s LSD with 95% confidence. Statistical significance is indicated with asterisks. (<b>C</b>,<b>E</b>,<b>G</b>,<b>I</b>) The EC50 is defined as the concentration of agonist that results in a response that is halfway between the baseline and maximum response. This concentration is used to measure each lectin’s potency to induce internalization of the copolymer. The dashed red line is the calculated simple linear regression using the GraphPad Prism software (version 10.3.1.509).</p>
Full article ">Figure 2
<p>Evaluating the concentration dependence of coumarin-153-loaded FA–DABA–SMA (NP-C153) internalization into cells in the presence of 100 µg/mL MAL2 lectin. Cells were plated in a 24-well plate and incubated overnight. MAL-2 was added to wells, and the cells were incubated for 35 min. Then, they were washed with 1× PBS. Cells were incubated in 150 <math display="inline"><semantics> <mrow> <mi mathvariant="sans-serif">μ</mi> <mi mathvariant="normal">g</mi> <mo>/</mo> <mi mathvariant="normal">m</mi> <mi mathvariant="normal">L</mi> </mrow> </semantics></math> of CellMask<sup>TM</sup> Deep Red plasma membrane stain diluted 1:1000 in 1× PBS for 7 min and washed three times in 1× PBS. C153-loaded FA–DABA–SMA was added to wells at different dilutions, incubated for 1 h, and washed three more times with 11× PBS. Samples were mounted onto glass slides with Fluoroshield Mounting Medium and imaged using a ZEISS Axio Imager M2 fluorescent microscope (Carl Zeiss AG, Oberkochen, Germany) at 40× magnification. The results were analyzed with Corel Photo-Paint X8 for background means, image means, and pixel measurements. The results are depicted as a bar graph for visualization of the mean intracellular NP-C153 fluorescent density corrected for background (bkg) <math display="inline"><semantics> <mrow> <mo>±</mo> <mtext> </mtext> </mrow> </semantics></math> SEM of varying dilutions of copolymer with and without MAL-2. The mean fluorescent density of each dilution without MAL-2 was compared to the mean fluorescent density of the same dilution with MAL-2 by ANOVA with a Fisher’s LSD multiple comparisons test with 95% confidence. Asterisks indicate statistical significance. Abbreviations: NP-C153: C153-loaded FA–DABA–SMA; NP: nanoparticle; bkg: background; ns: not significant.</p>
Full article ">Figure 3
<p>Kinetics of binding and internalization of C153-loaded FA–DABA–SMA into PANC-1 cells. Images in green depict fluorescence for C153-FA–DABA–SMA, and red fluorescence depicts CellMask™ Deep red plasma membrane stain. CellMaskTM Deep Red, plasma membrane stain, diluted 1:1000 in 1× PBS, was added to cells at 150 µL on circular glass slides in 24-well tissue culture dishes for 7 min and washed three times in 1× PBS. C153-FA–DABA–SMA copolymers were added to wells at indicated time points at 30, 60, 90, and 120 s and washed three times with 1× PBS. Cells on glass slides were mounted on microscope slides containing Fluoroshield Mounting Medium. Images were visualized using a ZEISS Axio Imager M2 fluorescent microscope (Carl Zeiss AG, Oerkochen, Germany) at 40× magnification.</p>
Full article ">Figure 4
<p>Colocalization of binding of C153-loaded FA–DABA–SMA to the (<b>A</b>) cell membrane and (<b>B</b>) the FRα receptor in PANC-1 cells. Images in green depict fluorescence for C153-FA–DABA–SMA, and red fluorescence depicts CellMask™ Deep red plasma membrane stain. Illustrated boxes signify the enlargement of the area for visualization. (<b>A</b>) CellMaskTM Deep Red, plasma membrane stain, diluted 1:1000 in 1× PBS, was added to cells at 150 µL on circular glass slides in 24-well tissue culture dishes for 7 min and washed three times in 1× PBS. (<b>B</b>) Cells (50,000 cells) were plated on 12 mm circular glass slides in culture media containing 10% fetal calf sera for 24 h and treated with C153-loaded FA–DABA–SMA. Cells were fixed, permeabilized, and immunostained with mouse monoclonal IgG anti-hFOLR1, washed in 1× PBS, and followed with goat anti-mouse conjugated with Alexa Fluor594. Stained cells were visualized using a Zeiss M2 imager fluorescent microscope with a 40× objective. Arrows represent the yellow color of green (C153-FA–DABA–SMA) and red (anti-FR conjugated Alexa 594) colocalization. The Pearson correlation coefficient was measured on a total image of cells and expressed as the Pearson coefficient R-value, which was calculated using Zeiss M2 imager software (Carl Zeiss™ AxioVision Rel. 4.8.2). The data represent one of three independent experiments showing similar results.</p>
Full article ">Figure 5
<p>Live cell microscopy using an inverted microscope (Leica DMi8) equipped with a Photron Fastcam SA-Z high-speed camera. PANC-1 cells (50,000 cells) were plated on a 35 mm MatTek dish, No. 1.5 gridded coverslip at 14 mm glass diameter in culture media containing 10% fetal calf sera for 24 h and left untreated. Live cells were washed with 1× PBS and incubated in 150 µL of CellMaskTM Deep Red plasma membrane stain diluted 1:1000 in 1× PBS for 7 min and washed three times in 1× PBS. (<b>A</b>) 350 kDa NP, (<b>B</b>) 350 kDa NP video, (<b>C</b>) 20 kDa NP, and (<b>D</b>) 20 kDa NP video of C153-loaded FA–DABA–SMA particles (green) were added to wells, then incubated for 1 h, and washed another three times with 1× PBS. Samples were then mounted onto glass slides using Fluoroshield Mounting Medium. Images were visualized using a Leica DMi8 fitted with a high-speed camera from Photron Fastcam SA-Z with a 100× oil objective. The kinetic expression of the cell membrane (red) and FA–DABA–SMA particles loaded with C153 (green) binding were taken every second and recorded as a <a href="#app1-receptors-03-00023" class="html-app">Video S1</a> (<b>B</b>) for the 350 kDa copolymer and <a href="#app1-receptors-03-00023" class="html-app">Video S2</a> (<b>D</b>) for the 20 kDa copolymer.</p>
Full article ">Figure 6
<p>The efficacy of the internalization of the 350 kDa and 20 kDa C153-loaded FA–DABA–SMA NP into PANC-1 cells following treatment with (<b>A</b>) MAL2, (<b>B</b>) SNA-1, (<b>C</b>) PNA, and (<b>D</b>) WGA lectins at the indicated concentrations. PANC-1 cells were plated at a density of 100,000 to 200,00 cells/mL on glass coverslips in 24-well plates. Cells were treated with 200 µL of lectin at concentrations of 100 <math display="inline"><semantics> <mrow> <mi mathvariant="sans-serif">μ</mi> </mrow> </semantics></math>g/mL, 10 <math display="inline"><semantics> <mrow> <mi mathvariant="sans-serif">μ</mi> </mrow> </semantics></math>g/mL, and 1 <math display="inline"><semantics> <mrow> <mi mathvariant="sans-serif">μ</mi> </mrow> </semantics></math>g/mL and incubated for 35 min. Cells were then washed with 1× PBS and incubated in CellMask™ Deep Red Plasma membrane stain and diluted to a concentration of 1:1000 in 1× PBS for 7 min. Cells were washed 3 times with 1× PBS and then treated with 170 <math display="inline"><semantics> <mrow> <mi mathvariant="sans-serif">μ</mi> </mrow> </semantics></math>L of 20 kDa and 350 kDa C153-loaded FA–DABA–SMA per well and incubated for 1 h. Cells were subsequently washed with 1× PBS and mounted onto glass slides with Fluoroshield Mounting Medium. The images were visualized using a ZEISS Axio Imager M2 fluorescent microscope (Carl Zeiss AG, Oberkochen, Germany) at 40× magnification. The results are depicted as scatter bar graphs of the mean fluorescent density corrected for background luminescence ± SEM of 3 independent experiments performed in triplicates. The results of the treatments with 20 kDa and 350 kDa copolymers were compared using the one-way ANOVA Fisher (LSD) test comparisons with 95% confidence, indicated by asterisks for statistical significance. ns: not significant.</p>
Full article ">Figure 7
<p>SMA in the nanotube and nanosheet configuration. (<b>A</b>) Schematic and molecular modeling representations of 20 kDa SMA self-assembly in neutral water. (<b>B</b>) Dynamic light scattering (DLS) spectrum of 20 kDa SMA in neutral water from 10 to 80 °C showing the stability of the structure with a ~100 nm hydrodynamic radius. (<b>C</b>) Schematic and molecular modeling representations of 350 kDa SMA self-assembly in neutral water. (<b>D</b>) DLS spectrum of 350 kDa SMA in neutral water from 10 to 80 °C showing the stability of the structure with a ~1000 nm hydrodynamic radius and unassociated SMA chains at ~10–20 nm. Images are adapted with permission of Taylor &amp; Francis from Molecular Simulation, Characterization of a novel self-association of an alternating copolymer into nanotubes in solution, Malardier-Jugroot, C., van de Ven, T. G. M., and Whitehead, M. A., 31, 2–1, 2005; permission conveyed through Copyright Clearance Center, Inc. [<a href="#B12-receptors-03-00023" class="html-bibr">12</a>], and adapted from Chemical Physics Letters, 636, McTaggart, M., Malardier-Jugroot, C., and Jugroot, M., Self-assembled biomimetic nanoreactors I: polymeric template, 206–220, Copyright (2015), with permission from Elsevier [<a href="#B25-receptors-03-00023" class="html-bibr">25</a>] and by AIP Publishing (<a href="https://doi.org/10.1063/5.0046081" target="_blank">https://doi.org/10.1063/5.0046081</a>, accessed on 15 August 2024).</p>
Full article ">Figure 8
<p>Graphical representation to visualize the structure of the folic acid receptor (FR<math display="inline"><semantics> <mrow> <mi>α</mi> </mrow> </semantics></math>). Green structures depict amino acid chains for FR<math display="inline"><semantics> <mrow> <mi>α</mi> <mo>,</mo> </mrow> </semantics></math> and N-acetyl-D-glucosamine (GlcNAc) sites are shown as purple projections from the amino acid chains. The folic acid binding pocket is labeled in red, and folic acid is labeled in grey. (<b>A</b>) The N139 glycosylation site, specifically with the bright green region highlighting the connection of this site to the binding pocket. At the same time, image (<b>B</b>) shows the full structure of the folic acid receptor with the binding pocket exposed, and image (<b>C</b>) shows the structure rotated 180° relative to (<b>B</b>). Images adapted from the RCSB Protein Data Bank [<a href="https://doi.org/10.2210/pdb4LRH/pdb" target="_blank">https://doi.org/10.2210/pdb4LRH/pdb</a>, accessed on 15 August 2024] are distributed under a CC0 1.0 Universal (CC0 1.0) Public Domain Dedication license [<a href="https://creativecommons.org/publicdomain/zero/1.0/" target="_blank">https://creativecommons.org/publicdomain/zero/1.0/</a>, accessed on 15 August 2024] [<a href="#B1-receptors-03-00023" class="html-bibr">1</a>].</p>
Full article ">
13 pages, 2240 KiB  
Review
Nuclear Receptors: Mechanistic Insights into Endocrine Resistance in Prostate and Breast Cancers
by Macrina Beatriz Silva-Cázares, Stephanie I. Nuñez-Olvera, Ricardo Hernández-Barrientos, Enoc Mariano Cortés-Malagón, María Elizbeth Alvarez-Sánchez and Jonathan Puente-Rivera
Receptors 2024, 3(4), 444-456; https://doi.org/10.3390/receptors3040022 - 14 Oct 2024
Viewed by 839
Abstract
This review focuses on the pivotal roles of nuclear receptors (NRs) in driving endocrine resistance in prostate and breast cancers. In prostate cancer (PCa), androgen receptor (AR) amplification, mutations, and altered coactivator interactions sustain tumor growth under androgen deprivation therapy (ADT), leading to [...] Read more.
This review focuses on the pivotal roles of nuclear receptors (NRs) in driving endocrine resistance in prostate and breast cancers. In prostate cancer (PCa), androgen receptor (AR) amplification, mutations, and altered coactivator interactions sustain tumor growth under androgen deprivation therapy (ADT), leading to castration-resistant prostate cancer (CRPC). Orphan NRs like RORβ, TLX, and COUP-TFII further contribute to CRPC by regulating stemness and therapeutic resistance mechanisms. In breast cancer, NRs, including estrogen receptor alpha (ERα), androgen receptor (AR), glucocorticoid receptor (GR), and liver receptor homolog-1 (LRH-1), modulate estrogen signaling pathways and alternative survival mechanisms like PI3K/AKT/mTOR and NFκB, promoting resistance to endocrine therapies such as tamoxifen. Understanding these NR-mediated mechanisms is critical for developing targeted therapies to overcome endocrine resistance and improve patient outcomes in hormone-dependent cancers. Full article
Show Figures

Figure 1

Figure 1
<p>Impact of elevated nuclear receptor expression on the Progression of Castration-Resistant Prostate Cancer (CRPC). Upregulation (up red arrows) of nuclear receptors and their role in CRPC development. These receptors contribute to key processes such as tumor growth and metastasis (GR), maintenance of stem cell populations, cellular proliferation and migration (RORβ, TLX, COUP-TFII), migration, invasion, androgen biosynthesis (NURR1, LRH1), and tumor progression (ERRα), ultimately leading to resistance against androgen deprivation therapy (ADT). Each receptor activates (black arrows) specific pathways that enhance tumor aggressiveness and resistance mechanisms, highlighting their potential as therapeutic targets for CRPC. Created with <a href="http://BioRender.com" target="_blank">BioRender.com</a> (accessed on 1 June 2024).</p>
Full article ">Figure 2
<p>Modulation of nuclear receptor functions in breast cancer endocrine resistance. (<b>A</b>) The glucocorticoid receptor (GR) and estrogen receptor (ER) synergistically modulate gene expression, enhancing ZBTB16 expression (green arrow) and contributing to a reduction in tamoxifen resistance (down green arrow). In contrast, GR and ER negatively regulate CDKs (CDK1, CDK2, and CDK6) (red inhibition arrow), leading to inhibition of proliferation and a decrease in tumor volume. (<b>B</b>) NFκB activation (up green arrow) by LRH-1 and inactivation (red inhibition arrow) by COUP-TFII promote stemness and endocrine resistance, suggesting a regulatory role in mechanisms of endocrine resistance in breast cancer. (<b>C</b>) NURR1 downregulation (down red arrow) and SPY1 upregulation (result in the activation of the ERK1/2 signaling pathway, which in turn activates (black arrow) the estrogen receptor (ER). The activation of ER is also associated with a decrease in tamoxifen resistance (down green arrow). Created with <a href="http://BioRender.com" target="_blank">BioRender.com</a> (accessed on 1 June 2024).</p>
Full article ">
19 pages, 2436 KiB  
Review
Receptor-Based Strategies for Overcoming Resistance in Cancer Therapy
by Naresh Sah, Abdul Althaf Shaik, Ganesh Acharya, Manikantha Dunna, Ashok Silwal, Sejal Sharma, Sabiha Khan and Sounak Bagchi
Receptors 2024, 3(4), 425-443; https://doi.org/10.3390/receptors3040021 - 24 Sep 2024
Viewed by 1027
Abstract
This review article explores the fundamental role of receptor targeting in overcoming drug resistance in cancer therapy, an area of critical concern given the persistently high rates of cancer morbidity and mortality globally. We highlight how receptor biology intersects with the development of [...] Read more.
This review article explores the fundamental role of receptor targeting in overcoming drug resistance in cancer therapy, an area of critical concern given the persistently high rates of cancer morbidity and mortality globally. We highlight how receptor biology intersects with the development of therapeutic resistance with a specific focus on anti-angiogenic agents, immune checkpoint inhibitors, and monoclonal antibodies, which directly or indirectly influence receptor pathways. We also explore how other receptor tyrosine kinases can initially suppress tumor growth, yet often lead to resistance, underscoring the need for novel combinatorial approaches that incorporate advanced receptor modulation techniques. Further, the review delves into the mechanisms by which modulation of the tumor microenvironment and immune system via receptor pathways can overcome resistance to traditional immunotherapies. Additionally, emerging technologies in receptor-targeted nanomedicine are also highlighted, showcasing their potential to revolutionize drug delivery and improve therapeutic outcomes by targeting specific receptor interactions. Ultimately, this review calls for a deeper understanding of receptor dynamics to develop more precise interventions, including insights from various healthcare settings that can prevent or circumvent drug resistance, thus enhancing patient outcomes in oncology. Full article
Show Figures

Figure 1

Figure 1
<p>An overview of receptor-based strategies for overcoming resistance in cancer therapy is summarized in this review article.</p>
Full article ">Figure 2
<p>Common scenario of anti-angiogenesis treatments. Anti-angiogenesis treatments initially inhibit primary tumor growth but often lead to eventual relapse due to the development of therapy resistance. Tumor cells resist through hypoxia and the upregulation of alternative pro-angiogenic factors, like Ang1/2, FGF, PDGF, EGF, HGF, and VEGF. Additionally, recruited vascular modulator cells, like TAMs, MDSCs, and EPCs, promote new blood vessel formation. Tumor cells adapt to energy stress and survival mechanisms, like autophagy, dormancy, and cancer stem cells. Resistance to anti-angiogenic drugs could be overcome by various approaches. This includes targeting alternative angiogenic pathways, anti-angiogenic immunotherapy, tumor microenvironment modulation, drug delivery optimization, and therapeutic combinations with angiogenic inhibitors. Combination therapies aim to enhance treatment efficacy and prevent tumor relapse.</p>
Full article ">Figure 3
<p>Schematic depiction detailing how nanodrug delivery combats immune checkpoint blockade resistance in hepatocellular carcinoma (HCC) through their antitumor mechanisms.</p>
Full article ">Figure 4
<p>Mechanisms and strategies to counter osimertinib resistance.</p>
Full article ">
Previous Issue
Next Issue
Back to TopTop