Skip to main content
Iranian Journal of Basic Medical Sciences logoLink to Iranian Journal of Basic Medical Sciences
. 2022 Oct;25(10):1222–1227. doi: 10.22038/IJBMS.2022.66737.14633

Mesenchymal stem cells cause induction of granulocyte differentiation of rat bone marrow C-kit+ hematopoietic stem cells through JAK3/STAT3, ERK, and PI3K signaling pathways

Ezzatollah Fathi 1, Seyed Alireza Mesbah-Namin 2, Ilja Vietor 3, Raheleh Farahzadi 4,*
PMCID: PMC9588313  PMID: 36311196

Abstract

Objective(s):

Hematopoietic stem cells (HSCs) are the cells that give rise to different types of blood cells during the hematopoiesis process. Mesenchymal stromal cells (MSCs) as key elements in the bone marrow (BM) niche interact with hematopoietic progenitor cells (HPCs) by secreting cytokines, which control HPCs maintenance and fate. Here we report that BM-MSCs are capable of inducing granulocytic differentiation of the C-Kit+ HSCs via activating JAK3/STAT3, ERK, and PI3K signaling pathways.

Materials and Methods:

For this purpose, BM-MSCs and C-kit+ HSCs were isolated. Next, cells were divided into two groups and differentiated into granulocytes: C-kit+ HSCs alone (control group) and co-cultured C-kit+ HSCs with MSCs (experimental group). Afterward, the gene and protein expression were assessed by real-time PCR and western blotting, respectively.

Results:

It was found that BM-MSCs resulted in increased JAK3/STAT3, ERK, and PI3K protein expression in granulocyte differentiated C-kit+ HSCs.

Conclusion:

It should be concluded that MSCs could affect the granulocyte differentiation of C-kit+ HSCs via increasing JAK3/STAT3, ERK, and PI3K signaling pathways.

Key Words: BM C-kit+ HSCs, Cell therapy, Granulocyte differentiation, Mesenchymal stem cells, Signaling pathways

Introduction

Mesenchymal stem cells (MSCs) are multipotent cells that can differentiate into different lineage cells including adipocytes, osteocytes, chondrocytes, etc. Today, these cells are isolated from various tissues like adipose, bone marrow (BM), umbilical cord blood (UCB), amniotic fluid, heart, liver, etc. (1, 2). Researchers and clinicians focus on BM as a significant source for experimental purposes. In addition to MSCs, BM is composed of different types of cells such as blood cells, epithelial cells (EPCs), and hematopoietic stem cells (HSCs) (3). The BM microenvironment plays a key role in the proliferation and differentiation of HSCs, MSCs, and stromal cells (4). In the BM, MSCs present a network of cytokines and interact with HSCs which cause physical support for differentiation of these through the secretion of cytokines (5). The role of MSCs in promoting HSCs proliferation and expansion has been reported in previous in vitro studies (6, 7). In one study it was pointed out that BM-derived stromal cells cause expansion of BM and CB resident CD34+ HSCs. In more detail, the role of different cytokines was proved in the study (6, 8). In 2005, it was first reported that co-transplantation of cultured MSCs and CD34+ HSCs could assist engraftment due to the supportive role of MSCs in hematopoiesis (9). In tissue engineering, it was shown that MSCs can repair damaged tissues (10-12). So, these cells are considered effective tools in therapeutic approaches (13). However, the role of different cell types in the BM niche in the differentiation of HSCs is yet to be known. Previous studies indicated that MSCs cause increased myeloid and lymphoid lineage differentiation of progenitor cells (PCs) (14). Chen et al. (2013) demonstrated that UCB-MSCs are responsible for the granulocytic differentiation process of primary acute promyelocytic leukemia by secreting interleukin (IL)-6 (15). Following that, Nikkhah et al. (2018) pointed out that BM-MSCs increased the CD11b expression in cell line HL-60 and eventually cause granulocyte differentiation (16).

The role of different signaling pathways in proliferation, differentiation, maturation, and apoptosis of HSCs was previously reported (17). Following the HSCs differentiation, a number of signal pathway proteins such as AKT, Janus family kinases (JAK), signal transducer and activator of transcription (STAT), extracellular signal-regulated kinase (ERK), phosphatidylinositol 3-kinases (PI3K), etc., have been pointed to become activated (18, 19). Investigations related to in vitro expansion of HSCs with a cell transplantation approach are among the goals of new research (20). As we know, the BM microenvironment is derived from common progenitors of mesenchymal origin, and MSCs as precursors of cellular components have an effective role in differentiation of HSCs (21).

This study aims to investigate the role of MSCs in the granulocyte differentiation potential of BM-C-kit+ HSCs via some signaling pathways.

Materials and Methods

Except for the cell culture materials which were purchased from SPL and Gibco, the rest of the materials are specified in the text of the manuscript.

The cells were divided into two groups: control group (C-kit+ HSCs alone) and experimental group (co-cultured C-kit+ HSCs with MSCs).

Isolation of BM-MSCs

BM-MSCs were isolated as previously reported by Fathi et al. (2021).(20) In brief, after obtaining the ethical code, 5 male Rattus norvegicus were euthanized, femur and tibia bones were collected, and BM was flushed. Next, the BM content was layered over the Ficoll-Paque. Mononuclear cells (MNCs) were seeded in 6-well plates containing complete culture media (DMEME low glucose+10% FBS+Pen/Strep). Cells were cultured until they reached 70–80% confluence (22).

Characterization of BM-MSCs

BM-MSCs were cultured, trypsinized, and incubated with FITC antibodies CD44, CD90, CD31, and CD34 (BD Phar Mingen, San Diego, CA, USA) in washing buffer (PBS containing 5% FBS). Next, a fluorescence-activated cell sorter was used for characterizing cells (23, 24).

Isolation of BM C-kit + HSCs

For isolating the BM-resident C-kit+ HSCs, MNCs obtained from the previous step were incubated with C-kit+ microbeads (Miltenyi Biotec, Germany). Next, re-suspended cells were passed through one LS MACS column and enriched C-kit+ cells were retrieved by flushing the column (25).

Characterization of BM C-kit + HSCs

Flow cytometry and immunocytochemistry (ICC) were performed as previously described by Fathi and Farahzadi (2022) for characterizing the BM C-kit+ HSCs (24).

Co-culture of BM C-kit + HSCs and BM-MSCs

To perform the co-culture procedure, BM-MSCs were plated into trans-well plates as two control and experimental groups (20). The granulocyte differentiation of C-kit+ HSCs was induced by 50 ng granulocyte colony-stimulating factor (GCSF) (Biolegend, Cat no: 775002). At the end of the 7th day, granulocyte-differentiated C-kit+ HSCs were subjected to Real-time PCR analysis for evaluating the granulocyte-related gene expression.

Gene expression assessment

At the end of the 7th day, granulocyte-differentiated C-kit+ HSCs were collected, total RNA was extracted and cDNA was synthesized. The mRNA expressions of CD11b, CD16, CD18, and CD34 genes were performed using Real-time PCR. Fluorescence data was calculated in relation to β-actin CT values by the 2-ΔΔCT method. Primers were listed in Table 1 (26).

Table 1.

Characteristics of primer sequences used for the Real time-PCR assays

No. Gene Primer pair sequence (5'-3') Product length (bp)
NM_012711.1 CD11b AGCCAGTTTCATCAACACAACC
GAGGTGCCCCTAAAACCAAGC
116
NM_207603.2 CD16 TCTCCAAAAGGCTGTGGTG
GACATAGTTGGCGTCCTG
160
NM_001037780.2 CD18 ACCTCTCCTACTCTATGC
AACGGAGGCTGGCAGGCTT
205
NM_001107202.2 CD34 AGACCACACCAGCCATCTC
CTTCTGGAGTAGAAGTACTG
153
NM_001101.5 β-actin TCCTCTCCCAAGTCCACACAGG
GGGCACGAAGGCTCATCATTC
131

Western blotting

At the end of the 7th day, the protein of granulocyte-differentiated C-kit+ HSCs was extracted. 50 μg of both groups were loaded to SDS-PAGE and transferred to polyvinylidene difluoride (PVDF) membranes. Next, membranes were incubated with primary antibodies β-actin (1:1000, sc-69879), AKT (1:1000, E-AB-30471), ERK 1/2 (1:1000, sc-292838), p-ERK 1/2 (1:1000, sc-16981-R), JAK3 (1:1000, sc-6932), STAT3 (1:1000, sc-8019), and PI3K (1:1000, sc-8010), and incubated with secondary antibodies (1:5000). Antigen-antibody complex was detected by enhanced chemiluminescence (ECL) reagent (27).

Statistical analysis

The results were analyzed using a t-test to determine the significant difference among groups.

Results

Characterization of BM-MSCs

BM-MSCs are spindle-shaped fibroblasts (Figures 1A-C). Also, as shown in Figures 2 A and B, the immunophenotypical characterization of BM-MSCs showed high expression levels of CD44 and CD90 and low expression levels of CD31 and CD34 surface markers (Figures 2 C and D).

Figure 1.

Figure 1

Morphology of bone marrow mesenchymal stem cells. (A) View of cells after isolation from bone marrow (scale bar = 20 μm); (B) Spindle-shaped morphology of cells (bar = 40 μm); and (C) More confluent cells (scale bar = 20 μm)

Figure 2.

Characterization of bone marrow mesenchymal stem cells. (A) BM-MSCs were positive for (A) CD44 and (B) CD90, and negative for (C) CD31 and (D) CD34. Isotype

Figure 2

control is seen with red diagrams

Characterization of BM C-kit + HSCs

Figures 3A and B show the total cell population and shift of the C-kit+ cell population (blue dots) from the isotype control (red blots), respectively. These figures demonstrated that enriched C-kit+ cells had high levels of expression of C-kit+ (94%). Also, monitoring the protein level of C-kit-related marker by ICC revealed the PE-conjugated C-kit cells (Figures 3C-E). The Fluorescence of the C-kit marker and DAPI staining of nuclei are colored in red and blue, respectively.

Figure 3.

Figure 3

Characterization of isolated bone marrow derived C-kit + cells. (A) Total population of cells for flow cytometry analysis; (B) 94% of cells were positive for C-kit marker; (C-E) Data confirmed the existence of C-kit, indicated by positive color cells by immunofluorescence imaging; Blue = DAPI; Red = PE-conjugated C-kit (bar = 20 μm)

MSCs cause increased gene expression of granulocyte markers CD11b, CD16, CD18, and CD34

To investigate the effect of MSCs on the granulocyte differentiation of BM C-kit+ HSCs, the gene expression of granulocyte markers was examined (20). The results revealed that gene expression of CD11b, CD16, CD18, and CD34 significantly increased by about 2.92, 2.59, 3.15, and 2.35 times in the experimental group (co-cultured C-kit+ HSCs with MSCs) as compared with the control group (C-kit+ HSCs alone), respectively (Figure 4). In other words, it could be claimed that MSCs play an important role in the expression of granulocyte markers of the C-kit+ HSCs with MSCs.

Figure 4.

Figure 4

Effect of BM-MSCs on (A) CD11b, (B) CD16, (C) CD18, and (D) CD34 gene expression in granulocyte differentiated BM-derived C-kit+ cells. Values are mean ± SD from independent experiments (*P<0.05 and **P<0.01, n = 3)

Western blot analysis

Following the co-culturing, granulocyte differentiated C-kit+ cells were collected and JAK3/STAT3, ERK, and PI3K as signaling components were investigated by western blotting. The expression levels of AKT, JAK3, STAT3, ERK1/2, p-ERK1/2, and PI3K were significantly increased 1.35-, 1.12-, 1.24-, 1.3-, 4.93-, and 2.1-fold, respectively) in the experimental group (Figure 5) (*P<0.05 and ***P<0.001).

Figure 5.

Figure 5

Effect of bone marrow-derived MSCs on expression of AKT, JAK3, STAT3, ERK1/2, p-ERK1/2, and PI3K in granulocyte differentiated BM-derived C-kit+ cells. Values are mean ± SD from independent experiments (*P<0.05 and ***P<0.001)

Discussion

Stem cells can be of interest in the treatment of different diseases including genetic disease, improving hematopoiesis in cancer patients under treatment, bone regeneration, repairing necrotic tissue in myocardial infarction patients, etc., thanks to their self-renewability properties and their ability to differentiate into various tissues (5, 11). Meanwhile, BM contains HSCs, MSCs, and stromal cells. HSCs residing in the BM communicate with the BM microenvironment and can produce different types of blood cells. MSCs contribute to stimulating the proliferation and differentiation of HSCs to various bloodlines through secreting growth factors, cytokines, and various chemokines such as IL-6, FLT3L, SCF, GCSF, GMCSF, etc (23). Thus, direct cell-to-cell contact as well as cytokines secreted from MSCs are involved in the co-culture of mesenchymal cells and HSCs in the hematopoiesis process and determine the fate of HSCs.

Infections are still a major challenge among cancer patients with severe long-term neutropenia following chemotherapy or transplantation with HSCs. With the emergence of granulocyte colony-stimulating factor (GCSF) to mobilize neutrophils, granulocyte injection is widely done to prevent or treat life-threatening infections among patients with neutropenia with high fever or impaired neutrophils (28).

Meanwhile, injection of granulocytes is associated with some limitations including pyrogenic reactions in response to HLA incompatibility; in case this proposal succeeds, through direct injection of cytokines secreted from MSCs, in stimulating HSCs, which are a part of BM single-nucleus cells in patients with neutropenia (for example, the neutropenic phase before BM transplantation is associated with neutropenia), a significant role can be done towards granulocyte line (28). This is because we know that reduction of granulocytes as the main part of the innate immune system in individuals undergoing chemotherapy or treatments such as BM transplantation is one of the major risks and complications, causing the incidence of numerous problems such as opportunistic infections. Reduction of the duration of renewal of new cells is one of the challenges ahead of medical research, and the positive results obtained from research in this regard can significantly help in driving therapeutic goals and providing newer methods with a more intensive and effective treatment model against malignant tissues.

Meanwhile, other studies have been reported regarding the positive effect of injecting MSCs alongside or together with BM mononuclear cells in cell transplantation. Possibly, the reason behind this positive effect is cytokines and factors secreted from MSCs. This highlights the necessity of the usage of research on cytokines affected or secreted by MSCs (19). The role of MSCs in differentiation to erythroid and myeloid lines is possibly due to the secretion of specific cytokines (29). However, so far the role of cytokines secreted from MSCs in the induction of granulocyte differentiation of HSCs has not been studied.

Various studies have been performed on the role of MSCs in the differentiation of HSCs. Tocci and Forte (2003) reported that BM-MSCs can maintain long hematopoiesis under in vitro conditions (30). Further, the signaling pathways involved in the induction of differentiation of hematopoiesis of HSCs have been examined in several studies. In this regard, pathways such as ERK, Wnt, Notch, SMAD, etc., have been reported as important pathways in self-renewal, differentiation, proliferation, apoptosis, and aging of HSCs. The effective factors in the interaction between MSCs and HSCs have also been reported in other studies. Kikuchi et al. (2011) reported that when HSCs are subject to MSCs, the expression of Notch ligands in MSCs increases through the Wnt pathway in HSCs (31). In another study by Simmons and Torok-Storb in 1991, it was found that MSCs in the BM create signals for differentiation and proliferation of HSCs (32). Also, in the study by Moreau et al. (2005), the role of cytokines as well as factors secreted and affected by MSCs on HSCs have been reported (33). In the study by Dilva et al. in 2005, CD34+ HSCs were cultured in a medium fortified with various cytokines in the presence and absence of MSCs (34). It was observed that MSCs supported the growth and differentiation induction of HSCs. In another study by Mehrasa et al. (2014), HSCs of the umbilical cord were subjected to co-culture conditions alongside MSCs (35). They found that the MSCs led to high proliferation and reduced apoptosis of umbilical cord HSCs due to secretion of cytokines and various growth factors (35). Meanwhile, another study reported that differentiation of HSCs during their development increases the aging process and cellular death, but this direct contact between cells and MSCs indicates their role in hematopoiesis versus differentiation. More comprehensively based on studies, MSCs produce diverse cytokines and factors affecting the differentiation of HSCs.

In the present study, the results showed that MSCs cause a significant increase in the protein expression levels of JAK3/STAT3, ERK, and PI3K. In a similar investigation by Fathi et al. (2021), it was reported that co-culturing of MNCs with BM-MSCs causes an increase in the expression levels of granulocyte markers of MNCs, e.g., CD34, CD16, CD11b, and CD18. Based on these data, one can conclude that MSCs may affect the granulocyte differentiation of MNCs via ERK protein expression (20).

Conclusion

Due to problems in the granulocyte injection pathway using GCSF, further research is required regarding alternative pathways. Since MSCs have a supportive role in BM and produce effective growth factors in hematopoiesis, thus there is the possibility of inducing the differentiative effect of these cells on HSCs. In addition, the interaction between two cell lines and the signaling pathways activated resulting from this interaction provides the possibility of proliferation and development of HSCs in the undifferentiated state.

Authors’ Contributions

EF as the executive of the project had main contribution in conception and design, data analysis, and manuscript writing; IV as the main colleague of the project in providing the kits and some materials needed for this project as well as interpretation of experimental procedure; RF and SAM-N as colleagues involved in performance of experiments, data analysis and manuscript writing.

Funding Statment

This work has been supported by the Center for International Scientific Studies & Collaboration (CISSC), Ministry of Science Research and Technology.

Data Availability Statement

The data sets used and/or analyzed during the current study are available from the corresponding author upon reasonable request.

Ethics Approval and Consent to Participate

Ethical consent was given by the ethics committee at Tabriz University of Medical Sciences, (Ethics code no: IR.TBZMED.VCR.REC.1397.322) in accordance with the guidelines of the Helsinki-Ethical Principles.

Conflicts of Interest

The authors declare that they have no conflicts of interest.

Acknowledgment

The authors wish to thank staff of the Faculty of veterinary Medicine, University of Tabriz, Tabriz, Iran and Medical University Innsbruck, Innrain 80-82, A-6020, Innsbruck, Austria.

References

  • 1.Heidari HR, Fathi E, Montazersaheb S, Mamandi A, Farahzadi R, Zalavi S, et al. Mesenchymal stem cells cause telomere length reduction of molt-4 cells via caspase-3, bad and p53 apoptotic pathway. Int J Mol Cell. 2021;10:1–10. doi: 10.22088/IJMCM.BUMS.10.2.113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Hass R, Kasper C, Böhm S, Jacobs R. Different populations and sources of human mesenchymal stem cells (MSC): A comparison of adult and neonatal tissue-derived MSC. Cell Commun Signal. 2011;9:1–14. doi: 10.1186/1478-811X-9-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Galán-Díez M, Cuesta-Domínguez Á, Kousteni S. The bone marrow microenvironment in health and myeloid malignancy. Cold Spring Harb Perspect Med. 2018;8:a031328. doi: 10.1101/cshperspect.a031328. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Montazersaheb S, Ehsani A, Fathi E, Farahzadi R, Vietor I. An overview of autophagy in hematopoietic stem cell transplantation. Front Bioeng Biotechnol. 2022;10:849768. doi: 10.3389/fbioe.2022.849768. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Wu KH, Wu HP, Chan CK, Hwang SM, Peng CT, Chao YH. The role of mesenchymal stem cells in hematopoietic stem cell transplantation: from bench to bedsides. Cell Transplant. 2013;22:723–729. doi: 10.3727/096368912X655217. [DOI] [PubMed] [Google Scholar]
  • 6.Li N, Feugier P, Serrurrier B, Latger-Cannard V, Lesesve JF, Stoltz JF, et al. Human mesenchymal stem cells improve ex vivo expansion of adult human CD34+ peripheral blood progenitor cells and decrease their allostimulatory capacity. Exp Hematol. 2007;35:507–515. doi: 10.1016/j.exphem.2006.10.015. [DOI] [PubMed] [Google Scholar]
  • 7.Ito Y, Hasauda H, Kitajima T, Kiyono T. Ex vivo expansion of human cord blood hematopoietic progenitor cells using glutaraldehyde-fixed human bone marrow stromal cells. J Biosci Bioeng. 2006;102:467–469. doi: 10.1263/jbb.102.467. [DOI] [PubMed] [Google Scholar]
  • 8.da Silva CL, Gonçalves R, Crapnell KB, Cabral JM, Zanjani ED, Almeida-Porada G. A human stromal-based serum-free culture system supports the ex vivo expansion/maintenance of bone marrow and cord blood hematopoietic stem/progenitor cells. Exp Hematol. 2005;33:828–835. doi: 10.1016/j.exphem.2005.03.017. [DOI] [PubMed] [Google Scholar]
  • 9.Lazarus HM, Koc ON, Devine SM, Curtin P, Maziarz RT, Holland HK, et al. Cotransplantation of HLA-identical sibling culture-expanded mesenchymal stem cells and hematopoietic stem cells in hematologic malignancy patients. Biol Blood Marrow Transplant. 2005;11:389–398. doi: 10.1016/j.bbmt.2005.02.001. [DOI] [PubMed] [Google Scholar]
  • 10.Ebrahimi T, Abasi M, Seifar F, Eyvazi S, Hejazi MS, Tarhriz V, et al. Transplantation of stem cells as a potential therapeutic strategy in neurodegenerative disorders. Curr Stem Cell Res Ther. 2020;16:133–144. doi: 10.2174/1574888X15666200628141314. [DOI] [PubMed] [Google Scholar]
  • 11.Fathi E, Sanaat Z, Farahzadi R. Mesenchymal stem cells in acute myeloid leukemia: a focus on mechanisms involved and therapeutic concepts. Blood research. 2019;54:165–174. doi: 10.5045/br.2019.54.3.165. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Fathi E, Valipour B, Farahzadi R. Targeting the proliferation inhibition of chronic myeloid leukemia cells by bone marrow derived-mesenchymal stem cells via erk pathway as a therapeutic strategy. Acta Medica Iranica. 2020;58:198–206. [Google Scholar]
  • 13.Fathi E, Vandghanooni S, Montazersaheb S, Farahzadi R. Mesenchymal stem cells promote caspase-3 expression of SHSY5Y neuroblastoma cells via reducing telomerase activity and telomere length. Iran J Basic Med Sci. 2021;24:1583–1589. doi: 10.22038/IJBMS.2021.59400.13187. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Briquet A, Dubois S, Bekaert S, Dolhet M, Beguin Y, Gothot A. Prolonged ex vivo culture of human bone marrow mesenchymal stem cells influences their supportive activity toward NOD/SCID-repopulating cells and committed progenitor cells of B lymphoid and myeloid lineages. Haematologica. 2010;95:47–56. doi: 10.3324/haematol.2009.008524. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Chen F, Zhou K, Zhang L, Ma F, Chen D, Cui J, et al. Mesenchymal stem cells induce granulocytic differentiation of acute promyelocytic leukemic cells via IL-6 and MEK/ERK pathways. Stem Cells Dev. 2013;22:1955–1967. doi: 10.1089/scd.2012.0621. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Nikkhah H, Safarzadeh E, Shamsasenjan K, Yousefi M, Lotfinejad P, Talebi M, et al. The effect of bone marrow mesenchymal stem cells on the granulocytic differentiation of HL-60 cells. Turk J Hematol. 2018;35:42–48. doi: 10.4274/tjh.2016.0498. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Richter J, Traver D. The role of Wnt signaling in hematopoietic stem cell development. Crit Rev Biochem Mol Biol. 2017;52:414–424. doi: 10.1080/10409238.2017.1325828. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Miranda M, Johnson D. Signal transduction pathways that contribute to myeloid differentiation. Leukemia. 2007;21:1363–1377. doi: 10.1038/sj.leu.2404690. [DOI] [PubMed] [Google Scholar]
  • 19.Montazersaheb S, Fathi E, Farahzadi R. Cytokines and signaling pathways involved in differentiation potential of hematopoietic stem cells towards natural killer cells. Tissue Cell. 2021;70:101501. doi: 10.1016/j.tice.2021.101501. [DOI] [PubMed] [Google Scholar]
  • 20.Fathi E, Azarbad S, Farahzadi R, Javanmardi S, Vietor I. Effect of rat bone marrow derived-mesenchymal stem cells on granulocyte differentiation of mononuclear cells as preclinical agent in cellbased therapy. Curr Gene Ther. 2021;22:152–161. doi: 10.2174/1566523221666210519111933. [DOI] [PubMed] [Google Scholar]
  • 21.Aqmasheh S, Shamsasanjan K, Akbarzadehlaleh P, Pashoutan Sarvar D, Timari H. Effects of mesenchymal stem cell derivatives on hematopoiesis and hematopoietic stem cells. Adv Pharm Bull. 2017;7:165–177. doi: 10.15171/apb.2017.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Adibkia K, Ehsani A, Jodaei A, Fathi E, Farahzadi R, Barzegar-Jalali M. Silver nanoparticles induce the cardiomyogenic differentiation of bone marrow derived mesenchymal stem cells via telomere length extension. Beilstein J Nanotechnol. 2021;12:786–797. doi: 10.3762/bjnano.12.62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Fathi E, Vietor I. Mesenchymal stem cells promote caspase expression in Molt-4 leukemia cells Via GSK-3α/and ERK1/2 signaling pathways as a therapeutic strategy. Curr Gene Ther. 2021;21:81–88. doi: 10.2174/1566523220666201005111126. [DOI] [PubMed] [Google Scholar]
  • 24.Fathi E, Farahzadi R. Mesenchymal stem cells as a cell-based therapeutic strategy targeting the telomerase activity of KG1 acute myeloid leukemia cells. Acta Medica Iranica. 2022;60:71–77. [Google Scholar]
  • 25.Czarna A, Sanada F, Matsuda A, Kim J, Signore S, Pereira JD, et al. Single-cell analysis of the fate of c-kit-positive bone marrow cells. NPJ Regen Med. 2017;2:27–42. doi: 10.1038/s41536-017-0032-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Montazersaheb S, Avci ÇB, Bagca BG, Ay NPO, Tarhriz V, Nielsen PE, et al. Targeting TdT gene expression in Molt-4 cells by PNA-octaarginine conjugates. Int J Biol Macromol. 2020;164:4583–4590. doi: 10.1016/j.ijbiomac.2020.09.081. [DOI] [PubMed] [Google Scholar]
  • 27.Bagheri Y, Barati A, Nouraei S, Namini NJ, Bakhshi M, Fathi E, et al. Comparative study of gavage and intraperitoneal administration of gamma-oryzanol in alleviation/attenuation in a rat animal model of renal ischemia/reperfusion-induced injury. Iran J Basic Med Sci. 2021;24:175–183. doi: 10.22038/IJBMS.2020.51276.11642. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Chen J, Pan Y. The safety and clinical efficacy of recombinant human granulocyte colony stimulating factor injection for colon cancer patients undergoing chemotherapy. Rev Assoc Med Bras. 2017;63:1061–1064. doi: 10.1590/1806-9282.63.12.1061. [DOI] [PubMed] [Google Scholar]
  • 29.Fathi E, Farahzadi R, Valipour B, Sanaat Z. Cytokines secreted from bone marrow derived mesenchymal stem cells promote apoptosis and change cell cycle distribution of K562 cell line as clinical agent in cell transplantation. PLoS One. 2019;14:e0215678. doi: 10.1371/journal.pone.0215678. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Tocci A, Forte L. Mesenchymal stem cell: use and perspectives. Hematol J. 2003;4:92–96. doi: 10.1038/sj.thj.6200232. [DOI] [PubMed] [Google Scholar]
  • 31.Kikuchi Y, Kume A, Urabe M, Mizukami H, Suzuki T, Ozaki K, et al. Reciprocal upregulation of Notch signaling molecules in hematopoietic progenitor and mesenchymal stromal cells. J Stem Cells Regen Med. 2011;7:61–68. doi: 10.46582/jsrm.0702010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Simmons PJ, Torok-Storb B. Identification of stromal cell precursors in human bone marrow by a novel monoclonal antibody, STRO-1. Blood. 1991;78:55–62. [PubMed] [Google Scholar]
  • 33.Moreau JE, Chen J, Horan RL, Kaplan DL, Altman GH. Sequential growth factor application in bone marrow stromal cell ligament engineering. Tissue Eng. 2005;11:1887–1897. doi: 10.1089/ten.2005.11.1887. [DOI] [PubMed] [Google Scholar]
  • 34.da Silva CL, Gonçalves R, dos Santos F, Andrade PZ, Almeida-Porada G, Cabral JM. Dynamic cell-cell interactions between cord blood haematopoietic progenitors and the cellular niche are essential for the expansion of CD34+, CD34+CD38- and early lymphoid CD7+ cells. J Tissue Eng Regen Med. 2010;4:149–158. doi: 10.1002/term.226. [DOI] [PubMed] [Google Scholar]
  • 35.Mehrasa R, Vaziri H, Oodi A, Khorshidfar M, Nikogoftar M, Golpour M, et al. Mesenchymal stem cells as a feeder layer can prevent apoptosis of expanded hematopoietic stem cells derived from cord blood. Int J Mol Cell Med. 2014;3:1–10. [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

The data sets used and/or analyzed during the current study are available from the corresponding author upon reasonable request.


Articles from Iranian Journal of Basic Medical Sciences are provided here courtesy of Mashhad University of Medical Sciences

RESOURCES