Next Article in Journal
Analysis of Relationships between Immune Checkpoint and Methylase Gene Polymorphisms and Outcomes after Unrelated Bone Marrow Transplantation
Next Article in Special Issue
Targeted Treatment of Head and Neck (Pre)Cancer: Preclinical Target Identification and Development of Novel Therapeutic Applications
Previous Article in Journal
Diversity of Epithelial-Mesenchymal Phenotypes in Circulating Tumour Cells from Prostate Cancer Patient-Derived Xenograft Models
Previous Article in Special Issue
De-Escalation Strategies of (Chemo)Radiation for Head-and-Neck Squamous Cell Cancers—HPV and Beyond
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Human Adipose-Derived Stem/Stromal Cells Promote Proliferation and Migration in Head and Neck Cancer Cells

1
Department of Otolaryngology, University Hospital, LMU Munich, Marchioninistrasse 15, 81377 Munich, Germany
2
Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Marchioninistrasse 15, 81377 Munich, Germany
3
Department of Gynecological Endocrinology and Reproductive Medicine, University Hospital of Schleswig-Holstein, 23562 Lübeck, Germany
4
Clinical Cooperation Group “Personalized Radiotherapy in Head and Neck Cancer“, Helmholtz Zentrum München, 85764 Neuherberg, Germany
*
Authors to whom correspondence should be addressed.
Cancers 2021, 13(11), 2751; https://doi.org/10.3390/cancers13112751
Submission received: 2 May 2021 / Revised: 25 May 2021 / Accepted: 27 May 2021 / Published: 1 June 2021

Abstract

:

Simple Summary

Fat grafts obtained from a minimal invasive liposuction device contain multipotent stem cells termed adipose-derived stem/stromal cells (ASCs). ASCs can be used for their proposed wound healing relevant characteristics, including for tissue defects in cancer patients. For head and neck cancers, little is known about the effects of ASCs on tumor cells. Using supernatants of ASCs from five patients in different functional experiments, this study aimed to investigate how ASCs influence tumor growth, invasive properties, and neoangiogenesis. The data show that all mentioned characteristics are promoted by fat graft stem cells in vitro in head and neck cancer cell lines. Although clinical relevance of these in vitro findings is unclear, due to the lack of in vivo and clinical data, fat grafts should be used cautiously and complete removal of tumor should be ensured before augmentation in head and neck cancer patients is performed.

Abstract

Human adipose-derived stem/stromal cells (ASCs) are increasingly used as auto-transplants in regenerative medicine to restore tissue defects or induce wound healing, especially in cancer patients. The impact of ASCs on squamous cell carcinoma of the upper aerodigestive tract (UAT) including head and neck and esophageal squamous cell carcinoma (HNSCC and ESCC) is not yet fully understood. ASCs were cultured from subcutaneous, abdominal lipoaspirates of five patients, who received auto-transplants to the head and neck. Supernatants were tested for paracrine effects in functional in vitro assays of proliferation of HNSCC tumor cell line FaDu and ESCC cell line Kyse30, and their cell migration/invasion capacities in Boyden chambers, in addition to endothelial tube formation assay using human umbilical vein endothelial cells (HUVECs). All ASC-derived supernatants enhanced proliferation of FaDu cells, invasive migration, and tube formation by HUVECs, compared to controls. Of five patients’ lipoaspirates, ASC-derived supernatants of four patients increased proliferation and invasive migration in Kyse30 cells. The data suggests that ASCs can promote tumor cell proliferation, invasiveness, and neo-angiogenesis in these tumor cell lines of the UAT and HUVEC in a paracrine manner. Although clinical studies on the subject of oncological safety are still needed, these findings emphasize the importance of complete tumor removal before ASCs are used in the head and neck.

1. Introduction

Stem cell-based therapies offer a new approach for the repair of volume defects and support wound healing. The work with either embryonic stem cells or induced pluripotent stem cells is highly restricted in the clinical setting in most countries due to regulations and ethical discourse, even though these cells are theoretically highly advantageous [1,2]. Mesenchymal stromal cells (MSCs) are non-hematopoietic, pluripotent progenitor cells that have been shown to reside in various tissues and organs, such as adipose tissue, umbilical cord matrix, synovial membrane, and dental tissues [3,4]. Because the isolation of MSCs from adipose tissue is comparably uncomplicated, adipose-derived stem cells (ASCs, also adipose-derived stromal cells) are one of the most widely used types of stem cells in clinical application to date [5]. Adipose tissue has been used for tissue augmentation since 1893, first performed by the German surgeon Neuber to fill a depressed scar on the face [6].
ASCs are localized in the perivascular niche of adipose tissues and are considered a suitable source for stem cell-based therapies, mainly because they can be collected in significant quantities during surgical fat transfer or minimally invasive procedures with techniques such as lipoaspiration [7,8,9]. In contrast to, e.g., breast augmentation in plastic surgery, which uses large amounts of lipoaspirates, only small volumes are usually required in head and neck surgery [9,10]. The volumes of lipoaspirates that are needed for procedures such as vocal fold augmentation typically amount to a few milliliters [11]. ASCs are part of the stromal vascular fraction that is extracted after enzymatic digestion (or mechanical dissociation) and centrifugation of adipose tissue [12,13].
The wound healing properties of ASCs are mostly attributed to their paracrine effects [13,14]. Soluble growth factors and cytokines secreted by ASCs, such as Vascular Endothelial Growth Factor (VEGF) and Transforming Growth Factor Beta 3 (TGF-β3), modulate various aspects of the wound healing process such as neovascularization, homing of stromal cells, and the inhibition of fibrosis [13,15]. Some experimental studies suggest that ASCs are responsible for tumorigenesis and tumor development promoted by inflammatory cytokines that are secreted from the ASCs [16,17,18]. Further studies hypothesized that adipocyte and ASC secretions can stimulate tumor cell growth and that tumor–stroma interaction can potentially trigger cancer recurrence by activating residual cancer cells in the tumor bed [19,20].
Generally, adipose tissue transplantation is considered a safe procedure in different clinical settings, especially in cancer patients [21,22,23]. Head and neck squamous cell carcinoma (HNSCC) patients have frequent wound healing difficulties in many stages of their diseases [11,24]. Although ASCs facilitate wound healing, they may accelerate recurrence of cancer when applied to a localization with residual tumor cells [22]. Women with a history of breast cancer often desire reshaping or breast augmentation after surgery and autologous fat grafting is an established and frequently performed therapy in these patients [25]. Therefore, many studies have focused on oncological safety of autologous fat grafting in breast cancer patients and few studies have tried to examine effects and safety in other tumor entities [26,27]. To date, the data on the interaction of ASCs and breast cancer cells, and the oncological safety of ASCs in breast cancer patients, remain inconclusive [18,25,27,28,29,30,31]. Although many experimental studies showed that ASCs were able to counteract cancer therapy or promote tumorigenesis in breast cancer cells, meta-analysis of clinical studies found no differences in recurrence rates of breast cancer in patients treated with grafts or not [25,29,30]. For bladder cancer, in vitro data has shown that ASCs might promote proliferation and invasive properties of cancer cells [32].
To date, only eight studies have linked MSCs/ASCs and HNSCC [24,33,34,35,36,37,38,39]. Grønhøj et al. evaluated the salivary gland function after ASC injection in patients with radiation-induced xerostomia after HPV-related oropharyngeal squamous cell carcinoma, but oncological safety was not an endpoint [24]. Xia et al. concluded that gingiva-derived MSCs inhibit tumorigenesis of oral cavity squamous cell carcinoma of the tongue [33]. Chiu et al. tested paracrine effects of ASC-conditioned medium on oral squamous cell carcinoma and found that it might protect cancer cells from cisplatin-induced cell death but did not find alterations in tumor cell growth and migration [34]. In different in vitro approaches and an in vivo xenograft model, Danan et al. found that ASCs did not significantly stimulate HNSCC cell proliferation or migration, and only increased survival in one single cancer cell line tested [35]. Rowan et al. did not find alterations in growth of HNSCC cells in vitro or tumor xenografts ex vivo, but found stimulation of cancer cell migration, integration of ASCs to tumor–stroma, and early micrometastasis in vivo [36]. Using bone marrow-derived MSCs of different donors and different HNSCC cell lines, the data of Wessely et al. suggested increased invasion properties and prometastatic effects of MSCs on HNSCC [37]. In the most recent study and in agreement with an older study of their group, Sinha et al. found that bone marrow-derived MSCs but not ASCs contribute to tongue squamous cell carcinoma progression [38,39].
In summary, these studies show an inhomogeneous state of knowledge regarding the role of ASCs in HNSCC progression. It remains unclear how ASCs and HNSCC cells interact and how underlying mechanisms might facilitate tumor progression in HNSCC. In this project, we aimed to characterize the paracrine effects of ASCs on representative cancer cell lines of the UAT. Therefore, we performed experiments with supernatants from ASCs that were acquired from lipoaspirate samples of five patients and tested their influence on HNSCC cell line FaDu and ESCC cell line Kyse30. To gain further insights in the interaction of ASC and UAT cancers, different assays were performed to assess tumor cell proliferation, invasive properties, and the ability to grow new blood vessels under the influence of ASCs in vitro.

2. Materials and Methods

2.1. Patient Characteristics

Patients included in this study underwent elective autologous fat grafting to the head and neck area (n = 5). General patient characteristics of these five patients are summarized in Table S1.
All samples were obtained after written informed consent during routine surgery. The study was conducted according to the guidelines of the Declaration of Helsinki and approved by the institutional ethics committee of the local medical faculty (Ethikkommission der Medizinischen Fakultät der Ludwig-Maximilians-Universität, IRB approval number 17-567).

2.2. Acquistion of Lipoaspirates

Lipoaspirates were obtained from the abdominal fat tissue (n = 5) with a thin (2 mm diameter) liposuction cannula (Spiggle&Theis, Overath, Germany) as described previously [9]. Lipoaspirates were then centrifugated for 5 min at 3000 rpm and the resulting oily and saline fractions were discarded, and the stromal vascular fraction was transferred to 1 mL syringes for injection into the augmentation site [9]. A small amount was transferred to the laboratory facilities.

2.3. Explant Cultivation and Isolation of Adipose-Derived Mesenchymal Stem Cells

As described previously [40], lipoaspirate tissue was washed three times with 37 °C pre-warmed PBS (Apotheke Klinikum der Universität München, Munich, Germany) and centrifuged for 7 min at 900 rpm (Varifuge 3.OR, Heraeus, Hanau, Germany, rotor diameter = 21.5 cm). The resulting liquid phase and oily supernatants were discarded. The remaining cellular fraction was plated on 100 mm tissue culture dishes (Cell+, Sarstedt, Nürmbrecht, Germany). Approximately 1 mL of lipoaspirate was plated per dish. For better attachment, the tissue was left for 5 min before carefully adding 2.5 mL of pre-warmed Mesenchymal Stem Cell Growth Medium 2 (MSCM 2, PromoCell, Heidelberg, Germany) to each tissue culture plate. Thereafter, plates were maintained at 37 °C in a humidified atmosphere with 5% CO2. On the third day of cultivation, another 2 mL of MSCM 2 was added. On the sixth day, the conditioned medium was collected, centrifuged, and frozen. Hereafter, the medium was changed three times per week. Between day 12 and 14, the tissue was carefully removed and tissue culture plates were washed thoroughly with pre-warmed PBS. To test the MSC characteristics as described before, immunophenotyping of these cultured cells and differentiation assays were performed [13].

2.4. Acquistion of ASC-Derived Supernatants

At a partial confluency of 80–90%, cells were detached using a 0.5% trypsin/0.2% EDTA solution (Biochrom GmbH, Berlin, Germany), and further cultivated in tissue culture flasks (Sarstedt). After meeting the criteria for MSC as described before, these cells were referred to as passage 0 ASCs and were passaged [13]. In passage 1, the conditioned media were collected after 72 h of cell culture, centrifuged, and frozen.

2.5. HNSCC Tumor Cell Lines and Proliferation Assay

FaDu and Kyse30 cell lines were obtained from Leibniz Institute German Collection of Microorganisms and Cell Culture (DSMZ). Cells were maintained in DMEM (FaDu) or RPMI 1640 (Kyse30), 10% FCS, 1% penicillin/streptomycin, in a 5% CO2 atmosphere at 37 °C. In the proliferation assay, 105 tumor cells were seeded in 6-well plates, washed with PBS after 24 h and incubated with ASC-derived supernatants from the five above mentioned individual patients as verum treatments or (1) supernatant from FaDu/Kyse30 with MSCM 2, (2) DMEM/RPMI without FCS, or (3) DMEM/RPMI with FCS as controls (1–3). After 72 h, cells were counted in an EVE automatic cell counter (Nano Entek, VWR, Munich, Germany). Supernatant from FaDu/Kyse30 with MSCM was defined as the baseline control for the ASC-derived supernatants, and therefore, all collected data were normalized to the mean of this control.

2.6. Cell Migration Assay

To investigate cell migration over a microporous membrane, a “Boyden chamber” assay with an 8 µm pore size (QCM 24-Well Colorimetric Cell Migration Assay, Merck, Darmstadt, Germany) was performed [41,42]. A quantity of 2.5–3.0 × 106 FaDu or Kyse30 cells in 300 µL DMEM/RPMI without FCS were placed into the upper inserts, and ASC-derived supernatants from the five above mentioned individual patients as verum treatments or (1) supernatant from FaDu/Kyse30 with MSCM 2, (2) DMEM/RPMI without FCS, or (3) DMEM/RPMI with FCS as controls (1–3) were placed in the lower wells and incubated for 24 h. After incubation for 24 h, fluid of the lower well was analyzed using a colorimeter (VersaMax Microplate Reader, Molecular Devices, San José, CA, USA).
The DMEM/RPMI without FCS control was used as a “blank control” and its resultant values were subtracted from the corresponding verum or control experiment results automatically. The supernatant from FaDu/Kyse30 with MSCM 2 control was defined as the baseline control for the ASC-derived supernatants, and, therefore, all data were normalized to the mean of this control.

2.7. Neo-Angionesis Assay

To mimic angiogenesis, a tube formation assay was performed as described previously [43,44]. Matrigel Matrix GFR (Corning, Fisher Scientific, Schwerte, Germany) was added to a µ-plate angiogenesis 96-well (ibidi, Gräfelfing, Germany) following the manufacturer’s application guide. Human umbilical vein endothelial cells (HUVECs, PromoCell, Heidelberg, Germany) in passage 3–5 were grown to 70–80% confluence, trypsinated with trypsin-EDTA (Thermo Scientific, Schwerte, Germany), and centrifuged at 300× g for 5  min. Subsequently, cells were resuspended in either the ASC-derived supernatants (cultured with MSCM 2), fresh Endothelial Cell Growth Medium MV (ECGM, PromoCell, Heidelberg, Germany), fresh MSCM 2 (PromoCell, Heidelberg, Germany), or ECGM with suramin (30 nM, PromoCell, Heidelberg, Germany). Then, 15,000 cells/well were seeded into an ibidi μ-slide angiogenesis 96-well plate and incubated at 37 °C, 5% CO2, and 95% humidity. Photographs of each well were taken after 20 h with a Leica DMi8 microscope (Leica, Wetzlar, Germany) using 5× magnification and phase contrast Ph (PLAN 5X/0.12 DRY). Each group consisted of at least three technical replicates of each experiment; results from three independent experiments are shown. ImageJ “Angiogenesis Analyzer” was used to analyze the photographs of the tube formation assay. Here, branches were identified and calculated.

2.8. Statistical Analysis

R version 3.6.0 was utilized for statistical analysis [45]. For each assay type, we performed a Kruskal–Wallis H test to determine if differences between verum and control groups were present (“kruskal.test” function, R “stats” package [45]), followed by Wilcoxon rank sum test-based pairwise comparisons with Benjamini Hochberg p-value adjustment (“wilcox.test” and “p.adjust” functions, R “stats” package [45]). p-values < 0.05 indicated statistical significance.

3. Results

3.1. ASC-Derived Supernatants Promote UAT Tumor Cell Proliferation In Vitro

Initially, we characterized the cell lines obtained from lipoaspirates of five patients and assessed their stem cell capacities as described before [13]. Three of the five cell lines had already been characterized previously [13]. Cells from all five patients were characterized as ASCs. The scheme used to characterize cell lines is shown in Figure 1.
To assess the paracrine function of these ASCs on UAT tumor cell proliferation, FaDu and Kyse30 cells were cultured with ASC-derived supernatants of the five patients or controls. The mean ± SD FaDu cell proliferation rate was approximately increased in a range of 2.10- to 2.92-fold compared to a matched negative control (Figure 2; Patient 1: 2.44 ± 0.10, patient 2: 2.10 ± 0.15, patient 3: 2.16 ± 0.26, patient 4: 2.92 ± 0.19, patient 5: 2.90 ± 0.25 vs. FaDu supernatant: 1.00 ± 0.07; all p < 0.01, Wilcoxon rank sum test). In addition, the FaDu cells cultured with positive controls also demonstrated increased proliferation rates (DMEM without FCS: 2.01 ± 0.34, DMEM with FCS: 4.64 ± 0.46 vs. FaDu supernatant; both p < 0.01, Wilcoxon rank sum test).
In Kyse30 cells, the mean ± SD proliferation rate was increased in the presence of ASC-derived supernatants in a range of 1.37- to 2.52-fold compared to a negative control (Figure 2; Patient 1: 1.98 ± 0.41, patient 2: 1.37 ± 0.16, patient 3: 1.53 ± 0.31, patient 4: 2.52 ± 0.31, patient 5: 2.07 ± 0.20 vs. Kyse30 supernatant: 1.00 ± 0.23; all p < 0.05, Wilcoxon rank sum test). As already seen in FaDu cells, the highest proliferation rate in Kyse30 was found in a positive control (RPMI with FCS), whereas the RPMI control without FCS showed no significant difference (RPMI with FCS: 4.90 ± 1.06 vs. Kyse30 supernatant, p = 0.01; RPMI without FCS: 1.19 ± 0.42 vs. Kyse30 supernatant, p = 0.47, Wilcoxon rank sum test).
Overall, HNSCC tumor cell proliferation was significantly increased following incubation with ASC supernatants from five different patients compared to the respective negative control conditions.

3.2. ASC-Derived Supernatants Promote Invasion of HNSCC Tumor Cells in a Functional In Vitro Assay

To investigate cell migration of HNSCC over an 8 µm microporous membrane, we performed a colorimetric Boyden chamber assay according to the proliferation experiments above. After subtraction of the negative control results (DMEM without FCS), the data was again normalized and compared to the tumor cell supernatant control.
Here, FaDu cells migrated more strongly towards all five ASC-derived supernatants and the positive control (DMEM with FCS) than towards FaDu cell supernatants (Figure 3; Patient 1: 4.25 ± 1.08, patient 2: 3.36 ± 0.92, patient 3: 4.33 ± 1.29, patient 4: 8.16 ± 1.46, patient 5: 4.29 ± 1.48, DMEM with FCS: 2.12 ± 0.60 vs. FaDu supernatant: 1.00 ± 0.54; all p < 0.01 except DMEM with FCS: p = 0.01, Wilcoxon rank sum test).
In Kyse30 experiments, cancer cells showed higher migration towards four of the five ASC-derived supernatants and the positive control (RPMI with FCS) compared to migration towards Kyse30 supernatant (Figure 3; Patient 1: 1.49 ± 0.09, patient 2: 1.50 ± 0.13, patient 3: 1.33 ± 0.14, patient 4: 1.77 ± 0.13, patient 5: 1.04 ± 0.51, RPMI with FCS: 1.49 ± 0.17 vs. Kyse30 supernatant: 1.00 ± 0.07; all p < 0.01 except patient 5: p = 0.07, Wilcoxon rank sum test).
In conclusion, HNSCC tumor cells migrated more strongly towards ASC-derived supernatants than cognate controls. In concordance with the proliferation experiments, this effect was more pronounced in FaDu cells than Kyse30 cells.

3.3. ASC-Derived Supernatants Promote Neo-Angiogenesis in a Functional Endothelial Tube Formation Assay In Vitro

To mimic neo-angiogenesis, we assessed the influence of ASC-derived supernatants and respective controls on tube formation in a functional endothelial cell assay. HUVECs were incubated in the presence of ASC supernatants or the indicated controls and the number of branches during tube formation was assessed after 20 h. After subtraction of branch numbers of the negative control (ECGM with 30 nM Suramin), the resulting branch number data was normalized and compared to the MSCM2 control group mean.
Representative photographs of tube formation experiments are depicted in Figure 4. In comparison to the control group (MSCM 2), the mean ± SD number of branches in the tube formation assay was increased 2- to 3-fold when cultured with ASC-derived supernatants of the five patients, and increased approximately 1.5-fold when cultured with ECGM (Figure 4; Patient 1: 2.37 ± 0.54, patient 2: 2.31 ± 0.68, patient 3: 2.45 ± 1.02, patient 4: 2.21 ± 0.43, patient 5: 1.83 ± 0.66, ECGM: 1.48 ± 0.36 vs. MSCM 2: 1.00 ± 0.21; Patient 1–4: p <0.001, patient 5 and ECGM: p < 0.01, Wilcoxon rank sum test).
In conclusion, HUVECs built more branches in a functional tube formation assay under the influence of ASC-derived supernatants than various cell mediums including growth-promoting supplements.

4. Discussion

Generally, adipose tissue transplantation is used successfully in various indications, e.g., wound healing, cardiovascular, and even joint disorders [46]. In the head and neck area, adipose tissue transplantation belongs to the most common topical stem cell therapies [11,47]. For augmentation of volume defects and treatment of impaired wound healing, adipose tissue transplantation is an established regenerative and reconstructive approach in head and neck cancer patients [48]. Therefore, ASCs are injected into tissues that might still contain residual tumor cells; in the head and neck area, these are most likely from squamous cell carcinoma [35,49]. In a literature search using the search terms “ASC AND HNSCC”, “adipose stem cells AND HNSCC”, “MSC AND HNSCC” and “mesenchymal stem cells AND HNSCC”, we found only six studies that examined the role of ASCs on HNSCC progression [33,34,35,36,37,38]. Their results are controversial and support both the notion that ASCs do support HNSCC progression and the opposing assumption that ASCs do not alter HNSCC progression. Our literature search of ASC/MSC and ESCC (search terms esophageal cancer AND ASC/adipose stem cells/MSC/mesenchymal stem cells) did not find a study that directly investigated ASC/ESCC interactions. The closest example was a study that showed tumor-promoting properties of adipose tissue on ESCC [50].
The present study focused on the effects of ASCs on two different cancer cell lines: FaDu cells represent a cell line gained from HNSCC of the hypopharynx with an estimated doubling time of 50 h; Kyse30 cells are derived from an esophageal well differentiated squamous cell carcinoma (doubling time 30 h). Both are well-characterized squamous cell carcinoma cell lines and represent established cancer cell lines of the UAT [51,52].
According to our data, ASC-derived supernatants promote tumor cell proliferation of FaDu and Kyse30 cells using conditioned media from ASC. Overall, tumor cells grew significantly faster than under respective negative control conditions in all combinations of two different cancer cell lines and ASC-derived supernatants obtained from five different patients. Although our results on tumor cell proliferation are unambiguous, in contrast, none of the other studies found increased HNSCC proliferation when ASC-conditioned medium or ASCs were added [34,35,36,38], except for increased colony formation only in SCC9 cells [35]. Although not testing directly, Nakayama et al. hypothesized that adipose tissue might increase ESCC tumor growth and decrease apoptosis of ESCC [50]. Although it remains unclear why most experiments did not show increased tumor cell growth in HNSCC, our experiments with ESCC cell line Kyse30 support the hypothesis of Nakayama.
The promotion of cell growth and invasiveness are typical attributes of aggressive tumor behavior. This behavior was shown in a study by Su et al., who examined tumor-promoting effects of ASCs on prostate cancer cells [53]. To evaluate the invasion capacity of HNSCC tumor cells stimulated with ASC supernatants, a migration assay using microporous membranes was used. This assay revealed that ASC-derived supernatants promote migration of HNSCC tumor cells in vitro towards the ASC-derived supernatants. This finding is of outmost importance with respect to dormant residual tumor cells, which might be re-activated by the ASC secretome. According to proliferation experiments of our study, this effect was more pronounced in FaDu cells than Kyse30 cells. This might result from the different doubling times of the cell lines with the doubling time of Kyse30 cells already being short and, therefore, less prone to the impact of the ASC secretome [51,52].
In the literature about HNSCC–ASC interactions, pro-migratory effects of the ASC secretome were found in the study of Rowan et al. [36], whereas three other studies did not find effects on migration or invasiveness [34,35,38]. However, there are findings that indicate carcinogenic effects of ASCs in those afore-mentioned studies: ASC-conditioned medium triggered chemoresistance of SCC25 and Cal27 cells, possibly through upregulation of the IGF-1R/AKT/ERK signaling pathway [34]. Also using Cal27 cells, Rowan et al. showed that ASCs were built into the tumor–stroma when Cal27 cells and ASCs were co-injected into the flank of nude mice and promoted HNSCC micrometastases to the murine brain [36]. Moreover, our results on invasiveness are similar to results of groups that had studied interactions of bone marrow-derived MSCs and HNSCC [37,39], and bone marrow-derived MSCs and ESCC [54,55,56].
In general, various studies have discussed the safety of ASC applications in cancer patients because the pursued regenerative properties of MSCs might promote tumor relapse [22]. Therefore, another important finding of our study is that ASC-derived supernatants promote neo-angiogenesis in a functional endothelial cell tube formation assay, in addition to their tumorigenic effects on HNSCC. Angiogenesis and neovascularization are highly complex processes, regulated by multiple players. Vascular endothelial growth factor (VEGF) is a central component in this context because it regulates neovascularization by targeting endothelial cell migration and proliferation [57]. In a previous study of our group, we found elevated VEGF levels in the secretome of ASCs and were able to confirm this finding in the five ASC cultures used in this work [13]. VEGF and other soluble factors of the secretome likely explain the pro-angiogenetic effects of ASCs. In addition to the paracrine effects of ASCs on angiogenesis, the literature also describes direct differentiation of ASCs into endothelial cells [58]. The exact mechanisms depend most likely on the maturation and stage of differentiation of ASCs [59]. Regarding neo-vascularization in a potentially dormant tumor site independent from stem cell therapies, local and/or free flap surgery is a commonly accepted and well-studied reconstructive method, in which a well-vascularized tissue flap is in direct contact with the tumor bed and extensive local vascular remodeling occurs. Immediate autologous tissue flap reconstruction has not been correlated with increased recurrence rates, e.g., in breast cancer patients [60,61]. Therefore, increased local vascularization at the site of a tumor bed is not necessarily an independent risk factor for recurrence.
It is important to note that the current body of literature emphasizes the high potential of ASCs in wound healing and regeneration. Multiple studies showed enhanced vascularization and improved wound healing after the treatment of skin wounds with ASC applications [62,63]. Highly promising findings for ASC application are also described in the context of spinal cord regeneration and recovery after partial hepatectomy [64,65]. Mostly, the paracrine effects of soluble factors that are secreted from ASCs are thought to be accountable for these effects, but direct cell interactions might also play an important role. Recently, it has been shown that ASCs, cultured in supernatant rich in growth factors, had enhanced specific binding capacity on selected cancer cells of fibrosarcoma and glioblastoma [66]. Menezes et al. observed that the presence of ASCs and not only their conditioned medium is necessary to build new maturated vessels [65]. Most likely, not only pro-angiogenetic factors such as VEGF, but also other growth factors are necessary to achieve tissue regeneration. ASCs might also execute immuno-suppressive effects in direct cell–cell interactions. Our group found that many ASCs express CD273/PD-L2. which plays a role in negative regulation of the adaptive immune response [13,67]. In HNSCC patients, in particular, functional and aesthetic reconstruction is challenging. Augmentation and regenerative therapy with cells that are capable of mediating rapid vascularization of grafts is highly appreciated. According to the present literature, mesenchymal stem cells promote regeneration, in addition to tumor cell growth of high-grade active tumors. Whether dormant tumor cells can be activated by ASCs in vivo remains unclear [61].
We performed the experiments of this study with two immortalized established cancer cell lines. The data show aspects of the interaction of active, proliferating tumor cells and ASC supernatants in vitro. Therefore, there is a limitation with respect to the comparability to the in vivo setting of lipotransfer after tumor resection. In this context, it is hard to prove whether dormant tumor cells can be activated by ASCs or their secretome. Other studies that used direct co-culture experiments of ASCs and HNSCC cells did not show increased migration or invasiveness in in vitro scratch assays and spheroid assays [35,38]. In this context, labeling of cells might influence cell migration and also only partially mimic the complex clinical setting with more cell types, e.g., immune cells involved. Another important limitation of this study and previous studies performed on the interaction of MSCs or ASCs and tumor cells is the lack of patient-specific tumor cell lines. Establishing both primary tumor cell and ASC cell lines from individual patients receiving tumor surgery and lipotransfer would help to overcome some of the limitations of the current studies.
Moreover, observational studies regarding the oncological safety of ASCs of lipotransfer procedures to the head and neck in head and neck cancer patients are necessary, e.g., in the framework of an augmentation of the vocal cord in laryngeal cancer. In such studies, cancer-specific outcome measures, such as the risk of relapse or disease-free survival, are necessary to evaluate the specific risk of stem cell applications in head and neck cancer patients. To date, most studies of autologous lipotransfer to the head and neck have focused on technical safety, efficacy in augmentation, and patient and surgeon satisfaction, and have relatively short periods of observational follow-up [68]. When adipose tissue transplantation is planned in cancer patients, the existence of residual or dormant squamous cell carcinoma cells should be at least excluded with intraoperative frozen section of surgical margins or, if it remains unclear, observation for locoregional recurrence after several months, e.g., 6 or 12 months, or an intraoperative frozen section.

5. Conclusions

Our data unambiguously show that ASC-conditioned media of different patients promote both tumor cell proliferation and migration of established cancer cell lines of the UAT, in addition to neo-angiogenesis of HUVECs in vitro. Therefore, ASCs are likely able to foster tumor growth and disease progression in squamous cell carcinoma via paracrine mechanisms. Although the literature indicates the topic remains controversial, this study highlights the potential tumor-promoting role of adipose tissue transplantation in head and neck cancer patients. However, further clinical trials are needed to evaluate the interaction of adipose-derived and other mesenchymal cells, and residual or dormant squamous cell carcinoma cells.

Supplementary Materials

The following are available online at https://www.mdpi.com/article/10.3390/cancers13112751/s1; Table S1: Patient characteristics.

Author Contributions

Conceptualization, K.S. and F.H.; methodology, K.S., F.H., S.S.-Z., J.Z., O.G., A.L.; software, K.S., S.P.H., J.Z.; validation, T.K.E., S.P.H., S.S.-Z., J.Z., O.G., A.L., M.C., K.S., F.H.; formal analysis, S.P.H., S.S.-Z., J.Z., K.S.; investigation, S.S.-Z., J.Z.; resources, K.S., F.H., O.G., M.C.; data curation, K.S., S.P.H., T.K.E., S.S.-Z., J.Z., F.H.; writing—original draft preparation, K.S., T.K.E., A.L., F.H.; writing—review and editing, S.P.H., S.S.-Z., J.Z., O.G., M.C.; visualization, S.P.H., J.Z., K.S.; supervision, K.S., F.H., O.G., M.C.; project administration, K.S., F.H., O.G.; funding acquisition, K.S., F.H.. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Friedrich-Baur-Stiftung, grant number 35/19 to KS and partly by funding from the Deutsche Forschungsgemeinschaft, grant number Gi 540/3-2 to OG. APC were supported in part by a grant from Spiggle & Theis Medizintechnik GmbH.

Institutional Review Board Statement

The study was conducted according to the guidelines of the Declaration of Helsinki and approved by the Institutional Ethics Committee of the local medical faculty (Ethikkommission der Medizinischen Fakultät der Ludwig-Maximilians-Universität, IRB approval number 17-567, amendment from 23 October 2018).

Informed Consent Statement

Informed consent was obtained during routine surgery from all subjects involved in the study.

Data Availability Statement

The data presented in this study are available on request from the corresponding authors. The data are not publicly available due to restrictions of the institutional IRB statement in concordance to European/German legislation on data restriction.

Acknowledgments

We want to thank Antonia Kleinsasser for her assistance in the grand proposal at Friedrich-Baur-Stiftung and in the preliminary experiments of this study.

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, or in the decision to publish the results.

References

  1. Liu, G.; David, B.T.; Trawczynski, M.; Fessler, R.G. Advances in Pluripotent Stem Cells: History, Mechanisms, Technologies, and Applications. Stem Cell Rev. Rep. 2020, 16, 3–32. [Google Scholar] [CrossRef] [Green Version]
  2. Rajabzadeh, N.; Fathi, E.; Farahzadi, R. Stem cell-based regenerative medicine. Stem Cell Investig. 2019, 6, 19. [Google Scholar] [CrossRef] [PubMed]
  3. Ullah, I.; Subbarao, R.B.; Rho, G.J. Human mesenchymal stem cells—Current trends and future prospective. Biosci. Rep. 2015, 35. [Google Scholar] [CrossRef]
  4. Mushahary, D.; Spittler, A.; Kasper, C.; Weber, V.; Charwat, V. Isolation, cultivation, and characterization of human mesenchymal stem cells. Cytom. Part A J. Int. Soc. Anal. Cytol. 2018, 93, 19–31. [Google Scholar] [CrossRef] [Green Version]
  5. Sheykhhasan, M.; Wong, J.K.L.; Seifalian, A.M. Human Adipose-Derived Stem Cells with Great Therapeutic Potential. Curr. Stem Cell Res. Ther. 2019, 14, 532–548. [Google Scholar] [CrossRef]
  6. Neuber, G.A. Fett transplantation. Verl. Dtsch. Ges. Chir. 1893, 22, 66. [Google Scholar]
  7. De Francesco, F.; Ricci, G.; D’Andrea, F.; Nicoletti, G.F.; Ferraro, G.A. Human Adipose Stem Cells: From Bench to Bedside. Tissue Eng. Part B Rev. 2015, 21, 572–584. [Google Scholar] [CrossRef] [Green Version]
  8. Goldman, J.J.; Wang, W.Z.; Fang, X.H.; Williams, S.J.; Baynosa, R.C. Tumescent Liposuction without Lidocaine. Plast. Reconstr. Surg. Glob. Open 2016, 4, e829. [Google Scholar] [CrossRef] [PubMed]
  9. Haubner, F.; Lorenz, A.; Kummer, P.; Alvarez, J.C.P. A new atraumatic device for liposuction and injection represents a technical advance for primary fat injection after cordectomy. Laryngo Rhino Otol. 2018, 97, 163–165. [Google Scholar] [CrossRef]
  10. Delay, E.; Savu, T.; Atanasiu, M. Lipomodeling in breast reconstruction. Ann. Chir. Plast. Esthet. 2018, 63, 505–515. [Google Scholar] [CrossRef] [PubMed]
  11. Fishman, J.M.; Long, J.; Gugatschka, M.; de Coppi, P.; Hirano, S.; Hertegard, S.; Thibeault, S.L.; Birchall, M.A. Stem cell approaches for vocal fold regeneration. Laryngoscope 2016, 126, 1865–1870. [Google Scholar] [CrossRef]
  12. Corselli, M.; Crisan, M.; Murray, I.R.; West, C.C.; Scholes, J.; Codrea, F.; Khan, N.; Péault, B. Identification of perivascular mesenchymal stromal/stem cells by flow cytometry. Cytom. Part A J. Int. Soc. Anal. Cytol. 2013, 83, 714–720. [Google Scholar] [CrossRef]
  13. 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 Fac. Plast. Surg. 2019, 21, 526–534. [Google Scholar] [CrossRef] [PubMed]
  14. Domenis, R.; Lazzaro, L.; Calabrese, S.; Mangoni, D.; Gallelli, A.; Bourkoula, E.; Manini, I.; Bergamin, N.; Toffoletto, B.; Beltrami, C.A.; et al. Adipose tissue derived stem cells: In vitro and in vivo analysis of a standard and three commercially available cell-assisted lipotransfer techniques. Stem Cell Res. Ther. 2015, 6, 2. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Guillén, M.I.; Platas, J.; Pérez Del Caz, M.D.; Mirabet, V.; Alcaraz, M.J. Paracrine Anti-inflammatory Effects of Adipose Tissue-Derived Mesenchymal Stem Cells in Human Monocytes. Front. Physiol. 2018, 9, 661. [Google Scholar] [CrossRef]
  16. Zhao, Y.; Zhang, X.; Zhao, H.; Wang, J.; Zhang, Q. CXCL5 secreted from adipose tissue-derived stem cells promotes cancer cell proliferation. Oncol. Lett. 2018, 15, 1403–1410. [Google Scholar] [CrossRef]
  17. Park, J.; Euhus, D.M.; Scherer, P.E. Paracrine and endocrine effects of adipose tissue on cancer development and progression. Endocr. Rev. 2011, 32, 550–570. [Google Scholar] [CrossRef] [Green Version]
  18. Zhao, R.; Kaakati, R.; Liu, X.; Xu, L.; Lee, A.K.; Bachelder, R.; Li, C.Y.; Hollenbeck, S.T. CRISPR/Cas9-Mediated BRCA1 Knockdown Adipose Stem Cells Promote Breast Cancer Progression. Plast. Reconstr. Surg. 2019, 143, 747–756. [Google Scholar] [CrossRef] [PubMed]
  19. Scioli, M.G.; Storti, G.; D’Amico, F.; Gentile, P.; Kim, B.S.; Cervelli, V.; Orlandi, A. Adipose-Derived Stem Cells in Cancer Progression: New Perspectives and Opportunities. Int. J. Mol. Sci. 2019, 20, 3296. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Wang, Y.; Liu, J.; Jiang, Q.; Deng, J.; Xu, F.; Chen, X.; Cheng, F.; Zhang, Y.; Yao, Y.; Xia, Z.; et al. Human Adipose-Derived Mesenchymal Stem Cell-Secreted CXCL1 and CXCL8 Facilitate Breast Tumor Growth by Promoting Angiogenesis. Stem Cells 2017, 35, 2060–2070. [Google Scholar] [CrossRef] [Green Version]
  21. Rigotti, G.; Marchi, A.; Galie, M.; Baroni, G.; Benati, D.; Krampera, M.; Pasini, A.; Sbarbati, A. Clinical treatment of radiotherapy tissue damage by lipoaspirate transplant: A healing process mediated by adipose-derived adult stem cells. Plast. Reconstr. Surg. 2007, 119, 1409–1422; discussion 1423–1404. [Google Scholar] [CrossRef] [PubMed]
  22. Zimmerlin, L.; Park, T.S.; Zambidis, E.T.; Donnenberg, V.S.; Donnenberg, A.D. Mesenchymal stem cell secretome and regenerative therapy after cancer. Biochimie 2013, 95, 2235–2245. [Google Scholar] [CrossRef] [PubMed]
  23. Chae, D.S.; Han, S.; Son, M.; Kim, S.W. Stromal vascular fraction shows robust wound healing through high chemotactic and epithelialization property. Cytotherapy 2017, 19, 543–554. [Google Scholar] [CrossRef] [PubMed]
  24. Grønhøj, C.; Jensen, D.H.; Vester-Glowinski, P.; Jensen, S.B.; Bardow, A.; Oliveri, R.S.; Fog, L.M.; Specht, L.; Thomsen, C.; Darkner, S.; et al. Safety and Efficacy of Mesenchymal Stem Cells for Radiation-Induced Xerostomia: A Randomized, Placebo-Controlled Phase 1/2 Trial (MESRIX). Int. J. Radiat. Oncol. Biol. Phys. 2018, 101, 581–592. [Google Scholar] [CrossRef]
  25. Agha, R.A.; Fowler, A.J.; Herlin, C.; Goodacre, T.E.; Orgill, D.P. Use of autologous fat grafting for breast reconstruction: A systematic review with meta-analysis of oncological outcomes. J. Plast. Reconstr. Aesthet. Surg. 2015, 68, 143–161. [Google Scholar] [CrossRef]
  26. Koellensperger, E.; Bonnert, L.C.; Zoernig, I.; Marmé, F.; Sandmann, S.; Germann, G.; Gramley, F.; Leimer, U. The impact of human adipose tissue-derived stem cells on breast cancer cells: Implications for cell-assisted lipotransfers in breast reconstruction. Stem Cell Res. Ther. 2017, 8, 121. [Google Scholar] [CrossRef] [Green Version]
  27. Mazur, S.; Zolocinska, A.; Siennicka, K.; Janik-Kosacka, K.; Chrapusta, A.; Pojda, Z. Safety of adipose-derived cell (stromal vascular fraction—SVF) augmentation for surgical breast reconstruction in cancer patients. Adv. Clin. Exp. Med. Off. Organ Wroclaw Med. Univ. 2018, 27, 1085–1090. [Google Scholar] [CrossRef] [Green Version]
  28. Bielli, A.; Scioli, M.G.; Gentile, P.; Cervelli, V.; Orlandi, A. Adipose Tissue-Derived Stem Cell Therapy for Post-Surgical Breast Reconstruction—More Light than Shadows. Adv. Clin. Exp. Med. Off. Organ Wroclaw Med. Univ. 2015, 24, 545–548. [Google Scholar] [CrossRef]
  29. Bougaret, L.; Delort, L.; Billard, H.; Le Huede, C.; Boby, C.; De la Foye, A.; Rossary, A.; Mojallal, A.; Damour, O.; Auxenfans, C.; et al. Adipocyte/breast cancer cell crosstalk in obesity interferes with the anti-proliferative efficacy of tamoxifen. PLoS ONE 2018, 13, e0191571. [Google Scholar] [CrossRef] [Green Version]
  30. Goto, H.; Shimono, Y.; Funakoshi, Y.; Imamura, Y.; Toyoda, M.; Kiyota, N.; Kono, S.; Takao, S.; Mukohara, T.; Minami, H. Adipose-derived stem cells enhance human breast cancer growth and cancer stem cell-like properties through adipsin. Oncogene 2019, 38, 767–779. [Google Scholar] [CrossRef]
  31. Scioli, M.G.; Artuso, S.; D’Angelo, C.; Porru, M.; D’Amico, F.; Bielli, A.; Gentile, P.; Cervelli, V.; Leonetti, C.; Orlandi, A. Adipose-derived stem cell-mediated paclitaxel delivery inhibits breast cancer growth. PLoS ONE 2018, 13, e0203426. [Google Scholar] [CrossRef] [Green Version]
  32. Maj, M.; Kokocha, A.; Bajek, A.; Drewa, T. The interplay between adipose-derived stem cells and bladder cancer cells. Sci. Rep. 2018, 8, 15118. [Google Scholar] [CrossRef] [Green Version]
  33. Xia, L.; Peng, R.; Leng, W.; Jia, R.; Zeng, X.; Yang, X.; Fan, M. TRAIL-expressing gingival-derived mesenchymal stem cells inhibit tumorigenesis of tongue squamous cell carcinoma. J. Dent. Res. 2015, 94, 219–228. [Google Scholar] [CrossRef] [PubMed]
  34. Chiu, Y.J.; Yang, J.S.; Hsu, H.S.; Tsai, C.H.; Ma, H. Adipose-derived stem cell conditioned medium attenuates cisplatin-triggered apoptosis in tongue squamous cell carcinoma. Oncol. Rep. 2018, 39, 651–658. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Danan, D.; Lehman, C.E.; Mendez, R.E.; Langford, B.; Koors, P.D.; Dougherty, M.I.; Peirce, S.M.; Gioeli, D.G.; Jameson, M.J. Effect of Adipose-Derived Stem Cells on Head and Neck Squamous Cell Carcinoma. Otolaryngol. Head Neck Surg. 2018, 158, 882–888. [Google Scholar] [CrossRef] [PubMed]
  36. Rowan, B.G.; Lacayo, E.A.; Sheng, M.; Anbalagan, M.; Gimble, J.M.; Jones, R.K.; Joseph, W.J.; Friedlander, P.L.; Chiu, E.S. Human Adipose Tissue-Derived Stromal/Stem Cells Promote Migration and Early Metastasis of Head and Neck Cancer Xenografts. Aesthet. Surg. J. 2016, 36, 93–104. [Google Scholar] [CrossRef] [PubMed]
  37. Wessely, A.; Waltera, A.; Reichert, T.E.; Stöckl, S.; Grässel, S.; Bauer, R.J. Induction of ALP and MMP9 activity facilitates invasive behavior in heterogeneous human BMSC and HNSCC 3D spheroids. Faseb J. 2019, 33, 11884–11893. [Google Scholar] [CrossRef] [Green Version]
  38. Sinha, S.; Narjus-Sterba, M.; Tuomainen, K.; Kaur, S.; Seppänen-Kaijansinkko, R.; Salo, T.; Mannerström, B.; Al-Samadi, A. Adipose-Derived Mesenchymal Stem Cells do not Affect the Invasion and Migration Potential of Oral Squamous Carcinoma Cells. Int. J. Mol. Sci. 2020, 21, 6455. [Google Scholar] [CrossRef] [PubMed]
  39. Salo, S.; Bitu, C.; Merkku, K.; Nyberg, P.; Bello, I.O.; Vuoristo, J.; Sutinen, M.; Vähänikkilä, H.; Costea, D.E.; Kauppila, J.H.; et al. Human bone marrow mesenchymal stem cells induce collagen production and tongue cancer invasion. PLoS ONE 2013, 8, e77692. [Google Scholar] [CrossRef]
  40. Hendijani, F. Explant culture: An advantageous method for isolation of mesenchymal stem cells from human tissues. Cell Prolif. 2017, 50. [Google Scholar] [CrossRef] [Green Version]
  41. Anitua, E.; Sanchez, M.; Merayo-Lloves, J.; De la Fuente, M.; Muruzabal, F.; Orive, G. Plasma rich in growth factors (PRGF-Endoret) stimulates proliferation and migration of primary keratocytes and conjunctival fibroblasts and inhibits and reverts TGF-beta1-Induced myodifferentiation. Invest. Ophthalmol. Vis. Sci. 2011, 52, 6066–6073. [Google Scholar] [CrossRef] [PubMed]
  42. Park, C.S.; Kim, O.S.; Yun, S.-M.; Jo, S.A.; Jo, I.; Koh, Y.H. Presenilin 1/γ-Secretase Is Associated with Cadmium-Induced E-Cadherin Cleavage and COX-2 Gene Expression in T47D Breast Cancer Cells. Toxicol. Sci. 2008, 106, 413–422. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Arnaoutova, I.; Kleinman, H.K. In vitro angiogenesis: Endothelial cell tube formation on gelled basement membrane extract. Nat. Protoc. 2010, 5, 628–635. [Google Scholar] [CrossRef]
  44. Kubota, Y.; Kleinman, H.K.; Martin, G.R.; Lawley, T.J. Role of laminin and basement membrane in the morphological differentiation of human endothelial cells into capillary-like structures. J. Cell Biol 1988, 107, 1589–1598. [Google Scholar] [CrossRef]
  45. R Development Core Team. R: A Language and Environment for Statistical Computing; R Foundation for Statistical Computing: Vienna, Austria, 2019. [Google Scholar]
  46. Andia, I.; Maffulli, N.; Burgos-Alonso, N. Stromal vascular fraction technologies and clinical applications. Expert Opin. Biol. Ther. 2019, 19, 1289–1305. [Google Scholar] [CrossRef]
  47. Hong, S.J.; Lee, S.H.; Jin, S.M.; Kwon, S.Y.; Jung, K.Y.; Kim, M.K.; Park, H.; Lee, K.W. Vocal fold wound healing after injection of human adipose-derived stem cells in a rabbit model. Acta Otolaryngol. 2011, 131, 1198–1204. [Google Scholar] [CrossRef]
  48. Tran, E.K.; Juarez, K.O.; Long, J.L. Stem cell treatments for oropharyngeal dysphagia: Rationale, benefits, and challenges. World J. Stem Cells 2020, 12, 1001–1012. [Google Scholar] [CrossRef]
  49. Chi, A.C.; Day, T.A.; Neville, B.W. Oral cavity and oropharyngeal squamous cell carcinoma—An update. CA Cancer J. Clin. 2015, 65, 401–421. [Google Scholar] [CrossRef]
  50. 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. 2016, 186, 1180–1194. [Google Scholar] [CrossRef] [Green Version]
  51. Rangan, S.R. A new human cell line (FaDu) from a hypopharyngeal carcinoma. Cancer 1972, 29, 117–121. [Google Scholar] [CrossRef]
  52. Shimada, Y.; Imamura, M.; Wagata, T.; Yamaguchi, N.; Tobe, T. Characterization of 21 newly established esophageal cancer cell lines. Cancer 1992, 69, 277–284. [Google Scholar] [CrossRef]
  53. Su, F.; Ahn, S.; Saha, A.; DiGiovanni, J.; Kolonin, M.G. Adipose stromal cell targeting suppresses prostate cancer epithelial-mesenchymal transition and chemoresistance. Oncogene 2019, 38, 1979–1988. [Google Scholar] [CrossRef] [PubMed]
  54. Kumar, J.D.; Holmberg, C.; Kandola, S.; Steele, I.; Hegyi, P.; Tiszlavicz, L.; Jenkins, R.; Beynon, R.J.; Peeney, D.; Giger, O.T.; et al. Increased expression of chemerin in squamous esophageal cancer myofibroblasts and role in recruitment of mesenchymal stromal cells. PLoS ONE 2014, 9, e104877. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Wang, J.; Yang, W.; Wang, T.; Chen, X.; Wang, J.; Zhang, X.; Cai, C.; Zhong, B.; Wu, J.; Chen, Z.; et al. Mesenchymal Stromal Cells-Derived β2-Microglobulin Promotes Epithelial-Mesenchymal Transition of Esophageal Squamous Cell Carcinoma Cells. Sci. Rep. 2018, 8, 5422. [Google Scholar] [CrossRef]
  56. 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. Int. J. Exp. Cell. Physiol. Biochem. Pharmacol. 2018, 47, 1785–1799. [Google Scholar] [CrossRef]
  57. Gerhardt, H.; Golding, M.; Fruttiger, M.; Ruhrberg, C.; Lundkvist, A.; Abramsson, A.; Jeltsch, M.; Mitchell, C.; Alitalo, K.; Shima, D.; et al. VEGF guides angiogenic sprouting utilizing endothelial tip cell filopodia. J. Cell Biol. 2003, 161, 1163–1177. [Google Scholar] [CrossRef]
  58. Hutchings, G.; Janowicz, K.; Moncrieff, L.; Dompe, C.; Strauss, E.; Kocherova, I.; Nawrocki, M.J.; Kruszyna, Ł.; Wąsiatycz, G.; Antosik, P.; et al. The Proliferation and Differentiation of Adipose-Derived Stem Cells in Neovascularization and Angiogenesis. Int. J. Mol. Sci. 2020, 21, 3790. [Google Scholar] [CrossRef]
  59. Ratushnyy, A.; Ezdakova, M.; Buravkova, L. Secretome of Senescent Adipose-Derived Mesenchymal Stem Cells Negatively Regulates Angiogenesis. Int. J. Mol. Sci. 2020, 21, 1802. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Min, S.Y.; Kim, H.Y.; Jung, S.Y.; Kwon, Y.; Shin, K.H.; Lee, S.; Kim, S.W.; Kang, H.S.; Yun, Y.H.; Lee, E.S. Oncological safety and quality of life associated with mastectomy and immediate breast reconstruction with a latissimus dorsi myocutaneous flap. Breast J. 2010, 16, 356–361. [Google Scholar] [CrossRef]
  61. Donnenberg, V.S.; Zimmerlin, L.; Rubin, J.P.; Donnenberg, A.D. Regenerative therapy after cancer: What are the risks? Tissue Eng. Part B Rev. 2010, 16, 567–575. [Google Scholar] [CrossRef]
  62. Hur, W.; Lee, H.Y.; Min, H.S.; Wufuer, M.; Lee, C.W.; Hur, J.A.; Kim, S.H.; Kim, B.K.; Choi, T.H. Regeneration of full-thickness skin defects by differentiated adipose-derived stem cells into fibroblast-like cells by fibroblast-conditioned medium. Stem Cell Res. Ther. 2017, 8, 92. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Park, I.S. Enhancement of Wound Healing by Conditioned Medium of Adipose-Derived Stromal Cell with Photobiomodulation in Skin Wound. Int. J. Stem Cells 2021. [Google Scholar] [CrossRef]
  64. Jiao, Z.; Ma, Y.; Zhang, Q.; Wang, Y.; Liu, T.; Liu, X.; Piao, C.; Liu, B.; Wang, H. The adipose-derived mesenchymal stem cell secretome promotes hepatic regeneration in miniature pigs after liver ischaemia-reperfusion combined with partial resection. Stem Cell Res. Ther. 2021, 12, 218. [Google Scholar] [CrossRef] [PubMed]
  65. Menezes, K.; Rosa, B.G.; Freitas, C.; da Cruz, A.S.; de Siqueira Santos, R.; Nascimento, M.A.; Alves, D.V.L.; Bonamino, M.; Rossi, M.I.; Borojevic, R.; et al. Human mesenchymal stromal/stem cells recruit resident pericytes and induce blood vessels maturation to repair experimental spinal cord injury in rats. Sci. Rep. 2020, 10, 19604. [Google Scholar] [CrossRef] [PubMed]
  66. Agostini, F.; Vicinanza, C.; Di Cintio, F.; Battiston, M.; Lombardi, E.; Golinelli, G.; Durante, C.; Toffoli, G.; Dominici, M.; Mazzucato, M. Adipose mesenchymal stromal/stem cells expanded by a GMP compatible protocol displayed improved adhesion on cancer cells in flow conditions. Ann. Transl. Med. 2020, 8, 533. [Google Scholar] [CrossRef] [PubMed]
  67. Latchman, Y.; Wood, C.R.; Chernova, T.; Chaudhary, D.; Borde, M.; Chernova, I.; Iwai, Y.; Long, A.J.; Brown, J.A.; Nunes, R.; et al. PD-L2 is a second ligand for PD-1 and inhibits T cell activation. Nat. Immunol. 2001, 2, 261–268. [Google Scholar] [CrossRef] [PubMed]
  68. Krastev, T.K.; Beugels, J.; Hommes, J.; Piatkowski, A.; Mathijssen, I.; van der Hulst, R. Efficacy and Safety of Autologous Fat Transfer in Facial Reconstructive Surgery: A Systematic Review and Meta-analysis. JAMA Fac. Plast. Surg. 2018, 20, 351–360. [Google Scholar] [CrossRef]
Figure 1. Mesenchymal stromal cell characterization was performed as described previously (*) [13]: (A) Mean Plastic adherence was found in all patient cell lines. A representative photograph is shown. (B) Cell lines were tested for positivity or negativity of the indicated markers using flow cytometry of passage 2 cells. All patient cell lines were positive for CD73, CD90, and CD105 and negative for markers CD31 and CD34 (mean fluorescent intensity (MFI) ± SD), as expected for mesenchymal stromal cells. (C) ELISA results for VEGF in ASC culture supernatants collected in passage 1 are shown. Results for cell culture supernatants are normalized to ng/106 cells (mean ± SD). (D) Lipid vesicle accumulation visualized by Oil Red O staining in adipogenic differentiated ASCs after two weeks of incubation with adipogenic differentiation medium (magnification 200×). Alizarin S staining of extracellular calcium deposits in osteogenic differentiated ASCs after two weeks of incubation with osteogenic differentiation medium (magnification 200×). Alcian blue staining of cartilage extracellular matrix in ASCs cultured as three-dimensional spheroids after four weeks of incubation with chondrogenic differentiation medium (magnification 40×). Shown are representative images of n = 5 independent experiments.
Figure 1. Mesenchymal stromal cell characterization was performed as described previously (*) [13]: (A) Mean Plastic adherence was found in all patient cell lines. A representative photograph is shown. (B) Cell lines were tested for positivity or negativity of the indicated markers using flow cytometry of passage 2 cells. All patient cell lines were positive for CD73, CD90, and CD105 and negative for markers CD31 and CD34 (mean fluorescent intensity (MFI) ± SD), as expected for mesenchymal stromal cells. (C) ELISA results for VEGF in ASC culture supernatants collected in passage 1 are shown. Results for cell culture supernatants are normalized to ng/106 cells (mean ± SD). (D) Lipid vesicle accumulation visualized by Oil Red O staining in adipogenic differentiated ASCs after two weeks of incubation with adipogenic differentiation medium (magnification 200×). Alizarin S staining of extracellular calcium deposits in osteogenic differentiated ASCs after two weeks of incubation with osteogenic differentiation medium (magnification 200×). Alcian blue staining of cartilage extracellular matrix in ASCs cultured as three-dimensional spheroids after four weeks of incubation with chondrogenic differentiation medium (magnification 40×). Shown are representative images of n = 5 independent experiments.
Cancers 13 02751 g001
Figure 2. Box-whisker plots depicting normalized proliferation of FaDu (left) and Kyse30 cells (right), cultured with ASC-derived supernatants from patients 1–5 (“ASC (Pat. 1–5)”) or supernatant from FaDu/Kyse30 with MSCM 2 (“FaDu w/ MSC”, “Kyse30 w/ MSC”), DMEM/RPMI without FCS (“DMEM w/o FCS”, “RPMI w/o FCS”), and DMEM/RPMI with FCS (“DMEM w/ FCS”, “RPMI w/ FCS”) as controls. Shown are mean ± SD from n = 3 independent experiments. Statistical test: Kruskal–Wallis H test, p-values * < 0.05, ** < 0.01.
Figure 2. Box-whisker plots depicting normalized proliferation of FaDu (left) and Kyse30 cells (right), cultured with ASC-derived supernatants from patients 1–5 (“ASC (Pat. 1–5)”) or supernatant from FaDu/Kyse30 with MSCM 2 (“FaDu w/ MSC”, “Kyse30 w/ MSC”), DMEM/RPMI without FCS (“DMEM w/o FCS”, “RPMI w/o FCS”), and DMEM/RPMI with FCS (“DMEM w/ FCS”, “RPMI w/ FCS”) as controls. Shown are mean ± SD from n = 3 independent experiments. Statistical test: Kruskal–Wallis H test, p-values * < 0.05, ** < 0.01.
Cancers 13 02751 g002
Figure 3. Box-whisker plots depicting migration (normalized after subtraction of negative controls) of FaDu (left) and Kyse30 cells (right) towards ASC-derived supernatants from patients 1–5 (“ASC (Pat. 1–5)”) or supernatant from FaDu/Kyse30 with MSCM (“FaDu w/ MSC”, “Kyse30 w/ MSC”), and DMEM/RPMI with FCS (“DMEM w/ FCS”, “RPMI w/ FCS”) as controls. Shown are mean ± SD from n = 3 independent experiments. Statistical test: Kruskal–Wallis H test, p-values * < 0.05, ** < 0.01, ns: not significant.
Figure 3. Box-whisker plots depicting migration (normalized after subtraction of negative controls) of FaDu (left) and Kyse30 cells (right) towards ASC-derived supernatants from patients 1–5 (“ASC (Pat. 1–5)”) or supernatant from FaDu/Kyse30 with MSCM (“FaDu w/ MSC”, “Kyse30 w/ MSC”), and DMEM/RPMI with FCS (“DMEM w/ FCS”, “RPMI w/ FCS”) as controls. Shown are mean ± SD from n = 3 independent experiments. Statistical test: Kruskal–Wallis H test, p-values * < 0.05, ** < 0.01, ns: not significant.
Cancers 13 02751 g003
Figure 4. Top left: Branches (yellow lines) in each well were counted using ImageJ Angiogenesis Analyzer software. Top right: Box-whisker plots depicting tube formation (number of branches normalized to MSCM 2 control after subtraction of ECGM with 30 nM Suramin control) of HUVECs cultured with the MSCM 2 control, ECGM (without Suramin) control, or ASC-derived supernatants from five different patients (“ASC (Pat. 1–5)”). Lower half: Representative photographs depicting tube formation in wells corresponding to patients 1–5 (“ASC (Pat. 1–5)”) and controls. p-values ** < 0.01, *** < 0.001.
Figure 4. Top left: Branches (yellow lines) in each well were counted using ImageJ Angiogenesis Analyzer software. Top right: Box-whisker plots depicting tube formation (number of branches normalized to MSCM 2 control after subtraction of ECGM with 30 nM Suramin control) of HUVECs cultured with the MSCM 2 control, ECGM (without Suramin) control, or ASC-derived supernatants from five different patients (“ASC (Pat. 1–5)”). Lower half: Representative photographs depicting tube formation in wells corresponding to patients 1–5 (“ASC (Pat. 1–5)”) and controls. p-values ** < 0.01, *** < 0.001.
Cancers 13 02751 g004
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Sharaf, K.; Eggersmann, T.K.; Haider, S.P.; Schwenk-Zieger, S.; Zhou, J.; Gires, O.; Lechner, A.; Canis, M.; Haubner, F. Human Adipose-Derived Stem/Stromal Cells Promote Proliferation and Migration in Head and Neck Cancer Cells. Cancers 2021, 13, 2751. https://doi.org/10.3390/cancers13112751

AMA Style

Sharaf K, Eggersmann TK, Haider SP, Schwenk-Zieger S, Zhou J, Gires O, Lechner A, Canis M, Haubner F. Human Adipose-Derived Stem/Stromal Cells Promote Proliferation and Migration in Head and Neck Cancer Cells. Cancers. 2021; 13(11):2751. https://doi.org/10.3390/cancers13112751

Chicago/Turabian Style

Sharaf, Kariem, Tanja K. Eggersmann, Stefan P. Haider, Sabina Schwenk-Zieger, Jiefu Zhou, Olivier Gires, Axel Lechner, Martin Canis, and Frank Haubner. 2021. "Human Adipose-Derived Stem/Stromal Cells Promote Proliferation and Migration in Head and Neck Cancer Cells" Cancers 13, no. 11: 2751. https://doi.org/10.3390/cancers13112751

APA Style

Sharaf, K., Eggersmann, T. K., Haider, S. P., Schwenk-Zieger, S., Zhou, J., Gires, O., Lechner, A., Canis, M., & Haubner, F. (2021). Human Adipose-Derived Stem/Stromal Cells Promote Proliferation and Migration in Head and Neck Cancer Cells. Cancers, 13(11), 2751. https://doi.org/10.3390/cancers13112751

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop