Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 299
Filter
1.
Biochemistry ; 63(7): 926-938, 2024 Apr 02.
Article in English | MEDLINE | ID: mdl-38489495

ABSTRACT

Progesterone receptor membrane component 1 (PGRMC1) binds heme via a surface-exposed site and displays some structural resemblance to cytochrome b5 despite their different functions. In the case of PGRMC1, it is the protein interaction with drug-metabolizing cytochrome P450s and the epidermal growth factor receptor that has garnered the most attention. These interactions are thought to result in a compromised ability to metabolize common chemotherapy agents and to enhance cancer cell proliferation. X-ray crystallography and immunoprecipitation data have suggested that heme-mediated PGRMC1 dimers are important for facilitating these interactions. However, more recent studies have called into question the requirement of heme binding for PGRMC1 dimerization. Our study employs spectroscopic and computational methods to probe and define heme binding and its impact on PGRMC1 dimerization. Fluorescence, electron paramagnetic resonance and circular dichroism spectroscopies confirm heme binding to apo-PGRMC1 and were used to demonstrate the stabilizing effect of heme on the wild-type protein. We also utilized variants (C129S and Y113F) to precisely define the contributions of disulfide bonds and direct heme coordination to PGRMC1 dimerization. Understanding the key factors involved in these processes has important implications for downstream protein-protein interactions that may influence the metabolism of chemotherapeutic agents. This work opens avenues for deeper exploration into the physiological significance of the truncated-PGRMC1 model and developing design principles for potential therapeutics to target PGRMC1 dimerization and downstream interactions.


Subject(s)
Heme , Neoplasms , Receptors, Progesterone , Humans , Cell Proliferation , Heme/chemistry , Membrane Proteins/chemistry , Neoplasms/metabolism , Protein Multimerization/genetics , Receptors, Progesterone/chemistry , Receptors, Progesterone/metabolism
2.
J Biomol Struct Dyn ; 41(21): 12401-12410, 2023.
Article in English | MEDLINE | ID: mdl-36752314

ABSTRACT

Uterine fibroids (UF) are benign smooth muscle neoplasm of uterus that have a significant impact on a woman's quality of life as they perturb hormonal homeostasis resulting in heavy menstrual bleeding, impaired fertility, pregnancy complications and loss. UF can be surgically removed through invasive procedures, but their recurrence rate is often high. Progesterone receptor (PR) has an imperative role in UF management. Mifepristone, ulipristal acetate (UPA) and asoprisnil (ASO) are some selective progesterone receptor modulators (SPRMs), acts on PR, but due to their side effects in long term use, they were withdrawn from the market. Hence, there is a dire need for novel, highly efficient with least side effects, therapeutics for the treatment of UF. To contribute toward the drug discovery for UF, we made an extensive structural comparison of reported PR crystal structures, also elucidated the binding modes of four existing SPRMs against human PR through ensemble docking approach. Our studies revealed the presence of 5 highly repeating water molecules that has an important role in ligand binding and structural stability. Our ensemble docking and MD simulation revealed that studied ligands have preferential selectivity toward the specific conformation of PR. It is anticipated that our study will be a useful resource to all the drug discovery scientists who are engaged in the identification of lead molecules against UF.


Subject(s)
Leiomyoma , Receptors, Progesterone , Pregnancy , Female , Humans , Receptors, Progesterone/chemistry , Receptors, Progesterone/metabolism , Quality of Life , Leiomyoma/drug therapy
3.
Proc Natl Acad Sci U S A ; 119(31): e2200667119, 2022 08 02.
Article in English | MEDLINE | ID: mdl-35881789

ABSTRACT

Liquid-liquid phase separation (LLPS) is emerging as a key physical principle for biological organization inside living cells, forming condensates that play important regulatory roles. Inside living nuclei, transcription factor (TF) condensates regulate transcriptional initiation and amplify the transcriptional output of expressed genes. However, the biophysical parameters controlling TF condensation are still poorly understood. Here we applied a battery of single-molecule imaging, theory, and simulations to investigate the physical properties of TF condensates of the progesterone receptor (PR) in living cells. Analysis of individual PR trajectories at different ligand concentrations showed marked signatures of a ligand-tunable LLPS process. Using a machine learning architecture, we found that receptor diffusion within condensates follows fractional Brownian motion resulting from viscoelastic interactions with chromatin. Interestingly, condensate growth dynamics at shorter times is dominated by Brownian motion coalescence (BMC), followed by a growth plateau at longer timescales that result in nanoscale condensate sizes. To rationalize these observations, we extended on the BMC model by including the stochastic unbinding of particles within condensates. Our model reproduced the BMC behavior together with finite condensate sizes at the steady state, fully recapitulating our experimental data. Overall, our results are consistent with condensate growth dynamics being regulated by the escaping probability of PR molecules from condensates. The interplay between condensation assembly and molecular escaping maintains an optimum physical condensate size. Such phenomena must have implications for the biophysical regulation of other nuclear condensates and could also operate in multiple biological scenarios.


Subject(s)
Biomolecular Condensates , Cell Nucleus , Receptors, Progesterone , Single Molecule Imaging , Transcription Factors , Biomolecular Condensates/chemistry , Cell Nucleus/chemistry , Chromatin/chemistry , Ligands , Machine Learning , Motion , Receptors, Progesterone/chemistry , Transcription Factors/chemistry
4.
Biochem Biophys Res Commun ; 592: 1-6, 2022 02 12.
Article in English | MEDLINE | ID: mdl-35007844

ABSTRACT

Currently, semiconductor nanoparticles known as quantum dots (QDs) have attracted interest in various application fields such as those requiring sensing properties, binding assays, and cellular imaging and are the very important in the acceleration of drug discovery due to their unique photophysical properties. Here, we applied graphene quantum dots (GQDs) for the binding assay of membrane progesterone receptor alpha (mPRα), one of the probable membrane receptors that have potential in drug discovery applications. By coupling the amino groups of mPRα with GQDs, we prepared fluorogenic GQD-conjugated mPRα (GQD-mPRα). When mixed with a progesterone-BSA-fluorescein isothiocyanate conjugate (P4-BSA-FITC) to check the ligand receptor binding activity of GQD-mPRα, fluorescence at 520 nm appeared. The fluorescence at 520 nm was reduced by the addition of free progesterone into the reaction mixture. GQD-coupled BSA (GQD-BSA) did not show a reduction in fluorescence at 520 nm. The results demonstrated the formation of a complex of GQD-mPRα and P4-BSA-FITC with ligand receptor binding. We established a ligand binding assay for membrane steroid receptors that is applicable for high-throughput assays.


Subject(s)
Biological Assay/methods , Graphite/chemistry , Quantum Dots/chemistry , Receptors, Progesterone/metabolism , Steroids/metabolism , Fluorescein-5-isothiocyanate/metabolism , Fluorescence , Humans , Models, Molecular , Progesterone/metabolism , Receptors, Progesterone/chemistry , Serum Albumin, Bovine/metabolism
5.
Mol Microbiol ; 116(3): 890-908, 2021 09.
Article in English | MEDLINE | ID: mdl-34184334

ABSTRACT

The lipid mediators, platelet-activating factor (PAF) and lysophosphatidylcholine (LPC), play relevant pathophysiological roles in Trypanosoma cruzi infection. Several species of LPC, including C18:1 LPC, which mimics the effects of PAF, are synthesized by T. cruzi. The present study identified a receptor in T. cruzi, which was predicted to bind to PAF, and found it to be homologous to members of the progestin and adiponectin family of receptors (PAQRs). We constructed a three-dimensional model of the T. cruzi PAQR (TcPAQR) and performed molecular docking to predict the interactions of the TcPAQR model with C16:0 PAF and C18:1 LPC. We knocked out T. cruzi PAQR (TcPAQR) gene and confirmed the identity of the expressed protein through immunoblotting and immunofluorescence assays using an anti-human PAQR antibody. Wild-type and knockout (KO) parasites were also used to investigate the in vitro cell differentiation and interactions with peritoneal mouse macrophages; TcPAQR KO parasites were unable to react to C16:0 PAF or C18:1 LPC. Our data are highly suggestive that PAF and LPC act through TcPAQR in T. cruzi, triggering its cellular differentiation and ability to infect macrophages.


Subject(s)
Lysophosphatidylcholines/metabolism , Platelet Activating Factor/metabolism , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Trypanosoma cruzi/genetics , Trypanosoma cruzi/metabolism , Amino Acid Sequence , Animals , Cell Differentiation , Chagas Disease/parasitology , Gene Knockout Techniques/methods , Host-Parasite Interactions , Humans , Lysophosphatidylcholines/chemistry , Macrophages , Mice , Molecular Docking Simulation , Phylogeny , Platelet Activating Factor/chemistry , Protein Conformation , Protozoan Proteins/chemistry , Receptors, Adiponectin/chemistry , Receptors, Adiponectin/genetics , Receptors, Adiponectin/metabolism , Receptors, Progesterone/chemistry , Receptors, Progesterone/genetics , Receptors, Progesterone/metabolism , Trypanosoma cruzi/chemistry
6.
Sci Data ; 8(1): 100, 2021 04 12.
Article in English | MEDLINE | ID: mdl-33846359

ABSTRACT

Progesterone receptor (PR) isoforms, PRA and PRB, act in a progesterone-independent and dependent manner to differentially modulate the biology of breast cancer cells. Here we show that the differences in PRA and PRB structure facilitate the binding of common and distinct protein interacting partners affecting the downstream signaling events of each PR-isoform. Tet-inducible HA-tagged PRA or HA-tagged PRB constructs were expressed in T47DC42 (PR/ER negative) breast cancer cells. Affinity purification coupled with stable isotope labeling of amino acids in cell culture (SILAC) mass spectrometry technique was performed to comprehensively study PRA and PRB interacting partners in both unliganded and liganded conditions. To validate our findings, we applied both forward and reverse SILAC conditions to effectively minimize experimental errors. These datasets will be useful in investigating PRA- and PRB-specific molecular mechanisms and as a database for subsequent experiments to identify novel PRA and PRB interacting proteins that differentially mediated different biological functions in breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Receptors, Progesterone/metabolism , Amino Acids/chemistry , Cell Line, Tumor , Female , Humans , Isotope Labeling/methods , Mass Spectrometry/methods , Receptors, Progesterone/chemistry , Two-Hybrid System Techniques
7.
Eur J Hum Genet ; 29(8): 1206-1215, 2021 08.
Article in English | MEDLINE | ID: mdl-33867527

ABSTRACT

Inherited paediatric cataract is a rare Mendelian disease that results in visual impairment or blindness due to a clouding of the eye's crystalline lens. Here we report an Australian family with isolated paediatric cataract, which we had previously mapped to Xq24. Linkage at Xq24-25 (LOD = 2.53) was confirmed, and the region refined with a denser marker map. In addition, two autosomal regions with suggestive evidence of linkage were observed. A segregating 127 kb deletion (chrX:g.118373226_118500408del) in the Xq24-25 linkage region was identified from whole-genome sequencing data. This deletion completely removed a commonly deleted long non-coding RNA gene LOC101928336 and truncated the protein coding progesterone receptor membrane component 1 (PGRMC1) gene following exon 1. A literature search revealed a report of two unrelated males with non-syndromic intellectual disability, as well as congenital cataract, who had contiguous gene deletions that accounted for their intellectual disability but also disrupted the PGRMC1 gene. A morpholino-induced pgrmc1 knockdown in a zebrafish model produced significant cataract formation, supporting a role for PGRMC1 in lens development and cataract formation. We hypothesise that the loss of PGRMC1 causes cataract through disrupted PGRMC1-CYP51A1 protein-protein interactions and altered cholesterol biosynthesis. The cause of paediatric cataract in this family is the truncating deletion of PGRMC1, which we report as a novel cataract gene.


Subject(s)
Cataract/genetics , Membrane Proteins/genetics , Receptors, Progesterone/genetics , Animals , Cataract/metabolism , Cataract/pathology , Child , Gene Deletion , Humans , Male , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Pedigree , Protein Binding , Receptors, Progesterone/chemistry , Receptors, Progesterone/metabolism , Sterol 14-Demethylase/metabolism , Zebrafish
8.
J Med Chem ; 64(9): 5226-5251, 2021 05 13.
Article in English | MEDLINE | ID: mdl-33905258

ABSTRACT

Positron emission tomography (PET) is useful for noninvasive in vivo visualization of disease-related receptors, for evaluation of receptor occupancy to determine an appropriate drug dosage, and for proof-of-concept of drug candidates in translational research. For these purposes, the specificity of the PET tracer for the target receptor is critical. Here, we review work in this area, focusing on the chemical structures of reported PET tracers, their Ki/Kd values, and the physical properties relevant to target receptor selectivity. Among these physical properties, such as cLogP, cLogD, molecular weight, topological polar surface area, number of hydrogen bond donors, and pKa, we focus especially on LogD and LogP as important physical properties that can be easily compared across a range of studies. We discuss the success of PET tracers in evaluating receptor occupancy and consider likely future developments in the field.


Subject(s)
Positron-Emission Tomography/methods , Radiopharmaceuticals/metabolism , Receptors, Cell Surface/metabolism , Humans , Hydrophobic and Hydrophilic Interactions , Neoplasms/diagnostic imaging , Protein Binding , Radiopharmaceuticals/chemistry , Receptors, Cannabinoid/chemistry , Receptors, Cannabinoid/metabolism , Receptors, Cell Surface/chemistry , Receptors, Cholinergic/chemistry , Receptors, Cholinergic/metabolism , Receptors, Progesterone/chemistry , Receptors, Progesterone/metabolism
9.
Article in English | MEDLINE | ID: mdl-33276393

ABSTRACT

Antigestagens (antiprogestins) are functional competitors of progesterone (P4) that prevent P4 from mediating its biological functions either by suppressing its production or blocking its function. Among the latter are progesterone antagonists, competitors of P4 binding to its nuclear receptor PGR, which have found application in both human and veterinary medicine, in particular in small animal practice for the prevention of nidation and the interruption of pregnancy. Depending on their mode of action, progesterone receptor antagonists can be divided into 2 classes. Class I antagonists bind to the PGR but fail to induce its binding to promoters of target genes (competitive inhibitors). Class II antigestagens, including aglepristone used in veterinary medicine, bind to the PGR, activate its association with a promoter, but interfere with the downstream signalling cascades, e. g., by recruiting transcriptional repressors. They act thereby as transdominant repressors exerting negative effects on target gene expression. Importantly for experimental sciences, as active antagonists, class II antagonists do not require the presence of the natural ligand for their action. Besides their clinical application, antigestagens are used in research for investigating P4-dependent physiological and pathological processes. Here an overview of the history and the current usage of progesterone receptor antagonists in veterinary medicine and research is presented.


Subject(s)
Hormone Antagonists , Receptors, Progesterone , Veterinary Drugs , Animals , Biomedical Research , Dogs , Rabbits , Receptors, Progesterone/antagonists & inhibitors , Receptors, Progesterone/chemistry , Receptors, Progesterone/metabolism , Ruminants
10.
Mol Phylogenet Evol ; 148: 106814, 2020 07.
Article in English | MEDLINE | ID: mdl-32278076

ABSTRACT

The membrane-associated progesterone receptor (MAPR) family consists of heme-binding proteins containing a cytochrome b5 (cytb5) domain characterized by the presence of a MAPR-specific interhelical insert region (MIHIR) between helices 3 and 4 of the canonical cytb5-domain fold. Animals possess three MAPR genes (PGRMC-like, Neuferricin and Neudesin). Here we show that all three animal MAPR genes were already present in the common ancestor of the opisthokonts (comprising animals and fungi as well as related single-celled taxa). All three MAPR genes acquired extensions C-terminal to the cytb5 domain, either before or with the evolution of animals. The archetypical MAPR protein, progesterone receptor membrane component 1 (PGRMC1), contains phosphorylated tyrosines Y139 and Y180. The combination of Y139/Y180 appeared in the common ancestor of cnidarians and bilaterians, along with an early embryological organizer and synapsed neurons, and is strongly conserved in all bilaterian animals. A predicted protein interaction motif in the PGRMC1 MIHIR is potentially regulated by Y139 phosphorylation. A multilayered model of animal MAPR function acquisition includes some pre-metazoan functions (e.g., heme binding and cytochrome P450 interactions) and some acquired animal-specific functions that involve regulation of strongly conserved protein interaction motifs acquired by animals (Metazoa). This study provides a conceptual framework for future studies, against which especially PGRMC1's multiple functions can perhaps be stratified and functionally dissected.


Subject(s)
Eukaryota/metabolism , Membrane Proteins/metabolism , Receptors, Progesterone/metabolism , Amino Acid Sequence , Animals , Evolution, Molecular , Membrane Proteins/chemistry , Phylogeny , Protein Binding , Protein Domains , Receptors, Progesterone/chemistry , Receptors, Progesterone/genetics
11.
Biochem J ; 477(2): 477-489, 2020 01 31.
Article in English | MEDLINE | ID: mdl-31904842

ABSTRACT

Protein quality control is crucial for maintaining cellular homeostasis and its dysfunction is closely linked to human diseases. The post-translational protein quality control machinery mainly composed of BCL-2-associated athanogene 6 (BAG6) is responsible for triage of mislocalized membrane proteins (MLPs). However, it is unknown how the BAG6-mediated degradation of MLPs is regulated. We report here that PAQR9, a member of the Progesterone and AdipoQ receptor (PAQR) family, is able to modulate BAG6-mediated triage of MLPs. Analysis with mass spectrometry identified that BAG6 is one of the major proteins interacting with PAQR9 and such interaction is confirmed by co-immunoprecipitation and co-localization assays. The protein degradation rate of representative MLPs is accelerated by PAQR9 knockdown. Consistently, the polyubiquitination of MLPs is enhanced by PAQR9 knockdown. PAQR9 binds to the DUF3538 domain within the proline-rich stretch of BAG6. PAQR9 reduces the binding of MLPs to BAG6 in a DUF3538 domain-dependent manner. Taken together, our results indicate that PAQR9 plays a role in the regulation of protein quality control of MLPs via affecting the interaction of BAG6 with membrane proteins.


Subject(s)
Homeostasis/genetics , Membrane Proteins/genetics , Molecular Chaperones/genetics , Receptors, Progesterone/genetics , Humans , Membrane Proteins/chemistry , Molecular Chaperones/chemistry , Protein Binding/genetics , Protein Domains/genetics , Protein Transport/genetics , Proto-Oncogene Proteins c-bcl-2/chemistry , Proto-Oncogene Proteins c-bcl-2/genetics , Receptors, Progesterone/chemistry , Ubiquitination/genetics , Ubiquitins/chemistry , Ubiquitins/genetics
12.
J Mol Endocrinol ; 65(1): T1-T14, 2020 07.
Article in English | MEDLINE | ID: mdl-31809260

ABSTRACT

Progesterone's ability to maintain pregnancy in eutherian mammals highlighted this steroid as the 'hormone of pregnancy'. It was the unique 'pro-gestational' bioactivity of progesterone that enabled eventual purification of this ovarian steroid to crystalline form by Willard Myron Allen in the early 1930s. While a functional connection between normal progesterone responses ('progestational proliferation') of the uterus with the maintenance of pregnancy was quickly appreciated, an understanding of progesterone's involvement in the early stages of pregnancy establishment was comparatively less well understood. With the aforementioned as historical backdrop, this review focuses on a selection of key advances in our understanding of the molecular mechanisms by which progesterone, through its nuclear receptor (the progesterone receptor), drives the development of endometrial receptivity, a transient uterine state that allows for embryo implantation and the establishment of pregnancy. Highlighted in this review are the significant contributions of advanced mouse engineering and genome-wide transcriptomic and cistromic analytics which reveal the pivotal molecular mediators and modifiers that are essential to progesterone-dependent endometrial receptivity and decidualization. With a clearer understanding of the molecular landscape that underpins uterine responsiveness to progesterone during the periimplantation period, we predict that common gynecologic morbidities due to abnormal progesterone responsiveness will be more effectively diagnosed and/or treated in the future.


Subject(s)
Embryo Implantation , Endometrium/metabolism , Progesterone/metabolism , Receptors, Progesterone/metabolism , Signal Transduction , Animals , Female , Humans , Models, Biological , Receptors, Progesterone/chemistry
13.
Sci Rep ; 9(1): 17279, 2019 11 21.
Article in English | MEDLINE | ID: mdl-31754172

ABSTRACT

Uterine fibroids (UFs) are associated with irregular or excessive uterine bleeding, pelvic pain or pressure, or infertility. Ovarian steroid hormones support the growth and maintenance of UFs. Ulipristal acetate (UPA) a selective progesterone receptor (PR) modulator (SPRM) reduce the size of UFs, inhibit ovulation and lead to amenorrhea. Recent liver toxicity concerns with UPA, diminished enthusiasm for its use and reinstate the critical need for a safe, efficacious SPRM to treat UFs. In the current study, we evaluated the efficacy of new SPRM, EC313, for the treatment for UFs using a NOD-SCID mouse model. EC313 treatment resulted in a dose-dependent reduction in the fibroid xenograft weight (p < 0.01). Estradiol (E2) induced proliferation was blocked significantly in EC313-treated xenograft fibroids (p < 0.0001). Uterine weight was reduced by EC313 treatment compared to UPA treatment. ER and PR were reduced in EC313-treated groups compared to controls (p < 0.001) and UPA treatments (p < 0.01). UF specific desmin and collagen were markedly reduced with EC313 treatment. The partial PR agonism and no signs of unopposed estrogenicity makes EC313 a candidate for the long-term treatment for UFs. Docking studies have provided a structure based explanation for the SPRM activity of EC313.


Subject(s)
Cell Proliferation/drug effects , Contraceptive Agents, Female/administration & dosage , Leiomyoma/drug therapy , Progesterone Congeners/administration & dosage , Receptors, Progesterone/agonists , Uterine Neoplasms/drug therapy , Animals , Contraceptive Agents, Female/adverse effects , Contraceptive Agents, Female/chemistry , Estrenes/administration & dosage , Estrenes/adverse effects , Female , Humans , Leiomyoma/pathology , Mice , Molecular Docking Simulation , Molecular Structure , Norpregnadienes/administration & dosage , Norpregnadienes/adverse effects , Oximes/administration & dosage , Oximes/adverse effects , Progesterone Congeners/adverse effects , Progesterone Congeners/chemistry , Receptors, Progesterone/chemistry , Receptors, Progesterone/metabolism , Structure-Activity Relationship , Uterine Neoplasms/pathology , Uterus/drug effects , Uterus/pathology , Xenograft Model Antitumor Assays
14.
Bioconjug Chem ; 30(11): 2947-2957, 2019 11 20.
Article in English | MEDLINE | ID: mdl-31589412

ABSTRACT

ProGlo is an efficient steroid receptor-targeted magnetic resonance (MR) imaging contrast agent (CA). It has been shown to bind to the progesterone receptor (PR) and produce enhanced image contrast in PR-positive cells and tissues in vitro and in vivo. However, the hydrophobicity of the steroid targeting domain of ProGlo (logP = 1.4) limits its formulation and delivery at clinically relevant doses. In this work, a hydrophobic moiety was utilized to drive efficient adsorption onto nanodiamond (ND) clusters to form a water-soluble nanoconstruct (logP = -2.4) with 80% release in 8 h under biological conditions. In cell culture, the ND-ProGlo construct delivered increased concentrations of ProGlo to target cells compared to ProGlo alone. Importantly, these results were accomplished without the use of solvents such as DMSO, providing a significant advance toward formulating ProGlo for translational applications. Biodistribution studies confirm the delivery of ProGlo to PR(+) tissues with enhanced efficacy over untargeted controls. These results demonstrate the potential for a noncovalent ND-CA construct as a general strategy for solubilizing and delivering hydrophobic targeted MR CAs.


Subject(s)
Breast Neoplasms/pathology , Contrast Media/pharmacokinetics , Magnetic Resonance Imaging/methods , Nanoconjugates/chemistry , Nanodiamonds/administration & dosage , Receptors, Progesterone/metabolism , Animals , Breast Neoplasms/metabolism , Contrast Media/chemistry , Female , Humans , Mice , Nanodiamonds/chemistry , Receptors, Progesterone/chemistry , Solubility , Tissue Distribution , Tumor Cells, Cultured
15.
Steroids ; 152: 108503, 2019 12.
Article in English | MEDLINE | ID: mdl-31562879

ABSTRACT

The ovarian steroid hormone progesterone and its nuclear receptor, the Progesterone Receptor (PR), play an essential role in the regulation of cell proliferation and differentiation in the mammary gland. In addition, experimental and clinical evidence demonstrate their critical role in controlling mammary gland tumorigenesis and breast cancer development. When bound to its ligand, the main action of PR is as a transcription factor, which regulates the expression of target genes networks. PR also activates signal transduction pathways through a rapid or non-genomic mechanism in breast cancer cells, an event that is fully integrated with its genomic effects. This review summarizes the molecular mechanisms of the ligand-activated PR actions that drive epithelial cell proliferation and the regulation of the stem cell population in the normal breast and in breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Neoplastic Stem Cells/metabolism , Receptors, Progesterone/metabolism , Animals , Breast Neoplasms/pathology , Cell Proliferation , Female , Humans , Neoplastic Stem Cells/pathology , Receptors, Progesterone/chemistry
16.
Mol Cell Endocrinol ; 493: 110471, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31163202

ABSTRACT

The purpose of this review is to highlight recent developments in small molecules and peptides that block the binding of coactivators to steroid receptors. These coactivator binding inhibitors bind at the coregulator binding groove, also known as Activation Function-2, rather than at the ligand-binding site of steroid receptors. Steroid receptors that have been targeted with coactivator binding inhibitors include the androgen receptor, estrogen receptor and progesterone receptor. Coactivator binding inhibitors may be useful in some cases of resistance to currently prescribed therapeutics. The scope of the review includes small-molecule and peptide coactivator binding inhibitors for steroid receptors, with a particular focus on recent compounds that have been assayed in cell-based models.


Subject(s)
Peptides/pharmacology , Receptors, Steroid/metabolism , Small Molecule Libraries/pharmacology , Binding Sites/drug effects , Humans , Models, Molecular , Peptides/chemistry , Protein Binding/drug effects , Protein Conformation , Receptors, Androgen/chemistry , Receptors, Androgen/metabolism , Receptors, Estrogen/chemistry , Receptors, Estrogen/metabolism , Receptors, Progesterone/chemistry , Receptors, Progesterone/metabolism , Receptors, Steroid/chemistry , Small Molecule Libraries/chemistry
17.
Endocr Relat Cancer ; 26(8): R479-R497, 2019 08.
Article in English | MEDLINE | ID: mdl-31252411

ABSTRACT

Nuclear receptors are transcription factors that play critical roles in development, homeostasis and metabolism in all multicellular organisms. An important family of nuclear receptors comprises those members that respond to steroid hormones, and which is subdivided in turn into estrogen receptor (ER) isoforms α and ß (NR3A1 and A2, respectively), and a second subfamily of so-called oxosteroid receptors. The latter includes the androgen receptor (AR/NR3C4), the glucocorticoid receptor (GR/NR3C1), the mineralocorticoid receptor (MR/NR3C2) and the progesterone receptor (PR/NR3C3). Here we review recent advances in our understanding of the structure-and-function relationship of steroid nuclear receptors and discuss their implications for the etiology of human diseases. We focus in particular on the role played by AR dysregulation in both prostate cancer (PCa) and androgen insensitivity syndromes (AIS), but also discuss conditions linked to mutations of the GR gene as well as those in a non-steroidal receptor, the thyroid hormone receptor (TR). Finally, we explore how these recent results might be exploited for the development of novel and selective therapeutic strategies.


Subject(s)
Androgen-Insensitivity Syndrome/metabolism , Prostatic Neoplasms/metabolism , Receptors, Androgen/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Androgen-Insensitivity Syndrome/etiology , Androgen-Insensitivity Syndrome/pathology , Humans , Male , Prostatic Neoplasms/etiology , Prostatic Neoplasms/pathology , Protein Multimerization , Receptors, Androgen/chemistry , Receptors, Androgen/genetics , Receptors, Cytoplasmic and Nuclear/chemistry , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Glucocorticoid/chemistry , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Receptors, Mineralocorticoid/chemistry , Receptors, Mineralocorticoid/genetics , Receptors, Mineralocorticoid/metabolism , Receptors, Progesterone/chemistry , Receptors, Progesterone/genetics , Receptors, Progesterone/metabolism , Receptors, Thyroid Hormone/chemistry , Receptors, Thyroid Hormone/genetics , Receptors, Thyroid Hormone/metabolism , Steroids/metabolism
18.
Biol Reprod ; 100(6): 1571-1580, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30877763

ABSTRACT

Progesterone receptor membrane component 1 (PGRMC1) interacts with PGRMC2, and disrupting this interaction in spontaneously immortalized granulosa cells (SIGCS) leads to an inappropriate entry into the cell cycle, mitotic arrest, and ultimately cell death. The present study revealed that PGRMC1 and PGRMC2 localize to the cytoplasm of murine granulosa cells of nonatretric follicles with their staining intensity being somewhat diminished in granulosa cells of atretic follicles. Compared to controls (Pgrmc1fl/fl), the rate at which granulosa cells entered the cell cycle increased in nonatretic and atretic follicles of mice in which Pgrmc1 was conditionally deleted (Pgrmc1d/d) from granulosa cells. This increased rate of entry into the cell cycle was associated with a ≥ 2-fold increase in follicular atresia and the nuclear localization of nuclear factor-kappa-B transcription factor P65; (NFΚB/p65, or RELA). GTPase activating protein binding protein 2 (G3BP2) binds NFΚB/p65 through an interaction with NFΚB inhibitor alpha (IκBα), thereby maintaining NFΚB/p65's cytoplasmic localization and restricting its transcriptional activity. Since PGRMC1 and PGRMC2 bind G3BP2, studies were designed to assess the functional relationship between PGRMC1, PGRMC2, and NFΚB/p65 in SIGCs. In these studies, disrupting the interaction between PGRMC1 and PGRMC2 increased the nuclear localization of NFΚB/p65, and depleting PGRMC1, PGRMC2, or G3BP2 increased NFΚB transcriptional activity and the progression into the cell cycle. Taken together, these studies suggest that PGRMC1 and 2 regulate granulosa cell cycle entry in follicles by precisely controlling the localization and thereby the transcriptional activity of NFΚB/p65.


Subject(s)
Cell Membrane/physiology , Granulosa Cells/physiology , Membrane Proteins/metabolism , Mitosis/physiology , NF-kappa B/metabolism , Receptors, Progesterone/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Female , I-kappa B Kinase/genetics , I-kappa B Kinase/metabolism , Membrane Proteins/chemistry , Membrane Proteins/genetics , Mice , Mice, Knockout , NF-kappa B/genetics , Ovarian Follicle/physiology , Protein Subunits , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Receptors, Progesterone/chemistry , Receptors, Progesterone/genetics
19.
Biochim Biophys Acta Gene Regul Mech ; 1862(4): 522-533, 2019 04.
Article in English | MEDLINE | ID: mdl-30716532

ABSTRACT

The activation functions AF1 and AF2 of nuclear receptors mediate the recruitment of coregulators in gene regulation. AF1 is mapped to the highly variable and intrinsically unstructured N terminal domain and AF2 lies in the conserved ligand binding domain. The unstructured nature of AF1 offers structural plasticity and hence functional versatility in gene regulation. However, little is known about the key functional residues of AF1 that mediates its interaction with coregulators. This study focuses on the progesterone receptor (PR) and reports the identification of K464, K481 and R492 (KKR) as the key functional residues of PR AF1. The KKR are monomethylated and function cooperatively. The combined mutations of KKR to QQQ render PR isoform B (PRB) hyperactive, whereas KKR to FFF mutations abolishes as much as 80% of PR activity. Furthermore, the hyperactive QQQ mutation rescues the loss of PR activity due to E911A mutation in AF2. The study also finds that the magnitudes of the mutational effect differ in different cell types as a result of differential effects on the functional interaction with coregulators. Furthermore, KKR provides the interface for AF1 to physically interact with p300 and SRC-1, and with AF2 at E911. Intriguingly, the inactive FFF mutant interacts strikingly stronger with both SRC-1 and AF2 than wt PRB. We propose a tripartite model to describe the dynamic interactions between AF1, AF2 and SRC-1 with KKR of AF1 and E911 of AF2 as the interface. An overly stable interaction would hamper the dynamics of disassembly of the receptor complex.


Subject(s)
Amino Acids/chemistry , Receptors, Progesterone/chemistry , Receptors, Progesterone/metabolism , Cell Line, Tumor , Humans , Ligands , Methylation , Mutation , NF-kappa B/metabolism , Nuclear Receptor Coactivator 1/metabolism , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Receptors, Progesterone/genetics , Transcription Factor AP-1/metabolism , Transcription, Genetic , p300-CBP Transcription Factors/metabolism
20.
Sci Rep ; 9(1): 653, 2019 01 24.
Article in English | MEDLINE | ID: mdl-30679694

ABSTRACT

Progesterone receptor membrane component1 (PGRMC1) is a heme-binding protein involved in cancers and Alzheimer's disease. PGRMC1 consists of a short N-terminal extracellular or luminal domain, a single membrane-spanning domain, and a long cytoplasmic domain. Previously, we generated two monoclonal antibodies (MAbs) 108-B6 and 4A68 that recognize cell surface-expressed PGRMC1 (csPGRMC1) on human pluripotent stem cells and some cancer cells. In this study, flow cytometric analysis found that an anti-PGRMC1 antibody recognizing the N-terminus of PGRMC1 could not bind to csPGRMC1 on cancer cells, and 108-B6 and 4A68 binding to csPGRMC1 was inhibited by trypsin treatment, suggesting that the epitopes of 108-B6 and 4A68 are trypsin-sensitive. To examine the epitope specificity of 108-B6 and 4A68, glutathione-S-transferase (GST)-fused PGRMC1 mutants were screened to identify the epitopes targeted by the antibodies. The result showed that 108-B6 and 4A68 recognized C-terminal residues 183-195 and 171-182, respectively, of PGRMC1, where trypsin-sensitive sites are located. A polyclonal anti-PGRMC1 antibody raised against the C-terminus of PGRMC1 could also recognized csPGRMC1 in a trypsin-sensitive manner, suggesting that the C-terminus of csPGRMC1 is exposed on the cell surface. This finding reveals that csPGRMC1 has a non-conventional plasma membrane topology, which is different from that of intracellular PGRMC1.


Subject(s)
Antibodies, Monoclonal/immunology , Cell Membrane/metabolism , Epitope Mapping , Membrane Proteins/immunology , Membrane Proteins/metabolism , Receptors, Progesterone/immunology , Receptors, Progesterone/metabolism , Cell Line , Humans , Intracellular Space/metabolism , Membrane Proteins/chemistry , Receptors, Progesterone/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...