Skip to main content

MINI REVIEW article

Front. Immunol., 09 October 2023
Sec. Cancer Immunity and Immunotherapy

Mesenchymal stem/stromal cells- a principal element for tumour microenvironment heterogeneity

Li SunLi SunYongliang Yao*Yongliang Yao*
  • Department of Clinical Laboratory, Kunshan First People’s Hospital, Affiliated to Jiangsu University, Kunshan, China

The heterogeneity of the tumor microenvironment (TME) is a major obstacle in cancer treatment, making most therapeutic interventions palliative rather than curative. Previous studies have suggested that the reason for the low efficacy of immunotherapy and the relapse of the original responders over time may be due to the complex network of mesenchymal stem/stromal cells (MSCs), a population of multipotent progenitor cells existing in a variety of tissues. Cancer-associated MSCs (CA-MSCs) have already been isolated from various types of tumors and are characterized by their vigorous pro-tumorigenic functions. Although the roles of CA-MSCs from different sources vary widely, their origins are still poorly understood. Current evidence suggests that when local resident or distally recruited MSCs interact with tumor cells and other components in the TME, “naïve” MSCs undergo genetic and functional changes to form CA-MSCs. In this review, we mainly focus on the multiple roles of CA-MSCs derived from different sources, which may help in elucidating the formation and function of the entire TME, as well as discover innovative targets for anti-cancer therapies.

Introduction

The International Society for Cellular Therapy issued a set of minimal standards for the recognition of mesenchymal stem/stromal cells (MSCs) in 2006 based on three principles: Firstly, in typical culture settings, MSCs must adhere to the plastic. Secondly, it is essential that MSCs express CD105, CD90 and CD73, while they must be negative for CD 45, CD 34, CD 14 or 11b, CD 79α or 19, and HLA-DR. Lastly, MSCs are capable of differentiating into adipocytes, chondrocytes, and osteoblasts under certain circumstances (13). In healthy tissues, MSCs play a key role in wound healing and immune regulation. In the bone marrow niche, MSCs play an important role in hematopoietic stem cell maintenance and lineage determination by secreting factors, and contribute to the construction of the niche (4). Although MSCs account for a relatively small proportion of the tumor microenvironment (TME), with studies reporting that they make up 0.01–5% of the total cell population, they can amplify their effects by directly altering the matrix composition of the TME and influencing the behavior of their offspring given the capacity for multi-directional differentiation (47).

It has been found that MSCs that reside within tumor tissue have high tumorigenicity compared to MSCs from healthy tissue (8). In-depth knowledge of the origin, heterogeneity, and functions of MSCs is critical to understanding disease progression and elucidating potential targets for novel therapeutic interventions.

Origins of CA-MSCS

MSCs are a heterogeneous group of progenitor cells with both pro-tumor and anti-tumor effects (911). MSCs are recruited from adjacent tissues or circulating bone marrow to the tumor sites and reprogrammed into cancer-associated MSCs (CA-MSCs). Current evidence suggests that both recruited and local resident MSCs are major sources of CA-MSCs (12).

The recruitment and reprogramming of MSCS

Chemokines and cytokines secreted by tumor cells and the surrounding stroma have been shown to be involved in the mechanisms of migration or homing of the circulating bone marrow MSCs (BMMSCs) to the TME. It is well known that tumor-produced CCL2, CCL5, CXCL12 and CXCL16 play crucial roles in the chemotaxis of BMMSCs (13). In addition, cytokines such as PDGF, TGF-β, VEGF and TNF-α were also found to play important roles in BMMSCs homing (14, 15). For example, a study in prostate cancer showed that TGF-β1 is a key molecule that regulates the distant recruitment of BMMSCs into tumor sites (16). MSCs present in adjacent normal tissues express the homing spectrum of inflammatory signals from tumor cells, and are thus anchored in the tumor mass (17). However, the signaling pathways and underlying mechanisms by which MSCs migrate from adjacent normal tissues to tumor sites are still unknown.

Based on evidence, tumor cells can effectively reprogram the phenotype and function of “naïve” MSCs. Le et al. have demonstrated that ovarian cancer cells cause “naïve” MSCs to express CD73, CD90 and CD105, while CD14, CD20, CD34 and CD45 are absent. Moreover, the levels of CXCL1, CXCL2 and IL-8 secreted by MSCs increase through PI3K/Akt, MAPK and NF-κB signaling pathways, which in turn make tumor cells resistant to chemotherapy and the differentiation of monocytes into M2 type (18). Wang et al. have shown that gastric cancer cells induce “naïve” mesenchymal stem cells have pro-metastatic behavior by activating YAP signaling through the exosomal Wnt5a (19). Exosomes derived from gastric cancer cell cause abnormal activation of the NF-kB signaling pathway in MSCs, thus promoting the secretion of pro-inflammatory factors by macrophages and the activation of CD69 and CD25 on the surface of T cells (20). In addition to the tumor cells, the surrounding microenvironment also plays a critical role in the malignant transformation of “naïve” MSCs into CA-MSCs. For example, mononuclear/macrophage-derived TNF-a makes “naïve” MSCs similar to CA-MSCs in their chemokine profile and ability to promote tumorigenicity in lymphomas (21). Moreover, neutrophils treated with tumor cells activate the AKT and p38 pathways in “naïve” MSCs by secreting inflammatory factors such as IL-17, IL-23 and TNF-α, and transform them into CA-MSCs, thus significantly promoting the growth and metastasis of gastric cancer (22).

MSCS naturally residing in the TME

As one of the key stromal cell types in the tumor niche, CA-MSCs have been isolated from various tumors and have been found to play an important role in tumor progression (Table 1). MSCs isolated from gastric cancer tissues (GCMSCs) promote cancer progression and contribute to tumor immunotherapy tolerance through the CXCR2–HK2–PD-L1 pathway (23). IL-8 derived from GCMSCs induces the expression of PD-L1 in GC cells through c-Myc regulated by the STAT3 and mTOR signaling pathways and inhibits the anti-tumor immune response (24). GCMSCs also disrupt the Treg/Th17 balance in peripheral blood mononuclear cells (PBMCs) by inhibiting Th17 cell proliferation and inducing Tregs differentiation, thereby impairing the anti-tumor immune response (25). GCMSCs reduce the frequency of infiltrating NK cells and inhibit their effector function to promote tumor growth through the mTOR signaling pathway (26). Lung cancer-associated MSCs exert strong metastasis-promoting effects by producing complement C3, which is induced and maintained by Th2 cytokines in a STAT6-dependent manner and also promote tumor metastasis and tumorigenesis by induction of EMT and stem-like reprogram (27, 28). Single-cell transcriptional analysis of MSCs in invasive breast cancer TME revealed increased expression of the COL10A1 and COL8A1 genes, as well as pro-tumor effects driven by TGF-β-related signals (29). Breast cancer-associated MSCs also promote cancer proliferation and enhance mammosphere formation partially via EGF–EGFR–AKT pathway (30). MSCs residing in other solid tumors also reported to contribute to tumor progression, such as pancreatic cancer, cervical cancer, colon cancer, head and neck squamous cell cancer and neuroblastoma (3135). In general, MSCs residing in TME not only up-regulate the proliferation, migration and invasion ability of tumor cells and their PD-L1 expression, but also disrupt the balance between immune cells, thus inhibiting the anti-tumor immune response (Figure 1).

TABLE 1
www.frontiersin.org

Table 1 The functions and signaling pathways of MSCs naturally residing within the TME.

FIGURE 1
www.frontiersin.org

Figure 1 Schematic of proposed network of MSCs residing in TME inhibit anti-tumor immune responses and promote tumor progression by modulating tumor cells and immune cells through multiple signaling pathways. MSC, mesenchymal stem/stromal cell; PD-L1, programmed death-1 ligand; EVs, extracellular vesicles; IL, interleukin; STAT, signal transducer and activator of transcription; EGF, epidermal growth factor; EGFR, epidermal growth factor receptor; TGF-β, transforming growth factor-β; GM-CSF, granulocyte-macrophage colony-stimulating factor; HK2, hexokinase 2; mTOR, mammalian target of rapamycin; JAK2, janus kinase 2; Th, T helper; C3, complement 3; Treg, regulatory T cell; NK, natural killer.

Besides solid tumors, hematological malignancies also rely heavily on support of host cells, specifically MSCs (36). The development of multiple myeloma MSCs (MM-MSCs) may be the result of multiple factors, and changes may vary depending on the individual, lesion location, co-culture myeloma cell type, and cell subsets within the MSC population. MM-MSCs differ from non-diseased donors (ND-MSCs) in a number of ways, including cytokine production. MM-MSCs secrete higher levels of growth factors such as SCF, VEGF, and IL-6, than ND-MSCs. Recent studies have shown that, compared with ND-MSCs, MM-MSCs respond to myeloma cells with increased expression of IL-10, the TNF family member B-cell activating factor, and HGF (37, 38). MM-MSCs, but not ND-MSCs, can also induce bortezomib resistance via enhanced NF-κB activity in MM cells (39). MM-MSCs are spatially co-located with tumor cells and immune cells which express genes involved in tumor survival and immune regulation. Inflammatory MSCs (iMSCs) are specific for MM bone marrow. The development of iMSCs is the result of NF-κB signaling activated by inflammatory mediators (40).

Different properties between CA-MSCS and “naïve” MSCS

Interactions with tumor cells and other components in the TME lead to genetic and functional changes in “naïve” MSCs (18, 41). CA-MSCs show stronger proliferation and migration abilities and have stronger tumor-promoting effects than “naïve” MSCs. In addition, the number of G0/G1 phase cells is increased in CA-MSCs compared to their counterparts in neuroblastoma, suggesting that CA-MSCs play an important role in regulating cancer dormancy. Compared with “naïve” MSCs, CA-MSCs also exhibit different adipogenic differentiation and immunomodulatory abilities (12).

Regulatory effects of MSCS from different sources on tumor cells

Evidence indicates that the specific effects of MSCs on tumors depend on the origin and tumor type (Table 2).

TABLE 2
www.frontiersin.org

Table 2 The phenotypes and functions of MSCs on tumor cells from different sources.

Bone marrow MSCs

BMMSCs promote metastasis by activating the ABL–MMP9 signaling axis in lung cancer cells (42). BMMSCs secreted IL-6 contribute significantly to pancreatic cancer growth (43). However, BMMSCs-derived IL-28 trigger prostate cancer cell apoptosis via IL-28Rα–STAT1 signaling pathway (44). The plasticity of cancer stem-like cells (CSCs) is one of the primary elements responsible for the heterogeneity of the TME (63). Estrogen receptor-positive breast cancer cells make direct contact with BMMSCs and acquire CSCs phenotype with increased resistance to standard antiestrogenic drugs (45). Extracellular vesicles released by BMMSCs instruct breast cancer cells to become dormant CSCs with chemoresistance capacity (46). Cancer cell-stimulated BMMSCs also create a CSCs niche via the release of prostaglandin E2 (64). IL-6 secreted by tumor-stimulated BMMSCs, enhances the stemness and migration capacity of osteosarcoma cells (47). BMMSCs-derived exosomes promote colon CSCs traits via miR-142-3p (48).

Umbilical cord MSCs

Umbilical cord MSCs (UCMSCs) are known to facilitate the proliferation and movement of lung cancer cells through the ERK–phospho-c-Fos-S374 pathway and to induce malignant characteristics of cancer cells through the AMPK signaling pathway (49, 50). UCMSCs inhibit the growth and metastasis and promote the apoptosis of lung cancer cells by regulating the PI3K–Akt and NF-κB pathways (51). UCMSCs inhibit the proliferation, migration and invasion of glioblastoma cells by regulating the IL-6–JAK2–STAT3 signaling pathway, and promote the apoptosis of glioblastoma cells (52). Exosomes derived from UCMSCs have anti-proliferative, pro-apoptotic, and anti-angiogenic effects on hepatocellular carcinoma cells (53). Exosomes secreted by UCMSCs also inhibit macrophage M2 polarization and prevent liver metastasis of colorectal cancer by inducing miR-1827 to target SUCNR1 (54). Regulation of ITGA2/ITGA6 has been demonstrated to be the means by which UCMSCs suppress the proliferation, migration and invasion of colon cancer cells (55).

Adipose tissue-derived MSCs

Adipose tissue-derived MSCs (ADMSCs) can be used in the treatment of lung metastatic melanoma therapy by producing the anti-angiogenic factor TSP-1 (56). ADMSCs can increase the efficacy of chemotherapy in hepatocellular carcinoma treatment (57). ADMSCs also accelerate the progression of colon cancer by inducing an MSC-transformed cancer-associated fibroblast phenotype through the ICAM1–STAT3–AKT axis (58). ADMSCs promote the proliferation and migration of head and neck cancer cells (59). In addition, preclinical treatment of breast cancer is aided by ADMSCs, which exhibit anti-tumor activity (60). It has been demonstrated that CCL5 secreted by ADMSCs can induce PD-L1 expression in breast cancer cells (61). Exosomes derived from ADMSCs reduce glioblastoma proliferation and significantly down-regulate the invasion-related genes ITGα5 and ITGβ3, as well as the angiogenesis induction gene KDR (62).

Naturally residing MSCs

IL-6 derived from ameloblastoma MSCs (AMMSCs) induces epithelial–mesenchymal transition (EMT) and promotes the formation of CSCs through the STAT3 and ERK1/2 signaling pathways (65). Endometrial cancer-derived MSCs exhibit high expression of PD-L1 and PD-L2, which can be augmented by TNF-α and IFN-γ (66). Additionally, our previous studies have revealed that IL-8 derived from GCMSCs could promote immune escape by inducing PD-L1 expression in GC cells via c-Myc, which is regulated by the STAT3 and mTOR signaling pathways (24). Moreover, our findings have shown that GCMSCs upregulate PD-L1 expression not only in the cell membrane of GC cells, but also in the cytoplasm and nucleus. Checkpoint blocking antibodies only block immune checkpoints on cell membranes. Thus, their effects may be counteracted by checkpoints from cytoplasmic translocation (67). The accumulation of nuclear PD-L1, which is promoted by deacetylation, has been reported to activate genes involved in the immune response (68). Evidence has suggested that PD-L1 expression is linked to tumor cells’ intrinsic properties (69). GCMSCs can also induce changes in PD-L1 expression by regulating the intrinsic metabolism of tumor cells (23). We have shown that GCMSCs can augment the CSCs properties of GC cells through PD-L1, thus leading to GC cells’ resistance to chemotherapy (67).

Regulatory effects of MSCS from different sources on immune cells

MSCs not only regulate tumor cells to boost the growth and progression, but also have effects on a variety of immune cells to modify the complex immune microenvironment (Figure 2).

FIGURE 2
www.frontiersin.org

Figure 2 Framework diagram of MSCs derived from different sources exerting broad immunosuppressive effects through the interactions with immune cells, such as macrophages, dendritic cells, T cells, B cells and neutrophil. BMMSC, bone marrow mesenchymal stem/stromal cell; UCMSC, umbilical cord mesenchymal stem/stromal cell; ADMSC, adipose tissue-derived mesenchymal stem/stromal cell; PD-L1, programmed death-1 ligand; MΦ, macrophage; DC, dendritic cell; CTLA-1, cytotoxic T-lymphocyte antigen 4; Treg, regulatory T cell; Th, T helper; IL, interleukin; TGF-β, transforming growth factor-β; IFN, interferon; Ig, immunoglobulin; G-CSF, granulocyte colony-stimulating factor; FGF2, fibroblast growth factor 2; SERPINE1, serpin family E member 1; VEGF, vascular endothelial growth factor.

Bone marrow MSCs

The quantity and quality of tumor-killing T cells in the TME are essential for the success of immune therapeutics; however, due to the expression of immune checkpoints, the majority of infiltrating T cells are unable to identify and eliminate neighboring tumor cells (70, 71). The PD-1–PD-L1 axis is involved in the BMMSCs-mediated inhibition of T cell glycolysis by negatively regulating HK2 activity (72). IL-17 can also significantly increase the level of PD-L1 produced by BMMSCs through inducing iNOS expression to shape the immunosuppressive TME (73). Moreover, BMMSCs raise PD-1 expression on activated CD4+CD25 T cells and induce apoptosis of these cells (74). Endothelial cells were safeguarded from cytotoxic T lymphocyte-induced lysis by co-culturing with BMMSCs (75). Besides, BMMSCs help create an immunosuppressive microenvironment in tumors by reducing the proportion of Th17 cells and increasing the proportion of Tregs in PBMCs (76). Studies have demonstrated that BMMSCs can drastically impede the growth of CD4+ and CD8+ T cells through cell-contact and paracrine-dependent means (77). Moreover, MSCs not only have an effect on T cell viability and proliferation, but also impede the production and operation of Th1 and Th17 cells, while encouraging the production of Th2 cells and Tregs (78, 79). Dendritic cells educated by BMMSCs initiate a transition from Th1 to Th2, stimulating Tregs and leading to a decrease in pro-inflammatory cytokines such as IL-6, IL-17, and IFN-γ, as well as an upsurge in IL-4 and IL-10 production (80). Another important soluble factor derived from BMMSCs is IL-1RA; BMMSCs from IL-1RA-deficient mice do not inhibit B cell differentiation (81).

Umbilical cord MSCs

UCMSCs attenuate acute lung injury via PGE2-dependent reprogramming of macrophages to promote their PD-L1 expression (82). Besides, UCMSCs shift the cytokine profile of dendritic cells from proinflammatory to immunoregulatory (83). A study has reported that human UCMSCs disrupt the maturation of B cells by secreting TGF-β (84). Che et al. also have reported that UCMSCs inhibit the production of IgG and IgM in B cells (85). TGF-β secreted by UCMSCs enhances the induction of Tregs from “naïve” T cells to suppress colitis-associated colorectal cancer (86). UCMSCs inhibit the tumorigenesis of colon cancer by reducing the proportion of macrophages and inhibiting TNF-α and IL-6 secretion (87).

Adipose tissue-derived MSCs

ADMSCs promote the activity of neutrophils by inhibiting apoptosis mediated by the increased expression of TGF-β, IFN-α, and G-CSF (88). Moreover, ADMSCs-derived exosomes enhance the secretion of TGF-β and IL-10 and impede the secretion of IL-6 by dendritic cells (89). ADMSCs upregulate FGF2, SERPINE1, VEGFA and VEGFC in macrophages and supporting the proliferation of breast cancer cells (90). Exosomes secreted by ADMSCs enable macrophages to switch from the M1 to the M2 phenotype, thus possessing potent anti-inflammatory properties (91). In the presence of ADMSCs, the proportion of Tregs increases and the production of IL-10, IL-17, and TGF-β by T cells is enhanced (92).

Naturally residing MSCs

Endometrial cancer-derived MSCs inhibit the proliferation of PBMCs, which can be partially rescued by treatment with anti-PD-L1 antibodies (66). Glioma-associated MSCs enhance the immunosuppressive activity of MDSCs through a positive feedback loop of miR-21–SP1–DNMT1 (93). GCMSCs efficiently promote macrophage polarization toward the pro-tumor M2 subtype through several soluble molecules, including IL-6, IL-8, TGF-β, HGF, PGE2, IL-1RA, TSG6 and IDO (94). MSCs from head and neck squamous cell cancer inhibit the proliferation of CD4+ and CD8+ T cells through indoleamine 2,3 dioxygenase activity (34). GCMSCs inhibit the degranulation ability, perforin production, and cytotoxicity of NK cells by up-regulating the expression of fructose-bisphosphatase 1 (95). Moreover, Lung cancer-associated MSCs reduce NK cells cytotoxicity by expressing IL-6 and prostaglandin E2 (96).

Conclusions

MSCs from different sources play various biological roles, and different tumors react in different ways to MSCs. MSCs not only manipulate the stemness of tumor cells, but also change the expression of immune checkpoint molecules on tumor cells and have huge effects on immune cells. Compared with “naïve” MSCs, CA-MSCs exhibit several unique characteristics, including stronger pro-tumor effects; they are considered to be key regulators of tumor fate. Given the multifaceted role that MSCs play in the TME, they represent an untapped but promising target for improving cancer therapies. It is necessary to formulate targeted therapy plans according to individual conditions in the treatment of cancer patients.

Author contributions

LS: Funding acquisition, Writing – original draft. YY: Writing – review & editing.

Funding

The authors declare financial support was received for the research, authorship, and/or publication of this article. This work was supported by Natural Science Foundation of Jiangsu Province (BK20210136), National Natural Science Foundation of China (82203547) and China Postdoctoral Science Foundation (2022M721433).

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

1. Olmedo-Moreno L, Aguilera Y, Baliña-Sánchez C, Martín-Montalvo A, Capilla-González V. Heterogeneity of in vitro expanded mesenchymal stromal cells and strategies to improve their therapeutic actions. Pharmaceutics (2022) 14(5):1112. doi: 10.3390/pharmaceutics14051112

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Ishige I, Nagamura-Inoue T, Honda MJ, Harnprasopwat R, Kido M, Sugimoto M, et al. Comparison of mesenchymal stem cells derived from arterial, venous, and Wharton's jelly explants of human umbilical cord. Int J Hematol (2009) 90(2):261–9. doi: 10.1007/s12185-009-0377-3

PubMed Abstract | CrossRef Full Text | Google Scholar

3. 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) 9:12. doi: 10.1186/1478-811X-9-12

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Frisbie L, Buckanovich RJ, Coffman L. Carcinoma-associated mesenchymal stem/stromal cells: architects of the pro-tumorigenic tumor microenvironment. Stem Cells (2022) 40(8):705–15. doi: 10.1093/stmcls/sxac036

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Han X, Wang L, Li T, Zhang J, Zhang D, Li J, et al. Beyond blocking: engineering RNAi-mediated targeted immune checkpoint nanoblocker enables T-cell-independent cancer treatment. ACS Nano (2020) 14(12):17524–34. doi: 10.1021/acsnano.0c08022

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Heinrich S, Craig AJ, Ma L, Heinrich B, Greten TF, Wang XW. Understanding tumour cell heterogeneity and its implication for immunotherapy in liver cancer using single-cell analysis. J Hepatol (2021) 74(3):700–15. doi: 10.1016/j.jhep.2020.11.036

PubMed Abstract | CrossRef Full Text | Google Scholar

7. Restifo NP, Smyth MJ, Snyder A. Acquired resistance to immunotherapy and future challenges. Nat Rev Cancer (2016) 16(2):121–6. doi: 10.1038/nrc.2016.2

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Coffman LG, Pearson AT, Frisbie LG, Freeman Z, Christie E, Bowtell DD, et al. Ovarian carcinoma-associated mesenchymal stem cells arise from tissue-specific normal stroma. Stem Cells (2019) 37(2):257–69. doi: 10.1002/stem.2932

PubMed Abstract | CrossRef Full Text | Google Scholar

9. Chen Z, Xu P, Wang X, Li Y, Yang J, Xia Y, et al. MSC-NPRA loop drives fatty acid oxidation to promote stemness and chemoresistance of gastric cancer. Cancer Lett (2023) 565:216235. doi: 10.1016/j.canlet.2023.216235

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Goldstein RH, Reagan MR, Anderson K, Kaplan DL, Rosenblatt M. Human bone marrow-derived MSCs can home to orthotopic breast cancer tumors and promote bone metastasis. Cancer Res (2010) 70(24):10044–50. doi: 10.1158/0008-5472

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Zhu Y, Sun Z, Han Q, Liao L, Wang J, Bian C, et al. Human mesenchymal stem cells inhibit cancer cell proliferation by secreting DKK-1. Leukemia (2009) 23(5):925–33. doi: 10.1038/leu.2008.384

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Li W, Yang J, Zheng P, Li H, Zhao S. The origins and generation of cancer-associated mesenchymal stromal cells: an innovative therapeutic target for solid tumors. Front Oncol (2021) 11:723707. doi: 10.3389/fonc.2021.723707

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Pavon LF, Sibov TT, de Souza AV, da Cruz EF, Malheiros SMF, Cabral FR, et al. Tropism of mesenchymal stem cell toward CD133+ stem cell of glioblastoma in vitro and promote tumor proliferation in vivo. Stem Cell Res Ther (2018) 9(1):310. doi: 10.1186/s13287-018-1049-0

PubMed Abstract | CrossRef Full Text | Google Scholar

14. Belotti D, Capelli C, Resovi A, Introna M, Taraboletti G. Thrombospondin-1 promotes mesenchymal stromal cell functions via TGFβ and in cooperation with PDGF. Matrix Biol (2016) 55:106–16. doi: 10.1016/j.matbio.2016.03.003

PubMed Abstract | CrossRef Full Text | Google Scholar

15. Ishii M, Takahashi M, Murakami J, Yanagisawa T, Nishimura M. Vascular endothelial growth factor-C promotes human mesenchymal stem cell migration via an ERK-and FAK-dependent mechanism. Mol Cell Biochem (2019) 455(1-2):185–93. doi: 10.1007/s11010-018-3481-y

PubMed Abstract | CrossRef Full Text | Google Scholar

16. Barcellos-de-Souza P, Comito G, Pons-Segura C, Taddei ML, Gori V, Becherucci V, et al. Mesenchymal stem cells are recruited and activated into carcinoma-associated fibroblasts by prostate cancer microenvironment-derived TGF-β1. Stem Cells (2016) 34(10):2536–47. doi: 10.1002/stem.2412

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Berger L, Shamai Y, Skorecki KL, Tzukerman M. Tumor specific recruitment and reprogramming of mesenchymal stem cells in tumorigenesis. Stem Cells (2016) 34(4):1011–26. doi: 10.1002/stem.2269

PubMed Abstract | CrossRef Full Text | Google Scholar

18. Le Naour A, Prat M, Thibault B, Mével R, Lemaitre L, Leray H, et al. Tumor cells educate mesenchymal stromal cells to release chemoprotective and immunomodulatory factors. J Mol Cell Biol (2020) 12(3):202–15. doi: 10.1093/jmcb/mjz090

PubMed Abstract | CrossRef Full Text | Google Scholar

19. Wang M, Zhao X, Qiu R, Gong Z, Huang F, Yu W, et al. Lymph node metastasis-derived gastric cancer cells educate bone marrow-derived mesenchymal stem cells via YAP signaling activation by exosomal Wnt5a. Oncogene (2021) 40(12):2296–208. doi: 10.1038/s41388-021-01722-8

PubMed Abstract | CrossRef Full Text | Google Scholar

20. Shen Y, Xue C, Li X, Ba L, Gu J, Sun Z, et al. Effects of gastric cancer cell-derived exosomes on the immune regulation of mesenchymal stem cells by the NF-kB signaling pathway. Stem Cells Dev (2019) 28(7):464–76. doi: 10.1089/scd.2018.0125

PubMed Abstract | CrossRef Full Text | Google Scholar

21. Ren G, Zhao X, Wang Y, Zhang X, Chen X, Xu C, et al. CCR2-dependent recruitment of macrophages by tumor-educated mesenchymal stromal cells promotes tumor development and is mimicked by TNFα. Cell Stem Cell (2012) 11(6):812–24. doi: 10.1016/j.stem.2012.08.013

PubMed Abstract | CrossRef Full Text | Google Scholar

22. Zhang J, Ji C, Li W, Mao Z, Shi Y, Shi H, et al. Tumor-educated neutrophils activate mesenchymal stem cells to promote gastric cancer growth and metastasis. Front Cell Dev Biol (2020) 8:788. doi: 10.3389/fcell.2020.00788

PubMed Abstract | CrossRef Full Text | Google Scholar

23. Huang C, Chen B, Wang X, Xu J, Sun L, Wang D, et al. Gastric cancer mesenchymal stem cells via the CXCR2/HK2/PD-L1 pathway mediate immunosuppression. Gastric Cancer (2023) 26(5):691–707. doi: 10.1007/s10120-023-01405-1

PubMed Abstract | CrossRef Full Text | Google Scholar

24. Sun L, Wang Q, Chen B, Zhao Y, Shen B, Wang H, et al. Gastric cancer mesenchymal stem cells derived IL-8 induces PD-L1 expression in gastric cancer cells via STAT3/mTOR-c-Myc signal axis. Cell Death Dis (2018) 9(9):928. doi: 10.1038/s41419-018-0988-9

PubMed Abstract | CrossRef Full Text | Google Scholar

25. Wang M, Chen B, Sun XX, Zhao XD, Zhao YY, Sun L, et al. Gastric cancer tissue-derived mesenchymal stem cells impact peripheral blood mononuclear cells via disruption of Treg/Th17 balance to promote gastric cancer progression. Exp Cell Res (2017) 361(1):19–29. doi: 10.1016/j.yexcr.2017.09.036

PubMed Abstract | CrossRef Full Text | Google Scholar

26. Guo S, Huang C, Han F, Chen B, Ding Y, Zhao Y, et al. Gastric Cancer Mesenchymal Stem Cells Inhibit NK Cell Function through mTOR Signalling to Promote Tumour Growth. Stem Cells Int (2021) 2021:9989790. doi: 10.1155/2021/9989790

PubMed Abstract | CrossRef Full Text | Google Scholar

27. Zheng Z, Li YN, Jia S, Zhu M, Cao L, Tao M, et al. Lung mesenchymal stromal cells influenced by Th2 cytokines mobilize neutrophils and facilitate metastasis by producing complement C3. Nat Commun (2021) 12(1):6202. doi: 10.1038/s41467-021-26460-z

PubMed Abstract | CrossRef Full Text | Google Scholar

28. Yan C, Chang J, Song X, Qi Y, Ji Z, Liu T, et al. Lung cancer-associated mesenchymal stem cells promote tumor metastasis and tumorigenesis by induction of epithelial-mesenchymal transition and stem-like reprogram. Aging (Albany NY) (2021) 13(7):9780–800. doi: 10.18632/aging.202732

PubMed Abstract | CrossRef Full Text | Google Scholar

29. Gordon JAR, Evans MF, Ghule PN, Lee K, Vacek P, Sprague BL, et al. Identification of molecularly unique tumor-associated mesenchymal stromal cells in breast cancer patients. PLoS One (2023) 18(3):e0282473. doi: 10.1371/journal.pone.0282473

PubMed Abstract | CrossRef Full Text | Google Scholar

30. Yan XL, Fu CJ, Chen L, Qin JH, Zeng Q, Yuan HF, et al. Mesenchymal stem cells from primary breast cancer tissue promote cancer proliferation and enhance mammosphere formation partially via EGF/EGFR/Akt pathway. Breast Cancer Res Treat (2012) 132(1):153–64. doi: 10.1007/s10549-011-1577-0

PubMed Abstract | CrossRef Full Text | Google Scholar

31. Waghray M, Yalamanchili M, Dziubinski M, Zeinali M, Erkkinen M, Yang H, et al. GM-CSF mediates mesenchymal-epithelial cross-talk in pancreatic cancer. Cancer Discovery (2016) 6(8):886–99. doi: 10.1158/2159-8290.CD-15-0947

PubMed Abstract | CrossRef Full Text | Google Scholar

32. Ávila-Ibarra LR, Mora-García ML, García-Rocha R, Hernández-Montes J, Weiss-Steider B, Montesinos JJ, et al. Mesenchymal stromal cells derived from normal cervix and cervical cancer tumors increase CD73 expression in cervical cancer cells through TGF-β1 production. Stem Cells Dev (2019) 28(7):477–88. doi: 10.1089/scd.2018.0183

PubMed Abstract | CrossRef Full Text | Google Scholar

33. Zhang X, Hu F, Li G, Li G, Yang X, Liu L, et al. Human colorectal cancer-derived mesenchymal stem cells promote colorectal cancer progression through IL-6/JAK2/STAT3 signaling. Cell Death Dis (2018) 9(2):25. doi: 10.1038/s41419-017-0176-3

PubMed Abstract | CrossRef Full Text | Google Scholar

34. Liotta F, Querci V, Mannelli G, Santarlasci V, Maggi L, Capone M, et al. Mesenchymal stem cells are enriched in head neck squamous cell carcinoma, correlates with tumour size and inhibit T-cell proliferation. Br J Cancer (2015) 112(4):745–54. doi: 10.1038/bjc.2015.15

PubMed Abstract | CrossRef Full Text | Google Scholar

35. Di Matteo S, Avanzini MA, Pelizzo G, Calcaterra V, Croce S, Spaggiari GM, et al. Neuroblastoma tumor-associated mesenchymal stromal cells regulate the cytolytic functions of NK cells. Cancers (Basel) (2022) 15(1):19. doi: 10.3390/cancers15010019

PubMed Abstract | CrossRef Full Text | Google Scholar

36. Reagan MR, Ghobrial IM. Multiple myeloma mesenchymal stem cells: characterization, origin, and tumor-promoting effects. Clin Cancer Res (2012) 18(2):342–9. doi: 10.1158/1078-0432.CCR-11-2212

PubMed Abstract | CrossRef Full Text | Google Scholar

37. Corre J, Mahtouk K, Attal M, Gadelorge M, Huynh A, Fleury-Cappellesso S, et al. Bone marrow mesenchymal stem cells are abnormal in multiple myeloma. Leukemia (2007) 21(5):1079–88. doi: 10.1038/sj.leu.2404621

PubMed Abstract | CrossRef Full Text | Google Scholar

38. Arnulf B, Lecourt S, Soulier J, Ternaux B, Lacassagne MN, Crinquette A, et al. Phenotypic and functional characterization of bone marrow mesenchymal stem cells derived from patients with multiple myeloma. Leukemia (2007) 21(1):158–63. doi: 10.1038/sj.leu.2404466

PubMed Abstract | CrossRef Full Text | Google Scholar

39. Markovina S, Callander NS, O'Connor SL, Xu G, Shi Y, Leith CP, et al. Bone marrow stromal cells from multiple myeloma patients uniquely induce bortezomib resistant NF-kappaB activity in myeloma cells. Mol Cancer (2010) 9:176. doi: 10.1186/1476-4598-9-176

PubMed Abstract | CrossRef Full Text | Google Scholar

40. de Jong MME, Kellermayer Z, Papazian N, Tahri S, Hofste Op Bruinink D, Hoogenboezem R, et al. The multiple myeloma microenvironment is defined by an inflammatory stromal cell landscape. Nat Immunol (2021) 22(6):769–80. doi: 10.1038/s41590-021-00931-3

PubMed Abstract | CrossRef Full Text | Google Scholar

41. Atiya H, Frisbie L, Pressimone C, Coffman L. Mesenchymal stem cells in the tumor microenvironment. Adv Exp Med Biol (2020) 1234:31–42. doi: 10.1007/978-3-030-37184-5_3

PubMed Abstract | CrossRef Full Text | Google Scholar

42. Gu JJ, Hoj J, Rouse C, Pendergast AM. Mesenchymal stem cells promote metastasis through activation of an ABL-MMP9 signaling axis in lung cancer cells. PLoS One (2020) 15(10):e0241423. doi: 10.1371/journal.pone.0241423

PubMed Abstract | CrossRef Full Text | Google Scholar

43. Antoon R, Wang XH, Saleh AH, Warrington J, Hedley DW, Keating A. Pancreatic cancer growth promoted by bone marrow mesenchymal stromal cell-derived IL-6 is reversed predominantly by IL-6 blockade. Cytotherapy (2022) 24(7):699–710. doi: 10.1016/j.jcyt.2021.12.005

PubMed Abstract | CrossRef Full Text | Google Scholar

44. McGuire JJ, Frieling JS, Lo CH, Li T, Muhammad A, Lawrence HR, et al. Mesenchymal stem cell-derived interleukin-28 drives the selection of apoptosis resistant bone metastatic prostate cancer. Nat Commun (2021) 12(1):723. doi: 10.1038/s41467-021-20962-6

PubMed Abstract | CrossRef Full Text | Google Scholar

45. Buschhaus JM, Rajendran S, Humphries BA, Cutter AC, Muñiz AJ, Ciavattone NG, et al. Effects of iron modulation on mesenchymal stem cell-induced drug resistance in estrogen receptor-positive breast cancer. Oncogene (2022) 41(29):3705–18. doi: 10.1038/s41388-022-02385-9

PubMed Abstract | CrossRef Full Text | Google Scholar

46. Sandiford OA, Donnelly RJ, El-Far MH, Burgmeyer LM, Sinha G, Pamarthi SH, et al. Mesenchymal Stem Cell-Secreted Extracellular Vesicles Instruct Stepwise Dedifferentiation of Breast Cancer Cells into Dormancy at the Bone Marrow Perivascular Region. Cancer Res (2021) 81(6):1567–82. doi: 10.1158/0008-5472.CAN-20-2434

PubMed Abstract | CrossRef Full Text | Google Scholar

47. Cortini M, Massa A, Avnet S, Bonuccelli G, Baldini N. Tumor-activated mesenchymal stromal cells promote osteosarcoma stemness and migratory potential via IL-6 secretion. PLoS One (2016) 11(11):e0166500. doi: 10.1371/journal.pone.0166500

PubMed Abstract | CrossRef Full Text | Google Scholar

48. Li H, Li F. Exosomes from BM-MSCs increase the population of CSCs via transfer of miR-142-3p. Br J Cancer (2018) 119(6):744–55. doi: 10.1038/s41416-018-0254-z

PubMed Abstract | CrossRef Full Text | Google Scholar

49. Chang YW, Wang CC, Yin CF, Wu CH, Huang HC, Juan HF. Quantitative phosphoproteomics reveals ectopic ATP synthase on mesenchymal stem cells to promote tumor progression via ERK/c-Fos pathway activation. Mol Cell Proteomics (2022) 21(6):100237. doi: 10.1016/j.mcpro.2022.100237

PubMed Abstract | CrossRef Full Text | Google Scholar

50. Wang L, Ouyang M, Xing S, Zhao S, Liu S, Sun L, et al. Mesenchymal Stem Cells and their Derived Exosomes Promote Malignant Phenotype of Polyploid Non-Small-Cell Lung Cancer Cells through AMPK Signaling Pathway. Anal Cell Pathol (Amst) (2022) 2022:8708202. doi: 10.1155/2022/8708202

PubMed Abstract | CrossRef Full Text | Google Scholar

51. Ye H, Pan J, Gong E, Cai X, Xu C, Li Y, et al. Inhibitory effect of immunologically activated mesenchymal stem cells on lung cancer cell growth and metastasis. Cancer Biother Radiopharm (2023) 38(5):322–35. doi: 10.1089/cbr.2020.3855

PubMed Abstract | CrossRef Full Text | Google Scholar

52. Wang M, Zhang Y, Liu M, Jia Y, He J, Xu X, et al. Inhibition of STAT3 signaling as critical molecular event in HUC-MSCs suppressed Glioblastoma Cells. J Cancer (2023) 14(4):611–27. doi: 10.7150/jca.77905

PubMed Abstract | CrossRef Full Text | Google Scholar

53. ElBadre HM, El-Deek SEM, Ramadan HK, Elbadr MM, Sabry D, Ahmed NM, et al. Potential role of human umbilical cord stem cells-derived exosomes as novel molecular inhibitors of hepatocellular carcinoma growth. Apoptosis (2023) 28(9-10):1346–56. doi: 10.1007/s10495-023-01863-z

PubMed Abstract | CrossRef Full Text | Google Scholar

54. Chen J, Li Z, Yue C, Ma J, Cao L, Lin J, et al. Human umbilical cord mesenchymal stem cell-derived exosomes carrying miR-1827 downregulate SUCNR1 to inhibit macrophage M2 polarization and prevent colorectal liver metastasis. Apoptosis (2023) 28(3-4):549–65. doi: 10.1007/s10495-022-01798-x

PubMed Abstract | CrossRef Full Text | Google Scholar

55. Guo G, Tan Z, Liu Y, Shi F, She J. The therapeutic potential of stem cell-derived exosomes in the ulcerative colitis and colorectal cancer. Stem Cell Res Ther (2022) 13(1):138. doi: 10.1186/s13287-022-02811-5

PubMed Abstract | CrossRef Full Text | Google Scholar

56. Bagheri-Mohammadi S, Moradian-Tehrani R, Noureddini M, Alani B. Novel application of adipose-derived mesenchymal stem cells via producing antiangiogenic factor TSP-1 in lung metastatic melanoma animal model. Biologicals (2020) 68:9–18. doi: 10.1016/j.biologicals.2020.09.004

PubMed Abstract | CrossRef Full Text | Google Scholar

57. Lou G, Chen L, Xia C, Wang W, Qi J, Li A, et al. MiR-199a-modified exosomes from adipose tissue-derived mesenchymal stem cells improve hepatocellular carcinoma chemosensitivity through mTOR pathway. J Exp Clin Cancer Res (2020) 39(1):4. doi: 10.1186/s13046-019-1512-5

PubMed Abstract | CrossRef Full Text | Google Scholar

58. Xue C, Gao Y, Sun Z, Li X, Zhang M, Yang Y, et al. Mesenchymal stem cells derived from adipose tissue accelerate the progression of colon cancer by inducing a MTCAF phenotype via ICAM1/STAT3/AKT axis. Front Oncol (2022) 12:837781. doi: 10.3389/fonc.2022.837781

PubMed Abstract | CrossRef Full Text | Google Scholar

59. Sharaf K, Eggersmann TK, Haider SP, Schwenk-Zieger S, Zhou J, Gires O, et al. Human adipose-derived stem/stromal cells promote proliferation and migration in head and neck cancer cells. Cancers (Basel) (2021) 13(11):2751. doi: 10.3390/cancers13112751

PubMed Abstract | CrossRef Full Text | Google Scholar

60. Li T, Zhou X, Wang J, Liu Z, Han S, Wan L, et al. Adipose-derived mesenchymal stem cells and extracellular vesicles confer antitumor activity in preclinical treatment of breast cancer. Pharmacol Res (2020) 157:104843. doi: 10.1016/j.phrs.2020.104843

PubMed Abstract | CrossRef Full Text | Google Scholar

61. Aboulkheyr Es H, Bigdeli B, Zhand S, Aref AR, Thiery JP, Warkiani ME. Mesenchymal stem cells induce PD-L1 expression through the secretion of CCL5 in breast cancer cells. J Cell Physiol (2021) 236(5):3918–28. doi: 10.1002/jcp.30135

PubMed Abstract | CrossRef Full Text | Google Scholar

62. Gečys D, Skredėnienė R, Gečytė E, Kazlauskas A, Balnytė I, Jekabsone A. Adipose tissue-derived stem cell extracellular vesicles suppress glioblastoma proliferation, invasiveness and angiogenesis. Cells (2023) 12(9):1247. doi: 10.3390/cells12091247

PubMed Abstract | CrossRef Full Text | Google Scholar

63. Kapoor-Narula U, Lenka N. Cancer stem cells and tumor heterogeneity: Deciphering the role in tumor progression and metastasis. Cytokine (2022) 157:155968. doi: 10.1016/j.cyto.2022.155968

PubMed Abstract | CrossRef Full Text | Google Scholar

64. Li HJ, Reinhardt F, Herschman HR, Weinberg RA. Cancer-stimulated mesenchymal stem cells create a carcinoma stem cell niche via prostaglandin E2 signaling. Cancer Discovery (2012) 2(9):840–55. doi: 10.1158/2159-8290.CD-12-0101

PubMed Abstract | CrossRef Full Text | Google Scholar

65. Jiang C, Zhang Q, Shanti RM, Shi S, Chang TH, Carrasco L, et al. Mesenchymal stromal cell-derived interleukin-6 promotes epithelial-mesenchymal transition and acquisition of epithelial stem-like cell properties in ameloblastoma epithelial cells. Stem Cells (2017) 35(9):2083–94. doi: 10.1002/stem.2666

PubMed Abstract | CrossRef Full Text | Google Scholar

66. Wang KH, Chang YH, Harnod T, Ding DC. Endometrial cancer-infiltrating mesenchymal stem cells exhibit immunosuppressive effects. Cell Transplant (2022) 31:9636897221104452. doi: 10.1177/09636897221104452

PubMed Abstract | CrossRef Full Text | Google Scholar

67. Sun L, Huang C, Zhu M, Guo S, Gao Q, Wang Q, et al. Gastric cancer mesenchymal stem cells regulate PD-L1-CTCF enhancing cancer stem cell-like properties and tumorigenesis. Theranostics (2020) 10(26):11950–62. doi: 10.7150/thno.49717

PubMed Abstract | CrossRef Full Text | Google Scholar

68. Gao Y, Nihira NT, Bu X, Chu C, Zhang J, Kolodziejczyk A, et al. Acetylation-dependent regulation of PD-L1 nuclear translocation dictates the efficacy of anti-PD-1 immunotherapy. Nat Cell Biol (2020) 22(9):1064–75. doi: 10.1038/s41556-020-0562-4

PubMed Abstract | CrossRef Full Text | Google Scholar

69. Hsu JM, Xia W, Hsu YH, Chan LC, Yu WH, Cha JH, et al. STT3-dependent PD-L1 accumulation on cancer stem cells promotes immune evasion. Nat Commun (2018) 9(1):1908. doi: 10.1038/s41467-018-04313-6

PubMed Abstract | CrossRef Full Text | Google Scholar

70. Marigo I, Zilio S, Desantis G, Mlecnik B, Agnellini AHR, Ugel S, et al. T cell cancer therapy requires CD40-CD40L activation of tumor necrosis factor and inducible nitric-oxide-synthase-producing dendritic cells. Cancer Cell (2016) 30(3):377–90. doi: 10.1016/j.ccell.2016.08.004

PubMed Abstract | CrossRef Full Text | Google Scholar

71. Hadrup S, Donia M, Thor Straten P. Effector CD4 and CD8 T cells and their role in the tumor microenvironment. Cancer Microenviron (2013) 6(2):123–33. doi: 10.1007/s12307-012-0127-6

PubMed Abstract | CrossRef Full Text | Google Scholar

72. Kawasaki Y, Sato K, Mashima K, Nakano H, Ikeda T, Umino K, et al. Mesenchymal stromal cells inhibit aerobic glycolysis in activated T cells by negatively regulating hexokinase II activity through PD-1/PD-L1 interaction. Transplant Cell Ther (2021) 27(3):231. doi: 10.1016/j.jtct.2020.11.012

CrossRef Full Text | Google Scholar

73. Wang S, Wang G, Zhang L, Li F, Liu K, Wang Y, et al. Interleukin-17 promotes nitric oxide-dependent expression of PD-L1 in mesenchymal stem cells. Cell Biosci (2020) 10:73. doi: 10.1186/s13578-020-00431-1

PubMed Abstract | CrossRef Full Text | Google Scholar

74. Yan Z, Zhuansun Y, Liu G, Chen R, Li J, Ran P. Mesenchymal stem cells suppress T cells by inducing apoptosis and through PD-1/B7-H1 interactions. Immunol Lett (2014) 162(1 Pt A):248–55. doi: 10.1016/j.imlet.2014.09.013

PubMed Abstract | CrossRef Full Text | Google Scholar

75. Cahill EF, Sax T, Hartmann I, Haffner S, Holler E, Holler B, et al. Mesenchymal stromal cells protect endothelial cells from cytotoxic T lymphocyte-induced lysis. Scand J Immunol (2016) 84(3):158–64. doi: 10.1111/sji.12459

PubMed Abstract | CrossRef Full Text | Google Scholar

76. Yu Y, Valderrama AV, Han Z, Uzan G, Naserian S, Oberlin E. Human fetal liver MSCs are more effective than adult bone marrow MSCs for their immunosuppressive, immunomodulatory, and Foxp3 + T reg induction capacity. Stem Cell Res Ther (2021) 12(1):138. doi: 10.1186/s13287-021-02176-1

PubMed Abstract | CrossRef Full Text | Google Scholar

77. Krampera M, Glennie S, Dyson J, Scott D, Laylor R, Simpson E, et al. Bone marrow mesenchymal stem cells inhibit the response of naive and memory antigen-specific T cells to their cognate peptide. Blood (2003) 101(9):3722–9. doi: 10.1182/blood-2002-07-2104

PubMed Abstract | CrossRef Full Text | Google Scholar

78. Wu R, Fan X, Wang Y, Shen M, Zheng Y, Zhao S, et al. Mesenchymal stem cell-derived extracellular vesicles in liver immunity and therapy. Front Immunol (2022) 13:833878. doi: 10.3389/fimmu.2022.833878

PubMed Abstract | CrossRef Full Text | Google Scholar

79. Tang Y, Wu P, Li L, Xu W, Jiang J. Mesenchymal stem cells and their small extracellular vesicles as crucial immunological efficacy for hepatic diseases. Front Immunol (2022) 13:880523. doi: 10.3389/fimmu.2022.880523

PubMed Abstract | CrossRef Full Text | Google Scholar

80. Ge W, Jiang J, Arp J, Liu W, Garcia B, Wang H. Regulatory T-cell generation and kidney allograft tolerance induced by mesenchymal stem cells associated with indoleamine 2,3-dioxygenase expression. Transplantation (2010) 90(12):1312–20. doi: 10.1097/TP.0b013e3181fed001

PubMed Abstract | CrossRef Full Text | Google Scholar

81. Luz-Crawford P, Djouad F, Toupet K, Bony C, Franquesa M, Hoogduijn MJ, et al. Mesenchymal stem cell-derived interleukin 1 receptor antagonist promotes macrophage polarization and inhibits B cell differentiation. Stem Cells (2016) 34(2):483–92. doi: 10.1002/stem.2254

PubMed Abstract | CrossRef Full Text | Google Scholar

82. Tu C, Wang Z, Xiang E, Zhang Q, Zhang Y, Wu P, et al. Human umbilical cord mesenchymal stem cells promote macrophage PD-L1 expression and attenuate acute lung injury in mice. Curr Stem Cell Res Ther (2022) 17(6):564–75. doi: 10.2174/1574888X17666220127110332

PubMed Abstract | CrossRef Full Text | Google Scholar

83. Chen M, Peng J, Xie Q, Xiao N, Su X, Mei H, et al. Mesenchymal stem cells alleviate moderate-to-severe psoriasis by reducing the production of type I interferon (IFN-I) by plasmacytoid dendritic cells (pDCs). Stem Cells Int (2019) 2019:6961052. doi: 10.1155/2019/6961052

PubMed Abstract | CrossRef Full Text | Google Scholar

84. Park HH, Lee S, Yu Y, Yoo SM, Baek SY, Jung N, et al. TGF-β secreted by human umbilical cord blood-derived mesenchymal stem cells ameliorates atopic dermatitis by inhibiting secretion of TNF-α and IgE. Stem Cells (2020) 38(7):904–16. doi: 10.1002/stem.3183

PubMed Abstract | CrossRef Full Text | Google Scholar

85. Che N, Li X, Zhou S, Liu R, Shi D, Lu L, et al. Umbilical cord mesenchymal stem cells suppress B-cell proliferation and differentiation. Cell Immunol (2012) 274(1-2):46–53. doi: 10.1016/j.cellimm.2012.02.004

PubMed Abstract | CrossRef Full Text | Google Scholar

86. Tang RJ, Shen SN, Zhao XY, Nie YZ, Xu YJ, Ren J, et al. Mesenchymal stem cells-regulated Treg cells suppress colitis-associated colorectal cancer. Stem Cell Res Ther (2015) 6(1):71. doi: 10.1186/s13287-015-0055-8

PubMed Abstract | CrossRef Full Text | Google Scholar

87. Fu Y, Li J, Li M, Xu J, Rong Z, Ren F, et al. Umbilical cord mesenchymal stem cells ameliorate inflammation-related tumorigenesis via modulating macrophages. Stem Cells Int (2022) 2022:1617229. doi: 10.1155/2022/1617229

PubMed Abstract | CrossRef Full Text | Google Scholar

88. Park YS, Lim GW, Cho KA, Woo SY, Shin M, Yoo ES, et al. Improved viability and activity of neutrophils differentiated from HL-60 cells by co-culture with adipose tissue-derived mesenchymal stem cells. Biochem Biophys Res Commun (2012) 423(1):19–25. doi: 10.1016/j.bbrc.2012.05.049

PubMed Abstract | CrossRef Full Text | Google Scholar

89. Shahir M, Mahmoud Hashemi S, Asadirad A, Varahram M, Kazempour-Dizaji M, Folkerts G, et al. Effect of mesenchymal stem cell-derived exosomes on the induction of mouse tolerogenic dendritic cells. J Cell Physiol (2020) 235(10):7043–55. doi: 10.1002/jcp.29601

PubMed Abstract | CrossRef Full Text | Google Scholar

90. Kim J, Escalante LE, Dollar BA, Hanson SE, Hematti P. Comparison of breast and abdominal adipose tissue mesenchymal stromal/stem cells in support of proliferation of breast cancer cells. Cancer Invest (2013) 31(8):550–4. doi: 10.3109/07357907.2013.830737

PubMed Abstract | CrossRef Full Text | Google Scholar

91. Lo Sicco C, Reverberi D, Balbi C, Ulivi V, Principi E, Pascucci L, et al. Mesenchymal stem cell-derived extracellular vesicles as mediators of anti-inflammatory effects: endorsement of macrophage polarization. Stem Cells Transl Med (2017) 6(3):1018–28. doi: 10.1002/sctm.16-0363

PubMed Abstract | CrossRef Full Text | Google Scholar

92. Fakhimi M, Talei AR, Ghaderi A, Habibagahi M, Razmkhah M. Helios, CD73 and CD39 induction in regulatory T cells exposed to adipose derived mesenchymal stem cells. Cell J (2020) 22(2):236–44. doi: 10.22074/cellj.2020.6313

PubMed Abstract | CrossRef Full Text | Google Scholar

93. Qiu W, Guo Q, Guo X, Wang C, Li B, Qi Y, et al. Mesenchymal stem cells, as glioma exosomal immunosuppressive signal multipliers, enhance MDSCs immunosuppressive activity through the miR-21/SP1/DNMT1 positive feedback loop. J Nanobiotechnology (2023) 21(1):233. doi: 10.1186/s12951-023-01997-x

PubMed Abstract | CrossRef Full Text | Google Scholar

94. Li W, Zhao SL, Zheng P, Shi PQ, Zhou Y, Zhang T, et al. [Gastric cancer-derived mesenchymal stem cells regulate the M2 polarization of macrophages within gastric cancer microenvironment via JAK2/STAT3 signaling pathway]. Zhonghua Zhong Liu Za Zhi (2022) 44(7):728–36. doi: 10.3760/cma.j.cn112152-20200106-00008

PubMed Abstract | CrossRef Full Text | Google Scholar

95. Han F, Guo S, Huang C, Cui L, Zhao Y, Ma J, et al. Gastric cancer mesenchymal stem cells inhibit natural killer cell function by up-regulating FBP1. Cent Eur J Immunol (2021) 46(4):427–37. doi: 10.5114/ceji.2021.111753

PubMed Abstract | CrossRef Full Text | Google Scholar

96. Galland S, Vuille J, Martin P, Letovanec I, Caignard A, Fregni G, et al. Tumor-derived mesenchymal stem cells use distinct mechanisms to block the activity of natural killer cell subsets. Cell Rep (2017) 20(12):2891–905. doi: 10.1016/j.celrep.2017.08.089

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: tumor microenvironment, heterogeneity, cancer-associated mesenchymal stem/stromal cells, stemness, immune cells

Citation: Sun L and Yao Y (2023) Mesenchymal stem/stromal cells- a principal element for tumour microenvironment heterogeneity. Front. Immunol. 14:1274379. doi: 10.3389/fimmu.2023.1274379

Received: 09 August 2023; Accepted: 28 September 2023;
Published: 09 October 2023.

Edited by:

Juan Francisco Santibanez, University of Belgrade, Serbia

Reviewed by:

Abderrahim Naji, Kōchi University, Japan

Copyright © 2023 Sun and Yao. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Yongliang Yao, YYLGD313@126.com

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.