ISSN 0006-2979, Biochemistry (Moscow), 2024, Vol. 89, No. 5, pp. 883-903 © Pleiades Publishing, Ltd., 2024.
883
Molecular Changes in Immunological Characteristics
of Bone Marrow Multipotent Mesenchymal Stromal Cells
in Lymphoid Neoplasia
Nataliya A. Petinati
1,a
*
#
, Aleksandra V. Sadovskaya
1,2#
, Natalia V. Sats
1
,
Nikolai M. Kapranov
1
, Yulia O. Davydova
1
, Ekaterina A. Fastova
1
,
Aminat U. Magomedova
1
, Anastasia N. Vasilyeva
1
, Olga A. Aleshina
1
,
Georgiy P. Arapidi
3,4,5
, Viktoria O. Shender
3,4
, Igor P. Smirnov
3
, Olga V. Pobeguts
3
,
Maria A. Lagarkova
3
, Nina I. Drize
1
, and Elena N. Parovichnikova
1
1
National Medical Research Center for Hematology, Ministry of Health of the Russian Federation,
125167 Moscow, Russia
2
Lomonosov Moscow State University, 119991 Moscow, Russia
3
Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine,
Federal Medical Biological Agency, 119435 Moscow, Russia
4
Shemyakin–Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences,
117997 Moscow, Russia
5
Moscow Institute of Physics and Technology, 141700 Dolgoprudny, Russia
a
e-mail: loel@mail.ru
Received September 15, 2023
Revised November 22, 2023
Accepted November 23, 2023
AbstractImmune system and bone marrow stromal cells play an important role in maintaining normal he-
matopoiesis. Lymphoid neoplasia disturbs not only development of immune cells, but other immune response
mechanisms as well. Multipotent mesenchymal stromal cells (MSCs) of the bone marrow are involved in immune
response regulation through both intercellular interactions and secretion of various cytokines. In hematological
malignancies, the bone marrow stromal microenvironment, including MSCs, is altered. Aim of this study was to
describe the differences of MSCs’ immunological function in the patients with acute lymphoblastic leukemia (ALL)
and diffuse large B-cell lymphoma (DLBCL). In ALL, malignant cells arise from the early precursor cells localized
in bone marrow, while in DLBCL they arise from more differentiated B-cells. In this study, only the DLBCL patients
without bone marrow involvement were included. Growth parameters, surface marker expression, genes of in-
terest expression, and secretion pattern of bone marrow MSCs from the patients with ALL and DLBCL at the onset
of the disease and in remission were studied. MSCs from the healthy donors of corresponding ages were used as
controls. It has been shown that concentration of MSCs in the bone marrow of the patients with ALL is reduced at
the onset of the disease and is restored upon reaching remission; in the patients with DLBCL this parameter does
not change. Proliferative capacity of MSCs did not change in the patients with ALL; however, the cells of the DLBCL
patients both at the onset and in remission proliferated significantly faster than those from the donors. Expression
of the membrane surface markers and expression of the genes important for differentiation, immunological status
maintenance, and cytokine secretion differed significantly in the MSCs of the patients from those of the healthy
donors and depended on nosology of the disease. Secretomes of the MSCs varied greatly; a number of proteins
associated with immune response regulation, differentiation, and maintenance of hematopoietic stem cells were
Abbreviations: ALL, acute lymphoblastic leukemia; Ct,number of cycles required to reach the probe fluorescence threshold
in PCR; DLBCL,diffuse large B-cell lymphoma; HSCs,hematopoietic stem cells; MFI,mean fluorescence intensity; MSCs,mul-
tipotent mesenchymal stromal cells; REL,relative expression level.
* To whom correspondence should be addressed.
# These authors contributed equally to this work.
PETINATI et al.884
BIOCHEMISTRY (Moscow) Vol. 89 No. 5 2024
depleted in the secretomes of the cells from the patients. Lymphoid neoplasia leads to dramatic changes in the
functional immunological status of MSCs.
DOI: 10.1134/S0006297924050092
Keywords: multipotent mesenchymal stromal cells, acute lymphoblastic leukemia, diffuse large B-cell lymphoma,
gene expression, protein secretion
INTRODUCTION
Bone marrow is responsible for production and
maintenance of blood cell populations, including im-
mune cells, throughout the human life. In addition, it
acts as an important lymphoid organ, housing many
types of mature lymphocytes including B cells, T cells,
natural killer T cells, and innate immune cells [1].
Inbone marrow, lymphoid cells interact with the stro-
mal microenvironment and are involved in regulation
of hematopoiesis and immune response. Hematopoi-
etic stem cells (HSCs) reside in the specialized niches
that maintain them for lifelong blood cell production.
Niches also provide homing and survival of HSCs, reg-
ulate their dormant state, self-renewal, differentiation,
and proliferation. Many cell types are involved in for-
mation and functioning of these niches [2]. Main niche
components include mesenchymal stem cells, which
differentiate into numerous other stromal cells that
make up the niche, keep HSCs in it, and take part in
immune responses in the bone marrow [3].
B-cell lymphoproliferative disorders, which in-
clude multiple myeloma, Hodgkin’s lymphoma, and
chronic lymphocytic leukemia (CLL), as well as pre-
cursor conditions such as monoclonal B-cell lympho-
cytosis, are pathologies characterized by uncontrolled
growth of B-lymphocytes [4]. Lymphoproliferative dis-
orders can occur either in the lymphatic tissues (as
in lymphoma) or in the bone marrow (as in CLL and
multiple myeloma). Course of the disease and treat-
ment vary widely depending on the type of neoplasia
and other individual factors; however, even extramed-
ullary tumors affect the bone marrow stromal micro-
environment [5]. In addition, the bone marrow stroma
can be strongly affected by chronic viral infections.
For example, the CXCL12-abundant reticular cells (CAR
cells) die in the mice with chronic lymphocytic chori-
omeningitis due to production of IFN-α and IFN-γ by
the virus-specific CD8
+
T cells [6]. Chronic viral infec-
tions are associated with hematopoiesis suppression,
bone marrow failure, and depletion of the HSC pool
[7,8]. Combination of functional analysis with 3D mi-
croscopy demonstrated that chronic infection with
lymphocytic choriomeningitis virus leads to the death
of most mesenchymal CAR cells and pro-inflammatory
transcriptional remodeling of the remaining ones. This
causes long-term functional defects and reduced com-
petitive repopulation ability of HSCs. Bone marrow
immunopathology is caused by the virus-specific acti-
vated CD8
+
T cells that accumulate in the bone mar-
row through the interferon-dependent mechanisms.
Combined inhibition of the IFN type I and type  II
pathways by antibodies completely prevents CAR cell
degeneration and protects HSCs from chronic dysfunc-
tion. Thus, viral infections and subsequent immune
response have a lasting effect on the bone marrow ho-
meostasis, permanently reducing repopulation ability
of HSCs and disrupting secretion of the key stromal cy-
tokines that support hematopoiesis [9].
Acute lymphoblastic leukemia (ALL) is a neopla-
sia arising from early progenitors of B-cells(B-ALL) or
T-cells (T-ALL). The disease is characterized by uncon-
trolled proliferation of lymphoid progenitors in the
bone marrow and consequent appearance of large
numbers of immature lymphocytes, disrupting normal
hematopoiesis. B-ALL is considered a genetic disease,
but increasing evidence points to the ability of the
bone marrow microenvironment to significantly con-
tribute to maintenance, progression, response to treat-
ment, and possibly development of the disease, regard-
less of the presence of specific genetic abnormalities
in hematopoietic cells [10]. There is a large body of
evidence suggesting that B-ALL cells can modify the
bone marrow microenvironment creating conditions
conducive to the survival of malignant cells during
chemotherapy, leading to the disease recurrence. Leu-
kemic cells interact with components of the bone mar-
row microenvironment, including multipotent mesen-
chymal stromal cells(MSCs) [11]. Studying interactions
between the bone marrow microenvironment and
ALL cells has led to the discovery of potential thera-
peutic targets that include cytokines/chemokines and
their receptors, adhesion molecules, signal transduc-
tion pathways, and hypoxia-associated proteins [12].
Complex interactions between the leukemic cells and
components of the bone marrow microenvironment
lead to the involvement of MSCs in the suppression of
antitumor response, since these cells secrete cytokines
such as transforming growth factor-β (TGF-β) and he-
patocyte growth factor (HGF), which mediate suppres-
sion of T-cells [13].
Diffuse large B-cell lymphoma (DLBCL) is a hetero-
geneous group of diseases that differ in histological, im-
munohistochemical, and molecular characteristics[14].
LYMPHOID NEOPLASIA 885
BIOCHEMISTRY (Moscow) Vol. 89 No. 5 2024
In DLBCL, histological or molecular methods reveal
damage to the bone marrow only in 10-25% of pa-
tients [15]. It is believed that in the remaining patients
bone marrow is not involved in the malignant process.
However, bone marrow stromal progenitor cells in the
DLBCL patients without bone marrow involvement
are altered [16]. These changes cannot be attributed to
the contact interaction with tumor cells, as occurs in
leukemia [17, 18]. Many tumors secrete cytokines and
chemokines [19, 20], moreover, presence of a tumor
can be considered an inflammatory process [21, 22].
Inflammation is associated with active release of mul-
tiple factors that can activate cells of the bone marrow
stromal microenvironment, in particular MSCs [23].
MSCs activated by IL-1β, TNF, and IFN-γ secrete inhib-
itors and activators of the inflammatory process [24].
The aim of the study was to analyze the effect of
lymphoid neoplasia from early progenitor cells in di-
rect contact with the bone marrow stroma (ALL) and
more mature cells located exclusively extramedullary
(DLBCL) on immunological function of MSCs.
MATERIALS AND METHODS
Patient and donor bone marrow samples. Pa-
tient and donor samples were obtained in accordance
with the Declaration of Helsinki after the written in-
formed consent. The study was approved by the ethics
committee of the Federal State Budgetary Institution
National Medical Research Center for Hematology of
the Ministry of Health of the Russian Federation, pro-
tocol No.171 dated April27, 2023.
The work was performed with MSCs isolated from
the bone marrow of patients with ALL and DLBCL;
MSCs from the bone marrow of healthy donors were
used as controls. To account for the age difference,
two different donor groups were age-matched with
the patients with ALL and DLBCL. As such, the studied
parameters were normalized to the value of the corre-
sponding donor group median. Data on the number of
samples, patients, and healthy donors are presented
inTable1.
MSCs cultivation. Bone marrow was obtained
from the patients during diagnostic punctures and
from the hematopoietic stem cells donors during ex-
fusions after informed consent. To prevent clotting,
2-7 ml of bone marrow were placed in sterile tubes
with 1 ml of heparin (50  units/ml). The bone marrow
samples were diluted 2-fold with an α-MEM (ICN, Cana-
da) containing 0.2% methylcellulose (1500 cP, Sigma-Al-
drich, USA) and left for 40 min at room temperature.
Supernatant was collected and precipitated by centrif-
ugation at 450g for 10 min. Number of nuclear cells
was determined by counting after staining with gen-
tian violet solution (1% solution in 3% acetic acid) in a
Goryaev chamber. Cells (3×10
6
) were placed in a flask
with a bottom area of 25 cm
2
(Corning-Costar, USA)
Table 1. Characteristics of patients and donors
Diagnosis Group
Total number
of patients
Sex
Number
of patients
Age, years
Median
age, years
ALL
onset 31
male 15 19-72 29
female 16 18-55 31
remission 14
male 5 19-55 24
female 9 18-55 30
DLBCL
onset 40
male 14 30-78 48
female 26 34-79 60
remission 40
male 14 30-78 48
female 26 34-79 60
Healthy
donors
ALL 56
male 30
18-48 27
female 26
DLBCL 30
male 10
30-78 54
female 20
PETINATI et al.886
BIOCHEMISTRY (Moscow) Vol. 89 No. 5 2024
in 5 ml of complete α-MEM nutrient medium (ICN,
Canada) supplemented with 10% fetal bovine serum
(FBS) (Hyclone, USA), 2 mM L-glutamine (ICN, Cana-
da), 100 U/ml penicillin (Sintez, Russia), and 5  µg/ml
streptomycin (BioPharmGarant, Russia). MSCs were
cultivated at 37°C and 5% CO
2
. Culture medium was
changed twice a week. After reaching confluency,
the cells were passaged. To do this, the cells were
washed twice with 5ml of Versen’s solution and once
with 0.25 ml of 0.25% trypsin solution (PanEco, Rus-
sia). 0.25 ml of trypsin solution was added, and flasks
were left at room temperature until the cells detached
from the surface. The cells were resuspended in 1 ml
of the medium with FBS, and counted in 0.2% trypan
blue solution (Sigma-Aldrich) to determine their num-
ber and viability (trypan blue only stains dead cells).
During passage, 10
5
cells were seeded in a flask with a
bottom area of 25 cm
2
in 5 ml of the medium. Cultures
were maintained for 4 passages.
The time to P0 was defined as the number of days
from seeding bone marrow to reaching confluence for
the first time.
Calculation of cumulative cell production. Cu-
mulative cell production over 3 passages was calculat-
ed using the formula (1):
N
sum
= N0 + N0 · 
N1
200000
 +N1 · 
N2
200000
 +N2 · 
N3
200000
, (1)
where N0, N1, N2, and N3 are number of the cells re-
moved from 2 culture flasks at passages 0, 1, 2, and 3,
respectively.
Surface marker expression analysis by flow cy-
tometry. Surface phenotype of MSCs was studied at the
2nd passage by flow cytometry. After removing MSCs
from the flask, they were washed twice with CellWash
solution (BD Biosciences, USA) and then 2×10
4
cells
were incubated for 20 min in the dark with antibod-
ies. The antibody panels were as follows: 1) PE-labeled
anti-CD90 (5E10, BD Pharmingen, USA), FITC-labeled
anti-HLA-ABC (FN50, BioLegend, USA) and APC-labeled
anti-HLA-DR (L243, BioLegend); 2) anti-CD105, labeled
with FITC (43A3, BioLegend), anti-CD54, labeled with
APC (HA58, BioLegend), anti-CD146 PE-labeled (P1H12,
BD Pharmingen, USA); 3) PE-labeled anti-CD73 (AD2,
BD Pharmingen, USA). The analysis was performed
using a CytoFLEX flow cytometer (Beckman Coulter,
USA), data were analyzed with Kaluza Analysis 2.1
(Beckman Coulter). MSC population was determined
by forward and side light scattering. Mean fluores-
cence intensity (MFI) was assessed in APC, FITC, and
PE channels.
Relative level of gene expression analysis.
RNA isolation. To isolate RNA, the cells of the first
passage (10
5
-4.5×10
5
cells) were centrifuged at 300g.
The pellet was washed with 1 ml of phosphate buffer
and centrifuged at 300g. 400  µl of TriZol (Ambion by
Life Technologies, USA) was added to the pellet. Sam-
ples with TriZol were frozen at –70°C. After thawing,
120 µl of chloroform was added to the samples, after
which they were shaken, incubated for 2 min at room
temperature, and centrifuged for 15 min at 13,500g
and 4°C in a Centrifuge 5424 R (Eppendorf, Germany).
The resulting upper phase was transferred into new
tubes. 400 µl of isopropanol was added, the samples
were incubated for 10 min at room temperature and
centrifuged for 10 min at 13,500g and 4°C. The pellet
was washed with 1 ml of 75% ethanol, vortexed, and
centrifuged for 5 min at 13,500g at 4°C. The pellet
was left to dry for 5  min at room temperature. Next,
100 µl of DEPC-treated water was added to the pellet
and left for 30 min on ice for it to dissolve. After vor-
texing, 1 µl was taken to measure the amount of ex-
tracted RNA. The measurement was carried out with
a NanoDrop One spectrophotometer (Thermo Fisher
Scientific, USA) at a wavelength of 260 nm, RNA pu-
rity was determined by the ratio of 260/280 nm (it
should be in the range of 1.8-2.0). To the remaining
99 μl of the RNA solution, 10  μl of 3  M sodium acetate
and 250μl of 96% ethanol were added. Samples were
stored at –20°C.
cDNA synthesis. RNA in a mixture of ethanol and
sodium acetate was centrifuged for 10min at 13,500g
and 4°C. After that, the pellet was washed with 1 ml of
75% ethanol, mixed on a vortex, and centrifuged for
5 min at 13,500g and 4°C. The pellet was left to dry for
5 min at room temperature. 1  µl of DEPC-treated wa-
ter was added per 1 µg of RNA and the samples were
left on ice for 30 min for dissolution. Primers for re-
verse transcription (T13 primers and random hex-
amers) were annealed: 2 µl of RNA solution, 1.25  µl
of each primer (40 pmol/µl) and 5.5  µl of DEPC-treated
water were mixed, incubated in a Tertsik amplifier
(DNA-Technology) for 10  min at 70°C and 10 min at4°C.
After that, 15 μl of the reverse transcription mix
(5.5 µl milliQ water, 5 µl 5X M-MLV reversease buffer
(Promega, USA), 2.5 µl dNTPs mix, 1  µl each RNAsin
(Promega) and M-MLV reversease (Promega)) was add-
ed, and the samples were incubated in a Tertsik ampli-
fier at 42°C for 1 h. 75  μl of milliQ water were added.
The samples were stored at –20°C.
Real-time PCR. Real-time PCR in Taq-man modifi-
cation was performed with an AbiPrism Real Time PCR
System 7500 (Thermo Fisher Scientific) in a 96-well
plate; the reaction volume was 25µl. Each sample was
analyzed in triplicate; a positive control (a reference
mixture of cDNA from 117 donors) was used to assess
the quality of the reaction and correlate the results
of different PCRs, and a negative control was includ-
ed (water was added instead of cDNA). Sequences of
primers and probes are presented in Table  2. PCR re-
agents were mixed into a master mix (12.8 µl milliQ
water, 3.5 µl 25 mM  MgCl
2
(Thermo Fisher Scientific),
LYMPHOID NEOPLASIA 887
BIOCHEMISTRY (Moscow) Vol. 89 No. 5 2024
Table 2. Primers and probes sequences
Gene Purpose Sequence
BACT forward primer CAACCGCGAGAAGATGACC
BACT reverse primer CAGAGGCGTACAGGGATAGC
BACT probe ROX-AGACCTTCAACACCCCAGCCATGTACG-BHQ2
GAPDH forward primer GGTGAAGGTCGGAGTCAACG
GAPDH reverse primer TGGGTGGAATCATATTGGAACA
GAPDH probe ROX-CTCTGGTAAAGTGGATATTGTTGCCATCA-BHQ2
VEGFA forward primer AGGCGAGGCAGCTTGAGTTA
VEGFA reverse primer ACCCTGAGGGAGGCTCCTT
VEGFA probe FAM-CCTCGGCTTGTCACATCTGCAAGTACGT-RTQ1
FGF2 forward primer GAAGAGCGACCCTCACATCAAG
FGF2 reverse primer TCCGTAACACATTTAGAAGCCAGTA
FGF2 probe FAM-TCATAGCCAGGTAACGGTTAGCACACACTCCT-RTQ1
IL6 forward primer ACCTGAACCTTCCAAAGATG
IL6 reverse primer CTCCAAAAGACCAGTGATGA
IL6 probe FAM-ATTCAATGAGGAGACTTGCCTGGTG-RTQ1
IL8 forward primer ACCATCTCACTGTGTGTAAAC
IL8 reverse primer GTTTGGAGTATGTCTTTATGC
IL8 probe FAM-CAGTTTTGCCAAGGAGTGCTAAAG-RTQ1
PDGFRB forward primer CTCCCTTATCATCCTCATCA
PDGFRB reverse primer TCCACGTAGATGTACTCATG
PDGFRB probe FAM-TCACAGACTCAATCACCTTCCATC-RTQ1
SPP1 forward primer ATAGTGTGGTTTATGGACTGAG
SPP1 reverse primer ATTCAACTCCTCGCTTTCC
SPP1 probe FAM-CCAGTACCCTGATGCTACAGACGAG-RTQ1
BGLAP forward primer GCAGCGAGGTAGTGAAGAG
BGLAP reverse primer GAAAGCCGATGTGGTCAG
BGLAP probe FAM-CTCCCAGCCATTGATACAGGTAGC-RTQ1
PPARG forward primer TACTGTCGGTTTCAGAAATGC
PPARG reverse primer CAACAGCTTCTCCTTCTCG
PPARG probe FAM-CCATCAGGTTTGGGCGGATGCC-RTQ1
FGFR1 forward primer CAGAATTGGAGGCTACAAGG
PETINATI et al.888
BIOCHEMISTRY (Moscow) Vol. 89 No. 5 2024
Table 2 (cont.)
Gene Purpose Sequence
FGFR1 reverse primer TGATGCTGCCGTACTCATTC
FGFR1 probe FAM-CATCATAATGGACTCTGTGGTGC-RTQ1
FGFR2 forward primer CTCATTATGGAAAGTGTGGTC
FGFR2 reverse primer TGGGCCGGTGAGGCGATC
FGFR2 probe FAM-CAGGTGGTACGTGTGATTGATGGA-RTQ1
SOX9 forward primer AGCAAGACGCTGGGCAAG
SOX9 reverse primer GTTCTTCACCGACTTCCTC
SOX9 probe FAM-CTGGAGACTTCTGAACGAGAGC-RTQ1
SDF1 forward primer CTACAGATGCCCATGCCGAT
SDF1 reverse primer TAGCTTCGGGTCAATGCACA
SDF1 probe FAM-CAGTTTGGAGTGTTGAGAATTTTGAG-RTQ1
TGFB1 forward primer TGCGTCTGCTGAGGCTCAA
TGFB1 reverse primer CGGTGACATCAAAAGATAACC
TGFB1 probe FAM-AGGAATTGTTGCTGTATTTCTGGTAC-RTQ1
ICAM1 forward primer GCAATGTGCAAGAAGATAGC
ICAM1 reverse primer CTCCACCTGGCAGCGTAG
ICAM1 probe ROX-CACGGTGAGGAAGGTTTTAGCTGTT-RTQ2
2.5 µl 2.5  mM dNTPs mix, 2.5  µl 10X SmarTaq buffer
(Promega), 1 µl of forward and reverse primers each
(10 pmol/µl), 0.5  µl fluorescent probe (10 pmol/µl),
0.2  µl Taq polymerase (Promega) per reaction). 72 µl of
the master mix were placed into wells of a 96-well PCR
plate and 3 µl of the cDNA solution was added. The
samples were mixed and divided into 3 wells (25 µl
per well) to obtain triplicates. PCR started with 10-min
incubation at 95°C to activate the polymerase; 40 cy-
cles of PCR were performed for the BACT and GAPDH
genes, and 45 cycles for the remaining genes. Cycle pa-
rameters: 15 s at 95°C +  40 s at 60°C.
Calculation of relative gene expression level.
Relative expression level (REL) of genes was calculat-
ed using the modified ΔΔCt method [25]. The mean Ct
of three replicates was used for calculations. Ct is the
number of cycles required to reach the probe fluores-
cence threshold in PCR. For each gene, ΔCt was calcu-
lated using the formula(2):
ΔCt = Ct
sample
– Ct
control
. (2)
The RELs of the housekeeping genes (RELhk)
BACT and GAPDH– were calculated using the formu-
la(3):
REL
hk
= 2
–ΔCt
. (3)
Next, the sample normalization factor (NF) was
calculated using the formula (4):
NF =
REL
BACT
*
REL
GAPDH
. (4)
RELs for the genes of interest were calculated us-
ing the formula (5):
REL = 
(2
−ΔCt
)
NF
. (5)
Secretome analysis. Preparation of MSCs con-
ditioned medium. MSCs at the passages 2-3 were
seeded at 4×10
3
cells per cm
2
in T175 flasks (Costar,
USA). After the cells reached confluence (3-4 days),
the flasks were washed 5 times with phosphate buffer
LYMPHOID NEOPLASIA 889
BIOCHEMISTRY (Moscow) Vol. 89 No. 5 2024
without Ca
2+
/Mg
2+
(Invitrogen, USA). The cells were
then cultured for 24 h in RPMI1640 medium without
serum and phenol red (HyClone). The conditioned me-
dium was centrifuged at 400g and frozen at –70°C.
Sample preparation for analysis. A protease in-
hibitor cocktail (Halt Protease Inhibitor Cocktail, Ther-
mo Fisher Scientific) was added to each sample, which
were then centrifuged at 1500g for 10  min to remove
debris. Supernatants were immediately frozen and ly-
ophilized to reduce volume. The lyophilized samples
were resuspended for 30 min in a buffer containing
6 M Gd-HCl, 10  mM  Tris-HCl (pH 8) and 2  mM DTT.
Toprecipitate the insoluble fraction, the solutions were
centrifuged at 16,000g for 10min at 4°C. Samples were
concentrated using a centrifuge filter (Corning Spin-X
UF6, Sigma-Aldrich) to replace the buffer. Buffer (8 M
urea, 2 M thiourea, 10  mM Tris-HCl (pH  = 8)) was add-
ed to the concentrated samples at a ratio of 1: 3 and
incubated at room temperature for 30 min. Disulfide
bonds were reduced with 5 mM  DTT at room tempera-
ture for 40  min and next alkylated with 10 mM iodoac-
etamide in the dark at room temperature for 20 min.
Alkylated samples were diluted by adding 50 mM
NH
4
HCO
3
solution at a ratio of 1 :  4 followed by tryp-
sin addition (0.01 μg per 1 μg of protein) and incu-
bation at 37°C for 14 h. The reaction was stopped by
adding formic acid to a final concentration of 5%. The
peptides were desalted using Discovery DSC-18 tubes
(1 ml, 50  mg) (Sigma-Aldrich), dried under vacuum,
and stored at –80°C before analysis. Prior to LC-MS/MS,
samples were redissolved in 5% acetonitrile with 0.1%
trifluoroacetic acid and sonicated.
LC-MS/MS analysis. Analysis was performed us-
ing an Orbitrap Q Exactive HF-X mass spectrometer
equipped with a nano-ESI source and a high pressure
nanoflow chromatograph (UPLC Ultimate 3000) with a
C-18 (100µm) reverse phase column. ×300mm).
Bioinformatic processing of mass spectrome-
try data. Raw data from the mass spectrometer were
converted into .mgf files using MSConvert (ProteoWiz-
ard Software Foundation, USA) with the parameters
“--mgf --filter pickPicking true [1,2]”. To identify pro-
teins, a search was carried out using MASCOT (ver-
sion 2.5.1, Matrix Science Ltd., UK) and X!Tandem
(ALANINE, 2017.02.01, 2017.02.01, The Global Proteome
Machine Organization) in the UniProt human protein
database with concatenated backtrap dataset. Permis-
sible masses of the precursor and fragment were set to
20 ppm and 0.04  Da, respectively. Database search pa-
rameters included the following: tryptic digestion with
one possible gap [26], static modification for urea meth-
yl (C), and dynamic modifications for oxidation (M).
For X!Tandem, parameters were chosen that allowed
to quickly check for the acetylation of the N-terminal
residue of the protein, the loss of ammonia from the
N-terminal glutamine and water from the N-terminal
glutamic acid. The resulting files were processed in
Scaffold 5 (version 5.1.0). An algorithm for estimating
the local false discovery rate (FDR) with standard
grouping of proteins was used. To assess the hits of
peptides and proteins, FDR =  0.05 was chosen for both.
The samples annotated in the Swiss-Prot database were
marked as preferred.
Statistical analysis. Data sets are presented as in-
dividual values with indicated median. For each data
set, a normality test was performed using the Shapiro–
Wilk test (at p < 0.05, the distribution was taken to be
different from normal). Significance of differences was
analyzed using Mann–Whitney test for non-normal
distributions. Differences were considered statistical-
ly significant at p < 0.05. Statistical analysis was per-
formed using GraphPad Prism8.03.
RESULTS
Under the influence of lymphoid neoplasia, the
characteristics of MSCs change. Hematopoietic pro-
genitors that initiate tumor development in ALL and
DLBCL differ in the stage of differentiation: in ALL,
early bone marrow progenitors undergo tumor trans-
formation, while in DLBCL, it happens to more differ-
entiated B cells outside the bone marrow. MSCs only
from DLBCL patients without bone marrow involve-
ment were included in this study.
MSCs growth characteristics. Time required for
the MSCs of the patients at the onset of ALL to reach
P0 increased in comparison to the time required for
donor MSCs (p < 0.0001), which may indirectly reflect
decrease in the number of stromal progenitor cells in
the bone marrow. Upon reaching remission, this pa-
rameter returned to normal. MSCs of the patients with
DLBCL did not differ from those of the healthy do-
nors at the onset of the disease, and in remission they
reached P0 faster than those of the donors (p = 0.0107).
As a result, the time to P0 was longer in the cells from
the patients with ALL than in the MSCs from the pa-
tients with DLBCL at the onset of the disease (Fig.1a).
In remission, the time to P0 did not differ significantly.
Cumulative cell production of ALL patients’ MSCs
in 3 passages did not differ from that of the healthy
donors. When remission was achieved, total cell pro-
duction of patients with ALL increased significantly
compared to the donors (p = 0.0419). Cumulative cell
production of the MSCs from the patients with DLBCL
increased in comparison to the donors’ MSCs both at
the onset (p = 0.0119) and in remission of the disease
(p=  0.0011), and at the onset it was also significantly
higher than at the onset of ALL (Fig.1b).
Thus, we have shown that in ALL, the MSC growth
is suppressed before treatment and returns to nor-
mal in remission, while in DLBCL, on the contrary,
PETINATI et al.890
BIOCHEMISTRY (Moscow) Vol. 89 No. 5 2024
Fig. 1. Time to passage 0 (P0)(a) and cumulative cell production in 3 passages(b) of MSCs from patients with ALL and DLBCL
before treatment and in remission normalized to age-matched donor MSCs’ median value. Data are presented as scatterplots
with indicated median. Horizontal red line indicates the median values of healthy donors’ cells.
the MSCs grow more actively than the cells from the
healthy donors. Other studied characteristics of the
MSCs from the patients differed from the donors’ cells
as well.
Membrane surface marker analysis. MSCs from
the bone marrow of the patients with hematological
diseases and the healthy donors differ in the expres-
sion of surface markers.
Mean fluorescence intensity (MFI) of the surface
markers was studied on the cells of 6 patients before
treatment of ALL and 3 in remission of ALL, 9 patients
before treatment and in remission of DLBCL, and 10
donors for each of the patient groups. According to the
International Society for Cellular Therapy (ISCT) crite-
ria, MSCs express CD90, CD105, and CD73 [27]. These
markers were present on all the studied cells, but their
MFIs differed between the groups (Fig. 2, a-c). CD73
MFI was significantly increased on the MSCs from the
patients with ALL before treatment compared to do-
nors’ (Fig. 2c). This parameter did not differ from the
donors neither at the onset nor in the remission of
DLBCL. However, in the remission of DLBCL MFI of
CD73 on the MSCs became significantly higher than it
was before treatment. The MSCs of patients with DLBCL
in remission had increased MFI of CD105 (Fig. 2b). Be-
fore the treatment of ALL, the observed expression
of HLA-ABC on the cells was significantly increased
compared to donors; in remission its MFI decreased
(Fig. 2d). Before the treatment of DLBCL, HLA-ABC
MFI on the cells did not differ from donors’, but in
the remission it significantly increased. The same ef-
fect was observed for the class II histocompatibility
molecules, HLA-DR, in DLBCL (Fig. 2e). Expression of
CD146 on the surface of the MSCs in the patients with
ALL demonstrated an upward trend compared to the
donors (Fig. 2f). In the patients with DLBCL, it did not
differ from the donors before treatment, but signifi-
cantly increased when remission was achieved. There
were no significant differences in the CD54 expression
between the studied groups except for a significant
increase in the patients in remission of DLBCL com-
pared to the donors (Fig. 2g).
Significant changes in the expression of MSC sur-
face markers were detected.
Changes in gene expression of factors import-
ant for immunoregulatory function of MSCs. Pat-
terns of the MSCs gene expression were studied. The
genes selected for analysis encode factors directly in-
volved in the regulation of immune response –  IL6,
IL8, TGFB1; homing and adhesion factors –  SDF1,
ICAM1; growth factors and their receptors –  FGF2,
FGFR1, FGFR2, PDGFRB, VEGFA. Expression pattern
of the MSC differentiation marker genes BGLAP, SPP1,
SOX9, and PPARG was studied as well, since the differ-
entiated descendants of MSCs– osteoblasts, adipocytes,
etc.– participate in immune regulation.
In the patients with ALL, expression of the IL6
gene in MSCs was significantly increased in compari-
son to the donors at the onset of the disease (p < 0.0001)
and did not normalize after achieving remission
LYMPHOID NEOPLASIA 891
BIOCHEMISTRY (Moscow) Vol. 89 No. 5 2024
Fig. 2. Mean fluorescence intensity (MFI) of the surface markers CD 90(a), CD 105 (b), CD 73 (c), HLA-ABC(d), HLA-DR (e),
CD146(f), and CD54(g) on the MSCs of patients with ALL and DLBCL, and healthy donors of corresponding ages. Yaxis shows
MFI fold change relative to the respective median value for the donor cells. Data are normalized to the median values for the
donor groups and presented as scatterplots with marked median.
(p<  0.0001), and in DLBCL it increased after chemo-
therapy compared with the onset of disease (Fig.  3a)
and the healthy donors (p=  0.0018). At the same time,
at the disease onset, IL6 gene expression was higher in
ALL than in DLBCL.
Additionally, at the onset of DLBCL increase in
the IL8 expression level relative to the healthy donors
(p =  0.0220) was observed, and after treatment REL of
this gene increased even more (Fig. 3b). TGFB1 REL
was lowered in the MSCs of patients at the onset of
ALL compared to donor cells (p = 0.004) and decreased
even more in remission (p = 0.0002). In patients with
DLBCL there was also decrease in the REL of this gene
after treatment compared with the onset of the dis-
ease (Fig.3c). In the MSCs of the patients with DLBCL
at the onset and in the remission, the FGF2 REL was
increased compared to donors’ (p = 0.0121, p=  0.0206,
respectively), at the onset of the disease it was sig-
nificantly higher than in the MSCs of ALL patients
(Fig. 3d). FGF2 receptors differed in their expres-
sion in the patients with ALL and DLBCL (Fig. 3,  e, f).
At the onset of DLBCL, the MSCs had reduced FGFR1
REL (p=0.0166) and increased FGFR2 REL (p = 0.0017).
In the MSCs of the patients with ALL and DLBCL,
FGFR1 REL was insignificantly reduced at the onset
and remission of the disease, and FGFR2 expression