Targeting Troponin C with Small Molecules Containing Diphenyl Moieties: Calcium
Sensitivity Effects on Striated Muscle and Structure Activity Relationship
Eric R. Hantz
1
, Svetlana B. Tikunova
2
, Natalya Belevych
3
, Jonathan P. Davis
2
, Peter J. Reiser
3, §
,
Steffen Lindert
1, §,
*
1
Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH, 43210
2
Davis Heart and Lung Research Institute and Department of Physiology and Cell Biology, The
Ohio State University, Columbus, OH, 43210
3
Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH 43210
§ denotes co-senior authors
* Correspondence to:
Department of Chemistry and Biochemistry, Ohio State University
2114 Newman & Wolfrom Laboratory, 100 W. 18
th
Avenue, Columbus, OH 43210
614-292-8284 (office), 614-292-1685 (fax)
lindert.1@osu.edu
Abstract
Despite large investments from academia and industry, heart failure, which results from a
disruption of the contractile apparatus, remains a leading cause of death. Cardiac muscle
contraction is a calcium-dependent mechanism, which is regulated by the troponin protein complex
(cTn) and specifically by the N-terminal domain of its calcium binding subunit (cNTnC). There is
an increasing need for the development of small molecules that increase calcium sensitivity
without altering systolic calcium concentration, thereby strengthening cardiac function. Here, we
examined the effect of our previously identified calcium sensitizing small molecule, ChemBridge
compound 7930079, in the context of several homologous muscle systems. The effect of this
molecule on force generation in isolated cardiac trabeculae and slow skeletal muscle fibers was
measured. Furthermore, we explored the use of Gaussian accelerated molecular dynamics in
sampling highly predictive receptor conformations based on NMR derived starting structures.
Additionally, we took a rational computational approach for lead optimization based on lipophilic
diphenyl moieties. This led to the identification of three novel low affinity binders, which had
similar binding affinities to known positive inotrope trifluoperazine. The most potent identified
calcium sensitizer was compound 16 with an apparent affinity of
117 ± 17$𝜇𝑀
.
Introduction
The occurrence of heart failure has been shown to increase over time with aging of the population,
and its impact was estimated at $216 billion in direct cost from 2016 - 2017
1
. Heart failure (HF)
has been generally characterized as the heart’s inability to pump and/or fill with enough blood to
meet the demands of circulation
2
. Heart failure can be divided into three subtypes: HF with
reduced ejection fraction (HFrEF), heart failure with preserved ejection fraction (HFpEF), and HF
with mid-range ejection fraction (HFmrEF). These subtypes have been differentiated based on the
percentage of ejection fraction (EF) from the left ventricle, where HFrEF has an EF below 40%,
HFpEF has an EF greater than 50%, and HFmrEF has an EF ranging from 40-50%
3, 4
. HF has
been classically treated with diuretics and blood pressure medication to reduce blood volume.
However, the focus for many experts is the design of small molecules that restore cardiac
.CC-BY-NC-ND 4.0 International licenseavailable under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprintthis version posted February 6, 2023. ; https://doi.org/10.1101/2023.02.06.527323doi: bioRxiv preprint
contractility and functionality in efforts to treat HFrEF. A popular class of small molecules with
this intended purpose are positive inotropes
5, 6, 7, 8, 9
. Some of these compounds have been shown
to utilize the beta-adrenergic pathways to create an increased systole concentration of Ca
2+
ions.
However, increasing the systolic [Ca
2+
] can potentially lead to arrhythmias in patients
10, 11
. The
current direction of the field has shifted towards the identification and development of positive
inotropes that increase Ca
2+
sensitivity of the proteins involved in muscle contraction without
affecting the concentration of systolic Ca
2+ 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22
.
Ca
2+
-dependent heart muscle contraction is regulated by cardiac troponin (cTn). The cTn complex
consists of three subunits: troponin T (cTnT) which interacts with tropomyosin and anchors the
protein complex to the thin filament, troponin I (cTnI) known as the inhibitory subunit, and
troponin C (cTnC) which facilitates Ca
2+
binding. cTnI plays a key structural role in anchoring
cTnC to the rest of the troponin complex, in addition to having two amphiphilic peptides that play
important functional roles. Under unsaturated Ca
2+
conditions, the cTnI inhibitory peptide
(residues 137-148) interacts with actin, thereby inhibiting actomyosin binding
23
. Additionally, the
cTnI switch peptide (cTnI
sp
residues 149-164) interacts with the N-terminal domain of cTnC
stabilizing the open conformation of the domain when saturated with Ca
2+
23, 24, 25
. cTnC is a
dumbbell-shaped protein with two globular domains connected by a flexible linker, each
containing two EF-hand motifs
24, 26
. The C-terminal domain (cCTnC), termed the structural
domain, contains two high-affinity binding sites (sites III and IV) which are constantly saturated
by Ca
2+
or Mg
2+
under physiological conditions
27
. The N-terminal domain (cNTnC), known as
the regulatory domain, contains only one active binding site (site II)
28, 29
. Upon Ca
2+
binding to
site II, a large conformational change of the troponin complex is initiated by cNTnC. One feature
of this structural rearrangement is the opening of a hydrophobic patch (residues 20, 23, 24, 26, 27,
36, 41, 44, 48, 57, 60, 77, 80, and 81) in cNTnC, which becomes stabilized upon binding of the
cTnI
sp
. The hydrophobic patch serves as an attractive target for increasing Ca
2+
sensitivity via
small molecules. Additionally, the hydrophobic patch and entire troponin complex have been the
focus of numerous in silico studies to capture the proteins dynamics of hydrophobic patch opening
30, 31, 32, 33, 34, 35, 36, 37
, and cTnI
sp
binding
38, 39, 40, 41, 42
.
We have knowledge of several compounds that bind to the hydrophobic patch and modulate the
sensitivity of Ca
2+
binding: levosimendan
43
, one of the most well-known Ca
2+
sensitizers, and its
analog pimobendan
44
, 3-methyldiphenylamine (3-mDPA)
45
, bepridil
46
, W7
47, 48
, dfbp-o (2′,4′-
difluoro(1,1′-biphenyl)-4-yloxy acetic acid)
49
, trifluoperazine (TFP)
50
, National Cancer Institute
(NCI) database compounds NSC147866
18
, NSC600285
16
, NSC611817
16
, and ChemBridge
compounds 6872062, 7930079, and 9008625
17
. The two-dimensional structures of these
compounds are shown in Figure 1. There are currently no FDA approved Ca
2+
sensitizers, despite
several of these compounds proceeding to clinical trial. Levosimendan has been utilized in Europe
for decades
51
, but has yet to receive FDA approval. Compounds trifluoperazine and bepridil failed
clinical trials due to off-target effects
52
. This underscores the need for continual improvement in
positive inotrope efficacy and safety.
In previous work we identified three Ca
2+
sensitizing compounds with respective
𝐾
!
values < 100
𝜇𝑀
, with the lead compound being ChemBridge compound 7930079 (2-[(4,5-diphenyl-1H-
imidazol-2-yl)thio]-N-1,3-thiazol-2-ylacetamide). This compound is one of the most potent and
high affinity cNTnC binders known to date. In this work, we examined the sensitizing effects of
2-[(4,5-diphenyl-1H-imidazol-2-yl)thio]-N-1,3-thiazol-2-ylacetamide in rat cardiac trabeculae
and in slow skeletal muscle fibers. We found this compound induced a small but significant shift
.CC-BY-NC-ND 4.0 International licenseavailable under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprintthis version posted February 6, 2023. ; https://doi.org/10.1101/2023.02.06.527323doi: bioRxiv preprint
in cardiac trabeculae and a much larger (four-fold) shift in slow skeletal fibers. Furthermore, we
evaluated the use of Gaussian accelerated Molecular Dynamics (GaMD) in generating predictive
receptor conformations for virtual screening based on initial NMR model starting structures.
Additionally, we further explored the structure activity relationship of potential Ca
2+
sensitizers
based on the lipophilic character of the known high affinity Ca
2+
sensitivity modulators. We found
three compounds to slow the rate of Ca
2+
dissociation from cNTnC within a cTnC-cTnI chimera,
with apparent disassociation constants ranging from
117 482$𝜇𝑀
, similar to that of many
known Ca
2+
modulators.
Methods
Receptor Selection and Preparation
For our computational docking studies, we seek to identify cNTnC – cTnI
sp
conformations that are
highly predictive. We prioritized the inclusion of the cTnI
sp
to accurately model the cTnC-cTnI
chimera utilized in our biochemical studies. Thus, we performed an exhaustive search for human
cNTnC – cTnI
sp
structures with known Ca
2+
sensitivity modulating small molecules bound in the
hydrophobic patch (holo) and ligand unbound (apo) structures in the RCSB protein data bank. We
obtained apo structures from two PDB entries: 1MXL
53
where cNTnC is in complex with the
cardiac isoform of the TnI switch peptide and 2MKP
54
where cNTnC is in complex with the fast
skeletal TnI switch peptide. We obtained holo cNTnC conformations of several known calcium
sensitivity modulators: bepridil (PDB: 1LXF
55
), W7 (PDBs: 2KFX
56
, 2KRD
57
, and 6MV3
47
),
dfbp-o (PDB: 2L1R
49
), and 3-mDPA (PDBs: 5WCL
45
and 5W88
45
). All selected PDB entries
were obtained from NMR experiments and contained multiple conformers; each structure was
extracted resulting in 166 receptor conformations. We examined all 166 receptor conformers in
our docking studies.
All receptor conformations were imported into Schrödinger’s Maestro
58
and prepared using the
Protein Preparation Wizard
59
; where for all applicable conformers the C-terminus was capped by
the addition of an N-methyl amide and the N-terminus was capped by the addition of an acetyl
group. The protonation states of all titratable residues were assigned using EPIK
60, 61
with a pH
constraint of
7.4 ± 1.0
.
GaMD Simulations and Clustered Receptor Generation
In previous work we have demonstrated the utility of GaMD to generate highly predictive receptor
conformations based on X-ray crystallographic structures
62
. Here, we evaluated the impact of this
computational technique for increased sampling of troponin’s structural ensemble when the initial
frame was derived from an NMR experiment. For each of the beforementioned nine NMR PDB
deposited structures (1MXL, 2MKP, 1LXF, 2KFX, 2KRD, 6MV3, 2L1R, 5WCL, and 5W88), we
extracted model one (the representative receptor conformer) which served as the initial starting
frame for a 300 ns GaMD simulation performed with Amber20
63, 64
. For ligand-bound conformers,
the small molecule was parameterized using the second generation of the generalized amber
forcefield (GAFF2)
65
and Amber’s antechamber software. All protein complexes were
parameterized with the amber forcefield ff14SB
66
, solvated with TIP3P
67
water molecules in a 12
octahedron, and neutralized with sodium ions.
All systems were minimized with restraints (20 kcal/mol) on the protein and ligand (if applicable)
using 2500 steps of steepest decent minimization followed by 2500 steps of conjugate gradient
decent. A second unconstrained minimization was sequentially performed using 2500 steps of
steepest decent minimization followed by 2500 steps of conjugate gradient decent. The system
.CC-BY-NC-ND 4.0 International licenseavailable under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprintthis version posted February 6, 2023. ; https://doi.org/10.1101/2023.02.06.527323doi: bioRxiv preprint
was then heated to 310 K over a span of 1 ns using the Langevin thermostat
68
. For each system, a
short conventional MD simulation was performed for equilibration under constant temperature
(310 K) and pressure (1 bar) using the Langevin thermostat and Berendsen barostat
69
, prior to the
GaMD preparation simulations. During the GaMD preparation simulation statistics were collected
to calculate the appropriate boosts to apply to the dihedral and total potential energies. These
statistics were obtained from a second 10 ns conventional MD run, the initial boosts then applied,
and subsequently updated during a 50 ns GaMD biasing run. The final GaMD restart parameters
(VmaxP, VminP, VavgP, sigmaVP, VmaxD, VminD, VavgD, and sigmaVD) were then read in
for a 300 ns GaMD production run. The upper limit for the dihedral and total boost potentials was
set to 6 kcal/mol. All simulations were performed with a 12
cutoff for electrostatic and van der
Waals interactions, and a 2 fs timestep with the SHAKE algorithm
70
. Coordinates were saved
every 2 ps, resulting in 150,000 frames.
The final structures used for docking studies were obtained by clustering each of the 300 ns GaMD
simulations individually. Prior to clustering the individual trajectories, all water molecules, ions,
and ligands were removed. Clustering was performed over every fourth frame of the original
150,000 frames, resulting in 37,500 frames available for clustering. The density-based clustering
algorithm (DBScan)
71
implemented in Amber’s CPPTRAJ was used to cluster the processed
trajectories to obtain approximately 11 representative conformers per simulation. Trajectories were
clustered using the backbone atoms of the hydrophobic patch residues (residues 20, 23, 24, 26, 27,
36, 41, 44, 48, 57, 60, 77, 80, and 81). In total, 101 clustered conformers were generated and used
in further active/decoy docking studies (see below). All receptor conformers were loaded into
Maestro and prepared using the Protein Preparation Wizard tool, as detailed above.
Receptor Grid Generation
Receptor grids for ligand-bound receptor conformers were generated by selecting the ligand within
the Maestro workspace and using the center of mass of the ligand as the center of the receptor grid.
For apo receptor conformers, the center of the search space was determined by submitting a PDB
file of the conformer to the FTMap webserver
72, 73
, where fragments were globally docked to
identify potential small molecule binding sites. The resulting output file (PDB format) was
imported into PyMOL
74
, where the align function was utilized to overlay the FTMap PDB file
with a holo receptor conformer. Upon visual inspection, the fragment cluster that overlapped the
position of the known calcium modulator was identified, and the center of mass of the cluster was
calculated. The center of mass served as the three-dimensional coordinates for the center of the
receptor grid. The search area was centered on the ligand’s center of mass or the supplied three-
dimensional coordinates, respectively, and allowed the centroids of any docked compound to fully
explore a
10 × 10 × 10$Å
"
inner search space, while the periphery of the ligand was able to extend
out into a
20 × 20 × 20$Å
"
search space. The OPLS3e forcefield
75
was used to generate the search
grid and all hydroxyl groups were selected to be freely rotatable within the search area.
Ligand Preparation
The three-dimensional coordinates of all small molecules (known binders) were extracted from
the representative conformation of their respective PDB entries. Small molecules from the
Schrödinger decoy sets, the ChemBridge EXPRESS-Pick Collection, the ChemBridge Core
Library, and NCI database used in our docking protocol were obtained from their respective
databases in the form of SDF files containing their three-dimensional structural information. For
the known binders levosimendan, pimobendan, and trifluoperazine, we generated their respective
structures using Schrödinger’s 2D Sketcher tool. Schrödinger’s LipPrep
76
tool was used to prepare
.CC-BY-NC-ND 4.0 International licenseavailable under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprintthis version posted February 6, 2023. ; https://doi.org/10.1101/2023.02.06.527323doi: bioRxiv preprint
each ligand for docking. All tautomers, protomers, and stereoisomers were generated for each
ligand. Protonation states were assigned using EPIK with a pH value of
7.4 ± 1.0
, identical to that
of the protein preparation step.
Active/Decoy Screening and Receptor Performance Analysis
A total of 267 receptor conformations (166 NMR receptor conformers and 101 GaMD clustered
conformers) were evaluated by active/decoy screening. The 13 known sensitivity modulators were
used as known actives in the active/decoy docking process, and the presumed decoy small
molecules (1,000) were obtained from the Schrödinger decoy set
77
with an averaged molecular
weight of 360 g/mol. The decoy set was confirmed to match the molecular properties of the known
binders based on the following chemical properties: molecular weight, LogP, formal charge,
number of hydrogen bond donors, and number of hydrogen bond acceptors. These properties were
calculated using the RDKit (version 2020.09.1)
78
Chem Descriptor ExactMolWt, Crippen model,
GetFormalCharge, Lipinski NOCount, and Lipinski NHOHCount modules, respectively (see
Figure S1). All compounds were subject to ligand preparation as detailed above. The active and
decoys compounds post-LigPrep were docked into all receptor conformations using Schrödinger’s
Glide SP
77, 79, 80
. Default parameters from the Schrödinger 2018-3 release were utilized for all
docking simulations.
The docked poses of all small molecules were ranked by their respective docking score.
Subsequently, the top docked pose of each compound was kept, and all other
protomers/tautomers/stereoisomers of the compound removed. The performance of every receptor
conformer was evaluated with three metrics: receiver operating characteristic (ROC) curve,
enrichment factor, and a weighted enrichment factor. The ROC curve was generated by plotting
the true positive rate (TPR) against the false positive rate (FPR) and the area under the ROC curve
(AUC) was calculated using Python’s scikit-learn library (ver.0.22.1)
81
. The ROC AUC metric
evaluates the predictiveness of the receptor conformation for use in future blind screenings.
Enrichment factor (EF) was calculated to evaluate the number of known binders for a predefined
early recognition period (in our case: top 40 compounds). The early recognition period was
determined based on the number of compounds we typically screen in one iteration of our in vitro
studies. EF was calculated via the following equation:
𝐸𝐹 =
𝑁
#$%&'()*&+*%,-*./
40
×
𝑁
0($,1)
+ 𝑁
#$%&'()
𝑁
#$%&'()
𝐸𝐹 =
𝑁
#$%&'()*&+*%,-*./
40
×
1000 + 13
13
= 𝑁
#$%&'()*&+*%,-*./
× 1.948
For cNTnC, nine of the thirteen known actives were classified as poor binders (
𝐾
!
> 100𝜇𝑀
),
and four actives (ChemBridge compounds 6872062, 7930079, 9008625, and 3-mDPA) were
classified as high moderate affinity binders (
𝐾
!
< 100𝜇𝑀
). We aimed to minimize the influence
of the poor binders with regards to the EF score and subsequent conformer identification, as we
believed this would steer the computational screenings towards the identification of additional
poor binders. Therefore, in order to prioritize the identification of the high affinity known binders
in the early recognition period, we formulated a weighted enrichment factor (
𝐸𝐹
2(&34%(0
) score:
!"
!"#$%&"'
#
$
%
%#$%()**#+#&,(
)-&#."/(#+(&01(23
&'
($
%
40!()**#+#&,(
)-&#."/(#+(&01(23
&)
(
*+
&
%
'"-0,/
,
-
.
%
%#$%()**#+#&,()-&#."/(
&'
/
,%
40!()**#+#&,()-&#."/
0
%
)-&#."/
.CC-BY-NC-ND 4.0 International licenseavailable under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprintthis version posted February 6, 2023. ; https://doi.org/10.1101/2023.02.06.527323doi: bioRxiv preprint
56
!"#$%&"'
7
8
9
%#$%()**#+#&,(
)-&#."/(#+(&01(23
:;
<8
9
40!()**#+#&,(
)-&#."/(#+(&01(23
:=
<
>?
:
=???@=A
=A
7
8
9
%#$%()**#+#&,(
)-&#."/(#+(&01(23
:;
<8
9
40!()**#+#&,(
)-&#."/(#+(&01(23
:=
<
:=B>CD
Computational Investigation of Compounds with Diphenyl Groups
Based on the two-dimensional structure of the four known high affinity Ca
2+
sensitizers (3-mDPA
and ChemBridge compounds 6872062, 7930079, and 9008625), we hypothesized that the diphenyl
rings are crucial for ligand binding. In order to preserve the lipophilic interactions, we created
three chemical motifs (diphenyl-imidazole, diphenyl-triazine, and diphenylmethane; see Figure
S2) for two-dimensional structural similarity searches of large chemical libraries. In preparation
for our database searches, we filtered both the ChemBridge EXPRESS-Pick Library (501,916
compounds) and Core Library (812,681 compounds) with a cheminformatics approach. Similarly
to our previous work
17
, compounds found to have a molecular weight greater than 400 g/mol or a
calculated LogP value less than 2.0 or greater than 4.0 were removed. In a second filtering step,
compounds found to violate both of the following rules were removed as violators: number of
hydrogen bond donors > 5 and number of hydrogen bond acceptors > 10. Additionally, the libraries
were screened for Pan-assay interference (PAINS) compounds and violators were sequentially
removed. Upon completion of library filtering, the EXPRESS-Pick library was reduced to 206,726
compounds and the Core library was reduced to 412,866 compounds. For each of the remaining
compounds (619,592 compounds), we calculated the Tanimoto coefficient between that compound
and the three diphenyl motifs. We identified all compounds that had a Tanimoto coefficient
0.6
to any of the diphenyl functional groups, resulting in 89 compounds. The compounds were further
filtered by removing any compounds we had tested in our previous study
17
to avoid overlap,
compounds that did not contain a diphenyl motif via visual confirmation, and any compounds that
added functional groups exclusively to the diphenyl rings. The last filtering step was implemented
to prioritize identification of compounds with extensions from the imidazole, triazine, or methane
in efforts to retain similar protein-ligand interactions as compared to the thiazole acetamide tail of
2-[(4,5-diphenyl-1H-imidazol-2-yl)thio]-N-1,3-thiazol-2-ylacetamide (see Figure S3). These
criteria further reduced the number of compounds to 36 compounds. All remaining 36 compounds
were docked into the top three performing receptor conformations (1LXF M6, 1LXF M23, and
1LXF M28) based on the results from our active/decoy docking simulations. The compounds were
all subjected to ligand preparation as detailed above, and docked using Glide SP with default
settings, and the docking score was averaged across the three conformers to select a top set of
potentially promising compounds for experimental testing. Compounds that exhibited an averaged
docked score across the three conformers of
>$−5.0$𝑘𝑐𝑎𝑙/𝑚𝑜𝑙
were removed from experimental
testing consideration. This resulted in 23 compounds to be ordered for in vitro testing based on
Tanimoto coefficients.
In addition to identifying potential sensitizing compounds based on two-dimensional similarity,
we performed an exhaustive blind docking of the ChemBridge Core Library. We utilized the
filtered version of the Core Library as described above. The 412,866 compounds were prepared as
described in the Ligand Preparation methods section, and subsequentially docked into the top three
receptor conformations (1LXF M6, 1LXF M23, and 1LXF M28) using Glide SP with default
settings. Upon completion of docking, the docking Z-score of each compound in each receptor
was calculated and averaged over all conformers. We have previously shown that compound
selection with an unbiased ranking such as Z-score, can lead to high success rates in blind virtual
screenings
62, 82
. Based on ranking by averaged Z-score, the two-dimensional structures of the top
.CC-BY-NC-ND 4.0 International licenseavailable under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprintthis version posted February 6, 2023. ; https://doi.org/10.1101/2023.02.06.527323doi: bioRxiv preprint
40 compounds were visually inspected and diphenyl motifs were identified in 17 of the
compounds. We selected these 17 diphenyl containing compounds for experimental testing. In
total, 40 compounds (23 based on Tanimoto coefficients and 17 from the Core library blind
screening) were ordered from ChemBridge and tested in vitro.
Preparation of Proteins for Biochemical Studies
The cTnC-cTnI chimera (herein designated as chimera) was generated as previously described
17,
83
. The purified chimera was labeled with the environmentally sensitive fluorescent probe IAANS
on Cys53 of the cNTnC (within the chimera) with C35S, T53C and C84S substitutions, as
previously described
84
.
Stopped-Flow Fluorescence Measurements
All kinetic measurements were carried out in stopped-flow buffer (10 mM MOPS, 150 mM KCl,
pH 7.0) at 15
o
C using an Applied Photophysics Ltd. (Leatherhead, UK) model SX.18MV stopped-
flow apparatus with a dead time of ~1.4 ms. IAANS fluorescence was excited at 330 nm and
monitored using a 420-470 nm band-pass interference filter (Oriel, Stratford, CT). Ca
2+
chelator
EGTA (10 mM) in stopped-flow buffer was used to remove Ca
2+
(200 µM) from the chimera (0.5
µM) also in stopped-flow buffer in the absence or presence of compounds. Varying concentration
of each compound were individually added to both stopped-flow reactants. Data traces were fit
using a program by P.J. King (Applied Photophysics, Ltd.), that utilizes the non-linear Lavenberg-
Marquardt algorithm. Each k
off
represents an average of at least three separate experiments ±
standard error, each averaging at least three shots fit with a single exponential equation.
Calcium-sensitivity of Force Generation
The care and use of the animals (four male Sprague Dawley rats, 7-9 months old) used for this
project were approved by the Institutional Animal Care and Use Committee of Ohio State
University. The preparation of cardiac trabeculae (n=10) and slow skeletal muscle (soleus) fibers
(n=10) was as described in Tikunova et al
13
. The force measurements were also as described in
Tikunova et al
13
. Briefly, active force generation was measured in a series of activating solutions
with different pCa (-log of [Ca
2+
]) values, first without, then with the Compound 7930079 (20
µM). The force versus pCa data were fit as described in Mahmud et al. 2022
15
. The pCa that
corresponded to 50% of maximal force generation (pCa
50
) was determined from the curve fits and
the difference in the value between with and without the compound was calculated for each cardiac
trabecula and muscle fiber. The paired Student’s t-test was used to evaluate the statistical
significance of the difference in pCa
50
with and without the compound. The fiber type of the soleus
fibers that were used for the force/pCa measurements was determined with SDS-PAGE, as
described in Bergrin et al., 2006
85
.
Results and Discussion
Effect of Compound 7930079 on Force Generation in Isolated Cardiac Trabeculae and Slow
Skeletal Muscle Fibers.
All of the soleus fibers that were selected for force measurements were verified to be slow-type,
based upon the results from SDS-PAGE (Figure S4). A summary of the force/pCa data obtained
in cardiac trabeculae and in skeletal slow fibers, with and then without 20
𝜇
M 7930079, is shown
in Figure 2. This compound significantly (P<.01) shifted the curve to higher sensitivity in cardiac
trabeculae (0.05 ± 0.04 pCa unit) and in slow fibers (0.20 ± 0.03 pCa unit). We also tested whether
7930079 impacted maximal force generation in cardiac trabeculae and in slow fibers. Every third
activation in the series of force/pCa measurements was in pCa 4.0 activating solution which yields
.CC-BY-NC-ND 4.0 International licenseavailable under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprintthis version posted February 6, 2023. ; https://doi.org/10.1101/2023.02.06.527323doi: bioRxiv preprint
maximal force generation in both types of preparation. The peak force generated in pCa 4.0
solution was averaged (n=6 activations), without then with, the compound. Compound 7930079
had no effect (P=0.39) on average maximal force generation in slow fibers. There was a significant
(P=0.001) decrease of 8% in the average force in the presence of 7930079 in cardiac trabeculae.
However, in a control set of measurements in six other cardiac trabeculae, in which the complete
force/pCa relationship was measured twice with the compound, the average maximal force in pCa
4.0 activating solution was 6%, lower, on average, in the second, compared to the first, series of
measurements in each trabecula. We, therefore, conclude that 7930093 caused a very small (~2%)
reduction in maximal force generation in cardiac trabeculae. While these are promising results,
this underscores the need for further identification of Ca
2+
sensitizers and of particular interest in
this study, the exploration of structure-activity relationships around the diphenyl motifs.
Top Receptor Conformations Identified Based on Active/Decoy Docking.
Prior to virtual screens of small molecule libraries, we sought to further characterize the
predictiveness of PDB-deposited cNTnC-cTnI
sp
receptor conformations to correctly identify
known cNTnC binders. We identified 166 NMR-derived conformers for characterization,
expanding our search from previous works
16,17
. Additionally, we sought to determine the
effectiveness of Gaussian accelerated Molecular Dynamics (GaMD) generated conformers based
on NMR starting structures. Recently, we have shown that clustering over GaMD simulations may
result in the identification of additional highly predictive receptor conformations
62
. However, the
starting structures for this study were almost exclusively from x-ray crystallography structures
deposited in the PDB. Here, for the cNTnC-cTnI
sp
system, we aimed to determine if this enhanced
sampling technique could capture additional protein dynamics that are currently absent from
known NMR PDB structures. Therefore, based on the representative NMR model of each PDB
entry (as described in Methods), we performed a 300 ns GaMD simulation and clustered over each
individual trajectory, resulting in 101 additional receptor conformations.
In total 267 cNTnC-cTnI
sp
conformers were used in our active/decoy screening, where all
compounds were docked utilizing the Glide SP docking methodology. The 13 known cNTnC Ca
2+
sensitivity modulators served as the known actives and were docked along a set of 1,000 presumed
decoy small molecules. The decoy compounds were obtained from Schrödinger’s decoy set with
an average molecular weight of 360 g/mol. In order to avoid any unrealistic enrichment in the
active/decoy screenings, the decoys and actives were property-matched across five chemical
properties: molecular weight, LogP, formal charge, number of hydrogen bond acceptors and
number of hydrogen bond donors (see Figure S1). For all conformers, we calculated the respective
true positive rates (TPRs) and false positive rates (FPRs) and determined the AUC of the generated
ROC curves. The ROC AUC metric was used to evaluate the predictiveness of the conformer in
identifying known actives over the decoy compounds. Furthermore, we used enrichment factor
(EF) as a confidence metric for identifying the actives in a predefined early recognition period (top
40 compounds). However, particularly in the case of cTnC, many of the known actives can be
classified as weak binders (
𝐾
!
> 100$𝜇𝑀
) to the hydrophobic patch of cNTnC in the presence of
cTnI
sp
, such as: levosimendan (
𝐾
!
700$𝜇𝑀
)
86
, bepridil (
𝐾
!
= 380 ± 60$𝜇𝑀)
13
, W7
(𝐾
!
=
150 300$𝜇𝑀)
87
, dfbp-o
(𝐾
!
= 270 ± 30$𝜇𝑀)
49
, TFP (
𝐾
!
= 110 ± 20$𝜇𝑀)
13
, and NCI
compound NSC147866 (
𝐾
!
= 379 ± 50$𝜇𝑀
)
18
. The experimental binding affinities of the other
two NCI compounds (NSC600285 and NSC611817) were not reported but were predicted to be
weak binders. In order to dampen the influence of the weak binders for the purpose of receptor
selection, we developed a weighted enrichment factor (EF
weighted
) score to prioritize the early
identification of known cNTnC Ca
2+
sensitivity modulators with moderate to high affinity. Those
.CC-BY-NC-ND 4.0 International licenseavailable under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprintthis version posted February 6, 2023. ; https://doi.org/10.1101/2023.02.06.527323doi: bioRxiv preprint
included 3-mDPA (
𝐾
!
= 30$𝜇𝑀)
45
and ChemBridge compounds 6872062, 7930079, and
9008625 with respective apparent experimental binding affinities of
84 ± 30$𝜇𝑀
,
1.45 ±
0.09$𝜇𝑀
, and
34 ± 12$𝜇𝑀
17
. These four compounds contain diphenyl motifs, which served as the
inspiration for our two-dimensional similarity searches of the ChemBridge databases (see below).
The data for all three metrics (ROC AUC, EF, and EF
weighted
) for all 166 NMR conformers is
summarized in Table S1, and the data for all 101 clustered GaMD conformers is summarized in
Table S2.
Figure 3 shows the evaluation of all 267 receptor conformations, as well as the ROC curve of the
top performing receptor conformation (1LXF NMR model 6) with the respective values for ROC
AUC, EF, and EF
weighted
explicitly shown. The top three receptor conformations were determined
to be all NMR models of PDB entry 1LXF (1LXF model 6, 1LXF model 23, and 1LXF model
28), and were used in later virtual screens. Interestingly, the top performing conformers all belong
to the same PDB (1LXF), which contains cNTnC in complex with cTnI
sp
and bepridil. We believe
that our focus on the diphenyl motifs (through use of the weighted EF) led to implicit bias of
selecting 1LXF receptor conformations. 1LXF is the only deposited structure of the cNTnC
cTnI
sp
complexes with a small molecule containing a diphenyl motif (benzyl methylaniline).
Therefore, we speculated, the various receptor conformations belonging to this PDB entry could
capture more favorable protein-ligand interactions between the hydrophobic patch residues and
that of compounds with a diphenyl structure. Additionally, only one GaMD clustered conformer
(5WCL clustered conformer 6) exhibited a relatively high ROC AUC of 0.750; however, the EF
and EF
weighted
values were low (5.89 and 5.98 respectively). As shown before, GaMD is a powerful
technique for structure-based drug discovery, with the most benefit seen when x-ray
crystallographic structures served as the initial frame of the simulation. Unfortunately, in our case,
there was no improvement for capturing ideal receptor conformations for ligand binding compared
to preexisting NMR based structures. The top three receptor conformers (1LXF model 6, 1LXF
model 23, and 1LXF model 28) were used for all virtual screenings going forward.
Exploration of Diphenyl Motifs Leads to Identification of Three cNTnC Calcium Sensitizers.
Based on our previous screenings, we theorized lipophilic phenyl rings to be an important
functional group for ligand binding into the cNTnC hydrophobic patch. Therefore, we focused our
efforts on identification of small molecules containing diphenyl structures. We created three motifs
of interest: diphenyl-imidazole (based on 2-[(4,5-diphenyl-1H-imidazol-2-yl)thio]-N-1,3-thiazol-
2-ylacetamide), diphenyl-triazine, and diphenyl-methane (similar to 3-mDPA). The two-
dimensional structures of these motifs are shown in Figure S2. We focused on two small molecule
libraries from ChemBridge, the EXPRESS-Pick Library and Core Library, and filtered each library
with a cheminformatics approach as described in the Methods section. For each compound of the
filtered libraries (619,592 compounds) we calculated the Tanimoto coefficient to each of the three
diphenyl motifs. Compounds found to have a high structural similarity (Tanimoto coefficient
0.6) were further filtered to remove redundancies from previous testing by our group and based on
averaged Glide SP docking score across the three most predictive receptor conformations. This
method led to the identification of 23 compounds for further experimental testing. Additionally,
we performed a blind virtual screening of the filtered Core Library (412,866 compounds) and rank
ordered compounds based on averaged Z-Score across the three most predictive receptor
conformations. Upon visual inspection of the top 40 compounds, 17 were identified to have a
diphenyl motif and were selected for further testing. In total, we ordered 40 compounds from
ChemBridge for in vitro testing, the two-dimensional structures of all ordered compounds are
available in Figure S5.
.CC-BY-NC-ND 4.0 International licenseavailable under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprintthis version posted February 6, 2023. ; https://doi.org/10.1101/2023.02.06.527323doi: bioRxiv preprint
Initial stopped-flow kinetics revealed that 39 of the initial 40 compounds were soluble enough in
aqueous buffer that experimental measurements could be obtained. Of the 39 compounds that were
able to be experimentally tested, three showed at least a 15% decrease of the Ca
2+
dissociation rate
for the chimera. The average Ca
2+
dissociation rate observed for the chimera in the absence of
compounds was 68.6 ± 0.4 s
−1
. Compounds 2 (ChemBridge ID 7874460), 16 (ChemBridge ID
56598339), and 17 (ChemBridge ID 14233019) lead to a moderate slowing of the Ca
2+
dissociation
rate to
51.8 ± 0.4$𝑠
EF
,
49.0 ± 0.3$𝑠
EF
, and
57.6 ± 0.9$𝑠
EF
at
50$𝜇𝑀
, respectively. To further
characterize these hits, for each compound we performed stopped flow experiments using
increasing concentrations of the respective small molecule in order to get a dose response. Figure
4 illustrates the effects of each compound slowing the rate of Ca
2+
disassociation from the chimera,
where panel 4A depicts the dose response and panel 4B depicts a representative stopped-flow trace
for each of the hit compounds at a
100$𝜇𝑀
concentration of the compound. Of the three
compounds, compound 17 was determined to have the lowest apparent experimental affinity of
482 ± 300$𝜇𝑀
, a similar range to that of bepridil and NSC147866. Compound 2 was found to
have an improved apparent experimental affinity (
154 ± 23$𝜇𝑀
) compared to compound 17.
Compound 16 performed the best of the tested compounds with an apparent experimental affinity
of
117 ± 17$𝜇𝑀
. Compounds 2 and 16 had comparable affinities to known modulators TFP and
W7, and showed significant improvement over levosimendan, bepridil, dfbp-o, and NSC147866.
While we did not identify additional high affinity compounds, the three compounds that were
identified meaningfully expand our knowledge of moderate affinity cNTnC binders.
Of the 40 tested compounds only five (12.5%) contained a diphenyl-imidazole structure and of
these, one compound was identified to have moderate binding affinity: compound 2. Meanwhile
18 compounds (45%) contained diphenyl-triazine structures, which consistently failed to slow the
rate of Ca
2+
dissociation despite several compounds with a near-identical structure to previously
identified modulators. We believe the substitution of a six-member ring disrupts the electrostatic
and van der Waals protein-ligand interactions, thereby preventing the small molecule to bind
deeply in the hydrophobic patch. Compounds containing diphenylmethane moieties accounted for
42.5% (17 compounds) of the screened compounds, and two of those small molecules were
identified as medium to low affinity binders: compounds 16 and 17. Based on these results, we
believe compounds containing a five-member ring (i.e., imidazole or triazole) or diphenylmethane
are optimal for maximal lipophilic protein-ligand contact. On the other hand, our results seem to
suggest that six-member rings (i.e., triazines) alter the sterics of the ligand and prevent deeper
binding in the hydrophobic pocket. Representative docked poses of the two identified Ca
2+
sensitizers with moderate affinity in this study are shown in Figure 5. In this figure, high affinity
Ca
2+
sensitizer 2-[(4,5-diphenyl-1H-imidazol-2-yl)thio]-N-1,3-thiazol-2-ylacetamide was docked
into the cNTnC hydrophobic patch with the results of our virtual screenings overlayed, where
panel 5A shows compound 16 (ChemBridge ID 56598339) and panel 5B shows compound 2
(ChemBridge ID 7874460). Compounds 2 and 16 were shown to have the prioritized diphenyl ring
structure dock similarly to 2-[(4,5-diphenyl-1H-imidazol-2-yl)thio]-N-1,3-thiazol-2-ylacetamide.
To confirm this, we determined the symmetry-corrected heavy atom RMSD of the diphenyl rings
(utilizing spyrmsd
88
) between 2-[(4,5-diphenyl-1H-imidazol-2-yl)thio]-N-1,3-thiazol-2-
ylacetamide and compounds 2 and 16 to be
0.64$Å
and
0.91$Å
, respectively. While the diphenyl
structural contacts of focus were maintained, it is likely that compounds 2 and 16 are missing other
important protein-ligand interactions beyond the lipophilic structure. Therefore, future work
.CC-BY-NC-ND 4.0 International licenseavailable under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprintthis version posted February 6, 2023. ; https://doi.org/10.1101/2023.02.06.527323doi: bioRxiv preprint
should consider only five-member heterocycles or diphenylmethanes with a focus on optimizing
ligand tail interactions with the cTnI
sp
.
Conclusion
In this study we tested one of our previously identified cTnC Ca
2+
sensitizers (2-[(4,5-diphenyl-
1H-imidazol-2-yl)thio]-N-1,3-thiazol-2-ylacetamide) for Ca
2+
sensitization properties in the
context of two different muscle types, cardiac trabeculae and slow skeletal muscle fibers which
share the expression of cTnC and explored further virtual screening around a diphenyl motif. The
results demonstrate that compound 7930079 is a potent sensitizer of force generation in cardiac
trabeculae and in slow skeletal muscle fibers, with a much greater (four-fold) effect in the latter.
This demonstrates the potential of this compound as a possible therapeutic for the treatment of
heart failure and/or muscle weakness that is commonly associated with several myopathies,
including cancer cachexia, AIDS and sarcopenia associated with aging.
We examined the predictive utility of GaMD clustered models based on NMR initial starting
structures and, at least for cTnC, found no improvement compared to receptor conformers derived
directly from NMR experiments. Future work should focus on whether this trend can be
generalized across different receptor systems. Additionally, whether starting GaMD simulations
from the most predictive models of 1LXF might increase the AUC and EF could be explored.
Through further drug discovery efforts centered around diphenyl motifs, we identified additional
low affinity binders that increase Ca
2+
sensitivity in cNTnC site II. While our approach did not
lead to the desired success of identifying additional high affinity Ca
2+
modulators, the observed
binding affinities for two of the three identified compounds in this work were comparable to TFP,
and better than levosimendan, W7, dfbp-o, bepridil, and NCI compound NSC147866. Therefore,
we believe that the diphenyl-imidazole functional group is an important scaffold in cNTnC
binding, but in itself is insufficient for high affinity binding. Future work to improve upon 2-[(4,5-
diphenyl-1H-imidazol-2-yl)thio]-N-1,3-thiazol-2-ylacetamide will require collaboration with
medicinal chemists. Lead optimization efforts will be focused on optimizing the protein-ligand
interactions of the “tail” region, as this region was not sampled sufficiently in our present off the
shelf catalog-based optimization. The identification of three more cNTnC binders increases our
knowledge of the structure activity relationship in cTnC binding, which will be useful for follow
up work.
Acknowledgements
The authors would like to thank the members of the Lindert group for useful discussions. We
would like to thank the Ohio Supercomputer Center for valuable computational resources
89
. This
work was supported by the NIH (R01 HL137015 to S.L.).
.CC-BY-NC-ND 4.0 International licenseavailable under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprintthis version posted February 6, 2023. ; https://doi.org/10.1101/2023.02.06.527323doi: bioRxiv preprint
Figure 1. 2D structure of known cNTnC Ca
2+
sensitivity modulators.
Figure 2. The Force/pCa relationships were measured in both rat cardiac trabeculae and slow
skeletal rat muscle fibers (10 each) before and after the addition of compound 7930079, our highest
affinity compound to date. Compound 7930079 significantly (P<0.05) sensitized both cardiac, and
more potently, slow skeletal muscle to Ca
2+
, as evidenced by the increase in Ca
2+
sensitivity
(positive ΔpCa50) compared to the absence of the compound (internal control).
.CC-BY-NC-ND 4.0 International licenseavailable under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprintthis version posted February 6, 2023. ; https://doi.org/10.1101/2023.02.06.527323doi: bioRxiv preprint
Figure 3. cNTnC receptor conformation characterization using active/decoy screening. Right:
ROC AUC (x-axis) of receptor conformations plotted against enrichment factor (y-axis) and
weighted enrichment factor (z-axis). Receptor conformations obtained from NMR models are
shown with a circle marker, and clustered GaMD conformers are shown with a triangle marker.
Left: ROC curve for the best performing conformer (1LXF NMR model 6) where the TPR is
plotted against the FPR. The inset region shows the TPR and FPR of the top 40 docked compounds,
the calculated EF, EF
weighted
, and highlights true positives.
Figure 4. Effect of the hit compounds on the Ca
2+
dissociation rate from the chimera. Panel (A)
shows the plot of the apparent rates of Ca
2+
dissociation from the chimera in the presence of
increased concentration of compounds 2, 16 or 17. Each data point represents an average of at least
three measurements ± standard error. Data were fit with an asymmetric sigmoid curve. Panel (B)
shows representative stopped-flow traces as Ca
2+
is removed from the chimera in the absence or
presence of 100 µM of compounds 2, 16 or 17. The traces have been normalized and staggered for
clarity.
.CC-BY-NC-ND 4.0 International licenseavailable under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprintthis version posted February 6, 2023. ; https://doi.org/10.1101/2023.02.06.527323doi: bioRxiv preprint
Figure 5. Docked poses of Ca
2+
modulators in cNTnC hydrophobic pocket. PDB 1LXF model 6
is shown in gray surface representation with the protein backbone explicitly shown in cartoon
representation, calcium sensitizer 2-[(4,5-diphenyl-1H-imidazol-2-yl)thio]-N-1,3-thiazol-2-
ylacetamide shown in cyan overlayed with compound 16 shown in yellow (panel A) and
compound 2 shown in magenta (panel B).
.CC-BY-NC-ND 4.0 International licenseavailable under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprintthis version posted February 6, 2023. ; https://doi.org/10.1101/2023.02.06.527323doi: bioRxiv preprint
References
(1) Virani, S. S.; Alonso, A.; Aparicio, H. J.; Benjamin, E. J.; Bittencourt, M. S.; Callaway, C.
W.; Carson, A. P.; Chamberlain, A. M.; Cheng, S.; Delling, F. N.; et al. Heart Disease and Stroke
Statistics-2021 Update: A Report From the American Heart Association. Circulation 2021, 143
(8), e254-e743. DOI: 10.1161/CIR.0000000000000950.
(2) Coronel, R.; de Groot, J. R.; van Lieshout, J. J. Defining heart failure. Cardiovasc Res 2001,
50 (3), 419-422. DOI: 10.1016/s0008-6363(01)00284-x.
(3) Savarese, G.; Lund, L. H. Global Public Health Burden of Heart Failure. Card Fail Rev 2017,
3 (1), 7-11. DOI: 10.15420/cfr.2016:25:2.
(4) Nussbaumerová, B.; Rosolová, H. Diagnosis of heart failure: the new classification of heart
failure. Vnitr Lek 2018, 64 (9), 847-851.
(5) Bourge, R. C.; Fleg, J. L.; Fonarow, G. C.; Cleland, J. G.; McMurray, J. J.; van Veldhuisen,
D. J.; Gheorghiade, M.; Patel, K.; Aban, I. B.; Allman, R. M.; et al. Digoxin reduces 30-day all-
cause hospital admission in older patients with chronic systolic heart failure. Am J Med 2013,
126 (8), 701-708. DOI: 10.1016/j.amjmed.2013.02.001.
(6) Tacon, C. L.; McCaffrey, J.; Delaney, A. Dobutamine for patients with severe heart failure: a
systematic review and meta-analysis of randomised controlled trials. Intensive Care Med 2012,
38 (3), 359-367. DOI: 10.1007/s00134-011-2435-6.
(7) Tuttle, R. R.; Mills, J. Dobutamine: development of a new catecholamine to selectively
increase cardiac contractility. Circ Res 1975, 36 (1), 185-196. DOI: 10.1161/01.res.36.1.185.
(8) Packer, M.; Carver, J. R.; Rodeheffer, R. J.; Ivanhoe, R. J.; DiBianco, R.; Zeldis, S. M.;
Hendrix, G. H.; Bommer, W. J.; Elkayam, U.; Kukin, M. L. Effect of oral milrinone on mortality
in severe chronic heart failure. The PROMISE Study Research Group. N Engl J Med 1991, 325
(21), 1468-1475. DOI: 10.1056/NEJM199111213252103.
(9) Endoh, M. Amrinone, forerunner of novel cardiotonic agents, caused paradigm shift of heart
failure pharmacotherapy. Circ Res 2013, 113 (4), 358-361. DOI:
10.1161/CIRCRESAHA.113.301689.
(10) Amin, A.; Maleki, M. Positive inotropes in heart failure: a review article. Heart Asia 2012,
4 (1), 16-22. DOI: 10.1136/heartasia-2011-010068.
(11) Kass, D. A.; Solaro, R. J. Mechanisms and Use of Calcium-Sensitizing Agents in the Failing
Heart. Circulation 2006, 113 (2), 305-315. DOI: 10.1161/CIRCULATIONAHA.105.542407
(acccessed 2023/02/01).
(12) Perrone, S. V.; Kaplinsky, E. J. Calcium sensitizer agents: a new class of inotropic agents in
the treatment of decompensated heart failure. Int J Cardiol 2005, 103 (3), 248-255. DOI:
10.1016/j.ijcard.2004.12.012.
(13) Tikunova, S. B.; Cuesta, A.; Price, M.; Li, M. X.; Belevych, N.; Biesiadecki, B. J.; Reiser,
P. J.; Hwang, P. M.; Davis, J. P. 3-Chlorodiphenylamine activates cardiac troponin by a
mechanism distinct from bepridil or TFP. J Gen Physiol 2019, 151 (1), 9-17. DOI:
10.1085/jgp.201812131.
(14) Li, M. X.; Hwang, P. M. Structure and function of cardiac troponin C (TNNC1):
Implications for heart failure, cardiomyopathies, and troponin modulating drugs. Gene 2015, 571
(2), 153-166. DOI: 10.1016/j.gene.2015.07.074.
(15) Mahmud, Z.; Tikunova, S.; Belevych, N.; Wagg, C. S.; Zhabyeyev, P.; Liu, P. B.; Rasicci,
D. V.; Yengo, C. M.; Oudit, G. Y.; Lopaschuk, G. D.; et al. Small Molecule RPI-194 Stabilizes
Activated Troponin to Increase the Calcium Sensitivity of Striated Muscle Contraction. Front
Physiol 2022, 13, 892979. DOI: 10.3389/fphys.2022.892979.
.CC-BY-NC-ND 4.0 International licenseavailable under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprintthis version posted February 6, 2023. ; https://doi.org/10.1101/2023.02.06.527323doi: bioRxiv preprint
(16) Aprahamian, M. L.; Tikunova, S. B.; Price, M. V.; Cuesta, A. F.; Davis, J. P.; Lindert, S.
Successful Identification of Cardiac Troponin Calcium Sensitizers Using a Combination of
Virtual Screening and ROC Analysis of Known Troponin C Binders. J Chem Inf Model 2017, 57
(12), 3056-3069. DOI: 10.1021/acs.jcim.7b00536.
(17) Coldren, W. H.; Tikunova, S. B.; Davis, J. P.; Lindert, S. Discovery of Novel Small-
Molecule Calcium Sensitizers for Cardiac Troponin C: A Combined Virtual and Experimental
Screening Approach. J Chem Inf Model 2020, 60 (7), 3648-3661. DOI:
10.1021/acs.jcim.0c00452.
(18) Lindert, S.; Li, M. X.; Sykes, B. D.; McCammon, J. A. Computer-aided drug discovery
approach finds calcium sensitizer of cardiac troponin. Chem Biol Drug Des 2015, 85 (2), 99-106.
DOI: 10.1111/cbdd.12381.
(19) Li, M. X.; Gelozia, S.; Danmaliki, G. I.; Wen, Y.; Liu, P. B.; Lemieux, M. J.; West, F. G.;
Sykes, B. D.; Hwang, P. M. The calcium sensitizer drug MCI-154 binds the structural C-terminal
domain of cardiac troponin C. Biochem Biophys Rep 2018, 16, 145-151. DOI:
10.1016/j.bbrep.2018.10.012.
(20) Tikunova, S. B.; Davis, J. P. Designing calcium-sensitizing mutations in the regulatory
domain of cardiac troponin C. J Biol Chem 2004, 279 (34), 35341-35352. DOI:
10.1074/jbc.M405413200.
(21) Tikunova, S. B.; Liu, B.; Swindle, N.; Little, S. C.; Gomes, A. V.; Swartz, D. R.; Davis, J.
P. Effect of calcium-sensitizing mutations on calcium binding and exchange with troponin C in
increasingly complex biochemical systems. Biochemistry 2010, 49 (9), 1975-1984. DOI:
10.1021/bi901867s.
(22) Shettigar, V.; Zhang, B.; Little, S. C.; Salhi, H. E.; Hansen, B. J.; Li, N.; Zhang, J.; Roof, S.
R.; Ho, H.-T.; Brunello, L.; et al. Rationally engineered Troponin C modulates in vivo cardiac
function and performance in health and disease. Nature Communications 2016, 7 (1), 10794.
DOI: 10.1038/ncomms10794.
(23) Takeda, S.; Yamashita, A.; Maeda, K.; Maéda, Y. Structure of the core domain of human
cardiac troponin in the Ca(2+)-saturated form. Nature 2003, 424 (6944), 35-41. DOI:
10.1038/nature01780.
(24) Dvoretsky, A.; Abusamhadneh, E. M.; Howarth, J. W.; Rosevear, P. R. Solution structure of
calcium-saturated cardiac troponin C bound to cardiac troponin I. J Biol Chem 2002, 277 (41),
38565-38570. DOI: 10.1074/jbc.M205306200.
(25) Kobayashi, T.; Solaro, R. J. Calcium, thin filaments, and the integrative biology of cardiac
contractility. Annu Rev Physiol 2005, 67, 39-67. DOI:
10.1146/annurev.physiol.67.040403.114025.
(26) Herzberg, O.; Moult, J.; James, M. N. G. CONFORMATIONAL FLEXIBILITY OF
TROPONIN C11Supported by the Medical Research Council of Canada, an Alberta Heritage
Foundation of Medical Research Fellowship (to O.H.) and an Alberta Heritage Foundation for
Medical Research Visiting Scientist Fellowship (to J.M.). In Calcium-Binding Proteins in Health
and Disease, Norman, A. W., Vanaman, T. C., Means, A. R. Eds.; Academic Press, 1987; pp
312-322.
(27) Zot, H. G.; Potter, J. D. A structural role for the Ca2+-Mg2+ sites on troponin C in the
regulation of muscle contraction. Preparation and properties of troponin C depleted myofibrils. J
Biol Chem 1982, 257 (13), 7678-7683.
.CC-BY-NC-ND 4.0 International licenseavailable under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprintthis version posted February 6, 2023. ; https://doi.org/10.1101/2023.02.06.527323doi: bioRxiv preprint
(28) Sia, S. K.; Li, M. X.; Spyracopoulos, L.; Gagné, S. M.; Liu, W.; Putkey, J. A.; Sykes, B. D.
Structure of cardiac muscle troponin C unexpectedly reveals a closed regulatory domain. J Biol
Chem 1997, 272 (29), 18216-18221. DOI: 10.1074/jbc.272.29.18216.
(29) Spyracopoulos, L.; Li, M. X.; Sia, S. K.; Gagné, S. M.; Chandra, M.; Solaro, R. J.; Sykes, B.
D. Calcium-Induced Structural Transition in the Regulatory Domain of Human Cardiac Troponin
C. Biochemistry 1997, 36 (40), 12138-12146. DOI: 10.1021/bi971223d.
(30) Lindert, S.; Kekenes-Huskey, P. M.; Huber, G.; Pierce, L.; McCammon, J. A. Dynamics and
calcium association to the N-terminal regulatory domain of human cardiac troponin C: a
multiscale computational study. J Phys Chem B 2012, 116 (29), 8449-8459. DOI:
10.1021/jp212173f.
(31) Lindert, S.; Kekenes-Huskey, P. M.; McCammon, J. A. Long-timescale molecular dynamics
simulations elucidate the dynamics and kinetics of exposure of the hydrophobic patch in troponin
C. Biophys J 2012, 103 (8), 1784-1789. DOI: 10.1016/j.bpj.2012.08.058.
(32) Dewan, S.; McCabe, K. J.; Regnier, M.; McCulloch, A. D.; Lindert, S. Molecular Effects of
cTnC DCM Mutations on Calcium Sensitivity and Myofilament Activation-An Integrated
Multiscale Modeling Study. J Phys Chem B 2016, 120 (33), 8264-8275. DOI:
10.1021/acs.jpcb.6b01950.
(33) Bowman, J. D.; Lindert, S. Molecular Dynamics and Umbrella Sampling Simulations
Elucidate Differences in Troponin C Isoform and Mutant Hydrophobic Patch Exposure. J Phys
Chem B 2018, 122 (32), 7874-7883. DOI: 10.1021/acs.jpcb.8b05435.
(34) Bowman, J. D.; Coldren, W. H.; Lindert, S. Mechanism of Cardiac Troponin C Calcium
Sensitivity Modulation by Small Molecules Illuminated by Umbrella Sampling Simulations. J
Chem Inf Model 2019, 59 (6), 2964-2972. DOI: 10.1021/acs.jcim.9b00256.
(35) Wang, D.; Robertson, I. M.; Li, M. X.; McCully, M. E.; Crane, M. L.; Luo, Z.; Tu, A. Y.;
Daggett, V.; Sykes, B. D.; Regnier, M. Structural and functional consequences of the cardiac
troponin C L48Q Ca(2+)-sensitizing mutation. Biochemistry 2012, 51 (22), 4473-4487. DOI:
10.1021/bi3003007.
(36) Wang, D.; McCully, M. E.; Luo, Z.; McMichael, J.; Tu, A. Y.; Daggett, V.; Regnier, M.
Structural and functional consequences of cardiac troponin C L57Q and I61Q Ca(2+)-
desensitizing variants. Arch Biochem Biophys 2013, 535 (1), 68-75. DOI:
10.1016/j.abb.2013.02.006.
(37) Jayasundar, J. J.; Xing, J.; Robinson, J. M.; Cheung, H. C.; Dong, W. J. Molecular dynamics
simulations of the cardiac troponin complex performed with FRET distances as restraints. PLoS
One 2014, 9 (2), e87135. DOI: 10.1371/journal.pone.0087135.
(38) Lindert, S.; Cheng, Y.; Kekenes-Huskey, P.; Regnier, M.; McCammon, J. A. Effects of
HCM cTnI mutation R145G on troponin structure and modulation by PKA phosphorylation
elucidated by molecular dynamics simulations. Biophys J 2015, 108 (2), 395-407. DOI:
10.1016/j.bpj.2014.11.3461.
(39) Cheng, Y.; Rao, V.; Tu, A. Y.; Lindert, S.; Wang, D.; Oxenford, L.; McCulloch, A. D.;
McCammon, J. A.; Regnier, M. Troponin I Mutations R146G and R21C Alter Cardiac Troponin
Function, Contractile Properties, and Modulation by Protein Kinase A (PKA)-mediated
Phosphorylation. J Biol Chem 2015, 290 (46), 27749-27766. DOI: 10.1074/jbc.M115.683045.
(40) Cool, A. M.; Lindert, S. Umbrella Sampling Simulations Measure Switch Peptide Binding
and Hydrophobic Patch Opening Free Energies in Cardiac Troponin. J Chem Inf Model 2022.
DOI: 10.1021/acs.jcim.2c00508.
.CC-BY-NC-ND 4.0 International licenseavailable under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprintthis version posted February 6, 2023. ; https://doi.org/10.1101/2023.02.06.527323doi: bioRxiv preprint
(41) Cool, A. M.; Lindert, S. Computational Methods Elucidate Consequences of Mutations and
Post-translational Modifications on Troponin I Effective Concentration to Troponin C. J Phys
Chem B 2021, 125 (27), 7388-7396. DOI: 10.1021/acs.jpcb.1c03844.
(42) Genchev, G. Z.; Kobayashi, M.; Kobayashi, T.; Lu, H. Molecular dynamics provides new
insights into the mechanism of calcium signal transduction and interdomain interactions in
cardiac troponin. FEBS Open Bio 2021, 11 (7), 1841-1853. DOI: 10.1002/2211-5463.13009.
(43) Kivikko, M.; Lehtonen, L. Levosimendan: a new inodilatory drug for the treatment of
decompensated heart failure. Curr Pharm Des 2005, 11 (4), 435-455. DOI:
10.2174/1381612053382043.
(44) Takeda, N.; Hayashi, Y.; Arino, T.; Takeda, A.; Noma, K. Effect of pimobendan in patients
with chronic heart failure. Exp Clin Cardiol 2001, 6 (4), 195-199.
(45) Cai, F.; Li, M. X.; Pineda-Sanabria, S. E.; Gelozia, S.; Lindert, S.; West, F.; Sykes, B. D.;
Hwang, P. M. Structures reveal details of small molecule binding to cardiac troponin. J Mol Cell
Cardiol 2016, 101, 134-144. DOI: 10.1016/j.yjmcc.2016.10.016.
(46) MacLachlan, L. K.; Reid, D. G.; Mitchell, R. C.; Salter, C. J.; Smith, S. J. Binding of a
calcium sensitizer, bepridil, to cardiac troponin C. A fluorescence stopped-flow kinetic, circular
dichroism, and proton nuclear magnetic resonance study. J Biol Chem 1990, 265 (17), 9764-
9770.
(47) Cai, F.; Hwang, P. M.; Sykes, B. D. Structural Changes Induced by the Binding of the
Calcium Desensitizer W7 to Cardiac Troponin. Biochemistry 2018, 57 (46), 6461-6469. DOI:
10.1021/acs.biochem.8b00882.
(48) Adhikari, B. B.; Wang, K. Interplay of troponin- and Myosin-based pathways of calcium
activation in skeletal and cardiac muscle: the use of W7 as an inhibitor of thin filament
activation. Biophys J 2004, 86 (1 Pt 1), 359-370. DOI: 10.1016/S0006-3495(04)74112-0.
(49) Robertson, I. M.; Sun, Y. B.; Li, M. X.; Sykes, B. D. A structural and functional perspective
into the mechanism of Ca2+-sensitizers that target the cardiac troponin complex. J Mol Cell
Cardiol 2010, 49 (6), 1031-1041. DOI: 10.1016/j.yjmcc.2010.08.019.
(50) Ogawa, Y. Calcium binding to troponin C and troponin: effects of Mg2+, ionic strength and
pH. J Biochem 1985, 97 (4), 1011-1023. DOI: 10.1093/oxfordjournals.jbchem.a135143.
(51) Papp, Z.; Agostoni, P.; Alvarez, J.; Bettex, D.; Bouchez, S.; Brito, D.; Černý, V.; Comin-
Colet, J.; Crespo-Leiro, M. G.; Delgado, J. F.; et al. Levosimendan Efficacy and Safety: 20 Years
of SIMDAX in Clinical Use. J Cardiovasc Pharmacol 2020, 76 (1), 4-22. DOI:
10.1097/FJC.0000000000000859.
(52) Li, M. X.; Robertson, I. M.; Sykes, B. D. Interaction of cardiac troponin with cardiotonic
drugs: a structural perspective. Biochem Biophys Res Commun 2008, 369 (1), 88-99. DOI:
10.1016/j.bbrc.2007.12.108.
(53) Li, M. X.; Spyracopoulos, L.; Sykes, B. D. Binding of cardiac troponin-I147-163 induces a
structural opening in human cardiac troponin-C. Biochemistry 1999, 38 (26), 8289-8298. DOI:
10.1021/bi9901679.
(54) Robertson, I. M.; Pineda-Sanabria, S. E.; Holmes, P. C.; Sykes, B. D. Conformation of the
critical pH sensitive region of troponin depends upon a single residue in troponin I. Arch
Biochem Biophys 2014, 552-553, 40-49. DOI: 10.1016/j.abb.2013.12.003.
(55) Wang, X.; Li, M. X.; Sykes, B. D. Structure of the regulatory N-domain of human cardiac
troponin C in complex with human cardiac troponin I147-163 and bepridil. J Biol Chem 2002,
277 (34), 31124-31133. DOI: 10.1074/jbc.M203896200.
.CC-BY-NC-ND 4.0 International licenseavailable under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprintthis version posted February 6, 2023. ; https://doi.org/10.1101/2023.02.06.527323doi: bioRxiv preprint
(56) Hoffman, R. M.; Sykes, B. D. Structure of the inhibitor W7 bound to the regulatory domain
of cardiac troponin C. Biochemistry 2009, 48 (24), 5541-5552. DOI: 10.1021/bi9001826.
(57) Oleszczuk, M.; Robertson, I. M.; Li, M. X.; Sykes, B. D. Solution structure of the regulatory
domain of human cardiac troponin C in complex with the switch region of cardiac troponin I and
W7: the basis of W7 as an inhibitor of cardiac muscle contraction. J Mol Cell Cardiol 2010, 48
(5), 925-933. DOI: 10.1016/j.yjmcc.2010.01.016.
(58) Schrödinger Release 2021-1 : Maestro; Schrödinger, LLC: New York, NY, 2021.
(accessed.
(59) Sastry, G. M.; Adzhigirey, M.; Day, T.; Annabhimoju, R.; Sherman, W. Protein and ligand
preparation: parameters, protocols, and influence on virtual screening enrichments. J Comput
Aided Mol Des 2013, 27 (3), 221-234. DOI: 10.1007/s10822-013-9644-8.
(60) Schrödinger Release 2021-1 : Epik; Schrödinger, LLC: New York, NY, 2021 (accessed.
(61) Shelley, J. C.; Cholleti, A.; Frye, L. L.; Greenwood, J. R.; Timlin, M. R.; Uchimaya, M.
Epik: a software program for pK( a ) prediction and protonation state generation for drug-like
molecules. J Comput Aided Mol Des 2007, 21 (12), 681-691. DOI: 10.1007/s10822-007-9133-z.
(62) Hantz, E. R.; Lindert, S. Actives-Based Receptor Selection Strongly Increases the Success
Rate in Structure-Based Drug Design and Leads to Identification of 22 Potent Cancer Inhibitors.
J Chem Inf Model 2022. DOI: 10.1021/acs.jcim.2c00848.
(63) Amber20; University of California, San Francisco. , 2021. (accessed.
(64) Miao, Y.; Feher, V. A.; McCammon, J. A. Gaussian Accelerated Molecular Dynamics:
Unconstrained Enhanced Sampling and Free Energy Calculation. Journal of Chemical Theory
and Computation 2015, 11 (8), 3584-3595. DOI: 10.1021/acs.jctc.5b00436.
(65) Vassetti, D.; Pagliai, M.; Procacci, P. Assessment of GAFF2 and OPLS-AA General Force
Fields in Combination with the Water Models TIP3P, SPCE, and OPC3 for the Solvation Free
Energy of Druglike Organic Molecules. Journal of Chemical Theory and Computation 2019, 15
(3), 1983-1995. DOI: 10.1021/acs.jctc.8b01039.
(66) Maier, J. A.; Martinez, C.; Kasavajhala, K.; Wickstrom, L.; Hauser, K. E.; Simmerling, C.
ff14SB: Improving the Accuracy of Protein Side Chain and Backbone Parameters from ff99SB.
Journal of Chemical Theory and Computation 2015, 11 (8), 3696-3713. DOI:
10.1021/acs.jctc.5b00255.
(67) Jorgensen, W. L.; Madura, J. D. Quantum and statistical mechanical studies of liquids. 25.
Solvation and conformation of methanol in water. Journal of the American Chemical Society
1983, 105 (6), 1407-1413. DOI: 10.1021/ja00344a001.
(68) Loncharich, R. J.; Brooks, B. R.; Pastor, R. W. Langevin dynamics of peptides: the
frictional dependence of isomerization rates of N-acetylalanyl-N'-methylamide. Biopolymers
1992, 32 (5), 523-535. DOI: 10.1002/bip.360320508.
(69) Berendsen, H. J. C.; Postma, J. P. M.; van Gunsteren, W. F.; DiNola, A.; Haak, J. R.
Molecular dynamics with coupling to an external bath. The Journal of Chemical Physics 1984,
81 (8), 3684-3690. DOI: 10.1063/1.448118 (acccessed 2021/03/24).
(70) Ryckaert, J.-P.; Ciccotti, G.; Berendsen, H. J. C. Numerical integration of the cartesian
equations of motion of a system with constraints: molecular dynamics of n-alkanes. Journal of
Computational Physics 1977, 23 (3), 327-341. DOI: https://doi.org/10.1016/0021-
9991(77)90098-5.
(71) Ester, M.; Kriegel, H.-P.; Sander, J.; Xu, X. A density-based algorithm for discovering
clusters in large spatial databases with noise. Proc. Second Int. Conf. Knowledge Disc. Data
Mining (KDD-96) 1996, 226-231.
.CC-BY-NC-ND 4.0 International licenseavailable under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprintthis version posted February 6, 2023. ; https://doi.org/10.1101/2023.02.06.527323doi: bioRxiv preprint
(72) Brenke, R.; Kozakov, D.; Chuang, G. Y.; Beglov, D.; Hall, D.; Landon, M. R.; Mattos, C.;
Vajda, S. Fragment-based identification of druggable 'hot spots' of proteins using Fourier domain
correlation techniques. Bioinformatics 2009, 25 (5), 621-627. DOI:
10.1093/bioinformatics/btp036.
(73) Kozakov, D.; Grove, L. E.; Hall, D. R.; Bohnuud, T.; Mottarella, S. E.; Luo, L.; Xia, B.;
Beglov, D.; Vajda, S. The FTMap family of web servers for determining and characterizing
ligand-binding hot spots of proteins. Nat Protoc 2015, 10 (5), 733-755. DOI:
10.1038/nprot.2015.043.
(74) The PyMOL Molecular Graphics System; Schrödinger, LLC: (accessed.
(75) Roos, K.; Wu, C.; Damm, W.; Reboul, M.; Stevenson, J. M.; Lu, C.; Dahlgren, M. K.;
Mondal, S.; Chen, W.; Wang, L.; et al. OPLS3e: Extending Force Field Coverage for Drug-Like
Small Molecules. J Chem Theory Comput 2019, 15 (3), 1863-1874. DOI:
10.1021/acs.jctc.8b01026.
(76) Schrödinger Release 2021-1 : LigPrep; Schrödinger, LLC: New York, NY, 2021. (accessed.
(77) Halgren, T. A.; Murphy, R. B.; Friesner, R. A.; Beard, H. S.; Frye, L. L.; Pollard, W. T.;
Banks, J. L. Glide: a new approach for rapid, accurate docking and scoring. 2. Enrichment
factors in database screening. J Med Chem 2004, 47 (7), 1750-1759. DOI: 10.1021/jm030644s.
(78) RDKit: Open-source cheminformatics; https://www.rdkit.org (accessed.
(79) Friesner, R. A.; Murphy, R. B.; Repasky, M. P.; Frye, L. L.; Greenwood, J. R.; Halgren, T.
A.; Sanschagrin, P. C.; Mainz, D. T. Extra precision glide: docking and scoring incorporating a
model of hydrophobic enclosure for protein-ligand complexes. J Med Chem 2006, 49 (21), 6177-
6196. DOI: 10.1021/jm051256o.
(80) Friesner, R. A.; Banks, J. L.; Murphy, R. B.; Halgren, T. A.; Klicic, J. J.; Mainz, D. T.;
Repasky, M. P.; Knoll, E. H.; Shelley, M.; Perry, J. K.; et al. Glide: a new approach for rapid,
accurate docking and scoring. 1. Method and assessment of docking accuracy. J Med Chem
2004, 47 (7), 1739-1749. DOI: 10.1021/jm0306430.
(81) Pedregosa, F.; Varoquaux, G.; Gramfort, A.; Michel, V.; Thirion, B.; Grisel, O.; Blondel,
M.; Prettenhofer, P.; Weiss, R.; Dubourg, V.; et al. Scikit-learn: Machine Learning in Python.
The Journal of Machine Learning Research 2011, (12), 2825-2830.
(82) Kim, S. S.; Aprahamian, M. L.; Lindert, S. Improving inverse docking target identification
with Z-score selection. Chem Biol Drug Des 2019, 93 (6), 1105-1116. DOI:
10.1111/cbdd.13453.
(83) Siddiqui, J. K.; Tikunova, S. B.; Walton, S. D.; Liu, B.; Meyer, M.; de Tombe, P. P.;
Neilson, N.; Kekenes-Huskey, P. M.; Salhi, H. E.; Janssen, P. M.; et al. Myofilament Calcium
Sensitivity: Consequences of the Effective Concentration of Troponin I. Front Physiol 2016, 7,
632. DOI: 10.3389/fphys.2016.00632.
(84) Davis, J. P.; Norman, C.; Kobayashi, T.; Solaro, R. J.; Swartz, D. R.; Tikunova, S. B.
Effects of thin and thick filament proteins on calcium binding and exchange with cardiac
troponin C. Biophys J 2007, 92 (9), 3195-3206. DOI: 10.1529/biophysj.106.095406.
(85) Bergrin, M.; Bicer, S.; Lucas, C. A.; Reiser, P. J. Three-dimensional compartmentalization
of myosin heavy chain and myosin light chain isoforms in dog thyroarytenoid muscle. Am J
Physiol Cell Physiol 2006, 290 (5), C1446-1458. DOI: 10.1152/ajpcell.00323.2005.
(86) Robertson, I. M.; Baryshnikova, O. K.; Li, M. X.; Sykes, B. D. Defining the binding site of
levosimendan and its analogues in a regulatory cardiac troponin C-troponin I complex.
Biochemistry 2008, 47 (28), 7485-7495. DOI: 10.1021/bi800438k.
.CC-BY-NC-ND 4.0 International licenseavailable under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprintthis version posted February 6, 2023. ; https://doi.org/10.1101/2023.02.06.527323doi: bioRxiv preprint
(87) Hoffman, R. M.; Li, M. X.; Sykes, B. D. The binding of W7, an inhibitor of striated muscle
contraction, to cardiac troponin C. Biochemistry 2005, 44 (48), 15750-15759. DOI:
10.1021/bi051583y.
(88) Meli, R.; Biggin, P. C. spyrmsd: symmetry-corrected RMSD calculations in Python. J
Cheminform 2020, 12 (1), 49. DOI: 10.1186/s13321-020-00455-2.
(89) Ohio Supercomputer Center. Ohio Technology Consortium of the Ohio Board of Regents,
1987.
.CC-BY-NC-ND 4.0 International licenseavailable under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprintthis version posted February 6, 2023. ; https://doi.org/10.1101/2023.02.06.527323doi: bioRxiv preprint