Please select what you would like included for printing:
Copy the text below and then paste that into your favorite email application
This site is protected by reCAPTCHA and the Google Privacy Policy and Terms of Service apply
Service map data © OpenStreetMap contributors
Metrics details
Nuclear receptor (NR) transcription factors use a conserved activation function-2 (AF-2) helix 12 mechanism for agonist-induced coactivator interaction and NR transcriptional activation
ligand-induced corepressor-dependent NR repression appears to occur through structurally diverse mechanisms
We report two crystal structures of peroxisome proliferator-activated receptor gamma (PPARγ) in an inverse agonist/corepressor-bound transcriptionally repressive conformation
Helix 12 is displaced from the solvent-exposed active conformation and occupies the orthosteric ligand-binding pocket enabled by a conformational change that doubles the pocket volume
Paramagnetic relaxation enhancement (PRE) NMR and chemical crosslinking mass spectrometry confirm the repressive helix 12 conformation
PRE NMR also defines the mechanism of action of the corepressor-selective inverse agonist T0070907
and reveals that apo-helix 12 exchanges between transcriptionally active and repressive conformations—supporting a fundamental hypothesis in the NR field that helix 12 exchanges between transcriptionally active and repressive conformations
these structures—some of which are stabilized by crystal contacts between symmetry-related molecules within the crystal lattice
or by mutations or coregulator peptides that reduce helix 12 dynamics to facilitate crystal formation for structure determination—are not fully representative of the dynamic NR LBD conformational ensemble
Here we report two crystal structures of PPARγ bound to T0070907 and corepressor peptides that reveal a unique a transcriptionally repressive conformation compared with all other reported structures of corepressor-bound NRs
A comparative structural analysis to the transcriptionally active conformation and the apo-PPARγ conformational ensemble using solution paramagnetic relaxation enhancement (PRE) NMR validates the unique repressive conformation and provides evidence that helix 12 in an apo-NR exchanges between transcriptionally active and repressive conformations
a Fluorescence polarization coregulator interaction assay shows a robust interaction between PPARγ LBD with corepressor ID2 peptides from NCoR and SMRT as well as a TRAP220 coactivator peptide (n = 6)
b Mammalian two-hybrid assay in HEK293T cells measuring the effect of T0070907 (10 μM) on the interaction between the Gal4-PPARγ LBD and the VP16-NCoR RID (n = 6; mean ± s.d.)
c Luciferase transcriptional reporter assay measuring the ligand-dependent change in PPARγ transcription (n = 4; mean ± s.d.)
d TR-FRET biochemical assay measuring the ligand-dependent change in the interaction of PPARγ LBD with SMRT ID2
and TRAP220 ID2 peptides (n = 3; mean ± s.e.m.)
Source data are provided as a Source Data file
We therefore posited that T0070907 could be used to facilitate crystallization of the PPARγ LBD bound to peptides derived from NCoR and SMRT ID2 motifs
a, b Helical structural elements that show notable conformational changes in the crystal structures of a PPARγ LBD bound to rosiglitazone and TRAP220 ID2 peptide (PDB 6ONJ) in the active conformation and b PPARγ LBD bound to T0070907 and NCoR ID2 peptide (PDB 6ONI) in the repressive conformation
c Structural overlay of the active and repressive conformation PPARγ LBD crystal structures with arrows depicting the movement of the helical structural elements
Ligand-binding pocket volumes (red surfaces) were calculated and displayed using the program CASTp. Ligands were removed from the structures for all calculations. Helix 12 was removed from the repressive conformation structure (PDB 6ONI) to assess the relative pocket volume to the active conformation structures. Calculations for the active conformation structure (PDB 6ONJ)
which displays density for the Ω-loop region
were performed with and without the Ω-loop region for comparison to the repressive conformation structure
d Cell-based luciferase reporter assay in HEK293T cells treated with DMSO control or 5 μM T0070907 measuring the ligand-dependent change in wild-type or mutant PPARγ transcription; data normalized to each WT or mutant construct vehicle control (DMSO) condition (n = 4; mean ± s.e.m.)
These corepressor interaction interface structure-guided mutants support the PPARγ crystal structures whereby many interactions contribute to binding corepressor peptide
whereas the coactivator interaction is predominately mediated by the “charge clamp” residues
h Mammalian two-hybrid assay measuring the effect of T0070907 (10 μM) on the interaction between the Gal4-PPARγ LBD and the VP16-NCoR RID (n = 6; mean ± s.e.m.)
i Luciferase transcriptional reporter assay in HEK293T cells treated with DMSO control or 5 μM T0070907 measuring the ligand-dependent change in wild-type or mutant PPARγ transcription; data normalized to each WT or mutant construct vehicle control (DMSO) condition (n = 4–12; mean ± s.e.m.)
and +GAP extension all show no increase in NCoR ID2 affinity in the presence of T0070907
and many of the others show diminished corepressor-selective responsiveness to T0070907
mutant significantly weakens NCoR ID2 peptide affinity in both the apo- and T0070907-bound forms
The M364 thiol interacts with both the T0070907 nitro group (3.3 and 4.3 Å) and the C-terminal carboxylate of Y477 (3.5 Å)
the latter of which may be important for stabilizing the repressive helix 12 conformation in the orthosteric pocket
There is generally good agreement between the biochemical and cellular NCoR interaction profiles with the ability of T0070907 to repress the transcription of the mutant variants
the mutants that show relatively no T0070907-dependent change in NCoR ID2 peptide binding affinity either do not show T0070907-dependent transcriptional repression or in many cases show activation in the presence of T0070907
mutants that show a T0070907-dependent increase in NCoR ID2 peptide binding affinity generally show T0070907-dependent transcriptional repression
This suggests that the repressive helix 12 conformation within the orthosteric pocket may be the primary contribution to the T0070907-dependent increase in corepressor binding affinity and repression of PPARγ transcription
is not shown as this conformation is not consistent with the PRE NMR data) conformation PPARγ LBD crystal structures
but also indicate the apo-helix 12 exchanges between conformations similar to transcriptionally repressive (T0070907 and NCoR ID2 peptide bound) and active (rosiglitazone and TRAP220 ID2 peptide bound) states
a Distances between K474 on helix 12 and several lysine residues structurally proximal to helix 12 (K265 and K275) or further away (K301 and K457) in the crystal structure of PPARγ LBD bound to T0070907 and NCoR ID2 peptide (PDB 6ONI)
b Relative peak area of four K474-crosslinked peptides normalized to a control uncrosslinked peptide and to the highest mean peak area within each condition (mean ± s.e.m.; n = 3)
which likely inhibits the K301-K474 crosslink; K301 is part of the charge clamp involved in binding coactivator and corepressor-peptide motifs
active conformation K474-K457 (helix 12-helix 11) crosslinks were dimensioned in the NCoR ID2 peptide-bound form and further decreased in the presence of T0070907
These XL-MS data support the repressive helix 12 conformation within the orthosteric pocket that we captured in the crystal structures of T0070907-bound PPARγ LBD bound to corepressor peptides and validated with PRE NMR
a Structural overlay highlighting the T0070907 binding pose in the crystal structure of PPARγ LBD bound to T0070907 and NCoR ID2 peptide (orange; PDB 6ONI) compared with the crystal structure of T0070907-bound PPARγ LBD without a coregulator peptide (blue; PDB 6C1I)
c 2D [1H,15N]-TROSY-HSQC spectra from the PRE analysis of MTSL-labeled T0070907-bound [K474C]-PPARγ LBD without coregulator peptide
Purple boxes in (b) highlight residues shown the zoomed-in snapshots (c) showing that only one of two slowly exchanging populations corresponding to the active (labeled “a”) and repressive (labeled “r”) is affected by the MTSL spin label on helix 12
These PRE NMR findings are consistent with helix 12 occupying both the repressive conformation (e.g.
and G346 in the β-sheet are affected) and the active conformation (e.g.
one of the two peaks for G399 in the AF-2 surface is affected) when of PPARγ is bound to T0070907
solvent exposed) and repressive helix 12 conformation (i.e.
within the orthosteric pocket) may influence corepressor selectivity
the rate of exchange between active and repressive helix 12 conformations is relatively fast (intermediate NMR time scale)
Binding of T0070907 slows the rate of exchange considerably
allowing population of a long-lived helix 12 conformation within the orthosteric ligand-binding pocket that affords high corepressor binding affinity and transcriptional repression
GW9662-bound PPARγ likely populates a shorter-lived repressive helix 12 population within the orthosteric pocket that exchanges with an active-like conformation more quickly relative to T0070907-bound PPARγ but slower than apo-PPARγ
resulting in an intermediate NCoR ID2 affinity between T0070907-bound PPARγ and apo-PPARγ
In our corepressor-peptide-bound structures of PPARγ
the corepressor-selective inverse agonist T0070907 adopts a binding pose near the AF-2 surface that leaves a large portion of the orthosteric pocket open—double the pocket volume of the coactivator-bound active conformation—which
along with a kink in helix 3 relative to the active conformation
enables helix 12 to bind deep within the pocket near the β-sheet in a repressive conformation
This helix 12 conformation is unique compared with all other published NR crystal structures and enables PPARγ to interact with the ID2 motif of NCoR and SMRT corepressor proteins with high affinity
likely by sequestering helix 12 away from the AF-2 surface where it would physically clash with the bound corepressor ID2 peptide
the SR10171- and SR11023-bound crystal structures do not report on the conformation of helix 12 in an activity-dependent
Structural studies in the presence of corepressor peptides may provide additional insight into the mechanism of action of these ligands compared with covalent inverse agonists and antagonists (T0070907 and GW9662) that enable helix 12 to exchange between a solvent-exposed conformation and a buried conformation within the orthosteric ligand-binding pocket
It is therefore possible that other NRs could share a similar helix 12-dependent mechanism for transcriptional repression
though future studies would need to explore this in detail
but do not stabilize an active helix 12 conformation
Noncovalent orthosteric antagonists may maintain a conformation resulting in a transcriptionally neutral coregulator affinity balance that mimics the coregulator affinity of apo-PPARγ or PPARγ bound to endogenous ligands
noncovalent orthosteric inverse agonists may function by two related mechanisms: competing with endogenous PPARγ ligands that activate PPARγ transcription
and inducing a conformation that weakens coactivator binding without significantly increasing corepressor binding affinity
which in total maintains a relative preference for corepressor binding
the corepressor-selective inverse agonist mechanism of T0070907 we describe here is different
Our work suggests an corepressor-selective ligand design model in which ligands that are able to completely displace helix 12 from the AF-2 surface—e.g.
by stabilizing helix 12 within the orthosteric pocket—will robustly increase corepressor binding affinity resulting in transcriptional repression of PPARγ
Wild type or mutant human PPARγ ligand-binding domain (LBD) protein
was expressed from a pET46 Ek/LIC plasmid (Novagen) as a TEV-clevable N-terminal hexahistag fusion protein in Escherichia coli BL21(DE3) cells using autoinduction ZY media (unlabeled protein)
or using M9 minimal media (for NMR studies) supplemented with 15N ammonium chloride with or without 13C-glucose and D2O at 37 °C
cells were induced with 1.0 mM isopropyl β-D-thiogalactoside (M9) at an OD600nm of 0.6
grown for an additional 24–48 h at 18 °C then harvested
cells were grown for 5 h at 37 °C and additional 12–18 h at 22 °C then harvested
Cells were lysed and 6xHis-PPARγ LBD was purified using Ni-NTA affinity chromatography and gel filtration chromatography
TEV protease was used to cleave the histag for most experiments except protein used for TR-FRET and fluorescence polarization
The purified proteins were concentrated to 10 mg mL−1 in a buffer consisting of 20 mM potassium phosphate (pH 7.4)
Purified protein was verified by SDS-PAGE as >95% pure
Covalent binding of T0070907 to PPARγ LBD (wild type or mutant variants) was analyzed by ESI-MS using a LTQ XL linear Ion trap mass spectrometer (Thermo Scientific); samples were incubated with or without 2 molar equivalents of T0070907 (unless otherwise indicated below) at 4 °C overnight and diluted to 2–3 μM in 0.1% formic acid for ESI-MS analysis
HEK293T (ATCC CRL03216) were cultured in Dulbecco’s minimal essential medium (DMEM
Gibco) supplemented with 10% fetal bovine serum (FBS) and 50 units ml−1 of penicillin
Cells were plated 20,000 cells/well in a 96-well flat bottom cell culture plate and co-transfected with 100 ng pG5-UAS and 25 ng pCMV-Gal4-PPARγ (human residues 185–477
containing the hinge and LBD) along with pACT empty vector (Promega) expressing the VP16 transactivation domain only
pACT with VP16 fused to the mouse NCoR receptor interaction domain (RID
or pAct with VP16 fused to the NCoR RID with each critical residue of the ID2 motif (LEDIIRKAL) mutated to threonine (TEDTTRKAT)
Transfection solutions were prepared in Opti-MEM with Mirus Bio TransIT-LT1 transfection reagent
After 24 h incubation at 37 °C in a 5% CO2 incubator
DMSO or T0070907 was added at a final concentration of 0.01% or 10 μM
britelite plus (PerkinElmer) was added to each well
then transferred to a white-bottom 384-well plate and read on a BioTek Synergy Neo multimode plate reader
Data were plotted and analyzed using GraphPad Prism software
Cells were grown to 90% confluency in T-75 flasks; from this
2 million cells were seeded in a 10-cm cell culture dish for transfection using X-tremegene 9 (Roche) and Opti-MEM (Gibco) with full-length human PPARγ (isoform 2) expression plasmid (4 μg)
and a luciferase reporter plasmid containing the three copies of the PPAR-binding DNA response element (PPRE) sequence (3xPPRE-luciferase) (4 μg)
cells were transferred to white 384-well cell culture plates (Thermo Fisher Scientific) at 10,000 cells/well in 20 μL total volume/well
cells were treated in quadruplicate with 20 μL of either vehicle control (1.5% DMSO in DMEM media)
twofold serial dilution of TZDs for dose response experiments
cells were harvested with 20 μL Britelite Plus (PerkinElmer)
and luminescence was measured on a BioTek Synergy Neo multimode plate reader
Data were plotted in GraphPad Prism as luminescence vs
ligand concentration and fit to a sigmoidal dose response curve
Time-resolved fluorescence resonance energy transfer (TR-FRET) assays were performed in low-volume black 384-well plates (Greiner) using 23 μL final well volume
For the TR-FRET coregulator peptide interaction assay
each well contained 4 nM protein (WT or mutant 6xHis-PPARγ LBD)
1 nM LanthaScreen Elite Tb-anti-His Antibody (ThermoFisher #PV5895)
NCoR and SMRT peptide in TR-FRET buffer (20 mM potassium phosphoate
each well contained 1 nM protein (wild type or mutant 6xHis-PPARγ LBD)
1 nM LanthaScreen Elite Tb-anti-HIS Antibody (ThermoFisher #PV5895)
and 5 nM Fluormone Pan-PPAR Green (Invitrogen; #PV4894) in TR-FRET buffer at pH 8
compounds stocks were prepared via serial dilution in DMSO
and plates were read using BioTek Synergy Neo multimode plate reader after incubation at 25 °C for several time points between 1 and 24 h
and the acceptor FITC emission was measured at 520 nm
Data were plotted using GraphPad Prism as TR-FRET ratio (520 nm/495 nm) vs
The coregulator interaction data were fit to sigmoidal dose response curve equation
and ligand displacement data were fit to the one site–Fit Ki binding equation using the binding affinity of Fluormone Pan-PPAR Green (2.8 nM; Invitrogen #PV4894 product insert)
Wild type or mutant 6xHis-PPARγ LBD (with or without T0070907) was serially diluted in assay buffer (20 mM potassium phosphate
pH 8) and plated with 180 nM FITC-labeled TRAP220 ID2 or NCoR ID2 peptides in low-volume black 384-well plates (Greiner) in triplicate
For single PPARγ LBD concentration experiments
the final concentration of FITC-labeled SMRT (ID1 and ID2)
and TRAP220 ID2 peptides was 180 nM and wild-type 6xHis-PPARγ LBD concentration was 25 μM
and fluorescence polarization was measured on a BioTek Synergy Neo multimode plate reader at 485 nm emission and 528 nm excitation wavelengths
Data were plotted using GraphPad Prism as fluorescence polarization signal in millipolarization units vs
protein concentration and fit to a one site—total binding equation
To visualize the location of MTSL for the PRE NMR studies
residue K474 was first modeled into cysteine (C474)
then the MTSL ligand was imported into Coot as a SMILES string: CC1(C)C(CSS(C)(=O)=O)=CC(C)(C)N1[O]
The MTSL sulfinic acid leaving group was removed and placed adjacent to C474
after which it was regularized and covalently attached to C474
The parameters for pLink2 search were as follows: three missed cleavage sites for trypsin/chymotrypsin for each chain; peptide length 4–100 amino acid; pLink2 search results were filtered by requiring precursor tolerance (±10 p.p.m.) and fragment tolerance (±15 p.p.m.) and FDR below 5% was required for all identified XL-MS peaks
The ion intensities and peak areas of the crosslinked peptides from different experimental conditions were manually calculated and compared
The highest peak intensity across all indicated experimental conditions within each bar plot panel was normalized as 100%—first to raw peak area of a control peptide lacking a lysine residue (S289-VEAVQEITE-Y299) to normalize for sample injection
then to the highest mean peak area within each condition
Further information on research design is available in the Nature Research Reporting Summary linked to this article
All other datasets generated and/or analyzed during the current study are available from the corresponding author on reasonable request
Structural overview of the nuclear receptor superfamily: insights into physiology and therapeutics
Activation of nuclear receptors: a perspective from structural genomics
The antiparasitic drug ivermectin is a novel FXR ligand that regulates metabolism
Discovery of second generation rorγ inhibitors composed of an azole scaffold
A unique secondary-structure switch controls constitutive gene repression by retinoic acid receptor
A structural and in vitro characterization of asoprisnil: a selective progesterone receptor modulator
Ternary crystal structure of human RORγ ligand-binding-domain
an inhibitor and corepressor peptide provides a new insight into corepressor interaction
Ternary complex of human RORγ ligand-binding domain
inverse agonist and SMRT peptide shows a unique mechanism of corepressor recruitment
Molecular determinants of the recognition of ulipristal acetate by oxo-steroid receptors
Structure of Rev-erbα bound to N-CoR reveals a unique mechanism of nuclear receptor-co-repressor interaction
Molecular switch in the glucocorticoid receptor: active and passive antagonist conformations
X-ray crystal structures of the estrogen-related receptor-gamma ligand binding domain in three functional states reveal the molecular basis of small molecule regulation
Structural basis for antagonist-mediated recruitment of nuclear co-repressors by PPARα
Structural basis for retinoic X receptor repression on the tetramer
A revisited version of the apo structure of the ligand-binding domain of the human nuclear receptor retinoic X receptor alpha
Ligand binding and co-activator assembly of the peroxisome proliferator-activated receptor-gamma
NFκB selectivity of estrogen receptor ligands revealed by comparative crystallographic analyses
The human nuclear xenobiotic receptor PXR: structural determinants of directed promiscuity
Structural basis for the deactivation of the estrogen-related receptor gamma by diethylstilbestrol or 4-hydroxytamoxifen and determinants of selectivity
Small molecule modulation of nuclear receptor conformational dynamics: implications for function and drug discovery
Review of the structural and dynamic mechanisms of PPARγ partial agonism
Partial agonists activate PPARγ using a helix 12 independent mechanism
Structural basis for the activation of PPARγ by oxidized fatty acids
Ligand and receptor dynamics contribute to the mechanism of graded PPARγ agonism
Ligand-induced stabilization of PPARγ monitored by NMR spectroscopy: implications for nuclear receptor activation
The nuclear receptor corepressors NCoR and SMRT decrease peroxisome proliferator-activated receptor gamma transcriptional activity and repress 3T3-L1 adipogenesis
a selective ligand for peroxisome proliferator-activated receptor gamma
functions as an antagonist of biochemical and cellular activities
A structural mechanism for directing corepressor-selective inverse agonism of PPARγ
Defining a conformational ensemble that directs activation of PPARγ
PPARγ in complex with an antagonist and inverse agonist: a tumble and trap mechanism of the activation helix
Structural basis of the transactivation deficiency of the human PPARγ F360L mutant associated with familial partial lipodystrophy
Nitroxide labeling of proteins and the determination of paramagnetic relaxation derived distance restraints for NMR studies
Chemical crosslinking mass spectrometry reveals the conformational landscape of the activation helix of PPARγ; a model for ligand-dependent antagonism
Distance restraints from crosslinking mass spectrometry: mining a molecular dynamics simulation database to evaluate lysine-lysine distances
Pharmacological repression of PPARγ promotes osteogenesis
Cooperative cobinding of synthetic and natural ligands to the nuclear receptor PPARγ
Crystal structure of the ligand binding domain of the human nuclear receptor PPARγ
Structural mechanism for signal transduction in RXR nuclear receptor heterodimers
Analysis of ligand binding and protein dynamics of human retinoid X receptor alpha ligand-binding domain by nuclear magnetic resonance
The effect of antagonists on the conformational exchange of the retinoid X receptor alpha ligand-binding domain
Prediction of the tissue-specificity of selective estrogen receptor modulators by using a single biochemical method
Unique ligand binding patterns between estrogen receptor alpha and beta revealed by hydrogen-deuterium exchange
Hydrogen/deuterium exchange reveals distinct agonist/partial agonist receptor dynamics within vitamin D receptor/retinoid X receptor heterodimer
Helix 11 dynamics is critical for constitutive androstane receptor activity
Defining the communication between agonist and coactivator binding in the retinoid X receptor alpha ligand binding domain
Antidiabetic phospholipid-nuclear receptor complex reveals the mechanism for phospholipid-driven gene regulation
iMOSFLM: a new graphical interface for diffraction-image processing with MOSFLM
Overview of the CCP4 suite and current developments
The Phenix software for automated determination of macromolecular structures
Coot: model-building tools for molecular graphics
CASTp 3.0: computed atlas of surface topography of proteins
UCSF Chimera—a visualization system for exploratory research and analysis
Using chemical shift perturbation to characterise ligand binding
NMRFx Processor: a cross-platform NMR data processing program
Using NMRView to visualize and analyze the NMR spectra of macromolecules
Identification of cross-linked peptides from complex samples
Download references
We thank Paola Munoz-Tello and Ted Kamenecka for helpful discussions and input
This work was supported by National Institutes of Health (NIH) grants F32DK108442 (R.B.)
and R01DK101871 (D.J.K.); Richard and Helen DeVos graduate fellowship award (S.M.); and National Science Foundation (NSF) award 1359369 that funds the SURF program at Scripps Research Florida (S.M.)
Use of the Stanford Synchrotron Radiation Lightsource
Office of Basic Energy Sciences under Contract No
The SSRL Structural Molecular Biology Program is supported by the DOE Office of Biological and Environmental Research
National Institute of General Medical Sciences (including P41GM103393)
The contents of this publication are solely the responsibility of the authors and do not necessarily represent the official views of NIDDK
Department of Integrative Structural and Computational Biology
Skaggs Graduate School of Chemical and Biological Sciences
Summer Undergraduate Research Fellows (SURF) program
solved the crystal structures and performed NMR and biochemical assays
performed the mammalian two-hybrid assay; A.N
performed chemical crosslinking mass spectrometry experiments
supervised the research and wrote the manuscript along with J.S
and input from all authors who approved the final version
The authors declare no competing interests
Peer review information Nature Communications thanks the anonymous reviewer(s) for their contribution to the peer review of this work
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations
Download citation
DOI: https://doi.org/10.1038/s41467-020-14750-x
Anyone you share the following link with will be able to read this content:
a shareable link is not currently available for this article
Sign up for the Nature Briefing newsletter — what matters in science
Amazon’s first robotics centre in the Czech Republic has officially launched its operations and is up and running
with a total area just shy of 190,000 square metres
and is outfitted with cutting-edge robotic technology
it stands as one of the Czech Republic's most state-of-the-art logistics hubs
Designated as "BRQ2," the centre officially commenced operations in June of this year
It heralds the introduction of the latest technologies aimed at simplifying daily tasks for Amazon employees
while prioritising the safety of operations and offering high-quality working conditions for our staff
To showcase this cutting-edge facility to the public
we recently organised a special guided tour of the centre for members of the media
BRQ2's team prepared a robot performance for those in attendance
synchronising several robotic units to create the BRQ2 inscription on the ground
We were also honoured to host the Minister of Trade and Industry
along with the Olomouc Region Governor and the Mayor of Kojetín
They spoke in favour of the facility and praised it as a significant asset
not only for the region but also for the entire country
We’re committed to being a good neighbour in every region where we operate; we’ve donated €10,000 towards establishing a rescue service station near Kojetín
with the cheque received by the Governor of the Olomouc Region from the hands of Michal Šmíd
“Amazon’s investment in Kojetin is another demonstration of our commitment to the Czech Republic
creating up to 2,000 good new permanent roles when fully operational,” said Michal Šmíd
adding: “Amazon provides some of the most advanced workplaces of their kind in the world
and systems to ensure the wellbeing and safety of all employees
and I am delighted to launch this new building.”
Amazon has invested over 26 billion Czech crowns in the Czech Republic
A portion of this investment is dedicated to implementing advanced technologies that support safer work methods
and improve wellbeing by reducing the need for heavy lifting
Amazon’s European Advanced Technology team
has played a pivotal role in creating more than 550 new pieces of technology
with over €400 million invested across Amazon sites in Europe in three years
Bethel’s Core Value Mark of Distinction award highlights employees who consistently live out the core values of our university
these four Bethel community members were named as the 2019 award recipients
Doug Kojetin is honored as a Christ-follower and character-builder for his tireless hard work which ensures our facilities run smoothly
attests to Kojetin’s impact through his work
“Doug has a true servant heart,” she says
“He works tirelessly to serve others… Everyday no matter the challenges presented to him
and is always willing to tackle any kind of task
He is extremely hardworking and efficient with his use of time on a given task.”
Kojetin is praised for his commitment to getting the job done in the most efficient and effective way possible
making sure his solutions are always safe and cost-effective
He consistently goes above and beyond to serve the entire Bethel community
even going so far as to stay overnight on campus during snow emergencies in order to ensure everyone is safe and taken care of
“Doug’s example of excellence in service is far reaching with students
Kojetin’s work in Facilities Management reflects his own character and relationship with Christ everyday
Education Licensing & Advising Specialist
Nadine Johnson is praised by her colleagues and students for the joy and hospitality she radiates in the Education Department
Johnson is a source of light and God’s love to her department and students
Associate Professor of Education Geri Von Grey works alongside Johnson
“professing Jesus is Savior and Lord and modeling His character to others through such manners as being kind
but rather rejoicing in the right while staying rooted in Scripture.”
Johnson is a light in the Education Department
affecting everyone she comes in contact with as she works hard to ensure their success
Students consistently applaud her consistent ability to lift others up
She works hard to prepare the teachers that will come out of Bethel as people who will be ready to make a difference in the lives of their students.
Director of Conference & Event Services
Avis Soderstrom is the face of Bethel to many people outside our community through her role in Conference & Event Services
Soderstrom has built a team that not only advances Bethel’s mission and continues to strengthen our reputation
“Avis has developed a team that is superb at working with internal and external audiences to help them achieve their goals while they are at Bethel,” says John Addleman
“[She] is an excellent example of how our commitment to make a difference in the world can impact all those around her
Not only is Soderstrom a strong piece of Bethel’s Conference & Event Services
her work is a source of salt and light to internal and external audiences
“Avis is faithful to Jesus and His call on her life
one who recognizes that the little details make others feel special
much like salt that makes us notice flavors that we would have missed otherwise.”
“Avis is a wonderful ambassador for Bethel
She embodies Bethel’s values and mission,” says Addleman
faithful to Jesus and His call on her life… What a precious gift [she is] to all of Bethel and to the many outside of Bethel whom [she] meets in the course of fulfilling [her] duties here.”
Sue Gazich is consistently praised by her coworkers and supervisors for her ability to provide professional and comprehensive care to everyone that comes into Health Services
She leads the department in lab coding in order to ensure that the staff is equipped to give the highest level of care they can.
Gazich is a Christ-follower as she cares for her patients’ well being
Sue conveys personal interest and affection for the individual
not just about the issue that brings them into the clinic
Her genuine and caring personality sets students at ease and almost always brings them a smile
she is truly a source of healing love,” says Liz Miller
“She makes a connection with each [patient] and that gives them a warm feeling that they are welcomed and loved like Jesus did in His ministry,” says Judy Jackson
“Our Bethel students/patients deserve our best
Her connection with the Bethel community goes beyond her work as a nurse
I know she prays for the students and everyone in Health Services and Counseling Services know that we can ask Sue to pray for us and it will happen
She is genuinely interested in others and how she can be praying for them,” says Hannah Wassink
“If I could describe Sue in one word
and Gazich were recognized and given the Core Value Mark of Distinction Award at the Executive Leadership Team meeting by Bethel’s senior management
Each recipient received a check of $250 and a custom Bethel University fleece jacket
we share a common goal: to become whole and holy persons
we have created core values that will help us get there
We understand that in order to be come people who reflect the image of God
we must bring transformation into every part of our lives
you will see that our core values affect academics
Learn more
Support Bethel
© 2025 Bethel University | CA Student Disclosures | Privacy Policy
Bethel University’s Christ-centered mission includes a commitment to equal educational opportunity and equal employment opportunity
Bethel does not unlawfully discriminate on the basis of race
Within the intellectual powerhouse that is Vanderbilt University and Vanderbilt University Medical Center thrives a group of doctors
computer scientists and others who also are visionaries
Several of these change-makers also donate their time and expertise through the Vanderbilt Innovation Ambassadors Program to help colleagues in their departments recognize and pursue the world-altering potential of their own ideas.
“We’re like the kindling that gets the fire going,” said Ambassador Bryan Hartley, MD, associate professor of Clinical Radiology & Radiological Sciences at VUMC whose company, Pulmera
is developing novel imaging technology to improve the diagnosis of early-stage lung cancer
An extension of long-standing, campuswide efforts to encourage innovation and commercialization activities, the Innovation Ambassador Program was launched in 2022 by the Center for Technology Transfer and Commercialization (CTTC) in collaboration with VUMC, Vanderbilt University School of Medicine and The Wond’ry
“Having an ambassador program shows that innovation and entrepreneurship are valued and encouraged,” noted Philip Swaney
who co-directs the program with CTTC director and Vanderbilt University assistant vice chancellor Alan Bentley
When faculty members see their peers “having an impact in this space,” Swaney said
“they feel empowered and supported to act.”
Innovation Ambassadors receive about six hours of initial training in commercialization
as well as relevant university policies and procedures
They’re also kept abreast of new programs and processes that are being rolled out across campus
The goal is to enable them share with others in their departments the information and resources
that can help them protect and commercialize their ideas and inventions
Ambassadors commit to serve two-year terms
In addition to communicating what they’ve learned
they meet quarterly to brainstorm ways to boost innovation and entrepreneurship
An example of their outreach is the Sullivan Family Biomedical Ideator program, launched in January by Ambassador Daniel Fabbri
director of Informatics Innovation at VUMC
Program participants learn ways to evaluate the commercial value and tech-transfer potential of their research
Other examples from the Department of Otolaryngology-Head and Neck Surgery are the national Haynes Award, honoring resident physicians who have made notable innovations, and the Haynes Innovation Symposium, which, according to the department’s innovation webpage
“provides practical insights to aspiring innovators on the strategies and pathways toward commercialization and dissemination.”
Launched with the help of Alexander Langerman, MD, SM, the Otolaryngology Department’s Ambassador, both the award and symposium are named for David Haynes
holder of the Directorship in Otolaryngology for Relationship Development
and Chief Patient Experience Officer at VUMC
The brainchild of department chair Eben Rosenthal
the award and symposium celebrate the “best and the brightest of the next generation of leaders,” and highlight VUMC’s innovative work
an accomplished innovator in his own right
Langerman and Fabbri are among the first recipients of Vanderbilt University’s Innovation Catalyst Fund for innovations they have brought to their respective fields
the fund helps streamline faculty research into real-world applications
“The Innovation Ambassador Program provides a great resource for faculty to learn more about inventions and technology commercialization directly from their peers,” said Kenneth Holroyd, MD, MBA, CTTC’s medical director, and lead of the Brock Family Center for Applied Innovation at VUMC.
“It is an important and growing part of building VUMC’s entrepreneurial culture,” said Holroyd
who also is VUMC’s Vice President for Tech Transfer
“We are looking for additional faculty volunteers to serve from departments that are not currently participating in this program.”
Of the 20 current Innovation Ambassadors, 13 are primarily affiliated with VUMC, including Shabnam Eghbali
a first-year resident physician in Internal Medicine
who plans a dual career in medicine and biopharma investment
said she hopes as an Innovation Ambassador to help other physicians in specialty training learn how they can apply their knowledge to affect large-scale changes in health care
represent the departments of Biochemistry and Pharmacology in the School of Medicine Basic Sciences
TS Harvey, PhD, co-recipient of a $1 million grant from the Robert Wood Johnson Foundation to study cultural factors in health inequities
represents the Department of Anthropology in the College of Arts and Science
Ambassador Christopher Vanags
directs the Peabody Research Office in the Vanderbilt Peabody College of Education and Human Development
The Ambassadors program “is a grassroots approach to building out our innovations
Ambassador program leadership is helping a half dozen other universities launch similar ambassador-like programs of their own
Vanderbilt has long been a hotbed of invention
thanks in large part to the creativity of its faculty and the support of the CTTC
and 593 patents and 923 licenses were issued
generating more than $286 million in revenue.
Vanderbilt and VUMC ranked sixth in the country in adjusted gross income from technology licensing
according to the latest survey conducted by AUTM
a nationwide community of university technology transfer offices. In FY2023
licensing revenue topped a record $96 million.
technology transfer is more than a revenue stream — it can open the door to inventors who want to help change the world for the better.
Physicians want to have a positive impact on their patients’ lives. The Innovation Ambassadors show that “they can also have a great impact through innovation and entrepreneurship,” said Ambassador Ryan Buckley, MD, who also directs the Medical Innovators Development Program for Vanderbilt medical students
Yet “people really need examples, advice and informal conversations with fellow entrepreneurs to be successful,” said Webster, the Richard A. Schroeder Professor of Mechanical Engineering who has co-founded companies that introduced innovations to the robotic surgery and flexible endoscopy fields
Last year the National Institutes of Health awarded a four-year, $4 million grant to Webster and Charleson Bell, PhD, director of entrepreneurship and biomedical innovation at the Wond’ry, to establish the Mid-South Research Evaluation and Commercialization Hub (REACH)
Leveraging an additional $8 million in state and institutional matching funds
mentorship and financial support for aspiring entrepreneurs to hundreds of community colleges and minority-serving institutions in Kentucky
accustomed to receiving government support for their research and publishing results in professional journals
are reluctant to leap into entrepreneurship
But unless they take steps to protect their intellectual property
“Basically, you’re killing it,” warned Ambassador Robert Carnahan, PhD, associate director of the Vanderbilt Vaccine Center, who helped develop business modules for scientists offered through the School of Medicine’s BRET Office of Career Development
“Most things require a commercial platform,” Carnahan explained
“If (companies) are going to put a million dollars into it
they need to know they’re going to get a return on their investment
you’ve got to put some protections around it.”
Entrepreneurship is not for the faint of heart
“It is not easy to take a proof-of-principle result from the bench to the market,” cautioned Kostoulas
professor of the Practice of Engineering Management
who has extensive experience as a marketing professional in the semiconductor industry
“Most of the work and expense happens during that transition.”
Faculty inventors don’t have to shoulder this burden alone
By pointing the way to the tech transfer office and investment community
Innovation Ambassadors encourage crucial collaborations that can help turn vision into reality
“The Innovation Ambassadors program provides a dynamic network for all of those interactions to happen.” Webster said
“By building this into the fabric of what we do at Vanderbilt
we will ultimately better achieve Vanderbilt’s core missions of discovery and translating those discoveries to impact the real world.”
Colin Barker
Barker is an international leader in interventional cardiology and percutaneous heart valve therapies
His research has led to development of devices to prevent strokes and improve treatment of congestive heart failure
Ryan Buckley
For Buckley, who also directs the Medical Innovators Development Program for Vanderbilt medical students
innovation is about bringing people together to find new ways to alleviate the suffering of others
Robert Carnahan
Professor of Pediatrics and of Radiology and Radiological Sciences
A leader in developing monoclonal antibody treatments for a host of viral diseases
Carnahan also is known internationally for his contributions to rational vaccine design
Edward Chaum
A self-described “serial entrepreneur,” Chaum specializes in the medical and surgical management of retinal diseases
drug development and biomedical engineering
Shabnam Eghbali
Eghbali, who worked for two investment funds while in medical school, is co-founder of Theia Healthcare
a nonprofit dedicated to inspiring and empowering women entrepreneurs and investors in healthcare
Her goal is a dual career as physician and biopharma investor
Daniel Fabbri
Holder of the DBMI Directorship in Informatics Innovation at VUMC
Biomedical Informatics and Computer Science
Fabbri has developed and marketed tools to improve the security and efficient use of medical records and envisioned an “Ideator” program to evaluate the marketability of faculty research
Bryan Hartley
Clinical Radiology & Radiological Sciences
A Vanderbilt-trained radiologist, Hartley was inspired by his experience as a Stanford Biodesign Innovation Fellow to “take the next step” to entrepreneurship. He inspires others as a contributor to the BackTable Innovation podcast series
TS Harvey
Vanderbilt University’s first Ford Foundation Senior Fellow
Harvey’s research in Guatemala and elsewhere focuses on expanding scientific partnerships
and building local capacities to collaboratively tackle large-scale public health and environmental challenges experienced by vulnerable populations.
Douglas Kojetin
Kojetin, who joined the Vanderbilt faculty in 2023, uses “structural pharmacology” and computational biology methods to define the molecular mechanisms of drug action and to probe the regulation of nuclear receptors, which play a role in a broad spectrum of human disease.Yiorgos Kostoulas
Professor of the Practice of Engineering Management
Division of Engineering Science and Management
Kostoulas is a researcher (optical spectroscopy) who also has extensive professional experience in marketing in the semiconductor industry
Alexander Langerman
Associate Professor of Otolaryngology-Head & Neck Surgery
Langerman is a head-and-neck surgeon and was co-founder of Surgical Explorer
now part of GHX (Global Healthcare Exchange)
which operates a platform that enables surgical teams to access remote
medical device support directly from suppliers
George Nicholson
Trained in interventional cardiology procedures
Nicholson and his colleagues work with biomedical engineers in the design and development of tools
techniques and devices tailored to improve outcomes for children with congenital heart disease
Chris Vanags
Research Assistant Professor of Earth and Environmental Sciences
Vanags helped develop and implement widely adapted STEM enrichment programs
Jonathan Wanderer
Professor of Anesthesiology and Biomedical Informatics
Associate Chief Medical Information Officer, VUMC
Wanderer innovates clinical informatics-driven, perioperative interventions, around the time of surgery. He directs the eStar Physician Builders Program
which supports the devising of new content and tools for VUMC’s health and information technology system
Alex Waterson
Research Professor of Pharmacology and Chemistry
Associate Director for Medicinal Chemistry
Vanderbilt Center for Cancer Drug Discovery
Waterson’s drug discovery collaborations span the cancer
and include service on the scientific advisory board of Nashville-based Cumberland Emerging Technologies
Robert Webster
Schroeder Professor of Mechanical Engineering
Webster is investigating the use of surgical robotics in ear
James Weimer
Assistant Professor of Computer Science and Biomedical Engineering
A focus of Weimer’s research is cyber-physical systems in health care. He has cofounded companies that developed a non-invasive continuous stroke monitoring system (Neuralert), and novel technology to monitor the risk of postpartum hemorrhage during labor and delivery (Vasowatch)
Lauren Williamson
Research Assistant Professor of Pediatrics
Williamson’s method of stabilizing cell populations that play essential roles in autoimmune disorders won second place in the 2022 Innovation Cup sponsored by the German science and technology company Merck KGaA
She also consults for the biotech industry
Jesse Wrenn
Wrenn’s research is focused on applying computer science and informatics methods to improve clinical care. He is part of a team investigating whether a strategy to individualize diuretic therapy will improve outcomes for patients hospitalized with acute heart failure
Adam Yock
Holder of the Directorship in Technology and Innovation for Radiation Oncology
Yock is a medical physicist whose group is developing quality assurance devices and treatment planning software to improve the accuracy and outcomes of radiation therapy
assistant professor of Chemical and Biomolecular Engineering at Vanderbilt University
received a $50,000 research grant at the 2019 Vanderbilt-Ingram Cancer Center Ambassador Breakfast
Many clinical trials are stopped prematurely because they fail to recruit enough study participants
Vanderbilt University Medical Center has received a five-year
$14 million grant from the National Center for Advancing Translational Sciences at the National Institutes of Health to address this
Vanderbilt-Ingram Cancer Center Ambassadors surpassed the $1 million mark with the awarding of the group’s latest research grants
His Mass of Christian Burial will be 11:00 am
Mary in Willmar with interment in the church cemetery. Visitation will be Tuesday
with a Knights of Columbus Rosary at 5:00 pm at Harvey Anderson Funeral Home in Willmar. www.hafh.org Visitation will continue one hour prior to mass at the church on Wednesday. Memorials may be given to the Willmar Area Food Shelf or the Andrew Hatlestad Memorial Nursing Scholarship
and Angela (Kojetin) Frank in Renville County. He was the eldest of five children
Michael’s Catholic Church in Morgan. They made their home on a farm in Henryville Township in Renville County. Myron and Elaine worked hard and enjoyed life on the farm along with their 10 children. Myron’s specialty was raising pullets and sugar beets. Myron was a member of the Knights of Columbus
Catherine’s Church Council in Redwood Falls
and served as a board member for the Danube Farmers Elevator and Oil Company. Myron was also one of the founding members of Southern Minnesota Beet Sugar Cooperative
Myron served as treasurer of the board for the Willmar Senior Citizens
and playing cards. He shared his love and knowledge of the land
hard work and faith with his children and grandchildren. He also taught them many card games and was notorious for winning most of the time. Myron loved to drive around his farm and the countryside to check how the crops were growing. Such drives made his day complete even up until his last days
children; Kathleen (Steve) Chesney of Brooklyn Park
and Brenda Frank (Doug Knight) of Denver CO; Also surviving are his siblings: Donald (Veronica) Frank of Redwood Falls
Betty Miller of Redwood Falls and Susie Drexler of Willmar. Also surviving are his beloved 13 grandchildren and 19 great-grandchildren.
Credit: Photo courtesy of The Scripps Research Institute
2014 – Scientists from the Florida campus of The Scripps Research Institute (TSRI) have been awarded $2.1 million from the National Institutes of Health to study the therapeutic potential of safer and more effective alternatives to the current crop of anti-diabetic drugs
which have been limited in their use due to side effects including bone loss and congestive heart failure
is the principal investigator for the new five-year study
The study will take a cue from the two mainstays of type 2 diabetes treatment—pioglitazone (Actos) and rosiglitazone (Avandia)
Both drugs raise the body's sensitivity to insulin
increasing the amount of glucose or sugar absorbed by the cells
that while these and other recently developed drugs are designed to bind to a specific site on the PPAR gamma (PPARG) nuclear receptor
"This unexpected finding opens a lot of potential opportunities," Kojetin said
"We're looking to design a molecule that blocks both sites and can be used to probe what this alternative binding does on a molecular level—with the hope that this information will help us come up with a better drug model."
are not responsible for the accuracy of news releases posted to EurekAlert
by contributing institutions or for the use of any information through the EurekAlert system
Copyright © 2025 by the American Association for the Advancement of Science (AAAS)
Photo Contest
by Madeline Kopiecki | Oct 2024
we asked local residents to scroll down their digital files or flip through their film—whatever it took to find their favorite shots of the Lake Minnetonka community
From scenic landscapes to candid portraits
artistic abstractions to vibrant close-ups
Lake Minnetonka Magazine readers delivered their favorite scenes from the past year
We would like to extend our thanks to returning contestants and newcomers alike
we’re excited to share the winning photos with you
Please note that judges reserve the right to recategorize submissions if they feel the photograph is better suited to a different category
Title: Sunset Cruise – Lake Minnetonka
Title: Minnetonka Yacht Club In The Clouds
Sign up for our newsletter and receive email updates with our top stories
History, Noteworthy
Noteworthy, Recipes
Noteworthy, Shops, Style
© Copyright 2026 Local. All Rights Reserved | Privacy Policy
Read the May 2025 Lake Minnetonka Magazine Digital Edition
Metrics details
A subset of nuclear receptors (NRs) function as obligate heterodimers with retinoid X receptor (RXR)
allowing integration of ligand-dependent signals across the dimer interface via an unknown structural mechanism
Using nuclear magnetic resonance (NMR) spectroscopy
x-ray crystallography and hydrogen/deuterium exchange (HDX) mass spectrometry
here we show an allosteric mechanism through which RXR co-operates with a permissive dimer partner
peroxisome proliferator-activated receptor (PPAR)-γ
while rendered generally unresponsive by a non-permissive dimer partner
Amino acid residues that mediate this allosteric mechanism comprise an evolutionarily conserved network discovered by statistical coupling analysis (SCA)
This SCA network acts as a signalling rheostat to integrate signals between dimer partners
thereby affecting signal transmission in RXR heterodimers
These findings define rules guiding how NRs integrate two ligand-dependent signalling pathways into RXR heterodimer-specific responses
it is poorly understood how ligand binding to one LBD controls co-regulator recruitment to its dimer partner within a NR heterodimer complex
The structural mechanisms that generate this spectrum of signalling outcomes are unknown
where helix 5 functions more generally as a signalling rheostat that integrates signals with the dimer interface
(a) CV-1 cells co-transfected with PPARγ expression plasmid
RXRα expression plasmid and a PPAR-responsive luciferase reporter
10 μM 9cRA and/or 10 μM rosiglitazone for 24 h
Magenta and black bars are coloured to match NMR data in d
Luciferase activity is shown normalized to vehicle-treated cells and was performed in quadruplicate
plotted with the average (±s.e.m) and representative of at least three experiments
(b) CV-1 cells co-transfected with TRβ expression plasmid
RXRα expression plasmid and a TR-responsive luciferase reporter
100 nM LG100268 (LG268) and/or 1 μM T3 for 24 h
plotted with the average (±s.e.m) and representative of at least two experiments
Magenta and black bars are coloured to match NMR data in e
(c) Structural location of RXR residues mentioned in the NMR analysis
(d) Overlay of 2D [1H,15N]-TROSY-HSQC NMR data for [2H,13C,15N]-RXRα LBD in the apo form and coloured magenta—with the same bound to 9cRA and coloured black
(e) Overlay of 2D [1H,15N]-TROSY-HSQC NMR data for [2H,13C,15N]-RXRα LBD bound to 9cRA and heterodimerized to apo-PPARγ and coloured magenta—with the same bound to rosiglitazone (Rosi) and coloured black
(f) Overlay of 2D [1H,15N]-TROSY-HSQC NMR data for [2H,13C,15N]-RXRα LBD bound to 9cRA and heterodimerized to apo-TRβ and coloured magenta—with the same bound to T3 and coloured black
indicating this may be a general feature for ligand activation of NRs
To determine the mechanism through which PPARγ acts as a permissive dimer partner, we performed differential NMR analysis by adding unlabelled apo-PPARγ to 9cRA-bound isotopically labelled RXRα, with and without addition of the full PPARγ agonist, rosiglitazone (Fig. 1b,d)
NMR chemical shift changes in RXRα are observed on addition of apo-PPARγ to the 9cRA-bound RXRα
consistent with complete formation of a heterodimer complex
Addition of rosiglitazone causes subtle but significant NMR chemical shift changes in RXRα (for example
S355 and G429 in the dimer interface) and only minor changes in NMR resonance linewidths for select residues
although heterodimerization with and ligand binding to PPARγ perturbs the conformation of RXRα
neither of these events dramatically affects the μs–ms dynamics of RXRα
This is in contrast to what occurs with the non-permissive RXR partner
To determine the mechanism through which TRβ acts as a non-permissive RXR heterodimer partner, we performed differential NMR analysis by adding unlabelled apo-TRβ to 9cRA-bound isotopically labelled RXRα, with and without addition of the TRβ agonist, T3 (Fig. 1b,e)
addition of apo-TRβ exerts a profound effect on the μs–ms conformational dynamics of 9cRA-bound RXRα
where a large number of agonist-bound RXRα NMR resonances revert to an apo-like NMR profile
NMR resonances that are destabilized—missing or have broad linewidths indicating increased μs–ms motion—correspond to RXRα residues in the ligand-binding pocket (for example
G413 and G429) and other nearby regions such as helix 8 (for example
Even more striking is that the addition of the TRβ agonist
re-stabilizes these agonist-bound RXRα residues by decreasing motion on the μs–ms timescale
resulting in a reappearance of NMR resonances for these regions
many of the missing NMR resonances in the apo-TRβ/agonist-RXRα heterodimer correspond to residues in the apo-RXRα homodimer that are stabilized on binding 9cRA
Our NMR studies indicate that these residues are not affected by PPARγ heterodimerization or ligand binding to PPARγ
but they are significantly affected by TRβ heterodimerization and ligand binding to TRβ
these data indicate that the mechanism through which RXRα is allosterically silenced by TRβ but not PPARγ
involves conformational dynamics on the μs–ms timescale
(a) Structure of the TR/RXR LBD heterodimer is shown as ribbon with T3 as space filled
and the bound SRC-2 peptide is coloured blue and only binds to T3·TR
with the dimer interface coloured coral and helix 12 coloured purple
and RXR helix 12 adopts an inactive conformation positioned into the AF-2 co-activator-binding surface
(b) TR is superimposed on the RXR LBD homodimer (PDB 1MVC)
except dimers are superimposed via the RXR protomer
illustrating the shift in the TR dimer interface relative to the other RXR promoter in the RXR homodimer
The magenta circle highlights the only region that superimposes similarly between TR and RXR
The amino-terminal end of TRβ helix 11 is oriented similarly towards RXRα
10 and the carboxyl-terminal part of helix 11 are substantially shifted
this altered dimer interface induces conformational changes in RXRα
(a) Helix 11 of the dimer interface shown as Cα traces for TRβ·T3·SRC-2/apo-RXRα
RXR/PPARγ (PDB 1FM6) and RXR/LXR (PDB 1UHL) heterodimers were superimposed on the TRβ·T3·SRC-2/apo-RXRα structure using the RXR promoter molecule and are coloured grey
(b–d) The active conformation RXR homodimer (PDB 1MVC) superimposed on the TRβ·T3·SRC-2/apo-RXRα structure via RXR and coloured as in a
(b) TR helix 11 (green) induces a shift in the RXR helix 11 (coral) relative to the RXR homodimer (grey)
(c) The unique position of TR T426 in helix 11 induces a shift in RXR P423 in helix 11 and a rotation of the RXR helix 11 backbone
(d) The location of TR A433 in helix 11 away from the dimer interface compared with the equivalent residue in RXR
allows RXR L430 and the RXR helical backbone to rotate in context of the TRβ·T3·SRC-2/apo-RXRα heterodimer
(e) RXR in the active conformation (PDB 1MVC) with RXR ligand (MBS649) shown as space filled and SRC-2 peptide bound to the AF-2 co-activator-binding surface coloured red
RXR W305 in helix 5 mediates contacts with the ligand
M454 in helix 12 and the co-activator-binding site via L276 in helix 3
Colour is used to help differentiate secondary structural elements and provide depth for overlapping elements; helix 3 and 4 in cyan
(f) TRβ·T3·SRC-2/apo-RXRα was superimposed with the active conformation RXR homodimer (PDB 1MVC) and shown as Cα trace
The rotation of helix 5 in TRβ·T3·SRC-2/apo-RXRα repositions W305 such that it clashes with the active conformation of RXR L276
but showing the active conformation of helix 12
and the clash with the rotated position of W305 in the TRβ·T3·SRC-2/apo-RXRα heterodimer
Thus in the TRβ·T3/apo-RXRα structure the TRβ-induced rotation of RXRα helix 11 and helix 5 disables the active conformation of RXRα
although the structural mechanism that drives this effect at the atomic level remained unknown
(a) The TRβ·T3·SRC-2/apo-RXRα structure (green and coral) superimposed with the active conformation RXR homodimer (PDB 1MVC) via RXR and shown in grey
E307) are part of a network of co-evolved amino acids identified using a statistical coupling analysis (SCA)
(b) The SCA network amino acids shown as space filled on the TRβ·T3·SRC-2/apo-RXRα heterodimer link the dimer interface
ligand-binding pocket and AF-2 co-activator-binding site
(c) Active conformation RXR homodimer (PDB 1MVC) superimposed with RXR from the TRβ·T3·SRC-2/apo-RXRα heterodimer
Shown are helix 3 and helix 4 of the AF-2 surface
The rotation of helix 5 induces an altered conformation of the AF-2 surface via the SCA network amino acids
(d) Regions in RXR that are protected from HDX on heterodimerizaton with TR
The changes in HDX support our model where these regions direct allosteric signalling within the heterodimer resulting in the silencing of RXR by TR
(a,b) CV-1 cells co-transfected with TRβ expression plasmid
(a) RXRα or (b) RXRα L276V mutant expression plasmid
or the indicated dose of T3±1 μM 9cRA for 24 h
(c) Drosophila SL2 cells co-transfected with TRβ expression plasmid
RXRα or RXRα E434-mutant expression plasmid
the RXR agonist LG100268 (LG268; 100 nM) and/or 1 μM TR agonist (T3) for 24 h
(d) CV-1 cells co-transfected with TRβ expression plasmid
RXRα or RXRα E434N mutant expression plasmid
Cells were treated with vehicle or the indicated dose of T3 overnight
(e) Drosophila SL2 cells co-transfected with VDR expression plasmid
the RXR agonist LG100268 (LG268; 100 nM) and/or 1 μM VDR agonist (vitamin D3) for 24 h
Luciferase activity is shown normalized to vehicle-treated cells and was performed in quadruplicate; plotted with the average±s.e.m and representative of at least three experiments
the C terminus of RXRα helix 11 plays a critical role in regulating the response of the TRβ/RXRα heterodimer to T3
Although RXR ligand has no activity on its own
within the context of the VDR/RXRα heterodimer it is conditionally permissive because it enhances vitamin D3-induced transactivation
The helix 11 mutations selectively modulate the conditional activation by the combination of vitamin D3 and RXR agonists
and lead to both gain- and loss-of-function
these data suggest that similar helix 11-mediated mechanisms control allosteric signalling across the dimer interfaces of TR/RXR and VDR/RXR heterodimers
and that there are several mechanisms for heterodimer signal integration
NMR data are coloured grey for 9cRA-bound RXRα; black for 9cRA-bound RXRα heterodimerized to apo-PPARγ or the same bound to the following PPARγ ligands: rosiglitazone (magenta)
MRL24 (orange) or SR1664 (green); plotted on PPARγ/RXRα (PDB 1FM9)
(a) NMR data (left) focusing on residues in RXRα helix 7 and helix 10/11 dimer interface that are perturbed by ligand binding to PPARγ
which are plotted onto the PPARγ/RXRα crystal structure and coloured according to structural location (yellow for helix 7; blue for helix 10/11); coloured dark if shown in the NMR data to the left or light if not
(b) NMR data (left) focusing on residues in core of RXRα that are perturbed by ligand binding to PPARγ
which are plotted onto the PPARγ/RXRα crystal structure and coloured red; and coloured dark if shown in the NMR data to the left or light if not
(c) NMR data (left) focusing on residues in RXRα helix 12
the AF-2 surface and the ligand-binding pocket that are perturbed by ligand binding to PPARγ
which are plotted onto the PPARγ/RXRα crystal structure and coloured according to structural location (green for AF-2/helix 12; orange for the ligand-binding pocket); coloured dark if shown in the NMR data to the left or light if not
RXRα K431 in helix 11 forms a hydrogen bond with PPARγ Y477
RXRα residues affected differently by the graded PPARγ ligands are structurally close to this region
the effect of PPARγ full agonists on RXRα residues in the dimer interface are likely mediated through stabilization of PPARγ helix 12 and its interaction with RXRα
but notable changes in RXRα helix 3 residue L276
as well as AF-2 surface residues K284 and S290
which are part of a region that forms electrostatic interactions with the bound co-activator peptide in the crystal structures
RXRα helix 5 is a key part of the LBD core that transmits PPARγ ligand-induced allosteric signals from the dimer interface to the RXRα ligand-binding pocket and the AF-2 surface
also mediates PPARγ ligand-induced allosteric signalling across the dimer interface in a similar direction
Helix 9 forms part of the core that contacts the dimer interface
and stabilizes the AF-2 surface via interaction with helix 3 and helix 4 residues
PPARγ ligand-induced NMR chemical shift changes in RXRα helix 11 (for example
suggest that helix 9 may also transmit allosteric information from the dimer interface to the AF-2 surface
Additional NMR resonances showing specific changes in response to PPARγ ligands include A457 at the C terminus of RXRα helix 12 and A327 in the RXRα ligand-binding pocket
These effects are not only present at the dimer interface
but also extend through the core of the RXRα LBD
these structural regions involve a network of co-evolved amino acids in NRs
which are energetically coupled and mediate allosteric signalling in RXR heterodimers
(a) NMR chemical shift perturbations in PPARγ on heterodimerization with RXRα mapped onto the structure of the PPARγ LBD (PDB 2PRG)
(b) LXR homodimer (PDB 3IPU) coloured grey and superimposed with the LXR promoter from the LXR/RXR heterodimer (PDB 1UHL) coloured green and coral
shows that RXR induces rotation of LXR helix 11
(c) The RXR-induced shift in LXR helix 11 (PDB 1UHL) induces a rotation of LXR helix 5 relative to the LXR homodimer (PDB 3IPU)
allowing W443 in helix 12 to adopt an alternative conformation with greater van der Waals contacts and increased buried surface area
supporting a general role for helix 5 rotation in allosteric control of RXR heterodimers
helix 5 rotation and the evolutionarily conserved SCA network of amino acid residues provide a structural conduit for signalling from the dimer partner
to the ligand-binding pocket and co-regulator-binding surface
most of the structural features for allosteric signal integration have remained a mystery
limited in part by our insufficient structural understanding of signalling within the individual domains
(a) The SCA network residues plotted on RXR using the PPARγ/RXRα heterodimer structure as a model (PDB 1FM6)
(b) Schematic diagram summarizing our TRβ·T3·SRC-2/apo-RXRα crystal structure showing how TR structurally silences RXR
The signal that emanates from TR (i) induces a shift in RXR helix 11 (ii)
leading to a rotation of helix 5 (iii) resulting in structural arrangements that cause RXR helix 12 to adopt an inactive conformation (iv)
(c) Summary of residues affected in the NMR analysis of ligand-selective signalling in PPARγ/RXRα
Helix numbers are indicated for elements of interest
Arrows indicate the flow of the allosteric signal
These regions in general employ the network of co-evolved residues predicted by SCA
PPARγ full agonists appear to cause a more prominent effect
but the specific role of this structural conduit is not clear
It could mean that PPARγ full agonists may provide additional stabilization to the RXRα AF-2 surface
or alter the shape of the AF-2 to give preferences for certain co-activators
Our NMR data further suggests that this interlocking relay system is also modulated by the ligand
as one of the two tryptophan residues in the RXR LBD
was differentially sensitive to PPAR ligands
We thus envision that structural elements in helix 5 of RXR and the dimer partner can move in a coordinated way with the C-terminal region of the helix 11 dimer interface to coordinate both receptor- and ligand-specific signals into an integrated transcriptional response with the co-evolved amino acids playing a primary role
Our data suggest an extension of this model where the position of helix 11 is also controlled by the heterodimer partner helix 11
Our mutagenesis data further suggests that the C terminus of helix 11 is also positioned by the type of dimer partner
contributing to permissive versus non-permissive heterodimer signal integration
15N and 13C′ (carbonyl) NMR chemical shift differences between monomer and heterodimer
and mapped onto the PPARγ LBD crystal structure (PDB 2PRG)
Each HDX experiment was carried out in triplicate and the intensity-weighted average m/z value (centroid) of each peptide isotopic envelope was calculated
Data-dependent tandem mass spectroscopy was performed in the absence of exposure to deuterium for peptide identification in a separate experiment using a 60-min gradient
Peptides with a Mascot score of ≥20 were included in the peptide sets used for HDX
PPREx3-luc (PPAR-luc) or ADH-mSppx3-luc (VDR-luc)
RXR mutant expression plasmids were generated using the Stratagene QuikChange Site-Directed Mutagenesis kit and verified by DNA sequencing
Statistical analyses were performed with Graphpad Prism
Accession codes: Atomic coordinates and structure factors have been deposited in the Protein Data Bank with accession code 4ZO1
Retinoid x receptor heterodimers in the metabolic syndrome
Sensors and signals: a coactivator/corepressor/epigenetic code for integrating signal-dependent programs of transcriptional response
Molecular determinants for the tissue specificity of SERMs
Anti-diabetic drugs inhibit obesity-linked phosphorylation of PPARgamma by Cdk5
The therapeutic potential of nuclear receptor modulators for treatment of metabolic disorders: PPARgamma
DNA binding site sequence directs glucocorticoid receptor structure and activity
DNA binding alters coactivator interaction surfaces of the intact VDR-RXR complex
The glucocorticoid receptor dimer interface allosterically transmits sequence-specific DNA signals
A novel role for helix 12 of retinoid X receptor in regulating repression
Structural determinants of allosteric ligand activation in RXR heterodimers
The nuclear receptor signalling scaffold: insights from full-length structures
Nuclear receptor full-length architectures: confronting myth and illusion with high resolution
Ligand control of coregulator recruitment to nuclear receptors
The structural basis of estrogen receptor/coactivator recognition and the antagonism of this interaction by tamoxifen
Structure and specificity of nuclear receptor-coactivator interactions
Crystal structure of the RAR-gamma ligand-binding domain bound to all-trans retinoic acid
Structure of the intact PPAR-gamma-RXR-alpha nuclear receptor complex on DNA
Structural basis for antagonist-mediated recruitment of nuclear co-repressors by PPARalpha
Molecular basis of agonism and antagonism in the oestrogen receptor
Allosteric control of ligand selectivity between estrogen receptors alpha and beta: implications for other nuclear receptors
Structural characterization of a subtype-selective ligand reveals a novel mode of estrogen receptor antagonism
Coupling of receptor conformation and ligand orientation determine graded activity
NFkappaB selectivity of estrogen receptor ligands revealed by comparative crystallographic analyses
Partial agonists activate PPARgamma using a helix 12 independent mechanism
Antidiabetic actions of a non-agonist PPARgamma ligand blocking Cdk5-mediated phosphorylation
Co-regulator recruitment and the mechanism of retinoic acid receptor synergy
Characterization of the interaction between retinoic acid receptor/retinoid X receptor (RAR/RXR) heterodimers and transcriptional coactivators through structural and fluorescence anisotropy studies
The phantom ligand effect: allosteric control of transcription by the retinoid X receptor
Unique requirements for retinoid-dependent transcriptional activation by the orphan receptor LXR
Interactions controlling the assembly of nuclear-receptor heterodimers and co-activators
Peroxisome proliferator-activated receptors and retinoic acid receptors differentially control the interactions of retinoid X receptor heterodimers with ligands
Unique response pathways are established by allosteric interactions among nuclear hormone receptors
A permissive retinoid X receptor/thyroid hormone receptor heterodimer allows stimulation of prolactin gene transcription by thyroid hormone and 9cRA
Functional evidence for retinoid X receptor (RXR) as a nonsilent partner in the thyroid hormone receptor/RXR heterodimer
Mechanism of the nuclear receptor molecular switch
Dynamics and ligand-induced solvent accessibility changes in human retinoid X receptor homodimer determined by hydrogen deuterium exchange and mass spectrometry
Investigation of ligand interactions with human RXRalpha by hydrogen/deuterium exchange and mass spectrometry
Deuterium exchange and mass spectrometry reveal the interaction differences of two synthetic modulators of RXRalpha LBD
Ligand-specific structural changes in the vitamin D receptor in solution
Molecular recognition of agonist ligands by RXRs
Crystal structure of the heterodimeric complex of LXRalpha and RXRbeta ligand-binding domains in a fully agonistic conformation
Asymmetry in the PPARgamma/RXRalpha crystal structure reveals the molecular basis of heterodimerization among nuclear receptors
Probing protein ligand interactions by automated hydrogen/deuterium exchange mass spectrometry
Structural basis for autorepression of retinoid X receptor by tetramer formation and the AF-2 helix
Effect of heterodimer partner RXRalpha on PPARgamma activation function-2 helix in solution
Heterodimeric interaction between retinoid X receptor alpha and orphan nuclear receptor OR1 reveals dimerization-induced activation as a novel mechanism of nuclear receptor activation
Novel roles of retinoid X receptor (RXR) and RXR ligand in dynamically modulating the activity of the thyroid hormone receptor/RXR heterodimer
Modulation of RXR function through ligand design
Molecular mechanism of allosteric communication in the human PPARalpha-RXRalpha heterodimer
An alternate binding site for PPARγ ligands
Benzoyl 2-methyl indoles as selective PPARgamma modulators
NMRPipe: a multidimensional spectral processing system based on UNIX pipes
PHENIX: building new software for automated crystallographic structure determination
Toward the structural genomics of complexes: crystal structure of a PE/PPE protein complex from Mycobacterium tuberculosis
X-ray structure determination at low resolution
Considerations for the refinement of low-resolution crystal structures
Optimal description of a protein structure in terms of multiple groups undergoing TLS motion
Presenting your structures: the CCP4mg molecular-graphics software
Methods for the analysis of high precision differential hydrogen deuterium exchange data
HDX workbench: software for the analysis of H/D exchange MS data
HD desktop: an integrated platform for the analysis and visualization of H/D exchange data
Download references
Mangelsdorf (the University of Texas Southwestern Medical Center) for discussions related to SCA
Support is acknowledged from an endowed fellowship from the Frenchman’s Creek Women for Cancer Research (S.S.); a fellowship from the Landenberger Foundation (D.P.M.); a NIH Pharmacological Sciences Training Grant (A.I.S); a NIH Medical Scientist Training Program Grant (A.I.S.); a New Investigator Award 1KN-09 from the James and Esther King Biomedical Research Program
Florida Department of Health (D.J.K.); the National Institutes of Health (NIH) National Research Service Award (NRSA) DK097890 (T.S.H.) and Pathway to Independence Award DK103116 (T.S.H.); NIH grants DK101871 (D.J.K.)
RR027270 (P.R.G.) and CA132022 (K.W.N.); and the State of Florida for institutional Scripps Florida start-up funds
Present address: Present address: Division of Pediatric Rheumatology
The Scripps Research Institute-Scripps Florida
University of Texas Southwestern Medical Center
expressed/purified protein and/or performed assays
wrote the manuscript with input from all the authors
Kendall Nettles is a consultant for Genentech
Supplementary Figures 1-8 and Supplementary Tables 1-2 (PDF 593 kb)
Download citation
went to be with our heavenly Father on April 10
A memorial service will be held at the Harvey Anderson & Johnson Funeral Home in Willmar
Visitation will be held one hour prior to the service at the funeral home
in Redwood Falls the daughter of Charles and Dorothy (Sanger) Malecek
received her first Communion and was confirmed at St
Mary's Catholic Church in Bechyn She grew up in Bechyn and attended country primary school and attended Danube High School graduating in the class of 1961
being a very independent individual with a sense of adventure and wanting to get away from the cold Minnesota winters
purchased a used vehicle and headed west by herself to California. Judy had several stops along the way: first in Colorado Springs
At both stops she took waitressing jobs until she had enough money to continue her ultimate destination – California.
William (Bill) Zaccheo. Judy and Bill enjoyed 51 years of married life until Bill passed away in 2016. While in California
Judy attended Pima Community College where she earned an Associate of Science Respiratory Therapist Degree
She worked as a respiratory therapist at hospitals in California
Judy was an excellent seamstress making most of her own clothes. Judy also very much enjoyed making dolls and embroidering pillows and towels as gifts for family and friends. Both Judy and Bill enjoyed traveling and took a cruise to Alaska and sightseeing train trips. However
Judy's biggest joys were being around family and friends. Judy would always have a big smile to greet someone and then she would follow up her smile with her sense of humor
Judy is survived by her siblings: Diane Wohnoutka
Charlene Malecek (special friend John Gilchrist)
and Rick Malecek; brother-in-law: Richard Zaccheo; sisters-in-law: Kathleen Zaccheo and Virginia Zaccheo; plus nieces and nephews
Charles and Dorothy (Sanger) Malecek; siblings: baby
Gordy Malecek; brothers-in-law: John Kojetin
Dominick Zaccheo; and sisters-in-law: Sandy Malecek and Connie Malecek
Blessed be her memory.
Metrics details
Nuclear receptors (NRs) are thought to dynamically alternate between transcriptionally active and repressive conformations
Most NR ligand series exhibit limited bias
primarily consisting of transcriptionally active agonists or neutral antagonists
but not repressive inverse agonists—a limitation that restricts understanding of the functional NR conformational ensemble
we report a NR ligand series for peroxisome proliferator-activated receptor gamma (PPARγ) that spans a pharmacological spectrum from repression (inverse agonism) to activation (agonism) where subtle structural modifications switch compound activity
While crystal structures provide snapshots of the fully repressive state
NMR spectroscopy and conformation-activity relationship analysis reveals that compounds within the series shift the PPARγ conformational ensemble between transcriptionally active and repressive conformations that are natively populated in the apo/ligand-free ensemble
Our findings reveal a molecular framework for minimal chemical modifications that enhance PPARγ inverse agonism and elucidate their influence on the dynamic PPARγ conformational ensemble
studies into the mechanisms of graded NR transcriptional repression have been hampered because most NR ligand series consist of compounds of a single pharmacological type (e.g.
Less common are reports of transcriptionally repressive NR inverse agonists and compounds that span the full pharmacological spectrum
which display distinct coregulator recruitment profiles or ligand bias
these static structures do not inform on the structural mechanism
behind the differential graded activity observed within a ligand series during structure-activity relationship development
as they do not elucidate how compounds with graded activity influence the dynamic PPARγ LBD conformational ensemble
we show that the 2-chloro-5-nitrobenzamide scaffold offers an opportunity to understand how ligands with graded activity across the entire pharmacological spectrum influence the PPARγ LBD conformational ensemble
We found that relatively minimal chemical modifications to this scaffold relative to the parent compound T0070907 yielded a ligand series spanning graded inverse agonism to agonism
Crystal structures of PPARγ LBD bound with ligands and corepressor peptide along with density functional theory (DFT) calculations provide structural insight into the structural basis of improved inverse agonist efficacy
NMR studies reveal that compounds within the ligand series influence the function of PPARγ by shifting the LBD conformational ensemble between transcriptionally active- and repressive-like states that are natively populated in the apo-LBD ensemble
Correlation analysis shows that biochemical activities and NMR structural analysis using purified PPARγ LBD protein can explain
ligand-dependent functions of full-length PPARγ-mediated transcription and gene expression in cells
a AF-2 surface differences between PPARγ LBD in the transcriptionally active conformation bound to agonist rosiglitazone and TRAP220/MED1 coactivator peptide (PDB 6ONJ) and repressive conformation bound to inverse agonist T0070907 and NCoR1 corepressor peptide (PDB 6ONI) peptides
Coactivator and corepressor peptides are shown in green and pink
b Pi stacking between the polar T0070907 pyridyl group and three residues (His323
and Tyr473) and a bridging water molecule form in the transcriptionally repressive conformation (PDB 6ONI)
c Compound scaffold of parent molecules containing polar pyridyl (T0070907) and hydrophobic phenyl (GW9662) groups in relation to the polar benzamide group in ZINC5672437
d TR-FRET NCoR1 corepressor biochemical interaction assay (n = 3 biological replicates; mean ± s.e.m.)
e Cellular luciferase transcriptional reporter assay in HEK293T cells treated with 10 µM compound (n = 4 biological replicates; mean ± s.e.m.)
f Crystal structures showing the compound flipped binding modes in transcriptionally active vs
repressive conformations when bound to GW9662 (PDB 3B0R and 8FHE)
g ZINC5672437 pi-stacking interaction in the repressive state (PDB 8FHE) where the benzamide replaces the bridging water in the T0070907-bound structure (PDB 6ONI)
h 2D [1H,15N]-TROSY-HSQC NMR focused on Gly399 of 15N-labeled PPARγ LBD bound to GW9662
or ZINC5672437 in the absence (solid lines) or presence (dashed lines) of NCoR1 corepressor peptide
i Data-informed design hypothesis to improve PPARγ inverse agonism
the compound analogs have a wide range of graded activities spanning transcriptional repression
with increased corepressor recruitment and binding affinity
with increased coactivator recruitment and binding affinity
Approximately half of the compounds display similar or improved inverse agonism compared to T0070907; of these
compounds 3 (SR33068) and 7 (SR36708) are the most efficacious in repressing aP2 expression in 3T3-L1 cells to levels similar to undifferentiated cells
Several analogs show properties of PPARγ agonism via increased coactivator peptide interaction
and increased transcription and aP2 expression
compared to the orthosteric full agonist rosiglitazone
the activity of compounds 17-20 classify these analogs as partial agonists
Some general features of chemical modifications that confer inverse agonism or partial agonism are apparent
all the improved inverse agonists contain a polar pyridyl aromatic ring
most of which are appended with a polar substitution (cyano
analogs with activities ranked worse than GW9662—the least efficacious inverse agonists and partial agonists— all contain a hydrophobic non-polar phenyl ring
Compounds 16 (SR36705) and 17 (SR33487) have extensions off a similar position of a polar pyridyl ring
indicating extensions at these positions may be unfavorable for inverse agonism
SAR on a larger ligand series would be needed to confirm these observations
Compounds are numbered via the rank order in efficacy in the TR-FRET NCoR1 corepressor peptide interaction assay data shown in Fig. 3.
All of the crystallized compounds contain a pyridyl nitrogen, which in T0070907 is at the fourth position, with additional R1 groups that interact with the aromatic triad residues (His323, His449, Tyr473) via pi-stacking interactions (Fig. 5b)
Compounds 3 and 11 contain a cyano group at the fourth position with a pyridyl nitrogen at the third and second positions
which substitutes for the water that bridges the interaction of T0070907 with the side chain of His323
Compound 2 contains a pyridyl nitrogen at the third position pointing towards the NCoR1 corepressor peptide bound at the AF-2 surface
and a cyano group at the fifth position that points towards helix 11 with no water-bridged interaction with the side chain of His323
Compound 5 contains a pyridyl nitrogen at the third position that points towards the NCoR1 corepressor peptide
but contains a fluorine at the fifth position instead of a cyano group that points towards helix 11
compound 4 contains a pyridyl nitrogen at the fourth position similar to T0070907
which retains the water-bridged interaction with His232
and a methyl substitution at the third position that points towards the NCoR1 corepressor peptide
the ligands could shift the ensemble towards and stabilize only the repressive-like conformation
while agonists would shift the ensemble towards an active-like conformation
we compared 2D [1H,15N]-TROSY-HSQC NMR data of 15N-labeled PPARγ LBD bound to the ligand series
Gly338 (β-sheet in the ligand-binding pocket)
This indicates the entire LBD is sensitive to the ligand-induced graded shift in the conformational ensemble between repressive- and active-like states
our findings demonstrate a platform that can assess
and potentially predict the cellular transcriptional activity of PPARγ compounds ranging from agonism to inverse agonism via biochemical and NMR-detected structural biology studies focused on the PPARγ LBD
T0070907 (CAS 313516-66-4) and GW9662 (CAS 22978-25-2)
Details for the compounds synthesized in this study can be found in Supplementary Methods
Peptides derived from human NCoR1 ID2 (2256-2278; DPASNLGLEDIIRKALMGSFDDK) and human TRAP220/MED1 ID2 (residues 638–656; NTKNHPMLM NLLKDNPAQD) were synthesized by LifeTein with an amidated C-terminus for stability
with or without a N-terminal FITC label and a six-carbon linker (Ahx)
Ligand concentrations for final functional profiling assays described below were chosen based on the observed potency values in biochemical TR-FRET coregulator and cellular transcriptional reporter assays after ~2 h ligand treatment (see Source Data 1)
Because 2-chloro-5-nitrobenzamide compound analogs bind via a covalent mechanism
their potency values will progressively left shift (become more potent) with time
HEK293T (ATCC #CRL-11268) and 3T3-1L (ATCC #CL-173) cells were cultured according to ATCC guidelines
HEK293T cells were grown at 37 °C and 5% CO2 in Dulbecco’s Modified Eagle Medium
Gibco) supplemented with 10% fetal bovine serum (FBS
100 μg/mL of streptomycin (Gibco) until 90–95% confluence in T-75 flasks prior to subculture or use
3T3-L1 cells were grown at 37 °C and 5% CO2 in DMEM
pyruvate (Gibco) supplemented with 10% bovine calf serum (BCS
100 μg/mL of streptomycin (Gibco) until 70% confluence in T-75 flasks prior to subculture or use
was expressed as a TEV-cleavable N-terminal hexa-his-tag fusion protein (6xHis-PPARγ LBD)
Expression was performed in BL21(DE3) Escherichia coli cells in either autoinduction ZY media (unlabeled protein) or using M9 minimal media supplemented with 15N ammonium chloride (for NMR studies)
and 18 °C for 16 h before harvesting by centrifugation (4000 g
cells were grown until the OD600 was 0.6 before adding 0.5 mM (final concentration) isopropyl β-D-thiogalactoside (IPTG) and incubating at 18 °C for 16 h before harvesting by centrifugation (4000 g
Cells were resuspended in lysis buffer (50 mM potassium phosphate (pH 7.4)
10 mM imidazole) and lysed by sonication on ice
Cell lysate was clarified by centrifugation (20,000 g
the protein was purified using Ni-NTA affinity chromatography followed by size exclusion chromatography (Superdex 75) on an AKTA pure in assay buffer (20 mM potassium phosphate
the His-tag was cleaved with TEV protease in dialysis buffer (20 mM potassium phosphate pH 7.4
The cleaved protein was reloaded on the Ni-NTA column
and further purified by size exclusion chromatography (Superdex 75) in assay buffer (20 mM potassium phosphate
Protein was confirmed to be >95% pure by SDS-PAGE
Assays were performed in black 384-well plates (Greiner) with 23 μL final well volume
1 nM LanthaScreen Elite Tb-anti-His Antibody (Thermo Fisher; dilution/amount used per product guidelines)
and 400 nM FITC-labeled NCoR1 ID2 or MED1 ID2 peptide in a buffer containing 20 mM potassium phosphate (pH 7.4)
Ligands were added as a single concentration (10 μM) or in dose-response format to determine EC50/IC50 values
and plates were read using BioTek Synergy Neo multimode plate reader after incubation at 25 °C for 1 h
The Tb donor was excited at 340 nm; the Tb donor emission was measured at 495 nm
ligand concentration; fit to a three-parameter sigmoidal dose-response equation to obtain potency values; and are representative of two or more independent experiments
Assays were performed using 6xHis-PPARγ LBD preincubated with or without 2 molar equivalents of compound at 4 °C overnight and buffer exchanged via centrifugal concentration using Amicon Ultra centrifugal filters to remove excess ligand
Protein samples were serially diluted into a buffer containing 20 mM potassium phosphate (pH 8)
and 0.01% Tween 20 and plated with 180 nM FITC-labeled NCoR1 ID2 or TRAP220/MED1 ID2 peptide in black 384-well plates (Greiner)
and FP was measured on a BioTek Synergy Neo multimode plate reader at 485 nm emission and 528 nm excitation wavelengths
Data were plotted using GraphPad Prism as FP signal in millipolarization units vs
protein concentration (n = 3 biological replicates); fit to a one site—total binding equation using a consistent
fixed Bmax value determined from a fit of the high-affinity interactions as binding for some conditions did not saturate at the highest protein concentration used (45 µM); and are representative of two or more independent experiments
HEK293T cells were cultured in Dulbecco’s minimal essential medium (DMEM
Gibco) supplemented with 10% fetal bovine serum (FBS) and 50 units mL−1 of penicillin
2 million cells were seeded in a 10-cm cell culture dish for transfection using X-tremegene 9 (Roche) and Opti-MEM (Gibco) with full-length human PPARγ isoform 2 expression plasmid (4 μg)
and a luciferase reporter plasmid containing the three copies of the PPAR-binding DNA response element (PPRE) sequence (3xPPRE-luciferase; 4 μg)
cells were treated in quadruplicate with 20 μL of either vehicle control (1.5% DMSO in DMEM media) or 5 μM ligand
or in dose-response format to determine EC50/IC50 values
where each ligand treated condition had separate control wells to account for plate location-based artifacts
Data were plotted in GraphPad Prism as luciferase activity vs
ligand concentration (n = 4 biological replicates); fit to a three-parameter sigmoidal dose-response equation to obtain potency values; and are representative of two or more independent experiments
3T3-L1 cells were cultured in DMEM medium supplemented with 10% FBS and 50 units mL−1 of penicillin
Cells were grown to 70% confluency and then seeded in 12-well dishes at 50,000 cells per well and incubated overnight at 37 °C
cells were treated with media supplemented with 0.5 mM 3-iso-butyl-1-methylxanthine
cells were treated with 10 μM compound in media supplemented with 877 nM insulin for 24 h
RNA was extracted using quick-RNA MiniPrep Kit (Zymo) and used to generate complementary DNA using qScript cDNA synthesis kit (Quantabio)
Expression levels of the PPARγ target gene aP2/FABP4 (forward primer: 5′-AAGGTGAAGAGCATCATAACCCT-3′) and the housekeeping gene TBP (forward primer: 5′-ACCCTTCACCAATGACTCCTATG-3′) used for normalization was measured using Applied Biosystems 7500 Real-Time PCR system
Relative gene expression was calculated via the ddCt method using Applied Biosystems Relative Quantitation Analysis Module Software
which reported values as mean with upper and lower limits
Data were plotted in GraphPad Prism and are representative of two or more independent experiments
Three of the compounds in our ligand series—GW9662
and 11 (SR33486)—and the control agonist rosiglitazone were not available or at the time these experiments were performed and left out of this analysis
Population-weighted 1H NMR chemical shift analysis was performed focusing on the NMR peaks observed for Gly399 in each ligand-bound NMR spectrum
Peak volumes for well-resolved peaks (e.g.
two peaks = two states/conformations in slow exchange) were calculated using an elliptical peak fitting algorithm
Population weighted average 1H NMR chemical shift values were calculated with Python using Jupyter Notebook using NumPy and Pandas packages using an equation (“lambda x: np.average(x.H1_P
weights=x.weighted_vol)”) with two inputs: peak volumes estimated the relative population sizes (weighted_vol) of each state
and the1H NMR chemical shift values of each state (H1_P)
the binding free energy was calculated by subtracting the free energy of the complex from the free energy sum of the ligand and binding site
Correlation data plotting and analysis of Spearman (s) correlation coefficients and two-sided alternative hypothesis p value testing (without adjustments for multiple comparisons) of the biochemical and cellular ligand profiling data were performed in Python using Jupyter Notebook using several libraries including Seaborn
Reported two-sided p values (p) represent the probability that the absolute value of the Spearman or Pearson coefficient of random (x
y) value drawn from the population with zero correlation would be greater than or equal to abs(r or s)
according to the Scipy stats documentation
Data in figure legends are reported as (n = X biological replicates
Further information on research design is available in the Nature Portfolio Reporting Summary linked to this article
A comprehensive map of molecular drug targets
The coregulator exchange in transcriptional functions of nuclear receptors
and allostery in nuclear receptors as transcription factors
A structural mechanism of nuclear receptor biased agonism
Chapter 4—Biased signaling and conformational dynamics in nuclear hormone receptors
Interactions governing transcriptional activity of nuclear receptors
The nuclear receptor superfamily: a structural perspective
The role of dynamic conformational ensembles in biomolecular recognition
Ligand-induced stabilization of PPARgamma monitored by NMR spectroscopy: implications for nuclear receptor activation
A dynamic mechanism of nuclear receptor activation and its perturbation in a human disease
Quantitative structural assessment of graded receptor agonism
Functional consequences of cysteine modification in the ligand binding sites of peroxisome proliferator activated receptors by GW9662
Probing the complex binding modes of the PPARγ partial agonist 2-chloro-N-(3-chloro-4-((5-chlorobenzo[d]thiazol-2-yl)thio)phenyl)-4-(trifluoromethyl)benzenesulfonamide (T2384) to orthosteric and allosteric sites with NMR spectroscopy
Modification of the orthosteric PPARγ covalent antagonist scaffold yields an improved dual-site allosteric inhibitor
Endogenous vitamin E metabolites mediate allosteric PPARγ activation with unprecedented co-regulatory interactions
Identification of a new type of covalent PPARγ agonist using a ligand-linking strategy
A molecular switch regulating transcriptional repression and activation of PPARγ
Biochemical and structural basis for the pharmacological inhibition of nuclear hormone receptor PPARγ by inverse agonists
Discovery and structure-based design of potent covalent PPARγ inverse-agonists BAY-4931 and BAY-0069
Discovery and characterization of orally bioavailable 4-chloro-6-fluoroisophthalamides as covalent PPARG inverse-agonists
Bladder-cancer-associated mutations in RXRA activate peroxisome proliferator-activated receptors to drive urothelial proliferation
Pparg signaling controls bladder cancer subtype and immune exclusion
Genomic activation of PPARG reveals a candidate therapeutic axis in bladder cancer
Evasion of immunosurveillance by genomic alterations of PPARγ/RXRα in bladder cancer
Definition of functionally and structurally distinct repressive states in the nuclear receptor PPARγ
The CoRNR motif controls the recruitment of corepressors by nuclear hormone receptors
The coactivator LXXLL nuclear receptor recognition motif
PPARγ corepression involves alternate ligand conformation and inflation of H12 ensembles
Structural mechanism underlying ligand binding and activation of PPARγ
a first-in-class inverse agonist of the peroxisome proliferator-activated receptor gamma (PPARG) lineage transcription factor
to potentially treat patients with the luminal subtype of advanced urothelial cancer (UC)
Abstract 2802: development of a surrogate tissue pharmacodynamic (PD) assay for clinical use with FX-909
a novel inhibitor of the urothelial luminal lineage transcription factor peroxisome proliferator-activated receptor gamma (PPARG)
GoodVibes: automated thermochemistry for heterogeneous computational chemistry data
Download references
This work was supported in part by the National Institutes of Health (NIH) grants R01DK124870 (D.J.K.) from the National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK) and R35GM146982 (Z.J.Y.) from the National Institute of General Medical Sciences (NIGMS)
The SSRL Structural Molecular Biology Program is supported by the DOE Office of Biological and Environmental Research and by the NIH NIGMS grant P30GM133894
These authors contributed equally: Brian S
Scripps Research and The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology
Department of Chemical and Biomolecular Engineering
expressed and purified protein and solved crystal structures
supervised the research and wrote the manuscript along with B.S
Nature Communications thanks Daniel Merk and the other anonymous reviewer(s) for their contribution to the peer review of this work
Download citation
DOI: https://doi.org/10.1038/s41467-025-57325-4
Metrics details
The nuclear receptor ligand-binding domain (LBD) is a highly dynamic entity
Crystal structures have defined multiple low-energy LBD structural conformations of the activation function-2 (AF-2) co-regulator-binding surface
yet it remains unclear how ligand binding influences the number and population of conformations within the AF-2 structural ensemble
we present a nuclear receptor co-regulator-binding surface structural ensemble in solution
viewed through the lens of fluorine-19 (19F) nuclear magnetic resonance (NMR) and molecular simulations
and the response of this ensemble to ligands
co-regulator peptides and heterodimerization
We correlate the composition of this ensemble with function in peroxisome proliferator-activated receptor-γ (PPARγ) utilizing ligands of diverse efficacy in co-regulator recruitment
While the co-regulator surface of apo PPARγ and partial-agonist-bound PPARγ is characterized by multiple thermodynamically accessible conformations
the full and inverse-agonist-bound PPARγ co-regulator surface is restricted to a few conformations which favor coactivator or corepressor binding
experimental evidence describing this ensemble is lacking and it remains poorly understood how binding of pharmacologically distinct ligands affects the ensemble of co-regulator-binding surface and helix 12 conformations
a direct observation of the ligand-dependent ensemble implied by these data is lacking
This raises the question: are there multiple long-lived conformations that correlate with functional efficacy (e.g.
co-regulator affinity) in nuclear receptors
using 19F NMR combined with biochemical co-regulator interaction analysis and molecular simulations
we define the ligand-dependent conformational ensemble of the co-regulator interaction surface
which controls the transcriptional activity of PPARγ
The data presented here indicate that helix 12 and the co-regulator-binding surface of apo PPARγ and partial-agonist-bound PPARγ is found in a broad energy well with multiple local minima of similar potential energy separated by relatively small kinetic barriers
allowing exchange on the μs to ms time scale
when PPARγ is bound to a full agonist or inverse agonist
helix 12 and the co-regulator-binding surface occupies narrow energy wells with fewer thermodynamically accessible conformations
simulations define some of the probable structures that compose these ensembles
These data better elucidate how ligands induce functional effects via nuclear receptors
the DMSO concentration is constant across the titration both in this figure and all other TR-FRET data presented
Q322C and K502C yielded well-functioning protein with pronounced ligand-inducible changes; we used these mutants to probe the conformational ensemble of the PPARγ co-regulator-binding surface (i.e.
any unlabeled portion of PPARγK502C-BTFA should not affect FP or TR-FRET results
these data indicate a diverse ligand-dependent helix 12 conformational ensemble
Helix 12 solvent exposure is distinct for the active and inactive helix 12 conformations
a 19F NMR spectra of delipidated apo PPARγC313A,K502C-BTFA bound to 0.2 molar equivalent of GW1929 (left)
and T0070907-bound PPARγK502C-BTFA with excess free BTFA
Due to the high binding affinity of GW1929 (4 nM)
all three expected peaks (two apoprotein peaks and one GW1929 peak) are present in slow exchange on the NMR time scale
allowing analysis of the three conformations simultaneously
change 19F NMR chemical shift values for various peaks; a dotted gray line is shown to highlight no D2O-induced change in 19F NMR chemical shift
these data indicate that some inverse agonists do more than simply displace activating lipids
but instead induce a distinct PPARγ LBD state with higher affinity for corepressors than delipidated apoprotein
Agonists reduce the conformational complexity of the co-regulator-binding surface
a Fluorine NMR spectra of PPARγC313A,Q322C-BTFA bound to the indicated ligands
The small sharp left-shifted peak in all the spectra is free BTFA
b Trajectory frame from a simulation of PPARγC313A,Q322C-BTFA bound to a coactivator peptide (MED1; green) with 322C-BTFA shown in orange as spheres (fluorine atoms are turquoise)
A single well-populated 19F NMR peak is observed for BTFA probes placed in two areas of the co-regulator-binding surface (helices 3 and 12) when PPARγ-BTFA is bound to a strong agonist such as GW1929 or rosiglitazone
multiple-well-populated 19F NMR peaks are observed in slow exchange on the NMR time scale indicating that the peaks represent distinct co-regulator-binding surface conformations with lifetimes on the order of milliseconds or longer
Chemical exchange saturation transfer indicates slow exchange between multiple helix 12 conformations
a A selective Gaussian pulse was used to saturate the 19F spectra bound to the indicated ligands at locations indicated by pink circles
The height of the pink circles indicates the height of the peak indicated by the black arrow when the selective pulse was carried out at the chemical shift of the pink circle
If exchange is occurring between the two peaks
b A selective Gaussian pulse was used to saturate the spectrum at an on resonance (a; orange box arrow) and off resonance (a; blue box arrow) location
the peak height of the most abundant resonance (a; black arrow) was monitored as a function of the duration of the saturation pulse
and the resulting peak intensities were fit to extract the exchange rate
Ninety-five percent confidence intervals for the fit of the calculated exchange rates are shown within parentheses
except for ciglitazone which used PPARγC313A,K502C-BTFA for increased signal to noise
b Plot of mean 19F NMR chemical shift values (PPARγK502C-BTFA) vs
and SMRT peptide dissociation constant (Kd) for PPARγK502C-BTFA (top panels) and wt PPARγ LBD (bottom panels) as measured by fluorescence polarization for a subset of the ligands in a
Linear regression fit is shown as a solid line and the correlation coefficient (R2) for the fitted line is indicated
The ligands with the highest efficacy for MED1 and NCoR recruitment for PPARγK502C-BTFA are highlighted
Error bars represent standard deviation of two (K502C-BTFA Kd and wt SMRT Kd) or three (wt MED1 and NCoR Kd) independent experiments
Agonist-bound PPARγ is changed little by MED1 binding
whereas NCoR binding changes the spectrum drastically and vice versa for inverse-agonist (T0070907)-bound PPARγ
*SMRT-induced mean chemical shift in GW1929-bound PPARγK502C-BTFA is the same as NCoR
Heterodimerization of PPARγ LBD with RXRα LBD favors coactivator binding
a 19F NMR of PPARγK502C-BTFA bound to T0070907 and PPARγC313A,502C-BTFA bound to MRL24 or GW1929 or with no ligand bound was performed in the presence (orange) or absence (black) of RXRα LBD
d The 19F NMR signal from the MRL24 ligand
Broadening and consequent reduction in signal intensity is expected as a consequence of the increased rotational correlation time of the heterodimer complex
c TR-FRET was used to measure interaction between wt PPARγ LBD and MED1 or NCoR in the presence or absence of equimolar concentrations of RXRα
Error bars represent standard deviation of two technical replicates within a single experiment
The experiment was repeated twice and gave similar results each time
e Heterodimerization favors MED1 binding (p = 0.0017) and disfavors NCoR binding (p = 0.0076) to apo PPARγ
visual and statistical comparison of NCoR and MED1 recruitment to PPARγ LBD saturated with ligand (four highest concentrations of ligands) indicates that RXRα affects co-regulator recruitment to T0070907-bound PPARγ (NCoR
All p values are derived from a two-tailed t test
e A model for helix 12 conformational diversity based on simulation and experiment
Apo PPARγ or partial-agonist-bound PPARγ helix 12 is found in many similar conformations of varying helical structure producing broad NMR peaks and rapid hydrogen deuterium exchange
while full agonist is found in a tighter cluster of conformations
Co-binding of the inverse agonist T0070907 and the corepressor NCoR produces one main conformation similar to an active conformation
Fuzziness implies intermediate exchange (μs to ms) between the conformations
these data reveal a correlation between ligand efficacy and the prevalence of at least three distinct structural ensembles using a diverse set of 16 pharmacologically distinct PPARγ ligands
it is encouraging that these non-converged simulations qualitatively agree with 19F NMR and provide a glimpse of possible conformations that comprise a portion of the observed 19F NMR spectra; however
quantitative comparison between the 19F NMR spectra and converged simulations remains a future challenge
Addition of NCoR or SMRT shifts the population from cluster 2 to cluster 3
while the mutations and labeling may shift the equilibrium population in the opposite direction toward the antagonist/partial agonist cluster (cluster 2)
it may be that helix 12 of wt PPARγ LBD bound to T0070907 is found in a conformation represented by the inverse agonist cluster (cluster 3; left-shifted narrower peak) to a larger degree than detected by PPARγK502C-BTFA
This work adds detail to how ligands in general control the activity of nuclear receptors
Our data not only confirm that helix 12 and the co-regulator-binding surface exist as a ligand-specific dynamic structural ensemble but also reveal the relative populations of sub-ensembles that comprise the overall structural ensemble and correlate function with this ensemble
Further definition of the conformational ensemble of the entire protein and the kinetics and thermodynamics of exchange between the members of the ensemble will build an accurate model of how ligands produce functional outputs via nuclear receptors and allow greater control of their function via ligands
A pET-46 plasmid carrying the genes for ampicillin resistance and N terminally 6xHis-tagged PPARγ containing a tobacco etch virus (TEV) nuclear inclusion protease recognition site between the His tag and protein of interest was transformed into chemically competent E
Cells were grown in either ZYP-5052 autoinduction media or terrific broth (TB)
Cells grown in TB at 37 °C were induced at an OD600 of approximately 0.8 by the addition of 0.5 mM isopropyl β-d-1-thiogalactopyranoside (IPTG) and the temperature lowered to 22 °C
Induction proceeded for 16 h prior to harvesting
Harvested cells were homogenized into 50 mM phosphate (pH 8.0)
and lysed using a C-5 Emulsiflex high-pressure homogenizer (Avestin)
Lysates were then clarified and passed through two Histrap FF 5 ml columns in series (GE Healthcare)
Protein was eluted using a gradient from 15 to 500 µM imidazole
Fast protein liquid chromatography was performed on either an NGC Scout system (Bio-Rad) or an ÄKTA Start (GE Healthcare)
Eight milligrams of recombinant 6xHis-tagged TEV was added to eluted protein followed by dialysis into 50 mM Tris (pH 8.0)
The protein was again passed through HisTrap FF columns in order to separate cleaved protein from TEV as well as the cleaved 6xHis tag
The cleavage step was only performed on protein which would be used for NMR or FP
but the protein used for TR-FRET did not have the 6xHis tag removed
The protein was then further purified by gel filtration using a HiLoad 16/600 Superdex 200 PG (GE Healthcare)
Size exclusion was performed in 25 mM MOPS (pH 8.0)
Protein was then dialyzed into 25 mM 3-(N-morpholino)propanesulfonic acid (MOPS) (pH 7.4)
Protein purity in excess of 95% was determined by gradient 4–20% sodium dodecyl sulfate-polyacrylamide gel electrophoresis analysis (NuSep)
Protein concentration was determined using ε280 = 12,045 M−1 cm−1
15N-labeled protein was grown in M9 minimal media containing 99% 15NH4Cl (Cambridge Isotope Laboratories) as the sole nitrogen source
cells were grown at 37 °C and 180 rpm until an OD600 of approximately 1.0 was reached
the temperature was dropped to 22 °C for 1 h
protein expression was induced by the addition of 500 µM IPTG during which induction cells remained at 22 °C
Protein expression and purification was then accomplished utilizing the same protocol as outlined above
purified protein was diluted to 0.8 mg ml−1 and batched with Lipidex 1000 (Perkin-Elmer) at an equal volume
This mixture was batched for 1 h at 37 °C and 100 rpm
protein was pulled through a gravity column by syringe
it was found that the speed of elution was important; protein could not remain on the resin at room temperature in excess of 3 min
Two more column volumes of pre-warmed 25 mM MOPS
and 1 mM EDTA were also pulled through in the same manner
Quality of delipidation was then estimated by 19F NMR
and loss of lipid can be most easily detected by a reduction in the peak at −84.1 ppm
When non-delipidated protein is used in this report it is labeled as bound to E
Mutations in PPARγ LBD were generated using the Quikchange Lightning site-directed mutagenesis kit (Agilent) using primers listed in Supplementary Table 6
The presence of expected mutations and absence of spurious mutations was confirmed by Sanger sequencing (Eurofins)
protein was incubated for 30–60 min and then buffer exchanged at least 100x using 10 kDa Amicon Ultra-15 concentrators (Merck Millipore) to remove excess unbound BTFA
and then mixed with appropriate amounts of H2O buffer that had been adjusted to read pH 7.4
ligands were added to the appropriate concentration as usual
The samples which contained only 10% D2O were prepared as usual with the standard addition of 10% buffered D2O to the final sample
Samples of PPARγK502C-BTFA were already loaded with ligand and labeled appropriately with BTFA prior to exchange into deuterated buffers
FP peptide binding assays were performed by plating a mixture of 50 nM peptide with an N-terminal FITC tag
12-point serial dilutions of PPARγ-LBD (wt
PPARγ-LBD and PPARγ ligands were added at a 1:1 ratio
This mixture was added to wells of low-volume 384-well black plates (Grenier Bio-one
catalog number 784076) to a final volume of 16 μL
Peptides were synthesized by Lifetein LLC (Somerset
sequence: NTKNHPMLMNLLKDNPAQD; and the NCoR peptide
sequence: GHSFADPASNLGLEDIIRKALMG (2251–2273)
Other peptides were purchased from ThermoFisher (Waltham
sequence: NTKNHPMLMNLLKDNPAQD (catalog number PV4549); CBP peptide
sequence: AASKHKQLSELLRGGSGSS (catalog number PV4596); and SMRT
sequence: HASTNMGLEAIIRKALMGKYDQW (catalog number PV4424)
All dilutions were made in 25 mM MOPS (pH 7.4)
0.01% fatty-acid-free bovine serum albumin (BSA) (EMD Millipore
Assay titrations were performed in duplicate
Plates were incubated in the dark at room temperature for 2 h before being read on a Synergy H1 microplate reader (BioTek)
FP was measured by excitation at 485 nm/20 nm and emission at 528 nm/20 nm for FITC
Data were fit using nonlinear regression (agonist vs
response – variable slope 4 parameters) in Prism 7.0b
we did two technical replicates and repeated these experiments independently in the lab for once (Fluormone) or 2 or more times FP and TR-FRET
We chose these number of technical replicates and independent experiment replicates based on our experience with the limited variability inherent in these biochemical assays
TR-FRET peptide recruitment assays were performed by plating a mixture of 8 nM 6xHis-PPARγ-LBD (wt
0.9 nM LanthaScreen Elite Tb-anti-His antibody (LifeTechnologies catalog number PV5863)
200 nM peptide (N terminally biotinylated and C terminally amidated)
and 12-point serial dilutions of PPARγ ligands from 50 μM to 1 pM
catalog number 784076) to a final volume of 20 μL
USA) for MED1 peptide; sequence: VSSMAGNTKNHPMLMNLLKDNPAQ; and NCoR peptide
TR-FRET was measured by excitation at 330 nm/80 nm and emission at 620 nm/10 nm for terbium and 665 nm/8 nm for d2
Change in TR-FRET was calculated by 665 nm/620 nm ratio
so as to exclude most effects of the second transition
the 50 μM SR2088 point was not run because we did not have sufficient ligand
PPARγ ligand inhibition constants (Ki) were measured using a protocol adapted from LanthaScreen TR-FRET PPARγ competitive binding assay (Invitrogen
Assay was performed by plating a mixture of 8 nM 6xHis-PPARγ-LBD
2.5 nM LanthaScreen Elite Tb-anti-His antibody
5 nM LanthaScreen Fluormone Pan-PPAR Green (Invitrogen
and 12-point serial dilutions of PPARγ ligands from 50 μM to 140 fM
This mixture was added to wells of low-volume 384-well black plates (Grenier Bio-one) to a final volume of 16 μL
Plates were incubated in the dark for 2 h at room temperature before being read on a Synergy H1 microplate reader (BioTek)
TR-FRET was measured by excitation at 330 nm/80 nm and emission at 495 nm/10 nm for terbium and 520 nm/25 nm for Fluormone
Change in TR-FRET was calculated by 520 nm/495 nm ratio
Nonlinear curve fitting was performed using Prism 7.0b (Graphpad Software Inc.) as described above for the TR-FRET data
including manual exclusion of highest two concentrations for nTZDpa
Thirty of the 1224 total data points for all three proteins (wt
and PPARγC313A,K502C-BTFA) were automatically excluded by Prism in the fits
where IC50 is the concentration of the ligand that produces 50% displacement of the Fluormone tracer
Lo is the concentration of Fluormone in the assay (5 nM)
and Kd is the binding constant of Fluormone to wt or the two BTFA-labeled mutants
and Lb is the concentration of bound Fluormone in the assay with no addition of test ligand
The affinity of Fluormone for the two BTFA-labeled mutant proteins was determined via TR-FRET by titration of Fluormone into each mutant bound to Elite Tb-anti-His antibody
Dissociation constants of Fluormone for wt was measured as 7.9 ± 0.2 for PPARγ LBD
and the variants were measured as 26 ± 3 nM for PPARγK502C-BTFA and 44 ± 4 nM for PPARγC313A,K502C-BTFA and 12 ± 1 nM PPARγC313A
Intermediate exchange effects and field inhomogeneity are likely present in some of these spectra
which will result in inaccuracies in the fitted models; however
the deconvolution method provides an objective view of the possible underlying spectral structure and populations
Two-dimensional [1H,15N]TROSY-HSQC NMR data were obtained using the trosyf3gpphsi19.2 pulse program
Select NMR spectra were replicated in two different ways: (1) Some NMR samples were measured via NMR initially and then days to weeks later to determine if certain parts of the spectrum changed
Any changes would indicate that non-reversible processes contribute to that part of the signal
such as unfolding or degradation of the protein
(2) Some spectra were run twice utilizing protein from the same batch as utilized for the first spectra or from an entirely different protein preparation
All production simulations were carried out using pmemd.cuda or pmemd.cuda.MPI
The best scoring docked binding mode overlaid well with GI262570
In this docked model in the helix 12-interacting region
RESP charges for GW1929 were derived using the RED server and force modification files were generated using GAFF parameters
1FM9 was modified (or not) to incorporate cysteine-BTFA (parameterized as described above) in place of K502 and docked with GW1929 in a similar way to that described above to build PPARγK502C-BTFA and PPARγ bound to GW1929
1PRG chain B was used to create the build for apo and 3BOR chain B was used to build T0070907-bound PPARγ LBD
3BOR contains GW9662 which differs from T0070907 by one atom
T0070907 has a nitrogen in place of a carbon atom in one of the ligand rings
This change was made in chimera and the ligand parameterized and incorporated into the structure as described above for BTFA
NCoR (same sequence as used in NMR and TR-FRET including N-terminal acetylation and C-terminal amidation) was added to these builds utilizing chimera using the following procedure
The core helix structure from NCoR (from 2OVM) was aligned to SMRT on a PPARα SMRT structure (1KKQ)
and then apo PPARγ chain B (1PRG) was aligned to PPARα and the PPARα/SMRT structure deleted leaving the aligned NCoR on apo PPARγ chain B
A similar procedure was used with the 3BOR structure to create T0070907 co-bound with NCoR on PPARγ
These PDBs were then used to create the final solvated
and equilibrated structure in a manner similar to that described above
and FP are publically available at https://osf.io/rqdpz/
Any other datasets generated during and/or analyzed during the current study are available from the corresponding author on reasonable request
Binding of ligands and activation of transcription by nuclear receptors
The structural basis of estrogen receptor/co-activator recognition and the antagonism of this interaction by tamoxifen
Crystal structure of the ligand-binding domain of the human nuclear receptor RXR-alpha
Nuclear receptor structure: implications for function
Distinct properties and advantages of a novel peroxisome proliferator-activated protein [gamma] selective modulator
(19)F-modified proteins and (19)F-containing ligands as tools in solution NMR studies of protein interactions
The role of ligands on the equilibria between functional states of a G protein-coupled receptor
The dynamic process of β2-adrenergic receptor activation
Structural insights into the dynamic process of β2-adrenergic receptor signaling
Biased signaling pathways in β2-adrenergic receptor characterized by 19F-NMR
Current applications of 19F NMR to studies of protein structure and dynamics
An antidiabetic thiazolidinedione is a high affinity ligand for peroxisome proliferator-activated receptor gamma (PPAR gamma)
A novel N-aryl tyrosine activator of peroxisome proliferator-activated receptor-γ reverses the diabetic phenotype of the Zucker diabetic fatty rat
a selective ligand for peroxisome proliferator-activated receptor-γ functions as an antagonist of biochemical and cellular activities
Medium chain fatty acids are selective peroxisome proliferator activated receptor (PPAR) gamma activators and Pan-PPAR partial agonists
Deconvolution of complex 1D NMR spectra using objective model selection
Tryptophan solvent exposure in folded and unfolded states of an SH3 domain by 19F and 1H NMR
Partial agonists activate PPARγ using a Helix 12 independent mechanism
19F NMR studies of solvent exposure and peptide binding to an SH3 domain
Mechanistic elucidation guided by covalent inhibitors for the development of anti-diabetic PPARγ ligands
Genome-wide profiling of PPARγ:RXR and RNA polymerase II occupancy reveals temporal activation of distinct metabolic pathways and changes in RXR dimer composition during adipogenesis
The shear viscosities of common water models by non-equilibrium molecular dynamics simulations
Conformational diversity of the helix 12 of the ligand binding domain of PPARγ and functional implications
Molecular determinants of nuclear receptor–corepressor interaction
Activation of the A2A adenosine G-protein-coupled receptor by conformational selection
Allosteric nanobodies reveal the dynamic range and diverse mechanisms of G-protein-coupled receptor activation
Use of glass electrodes to measure acidities in deuterium oxide
Detecting outliers when fitting data with nonlinear regression—a new method based on robust nonlinear regression and the false discovery rate
Colloidal drug formulations can explain “bell-shaped” concentration–response curves
Active Pin1 is a key target of all-trans retinoic acid in acute promyelocytic leukemia and breast cancer
Pharmacologic Analysis of Drug/Receptor Interaction 2nd edn (Raven
Simulation of NMR pulse sequences during equilibrium and non-equilibrium chemical exchange
in Introduction to Protein Structure Predictiction: Methods and Algorithms (eds Rangwala
Comparative protein modelling by satisfaction of spatial restraints
H++ 3.0: automating pK prediction and the preparation of biomolecular structures for atomistic molecular modeling and simulations
Server: A web service for deriving RESP and ESP charges and building force field libraries for new molecules and molecular fragments
Application of RESP charges To calculate conformational energies
Development and testing of a general amber force field
ff14SB: improving the accuracy of protein side chain and backbone parameters from ff99SB
Comparison of simple potential functions for simulating liquid water
Determination of alkali and halide monovalent ion parameters for use in explicitly solvated biomolecular simulations
PTRAJ and CPPTRAJ: software for processing and analysis of molecular dynamics trajectory data
AutoDock Vina: improving the speed and accuracy of docking with a new scoring function
Numerical integration of the Cartesian equations of motion of a system with constraints: molecular dynamics of n-alkanes
Download references
We thank James Aramini at the City University of New York Advanced Science Research Center (CUNY ASRC) for assistance in setting up the saturation transfer difference
Roe (NIH Laboratory of Computational Biology
NHLBI) for providing the minimization and equilibration script and simulation advice
NMR data presented herein were collected at the CUNY ASRC Biomolecular NMR Facility
This work was supported in part by National Institutes of Health (NIH) grants K99DK103116 (to T.S.H.)
and DK105825 (to P.R.G.); and National Science Foundation (NSF) award 1359369 (PI Karbstein) that funds the SURF program at Scripps Florida
These authors contributed equally: Michelle D
Biochemistry and Biophysics Graduate Program
Center for Biomolecular Structure and Dynamics
Department of Biomedical and Pharmaceutical Sciences
Summer Undergraduate Research Fellows (SURF) Program
Department of Pharmacology & Physiology
conceived the study and designed experiments
prepared samples and performed experiments
prepared samples and performed preliminary experiments
and characterization of ligand pharmacology
with molecular dynamics simulations and ligand parameterization
analyzed data and wrote the manuscript with input from all authors
Publisher's note: Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations
Download citation
DOI: https://doi.org/10.1038/s41467-018-04176-x
Metrics details
Small chemical modifications can have significant effects on ligand efficacy and receptor activity
but the underlying structural mechanisms can be difficult to predict from static crystal structures alone
Here we show how a simple phenyl-to-pyridyl substitution between two common covalent orthosteric ligands targeting peroxisome proliferator-activated receptor (PPAR) gamma converts a transcriptionally neutral antagonist (GW9662) into a repressive inverse agonist (T0070907) relative to basal cellular activity
and mutagenesis coupled to activity assays reveal a water-mediated hydrogen bond network linking the T0070907 pyridyl group to Arg288 that is essential for corepressor-selective inverse agonism
NMR spectroscopy reveals that PPARγ exchanges between two long-lived conformations when bound to T0070907 but not GW9662
including a conformation that prepopulates a corepressor-bound state
priming PPARγ for high affinity corepressor binding
Our findings demonstrate that ligand engagement of Arg288 may provide routes for developing corepressor-selective repressive PPARγ ligands
and it remains poorly understood how to design inverse agonists
Crystal structures of PPARγ bound to T0070907 or GW9662 reveal no major overall structural differences that explain the difference in efficacy
a water-mediated hydrogen bond network that uniquely links R288 to the T0070907 pyridyl group—an interaction that cannot occur with GW9662
which lacks a hydrogen bond acceptor—is essential for corepressor-selective cellular repression of PPARγ
NMR analysis shows that T0070907-bound PPARγ populates two long-lived structural conformations
one of which resembles the state populated by GW9662 and a unique state that is similar to the corepressor-bound state
thus revealing a structural mechanism for directing corepressor-selective repression of PPARγ
a Chemical structures of GW9662 and T0070907
b Cell-based full-length PPARγ luciferase transcriptional assay showing the effect of activating
and repressive PPARγ ligands (5 µM) on full-length PPARγ transcription in HEK293T cells
Individual points (n = 12) normalized to DMSO control (mean) are plotted as white circles on top of a box-and-whiskers plot; the box represents 25th
and the whiskers plot the entire range of values
c Relative quantitation of fold change in PPARγ target gene expression determined by qPCR normalized to TBP expression of 3T3-L1 cells treated with transcriptionally activating
plotted as relative log2 of data calculated using the 2-ΔΔCt method (n = 3) with error bars representative of the upper and lower limits
Data are representative of at least 3 independent experiments
Coregulator binding profiles of the activating
a Fluorescence polarization coregulator interaction assay showing the effect of ligands on the interaction between the PPARγ LBD and peptides derived from the TRAP220 coactivator or NCoR1 corepressor
plotted as the mean (n = 2) with error bars representative of s.d
b Kd values derived from the coregulator interaction assay
Dotted orange and purple lines note the DMSO/apo-PPARγ values
Orange and purple shaded areas note the affinity regions for an ideal inverse agonist (i.e.
orange circles in the orange square; for higher NCoR1 affinity
c Superposition of crystal structures of the PPARγ LBD bound to GW9662 (PDB code 3B0R; ligand in green
cartoon in blue) and SR1664 (PDB code 4R2U; ligand in yellow)
The location of the simple substitution between GW9662 (methine) vs
T0070907 (nitrogen) is marked with a black circle
and the SR2595 and SR10221 tert-butyl extension within the helix 12 subpocket towards F282 (blue) from the SR1664 parent compound is marked with a yellow circle
suggesting the corepressor-selective inverse agonist transcriptionally repressive profile of T0070907 may originate from a previously uncharacterized structural mechanism
A water-mediated hydrogen bond network connects Arg288 to T0070907
a Overall structure of T0070907-bound PPARγ LD (PDB code 6C1I; gray) and overlay with GW9662-bound crystal structure (PDB code 3B0R; blue)
b A water-mediated hydrogen bond network in the T0070907-bound crystal structure (chain B is shown) links the pyridyl group in T0070907 to the R288 side chain
which forms a bipartite hydrogen bond with the E295 side chain
c The GW9662-bound crystal structure (chain B is shown) lacks the R288-ligand hydrogen bond network but contains the R288-E295 hydrogen bond
d 2D strip corresponding to the R288 Nε-Hε group from a 3D 15N-NOESY-HSQC NMR experiment (τmix = 100 ms) of T0070907-bound 15N-labeled PPARγ LBD reveals a NOE signal at 4.77 p.p.m
e Pyridyl-water network hydrogen bonds populated during molecular dynamics simulations for T0070907- and GW9662-bound structures starting from crystallized (xtal) and modeled (model) conformations from 3–4 replicate simulations
as well as a model generated of T0070907-bound PPARγ from the GW9662-bound PPARγ crystal structure that was independently solvated without the crystallized waters (model)
the pyridyl group of T0070907 was hydrogen bonded to a water molecule for a significant fraction of the simulation (65–95%)
as was the water-bridged R288-T0070907 pyridyl (5–46%)
a direct interaction between R288 and the pyridyl group of T0070907 not mediated by water was lowly populated (<2%)
A direct (4–64%) and water-bridged (3–61%) R288-E295 interaction was also confirmed
The extensive pyridyl-based water-mediated hydrogen bond network is not possible to the hydrophobic phenyl group of GW9662
revealing a unique chemical feature in T0070907 that could confer corepressor-selective inverse agonism
Mutagenesis validates the R288-pyridyl interaction for conferring corepressor-selective inverse agonism
a HEK293T cells transfected with full-length PPARγ expression plasmid along with a 3 × -PPRE-luciferase reporter plasmid and treated with the indicated ligands (5 µM)
Individual points (n = 6) normalized to DMSO control (mean) are plotted on top of a box-and-whiskers plot; the box represents 25th
c Affinities determined from a fluorescence polarization assay of wild-type and mutant PPARγ LBDs preincubated with a covalent ligand (GW9662 or T0070907) or vehicle (DMSO) binding to FITC-labeled b TRAP220 or c NCoR1
Data plotted as the Kd value and error from fitting data of two experimental replicates using a one site binding equation
d Legend to the web of efficacy radar chart diagrams
Corepressor-selective inverse agonism is associated with data points populating the periphery of the plots
e Radar web of efficacy plots displaying assay data for wild-type PPARγ and mutant variants
Data normalized within the range of values for each assay (a–c)
Consistent with the cell-based transcription assay
the R288K and E295A mutants maintained the coregulator binding profile of T0070907 and rank ordering
the R288A and R288L mutants showed similar affinity for TRAP220 and NCoR1 when covalently bound to T0070907 or GW9662
This indicates the pyridyl-water network directs the corepressor-selective inverse agonism profile of T0070907
the lack of which results in an antagonist GW9662-like profile
This dramatic result reveals that R288-mediated pyridyl-water network directs the corepressor-selective inverse agonism profile conferred by T0070907
We therefore used NMR spectroscopy to assess the impact of T0070907 and GW9662 on the dynamics of the PPARγ LBD
NMR detected exchange between two long-lived T0070907-bound conformations
a Overlay of 2D [1H,15N]-TROSY-HSQC NMR spectra of 15N-labeled PPARγ LBD bound to GW9662 or T0070907
b Binding of T0070907 but not GW9662 stabilizes intermediate exchange (µs-ms time scale) dynamics (residues labeled in a shown in green spheres) and causes peak doubling (tan and pink spheres; G399 is colored pink for emphasis)
Data plotted on the T0070907-bound PPARγ crystal structure and important structural regions are highlighted as follows: AF-2 surface (black oval); an extended pyridyl-water hydrogen bond network (blue spheres
beyond the key pyridyl-water interaction (red sphere)
c Snapshot overlays of 2D [1H,15N]-TROSY-HSQC spectra of 15N-labeled PPARγ LBD bound to T0070907 or GW9662 shows co-linear shifting of peaks at the different temperatures
The spectral region displayed shows peaks conserved when PPARγ is bound to GW9662 or T0070907 (green arrows); a unique set of doubled peaks when bound to T0070907 (purple arrows); and absent peaks due to intermediate exchange on the NMR time scale when bound to GW9662 (dotted rectangles)
d Snapshots of ZZ-exchange [1H,15N]-HSQC NMR spectra (delay = 1 s) of T0070907-bound 15N-labeled PPARγ LBD focused on G399 at the indicated temperatures
Two G399 conformational states are denoted as A and B with the ZZ-exchange transfer crosspeaks as A–B and B–A
e ZZ-exchange NMR analysis build-up curve from for G399 at 310 K generated by plotting peak intensities of the state A and B peaks and exchange crosspeaks (A–B and B-A) as a function of delay time
f Schematic of the T0070907-bound PPARγ conformational ensemble defined by the NMR studies
including the β-sheet (G338) and helix 6 (R350
L356) within the ligand-binding pocket; a surface comprising helix 2a (R234) and the C-terminal region of helix 7 and the loop connecting helix 7 and 8 (K373
D380); helix 3 near the AF-2 surface (I303); and the loop connecting helix 8 and 9 near the AF-2 surface (S394)
the peak for G321 in the GW9662-bound conformation and one of the T0070907-bound conformations shows significant line broadening at the lower temperature
indicating these states may share conformational and dynamical features
T0070907 populates a shared conformation with GW9662 and a unique conformation
which is connected to the AF-2 coregulator interaction surface via water-mediated hydrogen bonds to N312 and D313 but does not directly interact with a coregulator peptide bound to the PPARγ LBD (PDB code 2PRG)
b Snapshot overlay of [1H,15N]-TROSY-HSQC NMR spectra of 15N-labeled PPARγ LBD (wild-type or R288 mutants) bound to GW9662 or T0070907 shows that the single GW9662-bound G399 peak has similar chemical shift values to one of the two T0070907-bound G399 peaks (state A) whereas state B is uniquely populated by T0070907
but not T0070907-bound R288A mutant PPARγ LBD
significantly populates the unique conformation
c Deconvoluted 19F NMR spectra of PPARγ LBD labeled with BTFA on helix 12 and covalently bound to GW9662 or T0070907
the 19F spectral profile of GW9662-bound PPARγ revealed two peaks corresponding to a major state (right peak; 78%) and minor state (left peak; 22%)
T0070907-bound PPARγ also showed two peaks with chemical shift values similar to GW9662-bound PPARγ
but the population magnitudes of the states are switched and skewed towards the left peak
this helix 12/AF-2 surface 19F NMR probe showed similar relative population sizes to that observed in the G399 ZZ-exchange analysis (34% and 66%
The right 19F NMR peak abundantly populated by GW9662 and moderately populated by T0070907 likely corresponds to the mutual G399 conformation from the 2D NMR analysis
the left 19F NMR peak likely corresponds to the unique G399 conformation; this peak is abundantly populated by T0070907 but lowly populated by GW9662
The low abundance of this peak when bound to GW9662 could explain in part why it was not detected by the 2D NMR analysis
which has lower overall sensitivity of signal-to-noise compared to the 19F NMR analysis
the BTFA probe attached to helix 12 may also be more sensitive to larger structural changes compared to backbone amide resonances
Coregulator binding preferences of the unique and mutual T0070907-bound conformations
a Snapshot overlay of 2D [1H,15N]-TROSY-HSQC spectra of T0070907-bound 15N-labeled PPARγ LBD titrated with NCoR1 peptide
b Extracted 1D planes of the NCoR1 spectra shown in a show the qualitative binding trends observed by NMR
whereas c an overlay of the extracted 1D planes is quantitatively described by d spectra using a 4-state model where the two slowly exchanging receptor populations (R and R*) are capable of binding to the peptide (RP and R*P)
and receptor isomerizes in both the free and peptide-bound states (R ↔ R*
(e–h) Analysis of performed for T0070907-bound 15N-labeled PPARγ LBD titrated with TRAP220 peptide performed similarly to the NCoR1 analysis described in a–d
which is also quantitatively described using the same 4-state model
Coregulator binding preferences of the single GW9662-bound conformation detected by NMR
a Snapshot overlay of 2D [1H,15N]-TROSY-HSQC spectra of GW9662-bound 15N-labeled PPARγ LBD titrated with NCoR1 peptide
b The 15N spectral planes of the 2D NMR titration are quantitatively described using a 3-state model (R + P ↔ RP ↔ R*P) where PPARγ LBD bound to GW9662 and NCoR1 slowly isomerizes between two states (RP and R*P)
c Snapshot overlay of 2D [1H,15N]-TROSY-HSQC spectra of GW9662-bound 15N-labeled PPARγ LBD titrated with TRAP220 peptide
d The 15N spectral planes of the 2D NMR titration are quantitatively described using a 2-state model (R + P ↔ RP)
These results reveal that the two long-lived T0070907-bound conformations have different binding preferences for NCoR1 and TRAP220
The NMR chemical shifts of the unique and mutual T0070907-bound conformations in the absence of coregulator peptide are similar to the NCoR1- and TRAP220-bound forms
the unique and mutual T0070907-bound states prepopulate a corepressor-like and coactivator-like bound conformation that afford high-affinity binding to NCoR1 and TRAP220
the chemical shift difference between the unique T0070907 conformation and NCoR1-bound state (i.e.
the degree of state B shifting) is much smaller than the mutual conformation and TRAP220-bound state (i.e.
indicating that the corepressor-like conformation prepopulated by T0070907 is more similar to the corepressor-bound state
NCoR1 binding to GW9662-bound PPARγ introduces a conformational frustration within the AF-2 surface: the AF-2 surface of GW9662-bound PPARγ does not prepopulate the corepressor-bound conformation and upon binding NCoR1is found in two slowly exchanging conformations similar to the corepressor- and coactivator-bound forms of T0070907-bound PPARγ
in addition to their utility as nondissociative orthosteric competitive ligands that inhibit binding of other orthosteric PPARγ ligands
our data suggest that T00709707 and GW9662 can be classified as a covalent corepressor-selective inverse agonist and a covalent antagonist
our work shows that the combination of different but complementary structural methods provides the full picture of ligand mechanism of action
Published findings and the data presented here indicate that PPARγ possesses constitutive activity and that binding of ligands differentially influences PPARγ activity
if indeed PPARγ lacks constitutive activity
our results should only require a minor change in nomenclature
as the corepressor-selective activity of T0070907 is well supported by structural and functional evidence provided by us and by others
our findings should inspire future work to develop and characterize corepressor-selective inverse agonists to probe the repressive functions of PPARγ
Human PPARγ LBD (residues 203–477 in isoform 1 numbering
which is commonly used in published structural studies and thus throughout this manuscript; or residues 231–505 in isoform 2 numbering)
and full-length retinoid x receptor α (RXRα) were expressed in Escherichia coli BL21(DE3) cells using enriched media (LB or autoinduction) or for NMR studies minimal media (M9 supplemented with 13C-glucose and/or 15NH4Cl) as TEV-cleavable hexahistidine-tagged fusion protein using a pET46 plasmid
cells were lysed in lysis buffer (500 mM potassium chloride
pH 7.5) supplemented with 5 µg/mL of DnaseI and lysozyme
Lysates were cleared by sonication (24,000 × g
1 h) and loaded onto 2 × 5 mL Histrap FF columns (GE Healthcare)
Protein was eluted using lysis buffer with 500 mM imidazole
protein was incubated at a 1:50 ratio with TEV protease overnight at 4 °C
loaded back onto the HisTrap FF column and collecting the flow through
Protein was concentrated and loaded onto a Superdex 200 prep grade 26/60 column (GE Healthcare)
The final storage buffer for LBD samples following size exclusion chromatography and subsequently frozen at −80 °C was 50 mM potassium chloride (pH 7.4)
and 0.5 mM EDTA; for full-length PPARγ was 25 mM MOPS (pH 7.4)
and 1 mM EDTA; or for full-length RXRα ws 25 mM MOPS (pH 7.4)
In most studies of covalent ligands (except TR-FRET studies)
PPARγ protein was pretreated with GW9662 or T0070907 overnight at 4 °C with a 2X molar excess of compound dissolved in d6-DMSO
Delipidation was performed using Lipidex 1000 resin (Perkin-Elmer): the protein was diluted to 0.8 mg ml−1
batched with an identical volume of resin at 37 °C and spun at 100 rpm for 45 min
and concentrated to a working concentration in 25 mM MOPS (pH 7.4)
and was either frozen immediately at −80 °C or used in the same day
Mammalian expression plasmids included Gal4-PPARγ-hinge-LBD (residues 185-477 in isoform 1 numbering; 213-505 in isoform 2 numbering) inserted in pBIND plasmid; and full-length PPARγ (residues 1-505; isoform 2) inserted in pCMV6-XL4 plasmid
Mutant proteins were generated using site directed mutagenesis of the aforementioned plasmids using primers listed in Supplementary Table 2
Peptides: LXXLL-containing motifs from TRAP220 (residues 638–656; NTKNHPMLMNLLKDNPAQD) and NCoR1 (2256–2278; DPASNLGLEDIIRKALMGSFDDK)
without a FITC-label or containing a N-terminal FITC label with a six-carbon linker (Ahx)
or NCoR1 (2251-2273; GHSFADPASNLGLEDIIRKALMG) containing an N-terminal biotin label and an amidated C-terminus for stability
Ligand (5 µM) or vehicle control was added (20 μL)
cells incubated for 18 hr and harvested for luciferase activity quantified using Britelite Plus (Perkin Elmer; 20 μL) or cell viability was tested using Celltiter-glo (Promega: 20 µL) on a Synergy Neo multimode plate reader (BioTek)
Data were analyzed using GraphPad Prism (luciferase activity vs
ligand concentration) and fit to a sigmoidal dose response curve
For western blot analysis of protein levels
HEK293T cells were transfected as described above
250,000 cells were transferred to 6-well plates (Corning)
Transfected cells were then lysed in TNT buffer (150 mM NaCl
Protein concentration was determined by BCA assay (Thermo Fisher)
and 20 µg of protein was loaded onto 4–15% gradient gels (Bio-Rad) and wet transferred onto PVDF
The membrane was blocked for 1 h at room temperature with Odyssey blocking buffer (Li-COR)
the membrane was incubated overnight at 4 °C with primary antibodies diluted in Odyssey blocking buffer: 1:1000 rabbit anti-PPARγ (Cell signaling technology; catalog #2443 S
1:1000 mouse anti-actin (EMD Millipore; catalog #MAB1501
The following day the blot was washed with PBST and treated with secondary antibodies (Li-COR; donkey anti-mouse-IgG IRDye 680LT
lot # C71201-15; goat anti-rabbit-IgG IR800CW
lot # C80546-08) at 1:2000 dilution in Odyssey blocking buffer for 1 hr at RT
The blot was then washed with PBST and visualized via multiplexed detection using the Odyssey 9120 infrared imaging system (Li-COR)
The assay was performed in black 384-well plates (Greiner) in assay buffer (20 mM potassium phosphate
His-PPARγ LBD was pre-incubated with or without a 2X molar excess of covalent ligand overnight at 4 °C
Noncovalent compounds were incubated with a constant concentration of 90 µM
equivalent to the maximum protein concentration
FITC-labeled NCoR1 and TRAP220 peptides were plated at a final concentration of 100 nM
Plates were incubated for 2 h at 4 °C and measured on a Synergy Neo multimode plate reader (BioTek) exciting at 495 nm and reading at 528 nm wavelengths
Data were plotted using GraphPad Prism and fit to one-site binding equation
We observed no significant changes in coregulator affinity when using native or delipidated protein
indicating any bacterial lipids retained during protein purification are bound substoichiometrically
FITC-labeled NCoR1 peptide was plated to a final concentration of 50 nM in wells to which was added delipidated full-length PPARγ loaded with a stoichiometric amount of ligand (protein concentration ranged from 50 µM to 24 nM by a 12 point 2-fold dilution) in a buffer containing 25 mM MOPS (pH 7.4)
0.01% fatty acid-free bovine serum albumin (BSA) (Millipore)
Plates were incubated in the dark for 2 h at room temperature and measured on a Synergy H1 microplate reader (BioTek) exciting at 495 nm and reading at 528 nm wavelengths
LanthaScreen® Elite Tb-anti-HIS Antibody (ThermoFisher; catalog #PV5863
and FITC-labeled NCoR1 peptide was prepared in assay buffer (20 mM potassium phosphate
Ligands were prepared as a 2X ligand stock
initially prepared via serial dilution in DMSO prior to addition to buffer
Equal volumes of the protein mixture and ligand were added to black 384 well plates (Greiner) for final concentrations of 4 nM protein
Final concentration of DMSO (vehicle) was constant in all wells at 0.5%
Plates were incubated for 2 h at 4 °C and measured on a Synergy Neo multimode plate reader (BioTek)
Data were plotted using GraphPad Prism (TR-FRET ratio 520 nm/495 nm vs
ligand concentration) and fit to sigmoidal dose response equation
Delipidated His-full-length PPARγ/full-length RXRα were plated at 31 nM with 0.9 nM LanthaScreen® Elite Tb-anti-HIS Antibody
and 12-point serial dilutions of T0070907 from 50 μM to 1 pM to a final volume of 16 μL
Plates were incubated for 4 h at room temperature and measured on Synergy H1 microplate reader (BioTek) at 620 nm for terbium and 665 nm for d2
a 1.25x molar excess of dsDNA of the Sult2A1 PPRE (5′-GTA AAA TAG GTG AAA GGT AA-3′; and its reverse complement) was added to each well
and the plate was incubated for 3 h prior to a subsequent reading
Data were plotted using GraphPad Prism (TR-FRET ratio 665 nm/620 nm vs
ligand concentration) and fit to a sigmoidal dose response equation
was used to simulate 1D NMR line shapes using MATLAB software (version R2018a) from the peptide titration experiments for G399 (15N planes vs
extracted 15N planes from 2D NMR data) using 2-state (U)
All simulations were performed using the 4-state model MATLAB script
parts of the 4-state model were turned off as described in the LineShapeKin manual
The 4-state simulations were performed considering the exchange rates and molar fractions of the two slowly exchanging PPARγ populations from the ZZ-exchange analysis
Scripts or non-commercially available programs used for the analysis in this study are publicly available, including MATLAB scripts used for the ZZ exchange NMR lineshape analyses [https://osf.io/4fmkd/], MATLAB from the LinShapeKin package [http://lineshapekin.net], and decon1d [https://github.com/hughests/decon1d]
The crystal structure of T0070907-bound PPARγ LBD is available in the Protein Data Bank (PDB accession code 6C1I). NMR chemical shift assignments used in our analysis were obtained from the Biological Magnetic Resonance Data Bank (BMRB accession codes 17975, 17976, 17977)
Thiazolidinediones and the promise of insulin sensitization in type 2 diabetes
A novel non-agonist peroxisome proliferator-activated receptor gamma (PPARgamma) ligand UHC1 blocks PPARgamma phosphorylation by cyclin-dependent kinase 5 (CDK5) and improves insulin sensitivity
PPARG post-translational modifications regulate bone formation and bone resorption
Pharmacological repression of PPARgamma promotes osteogenesis
Sirt1 promotes fat mobilization in white adipocytes by repressing PPAR-gamma
Antagonism of peroxisome proliferator-activated receptor gamma prevents high-fat diet-induced obesity in vivo
The PPARgamma antagonist T0070907 suppresses breast cancer cell proliferation and motility via both PPARgamma-dependent and -independent mechanisms
Potential of peroxisome proliferator-activated receptor gamma antagonist compounds as therapeutic agents for a wide range of cancer types
Inhibition of peroxisome proliferator-activated receptor gamma activity suppresses pancreatic cancer cell motility
Defining a conformational ensemble that directs activation of PPARgamma
Insights into the mechanism of partial agonism: crystal structures of the peroxisome proliferator-activated receptor gamma ligand-binding domain in the complex with two enantiomeric ligands
Crystal structure of the peroxisome proliferator-activated receptor gamma (PPARgamma) ligand binding domain complexed with a novel partial agonist: a new region of the hydrophobic pocket could be exploited for drug design
Co-crystal structure guided array synthesis of PPARgamma inverse agonists
A functional peroxisome proliferator-activated receptor-gamma ligand-binding domain is not required for adipogenesis
Transcription coactivator TRAP220 is required for PPAR gamma 2-stimulated adipogenesis
Medium chain fatty acids are selective peroxisome proliferator activated receptor (PPAR) gamma activators and pan-PPAR partial agonists
Accurate quantitation of water-amide proton exchange rates using the phase-modulated CLEAN chemical EXchange (CLEANEX-PM) approach with a Fast-HSQC (FHSQC) detection scheme
Ligand-directed signalling at beta-adrenoceptors
New concepts in pharmacological efficacy at 7TM receptors: IUPHAR review 2
An introduction to NMR-based approaches for measuring protein dynamics
NMR line shapes and multi-state binding equilibria
The necessary nitrogen atom: a versatile high-impact design element for multiparameter optimization
Subtleties in GPCR drug discovery: a medicinal chemistry perspective
Tactical approaches to interconverting GPCR agonists and antagonists
Modification of the orthosteric PPARgamma covalent antagonist scaffold yields an improved dual-site allosteric inhibitor
Probing the Complex Binding Modes of the PPARgamma partial agonist 2-chloro-N-(3-chloro-4-((5-chlorobenzo[d]thiazol-2-yl)thio)phenyl)-4-(trifluorome thyl)benzenesulfonamide (T2384) to orthosteric and allosteric sites with NMR spectroscopy
PPARgamma helix 12 exhibits an antagonist conformation
Zheng, J. et al. Chemical crosslinking mass spectrometry reveals the conformational landscape of the activation helix of PPARgamma; a model for ligand-dependent antagonism. Structure, in press, https://doi.org/10.1016/j.str.2018.07.007 (2018)
PPARgamma in complex with an antagonist and inverse agonist: a tumble and trap mechanism of the activation helix
Crystal structure of the ligand binding domain of the human nuclear receptor PPARgamma
The differential interactions of peroxisome proliferator-activated receptor gamma ligands with Tyr473 is a physical basis for their unique biological activities
Comparative protein structure modeling using Modeller
UCSF Chimera--a visualization system for exploratory research and analysis
H++: a server for estimating pKas and adding missing hydrogens to macromolecules
Server: a web service for deriving RESP and ESP charges and building force field libraries for new molecules and molecular fragments
Application of RESP charges to calculate conformational energies
Long-time-step molecular dynamics through hydrogen mass repartitioning
Numerical integration of the cartesian equations of motion of a system with constraints: molecular dynamics of n-alkanes
An improved hydrogen bond potential: impact on medium resolution protein structures
Using circular dichroism collected as a function of temperature to determine the thermodynamics of protein unfolding and binding interactions
Nuclear magnetic resonance methods for quantifying microsecond-to-millisecond motions in biological macromolecules
A slow conformational switch in the BMAL1 transactivation domain modulates circadian rhythms
Download references
We thank Paola Munoz-Tello for critical reading of the manuscript
This work was supported by National Institutes of Health (NIH) grants R01DK101871 (DJK)
and P20GM103546 (computational resources to TH); American Heart Association (AHA) fellowship award 16POST27780018 (RB); and the William R
Charitable Trust (TSRI High School Student Summer Internship Program)
A portion of this work (ZZ exchange NMR) was performed at the National High Magnetic Field Laboratory (NHMFL/MagLab)
which is supported by National Science Foundation (NSF) Cooperative Agreement No
19F NMR data presented herein were collected at the CUNY ASRC Biomolecular NMR Facility
High School Student Summer Internship Program
and M.D.N performed mutagenesis and/or purified proteins
performed cellular assays and mutagenesis assays
performed the molecular dynamics simulations
conceived the experiments and wrote the manuscript with input from all authors
Publisher’s note: Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations
Download citation
DOI: https://doi.org/10.1038/s41467-018-07133-w
* Assisted living and similar communities (e.g.
Residential care communities with missing data were excluded
† Participating administrators and directors of residential care communities were asked
"What is the total number of residents currently living at this residential care community
§ Participating administrators and directors of residential care communities were asked
Include both occupied and unoccupied beds."
there were 22,200 residential care communities serving 713,300 residents across the United States
Forty percent of residential care communities were smaller with 4–10 beds
but these communities housed only 7% of all residents
The largest residential care communities with more than 100 beds were only 9% of all communities but housed 33% of all residents
Source: Caffrey C, Harris-Kojetin L, Rome V, Sengupta M. Operating characteristics of residential care communities, by community bed size: United States, 2012. NCHS data brief, no 170. Hyattsville, MD: National Center for Health Statistics; 2014. Available at http://www.cdc.gov/nchs/data/databriefs/db170.htm
Reported by: Vincent Rome, MPH, vrome@cdc.gov
Alternate Text: The figure above is a bar chart showing that in 2012
Forty percent of residential care communities were smaller with 4-10 beds
but these communities housed only 7% of all residents
The largest residential care communities with more than 100 beds were only 9% of all communities but housed 33% of all residents
How do I view different file formats (PDF, DOC, PPT, MPEG) on this site?
This work, Soldier in Heroic Battle to Receive Silver Star, by SGT Nicole Kojetin, identified by DVIDS, must comply with the restrictions shown on https://www.dvidshub.net/about/copyright
This work, Face of Defense: Apache Pilot Receives Distinguished Flying Cross, must comply with the restrictions shown on https://www.dvidshub.net/about/copyright
Homeowner blames city for flooding; city says it's climate change
Longtime Edina resident Robert Tengdin's legal fight with the city over stormwater flooding his beloved tennis court has spilled into the community newspaper
with the two sides arguing their opposing views
"I ask you, is this the Edina others envy and of which we have been so proud?" Tengdin, 91, wrote in an open letter in the Edina Sun Current last month
The area east of Highlands Lake that abuts Tengdin's tennis court and several neighbors' backyards has become a swamp in recent years
and it likely will stay that way if the city wins its appeal next spring
was dry for decades and considered part of Highlands Park
with the city laying wood chips and maintaining nature walking paths
But the city claims it always has been a wetland and this is its natural state
"The city can't control the weather," City Manager Scott Neal wrote in a letter responding to Tengdin's full-page ad
The back-and-forth is the latest development in a battle now spanning seven years. Tengdin first complained to the city in 2014 when historic flooding in Edina damaged 48 homes. Five years later, neither Tengdin's concerns nor the water had receded, so he gathered nearly 100 signatures on a petition asking the city to fix the flooding.
Jeff Doom, a financial planner who moved across the street from Tengdin in 2007, said he was walking his dog Ole in the area until it became impassable two years ago.
"Some parts of the trails were muddy, then all of a sudden the trail is 2 feet under water," Doom said. "That's the puzzling thing. How in the heck did this happen? If you say, 'Well, it's a wetland, and this is just part of being a wetland,' ... but we've had a drought. Shouldn't it go back after a drought to at least where it was a few years ago? What's different?"
City staff declined to be interviewed for this story, citing the pending litigation.
In court documents, Edina argues it is not responsible for rising groundwater levels that have increased by 10 feet over the past decade, and it has no statutory duty "to lower water levels in a wetland that are the clear result of climate change."
The 88-year-old said if he were still in his role today, he would drain water into Highlands Lake. "It can be corrected, but it's going to cost a few dollars," Kojetin said.
Ken Powell, Wetland Conservation Act supervisor for the Minnesota Board of Water and Soil Resources, said he's seen an increase in similar litigation from homeowners downstream, or where stormwater drains, who are "getting the brunt" from a combination of climate change and more drainage.
"I don't know what the solution is. There is not a mechanism to stop the drainage. These landowners downstream have no option but to pursue legal remedies," Powell said.
Tengdin's attorney, Tamara O'Neill Moreland, argues the flooding is not only caused by a changing climate, but an increase in impervious surfaces from redevelopment. In addition, she says the basin is part of the city's stormwater management system, so Edina has an obligation to maintain and repair it.
"You don't have the ability as a city to sit back and say, 'We're going to store our water on your private property because this is what happens when it rains a lot,' " she said.
Now that portion of the park is only accessible when water freezes over. Tengdin refers to it as the Okefenokee Swamp. "All it needs is an alligator," he said. "It's a wonderful place to be able to walk to but not to wade through."
Tengdin wants Edina to fix the flooding so residents can enjoy the area again. Such a remedy also would improve the value of his home where he raised three sons. A 2019 assessment from Tengdin's real estate agent listed a recommended selling price of $850,000, but ReMax estimates the house is devalued by $400,000 without the flooding being addressed.
In summer 2020, the tennis court he built in 1967 was full of standing water and mallard ducks, instead of the friends and neighbors who played on the court for decades. That year, Tengdin retired after 68 years with Allison-Williams Co. investment banking firm, and he filed his lawsuit.
The city sought to throw out the case, arguing it can't be sued for enacting its own policies. Hennepin County District Judge Jacqueline Regis denied the motion, but the city appealed days before an August trial was set to begin. A decision from the appellate court is likely next spring.
Regis wrote in her order that the east basin "was dry enough that Edina built and maintained a park and trail system" from the 1960s through at least the 1990s. "In recent years, however, the water level has noticeably risen," she stated, adding that "numerous trees in the east basin died due to flooding and the trails are, at least partly, submerged."
A $68,000 Barr Engineering study conducted by the city found flooding wasn't caused by actions or inaction of the city. Four possible solutions from the study were rejected by the city: building a floodwall, draining stormwater into Highlands Lake, more frequent maintenance of the pump at the lake and pumping the east basin.
In February, a year after Tengdin sued, the Edina City Council adopted a policy stating that the city only pumps out stormwater "when floodwater threatened a principle (habitable) structure."
Before that, City Engineer Chad Millner wrote that "flooded tennis courts are currently not a priority" in a series of 2019 e-mails, after Mayor James Hovland and council members toured Tengdin's property and he raised his concerns with city staff.
Attorney Paul Reuvers is defending Edina through the League of Minnesota Cities Insurance Trust. The city made a "judgment call," he said, and drew the line with limited resources. "The city can't pump [at] everyone's request," he said.
Tengdin thinks the city is trying to drag out litigation long enough that he dies before the issue is settled in court.
But when asked whether he's lived his whole life in Edina, Tengdin replied, "Not yet."
Kim Hyatt reports on North Central Minnesota. She previously covered Hennepin County courts.
No Section
Peek inside homes for sale in the Twin Cities area
After falling behind 17-0 at halftime and being dominated most of the game
the Bulldogs may have locked up a spot in the College Football Playoff
* Denominators used to calculate percentages for adult day services centers
and residential care communities were derived from the number of residents/participants on a given day in 2012
Denominators used to calculate percentages for home health agencies and hospices were the number of patients whose episode of care in a home health agency ended at any time in 2011
and the number of patients who received care from Medicare-certified hospices at any time in 2011
† Participating administrators and directors of residential care communities and adult day services centers were asked
"Of the residents currently living at this community/participants enrolled at this center
about how many have been diagnosed with depression?"
the percentage of users of long-term care services with a diagnosis of depression was highest in nursing homes (49%) and home health agencies (35%)
and lowest in residential care communities (25%)
The percentage of users with a diagnosis of depression in nursing homes (49%) was approximately twice that of those in adult day services centers (24%) or residential care communities (25%) in 2012
Source: Harris-Kojetin L, Sengupta M, Park-Lee E, Valverde R. Long-term care services in the United States: 2013 overview. Hyattsville, MD: US Department of Health and Human Services, CDC; 2013. Available at http://www.cdc.gov/nchs/data/nsltcp/long_term_care_services_2013.pdf
For a long time, the sport of bandy was so obscure it barely registered a ripple on the U.S. landscape. After more than three decades of growth in the country, though, the ice sport — consider it a cross between ice hockey, field hockey and soccer — has gained enough traction to create a U.S. Bandy Hall of Fame. ¶ The inaugural class of six, deep with Minnesotans, will be inducted at 6 p.m. Wednesday night at the Aster Cafe in Minneapolis.
They are: Bob Kojetin, former head of Edina Park and Recreation, who is credited with helping introduce bandy — an 11-on-11 skating sport with Swedish roots played with a ball and bowed sticks on a rink roughly the size of a soccer field — to the United States; Magnus Skold, longtime General Manager of the U.S. team; Gunnar Fast, who brought the sport to Minnesota; and Chris Halden, Tom Howard, Chris Middlebrook, all longtime standout players.
The Hall of Fame will be housed at the John Rose Oval in Roseville, which is also the home rink for the U.S. National Team.
"Once we reached the 30-year mark, with hundreds of players, we formed a committee. We're not getting any younger," Halden said. "They decided to do the induction by decade, starting with the guys who really built the sport."
Like a lot of U.S. players, Halden is a converted hockey player. He and Middlebrook were college hockey teammates at Gustavus. Both gave bandy a shot in the early 1980s and became hooked. More than 30 years and countless international tournaments later, Halden still is going strong at age 58. Many of those international tournaments were in Russia, where he said the U.S. National Team players were treated like "rock stars" and often play in front of crowds of 25,000.
Locally, the 11-on-11 version is played at the Oval, while a modified 4-on-4 version at local hockey rinks is catching on among the next generation of college players looking for a diversion, but still a workout, over the summer. A league this summer attracted several top-tier players, and Halden is hoping to convince some of them to make the leap to the national team someday.
"Our formula is to get hockey players to convert to bandy," Halden said. "But we've had a youth program in the past few years with homegrown players. ... It's still a small cult sport. But it's really incredible. It's so fun to play."
Wolves
Minnesota passing on Curry (twice) or Ant’s list
Butler and the Warriors will play at Target Center in a second-round NBA playoff series
Owen Marsolek struck out 17 to lead the Hilltoppers to a 3-0 victory Monday at Siebert Field
Online retail giant Amazon is now the owner of a newly completed warehouse in central Moravia. Once its operations are in full swing next year, the Kojetín Industrial Park could offer up to 2,000 jobs
This will be the online retail giant’s second warehouse in Czechia, following one in Dobrovíz, Central Bohemia that opened in 2015
Built by industrial construction firm Panattoni
this is Czechia’s first multi-story distribution center
the building boasts a high level of energy self-sufficiency
The warehouse will have a special robotic unit to facilitate the work of employees
The four-story distribution center has a total area of more than 187,000 square meters
“I believe that the state-of-the-art logistics complex will help Amazon in its further expansion in the e-commerce sector
which has been experiencing a boom in recent years
The project will also be beneficial for the region thanks to a large number of job opportunities,” Sovička added
The new distribution hall was built on a brownfield formerly used by a sugar factory for its settling tanks
“Investment in the revitalization of aging or dilapidated sites is our specialty
I am proud that we have once again succeeded in creating an industrial zone on land with low ecological value
which will rank at the very top in the field of environmentally friendly and technological construction,” Accolade Group CEO Milan Kratina said
The logistics hall was built to meet strict environmental requirements. Accolade states it is aiming for BREEAM New Construction sustainable approach certification at the "Excellent" level
The certification takes place after the building is finished
BREEAM (Building Research Establishment Environmental Assessment Method) is the oldest and most widely recognized certification for the sustainability of buildings
The certification evaluates not only the energy efficiency of the building and other operational factors
but also the environmental impact of the construction process
Many of the building panels were made at a factory some 12 kilometers from the construction site and moved by rail
The area around the warehouse will feature a lizard habitat and bee hives
out of which around 300 will be planted at the warehouse and the rest in Kojetín
“We care about the operation being environmentally friendly
which is why we installed modern 4 MW photovoltaic panels on the roof
this exceeds ordinary home installations by three orders of magnitude,” Michal Šmíd
Your morning coffee deserves a great companion. Why not enjoy it with our daily newsletter? News from Czechia, curated insights, and inspiring stories in English.
Global internet retail giant Amazon is building a new logistics center in Kojetin industrial park near Prerov in the Olomouc Region
which should create over 2,000 jobs when it opens in 2023
and will be equipped with modern technology
Photo credit: Freepik (illustrative photo)
The ongoing construction of the new Amazon facility in the industrial park in Kojetin is scheduled for completion next year
The facility will be equipped with modern technology
General Manager of Amazon for the Czech Republic
the new distribution centre will allow the company to offer more products and support a growing number of independent small businesses that sell their goods through its e-shop
The Kojetin centre will prepare and dispatch smaller items to customers
Amazon has already started recruiting the first staff for the centre
The full recruitment process is planned for the second quarter of 2022
“The new jobs will complement 4,000 existing permanent jobs that we have already created in the Czech Republic
and this is a result we are proud of,” said Šmíd in a press release.
Amazon has been operating in the Czech Republic since 2013
The company’s first distribution centre in the country opened in 2015
and it now employs over 3,000 permanent staff
The company employs a further 1,000 specialized staff in corporate offices in the center of Prague
Advertise with us
Privacy Policy
Brno Daily is a Czech media outlet for expats
Our partners