ISSN 0006-2979, Biochemistry (Moscow), 2025, Vol. 90, No. 1, pp. 132-160 © Pleiades Publishing, Ltd., 2025.
Published in Russian in Biokhimiya, 2025, Vol. 90, No. 1, pp. 144-172.
132
Physiological Concentrations of Calciprotein Particles
Trigger Activation and Pro-Inflammatory Response
in Endothelial Cells and Monocytes
Daria Shishkova
1,a
, Victoria Markova
1,b
, Yulia Markova
1,c
, Maxim Sinitsky
1,d
,
Anna Sinitskaya
1,e
, Vera Matveeva
1,f
, Evgenia Torgunakova
1,g
,
Anastasia Lazebnaya
1,h
, Alexander Stepanov
1,i
, and Anton Kutikhin
1,k
*
1
Department of Experimental Medicine,
Research Institute for Complex Issues of Cardiovascular Diseases, 650002 Kemerovo, Russia
a
e-mail: shidk@kemcardio.ru 
b
e-mail: markve@kemcardio.ru 
c
e-mail: markyo@kemcardio.ru
d
e-mail: sinimu@kemcardio.ru 
e
e-mail: cepoav@kemcardio.ru 
f
e-mail: matvvg@kemcardio.ru
g
e-mail: torgea@kemcardio.ru 
h
e-mail: lazeai@kemcardio.ru 
i
e-mail: stepav@kemcardio.ru
j
e-mail: kytiag@kemcardio.ru
Received November 13, 2024
Revised December 3, 2024
Accepted December 5, 2024
AbstractSupraphysiological concentrations of calciprotein particles (CPPs), which are indispensable scav-
engers of excessive Ca
2+
and PO
4
3−
ions in blood, induce pro-inflammatory activation of endothelial cells
(ECs) and monocytes. Here, we determined physiological levels of CPPs (10 μg/mL calcium, corresponding to
10% increase in Ca
2+
in the serum or medium) and investigated whether the pathological effects of calcium
stress depend on the calcium delivery form, such as Ca
2+
ions, albumin- or fetuin-centric calciprotein mono-
mers (CPM-A/CPM-F), and albumin- or fetuin-centric CPPs (CPP-A/CPP-F). The treatment with CPP-A or CPP-F
upregulated transcription of pro-inflammatory genes (VCAM1, ICAM1, SELE, IL6, CXCL8, CCL2, CXCL1, MIF)
and promoted release of pro-inflammatory cytokines (IL-6, IL-8, MCP-1/CCL2, and MIP-3α/CCL20) and pro- and
anti-thrombotic molecules (PAI-1 and uPAR) in human arterial ECs and monocytes, although these results
depended on the type of cell and calcium-containing particles. Free Ca
2+
ions and CPM-A/CPM-F induced less
consistent detrimental effects. Intravenous administration of CaCl
2
, CPM-A, or CPP-A to Wistar rats increased
production of chemokines (CX3CL1, MCP-1/CCL2, CXCL7, CCL11, CCL17), hepatokines (hepassocin, fetuin-A,
FGF-21, GDF-15), proteases (MMP-2, MMP-3) and protease inhibitors (PAI-1) into the circulation. We concluded
that molecular consequences of calcium overload are largely determined by the form of its delivery and CPPs
are efficient inducers of mineral stress at physiological levels.
DOI: 10.1134/S0006297924604064
Keywords: calciprotein particles, calciprotein monomers, calcium ions, calcium stress, mineral stress, endothe-
lial cells, monocytes, endothelial dysfunction, endothelial activation, systemic inflammatory response
Abbreviations: CPMs, calciprotein monomers; CPPs, concentrations of calciprotein particles; ECs, endothelial cells.
* To whom correspondence should be addressed.
INTRODUCTION
Calciprotein particles (CPPs) and calciprotein
monomers (CPMs) are formed through the molecular
interactions between fetuin-A and nascent calcium
phosphate clusters. They scavenge of excessive Ca
2+
and PO
4
3−
ions, thus representing an elegant mech-
anism for the mineral homeostasis regulation [1-6].
While albumin (by far the most abundant serum pro-
tein) is mostly responsible for the clearance of circu-
lating Ca
2+
ions [5, 7], fetuin-A operates as a mineral
chaperone that either stabilizes calcium phosphate
CALCIPROTEIN PARTICLES 133
BIOCHEMISTRY (Moscow) Vol. 90 No. 1 2025
as a colloid by forming CPMs or secures its physio-
logical aggregation into corpuscular CPPs [5, 7]. CPPs
are then removed from the circulation by endothelial
cells (ECs) [8-15], monocytes [13], and liver or spleen
macrophages [16-19]. Generation of CPMs and CPPs is
an evolutionary mechanism aimed to prevent blood
supersaturation with Ca
2+
and PO
4
3−
ions (e.g., as a
result of bone resorption) and to avert extraskele-
tal calcification, a pathological condition that is fre-
quent in patients with chronic kidney disease [20-22].
Yet, internalization of CPPs by ECs and monocytes/
macrophages and their digestion in lysosomes induce
a chain of detrimental events including an increase
in cytosolic Ca
2+
, mitochondrial and endoplasmic re-
ticulum stress, nuclear factor (NF)-κB-mediated tran-
scriptional response, and release of pro-inflammatory
cytokines. such as interleukin (IL)-6, IL-8, and mono-
cyte chemoattractant protein  1/chemokine (C-C motif)
ligand  2 (MCP-1/CCL2), ultimately contributing to the
development of chronic low-grade inflammation [8-19,
23-26]. Treatment with infliximab, a selective inhibitor
of tumor necrosis factor (TNF)-α, reduced CPM and
CPP count in the serum of patients with autoimmune
diseases (inflammatory bowel disease, inflammatory
arthritis) [27], suggesting an efficacy of anti-inflam-
matory therapies in reducing CPP-related endothelial
and monocyte/macrophage activation.
Currently, experimental studies employ a variety
of CPP concentrations, from 25  µg/mL [13, 15] to 100
or 200  µg/mL calcium [16-18, 25, 28], depending on the
cell type and duration of exposure. Above-median lev-
els of ionized serum calcium (Ca
2+
) have been shown
as a significant risk factor of cardiovascular death, as
well as myocardial infarction and ischemic stroke [11,
29,  30]. The last two life-threatening conditions are
driven by atherosclerosis, the development of which
is triggered by endothelial activation and impaired en-
dothelial integrity [31-35]. Average interquartile range
between the risk (upper) and protective (lower) quar-
tiles of ionized calcium is 0.12  mmol/L (i.e., 10% of the
average reference value, or 4.8  µg/mL) [11], suggesting
that in order to obtain clinically relevant results, the
amount of calcium introduced to the cell culture or
experimental animals should not exceed these values.
Hence, an adequate quantification of CPP and CPM
physiological doses should include their recalculation
according to the respective mass of ionized calcium
(e.g., added as CaCl
2
) in order to reach a 10% increase
in the ionized calcium content in the medium.
Albeit the adverse consequences of calcium stress
have been well described [36-38], it remains unclear
whether its deleterious effects are determined by
a calcium source (free Ca
2+
ions, colloidal CPMs, or
corpuscular CPPs) or solely depend on the amount
of calcium in the microenvironment. Earlier studies
have reported that stimulation of calcium-sensing
receptor by increasing the concentration of extracel-
lular Ca
2+
promoted internalization of CPPs, leading
to the activation of NLRP3 (NLR family pyrin domain
containing3) inflammasome and IL-1β signaling path-
way [39]. Pathological effects of CPPs largely depend
on their crystallinity (amorphous primary CPPs and
crystalline secondary CPPs) and density (high-den-
sity CPPs that are precipitated at ≤16,000g and low-
density CPPs that are not precipitated at this centrif-
ugal force) [40]. The serum levels of high-density CPPs
are independently and positively associated with the
content of the pro-inflammatory cytokine eotaxin,
whereas the levels of low-density CPPs are negatively
associated with another potent inflammation inducer,
IL-8 [40]. Likewise, a higher hydrodynamic radius of
CPPs, which correlates with a reduced kidney function
and age-dependent vascular remodeling, is associated
with the cardiovascular mortality in patients with pe-
ripheral artery disease [41], as well as with vascular
calcification [42] and all-cause mortality in patients
with the end-stage renal disease [43]. The content of
primary and secondary CPPs is associated with the
vascular remodeling pathways, including those in-
volved in collagen assembly and extracellular matrix
formation [44]. CPP-induced vascular remodeling in-
cludes osteochondrogenic reprogramming of vascular
smooth muscle cells, which strongly depends on the
particle-size distribution, mineral composition, and
crystallinity of CPPs [45]. Recent studies demonstrat-
ed an association between increased CPP counts or
accelerated primary-to-secondary CPP transition with
chronic kidney disease  [44], ST-segment elevation myo-
cardial infarction  [46], and cardiovascular death in pa-
tients with the end-stage renal disease  [47] or type  2
diabetes mellitus  [48]. Removal of CPPs from blood
using specific columns ameliorated chronic inflamma-
tion, endothelial dysfunction, left ventricular hypertro-
phy, and vascular calcification [49]. Similarly, inhibi-
tion of primary-to-secondary CPP transition prevented
high phosphate-induced rat aortic calcification [50].
As indicated above, quantification of CPPs pri-
marily relies on determining the concentration of
calcium (µg) per unit volume (mL) [12,  14,  16]. Arti-
ficially synthesized calcium-free magnesiprotein par-
ticles (MPPs) did not exhibit any significant toxicity
after their introduction to cultured ECs cultures or
animals  [11], suggesting that calcium concentration
is a leading factor determining the consequences of
mineral stress. However, the spatiotemporal patterns
of intracellular calcium distribution might differ de-
pending on the calcium vehicle – from a steady and
controlled entry of Ca
2+
ions through the cell mem-
brane [51,  52] to a sharp and uncurbed influx of Ca
2+
ions into the cytosol after partial digestion of CPPs in
the lysosomes [11]. These features of calcium metabo-
lism may significantly affect transcriptional programs,
SHISHKOVA et al.134
BIOCHEMISTRY (Moscow) Vol. 90 No. 1 2025
and better understanding of cellular response to cir-
culating Ca
2+
ions, CPMs, and CPPs is required to elu-
cidate the pathophysiology of mineral homeostasis
disorders.
Here, we investigated whether the calcium deliv-
ery form dictates the response of ECs and monocytes
to physiologically relevant mineral stress that which
was achieved by adding 10  µg/mL calcium (an amount
sufficient to gain a 10% increase in ionized calcium) to
either cell culture medium or rat serum. We found that
incubation of primary human arterial ECs with albu-
min-centric CPPs (CPP-A) initiated their pro-inflamma-
tory activation manifested as an elevated production
of pro-inflammatory cytokines [IL-6, IL-8, MCP-1/CCL2,
macrophage inflammatory protein-3 alpha (MIP-3α),
plasminogen activator inhibitor-1 (PAI-1), and uroki-
nase-type plasminogen activator receptor (uPAR)] and
verified by an increased expression of genes encoding
cell adhesion molecules (VCAM1, ICAM1, E-selectin)
and pro-inflammatory cytokines [IL-6, CXCL8 (chemo-
kine (C-X-C motif) ligand 8), CCL2, and CXCL1]. Incu-
bation with fetuin-centric CPPs (CPP-F) also promoted
release of IL-6, IL-8, and MCP-1/CCL2 and upregulated
expression of genes coding for cell adhesion molecules
(VCAM1, ICAM1, SELE, and SELP) and pro-inflammato-
ry cytokines (IL6, CXCL1, and MIF). Likewise, incuba-
tion of monocytes with CPP-A in the flow culture sys-
tem promoted release of IL-6, IL-8, MIP-1α/1β, MIP-3α,
CXCL1, CXCL5, PAI-1, uPAR, lipocalin-2, and matrix
metalloproteinase-9 (MMP-9). However, addition of
free Ca
2+
ions and CPM-A caused only mild alterations
in the transcriptional program and cytokine release
by primary arterial ECs and monocytes. Intravenous
administration of excessive Ca
2+
ions (CaCl
2
), CPM-A,
or CPP-A to Wistar rats precipitated systemic inflam-
matory response including an elevation in the con-
tent of multiple cytokines, hepatokines, and proteases.
Wesuggest that the pathological effects of CPPs in  vitro
are determined by a local calcium overload, as CPPs
represent calcium vehicles with a single destination
(lysosomes). In contrast, the inflammatory response
to the intravenous calcium bolus is less dependent
on the form of calcium delivery. Nevertheless, even
physiological doses of CPPs induced pro-inflammatory
activation of ECs and monocytes, as well as systemic
inflammatory response in vivo.
MATERIALS AND METHODS
Synthesis and quantification of CPMs and CPPs.
To prepare a mixture for the synthesis of CPMs and
CPPs, 340  mg bovine serum albumin (BSA; Sigma-
Aldrich, USA) or 8  mg bovine serum fetuin-A (BSF;
Sigma- Aldrich) were dissolved in 4  mL of physiolog-
ical saline with the subsequent addition of 2  mL of
Na
2
HPO
4
(24mmol/L; Sigma-Aldrich) and 2  mL of CaCl
2
(40 mmol/L; Sigma-Aldrich). The mixture was resus-
pended after addition of each reagent. The final con-
centrations of reagents in the mixture were 42  mg/mL
for BSA or 1  mg/mL for BSF (equal to the median se-
rum level in a human population [11]), 10mmol/L for
CaCl
2
(3.2  mg of calcium), and 6mmol/L for Na
2
HPO
4
.
The suspension was then aliquoted into 8 microtubes
(1  mL per tube) that were placed into pre-heated
(37°C) heating block (Thermit, DNA-Technology, Rus-
sia) and incubated for 10  min. After this procedure,
the mixture contained three calcium sources: free Ca
2+
ions, CPMs (either CPM-A or CPM-F), and CPPs (either
CPMs-F or CPPs-F).
The resulting suspension was then aliquoted into
four ultracentrifuge tubes (2  mL per tube; Beckman
Coulter, USA) and centrifuged at 200,000g (OPTIMA
MAX-XP, Beckman Coulter) for 1  h to sediment CPP-A/
CPP-F which were then resuspended in sterile deion-
ized water and visualized by scanning electron mi-
croscopy (S-3400N, Hitachi, Japan) at an accelerating
voltage of 10 or 30  kV after 1  :  200 dilution. To com-
pare CPPs-A and CPPs-F with primary CPPs generated
from tissue extracts or biological fluids, we employed
atherosclerotic plaque-derived and serum-derived
CPPs that had been generated in T-25 flasks (Wuxi
NEST Biotechnology, China) for 6 weeks after adding
either 3  mL of plaque extract or 3  mL of human se-
rum, 1  mmol/L CaCl
2
, and 1  mmol/L Na
2
HPO
4
to 7  mL
of Dulbecco’s Modified Eagle’s Medium (DMEM; Pan-
Eco, Russia) containing 10% fetal bovine serum (FBS,
Capricorn Scientific, Germany), 1% L-glutamine–pen-
icillin–streptomycin solution (Thermo Fisher Scien-
tific, USA), and 0.4% amphotericin B (Thermo Fisher
Scientific). Plaque extracts were obtained as described
in  [8]. After incubation for 6  weeks, CPPs were sedi-
mented, prepared for scanning electron microscopy,
and visualized as described in [8]. The supernatant
with CPM-A/CPM-F and free Ca
2+
ions was transferred
into centrifugal filters with a 30-kDa molecular weight
cutoff (Guangzhou Jet Bio-Filtration, China) and cen-
trifuged at 1800g for 25  min to separate CPM-A/CPM-F
(retentate) and free Ca
2+
ions (filtrate).
The concentration of calcium in CPP-A/CPP-F,
CPM-A/CPM-F and of free Ca
2+
ions was measured by
using o-cresolphthalein complexone and diethanol-
amine-based colorimetric assay (CalciScore, AppScience
Products, Russia) after 1  :  30, 1  :  10, and 1  :  10 dilution,
respectively. Albumin concentration was measured us-
ing BCA Protein Assay Kit (Thermo Fisher Scientific)
after 1  :  200 dilution of the CPM-containing retentate;
the filtrate containing free Ca
2+
ions was not dilut-
ed before the measurement as it was expected to be
devoid of albumin. The results of colorimetric assays
were detected by spectrophotometry (Multiskan Sky,
Thermo Fisher Scientific) at 575  nm (calcium) and
CALCIPROTEIN PARTICLES 135
BIOCHEMISTRY (Moscow) Vol. 90 No. 1 2025
562  nm (albumin). All procedures were performed
under sterile conditions.
Dosage estimation. The amount calcium required
for a 10% increase in the ionized calcium content in
the milieu was estimated by adding 5, 10, 15, or 20  µg
of calcium (in a form of CaCl
2
) dissolved in aque-
ous BSA solution (300 mg/mL, average albumin con-
centration in the retentate) or aqueous BSF solution
(28  mg/mL, average fetuin-A concentration in the re-
tentate) per 1  mL of serum-free EndoLife cell culture
medium (EL1, AppScience Products) or by adding 10,
15, 20, or 40  µg of calcium dissolved in aqueous BSA
solution (300  µg/mL) per 1  mL rat serum. The mix-
ture was briefly resuspended and incubated for 1  h,
after which the concentration of ionized calcium Ca
2+
,
was measured (Konelab 70i, Thermo Fisher Scientific).
EndoLife medium and rat serum without CaCl
2
addi-
tion were used as respective controls. According to our
previous study, a 10% increase in the ionized calci-
um content [0.10-0.14mmol/L (from 4.0 to 5.6  µg/mL);
average, 0.12mmol/L (4.8  µg/mL) for human serum] is
equal to the interquartile range between the highest
(risk) and the lowest (protective) quartiles.
Cell culture. Primary human coronary artery
endothelial cells (HCAECs, Cell Applications, USA)
and human internal thoracic artery endothelial cells
(HITAECs, Cell Applications) were grown in T-75 flasks
according to the manufacturers protocol in EndoBoost
Medium (EB1, AppScience Products) using 0.25% tryp-
sin-EDTA solution (PanEco), and 10% FBS for trypsin
inhibition during subculturing. Immediately before
the experiments, EndoBoost Medium we replaced
with serum-free EndoLife Medium, during which the
cells were washed s twice with warm (37°C) Ca
2+
-and
Mg
2+
-free Dulbecco’s Phosphate Buffered Saline (DPBS)
(pH7.4, BioLot) to remove the residual serum compo-
nents. HCAECs and HITAECs were grown in parallel,
were seeded into flow culture chambers (Ibidi, Germa-
ny) or 6-well plates (Wuxi NEST Biotechnology), and
grown until reaching confluence.
Monocytes have been isolated from 5 healthy
volunteers (the authors of this study) by consecutive
extraction of peripheral blood mononuclear cells using
a Ficoll density gradient centrifugation (Ficoll solution,
1077  g/cm
3
; PanEco) and positive magnetic separation
of CD14
+
cells with an EasySep Magnet kit (STEMCELL
Technologies, USA) and monocyte isolation kit (STEM-
CELL Technologies) according to the manufacturers
instructions under sterile conditions. Monocyte count
was performed with an automated Countess  II cell
counter (Thermo Fisher Scientific) and cell counting
chamber slides (Thermo Fisher Scientific).
Internalization assay. To analyze internalization
of CPMs and CPPs by ECs, CPM-A and CPP-A were
labeled with fluorescein 5-isothiocyanate-conjugated
BSA (FITC-BSA, Thermo Fisher Scientific) either during
CPM/CPP synthesis (by adding 750  µg of FITC-BSA at
a 5  µg/µL concentration) or after the synthesis by in-
cubation of sedimented CPP-A with 125  µg (25  µL) of
FITC-BSA for 1  h at 4°C and subsequent incubation
of 500  µL of retentate (CPM-A) with 250  µg (50  µL) of
FITC-BSA for 1  h at 4°C after vortexing. The synthesis
of CPM-A and CPP-A was performed in the dark less
than 24  h before the experiment. After the labeling,
sedimented CPP-A were resuspended in DPBS, centri-
fuged at 13,000g (Microfuge 20R, Beckman Coulter) for
10  min to wash CPP-A from unbound FITC-BSA, and
resuspended in 400  µL of DPBS.
Laminar flow was established using Ibidi Pump
System Quad system (Ibidi) equipped with four sep-
arate flow culture units and Perfusion Set Yellow/
Green (Ibidi). Before starting the experiment, HCAECs
and HITAECs were cultured until confluence in flow
culture chambers (350,000 cells per chamber) and
exposed to a laminar flow (15  dyn/cm
2
) using a se-
rum-free EndoLife cell culture medium during 24  h.
Next, FITC-labeled CPM-A and CPP-A were added into
the system (10  µg of calcium per 1  mL medium; 150  µg
of calcium per unit). In total, three consecutive runs
were performed: (i)  with CPM-A and CPP-A labeled
during their synthesis; (ii)  with CPM-A and CPP-A
labeled after the synthesis; and (iii)  with unlabeled
CPM-A and CPP-A. ECs were incubated with CPM-A
and CPP-A for 1  h; nuclei were counterstained with
Hoechst 33342 (2  µg/mL, Thermo Fisher Scientific) for
5  min. FITC-labeled CPM-A and CPP-A were visualized
after thorough washing by confocal microscopy (LSM
700, Carl Zeiss, Germany).
To investigate colocalization of lysosomes and
FITC-labeled CPMs and CPPs, CPM-A, CPM-F, CPP-A,
and CPP-F were labeled with FITC after their synthesis
as described above. FITC-labeled CPM-A, CPM-F, CPP-A,
and CPP-F (10  µg calcium per 1  mL medium, 4  µg cal-
cium per well) were added to confluent of HCAECs
and HITAECs seeded into 8-well chambers (80826,
Ibidi) for 3  h, and then replaced the medium with a
fresh one containing the pH sensor LysoTracker Red
(1  µmol/L; Thermo Fisher Scientific) for 1  h. Unbound
FITC-BSA (60  µg) was used as a control; nuclei were
counterstained with Hoechst 33342 for 10  min. FITC-
labeled CPM-A, CPM-F, CPP-A, and CPP-F were visual-
ized after thorough washing by confocal microscopy.
Treatment of ECs and monocytes with free Ca
2+
ions, CPMs, and CPPs. To investigate the response of
ECs to equal calcium concentrations delivered by dif-
ferent distinct vehicles, we added DPBS (control), free
Ca
2+
ions (CaCl
2
as a vehicle), CPMs (either CPM-A or
CPM-F), or CPPs (either CPP-A or CPP-F) (10  µg of cal-
cium per 1  mL cell culture medium; 20  µg calcium per
well of a 6-well plate; n =  18 wells per group) to con-
fluent HCAEC and HITAEC cultures for 24  h. We also
added BSA (12  mg; i.e., average mass of albumin
SHISHKOVA et al.136
BIOCHEMISTRY (Moscow) Vol. 90 No. 1 2025
in added CPM-A) or BSF (0.33  mg; i.e., average mass
of fetuin-A in CPM-F) to all wells in the respective
experiments for negating potential protective effects
of these proteins. Serum-supplemented EndoBoost
medium was replaced with serum-free EndoLife me-
dium immediately before starting the experiment.
After incubation for 24  h, the cells were examined
by phase contrast microscopy; cell culture medium
was removed, and the cells were washed with ice-
cold (4°C) DPBS and lysed in TRIzol reagent (Thermo
Fisher Scientific) to extract RNA according to the man-
ufacturers protocols. Cell culture medium was centri-
fuged at 2000g (MiniSpin Plus, Eppendorf, Germany)
to remove cell debris, transferred into new tubes, and
frozen at −80°C.
To evaluate the cytotoxicity of different modali-
ties of calcium stress, we conducted colorimetric as-
say using water-soluble tetrazolium salt (WST)-8 and
annexin V/propidium iodide staining followed by flow
cytometry. For the WST-8 assay, HCAECs and HITAECs
were grown in 96-well plates (Wuxi NEST Biotechnol-
ogy) to confluency serum-supplemented EndoBoost
medium; next, the culture medium was replaced with
serum-free EndoLife medium, and added DPBS (con-
trol), free Ca
2+
ions (as CaCl
2
), CPMs (either CPM-A or
CPM-F), or CPPs (either CPP-A or CPP-F) were added
to the wells (10  µg calcium per 1  mL cell culture me-
dium; 2  µg calcium per well of 96-well plate; n = 12
wells per group) for 24  h. Next, the medium was re-
placed with 100  µL of fresh serum-free EndoLife me-
dium and 10  µL of WST-8 reagent (Wuhan Servicebio
Technology, China) was added for 2  h. The products
of reaction were detected spectrophotometrically
at 450 nm.
For annexin V/propidium iodide staining, HCAECs
and HITAECs were seeded into 6-well plates (Wuxi
NEST Biotechnology) and grown to confluency in se-
rum-supplemented EndoBoost medium. Next, the me-
dium was replaced with serum-free EndoLife medium,
and DPBS (control), free Ca
2+
ions (using CaCl
2
as a
vehicle), CPMs (either CPM-A or CPM-F), or CPPs (ei-
ther CPP-A or CPP-F) were added to the wells (10  µg
calcium per 1  mL cell culture medium, 20  µg calcium
per well of 6-well plate) for 24  h. The cells were then
detached using Accutase (Capricorn Scientific) and an-
alyzed by the annexin V/propidium iodide assay using
a respective kit (ab14085, Abcam, United Kingdom) ac-
cording to the manufacturers protocol. Flow cytome-
try was conducted with a CytoFlex instrument using
the CytExpert software (Beckman Coulter).
To study the monocyte response, we incubat-
ed monocytes (350,000 cells per unit) in serum-free
EndoLife medium with equal concentrations of free
Ca
2+
ions (CaCl
2
), CPM-A, or CPP-A (10  µg calcium per
1  mL culture medium; 150  µg calcium per unit; n = 5
donors/runs per group) in a flow culture system using
the above-mentioned perfusion set for 24  h. Similar
to the previous experiment, DPBS was used as a con-
trol and BSA (87  mg, an average mass of albumin in
added CPM-A) was added to all units for negating its
potential protective effects. Four experimental groups
(DPBS, Ca
2+
, CPM-A, and CPP-A) were distributed across
four units of the flow culture system. The experiment
was performed under sterile conditions. After 24  h of
incubation, cell culture medium was collected, centri-
fuged at 220g (5804R, Eppendorf) to sediment mono-
cytes and then at 2000g to remove cell debris, and
then frozen at −80°C.
Gene expression analysis. Gene expression in
Ca
2+
, CPM-A/CPM-F, or CPP-A/CPP-F-treated HCAECs
and HITAECs was analyzed by reverse transcrip-
tion-polymerase chain reaction (RT-qPCR). Briefly,
cDNA was synthesized with M-MuLV–RH First Strand
cDNA Synthesis Kit (R01-250, Evrogen, Russia) and
reverse transcriptase M-MuLV–RH (R03-50, Evrogen),
and RT-qPCR was carried out with customized prim-
ers (500nmol/L each, Evrogen, TableS1 in the Online
Resource  1), (20  ng), and BioMaster HS-qPCR Lo-ROX
SYBR Master Mix (MHR031-2040, Biolabmix, Russia)
according to the manufacturers protocol. The levels
of mRNAs (VCAM1, ICAM1, SELE, SELP, IL6, CXCL8,
CCL2, CXCL1, MIF, NOS3, SNAI1, SNAI2, TWIST1, and
ZEB1 genes) were quantified by calculating ΔCt using
the 2
−ΔΔCt
method and normalized to the average ex-
pression level of three housekeeping genes (GAPDH,
ACTB, and B2M) and to the DPBS-treated group (2
−ΔΔCt
).
Administration of free Ca
2+
ions, CPMs, and
CPPs to Wistar rats. The animal study protocol was
approved by the Local Ethical Committee of the Re-
search Institute for Complex Issues of Cardiovascular
Diseases (protocol code, 042/2023; date of approval,
April 4, 2023). Animal experiments were performed in
accordance with the European Convention for the Pro-
tection of Vertebrate Animals (Strasbourg, 1986) and
Directive 2010/63/EU of the European Parliament on
the protection of animals used for scientific purpos-
es. Male Wistar rats (body weight, ~300  g; estimated
blood volume, ~20  mL, i.e., 6.5% of body weight) were
used in the experiments. To investigate the response
to the intravenous administration of various calcium
sources, DPBS (control), free Ca
2+
ions (CaCl
2
), CPM-A,
or CPP-A (10  µg calcium per 1  mL rat blood; 200  µg
calcium per rat; n =  5 rats per group, n =  20 rats in
total) were injected into the rat tail vein. BSA was
added to all injections (average mass of albumin add-
ed to CPM-A, 120  mg,) for adjustment of the possible
immune response to BSA. After 1  h, all rats were eu-
thanized by intraperitoneal injection of sodium pento-
barbital (100  mg/kg body weight). Serum was obtained
by centrifuging rat blood at 1700g for 15  min.
Dot blotting and enzyme-linked immunosor-
bent assay (ELISA). Protein levels in the cell culture
CALCIPROTEIN PARTICLES 137
BIOCHEMISTRY (Moscow) Vol. 90 No. 1 2025
medium were measured by dot blotting and ELISA.
Dot blotting was conducted using Proteome Profiler
Human XL Cytokine Array Kit (ARY022B, R&D Sys-
tems, USA) and Proteome Profiler Rat XL Cytokine
Array (ARY030, R&D Systems) according to the manu-
facturers instructions; proteins were visualized using
chemiluminescence detection with an Odyssey XF im-
aging system (LI-COR Biosciences, USA). Densitometric
quantification was performed using the ImageJ soft-
ware (National Institutes of Health, USA). To increase
dot blotting sensitivity, cell culture medium was con-
centrated using HyperVAC-LITE vacuum centrifugal
concentrators (Gyrozen, Republic of Korea) before
the measurements. Rat serum was assessed without
preliminary concentrating. All culture medium sam-
ples were concentrated to the same extent: 7-fold from
medium from monocytes (from 14  mL to 2  mL) and
3-fold for medium from endothelial cells (from 3  mL
to 1  mL). Next, 1  mL of the concentrated medium or
non-concentrated rat serum were loaded for dot blot-
ting. The content of IL-8, IL-6, and MCP-1/CCL2 was
determined by ELISA using the corresponding kits
(A-8768, A-8762, and A-8782, Vector-Best, Russia) ac-
cording to the manufacturers protocols. Colorimetric
detection of ELISA results was conducted spectro-
photometrically at 450 nm. For ELISA measurement,
100  µL of non-concentrated cell culture medium was
used for all samples.
Statistical analysis was performed with Graph-
Pad Prism 8 (GraphPad Software, USA). For RT-qPCR,
the data are presented as mean ± standard deviation
(SD). Four independent groups were compared by the
ordinary one-way analysis of variance (ANOVA) and
subsequent Dunnett’s multiple comparison test with
a single pooled variance. The results of ELISA mea-
surements are presented as median, 25th and 75th
percentiles, and range. Four independent groups were
compared by the Kruskal–Wallis test with subsequent
Dunn’s multiple comparison test. The differences were
considered as statistically significant at p ≤ 0.05.
RESULTS
Physiological relevance of CPM and CPP syn-
thesis under conditions of mineral stress. To inves-
tigate the effects of different calcium delivery forms
on ECs and monocytes, we created a rection mixture
containing physiological concentration of BSA, phys-
iological saline (NaCl), and supraphysiological levels
of Na
2
HPO
4
, and CaCl
2
for simultaneous generation
of albumin-centric CPMs (CPM-A) and CPPs (CPP-A).
Previously, similar mineral stress conditions have
been used to produce fetuin-centric CPMs (CPM-F)
and CPPs (CPP-F) [18]. Next, we used ultracentrifuga-
tion to isolate CPPs followed by ultrafiltration to sep-
arate CPMs (yellow retentate) from free ions and salts
(transparent filtrate). Therefore, calcium was repre-
sented by (i)free Ca
2+
ions, (ii) CPMs (colloidal form),
and (iii) CPPs (corpuscular form). We used albumin
to assemble CPMs (CPM-A) and CPPs (CPP-A) because
a below-median content of serum albumin has been
demonstrated as an independent risk factor for the
coronary artery disease and ischemic stroke (in con-
junction with above-median serum levels of Ca
2+
) [11].
Low serum albumin levels were found to correlate
with a higher serum calcification propensity (i.e., CPP
precipitation), while the content of albumin showed
a positive correlation with total calcium (fetuin and
phosphate did not display such associations) [11]. How-
ever, because fetuin-A plays a pivotal role as a mineral
chaperone and governs formation of CPMs and CPPs in
human blood, we also used CPM-F and CPP-F in most
of the experiments. CPM-F and CPP-F were synthesized
using the protocol described except that bovine serum
fetuin (BSF) was used instead of BSA.
Scanning electron microscopy of CPP-A showed
their sponge-like structure and irregular shape, which
differed from spherical and needle-shaped appearance
of primary and secondary blood-derived CPPs, respec-
tively (Fig.1). CPP-F had a spherical shape and sponge-
like structure, thus closely resembling atherosclerotic
plaque- and serum-derived primary CPPs [11]. These
observations were in agreement with our previous
data on the comparison of albumin-centric, fetuin-cen-
tric, plaque-derived, and serum-derived CPPs [8] and
can be explained by the different cooperation of acidic
serum proteins during CPP generation in the blood.
CPPs and CPMs absorbed ~30 and ~20% of calci-
um, respectively, whereas ~50% of calcium remained
in the solution as free Ca
2+
ions. This distribution was
in agreement with the physiological ratio between ion-
ized calcium (Ca
2+
) and protein- and phosphate-bound
calcium in human serum (1  :  1). CPPs contained from
11 to 17% of total albumin, whereas 83 to 89% of al-
bumin remained in the retentate, thus retaining the
Ca
2+
-binding ability. The filtrate contained no BSA or
BSF, which confirmed the efficiency of the ultrafiltra-
tion procedure. Taken together, these data confirmed
the physiological relevance of the procedure devel-
oped for artificial synthesis of CPMs and CPPs under
conditions of mineral stress.
Physiological concentrations of CPPs cause
pro-inflammatory activation of ECs and monocytes.
To determine the amount of calcium that has to be
added to ensure physiological elevation in the ion-
ized calcium content, we calculated the dose-response
curve. Thus, an addition of 10  µg of calcium per 1  mL
of serum-free cell culture medium (Fig.  2a) or rat se-
rum (Fig.  2b) was sufficient to achieve a 10% increase
in the concentration of ionized calcium (i.e., the in-
terquartile range between the risk and protective
SHISHKOVA et al.138
BIOCHEMISTRY (Moscow) Vol. 90 No. 1 2025
Fig.  1. Scanning electron microscopy images of albumin-centric (CPP-A), fetuin-centric (CPP-F), calcified atherosclerotic
plaque-derived (CPP-PD), and serum-derived (CPP-SD) CPPs. Secondary electron mode; acceleration voltage, 10  kV (CPP-A)
or 30 kV (CPP-F, CPP-PD, and CPP-SD); magnification, ×30,000; scale bar: 1 µm.
Fig. 2. Increase in the ionized calcium (Ca
2+
) concentration in (a) cell culture medium and (b) rat serum upon addition
of increasing amounts of CaCl
2
; x-axis, concentration of added calcium; y-axis, increase in Ca
2+
concentration relative to
the control medium or serum without calcium addition. An increase in the Ca
2+
concentration by 10% (blue dashed line)
was achieved by the addition of 10 µg of calcium per 1 mL of cell culture medium or rat serum (red circle).
CALCIPROTEIN PARTICLES 139
BIOCHEMISTRY (Moscow) Vol. 90 No. 1 2025
Fig. 3. Internalization of FITC-BSA-labeled CPMs (FITC-CPMs) and CPPs (FITC-CPPs) by HCAECs and HITAECs. a) Comparison
of signal intensities of internalized FITC-CPMs and FITC-CPPs obtained by two different labeling techniques. ECs were treat-
ed for 1  h with unlabeled CPMs and CPPs (left panel), CPMs and CPPs that incorporated FITC-BSA during their synthesis
(central panel), and CPMs and CPPs that were incubated with FITC-BSA after their synthesis (right paned). Nuclei were
counterstained with Hoechst 33342. Confocal microscopy; magnification, ×630; scale bar, 5  µm. b) Lysosomes stained with
LysoTracker Red (LTR) in ECs treated for 4 h with CPMs (FITC-CPM-A and FITC-CPM-F) or CPPs (FITC-CPP-A and FITC-CPP-F):
left panel, free FITC-BSA; central panel: CPM-A and CPM-F co-incubated with FITC-BSA during their synthesis; right panel,
CPP-A and CPP-F co-incubated with FITC-BSA after their synthesis. Yellow arrows indicate CPP-A and CPP-F inside the cells.
Nuclei were counterstained with Hoechst 33342. Confocal microscopy; magnification, ×200; scale bar, 50  µm.
SHISHKOVA et al.140
BIOCHEMISTRY (Moscow) Vol. 90 No. 1 2025
Fig.  4. Bright-field microscopy (CPM-A/CPP-A, top) and phase-contrast microscopy (CPM-F/CPP-F, bottom) of HCAECs (left
panels) and HITAECs (right panels) treated with DPBS (control), free Ca
2+
ions, CPMs (CPM-A, top; CPM-F, bottom), or CPPs
(CPP-A, top; CPP-F, bottom) (10 µg of calcium per 1 mL serum-free cell culture medium) for 24 h; magnification, ×200; scale
bar, 100 µm.
quartiles in the population). Hence, we selected 10  µg/mL
as the optimal calcium concentration to model clini-
cally relevant mineral stress. Further experiments in-
cluded four groups: 1)  control (DPBS); 2)  free Ca
2+
ions
delivered as CaCl
2
; 3)  either CPM-A or CPM-F; 4)  either
CPP-A or CPP-F.
We then asked whether CPMs are internalized in
a flow system in a similar manner as CPPs. To address
this question, we labeled CPM-A and CPP-A with FITC-
BSA either during CPM-A/CPP-A generation (by adding
FITC-BSA to the solution) or after their formation by
incubation of sedimented CPP-A and separated CPM-A
with FITC-BSA. An intense green fluorescence was ev-
ident in ECs already 1  h after addition of FITC-labeled
CPM-A and CPP-A to the flow culture system (Fig. 3a).
CPM-A and CPP-A incubated with FITC-BSA after their
CALCIPROTEIN PARTICLES 141
BIOCHEMISTRY (Moscow) Vol. 90 No. 1 2025
Fig.  5. Cytotoxicity assay after incubation of HCAECs (left panel) and HITAEC (right panel) with DPBS (control), free Ca
2+
ions, CPMs (CPM-A or CPM-F), or CPPs (CPP-A or CPP-F) (10  µg of calcium per 1  mL serum-free cell culture medium)
for 24  h: a) WST-8 colorimetric assay (evaluation of WST-8 reduction by intracellular dehydrogenases to a water-soluble
orange- yellow formazan compound with the maximum absorption at 450  nm). b) Annexin V and propidium iodide assay
(lower left quadrant Q2-LL, normal cells; lower right quadrant Q2-LR, early apoptotic cells; upper right quadrant Q2-UR,
late apoptotic cells; upper left quadrant Q2-UL, necrotic cells). Top panel, statistical analysis of the content of intact and
late apoptotic cells. Bottom panel: representative flow cytometry plots.
SHISHKOVA et al.142
BIOCHEMISTRY (Moscow) Vol. 90 No. 1 2025
synthesis produced a significantly higher fluorescence
signal comparison to those that incorporated FITC-
BSA during their synthesis (Fig. 3a). Similar fluores-
cence intensity for CPM-A and CPP-A suggested that
FITC-BSA had the same affinity for both these spe-
cies (Fig. 3a). Identification of FITC-labeled CPMs/CPPs
in lysosomes stained with the pH sensor LysoTrack-
er Red confirmed internalization of CPM-A, CPM-F,
CPP-A, and CPP-F by HCAECs and HITAECs after 4  h
of incubation, while FITC-BSA itself did not enter ECs
(Fig. 3b).
To compare the pathological effects of different
calcium delivery forms on ECs, we added Ca
2+
, CPM-A/
CPM-F, or CPP-A/CPP-F (10  µg/mL) to HCAECs and
HITAECs. Using bright-field and phase contrast mi-
croscopies, we observed pathological alterations (i.e.,
loss of intercellular contacts, cell shrinkage and de-
tachment) in ECs after incubation with CPP-A or CPP-F,
but not with Ca
2+
or CPM-A/CPM-F (Fig. 4).
To further investigate the cytotoxicity of CPP-A
and CPP-F, we assessed a decrease in the cell viability
and metabolism in different modes of calcium stress
using WST-8 colorimetric assay. After 24-hour incuba-
tion with CPP-A or CPP-F, the intensity of cell metab-
olism dropped in both HCAECs and HITAECs (Fig. 5a).
Flow cytometry analysis of cell death using annexin V
and propidium iodide staining identified that a signifi-
cant proportion of ECs underwent apoptosis after 24  h
of treatment with CPP-A or CPP-F (Fig. 5b).
RT-qPCR demonstrated a significant increase in
the expression of genes encoding cell adhesion mol-
ecules (VCAM1, ICAM1, and SELE) and pro-inflam-
matory cytokines (IL6, CXCL8, CCL2, and CXCL1) in
HCAECs treated with CPP-A (Table  1). Exposure to
CPP-F triggered a similar response, which included el-
evated expression of VCAM1, SELP, IL6, and MIF genes
along with a trend towards a significant increase in
the expression of ICAM1 and SELE genes (Table  2).
Table 1. Relative gene expression (ΔCt; fold change; p-value) in HCAECs and HITAECs treated with DPBS (control),
free Ca
2+
ions, CPM-A, or CPP-A (10  µg of calcium per 1  mL of serum-free cell culture medium) for 24  h
Gene Metrics HCAEC HITAEC
DPBS Ca
2+
CPM-A CPP-A DPBS Ca
2+
CPM-A CPP-A
VCAM1
ΔCt
0.0003 ±
0.0006
0.0001 ±
0.0001
0.0002 ±
0.0001
0.0015 ±
0.0010
0.0003 ±
0.0002
0.0011 ±
0.0011
0.0006 ±
0.0004
0.0010 ±
0.0008
fold
change
1 0.52 0.76 6.00 1 3.67 2.00 3.33
p-value 1.00 0.904 0.985 0.001 1.00 0.009 0.463 0.029
ICAM1
ΔCt
0.0148 ±
0.0066
0.0372 ±
0.0210
0.0118 ±
0.0034
0.1169 ±
0.0837
0.0338 ±
0.0213
0.0503 ±
0.0339
0.0404 ±
0.0244
0.0432 ±
0.0201
fold
change
1 2.52 0.80 7.90 1 1.49 1.20 1.28
p-value 1.00 0.320 0.994 0.001 1.00 0.148 0.782 0.559
SELE
ΔCt
0.0056 ±
0.0026
0.0089 ±
0.0036
0.0020 ±
0.0009
0.0134 ±
0.0065
0.0595 ±
0.0351
0.1049 ±
0.1387
0.0909 ±
0.1203
0.0943 ±
0.0872
fold
change
1 1.59 0.36 2.39 1 1.76 1.53 1.58
p-value 1.00 0.041 0.026 0.001 1.00 0.415 0.687 0.619
SELP
ΔCt
0.0077 ±
0.0067
0.0027 ±
0.0008
0.0015 ±
0.0009
0.0026 ±
0.0023
0.0009 ±
0.0006
0.0054 ±
0.0058
0.0056 ±
0.0055
0.0025 ±
0.0030
fold
change
1 0.35 0.19 0.34 1 6.00 6.22 2.78
p-value 1.00 0.001 0.001 0.001 1.00 0.008 0.005 0.567
CALCIPROTEIN PARTICLES 143
BIOCHEMISTRY (Moscow) Vol. 90 No. 1 2025
Table 1 (cont.)
Gene Metrics HCAEC HITAEC
DPBS Ca
2+
CPM-A CPP-A DPBS Ca
2+
CPM-A CPP-A
IL6
ΔCt
0.0182 ±
0.0131
0.0058 ±
0.0023
0.0072 ±
0.0045
0.1197 ±
0.0837
0.0085 ±
0.0043
0.0132 ±
0.0155
0.0197 ±
0.0214
0.0247 ±
0.0272
fold
change
1 0.32 0.40 6.58 1 1.55 2.32 2.91
p-value 1.00 0.729 0.796 0.001 1.00 0.803 0.196 0.035
CXCL8
ΔCt
0.0371 ±
0.0260
0.0441 ±
0.0152
0.0250 ±
0.0105
2.1412 ±
1.5287
0.1396 ±
0.0561
0.1801 ±
0.2005
0.1825 ±
0.1871
0.3279 ±
0.3681
fold
change
1 1.19 0.67 57.71 1 1.29 1.31 2.35
p-value 1.00 0.999 0.999 0.001 1.00 0.914 0.901 0.045
CCL2
ΔCt
0.7514 ±
0.6502
0.4398 ±
0.4293
0.6965 ±
0.6669
1.3616 ±
1.0636
0.8908 ±
0.4072
1.2866 ±
1.5286
1.4987 ±
1.6929
1.6162 ±
1.9876
Fold
change
1 0.59 0.93 1.81 1 1.44 1.68 1.81
p-value 1.00 0.448 0.992 0.042 1.00 0.777 0.494 0.353
CXCL1
ΔCt
0.1267 ±
0.0562
0.0436 ±
0.0408
0.0444 ±
0.0343
0.3486 ±
0.1551
0.0647 ±
0.0279
0.1520 ±
0.1842
0.0944 ±
0.0885
0.0983 ±
0.1052
fold
change
1 0.34 0.35 2.75 1 2.35 1.46 1.52
p-value 1.00 0.017 0.018 0.001 1.00 0.069 0.782 0.715
MIF
ΔCt
0.3853 ±
0.1660
0.2309 ±
0.1040
0.2731 ±
0.0839
0.4170 ±
0.2857
0.2753 ±
0.1576
0.7919 ±
0.9280
0.5270 ±
0.4019
0.4618 ±
0.4146
fold
change
1 0.60 0.71 1.08 1 2.88 1.91 1.68
p-value 1.00 0.031 0.155 0.910 1.00 0.018 0.386 0.618
NOS3
ΔCt
0.0094 ±
0.0063
0.0069 ±
0.0036
0.0069 ±
0.0033
0.0091 ±
0.0087
0.0031 ±
0.0018
0.0093 ±
0.0090
0.0119 ±
0.0126
0.0060 ±
0.0037
fold
change
1 0.73 0.73 0.97 1 3.00 3.84 1.94
p-value 1.00 0.473 0.475 0.997 1.00 0.050 0.005 0.547
SNAI1
ΔCt
0.0168 ±
0.0101
0.0100 ±
0.0040
0.0124 ±
0.0065
0.0344 ±
0.0291
0.0049 ±
0.0020
0.0129 ±
0.0110
0.0140 ±
0.0124
0.0094 ±
0.0100
fold
change
1 0.60 0.74 2.05 1 2.63 2.86 1.92
p-value 1.00 0.487 0.771 0.009 1.00 0.042 0.018 0.371
SHISHKOVA et al.144
BIOCHEMISTRY (Moscow) Vol. 90 No. 1 2025
Table 1 (cont.)
Gene Metrics HCAEC HITAEC
DPBS Ca
2+
CPM-A CPP-A DPBS Ca
2+
CPM-A CPP-A
SNAI2
ΔCt
0.0129 ±
0.0103
0.0038 ±
0.0009
0.0065 ±
0.0046
0.0047 ±
0.0032
0.0009 ±
0.0007
0.0033 ±
0.0022
0.0099 ±
0.0160
0.0030 ±
0.0055
fold
change
1 0.29 0.50 0.36 1 3.67 11.00 3.33
p-value 1.00 0.001 0.005 0.001 1.00 0.784 0.015 0.841
TWIST1
ΔCt
0.0015 ±
0.0012
0.0003 ±
0.0002
0.0002 ±
0.0001
0.0009 ±
0.0008
0.0004 ±
0.0004
0.0018 ±
0.0026
0.0037 ±
0.0077
0.0016 ±
0.0016
fold
change
1 0.20 0.13 0.60 1 4.50 9.25 4.00
p-value 1.00 0.001 0.001 0.150 1.00 0.742 0.170 0.874
ZEB1
ΔCt
0.2376 ±
0.1200
0.0779 ±
0.0561
0.1607 ±
0.0596
0.3277 ±
0.2237
0.1438 ±
0.0686
0.3697 ±
0.4382
0.4552 ±
0.4601
0.3559 ±
0.4082
fold
change
1 0.33 0.68 1.38 1 2.57 3.17 2.47
p-value 1.00 0.002 0.209 0.117 1.00 0.201 0.049 0.244
Note. Genes encoding pro-inflammatory cell adhesion molecules (VCAM1, ICAM1, SELE, SELP), pro-inflammatory cytokines
(IL6, CXCL8, CCL2, CXCL1, MIF), endothelial nitric oxide synthase (NOS3), and endothelial-to-mesenchymal transition tran-
scription factors (SNAI1, SNAI2, TWIST1, ZEB1) were analyzed. Significant fold change values and p-values are shown in
bold; ΔCt is shown as mean ± SD.
The same gene expression pattern was observed for
HITAECs, including CPP-A-induced activation of ex-
pression of VCAM1, IL6, and CXCL8 genes (Table 1),
while incubation with CPP-F upregulated expression
of SELE, SELP, CXCL1, and MIF genes (Table 2). Col-
lectively, these molecular signatures pointed towards
the development of pro-inflammatory endothelial
activation and suggested elevation in the content of
pro-inflammatory cytokines in cell culture medium.
In contrast to CPP-A/CPP-F, free Ca
2+
ions and CPM-A/
CPM-F induced a stochastic rather than a consis-
tent response which did not reflect endothelial dys-
function.
Treatment with CPP-A significantly increased ex-
pression of inducible endothelial pro-inflammatory
cytokines (IL-6, IL-8, and MCP-1/CCL2) in the cell cul-
ture medium for both HCAECs and HITAECs (Fig.  6),
while addition of free Ca
2+
ions and CPM-A caused no
stable pathological response at the protein level, thus
corroborating the results of gene expression profiling
(Fig. 6). Incubation with CPP-F also promoted expres-
sion of above-mentioned cytokines in HCAECs and
HITAECs, whereas free Ca
2+
ions and CPM-F caused
a cytokine response in HITAECs (Fig.  7).
To further explore the release of cytokines upon
calcium overload, we performed a semi-quantitative
dot blotting analysis of serum-free cell culture me-
dium from HCAECs and HITAECs treated with Ca
2+
,
CPM-A, and CPP-A. CPP-A increased production of
PAI-1 (also called serpin E1), chemokine (C-X-C mo-
tif) ligand 1 (CXCL1), MCP-1/CCL2, IL-8, macrophage
migration inhibitory factor (MIF), and soluble CD105
and CD147 proteins has been elevated in HCAECs, as
well as upregulated the synthesis of ST2 (suppression
of tumorigenicity  2) and RANTES/CCL5 [regulated
on activation, normal T  cell expressed and secreted/
chemokine (C-C motif) ligand  5] in HITAECs (Fig.  8
and Table S1 in the Online Resource 1). The levels of
soluble uPAR and MIP-3α/CCL20 were elevated in the
cell culture supernatant in both EC lines after incuba-
tion with CPP-A (Fig.  8 and Table  1), i.e., exposure to
CPP-A induced the release of 11 cytokines (Fig.  8 and
TableS1 in the Online Resource1). Six of these pro-in-
flammatory molecules (CXCL1, MCP-1/CCL2, MIF, uPAR,
sCD147, and ST2 protein) were also overrepresented
in the cell culture supernatant collected from CPM-A-
treated ECs, while five proteins [CXCL1, sCD147, ST2
protein, platelet-derived growth factor AA (PDGF-AA),
CALCIPROTEIN PARTICLES 145
BIOCHEMISTRY (Moscow) Vol. 90 No. 1 2025
Table  2. Relative gene expression (ΔCt, fold change, and p-value) in HCAECs and HITAECs treated with control
DPBS, free Ca
2+
ions, CPM-F, and CPP-F (5  µg of calcium per 1  mL of serum-free cell culture medium) for 24  h
Gene Metrics HCAEC HITAEC
DPBS Ca
2+
CPM-F CPP-F DPBS Ca
2+
CPM-F CPP-F
VCAM1
ΔCt
0.0020 ±
0.0015
0.0019 ±
0.0007
0.0018 ±
0.0009
0.0042 ±
0.0032
0.0004 ±
0.0003
0.0004 ±
0.0002
0.0005 ±
0.0003
0.0006 ±
0.0005
fold
change
1 0.95 0.90 2.1 1 0.95 1.28 1.54
p-value 1.00 0.998 0.984 0.008 1.00 0.995 0.628 0.113
ICAM1
ΔCt
0.0540 ±
0.0510
0.0165 ±
0.0133
0.0208 ±
0.0266
0.1011 ±
0.1201
0.0646 ±
0.0282
0.0906 ±
0.0503
0.0820 ±
0.0306
0.0885 ±
0.0519
fold
change
1 0.31 0.39 1.87 1 1.40 1.27 1.37
p-value 1.00 0.229 0.319 0.098 1.00 0.159 0.454 0.213
SELE
ΔCt
0.0048 ±
0.0037
0.0082 ±
0.0107
0.0028 ±
0.0019
0.0091 ±
0.0053
0.0018 ±
0.0007
0.0084 ±
0.0055
0.0069 ±
0.0032
0.0058 ±
0.0061
fold
change
1 1.71 0.58 1.90 1 4.67 3.83 3.22
p-value 1.00 0.441 0.879 0.375 1.00 0.001 0.003 0.023
SELP
ΔCt
0.0119 ±
0.0131
0.0060 ±
0.0060
0.0089 ±
0.0079
0.0318 ±
0.0276
0.0050 ±
0.0023
0.0032 ±
0.0016
0.0035 ±
0.0017
0.0078 ±
0.0053
fold
change
1 0.50 0.75 2.67 1 0.64 0.70 1.56
p
-value 1.00 0.534 0.895 0.002 1.00 0.227 0.374 0.025
IL6
ΔCt
0.0072 ±
0.0029
0.0100 ±
0.0097
0.0063 ±
0.0046
0.0233 ±
0.0188
0.0044 ±
0.0016
0.0045 ±
0.0045
0.0031 ±
0.0012
0.0064 ±
0.0042
fold
change
1 1.39 0.88 3.24 1 1.02 0.70 1.45
p-value 1.00 0.783 0.988 0.001 1.00 0.999 0.463 0.191
CXCL8
ΔCt
0.1330 ±
0.0572
0.2582 ±
0.1501
0.1892 ±
0.0573
0.1632 ±
0.0898
0.0294 ±
0.0175
0.0636 ±
0.0714
0.0366 ±
0.0185
0.0522 ±
0.0411
fold
change
1 1.94 1.42 1.23 1 2.16 1.24 1.78
p-value 1.00 0.001 0.203 0.668 1.00 0.053 0.924 0.272
CCL2
ΔCt
1.1073 ±
0.3168
2.8331 ±
1.9144
1.1255 ±
0.3747
1.3772 ±
0.8673
0.3096 ±
0.0759
0.5079 ±
0.3852
0.4625 ±
0.1941
0.5145 ±
0.3698
Fold
change
1 2.56 1.02 1.24 1 1.64 1.49 1.66
p-value 1.00 0.001 0.999 0.795 1.00 0.105 0.265 0.090
SHISHKOVA et al.146
BIOCHEMISTRY (Moscow) Vol. 90 No. 1 2025
Table 2 (cont.)
Gene Metrics HCAEC HITAEC
DPBS Ca
2+
CPM-F CPP-F DPBS Ca
2+
CPM-F CPP-F
CXCL1
ΔCt
0.3756 ±
0.1000
0.6205 ±
0.3781
0.4173 ±
0.1658
0.5612 ±
0.3499
0.0543 ±
0.0156
0.1053 ±
0.0885
0.0831 ±
0.0437
0.1477 ±
0.1538
fold
change
1 1.65 1.11 1.49 1 1.94 1.53 2.72
p-value 1.00 0.026 0.942 0.117 1.00 0.235 0.668 0.009
MIF
ΔCt
2.8076 ±
0.9811
2.6168 ±
1.6847
2.2889 ±
0.8340
5.5963 ±
3.2599
0.2999 ±
0.0856
0.4214 ±
0.3609
0.3627 ±
0.1458
0.6572 ±
0.5581
fold
change
1 0.93 0.82 1.99 1 1.41 1.21 2.19
p-value 1.00 0.983 0.763 0.001 1.00 0.583 0.904 0.007
NOS3
ΔCt
0.1599 ±
0.0597
0.1075 ±
0.0592
0.1686 ±
0.0643
0.2636 ±
0.0972
0.0176 ±
0.0056
0.0187 ±
0.0174
0.0170 ±
0.0060
0.0347 ±
0.0382
fold
change
1 0.67 1.05 1.65 1 1.06 0.97 1.97
p-value 1.00 0.082 0.969 0.001 1.00 0.997 0.999 0.050
SNAI1
ΔCt
0.0595 ±
0.0214
0.0513 ±
0.0284
0.0650 ±
0.0257
0.0943 ±
0.0377
0.0017 ±
0.0008
0.0020 ±
0.0017
0.0015 ±
0.0008
0.0031 ±
0.0024
fold
change
1 0.86 1.09 1.58 1 1.18 0.88 1.82
p-value 1.00 0.728 0.892 0.002 1.00 0.937 0.969 0.031
SNAI2
ΔCt
0.0162 ±
0.0088
0.0097 ±
0.0056
0.0196 ±
0.0100
0.0521 ±
0.0311
0.0003 ±
0.0003
0.0005 ±
0.0006
0.0002 ±
0.0001
0.0003 ±
0.0003
fold
change
1 0.60 1.21 3.22 1 1.57 0.63 1.01
p-value 1.00 0.596 0.901 0.001 1.00 0.451 0.856 0.999
TWIST1
ΔCt
0.0019 ±
0.0010
0.0016 ±
0.0009
0.0022 ±
0.0011
0.0029 ±
0.0014
0.00013 ±
0.0001
0.0002 ±
0.0001
0.0001 ±
0.0001
0.0003 ±
0.0003
fold
change
1 0.84 1.16 1.53 1 1.54 0.85 2.31
p-value 1.00 0.757 0.794 0.086 1.00 0.992 0.981 0.094
ZEB1
ΔCt
0.3158 ±
0.2627
0.2493 ±
0.0424
0.2738 ±
0.0783
0.2246 ±
0.2089
0.0468
± 0.0208
0.0734 ±
0.0970
0.0227 ±
0.0064
0.0589 ±
0.0651
fold
change
1 0.16 0.23 0.71 1 1.57 0.49 1.26
p-value 1.00 0.294 0.323 0.276 1.00 0.403 0.477 0.875
Note. For the analyzed genes, see note to Table 1.
CALCIPROTEIN PARTICLES 147
BIOCHEMISTRY (Moscow) Vol. 90 No. 1 2025
Fig. 6. Assessment of IL-6 (top panel), IL-8 (middle panel),
and MCP-1/CCL2 (bottom panel) in non-concentrated se-
rum-free cell culture medium from HCAECs and HITAECs
treated with DPBS (control; black), free Ca
2+
ions (blue),
CPM-A (green), and CPP-A (red) (10 µg of calcium per 1 mL
of serum-free cell culture medium) for 24 h (ELISA).
Fig.  7. Assessment of IL-6 (top panel), IL-8 (middle panel),
and MCP-1/CCL2 (bottom panel) in non-concentrated se-
rum-free cell culture medium from HCAECs and HITAECs
treated with DPBS (control, black), free Ca
2+
ions (blue),
CPM-F (green), and CPP-F (red) (10 µg of calcium per 1 mL
of serum-free cell culture medium) for 24 h (ELISA).
and RANTES/CCL5] were upregulated after addition of
CaCl
2
(Fig. 8 and Table S1 in the Online Resource 1).
The most upregulated molecules were soluble CD147
(also called extracellular matrix metalloproteinase
inducer, EMMPRIN or basigin; fold change, 11.41 in
HCAECs after exposure to CPP-A) and MIP-3α/CCL20
(fold change, 12.52 in HITAECs, also after exposure
to CPP-A).
Likewise, incubation of monocytes with CPP-A in-
duced release of serpin E1/PAI-1, CXCL1/growth regu-
lated protein alpha (GROα), chemokine (C-X-C motif)
ligand  5/epithelial neutrophil-activating protein  78
(CXCL5/ENA-78), adiponectin, neutrophil gelatinase-as-
sociated lipocalin (NGAL)/lipocalin-2, IL-6, chitinase  3-
like  1, apolipoprotein A-I, uPAR, MIP-3α/CCL20, and
MMP-9, in contrast to Ca
2+
and CPM-A, which triggered
SHISHKOVA et al.148
BIOCHEMISTRY (Moscow) Vol. 90 No. 1 2025
Fig.  8. Cytokine profiling of concentrated (3-fold) serum-free cell culture medium from HCAECs (top) and HITAECs (bottom)
treated with DPBS (control), free Ca
2+
ions, CPM-A, or CPP-A (10  µg of calcium per 1  mL of cell culture medium) for 24  h
(dot blotting). Green, serpin E1/PAI-1; light brown, CXCL1/GROα; gray, CD105/endoglin; dark blue, MCP-1/CCL2; red, IL-8;
violet, MIF; dark brown, uPAR; gold, MIP-3α/CCL20; light blue, CD147/ EMMPRIN/basigin; azure, ST2; pink, PDGF-AA; dark
green, RANTES/CCL5. Short, medium, and long arrows indicate fold change from 1.20 to 1.49, from 1.50 to 1.99, and ≥2.00,
respectively, as compared with the DPBS group.
stochastic alterations of cytokine release (Fig.  9 and
Table S2 in the Online Resource 1). CPP-A caused an
increased release of eleven abovementioned cytokines
into the milieu, while the effects of Ca
2+
and CPM-A
were limited to the induction of NGAL/lipocalin-2,
chitinase  3-like  1, and MMP-9 (Fig.  9 and Table S2
in the Online Resource  1). CXCL1, adiponectin, IL-6,
and apolipoprotein A-I were exclusively expressed
in the monocyte-derived culture medium upon the
incubation with CPP-P (amorphous primary CPPs).
CALCIPROTEIN PARTICLES 149
BIOCHEMISTRY (Moscow) Vol. 90 No. 1 2025
Fig. 9. Cytokine profiling of concentrated (7-fold) serum-free cell culture medium of human monocytes treated with DPBS
(control), free Ca
2+
ions, CPM-A, and CPP-A (10  µg of calcium per 1  mL of cell culture medium) for 24 hours (dot blotting).
Green, serpin E1/PAI-1; light brown, CXCL1/GROα); red, CXCL5/ENA-78; light blue, adiponectin; violet, NGAL/lipocalin-2;
dark blue, IL-6; sky blue, chitinase 3-like 1; pink, apolipoprotein A-I; brown, uPAR; gold, MIP-3α; dark green, MMP-9.
Short, medium, and long arrows indicate fold change from 1.20 to 1.49, 1.50 to 1.99, and ≥2.00, respectively, as compared
with the DPBS group.
Hence, we found that CPP-A caused pro-inflammatory
activation of ECs and monocytes, as evidenced by the
upregulation of expression of multiple cytokine genes
and increased release of corresponding proteins. Al-
though free Ca
2+
ions and CPM-A also induced release
of several cytokines by ECs, their pro-inflammatory
effects were less pronounced as compared to CPP-A
regardless of the cell line (HCAECs, HITAECs, and
monocytes).
All tested calcium delivery forms induce sys-
temic inflammatory response in rats in the ab-
sence of other cardiovascular risk factors. Finally,
we examined in vivo effects of various calcium stress
modalities after intravenous administration of CaCl
2
,
CPM-A, and CPP-A to normolipidemic and normoten-
sive Wistar rats (10  µg of calcium per 1  mL of blood).
In contrast to the in  vitro findings, all calcium delivery
forms caused an elevation in the content of pro-in-
flammatory cytokines in rat serum as evidenced by dot
blotting (22, 30, and 24 cytokines in the case of Ca
2+
,
CPM-A, and CPP-A injections, respectively; Fig.  10 and
Table S3 in the Online Resource  1). Among these mol-
ecules were granulocyte-macrophage colony-stimulat-
ing factor (GM-CSF), chemokines [chemokine (C-X3-C
motif) ligand  1 (CX3CL1, fractalkine), MCP-1/CCL2,
chemokine (C-X-C motif) ligand  7 (CXCL7), C-C motif
chemokine  11 (CCL11, eotaxin), C-C motif chemok-
ine  17 (CCL17)], serpin E1/PAI-1, matrix metallopro-
teinase  2 (MMP-2), matrix metalloproteinase  3 (MMP-3),
hepatokines [hepassocin, fetuin-A, fibroblast growth
factor (FGF-21), growth/differentiation factor  15
(GDF-15)], and proteins with pleiotropic effects [re-
ceptor for advanced glycation end-products (RAGE/
AGER), adiponectin, fibulin-3, galectin-1, and galec-
tin-3] (Fig.10 and Table S3 in the Online Resource 1).
Prolactin, GM-CSF, hepassocin, ciliary neurotroph-
ic factor (CNTF), MMP-3, CX3CL1/fractalkine, FGF-21,
fibuin-3, and GDF-15 were upregulated after all three
types of calcium intervention (Fig.  10 and Table  3
in the Online Resource  1). Yet, RAGE/AGER, fetuin-A,
MCP-1/CCL2, MMP-9, CCL17, and galectin-3 were up-
regulated exclusively after injections of CPM-A and
CPP-A, while release of hepatocyte growth factor
(HGF), CCL11/eotaxin, and galectin-1 was stimulated
only by CPM-A; serpin E1/PAI-1 was upregulated sole-
ly by CPP-A (Fig.  10 and Table  S3 in the Online Re-
source1). Among the cytokines overrepresented in the
cell culture medium from the CPM-A-treated ECs and
monocytes, MCP-1/CCL2 and MMP-9 remained elevat-
ed in the serum of rats injected with either CPM-A or
CPP-A (Table 3). The content of three proteins (uPAR,
CXCL1/GROα, and MIP-3α/CCL20) was increased
SHISHKOVA et al.150
BIOCHEMISTRY (Moscow) Vol. 90 No. 1 2025
Fig. 10. Cytokine profiling of nonconcentrated serum from rats treated with DPBS (control), free Ca
2+
ions, CPM-A, and
CPP-A (10  µg of calcium per 1  mL of blood) for 1  h. Top panel (red box): green, prolactin; dark green, GM-CSF; black,
RAGE/AGER; light blue, hepassocin/fibrinogen-like protein  1 (FGL-1); violet, fetuin-A; dark blue, ciliary neurotrophic factor
(CNTF); gold, MMP-3; pink, hepatocyte growth factor (HGF); brown, chemokine (C-X3-C motif) ligand 1 (CX3CL1)/fractalkine;
red, MCP-1/CCL2; azure, MMP-9. Bottom panel (blue box): green, fibroblast growth factor 21 (FGF21); dark green, chemo-
kine (C-X-C motif) ligand 7 (CXCL7); black, fibulin-3; light blue, cysteine-rich angiogenic inducer 61/CCN family member  1
(Cyr61/CCN1); violet, C-C motif chemokine 11 (CCL11)/eotaxin; dark blue, serpin E1/PAI-1; gold, C-C motif chemokine  17
(CCL17)/thymus and activation-regulated chemokine (TARC); pink, galectin-1; brown, galectin-3; red, growth/differentiation
factor  15 (GDF-15); azure, Pref-1/delta like non-canonical Notch ligand  1 (DLK1)/fetal antigen-1 (FA1). Short, medium, and
long arrows indicate fold change from 1.20 to 1.49, 1.50 to 1.99, and ≥2.00, respectively, as compared with the DPBS group.
in the cell culture medium from ECs and monocytes
treated with CPP-A (Table 3). uPAR and MIP-3α/CCL20
were upregulated in the culture medium from all
three types of cells (HCAECs, HITAECs, and mono-
cytes), but not in the rat serum. Serpin E1/PAI-1 was
the only molecule that was upregulated in all samples
(EC- and monocyte-derived cell culture medium and
rat serum) after CPP-A treatment (Table 3).
DISCUSSION
Disturbed mineral homeostasis, e.g., reduction
in serum albumin and increase in serum calcium or
phosphate, is an independent cardiovascular risk fac-
tor [11]. It a manifestation of chronic kidney disease
which also promotes endothelial dysfunction via ele-
vating serum concentrations of urea and creatinine
[53,  54]. In addition to these biochemical triggers, en-
dothelial dysfunction is exacerbated by internalization
of CPPs by vascular ECs [8-15] and liver sinusoidal
ECs [17,  18] resulting in the development of chronic
low-grade inflammation [13,  14]. CPP internalization
is associated with the increased levels of pro-inflam-
matory cytokines, dysregulated balance between va-
soconstriction and vasodilation (including decreased
NO production), and endothelial-to-mesenchymal tran-
sition [55]. In particular, internalization of CPPs by ECs
and monocytes triggers the release of inducible endo-
thelial cytokines (IL-6, IL-8, MCP-1/CCL2, and soluble
CALCIPROTEIN PARTICLES 151
BIOCHEMISTRY (Moscow) Vol. 90 No. 1 2025
Table 3. Specific and common cytokines elevated in EC- or monocyte-derived cell culture medium or rat serum
upon the treatment with free Ca
2+
ions, CPM-A, and CPP-A (10 µg of calcium per 1 mL of cell culture medium
or blood) for 24 h (cells) or 1 h (rats) compared with the control (DPBS)
Calcium stress inducer
In vitro (serum-free culture
medium from HCAECs
and HITAECs)
In vitro (serum-free
culture medium
from monocytes)
In vivo (rat serum)
Free Ca
2+
ions
delivered by CaCl
2
CXCL1/GROα,
CD147/EMMPRIN/basigin,
ST2, PDGF-AA, MIP-3α/CCL20
NGAL/lipocalin-2,
chitinase 3-like 1,
MMP-9
prolactin, GM-CSF,
hepassocin/FGL1,
CNTF, MMP-3,
CX3CL1/fractalkine,
FGF-21, CXCL7, fibulin-3,
Cyr61/CCN, GDF-15
CPM-A
MCP-1/CCL2, CXCL1/GROα,
MIF, uPAR,
CD147/EMMPRIN/basigin,
ST2
MMP-9,
NGAL/lipocalin-2,
chitinase  3-like 1
MCP-1/CCL2, MMP-9,
prolactin, GM-CSF,
RAGE/AGER,
hepassocin/FGL1, fetuin-A,
CNTF, MMP-3, HGF,
CX3CL1/fractalkine,
FGF-21, CXCL7, fibulin-3,
Cyr61/CCN, CCL11/eotaxin,
CCL17/TARC, galectin-1,
galectin-3, GDF-15
CPP-A
Serpin E1/PAI-1, uPAR,
CXCL1/GROα, MIP-3α/CCL20,
MCP-1/CCL2,
CD105/endoglin, IL-8, MIF,
CD147/EMMPRIN/basigin,
ST2, RANTES/CCL5
Serpin E1/PAI-1,
uPAR, CXCL1/GROα,
MIP-3α/CCL20,
MMP-9, CXCL5/ENA-78,
adiponectin,
NGAL/lipocalin-2, IL-6,
chitinase  3-like  1,
apolipoprotein A-I
Serpin E1/PAI-1,
MCP-1/CCL2, MMP-9,
prolactin, GM-CSF,
RAGE/AGER,
hepassocin/FGL1, fetuin-A,
CNTF, MMP-3,
CX3CL1/fractalkine,
FGF-21, fibulin-3,
CCL17/TARC, galectin-3,
GDF-15, Pref-1/DLK1/FA1
CPM-A: upregulated in ECs
and rats
MCP-1/CCL2
CPM-A: upregulated
in monocytes and rats
MMP-9
CPP-A: upregulated in ECs
and monocytes
uPAR, CXCL1/GROα, MIP-3α/CCL20
CPP-A: upregulated in ECs
and rats
MCP-1/CCL2
CPP-A: upregulated
in monocytes and rats
MMP-9
CPP-A: upregulated in ECs,
monocytes, and rats
Serpin E1/PAI-1
intercellular adhesion molecule sICAM-1) and mono-
cyte-derived cytokines [MIP-1α, MIP-3α, cytokine-in-
duced neutrophil chemoattractant-1 (CINC-1), cyto-
kine-induced neutrophil chemoattractant-3 (CINC-3),
and C-X-C motif chemokine ligand  10 (CXCL10)] [13].
Since chronic low-grade inflammation and endothelial
dysfunction mutually promote each other [56-59], it is
difficult to distinguish the contribution of CPP-stimu-
lated ECs and monocytes, as well as specific cytokines
produced by these cells, to systemic inflammation.
According to the recent concepts, nanometer-
sized (~9-10  nm) CPMs act as building blocks for
SHISHKOVA et al.152
BIOCHEMISTRY (Moscow) Vol. 90 No. 1 2025
submicrometer-sized (~30-100  nm) primary CPPs (or
CPP-I), which, in turn, undergo aggregation and amor-
phous-to-crystalline transition to micrometer-sized
(100-300  nm) secondary CPPs (or CPP-II) [18,  27,  60,
61]. Initially amorphous and spherical, primary CPPs
mature into spindle- or needle-shaped crystalline sec-
ondary CPPs [18, 27, 60, 61]. While CPMs and primary
CPPs consist of amorphous calcium phosphate, second-
ary CPPs are composed of carbonate hydroxyapatite,
also called bioapatite. All these calcium and phosphate
scavengers can adsorb proteins from the surrounding
medium (e.g., circulating proteins from the blood) [18,
27,  60,  61]. Calcium and phosphate react with each
other in the presence of the mineral chaperone fe-
tuin-A and other acidic serum proteins with the for-
mation of amorphous calcium phosphate (CPMs or
primary CPPs) or carbonate hydroxyapatite (second-
ary CPPs) [22, 62-64]. CPPs act as mineral scavengers;
phosphate as an inherent component of hydroxyapa-
tite is an essential constituent of these compounds [22,
62-64]. It was reported that high phosphate concentra-
tions inhibit production of nitric oxide, thus impairing
vasodilation [65-67], induce apoptosis of endothelial
cells [65, 67], and promote systemic inflammation
in a dose-dependent manner by triggering oxidative
stress and stimulating expression of pro-inflammatory
cytokines [68]. Hence, high phosphate is considered as
an independent factor of endothelial dysfunction and
chronic low-grade inflammation [69-71].
Although it has been shown that exhaustion of
the serum Ca
2+
-binding capacity leads to the precip-
itation of CPPs and that mineral stress affects the
development of endothelial dysfunction and systemic
inflammation, it remained unclear whether the con-
sequences of calcium overload are solely determined
by the calcium amount or also by the form of calcium
delivery (free Ca
2+
ions, CPMs, and CPPs). Typically,
CPMs and CPPs are generated artificially by combin-
ing excessive concentrations of calcium and phosphate
with a protein source in a buffering solution. The
above question can be answered by applying a holis-
tic approach (by adding serum as a protein source)
or a reductionist approach (by adding a major serum
protein, e.g., albumin or fetuin-A, as a protein source).
The holistic approach recapitulates a scenario occur-
ring in human serum, while the reductionist approach
allows to analyze the mineral-buffering capacity of
each serum protein and to avoid potential negative
effects of other serum components. To better address
the task of modeling mineral stress, here we applied
the reductionist approach to synthesize simultaneous-
ly CPMs and CPPs. We chose BSA as a protein source
for CPM-A and CPP-A because (i)  the lower quartile
of serum albumin content is associated with cardio-
vascular disease and correlates with increased serum
calcification propensity  [11]; (ii)  the concentration of
serum albumin positively correlates with the concen-
tration of serum CPPs (measured with fluorescently
labeled bisphosphonate) and total calcium  [11]; (iii)  al-
bumin represents one of the two primary scavengers
of ionized calcium along with fetuin-A  [7]; (iv)  albu-
min is convenient to use as its yellow color facilitates
visual quality control. Beside BSA, we also used BSF
to synthesize CPM-F and CPP-F because fetuin-A is a
primary mineral chaperone that maintains generation
of CPMs and CPPs in human body [1-4].
When physiological concentrations of BSA were
mixed with supraphysiological concentrations of calci-
um and phosphate, the ratio (%) between the ionized,
protein-bound (CPMs), and phosphate-bound (CPPs)
calcium was 50  :  20  :  30, respectively. This validated
the physiological relevance of the implemented miner-
al stress model (as the distribution of ionized to bound
calcium was 1  :  1) and confirmed that circulating min-
eral depots – CPMs and CPPs – are able to maintain
their calcium-binding function at the physiological lev-
el even upon severe mineral stress. During mineral
stress, the amount of calcium in CPPs exceeded that
in CPMs, thus highlighting the primary role of CPPs
as mineral scavengers buffering the human blood
and controlling the level of ionized calcium. These
results are in accordance with the earlier reports on
the calcium ratio in CPPs and CPMs (1  :  1) [18,  72].
They suggest that CPPs represent an ultimate buffer
that aggregates excessive calcium and phosphate to
prevent blood supersaturation with ionized calcium
when other mineral buffers are exhausted. A relative-
ly low proportion of albumin bound to CPPs (~15%)
even at supraphysiological conditions of mineral stress
indicated that CPP generation, probably, does not af-
fect the functions of albumin in a living organism.
To compare the effects of different calcium vehi-
cles (CaCl
2
as a donor of Ca
2+
ions, CPM-A/CPM-F, and
CPP-A/CPP-F), we selected ECs and monocytes, which
are the first cell populations encountering CPPs in
an organism. In this study, we determined and used
the physiological dose of calcium (10  µg/mL), as this
parameter is strictly regulated in a body in order to
prevent arrhythmia and extraskeletal calcification
[73-75]. This dose induced a 10% increase in the ion-
ized calcium concentration, which corresponded to
the interquartile range of plasma ionized calcium in
the population (from 0.10 to 0.14 mmol/L, i.e., from
4.0 to 5.6  µg/mL) [11]. Internalization of CPPs is a
mandatory pre-requisite of their detrimental effects
[8-19]. We showed that CPMs are internalized by the
ECs in a flow similarly to CPPs [10, 13]. In accordance
with our previous findings [9-15], incubation of arte-
rial ECs with CPP-A upregulated expression of genes
encoding pro-inflammatory cell adhesion molecules
(VCAM1, ICAM1, and SELE) and pro-inflammatory cy-
tokines (IL6, CXCL8, CCL2, and CXCL1). The treatment
CALCIPROTEIN PARTICLES 153
BIOCHEMISTRY (Moscow) Vol. 90 No. 1 2025
of arterial ECs with CPP-F promoted expression of
VCAM1, ICAM1, SELE, SELP, IL6, MIF, and CXCL1
genes. Such molecular reconfiguration is typical for
dysfunctional ECs [55,  76,  77] and indicates develop-
ment of chronic low-grade inflammation [56-59] and
senescence-associated secretory phenotype [78-80].
Incubation of ECs and monocytes with CPP-A
or CPP-F (10  µg of calcium per 1 mL of cell culture
medium) promoted release of pro-inflammatory cyto-
kines. Among the molecules upregulated in arterial
ECs and monocytes after their exposure to CPP-A were
cytokines and chemokines (IL-6, IL-8, MCP-1/CCL2,
CXCL1/GROα, MIP-3α/CCL20), as well as pro- and an-
ti-thrombotic molecules (serpin E1/PAI-1 and uPAR).
Of these, uPAR, MIP-3α/CCL20, and serpin E1/PAI-1
were consistently upregulated in HCAECs, HITAECs,
and monocytes. To summarize the results of in  vitro
experiments, exposure to CPPs triggered the pro-in-
flammatory response characterized by the activation
of cytokine release and corresponding changes in the
gene expression. Although Ca
2+
, CPM-A, and CPM-F
also stimulated production of several cytokines by
ECs, this response was less pronounced and suggest-
ed limited pathogenic effects of these calcium sources.
Cytokine profiling by ELISA revealed a statistical-
ly significant elevation in the IL-6, IL-8, and MCP-1/
CCL2 production by both HCAECs and HITAECs after
their incubation with CPP-A. Yet, dot blot experiments
found an increase in the IL-8 and MCP-1/CCL2 content
exclusively in HCAECs; no IL-6 was detected in the
cell culture supernatant even after concentrating it
3-fold. The most probable reason for this discrepancy
is a limited sensitivity of semi-quantitative chemilu-
minescent dot blotting, as IL-6 levels did not exceed
175pg/mL (in comparison with 250-500pg/mL for IL-8
and 2500-6000pg/mL for MCP-1/CCL2). This suggestion
was partially confirmed in our previous study [13],
in which the concentration of IL-6 after 24-hour cul-
turing of CPP-treated ECs reached 300-450 pg/mL and
could be detected by dot blotting.
Previous studies consistently reported cytotox-
ic and pro-inflammatory (primarily NLRP3 inflam-
masome-mediated) effects of CPPs mediated by the
calcium and osmotic stress that resulted from a sharp
rise in the cytosolic Ca
2+
following the dissolution of
calcium in the lysosomes [11,  81,  82]. Bafilomycin A1,
a specific inhibitor of vacuolar H
+
ATPase (V-ATPase),
rescued ECs  [11] and vascular smooth muscle cells
[81] from the CPP-induced lysosome-dependent cell
death by preventing lysosomal acidification and
CPP dissolution. Similar cytoprotective effects were
reached by using inhibitors of calcium-sensing recep-
tor NLRP3 or caspase-1 during the calcium stress [82].
Likewise, pharmacological inhibition of cathepsin  B,
aprominent lysosomal protease, ameliorated the CPP-
related release of IL-1β by the macrophages  [25,  82].
Gene set enrichment analysis revealed an upregulation
of lysosome-related proteins, in particular, lysosomal
membrane proteins, in arterial ECs [13]. Moreover,
molecular terms related to the lysosome-mediated
calcium dissolution (e.g., vacuolar acidification, pH
regulation, regulation of proteolysis, Ca
2+
elevation
in cytosol, and mitochondrial outer membrane per-
meabilization) were also upregulated in CPP-treated
HCAECs and HITAECs [13]. Further proteomic analysis
suggested that lysosomal response to CPP internaliza-
tion involves pre-existing protein machinery rather
than employs transcriptional, post-transcriptional, and
translational regulation [15]. Although some studies
reported lysosomal alkalization and reduced hydro-
lase activity during CPP dissolution because of Ca
2+
overflow [26, 28], here we able to detected CPP-A
and CPP-F in EC lysosomes using standard pH sensor
(LysoTracker Red). Taken together lysosome-specific
distribution and cytotoxic and pro-apoptotic effects of
CPP-A and CPP-F, we suggested that their pathogenic
profile is similar to those of atherosclerotic plaque-
and serum-derived CPPs [8], as well as to CPP-P and
CPP-S (crystalline secondary CPPs) [9-13, 15].
Intravenous administration of Ca
2+
, CPM-A, or
CPP-A to Wistar rats free of other cardiovascular risk
factors, also triggered systemic inflammatory response
primarily mediated by chemokines (MCP-1/CCL2,
CX3CL1, CXCL7, CCL11, and CCL17), hepatokines (he-
passocin, fetuin-A, FGF-21, and GDF-15), proteases
(MMP-2 and MMP-3), and protease inhibitors (serpin
E1/PAI-1). Hence, our in  vitro and in  vivo findings sup-
ported a pronounced pro-inflammatory effect of CPPs.
For this study, we selected a 1-hour time point based
on the results of our previous works [13] and taking
into account rapid utilization of excessive calcium
from the blood through its clearance by acidic serum
proteins acting as Ca
2+
scavengers, removal of CPMs
by kidneys, and recycling of CPPs in the liver [5].
Notably, ionized calcium concentration represents one
of the most tightly regulated biochemical parameters in
the human blood (which can even be compared to pH),
as stable hypercalcemia is a relatively rare condition
and even transient hypercalcemia might lead to ar-
rhythmia [83-86]. Hence, the model of transient hyper-
calcemia, a condition that frequently occurs in patients
with hyperparathyroidism or excessive vitamin  D in-
take, is quite relevant [83-86]. The pathological effects
of calcium stress observed 1  h after the intravenous
administration of CaCl
2
, CPMs, or CPPs to Wistar rats,
were related to the low-grade systemic inflammation
defined as a minor to moderate increase in the cyto-
kine levels in the circulation. Such calcium stress-de-
rived transient increase leads to a pro-inflammatory
state which might negatively affect vascular health
by sustaining endothelial activation. If uncurbed (e.g.,
in elderly patients having more than one comorbid
SHISHKOVA et al.154
BIOCHEMISTRY (Moscow) Vol. 90 No. 1 2025
condition, such as diabetes mellitus or chronic kidney
disease), these transient elevations of pro-inflamma-
tory cytokines can contribute to the frailty syndrome,
in which biological age (i.e., age-related functional de-
cline) exceeds the chronological age [56-59].
Our results are in agreement with the data ob-
tained by Wilhelm Jahnen–Dechent’s group, who
showed the absence of CPM toxicity or pro-inflam-
matory effects after internalization by sinusoidal liver
ECs and proximal tubular epithelial cells, in contrast
to CPPs, which exhibited significant cytotoxic effects
and triggered rapid assembly of NLRP3 inflammasome
as early as within 2  h after CPP addition [18]. Com-
bined analysis of in  vitro and in  vivo results revealed
that serpin E1/PAI-1 was the only molecule upregulat-
ed in ECs, monocytes, and rat serum after CPP-A treat-
ment. The reasons behind this stable upregulation of
serpin E1/PAI-1 might include its relative abundance
(which permits to detect and increase in its release by
dot blotting) and calcium-dependent regulation. Other
cytokines overrepresented in the cell culture superna-
tant from the CPP-A/CPP-F-treated ECs or monocytes
or in the serum of CPP-A/CPP-F-treated rats (uPAR,
CXCL1/GROα, MIP-3α/CCL20, MCP-1/CCL2, and MMP-9)
exhibited less consistent expression across the exper-
imental models. For instance, uPAR and CXCL1/GR
produced only a moderate signal in the cell culture su-
pernatant from either ECs or monocytes, while MIP-3α/
CCL20 and MMP-9 were highly expressed in mono-
cytes but not in the control ECs. Similar to serpin E1/
PAI-1, MCP-1/CCL2 showed a relatively high expression
in all models but was not upregulated in monocytes.
Further, serpin E1/PAI-1 is produced [87,  88] and even
its activity is maintained [89] in a calcium-dependent
manner, suggesting an existence of a mechanism ensur-
ing activation of its release after the treatment with
CPP-A or CPP-F. Unfortunately, previous studies have
not examined production of serpin E1/PAI-1 bythe cell
populations after exposure to CPPs.
Future studies might further investigate the ef-
fects of CPMs and CPPs, considering a higher affin-
ity of fetuin-A for calcium and a unique function of
this protein as a mineral chaperone governing the
CPP formation, although the average fetuin-A level in
the serum (1  g/L) is significantly lower than that of
albumin (34  g/L) [90-93]. It might be promising to in-
vestigate combined effects of Ca
2+
and CPMs or CPPs,
as Ca
2+
-dependent calcium-sensing receptor promotes
internalization of CPPs  [39], which might exacerbate
their pathogenic effects. Another task is to define the
hierarchy in the mineral-binding ability of acidic se-
rum proteins (albumin, fetuin-A, osteonectin, osteo-
protegerin, osteopontin, matrix Gla protein, Gla-rich
protein, alpha-1-acid glycoprotein, transferrin, hapto-
globin, fibrinogen, ceruloplasmin, alpha-2-macroglob-
ulin, immunoglobulin  A, fibronectin, and antithrom-
bin  III). From the diagnostic viewpoint, differential
detection of CPMs and CPPs might be performed us-
ing the flow cytometry approach with fluorescently
labeled bisphosphonate (e.g., IVISense Osteo 680) and
artificially synthesized CPMs and CPPs used to set the
CPM- and CPP-specific gates. Measuring serum concen-
trations of CPMs and CPPs in healthy individuals and
in various diseases states might help to better under-
stand the pathophysiological importance of these pa-
rameters.
Our findings suggest that the adverse effects of
calcium stress are determined by the calcium delivery
mode rather than simply by the amount of calcium.
This might indicate the necessity to reconsider the
approaches for CPP quantification, albeit alternative
techniques (fluorescent labeling in combination with
flow cytometry, turbidimetry, nephelometry, dynam-
ic light scattering, and scanning electron microscopy)
are less standardized and their application for CPP
quantification in vitro is currently debated. Dynamic
light scattering and scanning electron microscopy are
time-consuming, while turbidimetry depends on the
particle-size distribution. Apparently, future studies
will need to address the development of new methods
for CPP quantification.
CONCLUSIONS
We found that physiological increase in the Ca
2+
concentration (by 10%, which is equal to the inter-
quartile range in a population) was achieved by add-
ing 10  µg of calcium to 1  mL of serum-free medium
or rat serum. Incubation of ECs and monocytes with
such amount of CPP-A or CPP-F initiated their pro-in-
flammatory activation that was manifested as tran-
scriptional reprogramming and increased release of
endothelium-derived (IL-6, IL-8, MCP-1/CCL2, uPAR,
MIP-3α/CCL20, serpin E1/PAI-1) and monocyte-derived
(IL-6, IL-8, MIP-1α/1β, MIP-3α/CCL20, uPAR, serpin E1/
PAI-1, CXCL1, CXCL5) cytokines. Addition of free Ca
2+
and CPM-A induced limited detrimental effects in ECs
and monocytes, although CPMs were internalized by
ECs in a flow similar to CPPs. All forms of calcium
delivery (free Ca
2+
ions, colloidal CPM-A, and cor-
puscular CPP-A) caused systemic inflammatory re-
sponse in normolipidemic and normotensive Wistar
rats (Ca
2+
: 22 cytokines, CPM-A: 30 cytokines, CPP-A:
24 cytokines). Serpin E1/PAI-1 was the only mole-
cule upregulated in all tested cells (ECs and mono-
cytes) and rat serum after the treatment with CPP-A.
The increase in the release of chemokines (CX3CL1,
MCP-1/CCL2, CXCL7, CCL11, CCL17) and hepatokines
(hepassocin, fetuin-A, FGF-21, GDF-15) at the back-
ground of upregulated cytokine expression suggested
chemokine burst and release of liver injury markers.
CALCIPROTEIN PARTICLES 155
BIOCHEMISTRY (Moscow) Vol. 90 No. 1 2025
Supplementary information. The online version
containing supplementary material is available at
https://doi.org/10.1134/S0006297924604064.
Contributions. Conceptualization, D.Sh. and A.K.
developed the study concept and performed data vali-
dation; D.Sh., Victoria M., Yu.M., M.S., A.S., Vera M., E.T.,
A.L., and A.S. developed the methodology, performed
the experiments, and analyzed the data; A.K. analyzed
and curated the data, acquired the funding, supervised
the project, prepared the figures, wrote and edited the
text of the article. All authors have read and agreed
to the published version of the manuscript.
Funding. This study was funded by the Russian
Science Foundation (project no. 22-15-00107 “Circula-
tion of calciprotein particles in human blood: patho-
genic consequences and molecular mechanisms”
toA.K.; https://rscf.ru/en/project/22-15-00107/).
Ethics approval and consent to participate.
Allanimal study protocols were approved by the Local
Ethical Committee of the Research Institute for Com-
plex Issues of Cardiovascular Diseases (protocol code:
042/2023; date of approval, April 4, 2023). All animal
experiments were performed in accordance with the
European Convention for the Protection of Vertebrate
Animals (Strasbourg, 1986) and Directive 2010/63/EU
of the European Parliament on the protection of ani-
mals used for scientific purposes.
Conflict of interest. The authors of this work de-
clare that they have no conflict of interest.
REFERENCES
1. Heiss, A., Pipich, V., Jahnen-Dechent, W., and
Schwahn,D. (2010) Fetuin-A is a mineral carrier pro-
tein: small angle neutron scattering provides new
insight on Fetuin-A controlled calcification inhibi-
tion, Biophys.J., 99, 3986-3995, https://doi.org/10.1016/
j.bpj.2010.10.030.
2. Jahnen-Dechent, W., Heiss, A., Schäfer,C., and Kettel-
er, M. (2011) Fetuin-A regulation of calcified matrix
metabolism, Circ. Res., 108, 1494-1509, https://doi.org/
10.1161/CIRCRESAHA.110.234260.
3. Jahnen-Dechent,W., Büscher,A., Köppert,S., Heiss,A.,
Kuro-O,M., and Smith, E.R. (2020) Mud in the blood:
the role of protein-mineral complexes and extracel-
lular vesicles in biomineralisation and calcification,
J. Struct. Biol., 212, 107577, https://doi.org/10.1016/
j.jsb.2020.107577.
4. Smith, E.R., Hewitson, T.D., and Jahnen-Dechent,W.
(2020) Calciprotein particles: mineral behaving bad-
ly? Curr. Opin. Nephrol. Hypertens., 29, 378-386, https://
doi.org/10.1097/MNH.0000000000000609.
5. Kutikhin, A.G., Feenstra,L., Kostyunin, A.E., Yuzhalin,
A.E., Hillebrands, J.L., and Krenning,G. (2021) Calcip-
rotein particles: balancing mineral homeostasis and
vascular pathology, Arterioscler. Thromb. Vasc. Biol., 41,
1607-1624, https://doi.org/10.1161/atvbaha.120.31569.
6. Jahnen-Dechent, W., and Pasch, A. (2023) Solving
the insoluble: calciprotein particles mediate bulk
mineral transport, Kidney Int., 103, 663-665, https://
doi.org/10.1016/j.kint.2023.01.011.
7. Bäck, M., Aranyi, T., Cancela, M. L., Carracedo, M.,
Conceição, N., Leftheriotis, G., Macrae, V., Martin, L.,
Nitschke, Y., Pasch, A., Quaglino, D., Rutsch, F.,
Shanahan, C., Sorribas, V., Szeri, F., Valdivielso, P.,
Vanakker, O., and Kempf, H. (2019) Endogenous cal-
cification inhibitors in the prevention of vascular
calcification: a consensus statement from the COST
Action EuroSoftCalcNet, Front. Cardiovasc. Med., 5,
196, https://doi.org/10.3389/fcvm.2018.00196.
8. Kutikhin, A. G., Velikanova, E. A., Mukhamadiyarov,
R. A., Glushkova, T. V., Borisov, V. V., Matveeva,
V. G., Antonova, L. V., Filip’ev, D. E., Golovkin, A. S.,
Shishkova, D. K., Burago, A. Y., Frolov, A. V., Dolgov,
V. Y., Efimova, O. S., Popova, A. N., Malysheva,
V. Y., Vladimirov, A. A., Sozinov, S. A., Ismagilov,
Z. R., Russakov, D. M., Lomzov, A. A., Pyshnyi, D. V.,
Gutakovsky, A. K., Zhivodkov, Y. A., Demidov, E. A.,
Peltek, S.E., Dolganyuk, V.F., Babich, O.O., Grigoriev,
E. V., Brusina, E. B., Barbarash, O. L., and Yuzhalin,
A. E. (2016) Apoptosis-mediated endothelial toxicity
but not direct calcification or functional changes in
anti-calcification proteins defines pathogenic effects
of calcium phosphate bions, Sci. Rep., 6, 27255, https://
doi.org/10.1038/srep27255.
9. Shishkova,D., Velikanova,E., Sinitsky,M., Tsepokina,A.,
Gruzdeva, O., Bogdanov, L., and Kutikhin, A. (2019)
Calcium phosphate bions cause intimal hyperpla-
sia in intact aortas of normolipidemic rats through
endothelial injury, Int. J. Mol. Sci., 20, 5728, https://
doi.org/10.3390/ijms20225728.
10. Shishkova,D., Markova,V., Sinitsky,M., Tsepokina,A.,
Velikanova, E., Bogdanov, L., Glushkova, T., and
Kutikhin, A. (2020) Calciprotein particles cause en-
dothelial dysfunction under flow, Int. J. Mol. Sci., 21,
8802, https://doi.org/10.3390/ijms21228802.
11. Shishkova, D. K., Velikanova, E. A., Bogdanov, L. A.,
Sinitsky, M. Y., Kostyunin, A. E., Tsepokina, A. V.,
Gruzdeva, O. V., Mironov, A. V., Mukhamadiyarov,
R. A., Glushkova, T. V., Krivkina, E. O., Matveeva,
V.G., Hryachkova, O.N., Markova, V.E., Dyleva, Y.A.,
Belik, E. V., Frolov, A. V., Shabaev, A. R., Efimova,
O. S., Popova, A. N., Malysheva, V. Y., Kolmykov,
R.P., Sevostyanov, O.G., Russakov, D. M., Dolganyuk,
V. F., Gutakovsky, A. K., Zhivodkov, Y. A., Kozhukhov,
A.S., Brusina, E.B., Ismagilov, Z.R., Barbarash, O.L.,
Yuzhalin, A.E., and Kutikhin, A.G. (2021) Calciprotein
particles link disturbed mineral homeostasis with car-
diovascular disease by causing endothelial dysfunc-
tion and vascular inflammation, Int. J. Mol. Sci., 22,
12458, https://doi.org/10.3390/ijms222212458.
SHISHKOVA et al.156
BIOCHEMISTRY (Moscow) Vol. 90 No. 1 2025
12. Bogdanov, L., Shishkova, D., Mukhamadiyarov, R.,
Velikanova,E., Tsepokina,A., Terekhov,A., Koshelev,V.,
Kanonykina, A., Shabaev, A., Frolov, A., Zagorod-
nikov, N., and Kutikhin, A. (2022) Excessive adven-
titial and perivascular vascularisation correlates
with vascular inflammation and intimal hyperpla-
sia, Int. J. Mol. Sci., 23, 12156, https://doi.org/10.3390/
ijms232012156.
13. Shishkova, D., Lobov, A., Zainullina, B., Matveeva, V.,
Markova,V., Sinitskaya,A., Velikanova,E., Sinitsky,M.,
Kanonykina, A., Dyleva, Y., and Kutikhin, A. (2022)
Calciprotein particles cause physiologically signifi-
cant pro-inflammatory response in endothelial cells
and systemic circulation, Int. J. Mol. Sci., 23, 14941,
https://doi.org/10.3390/ijms232314941.
14. Feenstra, L., Kutikhin, A. G., Shishkova, D. K., Buike-
ma, H., Zeper, L. W., Bourgonje, A. R., Krenning, G.,
and Hillebrands, J. L. (2023) Calciprotein particles
induce endothelial dysfunction by impairing endo-
thelial nitric oxide metabolism, Arterioscler. Thromb.
Vasc. Biol., 43, 443-455, https://doi.org/10.1161/
ATVBAHA.122.318420.
15. Shishkova, D., Lobov, A., Repkin, E., Markova, V.,
Markova,Y., Sinitskaya,A., Sinitsky,M., Kondratiev,E.,
Torgunakova,E., and Kutikhin,A. (2023) Calciprotein
particles induce cellular compartment-specific pro-
teome alterations in human arterial endothelial cells,
J. Cardiovasc. Dev. Dis., 11, 5, https://doi.org/10.3390/
jcdd11010005.
16. Herrmann, M., Schäfer, C., Heiss, A., Gräber, S.,
Kinkeldey, A., Büscher, A., Schmitt, M. M.,
Bornemann, J., Nimmerjahn, F., Herrmann, M.,
Helming, L., Gordon, S., and Jahnen-Dechent, W.
(2012) Clearance of fetuin-A-containing calcipro-
tein particles is mediated by scavenger receptor-A,
Circ. Res., 111, 575-584, https://doi.org/10.1161/
CIRCRESAHA.111.261479.
17. Köppert, S., Büscher, A., Babler, A., Ghallab, A., Buhl,
E. M., Latz, E., Hengstler, J. G., Smith, E. R., and
Jahnen-Dechent, W. (2018) Cellular clearance and bi-
ological activity of calciprotein particles depend on
their maturation state and crystallinity, Front. Immu-
nol., 9, 1991, https://doi.org/10.3389/fimmu.2018.01991.
18. Koeppert, S., Ghallab, A., Peglow, S., Winkler, C. F.,
Graeber, S., Büscher, A., Hengstler, J. G., and Jahnen-
Dechent, W. (2021) Live imaging of calciprotein par-
ticle clearance and receptor mediated uptake: role
of calciprotein monomers, Front. Cell Dev. Biol., 9,
633925, https://doi.org/10.3389/fcell.2021.633925.
19. Zeper, L. W., Bos, C., Leermakers, P. A., Franssen,
G. M., Raavé, R., Hoenderop, J. G. J., and de Baaij,
J. H. F. (2024) Liver and spleen predominantly me-
diate calciprotein particle clearance in a rat mod-
el of chronic kidney disease, Am. J. Physiol. Renal
Physiol., 326, F622-F634, https://doi.org/10.1152/
ajprenal.00239.2023.
20. Hutcheson, J.D., and Goettsch,C. (2023) Cardiovascu-
lar calcification heterogeneity in chronic kidney dis-
ease, Circ. Res., 132, 993-1012, https://doi.org/10.1161/
CIRCRESAHA.123.321760.
21. Sutton, N. R., Malhotra, R., St Hilaire, C., Aikawa, E.,
Blumenthal, R. S., Gackenbach, G., Goyal, P., John-
son,A., Nigwekar, S.U., Shanahan, C.M., Towler, D.A.,
Wolford, B.N., and Chen, Y. (2023) Molecular mecha-
nisms of vascular health: insights from vascular aging
and calcification, Arterioscler. Thromb. Vasc. Biol., 43,
15-29, https://doi.org/10.1161/ATVBAHA.122.317332.
22. Turner, M. E., Beck, L., Hill Gallant, K. M., Chen, Y.,
Moe, O. W., Kuro-O, M., Moe, S. M., and Aikawa, E.
(2024) Phosphate in cardiovascular disease: from new
insights into molecular mechanisms to clinical impli-
cations, Arterioscler. Thromb. Vasc. Biol., 44, 584-602,
https://doi.org/10.1161/ATVBAHA.123.319198.
23. Smith, E. R., Hanssen, E., McMahon, L. P., and Holt,
S.G. (2013) Fetuin-A-containing calciprotein particles
reduce mineral stress in the macrophage, PLoS One, 8,
e60904, https://doi.org/10.1371/journal.pone.0060904.
24. Peng, H.H., Wu, C.Y., Young,D., Martel,J., Young,A.,
Ojcius, D.M., Lee, Y. H., and Young, J.D. (2013) Phys-
icochemical and biological properties of biomimetic
mineralo-protein nanoparticles formed spontaneous-
ly in biological fluids, Small, 9, 2297-2307, https://doi.
org/10.1002/smll.201202270.
25. Anzai, F., Karasawa, T., Komada, T., Yamada, N.,
Miura,Y., Sampilvanjil,A., Baatarjav,C., Fujimura,K.,
Matsumura, T., Tago, K., Kurosu, H., Takeishi, Y.,
Kuro-O,M., and Takahashi,M. (2021) Calciprotein par-
ticles induce IL-1β/α-mediated inflammation through
NLRP3 inflammasome-dependent and -independent
mechanisms, Immunohorizons, 5, 602-614, https://
doi.org/10.4049/immunohorizons.2100066.
26. Murthy, S., Karkossa, I., Schmidt, C., Hoffmann, A.,
Hagemann,T., Rothe,K., Seifert,O., Anderegg,U., von
Bergen,M., Schubert,K., and Rossol,M. (2022) Danger
signal extracellular calcium initiates differentiation of
monocytes into SPP1/osteopontin-producing macro-
phages, Cell Death Dis., 13, 53, https://doi.org/10.1038/
s41419-022-04507-3.
27. Tiong, M.K., Smith, E.R., Toussaint, N.D., Al-Khayyat,
H. F., and Holt, S. G. (2021) Reduction of calciprotein
particles in adults receiving infliximab for chronic
inflammatory disease, JBMR Plus, 5, e10497, https://
doi.org/10.1002/jbm4.10497.
28. Kunishige, R., Mizoguchi, M., Tsubouchi, A.,
Hanaoka, K., Miura, Y., Kurosu, H., Urano, Y.,
Kuro-O, M., and Murata, M. (2020) Calciprotein par-
ticle-induced cytotoxicity via lysosomal dysfunction
and altered cholesterol distribution in renal epithelial
HK-2 cells, Sci. Rep., 10, 20125, https://doi.org/10.1038/
s41598-020-77308-3.
29. Kobylecki, C. J., Nordestgaard, B. G., and Afzal, S.
(2021) Plasma ionized calcium and risk of cardio-
CALCIPROTEIN PARTICLES 157
BIOCHEMISTRY (Moscow) Vol. 90 No. 1 2025
vascular disease: 106,774 individuals from the Co-
penhagen General Population Study, Clin. Chem., 67,
265-275, https://doi.org/10.1093/clinchem/hvaa245.
30. Kobylecki, C. J., Nordestgaard, B. G., and Afzal, S.
(2022) Low plasma ionized calcium is associated
with increased mortality: a population-based study
of 106,768 individuals, J.Clin. Endocrinol. Metab., 107,
e3039-e3047, https://doi.org/10.1210/clinem/dgac146.
31. Gimbrone, M. A., Jr., and García-Cardeña, G. (2016)
Endothelial cell dysfunction and the pathobiology
of atherosclerosis, Circ. Res., 118, 620-636, https://
doi.org/10.1161/CIRCRESAHA.115.306301.
32. Mundi, S., Massaro, M., Scoditti, E., Carluccio, M. A.,
van Hinsbergh, V. W. M., Iruela-Arispe, M. L., and
De Caterina, R. (2018) Endothelial permeability, LDL
deposition, and cardiovascular risk factors – a review,
Cardiovasc. Res., 114, 35-52, https://doi.org/10.1093/
cvr/cvx226.
33. Libby, P., Buring, J. E., Badimon, L., Hansson, G. K.,
Deanfield, J., Bittencourt, M. S., Tokgözoğlu, L., and
Lewis, E.F. (2019) Atherosclerosis, Nat. Rev. Dis. Prim-
ers, 5, 56, https://doi.org/10.1038/s41572-019-0106-z.
34. Tamargo, I.A., Baek, K.I., Kim,Y., Park,C., and Jo,H.
(2023) Flow-induced reprogramming of endothelial
cells in atherosclerosis, Nat. Rev. Cardiol., 20, 738-753,
https://doi.org/10.1038/s41569-023-00883-1.
35. Wang, X., Shen, Y., Shang, M., Liu, X., and Munn,
L. L. (2023) Endothelial mechanobiology in athero-
sclerosis, Cardiovasc. Res., 119, 1656-1675, https://doi.
org/10.1093/cvr/cvad076.
36. Giorgi, C., Marchi, S., and Pinton, P. (2018) The ma-
chineries, regulation and cellular functions of mito-
chondrial calcium, Nat. Rev. Mol. Cell Biol., 19, 713-
730, https://doi.org/10.1038/s41580-018-0052-8.
37. Walkon, L. L., Strubbe-Rivera, J. O., and Bazil, J. N.
(2022) Calcium overload and mitochondrial metab-
olism, Biomolecules, 12, 1891, https://doi.org/10.3390/
biom12121891.
38. De Nicolo, B., Cataldi-Stagetti, E., Diquigiovanni, C.,
and Bonora, E. (2023) Calcium and reactive oxygen
species signaling interplays in cardiac physiology and
pathologies, Antioxidants (Basel), 12, 353, https://doi.
org/10.3390/antiox12020353.
39. Werner, L. E., and Wagner, U. (2023) Calcium-sens-
ing receptor-mediated NLRP3 inflammasome activa-
tion in rheumatoid arthritis and autoinflammation,
Front. Physiol., 13, 1078569, https://doi.org/10.3389/
fphys.2022.1078569.
40. Mukai,H., Miura,Y., Kotani,K., Kotoda,A., Kurosu,H.,
Yamada, T., Kuro-O, M., and Iwazu, Y. (2022) The ef-
fects for inflammatory responses by CPP with dif-
ferent colloidal properties in hemodialysis patients,
Sci. Rep., 12, 21856, https://doi.org/10.1038/s41598-
022-26166-2.
41. Bojic,M., Cejka,D., Bielesz,B., and Schernthaner, G.H.
(2023) Secondary calciprotein particle size is associat-
ed with patient mortality in peripheral artery disease,
Atherosclerosis, 370, 12-17, https://doi.org/10.1016/j.
atherosclerosis.2023.02.006.
42. Chen, W., Anokhina, V., Dieudonne, G., Abramowitz,
M.K., Kashyap,R., Yan,C., Wu, T.T., de Mesy Bentley,
K. L., Miller, B. L., and Bushinsky, D. A. (2019) Pa-
tients with advanced chronic kidney disease and
vascular calcification have a large hydrodynamic
radius of secondary calciprotein particles, Nephrol.
Dial. Transplant., 34, 992-1000, https://doi.org/10.1093/
ndt/gfy117.
43. Chen,W., Fitzpatrick, J., Monroy-Trujillo, J.M., Sozio,
S. M., Jaar, B. G., Estrella, M. M., Serrano, J., Anokhi-
na, V., Miller, B. L., Melamed, M. L., Bushinsky, D. A.,
and Parekh, R. S. (2021) Associations of serum cal-
ciprotein particle size and transformation time with
arterial calcification, arterial stiffness, and mortality
in incident hemodialysis patients, Am. J. Kidney Dis.,
77, 346-354, https://doi.org/10.1053/j.ajkd.2020.05.031.
44. Feenstra,L., Reijrink,M., Pasch,A., Smith, E.R., Visser,
L. M., Bulthuis, M., Lodewijk, M. E., Mastik, M. F.,
Greuter, M. J. W., Slart, R. H. J. A., Mulder, D. J., Pol,
R. A., Te Velde-Keyzer, C. A., TransplantLines Investi-
gators, Krenning,G., and Hillebrands, J.L. (2024) Cal-
ciprotein particle counts associate with vascular re-
modelling in chronic kidney disease, Cardiovasc. Res.,
120, 1953-1966, https://doi.org/10.1093/cvr/cvae164.
45. Zeper, L.W., Smith, E.R., Ter Braake, A.D., Tinnemans,
P. T., de Baaij, J. H. F., and Hoenderop, J. G. J. (2023)
Calciprotein particle synthesis strategy determines
in vitro calcification potential, Calcif. Tissue Int., 112,
103-117, https://doi.org/10.1007/s00223-022-01036-1.
46. Mencke, R., Al Ali, L., de Koning, M. L. Y., Pasch, A.,
Minnion, M., Feelisch, M., van Veldhuisen, D. J., van
der Horst, I. C. C., Gansevoort, R. T., Bakker, S. J. L.,
de Borst, M. H., van Goor, H., van der Harst, P.,
Lipsic, E., and Hillebrands, J. L. (2024) Serum calci-
fication propensity is increased in myocardial in-
farction and hints at a pathophysiological role inde-
pendent of classical cardiovascular risk factors, Ar-
terioscler. Thromb. Vasc. Biol., 44, 1884-1894, https://
doi.org/10.1161/ATVBAHA.124.320974.
47. Mori, K., Shoji, T., Nakatani, S., Uedono, H., Ochi, A.,
Yoshida, H., Imanishi, Y., Morioka, T., Tsujimoto, Y.,
Kuro-O, M., and Emoto, M. (2024) Differential asso-
ciations of fetuin-A and calcification propensity with
cardiovascular events and subsequent mortality in
patients undergoing hemodialysis, Clin. Kidney J., 17,
sfae042, https://doi.org/10.1093/ckj/sfae042.
48. Van der Vaart, A., Eelderink, C., van Goor, H.,
Hillebrands, J. L., Te Velde-Keyzer, C. A., Bakker,
S.J.L., Pasch,A., van Dijk, P.R., Laverman, G.D., and
de Borst, M.H. (2024) Serum T50 predicts cardiovas-
cular mortality in individuals with type 2 diabetes: a
prospective cohort study, J.Intern. Med., 295, 748-758,
https://doi.org/10.1111/joim.13781.
SHISHKOVA et al.158
BIOCHEMISTRY (Moscow) Vol. 90 No. 1 2025
49. Miura,M., Miura,Y., Iwazu,Y., Mukai,H., Sugiura,T.,
Suzuki,Y., Kato,M., Kano,M., Nagata,D., Shiizaki,K.,
Kurosu,H., and Kuro-O,M. (2023) Removal of calcip-
rotein particles from the blood using an adsorption
column improves prognosis of hemodialysis minia-
ture pigs, Sci. Rep., 13, 15026, https://doi.org/10.1038/
s41598-023-42273-0.
50. Kawakami, K., Ohya, M., Yashiro, M., Sonou, T.,
Yamamoto, S., Nakashima, Y., Yano, T., Tanaka, Y.,
Ishida,K., Kobashi,S., Shigematsu,T., and Araki, S.I.
(2023) Bisphosphonate FYB-931 prevents high phos-
phate-induced vascular calcification in rat aortic
rings by altering the dynamics of the transformation
of calciprotein particles, Calcif. Tissue Int., 113, 216-
228, https://doi.org/10.1007/s00223-023-01086-z.
51. Rossi, A., Pizzo, P., and Filadi, R. (2019) Calcium,
mitochondria and cell metabolism: a functional tri-
angle in bioenergetics, Biochim. Biophys. Acta Mol.
Cell Res., 1866, 1068-1078, https://doi.org/10.1016/
j.bbamcr.2018.10.016.
52. Park, C. J., and Shin, R. (2022) Calcium channels and
transporters: roles in response to biotic and abiotic
stresses, Front. Plant Sci., 13, 964059, https://doi.org/
10.3389/fpls.2022.964059.
53. Harlacher, E., Wollenhaupt, J., Baaten, C. C. F. M. J.,
and Noels, H. (2022) Impact of uremic toxins on en-
dothelial dysfunction in chronic kidney disease: a sys-
tematic review, Int. J. Mol. Sci., 23, 531, https://doi.org/
10.3390/ijms23010531.
54. Roumeliotis,S., Mallamaci,F., and Zoccali,C. (2020) En-
dothelial dysfunction in chronic kidney disease, from
biology to clinical outcomes: a 2020 update, J. Clin.
Med., 9, 2359, https://doi.org/10.3390/jcm9082359.
55. Kutikhin, A. G., Shishkova, D. K., Velikanova, E. A.,
Sinitsky, M. Y., Sinitskaya, A. V., and Markova, V. E.
(2022) Endothelial dysfunction in the context of blood-
brain barrier modeling, J.Evol. Biochem. Physiol., 58,
781-806, https://doi.org/10.1134/S0022093022030139.
56. Ferrucci, L., and Fabbri, E. (2018) Inflammageing:
chronic inflammation in ageing, cardiovascular
disease, and frailty, Nat. Rev. Cardiol., 15, 505-522,
https://doi.org/10.1038/s41569-018-0064-2.
57. Liberale,L., Montecucco,F., Tardif, J.C., Libby,P., and
Camici, G. G. (2020) Inflamm-ageing: the role of in-
flammation in age-dependent cardiovascular disease,
Eur. Heart J., 41, 2974-2982, https://doi.org/10.1093/
eurheartj/ehz961.
58. Walker, K. A., Basisty, N., Wilson, D. M., 3rd, and
Ferrucci, L. (2022) Connecting aging biology and
inflammation in the omics era, J. Clin. Invest., 132,
e158448, https://doi.org/10.1172/JCI158448.
59. Li,X., Li,C., Zhang,W., Wang,Y., Qian,P., and Huang,H.
(2023) Inflammation and aging: signaling path-
ways and intervention therapies, Signal Transduct.
Target Ther., 8, 239, https://doi.org/10.1038/s41392-
023-01502-8.
60. Kuro-O,M. (2021) Phosphate as a pathogen of arterio-
sclerosis and aging, J. Atheroscler. Thromb., 28, 203-
213, https://doi.org/10.5551/jat.RV17045.
61. Tiong, M. K., Holt, S. G., Ford, M.L., and Smith, E. R.
(2022) Serum calciprotein monomers and chronic kid-
ney disease progression, Am.J. Nephrol., 53, 806-815,
https://doi.org/10.1159/000526609.
62. Kuro-O,M. (2021) Klotho and calciprotein particles as
therapeutic targets against accelerated ageing, Clin.
Sci. (Lond)., 135, 1915-1927, https://doi.org/10.1042/
CS20201453.
63. Gelli,R., Ridi,F., and Baglioni,P. (2019) The importance
of being amorphous: calcium and magnesium phos-
phates in the human body, Adv. Colloid Interface Sci.,
269, 219-235, https://doi.org/10.1016/j.cis.2019.04.011.
64. Pasch,A., Jahnen-Dechent,W., and Smith, E.R. (2018)
Phosphate, calcification in blood, and mineral stress:
the physiologic blood mineral buffering system and its
association with cardiovascular risk, Int. J. Nephrol.,
2018, 9182078, https://doi.org/10.1155/2018/9182078.
65. Di Marco, G. S., Hausberg, M., Hillebrand, U., Ruste-
meyer,P., Wittkowski,W., Lang,D., and Pavenstädt,H.
(2008) Increased inorganic phosphate induces human
endothelial cell apoptosis in vitro, Am. J. Physiol. Re-
nal Physiol., 294, F1381-F1387, https://doi.org/10.1152/
ajprenal.00003.2008.
66. Shuto, E., Taketani, Y., Tanaka, R., Harada, N.,
Isshiki,M., Sato,M., Nashiki,K., Amo,K., Yamamoto,H.,
Higashi, Y., Nakaya, Y., and Takeda, E. (2009) Dietary
phosphorus acutely impairs endothelial function,
J. Am. Soc. Nephrol., 20, 1504-1512, https://doi.org/
10.1681/ASN.2008101106.
67. Peng, A., Wu, T., Zeng, C., Rakheja, D., Zhu, J., Ye, T.,
Hutcheson, J., Vaziri, N. D., Liu, Z., Mohan, C., and
Zhou, X. J. (2011) Adverse effects of simulated hy-
per- and hypo-phosphatemia on endothelial cell
function and viability, PLoS One, 6, e23268, https://
doi.org/10.1371/journal.pone.0023268.
68. Yamada,S., Tokumoto,M., Tatsumoto,N., Taniguchi,M.,
Noguchi, H., Nakano, T., Masutani, K., Ooboshi, H.,
Tsuruya, K., and Kitazono, T. (2014) Phosphate over-
load directly induces systemic inflammation and
malnutrition as well as vascular calcification in ure-
mia, Am. J. Physiol. Renal Physiol., 306, F1418-F1428,
https://doi.org/10.1152/ajprenal.00633.2013.
69. Voelkl, J., Egli-Spichtig, D., Alesutan, I., and Wagner,
C. A. (2021) Inflammation: a putative link between
phosphate metabolism and cardiovascular disease,
Clin. Sci. (Lond)., 135, 201-227, https://doi.org/10.1042/
CS20190895.
70. Ding, M., Zhang, Q., Zhang, M., Jiang, X., Wang, M.,
Ni, L., Gong, W., Huang, B., and Chen, J. (2022) Phos-
phate overload stimulates inflammatory reaction
via PiT-1 and induces vascular calcification in ure-
mia, J. Ren. Nutr., 32, 178-188, https://doi.org/10.1053/
j.jrn.2021.03.008.
CALCIPROTEIN PARTICLES 159
BIOCHEMISTRY (Moscow) Vol. 90 No. 1 2025
71. Hetz,R., Beeler,E., Janoczkin,A., Kiers, S., Li, L., Wil-
lard, B.B., Razzaque, M.S., and He,P. (2022) Excessive
inorganic phosphate burden perturbed intracellular
signaling: quantitative proteomics and phosphopro-
teomics analyses, Front. Nutr., 8, 765391, https://
doi.org/10.3389/fnut.2021.765391.
72. Heiss, A., Eckert, T., Aretz, A., Richtering, W., van
Dorp, W., Schäfer, C., and Jahnen-Dechent, W. (2008)
Hierarchical role of fetuin-A and acidic serum pro-
teins in the formation and stabilization of calcium
phosphate particles, J. Biol. Chem., 283, 14815-14825,
https://doi.org/10.1074/jbc.M709938200.
73. Dridi, H., Kushnir, A., Zalk, R., Yuan, Q., Melville, Z.,
and Marks, A. R. (2020) Intracellular calcium leak in
heart failure and atrial fibrillation: a unifying mech-
anism and therapeutic target, Nat. Rev. Cardiol., 17,
732-747, https://doi.org/10.1038/s41569-020-0394-8.
74. Arnold,A., Dennison,E., Kovacs, C.S., Mannstadt,M.,
Rizzoli, R., Brandi, M. L., Clarke, B., and Thakker,
R. V. (2021) Hormonal regulation of biomineraliza-
tion, Nat. Rev. Endocrinol., 17, 261-275, https://doi.org/
10.1038/s41574-021-00477-2.
75. Collins, M.T., Marcucci,G., Anders, H.J., Beltrami,G.,
Cauley, J. A., Ebeling, P. R., Kumar, R., Linglart, A.,
Sangiorgi, L., Towler, D. A., Weston, R., Whyte, M. P.,
Brandi, M. L., Clarke, B., and Thakker, R. V. (2022)
Skeletal and extraskeletal disorders of biomineral-
ization, Nat. Rev. Endocrinol., 18, 473-489, https://
doi.org/10.1038/s41574-022-00682-7.
76. Shishkova, D. K., Sinitskaya, A. V., Sinitsky, M. Y.,
Matveeva, V. G., Velikanova, E. A., Markova, V. E.,
and Kutikhin, A. G. (2022) Spontaneous endotheli-
al-to-mesenchymal transition in human primary um-
bilical vein endothelial cells, Compl. Iss. Cardiovasc.
Dis., 11, 97-114, https://doi.org/10.17802/2306-1278-
2022-11-3-97-114.
77. Bogdanov, L. A., Velikanova, E.A., Kanonykina, A. Y.,
Frolov, A. V., Shishkova, D. K., Lazebnaya, A. I., and
Kutikhin, A. G. (2022) Vascular smooth muscle cell
contractile proteins as universal markers of ves-
sels of microcirculatory bed, Compl. Iss. Cardiovasc.
Dis., 11, 162-176, https://doi.org/10.17802/2306-1278-
2022-11-3-162-176.
78. Wang, B., Han, J., Elisseeff, J. H., and Demaria, M.
(2024) The senescence-associated secretory pheno-
type and its physiological and pathological implica-
tions, Nat. Rev. Mol. Cell Biol., 25, 958-978, https://
doi.org/10.1038/s41580-024-00727-x.
79. Mehdizadeh, M., Aguilar, M., Thorin, E., Ferbeyre, G.,
and Nattel,S. (2022) The role of cellular senescence in
cardiac disease: basic biology and clinical relevance,
Nat. Rev. Cardiol., 19, 250-264, https://doi.org/10.1038/
s41569-021-00624-2.
80. Di Micco, R., Krizhanovsky, V., Baker, D., and d’Adda
di Fagagna, F. (2021) Cellular senescence in ageing:
from mechanisms to therapeutic opportunities, Nat.
Rev. Mol. Cell Biol., 22, 75-95, https://doi.org/10.1038/
s41580-020-00314-w.
81. Ewence, A.E., Bootman, M., Roderick, H. L., Skepper,
J.N., McCarthy,G., Epple,M., Neumann,M., Shanahan,
C. M., and Proudfoot, D. (2008) Calcium phosphate
crystals induce cell death in human vascular smooth
muscle cells: a potential mechanism in atherosclerotic
plaque destabilization, Circ. Res., 103, e28-e34, https://
doi.org/10.1161/CIRCRESAHA.108.181305.
82. Jäger,E., Murthy,S., Schmidt,C., Hahn,M., Strobel,S.,
Peters,A., Stäubert,C., Sungur,P., Venus,T., Geisler,M.,
Radusheva, V., Raps, S., Rothe, K., Scholz, R., Jung, S.,
Wagner, S., Pierer, M., Seifert, O., Chang, W., Estrela-
Lopis, I., Raulien, N., Krohn, K., Sträter, N., Hoep-
pener, S., Schöneberg, T., Rossol, M., and Wagner, U.
(2020) Calcium-sensing receptor-mediated NLRP3 in-
flammasome response to calciprotein particles drives
inflammation in rheumatoid arthritis, Nat. Commun.,
11, 4243, https://doi.org/10.1038/s41467-020-17749-6.
83. Tournis, S., Makris, K., Cavalier, E., and Trovas, G.
(2020) Cardiovascular risk in patients with primary
hyperparathyroidism, Curr. Pharm. Des., 26, 5628-
5636, https://doi.org/10.2174/1381612824999201105
165642.
84. Bkaily, G., and Jacques, D. (2023) Calcium homeosta-
sis, transporters, and blockers in health and diseases
of the cardiovascular system, Int. J. Mol. Sci., 24, 8803,
https://doi.org/10.3390/ijms24108803.
85. Minisola, S., Arnold, A., Belaya, Z., Brandi, M. L.,
Clarke, B.L., Hannan, F. M., Hofbauer, L. C., Insogna,
K. L., Lacroix, A., Liberman, U., Palermo, A., Pepe, J.,
Rizzoli,R., Wermers,R., and Thakker, R.V. (2022) Ep-
idemiology, pathophysiology, and genetics of primary
hyperparathyroidism, J. Bone Miner. Res., 37, 2315-
2329, https://doi.org/10.1002/jbmr.4665.
86. Tonon, C.R., Silva, T.A.A.L., Pereira, F.W.L., Queiroz,
D. A. R., Junior, E. L. F., Martins, D., Azevedo, P. S.,
Okoshi, M. P., Zornoff, L. A. M., de Paiva, S. A. R.,
Minicucci, M.F., and Polegato, B.F. (2022) A review of
current clinical concepts in the pathophysiology, eti-
ology, diagnosis, and management of hypercalcemia,
Med. Sci. Monit., 28, e935821, https://doi.org/10.12659/
MSM.935821.
87. Lang, I. M., and Schleef, R. R. (1996) Calcium-de-
pendent stabilization of type I plasminogen activa-
tor inhibitor within platelet alpha-granules, J. Biol.
Chem., 271, 2754-2761, https://doi.org/10.1074/jbc.
271.5.2754.
88. Liu,Q., Möller,U., Flügel,D., and Kietzmann,T. (2004)
Induction of plasminogen activator inhibitor I gene
expression by intracellular calcium via hypoxia-
inducible factor-1, Blood, 104, 3993-4001, https://doi.
org/10.1182/blood-2004-03-1017.
89. Peiretti, F., Fossat, C., Anfosso, F., Alessi, M. C.,
Henry,M., Juhan-Vague,I., and Nalbone,G. (1996) In-
crease in cytosolic calcium upregulates the synthesis
SHISHKOVA et al.160
BIOCHEMISTRY (Moscow) Vol. 90 No. 1 2025
of type 1 plasminogen activator inhibitor in the hu-
man histiocytic cell line U937, Blood, 87, 162-173,
https://doi.org/10.1182/blood.V87.1.162.162.
90. Heiss, A., DuChesne, A., Denecke, B., Grötzinger, J.,
Yamamoto, K., Renné, T., and Jahnen-Dechent, W.
(2003) Structural basis of calcification inhibition by
alpha 2-HS glycoprotein/fetuin-A: formation of colloi-
dal calciprotein particles, J. Biol. Chem., 278, 13333-
13341, https://doi.org/10.1074/jbc.M210868200.
91. Schäfer, C., Heiss, A., Schwarz, A., Westenfeld, R.,
Ketteler, M., Floege, J., Müller-Esterl, W., Schinke, T.,
and Jahnen-Dechent, W. (2003) The serum protein
alpha 2-Heremans-Schmid glycoprotein/fetuin-A is a
systemically acting inhibitor of ectopic calcification,
J. Clin. Invest., 112, 357-366, https://doi.org/10.1172/
JCI17202.
92. Heiss, A., Jahnen-Dechent, W., Endo, H., and
Schwahn,D. (2007) Structural dynamics of a colloidal
protein-mineral complex bestowing on calcium phos-
phate a high solubility in biological fluids, Biointer-
phases, 2, 16-20, https://doi.org/10.1116/1.2714924.
93. Rochette, C.N., Rosenfeldt,S., Heiss,A., Narayanan,T.,
Ballauff,M., and Jahnen-Dechent, W. (2009) A shield-
ing topology stabilizes the early stage protein-miner-
al complexes of fetuin-A and calcium phosphate: a
time-resolved small-angle X-ray study, Chembiochem,
10, 735-740, https://doi.org/10.1002/cbic.200800719.
Publishers Note. Pleiades Publishing remains
neutral with regard to jurisdictional claims in published
maps and institutional affiliations. AI tools may have
been used in the translation or editing of this article.