Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
J Med Chem ; 65(7): 5821-5829, 2022 04 14.
Article in English | MEDLINE | ID: mdl-35302785

ABSTRACT

1α,25-dihydroxyvitamin D3 (1,25D3) regulates many physiological processes in vertebrates by binding to the vitamin D receptor (VDR). Phylogenetic analysis indicates that jawless fishes are the most basal vertebrates exhibiting a VDR gene. To elucidate the mechanism driving VDR activation during evolution, we determined the crystal structure of the VDR ligand-binding domain (LBD) complex from the basal vertebratePetromyzon marinus, sea lamprey (lVDR). Comparison of three-dimensional crystal structures of the lVDR-1,25D3 complex with higher vertebrate VDR-1,25D3 structures suggests that 1,25D3 binds to lVDR similarly to human VDR, but with unique features for lVDR around linker regions between H11 and H12 and between H9 and H10. These structural differences may contribute to the marked species differences in transcriptional responses. Furthermore, residue co-evolution analysis of VDR across vertebrates identifies amino acid positions in H9 and the large insertion domain VDR LBD specific as correlated.


Subject(s)
Lampreys , Receptors, Calcitriol , Animals , Lampreys/metabolism , Ligands , Phylogeny , Protein Binding , Receptors, Calcitriol/metabolism , Vitamin D
2.
Nat Commun ; 11(1): 6249, 2020 12 07.
Article in English | MEDLINE | ID: mdl-33288743

ABSTRACT

The bioactive vitamin D3, 1α,25(OH)2D3, plays a central role in calcium homeostasis by controlling the activity of the vitamin D receptor (VDR) in various tissues. Hypercalcemia secondary to high circulating levels of vitamin D3 leads to hypercalciuria, nephrocalcinosis and renal dysfunctions. Current therapeutic strategies aim at limiting calcium intake, absorption and resorption, or 1α,25(OH)2D3 synthesis, but are poorly efficient. In this study, we identify WBP4 as a new VDR interactant, and demonstrate that it controls VDR subcellular localization. Moreover, we show that the vitamin D analogue ZK168281 enhances the interaction between VDR and WBP4 in the cytosol, and normalizes the expression of VDR target genes and serum calcium levels in 1α,25(OH)2D3-intoxicated mice. As ZK168281 also blunts 1α,25(OH)2D3-induced VDR signaling in fibroblasts of a patient with impaired vitamin D degradation, this VDR antagonist represents a promising therapeutic option for 1α,25(OH)2D3-induced hypercalcemia.


Subject(s)
Calcium/metabolism , Hypercalcemia/metabolism , Receptors, Calcitriol/metabolism , Vitamin D/pharmacology , Animals , Calcitriol/analogs & derivatives , Calcitriol/pharmacology , Cell Line , Cell Line, Tumor , Cytosol/metabolism , Gene Expression/drug effects , HeLa Cells , Humans , Hypercalcemia/genetics , Hypercalcemia/prevention & control , Male , Mice, Inbred C57BL , Mice, Knockout , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Binding , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Rats , Receptors, Calcitriol/genetics , Vitamin D/analogs & derivatives
3.
PLoS One ; 15(10): e0240676, 2020.
Article in English | MEDLINE | ID: mdl-33052979

ABSTRACT

The impact of cancer on lifespan is significantly increasing worldwide. Enhanced activity of drug efflux pumps and the incidences of the tumor microenvironment such as the apparition of a hypoxic gradient inside of the bulk tumor, are the major causes of chemotherapy failure. For instance, expression of Hypoxia-inducible factor (HIF-1α) has been associated with metastasis, resistance to chemotherapy and reduced survival rate. One of the current challenges to fight against cancer is therefore to find new molecules with therapeutic potential that could overcome this chemoresistance. In the present study, we focused on the bioactive plant flavonoid quercetin, which has strong antioxidant and anti-proliferative properties. We examined the efficacy of combined treatments of quercetin and the anti-cancer drugs gemcitabine and doxorubicin, known to specifically act on human pancreatic and hepatic cancer cells, respectively. Moreover, our study aimed to investigate more in-depth the implication of the multidrug transporter MDR1 and HIF-1α n chemoresistance and if quercetin could act on the activity of the drug efflux pumps and the hypoxia-associated effects. We observed that the anti-cancer drugs, were more effective when administered in combination with quercetin, as shown by an increased percentage of dead cells up to 60% in both 2D and 3D cultures. In addition, our results indicated that the combination of anti-cancer drugs and quercetin down-regulated the expression of HIF-1α and increased the expression levels of the regulator of apoptosis p53. Moreover, we observed that quercetin could inhibit the efflux activity of MDR1. Finally, our in vitro study suggests that the efficiency of the chemotherapeutic activity of known anti-cancer drugs might be significantly increased upon combination with quercetin. This flavonoid may therefore be a promising pharmacological agent for novel combination therapy since it potentializes the cytotoxic activity of gemcitabine and doxorubicin on by targeting the chemoresistance developed by the pancreatic and liver cancer cells respectively.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antioxidants/pharmacology , Deoxycytidine/analogs & derivatives , Doxorubicin/pharmacology , Quercetin/pharmacology , ATP Binding Cassette Transporter, Subfamily B/metabolism , Adenocarcinoma/drug therapy , Cell Hypoxia/drug effects , Deoxycytidine/pharmacology , Drug Resistance, Neoplasm/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Hep G2 Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Liver Neoplasms/drug therapy , Pancreatic Neoplasms/drug therapy , Tumor Microenvironment/drug effects , Gemcitabine
4.
Nucleic Acids Res ; 48(19): 11199-11213, 2020 11 04.
Article in English | MEDLINE | ID: mdl-32990725

ABSTRACT

The MED1 subunit of the Mediator complex is an essential coactivator of nuclear receptor-mediated transcriptional activation. While structural requirements for ligand-dependent binding of classical coactivator motifs of MED1 to numerous nuclear receptor ligand-binding domains have been fully elucidated, the recognition of the full-length or truncated coactivator by full nuclear receptor complexes remain unknown. Here we present structural details of the interaction between a large part of MED1 comprising its structured N-terminal and the flexible receptor-interacting domains and the mutual heterodimer of the vitamin D receptor (VDR) and the retinoid X receptor (RXR) bound to their cognate DNA response element. Using a combination of structural and biophysical methods we show that the ligand-dependent interaction between VDR and the second coactivator motif of MED1 is crucial for complex formation and we identify additional, previously unseen, interaction details. In particular, we identified RXR regions involved in the interaction with the structured N-terminal domain of MED1, as well as VDR regions outside the classical coactivator binding cleft affected by coactivator recruitment. These findings highlight important roles of each receptor within the heterodimer in selective recognition of MED1 and contribute to our understanding of the nuclear receptor-coregulator complexes.


Subject(s)
DNA/metabolism , Mediator Complex Subunit 1 , Receptors, Calcitriol , Retinoid X Receptor alpha , Humans , Ligands , Mediator Complex Subunit 1/chemistry , Mediator Complex Subunit 1/metabolism , Protein Binding , Protein Domains , Protein Multimerization , Receptors, Calcitriol/chemistry , Receptors, Calcitriol/metabolism , Retinoid X Receptor alpha/chemistry , Retinoid X Receptor alpha/metabolism
5.
J Med Chem ; 63(17): 9457-9463, 2020 09 10.
Article in English | MEDLINE | ID: mdl-32787090

ABSTRACT

Vitamin D receptor (VDR) antagonists prevent the VDR activation function helix 12 from folding into its active conformation, thus affecting coactivator recruitment and antagonizing the transcriptional regulation induced by 1α,25-dihydroxyvitamin D3. Here, we report the crystal structure of the zebrafish VDR ligand-binding domain in complex with the ZK168281 antagonist, revealing that the ligand prevents optimal folding of the C-terminal region of VDR. This interference was confirmed by hydrogen-deuterium exchange mass spectrometry (HDX-MS) in solution.


Subject(s)
Calcitriol/analogs & derivatives , Receptors, Calcitriol/antagonists & inhibitors , Receptors, Calcitriol/metabolism , Animals , Calcitriol/metabolism , Calcitriol/pharmacology , Cell Line , Ligands , Models, Molecular , Protein Binding , Protein Domains , Rats , Receptors, Calcitriol/chemistry , Zebrafish
SELECTION OF CITATIONS
SEARCH DETAIL
...