Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2022 Dec 1.
Published in final edited form as: Biomed Pharmacother. 2021 Nov 5;144:112269. doi: 10.1016/j.biopha.2021.112269

Roles of mesenchymal stromal cells in the head and neck cancer microenvironment

Marcelo Coutinho de Miranda a, Mariane Izabella Abreu de Melo a, Pricila da Silva Cunha a, Jovino Gentilini Júnior a, Jerusa Araújo Quintão Arantes Faria b, Michele Angela Rodrigues c, Dawidson Assis Gomes a
PMCID: PMC8630686  NIHMSID: NIHMS1755128  PMID: 34794230

Abstract

Head and neck cancer (HNC), a common malignancy worldwide, is associated with high morbidity and mortality rates. Squamous cell carcinoma is the most common HNC type, followed by salivary gland carcinomas, head and neck sarcomas, and lymphomas. The microenvironment of HNCs comprises various cells that regulate tumor development. Recent studies have reported that the tumor microenvironment, which modulates cancer progression, regulates cancer treatment response. However, the presence of different types of stromal cells in cancers is a major challenge to elucidate the role of individual cells in tumor progression. The role of mesenchymal stromal cells (MSCs), which are a component of the tumor microenvironment, in HNC is unclear. The major impediment for characterizing the role of MSCs in cancer progression is the lack of MSC-specific markers and their phenotypic similarity with stromal cells. This review aimed to summarize the latest findings on the role of MSCs in the progression of HNC to improve our understanding of HNC pathophysiology.

Keywords: Cancer microenvironment, mesenchymal stromal cells, head and neck cancer, Squamous cell carcinoma, cancer metastasis

Graphical Abstract

graphic file with name nihms-1755128-f0001.jpg

1. Introduction

Head and neck cancers (HNCs) are a common type of malignancy associated with high mortality and morbidity rates [15]. At this anatomical site, squamous cell carcinoma (SCC) is the most common neoplasia in adults and accounts for 3% of all cancers and 1.5% of all cancer-related deaths in the United States of America [1,57]. SCC, which is typically observed in tobacco and alcohol users, is associated with a poor prognosis [1,8]. Lately, human papillomavirus (HPV) infection has increased the oropharyngeal SCC incidence in middle-aged patients with different progression and treatment responses, and better prognosis [9]. Lymph node-regional metastasis in head and neck SCC (HNSCC) decreases the overall survival rate by approximately 40%–50% [10]. The elucidation of various signaling pathways involved in the induction of metastasis can aid in mitigating the poor prognosis of patients with HNSCC. Previous studies have reported that the tumor microenvironment has a critical role in the metastasis of cancer cells [1115].

The tumor microenvironment is the ecosystem that surrounds and includes the tumor; it comprises the extracellular matrix (ECM), blood and lymph vessels, immune cells, fibroblasts/myofibroblasts, endothelial cells, pericytes, multipotent cells, and signaling molecules [16,17]. Recent studies have demonstrated that the modulation of the tumor microenvironment is a promising therapeutic strategy for cancer [14,18,19]. However, the role of many of the tumor microenvironment components in tumorigenesis is unclear. In particular, mesenchymal stromal cells (MSCs) are reported to exhibit dual roles in tumor progression [20,21].

MSCs, which are multipotent, mobile cells, are involved in tissue homeostasis, tissue/organ repair, and immune modulation [2225]. Inflammatory signals promote the migration of MSCs to the lesion site, where they secrete signaling molecules and differentiate into various cell types. Chronic inflammatory sites, such as tumors, promote the homing of MSCs, and these cells can be recovered from different cancer tissues [20,26]. These cells can be recognized in vitro based on several markers and their capacity to differentiate into different cell types [27]. However, these markers are not MSC-specific, which is a major impediment to examine MSC behavior in situ. This review aimed to summarize the role of the MSCs in the progression of HNCs, especially SCC.

2. MSCs identification

First identified in the bone marrow, the mesenchymal stem cells have the ability to differentiate into muscular, bone, and cartilage tissue in vivo [28,29]. Later, cells with similar capacities were recovered from different tissues; however, the true transdifferentiating capacity of these cells is still controversial [24]. Hence, the International Society for Cellular Therapy proposed that mesenchymal stroma cells are the best terminology for these cells. The term mesenchymal stem cells only should be used to indicate cells satisfying specific stemness criteria [30].

BM and adipose tissue stromal vascular fraction are the most common tissue sources for human MSC extraction [22,31,32]. BM-derived MSCs (BMMSCs) and adipose-derived MSCs (AMSCs) share several characteristics, such as the ability to divide symmetrically (generating two stem cells or two differentiated cells) or asymmetrically (generating one stem cell and one differentiated cell), differentiate into some tissue lineages, and migrate [23,27]. Stem cell populations are heterogeneous and often behave as individual cells.

MSCs are identified based on the expression of a subset of surface protein markers in vitro and the ability to adhere in culture, form colonies, and differentiate into at least three cell lineages after stimulation [22,27,33]. These features of MSCs are analyzed in vitro using a population of cells. Currently available markers cannot delineate the physiological roles of the MSCs in a specific tissue or disease, making the identification of MSCs in loco challenging. MSCs derived from different tissues are more closely related than those derived from other mesodermal lineages [34,35]. Crisan et al. (2008) reported that the morphology, proliferation kinetics, differentiation capacity, and surface markers of MSCs are similar to those of BMMSCs [36]. However, the phenotypic signatures of MSCs are similar to those of other cell lines, such as perivascular cells and fibroblasts [37,38].

Previous studies have reported that the surface markers of fibroblasts and microvascular pericytes are similar to those of MSCs, which further hinders the identification of MSCs [3941]. MSC-specific markers were observed in perivascular spaces around large arteries and veins, where fibroblast-like cells were identified in the outermost tunica adventitia (Figure 1) [42]. The proximity to the vasculature enables the MSCs to respond rapidly to injury or disease. Additionally, MSCs may have a distinct role in the tumor maintenance phase. The major challenge to elucidate the role of MSCs during the natural course of a disease is the lack of specific markers for differentiating different mesenchymal cell types.

Figure 1: Mesenchymal stromal cell (MSC) distribution.

Figure 1:

The major sources of MSCs are the bone marrow, adipose tissue, and dental pulp, although they can be obtained from different organs. MSCs are postulated to be localized in proximity to the blood vessels, such as permeating arterioles and inside the tunica adventitia in arteries. Stimulatory signals promote the migration of MSCs to the injured/inflamed site and regulate the host response or promote MSC differentiation.

Among the several markers proposed for MSC identification, some are strategic markers. For example, smooth muscle actin (SMA) can differentiate MSCs from activated fibroblast/myofibroblast [43,44]. However, the physiological levels of SMA have not been reported and SMA may exhibit varying protein and mRNA levels in MSCs in vitro [45]. It was reported that the expression of SMA in non-activated dermal fibroblasts was higher than that in matched MSCs (log2 (fold change) = 2.036, adjusted p-value = 1.1487 × 10−4; RNAseq) [38]. The upregulated expression of SMA promotes the differentiation of MSCs into myofibroblasts and cancer-activated fibroblasts (CAFs), which are involved in the development of various pathologies [43,46,47]. The co-expression of fibroblast activation protein alpha and fibroblast specific protein 1 (FSP, also known as S100A4) has been successfully used to differentiate fibroblasts from mesenchymal cells [48,49]. RNAseq analysis of MSCs and dermal fibroblasts revealed that the expression of S100A4 was downregulated in MSCs (log2 (fold change) = −2.808, adjusted p-value = 4.92 × 10−6) [38]. Gremlin-1 (GREM1) is also reported to be a candidate marker of MSCs [50,51]. RNAseq analysis revealed that the expression of GREM1 was not markedly different between MSCs and dermal fibroblasts (log2 (fold change) = 0.644, adjusted p-value = 0.5182) [38]. However, a refined analysis must be performed to exclude post-translation modifications of GREM1. Several studies have also proposed various markers, such as Meflin (ISLR), PDPN (podoplanin), STRO-1, and stage-specific embryonic antigen 4 (SSEA-4) [5155]. However, these markers are associated with some limitations and may be expressed in other cell lineages. The limitations and reliability of MSC markers have been reviewed by Lv et al. [55].

3. Physiological role of MSCs

In addition to tissue repair, MSCs are involved in various biological processes, including tissue homeostasis regulation and maintenance of blood vessel integrity [23,36]. Tissue injury induces the immune cells, endothelial cells, and fibroblasts to secrete several chemokines and mediators, which promote the mobilization of MSCs. These MSCs can undergo differentiation in several stromal components. Additionally, MSCs can produce factors that stimulate innate tissue repair and modulate the immune response and inflammation.

MSCs can be detected at the injured tissue site (virtually in every blood vessel) or mobilized from the BM through a poorly understood mechanism [23,56]. Endothelial cell selectins are reported to promote MSC homing [57,58]. CD44 and CD24 expressed on the MSC surface can interact with yet unknown endothelial selectins and initiate the rolling of the MSCs after chemotaxis [59,60]. The identification of vascular selectin is an active area of research. P-selectin (CD24/CD44 binding) and Galectin-1 (CD44 binding) are reported to promote MSC-endothelial cell interaction [58,61,62]. Integrins, such as α4β1 are activated after chemotaxis and bind to VCAM-1 on endothelial cells, which results in MSC arrest [63,64]. The release of matrix metalloproteinases (MMPs) promotes the transmigration of MSCs into the fenestrated endothelium of the stromal tissue [64]. The inflammatory cytokines released from the injured tissue can upregulate MMP expression in MSCs and consequently promote cell migration. The knockdown of MMP2, MMP14 (MT-MMP), and TIMP2 inhibits the migration of MSCs through the interstitium [65]. Mediators, such as interleukin-8 (IL-8), platelet-derived growth factor-AB (PGDF-AB), insulin-like growth factor (IGF)-1, and vascular endothelial growth factor (VEGF) can promote the migration of MSCs into the stroma [6668].

At the lesion site, the MSCs and surrounding cells modulate each other. For example, interferon-gamma (INF-γ), which enhances macrophage activation and M1 polarization, neutrophil mobility, and CD8+ T cell and natural killer (NK) cell activity, promotes the defense response against infection and tumors [69]. INF-γ-stimulated MSCs exert immunosuppressive effects and induce the expression of indoleamine 2,3-dioxygenase (IDO1), which results in the mitigation of inflammation and the stimulation of anti-inflammatory proteins [7072]. Moreover, the co-culture of MSCs with lymphocytes inhibits T cell proliferation without apoptosis induction [73]. Additionally, MSCs shift the activity of mature immune cells toward an anti-inflammatory state, which results in the augmentation of T regulatory (Treg) cells, T helper-2 (Th2) cells, and type 2 dendritic cells (DC2) polarization. Furthermore, MSCs decrease the number of Th1 and NK cells, promote M2 polarization of macrophages, and inhibit the secretion of immunoglobulin from plasma cells [7482].

4. Dual roles of MSCs in the HNC microenvironment

The solid tumor is a complex system comprising heterogeneous transformed cells, stromal cells, immune cells, blood vessels, and lymph vessels. The variability of individual tumors can be attributed to their non-structured, variable, and pathological microenvironment, which is responsible for poor treatment response and cancer progression [17,19,8386]. Each cell can communicate with the microenvironment components through cytokines and mediators in a paracrine fashion or cell-cell interaction [87]. Cytokines and mediators confer plasticity, variability, and uniqueness to the microenvironment. Recent studies have shown that the tumor microenvironment content is a part of the repertoire for cancer treatment [88].

MSCs associated with the tumor exert dual effects on tumor progression in different models [20,89,90]. The role of MSCs in disease progression has not been completely elucidated. MSCs can contribute to HNC formation through distinct mechanisms. For example, MSCs can exert their activity by secreting biomolecules, promoting cell-cell contact, suppressing protective activities of immune cells, enhancing angiogenesis, or undergoing differentiation in other tumor stroma components, such as CAF.

5. Effect of the crosstalk between MSCs and HNC cells: Paracrine signaling, cell-cell interaction, and immunomodulation

Various studies have characterized the biomolecules secreted from MSCs and cancer cells, which are termed secretome [60,91,92]. The crosstalk between cancer cells and MSCs can modify the secretion of biomolecules involved in altering cell behavior (Table 1) [68,9396]. Watts et al. (2016) characterized the secretion profile of HNSCC (JHU-011, JHU-012, and JHU-019 cells), which included stromal cell-derived factor 1 (SDF-1 or CXCL-12), VEGF, growth-regulated alpha protein (Gro-α or CXCL1), IL-8, IL-6, and platelet-derived growth factors (PDGF). The exposure of BMMSCs to the HNSCC-derived secretory molecules increased the migration and invasion of the MSCs toward cancer cells. Conversely, the inhibition of IL-6 and PDGF receptor alpha decreased the migration and invasion of MSCs toward the cancer cells [93]. These results indicate that cancer cell-secreted IL-6 and PDGF in the tumor microenvironment are sufficient to induce MSC migration into the head and neck tumor stroma. Kansy et al. (2014) reported the presence of IL-1β, IL-2, IL-4, IL-6, IL-8, granulocyte-macrophage colony-stimulating factor (GM-CSF), granulocyte colony-stimulating factor (G-CSF), INF-γ, macrophage inflammatory protein 1β (MIP1β or CCL4), SDF-1, and tumor necrosis factor-alpha (TNF-α) in the secretome of MSCs derived from primary HNSCC [97]. Ji et al. (2016) reported a similar secretome profile of MSCs derived from gingival tissue upon supplementation of IL-10, a potent anti-inflammatory cytokine [98,99].

Table 1:

Effects of the secretome on head and neck cancer.

Secretome origin Molecule Phenotype Target cell
BMMSC IL-6

B2M

CCN2

FGF19

EV

POSTN

CCL5

Not identified
Proliferation

Invasion / Migration

Invasion / Migration / ↑MMP9 / ↑MMP2 / EMP

Proliferation

Migration

EMP

Proliferation

Death resistance

↑MMP9 / ↑MMP2 / ColA1

Decreased proliferation / decreased invasion
FaDu/HLaC78

TE1/Eca109

TSCCa/ Cal27

CNE2/5-8F/6-10B

CNE1/CNE2/5-8F

CNE2

Cal27

Cal27/HN4

HSC3

Eca109
Gingival-MSC SCGF-β

Not identified
Death resistance

Decreased proliferation / Cell death
ISCCI24

NH6/Cal27
AMSC TGF-β ↑MMP14 PCI13
HNSCC

(JHU-011/-012/-019)
IL-6

PDGF
Invasion / Migration

Invasion / Migration
BMMSC
ACC TGF-β / SDF-1 Migration BMMSC

BMMSC – bone marrow mesenchymal stromal cell; PDGF – plated-derived growth factor; B2M – beta 2 microglobulin; CCN2 – cellular communication network factor 2; FGF19 – fibroblast growth factor 19; EV – extracellular vesicle; POSTN – periostin; SCGF-β – stromal cell growth factor-beta; TGF-β – transforming growth factor-beta; SDF-1 – stromal cell-derived factor 1; MMP – matrix metalloproteinases; EMP – epithelial-mesenchymal plasticity; MSC – mesenchymal stromal cell; AMSC – adipose-derived mesenchymal stromal cell; ACC – acinic cell carcinoma-derived cell.

Wang et al. (2018) identified beta-2-microglobulin (B2M) in the secretome of BMMSCs. The knockdown of B2M-encoding gene on BMMSCs inhibited the invasion and migration of HNSCC cells (TE1 and Eca109 cells) exposed to the MSC secretome [100]. Similarly, the exposure of HNSCC cells (TSCCa and Cal-27 cells) to the MSC secretome upregulated the levels of cellular communication network factor 2 (CCN2) in BMMSCs. CCN2 knockdown in MSCs inhibited cancer cell migration and invasion and downregulated the levels of MMP9, MMP2, and epithelial-mesenchymal plasticity markers. The expression of CCN2 in HNSCC tissues was higher than that in the adjacent non-tumorous tissues [101]. The inhibition of IL-6 in the BMMSC secretome decreased HNSCC cell (FaDu and HLaC78 cells) proliferation, which was partly dependent on the MAPK (ERK) signaling pathway [96].

AMSCs secrete high levels of VEGF and tumor growth factor-beta (TGF-β) [102]. TGF-β promoted the invasion of HNSCC cells by upregulating the expression of MMP14 [103]. Previous studies have reported that TGF-β and SDF-1 induce BMMSC migration toward acinic cell carcinoma cell lineage [104].

Coccè et al. (2017) reported that paclitaxel or gemcitabine promoted the secretion of stem growth factor-beta (SCGF-β) from MSCs derived from gingival tissue, which protects cancer cells (SCC154 cells) against these anti-neoplastic drugs in vitro [105]. In vitro approaches related to the MSC effects on HNC are summarized in table 2.

Table 2:

In vitro role of MSC on HNC.

Observed effect Effector cells Publication
Migration and invasion of BMMSC JHU-011/012 derived IL-6 [93]
Migration and invasion of TE-1 and Eca109 HNSCC BMMSC derived beta-2-microglobulin [100]
Proliferation of FaDu and HlaC78 cells BMMSC derived IL-6 [96]
BMMSC migration Acinic-cell carcinoma derived TGF-β and SDF-1 [104]
Anti-apoptotic effect on SCC154 cells SCGF-β secreted by gingival derived MSC exposed to paclitaxel or gemcitabine [105]
Anti-apoptotic effect on JHU-012 cells BMMSC cultured with cisplatin [106]
Anti-apoptotic effect on Eca109 and TE-1 BMMSC cultured with carboplatin [100]
Cell proliferation, EMP, and paclitaxel resistance (SCC25) BMMSC [107]
Proliferation, survival, and EMP of HN4 and Cal27 cells BMMSC derived POSTN [108]
Increased expresson of MMP9, MMP2, and collagen (1A2, 6A3, 1A1, 3A1) BMMSC derived CCL5 [95]
EMP on ameloblastoma cells IL-6, IL-8, angiogenin from MSC isolated from ameloblastoma [109]
Migration and invasion of SSC15 cells miR-8485 on EV-Leukoplakia-MSC and EV-HNSCC [110]
Proliferation (CNE2, 5-8F, 6-10B), migration (CNE1, CNE2, 5-8F) and EMP (CNE2) FGF-19 on EV-BMMSC [111]
Inhibition of proliferation, induction of cell death of Cal27 and HN6 cells Gengival derived-MSC [98]
Inhibition of proliferation and cell death of Eca109 cells BMMSC [112]
Proliferation, migration, and invasion (Eca109, TE-1)) MSC derived GREM-1 [113]
Invasion of TE-9 cells AMSC [114]
Proliferation of TPC1 cells BMMSC [115]
Increased expression of DUSP6, cell death Umbilical cord MSC-HNSCC fusion [116]
Reduction on TCD4+ and TCD8+ cells; reduction of INF-γ and TNF-α expression BMMSC-IDO [117]
Reduction on T cell proliferation HNSCC-MSC-IDO and IL4I1 [118]
Reduction on T cell (CD3+) cell proliferation MSC [119]
Reduction on CD73 expression, reduction on T cell proliferation HNSCC derived MSC [120]

BMMSC – Bone marrow mesenchymal stromal cell; AMSC – Adipose tissue derived-mesenchymal stromal cell; TGF-β – Transforming growth factor-beta; SDF-1 – Stromal cell-derived factor 1; SCGF-β – Stromal cell growth factor-beta; EMP – Epithelial-mesenchymal plasticity; POSTN – Periostin; MMP – Matrix metalloproteinases; EV – Extracellular vesicles; FGF-19 – Fibroblast growth factor 19; GREM-1 – Gremlin 1; DUSP6 – Dual Specificity Phosphatase 6; IDO – Indoleamine 2,3-dioxygenase; IL4I1 – Interleukin 4 Induced 1.

Wang et al. demonstrated that co-culturing JHU-012 cells with BMMSCs promoted cisplatin resistance and upregulated the expression of BCL-2, an anti-apoptotic protein [106]. BMMSCs also confers paclitaxel resistance in SSC25 cells with upregulation of BCL-2, periostin (POSTN), multidrug resistance-associated protein 1, and ABCG transporter [107]. Conversely, treatment with the secretome of doxorubicin-treated MSCs increased tumor cell death [105]. Additionally, the BMMSC secretome protects HNSCC against the growth-inhibitory effects of carboplatin [100]. These findings indicate that the stromal cells regulate the treatment response in patients with cancer.

Moreover, POSTN secreted by BMMSCs induced Cal27 cell proliferation and inhibited the death of HN4 and Cal27 cells through PI3K signaling and upregulation of epithelial-mesenchymal plasticity markers [108]. CCL5 secreted from BMMSCs upregulated the expression of MMP9, MMP2, and collagen (col1A2, col1A1, col6A3, and col3A1). The upregulated collagen expression (desmoplasia) in the tumor front is correlated with decreased patient survival [95]. Compared with those derived from the BMMSCs and oral mucosa-derived MSCs, the levels of MMP9 were higher and the levels of collagen IV were lower in MSCs obtained from the tissues of patients with leukoplakia comprising microscopically visible dysplasia or hyperplasia [121].

In addition to the SCC stroma, MSCs have been detected elsewhere. Jiang et al (2017) isolated MSC-like cells from follicular ameloblastoma, an aggressive odontogenic tumor with high recurrence rates [4,109,122124]. The secretome of ameloblastoma-derived MSCs comprised increased levels of IL-6, IL-8, and angiogenin, which contribute to mesenchymal-epithelial plasticity in ameloblastoma cells [109]. The cells can secrete biomolecules directly or through vesicles.

Extracellular vesicles (EVs) facilitate cell-cell communication and transport biomolecules, such as proteins, micro RNAs (miRNAs), long non-coding RNAs (lncRNAs), and messenger RNAs (mRNAs) [125]. EVs of MSC (MSC-EV) derived from a premalignant lesion and oral squamous cell carcinoma increased SCC15 cell migration and invasion by upregulating the levels of miR-8485. Although the miR-8485 interaction sites were not identified, transfection with the miR-8485 mimic restored the aggressive phenotype in the SCC cell line. This indicated that miR-8485 is involved in the carcinogenesis of HNC. The expression of miR-8485 was upregulated in both leukoplakia and carcinoma lesions [110]. As a limited number of these premalignant lesions progress to carcinoma, it would be interesting to understand if this microRNA correlates with malignization, validating its expression as an HNSCC progression marker. BMMSC-EV promoted pharyngeal SCC cell proliferation (CNE2, 5–8F, and 6–10B cells), migration (CNE1, CNE2, and 5–8F cells), and epithelial-mesenchymal plasticity (CNE2 cells) [111]. Fibroblast growth factor-19 (FGF-19) encapsulated within MSC-EV promotes cell proliferation by activating the ERK signaling pathway through FGF receptor 4 (FGFR4). However, MSC-EVs differentially affect the pharyngeal cell lineages, which indicates the diverse response of different cancers to the microenvironment.

In contrast, Ji et al. (2016) demonstrated that the MSC secretome decreased oral cancer cell proliferation and increased cell death (Cal27 and HN6 cells) [98]. Additionally, the MSC secretome downregulated the levels of Cyclin D1, phosphorylated STAT3, and ERK and upregulated the phosphorylation of JNK. Similar to treatment with the MSC secretome, treatment with the JNK signaling inhibitor promoted cell proliferation. However, the components of the MSC secretome slightly increased cell proliferation or did not consistently increase the proliferation of the cell lines. This can be attributed to the complex availability of each component in the conditioned media and the metabolic profiles of different cell lines. Li et al. (2011) reported similar results using BMMSC secretome, which decreased cell proliferation and invasion (Eca109 cells) and increased cell death. The downregulated expression of Cyclin E, phosphorylated Rb, and MMP2 was consistent with the phenotypic characteristics [112]. These findings suggest that both MSCs and HNC secretome comprise modulators that promote stromal MSC enrichment and modulate the proliferation and migration/invasion of cancer cells.

During migration, cancer cells are in close contact with stromal components. This cell-cell interaction can induce epithelial-mesenchymal plasticity, cell proliferation, and tumor invasion [105,126128]. Hong et al. (2018) reported that the expression of GREM1 was upregulated in esophageal SCC [126]. GREM1 knockdown in MSCs downregulated the expression of epithelial-mesenchymal plasticity markers, TGF-β, and activated Smad-2 and Smad-3 and the proliferation and invasion of Eca109 and TE-1 cells and upregulated the levels of bone morphogenic protein-4 (BMP4), phosphorylated Smad-1, Smad-5, and Smad-8. The inhibition of TGF-β mimics the GREM1 knockdown phenotype. The authors suggest that MSC-GREM1 inhibits BMP4 that can activate Smad-1, Smad-5, and Smad-8 and consequently inhibit the protective effect of this pathway. The paracrine stimulation of TGF-β released by MSCs and cell contact-dependent pathway can increase cancer cell aggressive behavior. Additionally, direct contact between AMSCs and SCC cells (TE-9 cells) increased cancer invasion into the gel-collagen matrix and upregulated the expression of MMP9 and laminin 5 [114]. The invasive SSC tissues exhibited upregulated expression of laminin 5 (gamma-2 chain), co-localized with stromal vimentin-positive cells, and promoted the expression of MMP2 and MMP14 on cancer cells in contact with these mesenchymal cells [126]. The enhanced activation of IGF receptor beta followed by phosphorylation of ERK and AKT signaling pathway could explain the observed phenotype. Unpublished preliminary data from our group indicated that the contact between AMSCs and SCC (HN12 cells) promoted MMP9 bidirectional expression and the invasion of cancer cells into the collagen/Matrigel matrix, which was partly dependent on integrin alpha 6. Moreover, MSCs increased thyroid cancer cell (TPC1 cells) proliferation through the epidermal growth factor receptor (EGFR), ERK, c-Jun, and p38 MAPK pathways [115]. However, Wang et al. (2012) reported that MSCs and HNSCC can fuse, which leads to the upregulation of dual-specificity phosphatase 6 (DUSP6). The upregulated expression of DUSP6 promotes cell death in these fused cells and consequently regulates cancer growth [116].

MSC-mediated immunomodulation is an active area of research. Previous studies have reported that MSC-mediated immunomodulation supports cancer growth [26,85,90,131]. HNSCC-derived MSCs decreased the proliferation of CD4+ and CD8+ T cells and downregulated the expression of INFγ and TNF-α [117]. The inhibition of IDO1 activity suppressed the HNSCC-derived MSC-mediated T cell proliferation arrest, which indicated that IDO1 of MSCs is involved in immunosuppression in HNC. Similarly, Mazzoni et al. (2021) reported that HNSCC-MSCs decreased T cell proliferation, which could be restored by IDO1 inhibition [118]. The inhibition of IL-4 induced gene 1 (IL4I1), which is secreted by HNSCC-MSCs and contributes to T cell proliferation inhibition, partially restores T cell proliferation capacity [118]. In contrast, treatment with the HNSCC secretome differentially affected the expression of CD39 and CD73 in MSCs and HNSCC-derived MSCs. CD39 and CD73 hydrolyze extracellular dNTP and AMP, respectively [120]. Extracellular ATP (eATP) and metabolites elicit an inflammatory response, promote cell proliferation, and activate immune cells [128]. MSCs derived from HNSCC exhibit downregulated expression of CD73 and metabolism of ATP [134]. Consequently, the expression of extracellular adenosine (ADO), an eATP metabolite known to inhibit T cell proliferation, is downregulated. The authors suggest that MSCs generate an inflammatory and tumor-friendly microenvironment. This conclusion must be carefully interpreted as different concentrations of ATP and ADO can exert differential effects on the immune cell subtypes. For example, ADO favors the M2-like macrophage phenotype, while eATP, M1-like polarization, and M1 macrophages promote tumor destruction. Meanwhile, eATP modulates lymphocyte (CD4+, CD8+, and Treg cells) infiltration into the tumor microenvironment and promotes cancer growth [135]. A detailed analysis of the effect of eATP on HNSCC and MSCs is needed.

The interaction and secretome among tumor cells and MSCs are associated with the differentiation of the mesenchymal stromal cells in cancer-associated fibroblasts [130132] These cancer-associated fibroblasts can release factors and interact with HNC cells, which turns the tumor more aggressive [132]. However, these CAFs can also be derived from resident fibroblasts, epithelial cells, endothelial cells, cancer stem cells, adipocytes, and pericytes in a complex system [131]. Hence, a detailed discussion on CAF is out of the scope of this review.

6. In vivo studies in MSCs and HNC interactions

Various animal models have been used to study cancer microenvironments (Table 3). Studies on the xenograft tumor models have partially elucidated the role of the MSCs in a complex environment. In the in vitro models, the cells were exposed to a relatively homogeneous environment characterized by the same oxygen concentration and nutrients and limited interactions with different cells. Although in vitro models provided valuable information, they cannot completely mimic the cancer microenvironment. Mouse xenograft models can mimic the in vivo tumor microenvironment. This environment involves communication with stromal cells, such as cells from the innate immune system, even in immunodeficient mice [136]. A similar outcome was reported when HNC cells were co-injected with MSCs from different sources, including those derived from HNSCC although different protocols were used [97,100,108,134]. For example, Rowan et al. (2015) reported that compared with those in the HNC cell lines, the tumor volume and vascularization were higher and the expression of MMP2 and MMP9 was upregulated in HNSCC cells (Cal27 and SCC4 cells) co-cultured with AMSCs [137]. Li et al. reported that the growth and angiogenesis were upregulated in xenograft tumors derived from HNSCC cells (Eca-109 cells) co-cultured with BMMSCs although the in vitro paracrine communication revealed decreased tumor cell proliferation [112]. Additionally, MSC migration toward cancer increased in the xenograft HNSCC model after radiation therapy, which indicated the protective effect of the MSCs during radiotherapy [135].

Table 3:

In vivo evidence of MSC on HNC.

Observed effect Effector cells Publication

Increased tumor size (FaDU) HNSCC-MSC [97]
(Cal27) BMMSC [108]
(Eca109, TE-1) BMMSC [100]

Vascularization, increased tumor size, increased MMP2/9 expression (Cal27, SCC4) AMSC [134]

Vascularization, increased tumor size (Eca109) BMMSC [112]

Increased tumor size after radiation (UMSCC1) MSC [135]

Increased tumor size, lymph node metastasis (Cal27) BMMSC [108]

Increased tumor size (CCN2) BMMSC [101,113]

Increased tumor size (SCC25) and paclitaxel resistance BMMSC [107]

Reduced tumor size, cellular fusion, increased DUSP6 expression (EC9706) MSC (commercial) [116]

Reduced tumor size (Cal27) Gingival derived-MSC [98]

Increased tumor size (TPC1) EV-BMMSC [111]

No influence on tumor growth (FaDu) EV-MSC-Myc [136]

Increased MSC migration toward tumor enriched with myofibroblast (OE21) BMMSC, myofibroblast [137]

DMBA induced tumor – proliferation reduction and increased cell death MSC [138]

DMBA induced tumor nodule stage – tumor progression;

DMBA induced tumor hyperplasia stage – tumor regression
MSC [139]

4NQO induced tumor – reduction on CD3+ and CD45+ cells MSC [119]

MMP – Matrix metalloproteinases; AMSC – Adipose tissue derived-mesenchymal stromal cell; BMMSC – Bone marrow mesenchymal stromal cell; DUSP6 – Dual Specificity Phosphatase 6; EV – Extracellular vesicles; MSC-Myc – immortalized mesenchymal stromal cell; DMBA - 7,12-Dimethylbenz[a]anthracene; 4NQO – 4-Nitroquinoline-1-oxide.

Moreover, lymph node and distant metastases increased in tumors cultured with MSCs [108,140,141]. Several studies have proposed the molecular pathways involved in these outcomes. The mRNA levels of POSTN were upregulated in BMMSC-HNSCC (Cal27 cells) tumors with high proliferation rates, volume, and lymph node metastasis [108]. The downregulation of the expression of GREM1 and CCN2 in MSCs decreased tumor volume, which indicated that these proteins were involved in MSC-dependent HNSCC growth [109,126]. Conversely, overexpression of DUSP6 in MSC-SCC fusion decreased tumorigeneses, angiogenesis, proliferation, and metastasis [116]. Additionally, gingival-derived MSCs inhibited tumor formation in oral SCC [98]. However, the clinical contribution of these findings needs further clarification.

Shi et al. (2016) reported the contribution of EV to cancer progression using an in vivo model [111]. The authors reported that compared with those in animals injected with phosphate-buffered saline, the tumor growth was rapid and the tumor volume was higher in animals injected daily with BMMSC-EV. In contrast, Tan et al. (2021) reported no difference of intraperitoneally injected animals with EV of MYC-immortalized MSCs derived from HNSCC xenograft (FaDu cells) [136]. Kumar et al. (2014) isolated myofibroblasts from HNSCC and co-injected it with an esophageal SCC cell line (OE21) to examine the role of the stroma in MSC homing. Fluorescence probe-labeled BMMSCs were intravascularly injected to allow posterior analysis [137]. The authors reported that the MSCs in the tumor injected with myofibroblasts exhibited increased migration toward the xenograft tumor, which was dependent on chemerin (RARRES2) and its receptor chemR23.

Furthermore, animal studies have been performed to examine the role of MSC in HNSCC carcinogenesis. Bruna et al. (2016) used the Syrian golden hamsters treated with 0.5% 7,12-dimethylbenz[a]anthracene (DMBA) to examine the role of MSCs in oral carcinogenesis [138]. The hamster-derived MSCs were injected into the mucosa around the local nodule after DMBA treatment. MSC injection decreased the tumor size, reduced the number of Ki67-positive cells, and promoted cell death. The same research group also reported the effect of systemic administration of MSCs on oral carcinogenesis [139]. The experimental groups were defined as animals with hyperplasia (4–5 weeks) or hyperplasia and dysplasia during two different time points (6–8 and 9–13 weeks). MSCs dose-dependently promoted the progression of initial nodules to SCC after 13 weeks of treatment in all animals. Surprisingly, the tumor volume in animals injected with an intermediate MSC concentration was lower than that in control animals. Animals injected with low and intermediate MSC concentrations exhibited hyperplasia and dysplasia in approximately 25% of the samples. The terminal point analysis revealed that almost all hyperplasia plus dysplasia groups injected with MSCs did not progress to SCC. After 13 weeks, the control animals were diagnosed with SCC, which indicated the tumor-protective role of MSCs. The clinical manifestation of leukoplakia was observed only in tissues with hyperplasia + dysplasia, which does not completely mimic the clinical and microscopic characteristics of human patients [142]. The authors suggest that systemic administration of MSCs can influence tumor progression in later phases of carcinogenesis because chronic inflammation persists in the tissue, which augments MSCs homing. High doses of MSCs administration can elicit an immune response in healthy animals and promote tumor growth.

Female rats with oral lesions induced by administering 0.05 g/L 4-nitroquinoline-1-oxide (4NQO) for 22 weeks exhibited increased numbers of MSCs in premalignant SCC tissue with dysplasia [119]. Analysis of the MSC secretome profile derived from these tissues revealed upregulated secretion of CCL21 and SDF1. Interestingly, the cancer samples exhibited decreased numbers of lymphocytes (CD3+) and leukocytes (CD45+). The MSC content was weakly and inversely correlated with CD3+ (R2 = 0.1371; p-value = 0.2361) or C45+ cells (R2 = 0.0308; p-value = 0.5849). This indicated that MSCs and other stromal cells could contribute to the decreased CD3+ number. The proliferation of CD3+ cells was significantly inhibited upon co-culturing with cancer-derived MSCs. Ouchi et al. reported the potential role of MSCs and their derivatives in HNC. They demonstrated the enrichment of CD90+/CD105+/CD73+ cells in recurrent spindle cell carcinoma, a high-grade SCC subtype, of the tongue [143].

7. Role of MSCs in HNC progression: Conclusions and perspectives

The findings of previous studies support the dual role of MSCs in HNC progression with a tendency to promote cancer aggressiveness. MSCs have a differential effect on different phases of carcinogenesis. However, this cannot be a conclusive finding as different studies use different models. In summary, MSCs isolated from different tissue sources exert their effects through the secretion of IL-6, VEGF, B2M, CCN2, TGF-β, SDF-1, POSTN, FGF19, miR-8485, IDO1, IL4I1, and EGF. These ligands act on PDGFR, VEGFR, FGFR, and EGFR on the cancer cell surface to exert various effects, inducing upregulation of MMP9, MMP2, MMP14, and epithelial-mesenchymal plasticity markers. The induction of MMP expression promotes the MSC-mediated and cancer cell-mediated ECM modifications and facilitates the migration of cancer cells into the stroma [11,144,145]. MMP may aid the extravasation of cancer cells from the blood and lymph vessels, which enables the migration of cancer cells to regional or distant sites [11,146]. Epithelial-mesenchymal plasticity is a cellular process in which epithelial cells lose the epithelial phenotype and acquire the mesenchymal phenotypes and behavior [147]. During this process, the expression of mesenchymal markers, such as vimentin and N-cadherin is upregulated, whereas that of epithelial markers (such as E-cadherin) is downregulated in epithelial cells. Acquiring mesenchymal characteristics promotes the migration of cancer cells. The downregulation of adhesive epithelial proteins facilitates the detachment from other epithelial cells, which promotes cell migration. HNC exhibited partial epithelial-mesenchymal characteristics dependent on the stromal content and correlated with lymph node metastasis [49]. MSCs can induce the expression of laminin 5, which facilitates cancer cell migration. Additionally, MSCs can mitigate HNC progression through some unknown signaling pathways. These unknown signaling (non-exclusively dependent on TGFRIII, ERK, and AKT) pathways downregulate the expression of MMP and collagen IV, inhibit proliferation, and upregulate DUSP6 (in MSC-HNC fusion).

Previous studies have begun to elucidate the characteristics of the HNC microenvironment and the role of MSCs in tumor progression (Figure 2). However, translational research could be conducted in the future to address various knowledge gaps, such as the role of MSCs in the molecular pathways involved in MSC-mediated metastasis induction, tumor radiotherapy resistance, and immunotherapy response in human cancers.

Figure 2: Effect of mesenchymal stromal cells (MSCs) on head and neck cancer (HNC).

Figure 2:

MSCs can affect both tumor progression and arrest with a tendency to promote the former. Various molecules can regulate the progression of HNC by modulating MSC behavior. IL-6, platelet-derived growth factor (PDGF), and matrix metalloproteinase (MMP) secreted by HNC cells influence MSC behavior. Similarly, B2M, CCN2, vascular endothelial growth factor, TNFβ, SDF1, SCGFβ, POSTN, CCL5, IL-6, FGF19, miR-8485, and MMP modulate HNC progression and affect cell survival, proliferation, migration, invasion, and epithelial-mesenchymal plasticity (EMP). Additionally, MSCs regulate immune response (by regulating the expression of IDO, CD39, CD73), differentiate into specific cell types (fibroblasts and myofibroblasts), and consequently modulate HNC behavior. GREM1, BMP4, TGFβ, MMP, laminin-5, integrin, and EGFR promote cancer cell migration and invasion and EMP. The fusion of MSCs and cancer cells leads to the production of DUSP6 and decreased cancer cell survival.

Highlights.

  • We review evidences for the MSC secretome on the HNC microenviroment and progression

  • We describe bidirecional communication between MSC and HNC on cell phenotypes.

  • We summarize in vitro and in vivo studies perfomed in HNC model

Acknowledgments

The authors would like to thank FAPEMIG, CAPES, and CNPq to support this work. All the figures were designed using BioRender. We would like to thank Editage (www.editage.com) for English language editing.

Funding

This work was supported by NIH grant 1R03TW008709 and by grants from the “INCT-Regenera”, “Rede Mineira de Engenharia de Tecidos e Terapia Celular (REMETTEC, RED-00570-16)” and FAPEMIG (APQ-03132-18).

Footnotes

Declaration of interests

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

Conflicts of interest

The authors declare no conflict of interest.

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • [1].Gupta B, Johnson NW, Kumar N, Global Epidemiology of Head and Neck Cancers: A Continuing Challenge, Oncol. 91 (2016) 13–23. doi: 10.1159/000446117. [DOI] [PubMed] [Google Scholar]
  • [2].Arboleda LPA, de Mendonça RMH, Lopez EEM, Araújo ALD, Palmier NR, de Pauli Paglioni M, Fonseca JM, Hoffmann IL, Cardinalli IA, Chaves ALF, Aranda S, Brandão TB, Lopes MA, Ribeiro ACP, Troconis CCM, Santos-Silva AR, Global frequency and distribution of head and neck cancer in pediatrics, a systematic review, Crit. Rev. Oncol. Hematol. 148 (2020). doi: 10.1016/j.critrevonc.2020.102892. [DOI] [PubMed] [Google Scholar]
  • [3].Tian Z, Li L, Wang L, Hu Y, Li J, Salivary gland neoplasms in oral and maxillofacial regions: a 23-year retrospective study of 6982 cases in an eastern Chinese population, Int. J. Oral Maxillofac. Surg. 39 (2010) 235–242. doi: 10.1016/j.ijom.2009.10.016. [DOI] [PubMed] [Google Scholar]
  • [4].Rizzitelli A, Smoll NR, Chae MP, Rozen WM, Hunter-Smith DJ, Incidence and overall survival of malignant ameloblastoma, PLoS One. 10 (2015). doi: 10.1371/journal.pone.0117789. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [5].Mifsud M, Eskander A, Irish J, Gullane P, Gilbert R, Brown D, de Almeida JR, Urbach DR, Goldstein DP, Evolving trends in head and neck cancer epidemiology: Ontario, Canada 1993–2010, Head Neck. 39 (2017) 1770–1778. doi: 10.1002/hed.24829. [DOI] [PubMed] [Google Scholar]
  • [6].Tshering P, Dorjee S, Dendup T, Dorji T, Wangmo D, Epidemiological and histopathological characteristics of head and neck cancers in Bhutan from 2011 to 2017: a retrospective descriptive study, Ecancermedicalscience. 14 (2020). doi: 10.3332/ecancer.2020.1024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [7].Siegel RL, Miller KD, Jemal A, Cancer statistics, 2018, CA. Cancer J. Clin. 68 (2018) 7–30. doi: 10.3322/caac.21442. [DOI] [PubMed] [Google Scholar]
  • [8].Ng JH, Iyer NG, Tan M-H, Edgren G, Changing epidemiology of oral squamous cell carcinoma of the tongue: A global study, Head Neck. 39 (2017) 297–304. doi: 10.1002/hed.24589. [DOI] [PubMed] [Google Scholar]
  • [9].Chow LQM, Head and neck cancer, N. Engl. J. Med. 382 (2020) 60–72. doi: 10.1056/NEJMra1715715. [DOI] [PubMed] [Google Scholar]
  • [10].Ho AS, Kim S, Tighiouart M, Gudino C, Mita A, Scher KS, Laury A, Prasad R, Shiao SL, Van Eyk JE, Zumsteg ZS, Metastatic lymph node burden and survival in oral cavity cancer, J. Clin. Oncol. 35 (2017) 3601–3609. doi: 10.1200/JCO.2016.71.1176. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [11].Geiger TR, Peeper DS, Metastasis mechanisms, Biochim. Biophys. Acta - Rev. Cancer. 1796 (2009) 293–308. doi: 10.1016/j.bbcan.2009.07.006. [DOI] [PubMed] [Google Scholar]
  • [12].Gupta GP, Massagué J, Cancer Metastasis: Building a Framework, Cell. 127 (2006) 679–695. doi: 10.1016/j.cell.2006.11.001. [DOI] [PubMed] [Google Scholar]
  • [13].Mbeunkui F, Johann DJ, Cancer and the tumor microenviroment: a review of an essential relationship, 63 (2010) 571–582. doi: 10.1007/s00280-008-0881-9.Cancer. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [14].Mondino A, Vella G, Icardi L, Targeting the tumor and its associated stroma: One and one can make three in adoptive T cell therapy of solid tumors, Cytokine Growth Factor Rev. 36 (2017) 57–65. doi: 10.1016/j.cytogfr.2017.06.006. [DOI] [PubMed] [Google Scholar]
  • [15].Peltanova B, Raudenska M, Masarik M, Effect of tumor microenvironment on pathogenesis of the head and neck squamous cell carcinoma: A systematic review, Mol. Cancer. 18 (2019). doi: 10.1186/s12943-019-0983-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [16].Taddei ML, Giannoni E, Comito G, Chiarugi P, Microenvironment and tumor cell plasticity: An easy way out, Cancer Lett. 341 (2013) 80–96. doi: 10.1016/j.canlet.2013.01.042. [DOI] [PubMed] [Google Scholar]
  • [17].Balkwill FR, Capasso M, Hagemann T, The tumor microenvironment at a glance, J. Cell Sci. 125 (2012) 5591–5596. doi: 10.1242/jcs.116392. [DOI] [PubMed] [Google Scholar]
  • [18].Quail DF, Joyce JA, Microenvironmental regulation of tumor progression and metastasis., Nat. Med. 19 (2013) 1423–37. doi: 10.1038/nm.3394. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [19].Dogan V, Rieckmann T, Münscher A, Busch CJ, Current studies of immunotherapy in head and neck cancer, Clin. Otolaryngol. 43 (2018) 13–21. doi: 10.1111/coa.12895. [DOI] [PubMed] [Google Scholar]
  • [20].Cuiffo BG, Karnoub a E., Mesenchymal stem cells in tumor development: emerging roles and concepts, Cell Adh Migr. 6 (2012) 220–230. doi: 10.4161/cam.20875. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [21].Galland S, Stamenkovic I, Mesenchymal stromal cells in cancer: a review of their immunomodulatory functions and dual effects on tumor progression, J. Pathol. 250 (2020) 555–572. doi: 10.1002/path.5357. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [22].Pittenger MF, Mackay AM, Beck SC, Jaiswal RK, Douglas R, Mosca JD, Moorman MA, Simonetti DW, Craig S, Marshak DR, Multilineage potential of adult human mesenchymal stem cells, Science (80-. ). 284 (1999) 143–147. doi: 10.1126/science.284.5411.143. [DOI] [PubMed] [Google Scholar]
  • [23].Bianco P, “Mesenchymal” Stem Cells, Annu. Rev. Cell Dev. Biol. 30 (2014) 677–704. doi: 10.1146/annurev-cellbio-100913-013132. [DOI] [PubMed] [Google Scholar]
  • [24].Kfoury Y, Scadden DT, Mesenchymal cell contributions to the stem cell niche, Cell Stem Cell. 16 (2015) 239–253. doi: 10.1016/j.stem.2015.02.019. [DOI] [PubMed] [Google Scholar]
  • [25].Spees JL, Lee RH, Gregory CA, Mechanisms of mesenchymal stem/stromal cell function, Stem Cell Res. Ther. 7 (2016). doi: 10.1186/s13287-016-0363-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [26].Coussens LM, Werb Z, Inflammation and cancer., Nature. 420 (2002) 860–7. doi: 10.1038/nature01322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [27].Dominici M, Le Blanc K, Mueller I, Slaper-Cortenbach I, Marini F, Krause D, Deans R, Keating a, Prockop D, Horwitz E, Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement., Cytotherapy. 8 (2006) 315–7. doi: 10.1080/14653240600855905. [DOI] [PubMed] [Google Scholar]
  • [28].Linard C, Brachet M, Strup-Perrot C, Busson E, Bonneau M, Lataillade J-J, Bey E, Benderitter M, Long-term effectiveness of local BMMSCs for skeletal muscle regeneration: a proof of concept obtained on a pig model of severe radiation burn, (n.d.). doi: 10.1186/s13287-018-1051-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [29].Mckenzie JL, Gan OI, Doedens M, Dick JE, Human short-term repopulating stem cells are efficiently detected following intrafemoral transplantation into NOD/SCID recipients depleted of CD122 cells, (2005). doi: 10.1182/blood-2005-03-1081. [DOI] [PubMed] [Google Scholar]
  • [30].Horwitz EM, Le Blanc K, Dominici M, Mueller I, Slaper-Cortenbach I, Marini FC, Deans RJ, Krause DS, Keating A, Clarification of the nomenclature for MSC: The International Society for Cellular Therapy position statement, Cytotherapy. 7 (2005) 393–395. doi: 10.1080/14653240500319234. [DOI] [PubMed] [Google Scholar]
  • [31].Halvorsen YC, Wilkison WO, Gimble JM, Adipose-derived stromal cells—their utility and potential in bone formation, Int. J. Obes. 24 (2000) S41–S44. doi: 10.1038/sj.ijo.0801503. [DOI] [PubMed] [Google Scholar]
  • [32].Zuk PA, Zhu M, Ashjian P, De Ugarte DA, Huang JI, Mizuno H, Alfonso ZC, Fraser JK, Benhaim P, Hedrick MH, Human Adipose Tissue Is a Source of Multipotent Stem Cells, Mol. Biol. Cell. 13 (2002) 4279–4295. doi: 10.1091/mbc.E02. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [33].Bourin P, Bunnell B, Casteilla L, Dominici M, Katz A, March K, Redl H, Rubin J, Yoshimura K, Gimble J, Stromal cells from the adipose tissue-derived stromal vascular fraction and culture expanded adipose tissue-derived stromal/stem cells: a joint statement of the International Federation for Adipose Therapeutics and Science (IFATS) and the International So, Cytotherapy. 15 (2013) 641–648. doi: 10.1016/j.jcyt.2013.02.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [34].Wagner W, Wein F, Seckinger A, Frankhauser M, Wirkner U, Krause U, Blake J, Schwager C, Eckstein V, Ansorge W, Ho AD, Comparative characteristics of mesenchymal stem cells from human bone marrow, adipose tissue, and umbilical cord blood, Exp. Hematol. 33 (2005) 1402–1416. doi: 10.1016/j.exphem.2005.07.003. [DOI] [PubMed] [Google Scholar]
  • [35].Abu Kasim NH, Govindasamy V, Gnanasegaran N, Musa S, Pradeep PJ, Srijaya TC, Aziz ZACA, Unique molecular signatures influencing the biological function and fate of post-natal stem cells isolated from different sources, J. Tissue Eng. Regen. Med. 9 (2015) E252–E266. doi: 10.1002/term.1663. [DOI] [PubMed] [Google Scholar]
  • [36].Crisan M, Yap S, Casteilla L, Chen CW, Corselli M, Park TS, Andriolo G, Sun B, Zheng B, Zhang L, Norotte C, Teng PN, Traas J, Schugar R, Deasy BM, Badylak S, Buhring HJ, Giacobino JP, Lazzari L, Huard J, Péault B, A Perivascular Origin for Mesenchymal Stem Cells in Multiple Human Organs, Cell Stem Cell. 3 (2008) 301–313. doi: 10.1016/j.stem.2008.07.003. [DOI] [PubMed] [Google Scholar]
  • [37].Covas DT, Panepucci RA, Fontes AM, Silva WA, Orellana MD, Freitas MCC, Neder L, Santos ARD, Peres LC, Jamur MC, Zago MA, Multipotent mesenchymal stromal cells obtained from diverse human tissues share functional properties and gene-expression profile with CD146+ perivascular cells and fibroblasts, Exp. Hematol. 36 (2008) 642–654. doi: 10.1016/j.exphem.2007.12.015. [DOI] [PubMed] [Google Scholar]
  • [38].Abreu de Melo MI, da Silva Cunha P, Coutinho de Miranda M, Faraco CCF, Barbosa JL, da Fonseca Ferreira A, Kunrath Lima M, Faria JAQA, Rodrigues MÂ, de Goes AM, Gomes DA, Human adipose-derived stromal/stem cells are distinct from dermal fibroblasts as evaluated by biological characterization and RNA sequencing, Cell Biochem. Funct. (2021). doi: 10.1002/cbf.3610. [DOI] [PubMed] [Google Scholar]
  • [39].Schwab KE, Gargett CE, Co-expression of two perivascular cell markers isolates mesenchymal stem-like cells from human endometrium, Hum. Reprod. 22 (2007) 2903–2911. doi: 10.1093/humrep/dem265. [DOI] [PubMed] [Google Scholar]
  • [40].da Silva Meirelles L, Caplan AI, Nardi NB, In Search of the In Vivo Identity of Mesenchymal Stem Cells, Stem Cells. 26 (2008) 2287–2299. doi: 10.1634/stemcells.2007-1122. [DOI] [PubMed] [Google Scholar]
  • [41].Denu RA, Nemcek S, Bloom DD, Goodrich AD, Kim J, Mosher DF, Hematti P, Fibroblasts and Mesenchymal Stromal/Stem Cells Are Phenotypically Indistinguishable, Acta Haematol. 136 (2016) 85–97. doi: 10.1159/000445096. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [42].Corselli M, Chen CW, Sun B, Yap S, Rubin JP, Péault B, The tunica adventitia of human arteries and veins as a source of mesenchymal stem cells, Stem Cells Dev. 21 (2012) 1299–1308. doi: 10.1089/scd.2011.0200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [43].Mishra PJ, Mishra PJ, Humeniuk R, Medina DJ, Alexe G, Mesirov JP, Ganesan S, Glod JW, Banerjee D, Carcinoma-associated fibroblast-like differentiation of human mesenchymal stem cells, Cancer Res. 68 (2008) 4331–4339. doi: 10.1158/0008-5472.CAN-08-0943. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [44].Mishra PJ, Banerjee D, Activation and differentiation of mesenchymal stem cells, in: Methods Mol. Biol., Humana Press Inc., 2017: pp. 201–209. doi: 10.1007/978-1-4939-6759-9_13. [DOI] [PubMed] [Google Scholar]
  • [45].Dingal PCDP, Bradshaw AM, Cho S, Raab M, Buxboim A, Swift J, Discher DE, Fractal heterogeneity in minimal matrix models of scars modulates stiff-niche stem-cell responses via nuclear exit of a mechanorepressor, Nat. Mater. 14 (2015) 951–960. doi: 10.1038/nmat4350. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [46].Tang H, Chu Y, Huang Z, Cai J, Wang Z, The metastatic phenotype shift toward myofibroblast of adipose-derived mesenchymal stem cells promotes ovarian cancer progression, Carcinogenesis. (2019). doi: 10.1093/carcin/bgz083. [DOI] [PubMed] [Google Scholar]
  • [47].Matsumoto K, Xavier S, Chen J, Kida Y, Lipphardt M, Ikeda R, Gevertz A, Caviris M, Hatzopoulos AK, Kalajzic I, Dutton J, Ratliff BB, Zhao H, Darzynkiewicz Z, Rose-John S, Goligorsky MS, Instructive Role of the Microenvironment in Preventing Renal Fibrosis, Stem Cells Transl. Med. 6 (2017) 992–1005. doi: 10.5966/sctm.2016-0095. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [48].Kahounová Z, Kurfürstová D, Bouchal J, Kharaishvili G, Navrátil J, Remšík J, Šimečková Š, Študent V, Kozubík A, Souček K, The fibroblast surface markers FAP, anti-fibroblast, and FSP are expressed by cells of epithelial origin and may be altered during epithelial-to-mesenchymal transition, Cytom. Part A. 93 (2018) 941–951. doi: 10.1002/cyto.a.23101. [DOI] [PubMed] [Google Scholar]
  • [49].Puram SV, Tirosh I, Parikh AS, Patel AP, Yizhak K, Gillespie S, Rodman C, Luo CL, Mroz EA, Emerick KS, Deschler DG, Varvares MA, Mylvaganam R, Rozenblatt-Rosen O, Rocco JW, Faquin WC, Lin DT, Regev A, Bernstein BE, Single-Cell Transcriptomic Analysis of Primary and Metastatic Tumor Ecosystems in Head and Neck Cancer, Cell. 171 (2017) 1611–1624.e24. doi: 10.1016/j.cell.2017.10.044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [50].Worthley DL, Churchill M, Compton JT, Tailor Y, Rao M, Si Y, Levin D, Schwartz MG, Uygur A, Hayakawa Y, Gross S, Renz BW, Setlik W, Martinez AN, Chen X, Nizami S, Lee HG, Kang HP, Caldwell JM, Asfaha S, Westphalen CB, Graham T, Jin G, Nagar K, Wang H, Kheirbek MA, Kolhe A, Carpenter J, Glaire M, Nair A, Renders S, Manieri N, Muthupalani S, Fox JG, Reichert M, Giraud AS, Schwabe RF, Pradere JP, Walton K, Prakash A, Gumucio D, Rustgi AK, Stappenbeck TS, Friedman RA, Gershon MD, Sims P, Grikscheit T, Lee FY, Karsenty G, Mukherjee S, Wang TC, Gremlin 1 identifies a skeletal stem cell with bone, cartilage, and reticular stromal potential, Cell. 160 (2015) 269–284. doi: 10.1016/j.cell.2014.11.042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [51].Gang EJ, Bosnakovski D, Figueiredo CA, Visser JW, Perlingeiro RCR, SSEA-4 identifies mesenchymal stem cells from bone marrow, Blood. 109 (2007) 1743–1751. doi: 10.1182/blood-2005-11-010504. [DOI] [PubMed] [Google Scholar]
  • [52].Maeda K, Enomoto A, Hara A, Asai N, Kobayashi T, Horinouchi A, Maruyama S, Ishikawa Y, Nishiyama T, Kiyoi H, Kato T, Ando K, Weng L, Mii S, Asai M, Mizutani Y, Watanabe O, Hirooka Y, Goto H, Takahashi M, Identification of Meflin as a Potential Marker for Mesenchymal Stromal Cells, Sci. Rep. 6 (2016). doi: 10.1038/srep22288. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [53].Aggoune D, Sorel N, Bonnet ML, Goujon JM, Tarte K, Hérault O, Domenech J, Réa D, Legros L, Johnson-Ansa H, Rousselot P, Cayssials E, Guerci-Bresler A, Bennaceur-Griscelli A, Chomel JC, Turhan AG, Bone marrow mesenchymal stromal cell (MSC) gene profiling in chronic myeloid leukemia (CML) patients at diagnosis and in deep molecular response induced by tyrosine kinase inhibitors (TKIs), Leuk. Res. 60 (2017) 94–102. doi: 10.1016/j.leukres.2017.07.007. [DOI] [PubMed] [Google Scholar]
  • [54].Montelatici E, Baluce B, Ragni E, Lavazza C, Parazzi V, Mazzola R, Cantarella G, Brambilla M, Giordano R, Lazzari L, Defining the identity of human adipose-derived mesenchymal stem cells, 82 (2015) 74–82. doi: 10.1139/bcb-2014-0094. [DOI] [PubMed] [Google Scholar]
  • [55].Lv FJ, Tuan RS, Cheung KMC, Leung VYL, Concise review: The surface markers and identity of human mesenchymal stem cells, Stem Cells. 32 (2014) 1408–1419. doi: 10.1002/stem.1681. [DOI] [PubMed] [Google Scholar]
  • [56].Caplan AI, Adult Mesenchymal Stem Cells: When, Where, and How., Stem Cells Int. 2015 (2015) 628766–628767. doi: 10.1155/2015/628767. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [57].Rüster B, Göttig S, Ludwig RJ, Bistrian R, Müller S, Seifried E, Gille J, Henschler R, Mesenchymal stem cells display coordinated rolling and adhesion behavior on endothelial cells, Blood. 108 (2006) 3938–3944. doi: 10.1182/blood-2006-05-025098. [DOI] [PubMed] [Google Scholar]
  • [58].Suila H, Hirvonen T, Kotovuori A, Ritamo I, Kerkelä E, Anderson H, Natunen S, Tuimala J, Laitinen S, Nystedt J, Räbinä J, Valmu L, Human umbilical cord blood-derived mesenchymal stromal cells display a novel interaction between P-selectin and galectin-1, Scand. J. Immunol. 80 (2014) 12–21. doi: 10.1111/sji.12179. [DOI] [PubMed] [Google Scholar]
  • [59].Sackstein R, Merzaban JS, Cain DW, Dagia NM, Spencer JA, Lin CP, Wohlgemuth R, Ex vivo glycan engineering of CD44 programs human multipotent mesenchymal stromal cell trafficking to bone, Nat. Med. 14 (2008) 181–187. doi: 10.1038/nm1703. [DOI] [PubMed] [Google Scholar]
  • [60].Moravcikova E, Meyer EM, Corselli M, Donnenberg VS, Donnenberg AD, Proteomic Profiling of Native Unpassaged and Culture-Expanded Mesenchymal Stromal Cells (MSC), Cytom. Part A. 93 (2018) 894–904. doi: 10.1002/cyto.a.23574. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [61].Bailey AM, Lawrence MB, Shang H, Katz AJ, Peirce SM, Agent-based model of therapeutic adipose-derived stromal cell trafficking during ischemia predicts ability to roll on p-selectin, PLoS Comput. Biol. 5 (2009). doi: 10.1371/journal.pcbi.1000294. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [62].Ciuculescu F, Giesen M, Deak E, Lang V, Seifried E, Henschler R, Variability in chemokine-induced adhesion of human mesenchymal stromal cells., Cytotherapy. 13 (2011) 1–8. doi: 10.3109/14653249.2011.602339. [DOI] [PubMed] [Google Scholar]
  • [63].Segers VFM, Van Riet I, Andries LJ, Lemmens K, Demolder MJ, De Becker AJML, Kockx MM, De Keulenaer GW, Mesenchymal stem cell adhesion to cardiac microvascular endothelium: Activators and mechanisms, Am. J. Physiol. - Hear. Circ. Physiol. 290 (2006). doi: 10.1152/ajpheart.00523.2005. [DOI] [PubMed] [Google Scholar]
  • [64].Steingen C, Brenig F, Baumgartner L, Schmidt J, Schmidt A, Bloch W, Characterization of key mechanisms in transmigration and invasion of mesenchymal stem cells, J. Mol. Cell. Cardiol. 44 (2008) 1072–1084. doi: 10.1016/j.yjmcc.2008.03.010. [DOI] [PubMed] [Google Scholar]
  • [65].Ries C, Egea V, Karow M, Kolb H, Jochum M, Neth P, MMP-2, MT1-MMP, and TIMP-2 are essential for the invasive capacity of human mesenchymal stem cells: Differential regulation by inflammatory cytokines, Blood. 109 (2007) 4055–4063. doi: 10.1182/blood-2006-10-051060. [DOI] [PubMed] [Google Scholar]
  • [66].Hou Y, Ryu CH, Jun JA, Kim SM, Jeong CH, Jeun SS, IL-8 enhances the angiogenic potential of human bone marrow mesenchymal stem cells by increasing vascular endothelial growth factor, Cell Biol. Int. 38 (2014) 1050–1059. doi: 10.1002/cbin.10294. [DOI] [PubMed] [Google Scholar]
  • [67].Ponte AL, Marais E, Gallay N, Langonné A, Delorme B, Hérault O, Charbord P, Domenech J, The In Vitro Migration Capacity of Human Bone Marrow Mesenchymal Stem Cells: Comparison of Chemokine and Growth Factor Chemotactic Activities, Stem Cells. 25 (2007) 1737–1745. doi: 10.1634/stemcells.2007-0054. [DOI] [PubMed] [Google Scholar]
  • [68].Scherzed A, Hackenberg S, Froelich K, Rak K, Schendzielorz P, Gehrke T, Hagen R, Kleinsasser N, The differentiation of hMSCs counteracts their migration capability and pro-angiogenic effects in vitro, Oncol. Rep. 35 (2016) 219–226. doi: 10.3892/or.2015.4383. [DOI] [PubMed] [Google Scholar]
  • [69].Schroder K, Hertzog PJ, Ravasi T, Hume DA, Interferon-γ: an overview of signals, mechanisms and functions, J. Leukoc. Biol. 75 (2004) 163–189. doi: 10.1189/jlb.0603252. [DOI] [PubMed] [Google Scholar]
  • [70].Krampera M, Cosmi L, Angeli R, Pasini A, Liotta F, Andreini A, Santarlasci V, Mazzinghi B, Pizzolo G, Vinante F, Romagnani P, Maggi E, Romagnani S, Annunziato F, Role for Interferon-γ in the Immunomodulatory Activity of Human Bone Marrow Mesenchymal Stem Cells, Stem Cells. 24 (2006) 386–398. doi: 10.1634/stemcells.2005-0008. [DOI] [PubMed] [Google Scholar]
  • [71].Choi H, Lee RH, Bazhanov N, Oh JY, Prockop DJ, Anti-inflammatory protein TSG-6 secreted by activated MSCs attenuates zymosan-induced mouse peritonitis by decreasing TLR2/NF-κB signaling in resident macrophages, Blood. 118 (2011) 330–338. doi: 10.1182/blood-2010-12-327353. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [72].Domenis R, Cifù A, Quaglia S, Pistis C, Moretti M, Vicario A, Parodi PC, Fabris M, Niazi KR, Soon-Shiong P, Curcio F, Pro inflammatory stimuli enhance the immunosuppressive functions of adipose mesenchymal stem cells-derived exosomes, Sci. Rep. 8 (2018) 1–11. doi: 10.1038/s41598-018-31707-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [73].Di Nicola M, Carlo-Stella C, Magni M, Milanesi M, Longoni PD, Matteucci P, Grisanti S, Gianni AM, Human bone marrow stromal cells suppress T-lymphocyte proliferation induced by cellular or nonspecific mitogenic stimuli, Blood. 99 (2002) 3838–3843. doi: 10.1182/blood.V99.10.3838. [DOI] [PubMed] [Google Scholar]
  • [74].Le Blanc K, Davies LC, Mesenchymal stromal cells and the innate immune response, Immunol. Lett. 168 (2015) 140–146. doi: 10.1016/j.imlet.2015.05.004. [DOI] [PubMed] [Google Scholar]
  • [75].Shi Y, Wang Y, Li Q, Liu K, Hou J, Shao C, Wang Y, Immunoregulatory mechanisms of mesenchymal stem and stromal cells in inflammatory diseases, Nat. Rev. Nephrol. 14 (2018) 493–507. doi: 10.1038/s41581-018-0023-5. [DOI] [PubMed] [Google Scholar]
  • [76].Blazar BR, MacDonald KPA, Hill GR, Immune regulatory cell infusion for graft-versus-host disease prevention and therapy, Blood. 131 (2018) 2651–2660. doi: 10.1182/blood-2017-11-785865. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [77].Reis M, Mavin E, Nicholson L, Green K, Dickinson AM, Wang XN, Mesenchymal stromal cell-derived extracellular vesicles attenuate dendritic cell maturation and function, Front. Immunol. 9 (2018). doi: 10.3389/fimmu.2018.02538. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [78].Petri RM, Hackel A, Hahnel K, Dumitru CA, Bruderek K, Flohe SB, Paschen A, Lang S, Brandau S, Activated Tissue-Resident Mesenchymal Stromal Cells Regulate Natural Killer Cell Immune and Tissue-Regenerative Function, Stem Cell Reports. 9 (2017) 985–998. doi: 10.1016/j.stemcr.2017.06.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [79].Madrigal M, Rao KS, Riordan NH, A review of therapeutic effects of mesenchymal stem cell secretions and induction of secretory modification by different culture methods., J. Transl. Med. 12 (2014) 260. doi: 10.1186/s12967-014-0260-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [80].Ma S, Xie N, Li W, Yuan B, Shi Y, Wang Y, Immunobiology of mesenchymal stem cells., Cell Death Differ. 21 (2014) 216–25. doi: 10.1038/cdd.2013.158. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [81].Wang D, DuBois RN, Immunosuppression associated with chronic inflammation in the tumor microenvironment., Carcinogenesis. 36 (2015) 1085–93. doi: 10.1093/carcin/bgv123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [82].Rafei M, Hsieh J, Fortier S, Li MY, Yuan S, Birman E, Forner K, Boivin MN, Doody K, Tremblay M, Annabi B, Galipeau J, Mesenchymal stromal cell derived CCL2 suppresses plasma cell immunoglobulin production via STAT3 inactivation and PAX5 induction, Blood. 112 (2008) 4991–4998. doi: 10.1182/blood-2008-07-166892. [DOI] [PubMed] [Google Scholar]
  • [83].Weber CE, Kuo PC, The tumor microenvironment, Surg. Oncol. 21 (2012) 172–177. doi: 10.1016/j.suronc.2011.09.001. [DOI] [PubMed] [Google Scholar]
  • [84].Ribatti D, Tamma R, A revisited concept. Tumors: Wounds that do not heal, Crit. Rev. Oncol. Hematol. 128 (2018) 65–69. doi: 10.1016/j.critrevonc.2018.05.016. [DOI] [PubMed] [Google Scholar]
  • [85].Patocs A, Zhang L, Xu Y, Weber F, Caldes T, Mutter GL, Platzer P, Eng C, Breast-cancer stromal cells with TP53 mutations and nodal metastases, N. Engl. J. Med. 357 (2007) 2543–2551. doi: 10.1056/NEJMoa071825. [DOI] [PubMed] [Google Scholar]
  • [86].Liu ET, Stromal effects in breast cancer., N. Engl. J. Med. 357 (2007) 2537–8. doi: 10.1056/NEJMp0707576. [DOI] [PubMed] [Google Scholar]
  • [87].Mao Y, Keller ET, Garfield DH, Shen K, Wang J, Stromal cells in tumor microenvironment and breast cancer, Cancer Metastasis Rev. 32 (2013) 303–315. doi: 10.1007/s10555-012-9415-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [88].Waldman AD, Fritz JM, Lenardo MJ, A guide to cancer immunotherapy: from T cell basic science to clinical practice, Nat. Rev. Immunol. 2020 2011. 20 (2020) 651–668. doi: 10.1038/s41577-020-0306-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [89].Cuiffo BG, Campagne A, Bell GW, Lembo A, Orso F, Lien EC, Bhasin MK, Raimo M, Hanson SE, Marusyk A, El-Ashry D, Hematti P, Polyak K, Mechta-Grigoriou F, Mariani O, Volinia S, Vincent-Salomon A, Taverna D, Karnoub AE, MSC-regulated microRNAs converge on the transcription factor FOXP2 and promote breast cancer metastasis, Cell Stem Cell. 15 (2014) 762–774. doi: 10.1016/j.stem.2014.10.001. [DOI] [PubMed] [Google Scholar]
  • [90].Ohkouchi S, Block GJ, Katsha AM, Kanehira M, Ebina M, Kikuchi T, Saijo Y, Nukiwa T, Prockop DJ, Mesenchymal Stromal Cells Protect Cancer Cells From ROS-induced Apoptosis and Enhance the Warburg Effect by Secreting STC1, Mol. Ther. 20 (2012) 417–423. doi: 10.1038/mt.2011.259. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [91].Kupcova Skalnikova H, Proteomic techniques for characterisation of mesenchymal stem cell secretome, Biochimie. 95 (2013) 2196–2211. doi: 10.1016/j.biochi.2013.07.015. [DOI] [PubMed] [Google Scholar]
  • [92].Tachida Y, Sakurai H, Okutsu J, Proteomic Comparison of the Secreted Factors of Mesenchymal Stem Cells from Bone Marrow, Adipose Tissue and Dental Pulp, J. Proteomics Bioinform. 8 (2015). doi: 10.4172/jpb.1000379. [DOI] [Google Scholar]
  • [93].Watts TL, Cui R, Szaniszlo P, Resto VA, Powell DW, Pinchuk IV, PDGF-AA mediates mesenchymal stromal cell chemotaxis to the head and neck squamous cell carcinoma tumor microenvironment, J. Transl. Med. 14 (2016). doi: 10.1186/s12967-016-1091-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [94].Al-Toub M, Almusa A, Almajed M, Al-Nbaheen M, Kassem M, Aldahmash A, Alajez NM, Pleiotropic effects of cancer cells’ secreted factors on human stromal (mesenchymal) stem cells, Stem Cell Res. Ther. 4 (2013). doi: 10.1186/scrt325. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [95].Salo S, Bitu C, Merkku K, Nyberg P, Bello IO, Vuoristo J, Sutinen M, Vähänikkilä H, Costea DE, Kauppila J, Lehenkari P, Dayan D, Vered M, Risteli J, Salo T, Human Bone Marrow Mesenchymal Stem Cells Induce Collagen Production and Tongue Cancer Invasion, PLoS One. 8 (2013). doi: 10.1371/journal.pone.0077692. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [96].Scherzed A, Hackenberg S, Radeloff A, Froelich K, Rak K, Hagen R, Kleinsasser N, Human mesenchymal stem cells promote cancer motility and cytokine secretion in vitro, Cells Tissues Organs. 198 (2014) 327–337. doi: 10.1159/000356731. [DOI] [PubMed] [Google Scholar]
  • [97].Kansy BA, Dißmann PA, Hemeda H, Bruderek K, Westerkamp AM, Jagalski V, Schuler P, Kansy K, Lang S, Dumitru CA, Brandau S, The bidirectional tumor - mesenchymal stromal cell interaction promotes the progression of head and neck cancer., Stem Cell Res. Ther. 5 (2014) 95. doi: 10.1186/scrt484. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [98].Ji X, Zhang Z, Han Y, Song J, Xu X, Jin J, Su S, Mu D, Liu X, Xu S, Cui H, Zhao Z, Wang Y, Liu H, Mesenchymal stem cells derived from normal gingival tissue inhibit the proliferation of oral cancer cells in vitro and in vivo, Int. J. Oncol. 49 (2016) 2011–2022. doi: 10.3892/ijo.2016.3715. [DOI] [PubMed] [Google Scholar]
  • [99].Couper KN, Blount DG, Riley EM, IL-10: The Master Regulator of Immunity to Infection, J. Immunol. 180 (2008) 5771–5777. doi: 10.4049/jimmunol.180.9.5771. [DOI] [PubMed] [Google Scholar]
  • [100].Wang J, Yang W, Wang T, Chen X, Wang J, Zhang X, Cai C, Zhong B, Wu J, Chen Z, Xiang AP, Huang W, Mesenchymal Stromal Cells-Derived β2-Microglobulin Promotes Epithelial-Mesenchymal Transition of Esophageal Squamous Cell Carcinoma Cells, Sci. Rep. 8 (2018). doi: 10.1038/s41598-018-23651-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [101].Wu YL, Li HY, Zhao XP, Jiao JY, Tang DX, Yan LJ, Wan Q, Bin Pan C, Mesenchymal stem cell-derived CCN2 promotes the proliferation, migration and invasion of human tongue squamous cell carcinoma cells, Cancer Sci. 108 (2017) 897–909. doi: 10.1111/cas.13202. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [102].Sharaf K, Kleinsasser A, Schwenk-Zieger S, Gires O, Schinke H, Kohlbauer V, Jakob M, Canis M, Haubner F, Molecular Characterization of Lipoaspirates Used in Regenerative Head and Neck Surgery, JAMA Facial Plast. Surg. 21 (2019) 526–534. doi: 10.1001/jamafacial.2019.0851. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [103].Böhrnsen F, Godek F, Kiesel J, Kramer FJ, Brockmeyer P, Schliephake H, Influence of TGF-β1 on tumor transition in oral cancer cell and BMSC co-cultures, J. Cranio-Maxillofacial Surg. 45 (2017) 731–740. doi: 10.1016/j.jcms.2017.02.009. [DOI] [PubMed] [Google Scholar]
  • [104].Ma H, Zhang M, Qin J, Probing the role of mesenchymal stem cells in salivary gland cancer on biomimetic microdevices, Integr. Biol. 4 (2012) 522–530. doi: 10.1039/c2ib20026k. [DOI] [PubMed] [Google Scholar]
  • [105].Coccè V, Farronato D, Brini AT, Masia C, Giannì AB, Piovani G, Sisto F, Alessandri G, Angiero F, Pessina A, Drug Loaded Gingival Mesenchymal Stromal Cells (GinPa-MSCs) Inhibit In Vitro Proliferation of Oral Squamous Cell Carcinoma, Sci. Rep. 7 (2017). doi: 10.1038/s41598-017-09175-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [106].Wang J, Cui R, Clement CG, Nawgiri R, Powell DW, Pinchuk IV, Watts TL, Activation PDGFR-α/AKT Mediated Signaling Pathways in Oral Squamous Cell Carcinoma by Mesenchymal Stem/Stromal Cells Promotes Anti-apoptosis and Decreased Sensitivity to Cisplatin, Front. Oncol. 10 (2020) 552. doi: 10.3389/fonc.2020.00552. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [107].Liu C, Billet S, Choudhury D, Cheng R, Haldar S, Fernandez A, Biondi S, Liu Z, Zhou H, Bhowmick NA, Bone marrow mesenchymal stem cells interact with head and neck squamous cell carcinoma cells to promote cancer progression and drug resistance, Neoplasia. 23 (2021) 118–128. doi: 10.1016/J.NEO.2020.11.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [108].Liu C, Feng X, Wang B, Wang X, Wang C, Yu M, Cao G, Wang H, Bone marrow mesenchymal stem cells promote head and neck cancer progression through Periostin-mediated phosphoinositide 3-kinase/Akt/mammalian target of rapamycin, Cancer Sci. 109 (2018) 688–698. doi: 10.1111/cas.13479. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [109].Jiang C, Zhang Q, Shanti RM, Shi S, Chang TH, Carrasco L, Alawi F, Le AD, Mesenchymal Stromal Cell-Derived Interleukin-6 Promotes Epithelial–Mesenchymal Transition and Acquisition of Epithelial Stem-Like Cell Properties in Ameloblastoma Epithelial Cells, Stem Cells. 35 (2017) 2083–2094. doi: 10.1002/stem.2666. [DOI] [PubMed] [Google Scholar]
  • [110].Li W, Han Y, Zhao Z, Ji X, Wang X, Jin J, Wang Q, Guo X, Cheng Z, Lu M, Wang G, Wang Y, Liu H, Oral mucosal mesenchymal stem cell-derived exosomes: A potential therapeutic target in oral premalignant lesions, Int. J. Oncol. 54 (2019) 1567–1578. doi: 10.3892/ijo.2019.4756. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [111].Shi S, Zhang Q, Xia Y, You B, Shan Y, Bao L, Li L, You Y, Gu Z, Mesenchymal stem cell-derived exosomes facilitate nasopharyngeal carcinoma progression, Am. J. Cancer Res. 6 (2016) 459–472. https://pubmed.ncbi.nlm.nih.gov/27186416/ (accessed April 29, 2020). [PMC free article] [PubMed] [Google Scholar]
  • [112].Li L, Tian H, Yue W, Zhu F, Li S, Li W, Human mesenchymal stem cells play a dual role on tumor cell growth in vitro and in vivo, J. Cell. Physiol. 226 (2011) 1860–1867. doi: 10.1002/jcp.22511. [DOI] [PubMed] [Google Scholar]
  • [113].Hong D, Liu T, Huang W, Liao Y, Wang L, Zhang Z, Chen H, Zhang X, Xiang Q, Gremlin1 delivered by mesenchymal stromal cells promoted epithelial-mesenchymal transition in human esophageal squamous cell carcinoma, Cell. Physiol. Biochem. 47 (2018) 1785–1799. doi: 10.1159/000491060. [DOI] [PubMed] [Google Scholar]
  • [114].Nakayama A, Aoki S, Uchihashi K, Nishijima-Matsunobu A, Yamamoto M, Kakihara N, Iwakiri R, Fujimoto K, Toda S, Interaction between Esophageal Squamous Cell Carcinoma and Adipose Tissue in Vitro, Am. J. Pathol. 186 (2016) 1180–1194. doi: 10.1016/j.ajpath.2016.01.003. [DOI] [PubMed] [Google Scholar]
  • [115].Castellone MD, Laatikainen LE, Laurila JP, Langella A, Hematti P, Soricelli A, Salvatore M, Laukkanen MO, Brief Report: Mesenchymal Stromal Cell Atrophy in Coculture Increases Aggressiveness of Transformed Cells, Stem Cells. 31 (2013) 1218–1223. doi: 10.1002/stem.1361. [DOI] [PubMed] [Google Scholar]
  • [116].Wang Y, Fan H, Zhou B, Ju Z, Yu L, Guo L, Han J, Lu S, Fusion of human umbilical cord mesenchymal stem cells with esophageal carcinoma cells inhibits the tumorigenicity of esophageal carcinoma cells, Int. J. Oncol. 40 (2012) 370–377. doi: 10.3892/ijo.2011.1232. [DOI] [PubMed] [Google Scholar]
  • [117].Liotta F, Querci V, Mannelli G, Santarlasci V, Maggi L, Capone M, Rossi MC, Mazzoni A, Cosmi L, Romagnani S, Maggi E, Gallo O, Annunziato F, Mesenchymal stem cells are enriched in head neck squamous cell carcinoma, correlates with tumour size and inhibit T-cell proliferation, Br. J. Cancer. 112 (2015) 745–754. doi: 10.1038/bjc.2015.15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [118].Mazzoni A, Capone M, Ramazzotti M, Vanni A, Locatello LG, Gallo O, De Palma R, Cosmi L, Liotta F, Annunziato F, Maggi L, IL4I1 Is Expressed by Head–Neck Cancer-Derived Mesenchymal Stromal Cells and Contributes to Suppress T Cell Proliferation, J. Clin. Med. 10 (2021) 2111. doi: 10.3390/JCM10102111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [119].Chen Y, Wang X, Fang J, Song J, Ma D, Luo L, He B, Xia J, Lui VWY, Cheng B, Wang Z, Mesenchymal stem cells participate in oral mucosa carcinogenesis by regulating T cell proliferation, Clin. Immunol. 198 (2019) 46–53. doi: 10.1016/j.clim.2018.12.001. [DOI] [PubMed] [Google Scholar]
  • [120].Schuler PJ, Westerkamp AM, Kansy BA, Bruderek K, Dissmann PA, Dumitru CA, Lang S, Jackson EK, Brandau S, Adenosine metabolism of human mesenchymal stromal cells isolated from patients with head and neck squamous cell carcinoma, Immunobiology. 222 (2017) 66–74. doi: 10.1016/j.imbio.2016.01.013. [DOI] [PubMed] [Google Scholar]
  • [121].Zhang Z, Song J, Han Y, Mu D, Su S, Ji X, Liu H, Impairment of mesenchymal stem cells derived from oral leukoplakia, Int. J. Clin. Exp. Pathol. 8 (2015) 10026–10037. [PMC free article] [PubMed] [Google Scholar]
  • [122].Almeida RDAC, Andrade ESDS, Barbalho JC, Vajgel A, Vasconcelos BCDE, Recurrence rate following treatment for primary multicystic ameloblastoma: Systematic review and meta-analysis, Int. J. Oral Maxillofac. Surg. 45 (2016) 359–367. doi: 10.1016/j.ijom.2015.12.016. [DOI] [PubMed] [Google Scholar]
  • [123].Hendra FN, Van Cann EM, Helder MN, Ruslin M, de Visscher JG, Forouzanfar T, de Vet HCW, Global incidence and profile of ameloblastoma: A systematic review and meta-analysis, Oral Dis. 26 (2020) 12–21. doi: 10.1111/odi.13031. [DOI] [PubMed] [Google Scholar]
  • [124].Reichart PA, Philipsen HP, Sonner S, Ameloblastoma: Biological profile of 3677 cases, Eur. J. Cancer. Part B Oral Oncol. 31 (1995) 86–99. doi: 10.1016/0964-1955(94)00037-5. [DOI] [PubMed] [Google Scholar]
  • [125].Abels ER, Breakefield XO, Introduction to Extracellular Vesicles: Biogenesis, RNA Cargo Selection, Content, Release, and Uptake, Cell. Mol. Neurobiol. 36 (2016) 301–312. doi: 10.1007/s10571-016-0366-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [126].Franz M, Richter P, Geyer C, Hansen T, Acuña LD, Hyckel P, Böhmer FD, Kosmehl H, Berndt A, Mesenchymal cells contribute to the synthesis and deposition of the laminin-5 γ2 chain in the invasive front of oral squamous cell carcinoma, J. Mol. Histol. 38 (2007) 183–190. doi: 10.1007/s10735-007-9086-5. [DOI] [PubMed] [Google Scholar]
  • [127].Tlsty TD, Coussens LM, Tumor Stroma and Regulation of Cancer Development, Annu. Rev. Pathol. Mech. Dis. 1 (2006) 119–150. doi: 10.1146/annurev.pathol.1.110304.100224. [DOI] [PubMed] [Google Scholar]
  • [128].Allard B, Longhi MS, Robson SC, Stagg J, The ectonucleotidases CD39 and CD73: Novel checkpoint inhibitor targets, Immunol. Rev. 276 (2017) 121–144. doi: 10.1111/imr.12528. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [129].Feng LL, Cai YQ, Zhu MC, Xing LJ, Wang X, The yin and yang functions of extracellular ATP and adenosine in tumor immunity, Cancer Cell Int. 20 (2020) 1–11. doi: 10.1186/s12935-020-01195-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [130].B. L, N. R, S. MA, F. GE, S. R, M. J, B. L, S. H, A. S, S. RC, D. YA, Cancer-Associated Fibroblasts Share Characteristics and Protumorigenic Activity with Mesenchymal Stromal Cells, Cancer Res. 77 (2017) 5142–5157. doi: 10.1158/0008-5472.CAN-16-2586. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [131].Ping Q, Yan R, Cheng X, Wang W, Zhong Y, Hou Z, Shi Y, Wang C, Li R, Cancer-associated fibroblasts: overview, progress, challenges, and directions, Cancer Gene Ther. 2021 289. 28 (2021) 984–999. doi: 10.1038/s41417-021-00318-4. [DOI] [PubMed] [Google Scholar]
  • [132].Bienkowska KJ, Hanley CJ, Thomas GJ, Cancer-Associated Fibroblasts in Oral Cancer: A Current Perspective on Function and Potential for Therapeutic Targeting, Front. Oral Heal. 0 (2021) 40. doi: 10.3389/FROH.2021.686337. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [133].Okada, Vaeteewoottacharn, Kariya, Application of Highly Immunocompromised Mice for the Establishment of Patient-Derived Xenograft (PDX) Models, Cells. 8 (2019) 889. doi: 10.3390/cells8080889. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [134].Rowan BG, Lacayo EA, Sheng M, Anbalagan M, Gimble JM, Jones RK, Joseph WJ, Friedlander PL, Chiu ES, Human Adipose Tissue-Derived Stromal/Stem Cells Promote Migration and Early Metastasis of Head and Neck Cancer Xenografts, Aesthetic Surg. J. 36 (2015) 93–104. doi: 10.1093/asj/sjv090. [DOI] [PubMed] [Google Scholar]
  • [135].Zielske SP, Livant DL, Lawrence TS, Radiation Increases Invasion of Gene-Modified Mesenchymal Stem Cells into Tumors, Int. J. Radiat. Oncol. Biol. Phys. 75 (2009) 843–853. doi: 10.1016/j.ijrobp.2008.06.1953. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [136].Tan TT, Lai RC, Padmanabhan J, Sim WK, Choo ABH, Lim SK, Assessment of Tumorigenic Potential in Mesenchymal-Stem/Stromal-Cell-Derived Small Extracellular Vesicles (MSC-sEV), Pharm. 2021, Vol. 14, Page 345. 14 (2021) 345. doi: 10.3390/PH14040345. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [137].Kumar JD, Holmberg C, Kandola S, Steele I, Hegyi P, Tiszlavicz L, Jenkins R, Beynon RJ, Peeney D, Giger OT, Alqahtani A, Wang TC, Charvat TT, Penfold M, Dockray GJ, Varro A, Increased expression of chemerin in squamous esophageal cancer myofibroblasts and role in recruitment of mesenchymal stromal cells, PLoS One. 9 (2014). doi: 10.1371/journal.pone.0104877. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [138].Bruna F, Arango-Rodríguez M, Plaza A, Espinoza I, Conget P, The administration of multipotent stromal cells at precancerous stage precludes tumor growth and epithelial dedifferentiation of oral squamous cell carcinoma, Stem Cell Res. 18 (2017) 5–13. doi: 10.1016/j.scr.2016.11.016. [DOI] [PubMed] [Google Scholar]
  • [139].Bruna F, Plaza A, Arango M, Espinoza I, Conget P, Systemically administered allogeneic mesenchymal stem cells do not aggravate the progression of precancerous lesions: A new biosafety insight, Stem Cell Res. Ther. 9 (2018). doi: 10.1186/s13287-018-0878-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [140].Meng W, Wu Y, He X, Liu C, Gao Q, Ge L, Wu L, Liu Y, Guo Y, Li X, Liu Y, Chen S, Kong X, Liang Z, Zhou H, A systems biology approach identifies effective tumor-stroma common targets for oral squamous cell carcinoma, Cancer Res. 74 (2014) 2306–2315. doi: 10.1158/0008-5472.CAN-13-2275. [DOI] [PubMed] [Google Scholar]
  • [141].Rowan BG, Gimble JM, Sheng M, Anbalagan M, Jones RK, Frazier TP, Asher M, Lacayo EA, Friedlander PL, Kutner R, Chiu ES, Human adipose tissue-derived stromal/stem cells promote migration and early metastasis of triple negative breast cancer xenografts, PLoS One. 9 (2014). doi: 10.1371/journal.pone.0089595. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [142].Farah CS, Fox SA, Dysplastic oral leukoplakia is molecularly distinct from leukoplakia without dysplasia, Oral Dis. 25 (2019) 1715–1723. doi: 10.1111/odi.13156. [DOI] [PubMed] [Google Scholar]
  • [143].Ouchi T, Morikawa S, Shibata S, Takahashi M, Yoshikawa M, Soma T, Miyashita H, Muraoka W, Kameyama K, Kawana H, Arima Y, Saya H, Okano H, Nakagawa T, Asoda S, Recurrent Spindle Cell Carcinoma Shows Features of Mesenchymal Stem Cells, J. Dent. Res. 97 (2018) 779–786. doi: 10.1177/0022034518759278. [DOI] [PubMed] [Google Scholar]
  • [144].T.M. and Collins SWHM, Spectrum of matrix metalloproteinase expression in primary and metastatic colon cancer: relationship to the tissue inhibitors of metalloproteinases and membrane type-1-matrix metalloproteinase, Br. J. Cancer. 84 (2001) 1664–1670. doi: 10.1054/bjoc.2001.1831. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [145].Klein T, Bischoff R, Physiology and pathophysiology of matrix metalloproteases., Amino Acids. 41 (2011) 271–90. doi: 10.1007/s00726-010-0689-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [146].Hanahan D, Weinberg RA, Hallmarks of cancer: The next generation, Cell. 144 (2011) 646–674. doi: 10.1016/j.cell.2011.02.013. [DOI] [PubMed] [Google Scholar]
  • [147].Lu W, Kang Y, Epithelial-Mesenchymal Plasticity in Cancer Progression and Metastasis, Dev. Cell. 49 (2019) 361–374. doi: 10.1016/j.devcel.2019.04.010. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES