Skip to main content

Xeno-free protocol for GMP-compliant manufacturing of human fetal pancreas-derived mesenchymal stem cells

Abstract

Background

Mesenchymal stem cells (MSCs) have been suggested as an appropriate source for diabetes cell-based therapies. The high proliferation and differentiation capacity of fetal MSCs and the role of fetal pancreatic-derived MSCs (FPMSCs) in islet generation make them good candidates for diabetes treatment. To manufacture clinical-grade MSCs, animal-free culture protocols are preferred. The current study aimed to establish a xeno-free/GMP-compliant protocol for FPMSCs manufacturing. The focus was on the effects of fetal bovine serum (FBS) replacement with pooled human serum (HS).

Material and methods

FPMSCs were isolated and expanded from the pancreas of legally aborted fetuses with few modifications in our previously established protocol. The cells were expanded in two different culture media, including DMEM supplemented with 10% FBS or 10% pooled HS. A side-by-side comparison was made to evaluate the effect of each serum on proliferation rate, cell cycle, senescence, multi-lineage differentiation capacity, immunophenotype, and tumorigenesis of FPMSCs.

Results

Flow cytometry analysis and three-lineage differentiation ability demonstrated that fibroblast-like cells obtained from primary culture had MSCs’ characteristics. The FPMSCs displayed similar morphology and CD markers expression in both sera. HS had a higher proliferative effect on FPMSCs than FBS. In FBS, the cells reached senescence earlier. In addition to normal karyotypes and anchorage-dependent growth, in vivo tumor formation was not seen.

Conclusion

Our results demonstrated that HS was a better serum alternative than FBS for in vitro expansion of FPMSCs. Compared with FBS, HS increased FPMSCs’ proliferation rate and decreased their senescence. In conclusion, HS can effectively replace FBS for clinical-grade FPMSCs manufacturing.

Graphical abstract

Introduction

Type 1 diabetes mellitus (T1DM) is an autoimmune disease caused by the destruction of insulin-producing beta cells. The current treatment modalities aim to control diabetes and its complications effectively [1]. The socioeconomic impact of T1DM has motivated scientists to search for curative treatment. As a new curative approach, islet transplantation has several limitations: the scarcity of islets, transplant rejection, immunosuppression, difficulties related to islet isolation, cell loss during the separation and transplantation process, and high costs [2]. Therefore, finding alternative cell sources for regenerating or replacing damaged islets is crucial. In recent decades, stem cell-based therapies have raised hopes for treating incurable diseases such as diabetes [3]. Mesenchymal stem cells (MSCs) have received more attention due to their proliferation and differentiation capacity and low immunogenicity. MSCs can migrate to the pancreas, reduce apoptosis and residual beta cell mass loss, and stimulate pancreatic stem cells to proliferate and differentiate into endocrine cells [4, 5]. Despite similar phenotypes and the common mechanisms of tissue regeneration, the source of MSCs can play a crucial role in their therapeutic effects [6]. It has been suggested that tissue-matched MSCs may increase the efficacy of their regenerative effects [7]. Studies have shown that diversity in the microenvironment of MSCs and, subsequently, the expression of different genes lead to differences in their function and behavior [8]. It is suggested that MSCs isolated from each tissue may be a more appropriate source for regenerating the same tissue. For instance, studies have shown that pancreatic-derived MSCs can play a more supportive and regenerative role for pancreatic islets [9,10,11]. Fetal pancreatic-derived MSCs (FPMSCs) are highly proliferative cells that play a crucial role in fetal pancreas development [12]. In vivo studies have been conducted to evaluate the therapeutic effect of FPMSCs in diabetes. These studies showed that they could be engrafted, differentiated, and exhibited islet cell-like function in the animal model. Furthermore, they have less immunogenicity and higher differentiation and proliferation capacity than adult MSCs [13, 14]. In our recent study, we examined the impact of the three-dimensional co-culture of FPMSCs on human embryonic stem cell-derived pancreatic progenitors’ development. This study revealed higher expression of NGN3 and INSULIN in FPMSCs than bone marrow-derived MSCs group [10]. Before clinical application, FPMSCs should be expanded in vitro to make an appropriate dose. Optimal MSCs’ expansion requires a 10–20% serum in a culture medium that significantly enhances proliferation and differentiation [15]. The source of serum is one of the most important safety considerations in clinical-grade cell manufacturing. Fetal bovine serum (FBS) is widely used for MSCs’ expansion [16, 17]. The use of animal derivatives can lead to adverse immune reactions such as anaphylactic shock or cardiac arrhythmia and may transmit viral and bacterial infections [18]. Studies showed that xeno-free sera such as human serum (HS) or human platelet lysate (hPL) could be used for MSCs’ culture. Indeed, MSCs’ expansion for clinical application should be carried out according to current good manufacturing practices (cGMP) [19]. GMP covers all aspects of the manufacturing process, including raw material selection and qualification. It emphasizes that all materials that come in direct contact with cell therapy products should be of appropriate quality. Additionally, there are more specific considerations for animal-sourced products [20]. Hence, to increase the safety of cell therapy products, most regulatory agencies recommend omitting animal-derived materials in the cell manufacturing process [21,22,23]. Regarding the possible importance of MSCs in support of islet regeneration and considering the unique potential of FPMSCs in the treatment of diabetes, we aimed to optimize the FPMSCs’ expansion under xeno-free/GMP-compliant conditions.

Materials and methods

Isolation and expansion of FPMSCs

To procure the legally aborted fetus, written informed consent was obtained from parents according to the procedures approved by the Ethics Committee of Endocrinology and Metabolism Research Institute, Tehran University of Medical Sciences, Tehran, Iran (IRB code: EC-00264). In addition to routine donor evaluation, HIV, HBV, HCV, CMV, EBV, HTLV, Toxoplasma, and venereal diseases were examined in the mother’s blood sample. FPMSCs were isolated and expanded according to our previously described method with few modifications [24]. Briefly, the pancreas was aseptically harvested from the aborted fetus (14–18 weeks) by a midline laparotomy following the left subcostal extension. The harvested tissue was washed three times with PBS/EDTA buffer (CliniMACS, Miltenyi Biotec, Germany) containing 200 units/ml penicillin, 0.2 mg/ml streptomycin, and 0.5 µg/ml amphotericin B (antibiotic/antimycotic solution, Biowest, France), trimmed, minced in small fragments, and partially digested using animal-free collagenase CLSAFA/AF (Worthington, USA) at 37 °C for 10 min. The digestion process was terminated by adding an equal volume of cold PBS/EDTA and allowing fragments to settle on ice for 10 min. The small tissue fragments were washed with PBS/EDTA and centrifuged for 5 min at 300 × g. The pellet was resuspended in DMEM-LG supplemented with 15% FBS (Both from Biowest, France), seeded into a 25 cm2 culture flask, and incubated at 37 °C, 5% CO2, and 95% humidity. To deplete non-adherent cells, the culture medium was changed after 72 h, and cell expansion was continued up to 90% confluency (10–14 days). Then, adherent cells were harvested using TrypLE™ solution (Thermo Fisher, USA) and seeded into new culture flasks containing DMEM-LG + 10% FBS (1 to 4 ratio). At the third passage (P3), the cells were harvested, counted, and cryopreserved (10% DMSO + 90% culture medium (DMEM-LG + 10% FBS)) for future studies. All tissue processing steps and cell manufacturing were performed under aseptic conditions using a Class II biological safety cabinet located in a Grade B cleanroom. Samples from each batch were examined for identity, potency, sterility, mycoplasma, and endotoxin. To evaluate and compare the effects of different sera, fully characterized FPMSCs were thawed, washed, and resuspended in PBS/EDTA. Then, 3 × 103 cells/cm2 were seeded into culture flasks containing DMEM-LG + 10% FBS (FBS group) or DMEM-LG + 10% HS (pooled AB serum, Biowest, France, (HS group). Before a side-by-side comparison, the cells were adapted to the new serum for three consecutive subcultures.

Characterization of FPMSCs

To characterize FPMSCs, each cryopreserved batch was evaluated at the third passage (P3) for CD markers’ expression, morphology, and three-lineage differentiation capacity [25]. Then, fully characterized FPMSCs were used for the experiments.

Flow cytometry analysis

At P3, FPMSCs (1 × 106) were incubated for an hour in PBS containing BSA 3% (Atocel, Austria) at 4 °C. Then, the cells were labeled with specific conjugated antibodies or isotype-matched control (all from eBioscience, Inc., USA) against CD markers according to the manufacturer’s instructions for 45 minutes at 4 °C. The labeled cells were resuspended in FACS buffer (FACSCaliburTM, BD Biosciences, USA) after washing twice with PBS (Biowest, France). For FPMSCs characterization, CD11b-FITC, CD45-FITC, CD19-PE, CD34-PE, HLA-DR-PE, CD105-PE, CD73-PE, and CD90-PE were examined. Moreover, to evaluate the effect of serum supplementation on FPMSCs’ phenotype, CD105-PE, CD73-PE, CD90-PE, and HLA-DR-PE were assayed in each study group. Flow cytometry analysis was done by Saba Biomedical laboratory (Tehran, Iran)

Multi‑lineage differentiation

StemPro® Osteogenesis, Adipogenesis, and Chondrogenesis Differentiation Kits were used to evaluate the differentiation capacity of FPMSCs (all from Invitrogen, USA). For the osteogenic differentiation study, FPMSCs were seeded into 4-well plates (1.5 × 104 cells/well). After 21 days of incubation, the cells were fixed with 4% PFA (paraformaldehyde) for 20 minutes at room temperature (RT), and calcium deposition was stained with an Alizarin Red S solution (Sigma, USA). For adipogenic differentiation, 2.5 × 104 cells/well were seeded in 4-well plates. After 14 days of culture, the cells were fixed, and the formation of lipid vacuoles in FPMSCs was examined using Oil Red O (Sigma, USA) staining. Chondrogenesis media was added to 2.5 × 105 FPMSCs, and the suspension was centrifuged (200 × g/5 minutes) to evaluate chondrogenic differentiation. The supernatant was discarded without disturbing the pellet, and 0.5 ml of differentiation media was added. The tubes were incubated with loose caps in an upright position at 37 °C and 5% CO2. After 28 days, the cell nodules were fixed in 4% PFA overnight. The paraffin blocks were prepared from fixed nodules and then stained with Alcian blue to evaluate cartilage matrix formation (the Pathology Department of the Shariati Hospital, Tehran, Iran). FPMSCs from each group were examined for osteogenic and adipogenic differentiation capacity to assess the effect of serum supplementation. Images were taken using an inverted microscope (Eclipse TS100, Nikon, Japan).

Growth kinetic

The proliferation rate and growth pattern of FPMSCs were evaluated by cell cycle analysis, population doubling time (PDT) calculation, and plotting the growth curve. For cell cycle analysis, 5 × 105 FPMSCs were fixed in cold 70% ethanol (4 °C, 1 h). After washing twice with cold PBS (800 × g /5 min), the cell samples were sent to the flow cytometry department of Saba Biomedical Laboratory (Tehran, Iran). Further processing of the samples was done according to their protocols, and the percentage of FPMSCs in S + G2 phases was determined. To calculate the PDT, 7 × 103 cells/well were seeded into 12-well culture plates. At 80% confluency, the cells were harvested and counted, and the PDT was calculated by this formula:

$${\text{PDT}} = \frac{{{\text{Culture duration}} \left( h \right) \times \log \left( 2 \right)}}{{\log \left( {\text{Final concentration}} \right) - \log \left( {\text{Initial concentration}} \right)}}$$

Cell expansion was continued for five consecutive passages (P3 to P8), and the mean of the PDT was calculated. FPMSCs were seeded at a density of 7 × 103 cells/well in 12-well plates and were incubated at 37 °C and 5% CO2 to plot the growth curve. The cells were counted daily, and the mean numbers were used to draw the growth curve.

Senescence assay

Senescence-associated β-galactosidase (SA-βG) assay was applied to evaluate FPMSCs’ senescence. The cells were seeded into 12-well plates at 4 × 104 cells/well in different media and cultured up to 60% confluency. After fixation with 2% PFA and 0.2% glutaraldehyde, the cells were stained using X-gal (5-Bromo-4-chloro-3-indolyl β-D-galactopyranoside) staining kit (Sigma-Aldrich, USA) according to the manufacturer’s instruction. The presence of SA-βG (a blue precipitate in the cytoplasm) was investigated by an inverted microscope.

Immunocytochemistry

To evaluate the effect of serum on FPMSCs’ protein expression, immunocytochemistry (ICC) was performed. The cells from each group were seeded into 4-well plates at 1 × 104 cells/well. At 50–60% confluency, the cells were fixed with 4% PFA, washed three times in 0.1% PBS/Tween20 (PBST), permeabilized using PBS + 0.25% Triton X-100 (10 min /RT), and blocked by blocking buffer (10% goat serum and 1% Albumin in PBST) for 1 h at RT. Finally, according to the manufacturer’s instructions, the primary antibodies were diluted in dilution buffer (PBST + 1% Albumin) and added to each sample. For this experiment, anti-Nestin (14–9843-80), anti-Vimentin (14–9897-80), anti-Ki67 (14–5699-80), anti-HLA-ABC (14–9983-80), and Mouse IgG1 K Isotype Control (14–4714-81) antibodies were used (all from eBioscience, USA). The cells were incubated in primary antibodies for an hour at 37 °C. EnVision Detection Kit (Dako, Denmark) was used to visualize the antigen–antibody reaction. Briefly, the secondary antibody (goat-HRP anti-Rabbit/Mouse Ab) was added (incubation at 37 °C for 30 min). After washing with PBST, 50 µl of DAB chromogen solution and 1 ml substrate buffer were added (5–10 min, RT), and the cells were examined under an inverted microscope. To quantify the Ki67 expression, the number of stained cells was calculated in 10 different microscopic fields (20X objective) with ImageJ software (v1.53f) using the “Cell Counter” plugin (plugins → analysis → cell counter) [26].

Tumorigenesis assay

Tumorigenesis potential of in vitro expanded FPMSCs was examined according to the recommendation of WHO technical report series No. 978 /61st report, 2013 [27].

Karyotype analysis

FPMSCs (at different passages) were sent to the Genetics Laboratory of Sarem Hospital (Tehran, Iran) for G-banding karyotyping.

Soft agar colony formation assay

The anchorage-independent growth ability of FPMSCs was examined by a soft agar colony formation assay using a previously published protocol [28]. Briefly, 1% sterile agar solution (UltraPure™ Agarose, 16500100, Thermo Fisher, USA) was mixed with 2X DMEM+20% FBS in a 1:1 ratio and added to a non-treated 6-well plate (base agar). To prepare top agar, three different numbers of FPMSCs (50, 20, and 10 × 104 cells) at 6th and 10th passages were mixed with 2X DMEM+20% FBS and 6% sterile agarose and added to base agar. The plates were incubated in 5% CO2, 37 ºC, and 95% humidity for 21 days. Colony formation was examined under an inverted microscope after crystal violet staining (0.1% W⁄V). HEK 293T cells (donated by Sabz Biomedicals, Tehran, Iran) were used as a positive control.

In vivo tumorigenicity assay

According to relevant ethical and institutional guidelines, the animal experiment was performed by the Gene Therapy Research Center (Digestive Disease Research Institute, Tehran University of Medical Sciences, Tehran, Iran). Briefly, 10 × 106 viable cells were suspended in 0.1 ml PBS and injected intramuscularly into 10 athymic mice (4–7 weeks old). Considering the principle of “reduction” in animal experiments, we decided to perform in vivo assay only for HS-supplemented group (as the optimal protocol). The HEK 293T cells were used as a positive control. All animals were examined weekly (up to 16 weeks) for any evidence of nodule formation at the injection site. At the end of the experiment, all animals were euthanized and examined for gross and microscopic evidence of the proliferation of inoculated cells at the injection site and vital organs.

Statistical analysis

The FPMSCs were isolated from three different donors, and all in vitro experiments were performed in triplicates. Paired T test was used to compare the quantitative data of each group. Results were reported as the mean ± SD and P value < 0.05 were considered significant.

Results

Isolation, expansion, and characterization of FPMSCs

FPMSCs were successfully isolated from three consecutive fetal pancreases. At P3, a homogenous population of spindle-shaped cells was seen. CD marker expression panel demonstrated that they express mesenchymal markers and lack hematopoietic markers and HLA-DR (Fig. 1A). Furthermore, they could differentiate into adipocytes, osteocytes, and chondrocytes using appropriate media (Fig. 1B).

Fig. 1
figure 1

FPMSCs’ characterization at the third subculture: A CD marker expression of FPMSCs isolated from different donors, B three-lineage differentiation of FPMSCs. The expression of mesenchymal CD markers and HLA-DR in both study groups C. The effect of different sera on FPMSCs’ differentiation to osteocytes D and adipocytes E

CD marker expression and differentiation capacity in different sera

After three consecutive passages in FBS or HS, no changes were seen in the expression of PLMSCs’ CD markers (Fig. 1C) or their differentiation capacity to osteocytes (Fig. 1D) and adipocytes (Fig. 1E). In both groups, PLMSCs showed spindle-shaped morphology. However, the cells in the HS group were thinner and smaller than in the FBS group (Fig. 2A).

Fig. 2
figure 2

A FPMSCs’ morphology in different sera. B Growth curve of FPMSCs in HS- and FBS-supplemented media. C The results of cell cycle analysis. D and E SA-βG assay of FPMSCs from two batches

Growth kinetic and senescence of FPMSCs in different sera

After several passages (P8), our results showed that the mean of PDT in the HS group (77.74 \(\pm \hspace{0.17em}\)22.52 h) was significantly (P < 0.0001) lower than in the FBS group (94.32 \(\pm \hspace{0.17em}\)24.48 h). In addition, the cell yield/cm2 of the culture area was 22,609.78 \((\pm \hspace{0.17em}\)6397.84) in the HS group and 16,864 \((\pm \hspace{0.17em}\)5993.90) in the FBS group (P < 0.0001). The growth curves showed that HS had increased the proliferation rate of FPMSCs (Fig. 2B). Cell cycle analysis revealed that the mean of FPMSCs in S + G2 phases in the HS group (32.98 \(\pm \hspace{0.17em}\)8.55) was significantly (P < 0.05) higher than in the FBS group (22.70 \(\pm \hspace{0.17em}\)4.22). Figure 2C illustrates the results of cell cycle analysis in one representative sample. The results of the SA-βG assay revealed that FBS accelerated the senescence of serially passaged FPMSCs. In one sample, we found early senescence of FPMSCs in FBS at P7, while they could be expanded in HS up to P17. In two other samples, the cells were proliferative in FBS up to P13 and P15 and in HS up to P18 and P20, respectively. Figure 2D, E illustrates the expression of SA-βG in each study group of two different PLMSC samples.

Proteins expression profile

ICC studies demonstrated that Vimentin (Fig. 3A) and HLA-ABC (Fig. 3B) were expressed equally in both study groups. The expression of Ki67 (Fig. 3C) and Nestin (Fig. 3D) was higher in the HS group. The mean of Ki67 expression was 54%\((\pm \hspace{0.17em}\)8.04) in the HS group and 44%\((\pm \hspace{0.17em}\)4.24) in the FBS group (P < 0.01).

Fig. 3
figure 3

ICC assay of FPMSCs in both study groups. A Vimentin, B HLA-ABC, C Ki67, and D Nestin. The white arrows show the Nestin expression in FBS-cultured cells

Tumorigenicity assay

G-banding karyotyping demonstrated chromosomal stability of in vitro expanded FPMSCs in both groups at different passages. All samples showed a normal 46XY karyotype (Fig. 4A). Figure 4B illustrates the results of soft agar colony formation assays in the HS and FBS group at the sixth passage. The assay revealed that HEK 293 T (positive control) could generate large colonies after 14 days. In contrast, no colony formation was seen in FPMSCs’ groups until the 28th day of culture. The same results were observed for FPMSCs in the 10th passage. Similarly, in vivo studies revealed that HS-cultured FPMSCs did not induce tumor formation in nude mice up to 4 months post-transplantation. In positive controls, nodule formation was seen after 1 week and was prominent in the second-week post-transplantation (Fig. 4C).

Fig. 4
figure 4

Tumorigenicity assay of FPMSCs in HS- and FBS-supplemented media. A G-banding karyotyping, B soft agar colony formation assay, C tumor formation in nude mice. The white arrows show early colony formation of HEK 293 T cells

Discussion

For decades, exogenous insulin injection has been suggested as the main treatment option for patients with T1DM. Despite regular insulin injections, fewer than 40% of patients can reach and maintain a stable euglycemic state. Recently, many researchers have focused on finding more efficient therapies for T1DM. The most promising approach is to restore beta cell mass or functionality using regenerative medicine technologies. Cell-based therapies and tissue engineering aim to improve the length and quality of patients’ life by regenerating, preserving, or enhancing beta cell functions [29]. In the last two decades, many protocols have been successfully developed to generate insulin-producing cells (IPCs) or islet organoids in vitro. Embryonic stem cells (ESCs) and MSCs have shown better outcomes among different cell types. In addition to IPCs’ generation, MSCs have been co-cultivated with ESCs to support islet organoid formation. Previous studies have shown that FPMSCs express different genes, proteins, and signaling pathway factors than other mesenchymal sources, suggesting them as a more suitable candidate for diabetes treatment [10]. In autoimmune and systemic diseases (like diabetes), the proliferation and differentiation capacity of autologous MSCs are significantly impaired. In such cases, allogeneic MSCs may have better therapeutic effects. Among different sources of allogeneic cells, priority should be given to those with higher potency, more safety, and lower immunogenicity. Fetal-derived MSC seems a suitable source for regenerative therapies because they are safer than ESCs and have higher potency and less immunogenicity than adult SCs [30, 31]. To prepare a therapeutic dose, MSCs should be expanded in vitro with the principles of GMP [32]. Along with other principles of GMP, careful selection of ancillary materials (AMs) is essential to assure the product’s safety and efficacy [33]. It is preferred to omit xenogeneic materials from the manufacturing process when applicable. The current study describes a xeno-free/GMP-compliant protocol for the in vitro expansion of FPMSCs. Our results demonstrated that FPMSCs were expanded efficiently using our xeno-free protocol. In addition, we could successfully modify our previous isolation protocol [24] using an animal-origin-free collagenase. FPMSCs’ characterization studies revealed no significant difference between FBS and HS groups. Both culture conditions yielded a pure population of FPMSCs, which lacked HLA-DR, expressed Vimentin, HLA-ABC, CD 90, CD105, and CD73, and were differentiated into osteocytes and adipocytes. The cells in both groups showed spindle shape morphology, but their size was smaller in the HS group (Fig. 2A). Some studies have shown that HS and FBS have similar effects on the differentiation potential of MSCs [34, 35]. However, few studies have indicated that HS increased the osteogenic differentiation of MSCs [19, 36]. Our differentiation studies aimed to evaluate the effect of serum supplementation on FPMSCs’ characteristics. To assess the effect of different sera on the differentiation capacity of MSCs, quantitative studies are needed. The growth curves demonstrated that FPMSCs proliferate more rapidly in HS-supplemented media. Similarly, cell cycle analysis revealed that the average of cells in S + G2 phases was significantly higher in the HS group (P < 0.05). Moreover, FPMSCs in the HS group showed higher expression of the Ki67 proliferation marker (P < 0.05). These findings imply that HS has a more proliferative effect on FPMSCs than FBS (Fig. 2C). From the 10th passage, FPMSCs in the HS group had a lattice growth pattern and were spindle-shaped until the 15th. In a similar study, ADMSCs cultured in HS showed a lattice growth pattern from the fourth passage with faster growth and smaller size than FBS [22]. Similarly, de Paula et al. reported that ADMSCs proliferated more rapidly in HS, and the cell density per unit area was higher than FBS [37]. In addition to the proliferation assays, we compared FPMSCs’ senescence during serial passages. The results of the SA-βG assay showed accelerated cellular senescence in FBS compared with the HS group. To sum up, the HS supplementation increases the yield of FPMSCs by boosting their proliferation, changing their size, and delaying their senescence. It is strongly in favor of clinical dose manufacturing. Both autologous and allogeneic human serum has been previously used for MSCs culture with diverse results [35, 38, 39]. These may be due to batch-to-batch variation of autologous and single donor allogenic HS. Therefore, the use of pooled HS is recommended. In this study, a commercial pooled HS was used, leading to more consistent results. The immunocytochemistry results showed that Nestin expression was considerably higher in the HS group (Fig. 3D). Nestin is a type VI intermediate filament protein expressed in many cells in the early stages of fetal development and may represent a specific group of multipotent precursor cells. Studies on damaged cells have shown that Nestin expression plays a crucial role in their repair and regeneration process [40]. Moreover, recent studies have indicated that Nestin-positive cells, with MSCs characteristics, resided in pancreatic islets and could differentiate into IPCs and islet-like cell clusters [41]. Interestingly, Nestin expression was observed in most FPMSCs cultured in FBS in previous studies. Only one study reported low expression of Nestin in FPMSCs [42]. Like our study, they had derived FPMSCs from pancreatic islet-like clusters. It suggests that the source and method of FPMSCs isolation may affect Nestin expression. The in vitro manipulation and expansion of cells raise numerous concerns, including the possibility of tumor formation or genomic alterations. We evaluate the tumorigenicity potential of in vitro cultured FPMSCs by G-banding karyotyping and soft agar colony formation at various passages. The results did not show colony formation or karyotype instability in both groups. Furthermore, no tumor formation was seen up to 4 months after the transplantation of FPMSCs cultured in HS-supplemented media. The current study is the first report on establishing a xeno-free/GMP-compliant protocol for FPMSCs’ expansion . Despite that, our study had some limitations, which should be considered in future experiments. To harvest the pancreas, we used donated fetuses that had been legally aborted. The aborted fetuses were rejected in many cases because of chromosomal abnormalities, long ischemic time, and damaged bodies/tissues. Consequently, we could not find appropriate donors below 14 weeks. On the other hand, we had legal and ethical limitations in using the fetuses in the third trimester. The results of our study could have been more conclusive if we had been able to examine FPMSCs derived from the first and the third trimesters. Considering the fetal pancreas shortage and to minimize the risk of failure, we used our previously established protocol for FPMSCs derivation. In that protocol, FBS was used as a serum supplementation. Similar to the previous studies, we adapted FPMSCs to the new serum (HS) for three consecutive passages, and then a side-by-side comparison was made. We can completely omit FBS from our protocol in future studies regarding the positive effect of HS supplementation on FPMSCs’ expansion. As FPMSCs could be a potential source for beta cell generation, future studies should focus on HS’s effect(s) on FPMSCs differentiation to IPCs.

Conclusions

Considering the results of this study, we can conclude that HS is a suitable FBS alternative for clinical-grade FPMSCs manufacturing. Supplementation of culture medium with 10% HS can yield a pure, safe, potent, and scalable population of FPMSCs. Nevertheless, more in vitro and in vivo studies are needed to complete the safety profile of FPMSCs before their clinical applications.

Availability of data and materials

Not applicable.

Abbreviations

AMs:

Ancillary materials

CD markers:

Cluster of differentiation markers

DMEM-LG:

Dulbecco’s modified eagle medium—low glucose

EDTA:

Ethylenediaminetetraacetic acid

ESCs:

Embryonic stem cells

FBS:

Fetal bovine serum

FPMSCs:

Fetal pancreatic-derived MSCs

GMP:

Good manufacturing practice

HLA-DR:

Human leukocyte antigen—DR isotype

HS:

Human serum

ICC:

Immunocytochemistry

IPCs:

Insulin-producing cells

MSCs:

Mesenchymal stem cells

PBS:

Phosphate-buffered solution

PDT:

Population doubling time

RT:

Room temperature

SCs:

Stem cells

T1DM:

Type 1 diabetes mellitus

X-gal:

5-Bromo-4-chloro-3-indolyl β-D-galactopyranoside

References

  1. Bluestone JA, Buckner JH, Fitch M, Gitelman SE, Gupta S, Hellerstein MK, et al. Type 1 diabetes immunotherapy using polyclonal regulatory T cells. Sci Transl Med. 2015. https://doi.org/10.1126/scitranslmed.aad4134.

    Article  PubMed  PubMed Central  Google Scholar 

  2. Shin J-Y, Jeong J-H, Han J, Bhang SH, Jeong G-J, Haque MR, et al. Transplantation of heterospheroids of islet cells and mesenchymal stem cells for effective angiogenesis and antiapoptosis. Tissue Eng Part A. 2015;21(5–6):1024–35.

    Article  CAS  Google Scholar 

  3. Chhabra P, Brayman KL. Stem cell therapy to cure type 1 diabetes: from hype to hope. Stem Cells Transl Med. 2013;2(5):328–36.

    Article  CAS  Google Scholar 

  4. Wei X, Yang X, Han ZP, Qu FF, Shao L, Shi YF. Mesenchymal stem cells: a new trend for cell therapy. Acta Pharmacol Sinica. 2013;34(6):747–54.

    Article  CAS  Google Scholar 

  5. Yeung TY, Seeberger KL, Kin T, Adesida A, Jomha N, Shapiro AJ, et al. Human mesenchymal stem cells protect human islets from pro-inflammatory cytokines. PLoS ONE. 2012;7(5):e38189.

    Article  CAS  Google Scholar 

  6. Jabbarpour Z, Kiani J, Keshtkar S, Saidijam M, Ghahremani MH, Ahmadbeigi N. Effects of human placenta-derived mesenchymal stem cells with NK4 gene expression on glioblastoma multiforme cell lines. J Cell Biochem. 2020;121(2):1362–73.

    Article  CAS  Google Scholar 

  7. Sagaradze GD, Basalova NA, Efimenko AY, Tkachuk VA. Mesenchymal stromal cells as critical contributors to tissue regeneration. Front Cell Develop Biol. 2020. https://doi.org/10.3389/fcell.2020.576176.

    Article  Google Scholar 

  8. Sangeetha K, Vennila R, Secunda R, Sakthivel S, Pathak S, Jeswanth S, et al. Functional variations between mesenchymal stem cells of different tissue origins: a comparative gene expression profiling. Biotech Lett. 2020;42(7):1287–304.

    Article  CAS  Google Scholar 

  9. Lee S, Jeong S, Lee C, Oh J, Kim S-C. Mesenchymal stem cells derived from human exocrine pancreas spontaneously express pancreas progenitor-cell markers in a cell-passage-dependent manner. Stem Cells Int. 2016;2016:1–12.

    Google Scholar 

  10. Ghezelayagh Z, Zabihi M, Zarkesh I, Gonçalves CAC, Larsen M, Hagh-parast N, et al. Improved differentiation of hESC-derived pancreatic progenitors by using human fetal pancreatic mesenchymal cells in a micro-scalable three-dimensional co-culture system. Stem Cell Rev Rep. 2022;18(1):360–77.

    Article  CAS  Google Scholar 

  11. Ahmed SM, El-Badri N. Pancreatic mesenchymal stromal cells: characteristics and possible origin. Liver Pancreat Sci. 2016;1(1):1–5.

    Article  Google Scholar 

  12. Hu Y, Liao L, Wang Q, Ma L, Ma G, Jiang X, et al. Isolation and identification of mesenchymal stem cells from human fetal pancreas. J Lab Clin Med. 2003;141(5):342–9.

    Article  CAS  Google Scholar 

  13. Ersek A, Pixley JS, Goodrich AD, Porada CD, Almeida-Porada G, Thain DS, et al. Persistent circulating human insulin in sheep transplanted in utero with human mesenchymal stem cells. Exp Hematol. 2010;38(4):311–20.

    Article  CAS  Google Scholar 

  14. Ma MT, Leung KK, Tsang KS, Leung PS. Reduced immunogenicity of pancreatic progenitor cells derived from first-trimester human fetal pancreas. Int J Biochem Cell Biol. 2011;43(5):812–20.

    Article  CAS  Google Scholar 

  15. Sensebé L, Bourin P. Mesenchymal stem cells for therapeutic purposes. Transplantation. 2009;87(9 Suppl):S49-53.

    Article  Google Scholar 

  16. Doucet C, Ernou I, Zhang Y, Llense JR, Begot L, Holy X, et al. Platelet lysates promote mesenchymal stem cell expansion: a safety substitute for animal serum in cell-based therapy applications. J Cell Physiol. 2005;205(2):228–36.

    Article  CAS  Google Scholar 

  17. Halme DG, Kessler DA. FDA regulation of stem-cell-based therapies. N Engl J Med. 2006;355(16):1730.

    Article  CAS  Google Scholar 

  18. Turnovcova K, Ruzickova K, Vanecek V, Sykova E, Jendelova P. Properties and growth of human bone marrow mesenchymal stromal cells cultivated in different media. Cytotherapy. 2009;11(7):874–85.

    Article  CAS  Google Scholar 

  19. Aldahmash A, Haack-Sørensen M, Al-Nbaheen M, Harkness L, Abdallah BM, Kassem M. Human serum is as efficient as fetal bovine serum in supporting proliferation and differentiation of human multipotent stromal (mesenchymal) stem cells in vitro and in vivo. Stem Cell Rev Rep. 2011;7(4):860–8.

    Article  Google Scholar 

  20. Pharmaceutical Inspection Convention and Pharmaceutical Inspection Co-operation Scheme. Guide to Good Manufacturing Practice for medicinal products. Annex 2A Manufacture of advanced therapy medicinal products for human use. Geneva: PIC/S; 2021.

  21. Al-Saqi SH, Saliem M, Quezada HC, Ekblad Å, Jonasson AF, Hovatta O, et al. Defined serum-and xeno-free cryopreservation of mesenchymal stem cells. Cell Tissue Bank. 2015;16(2):181–93.

    Article  CAS  Google Scholar 

  22. Kocaoemer A, Kern S, Klüter H, Bieback K. Human AB serum and thrombin-activated platelet-rich plasma are suitable alternatives to fetal calf serum for the expansion of mesenchymal stem cells from adipose tissue. Stem cells. 2007;25(5):1270–8.

    Article  CAS  Google Scholar 

  23. Chimenti I, Gaetani R, Forte E, Angelini F, De Falco E, Zoccai GB, et al. Serum and supplement optimization for EU GMP-compliance in cardiospheres cell culture. J Cell Mol Med. 2014;18(4):624–34.

    Article  CAS  Google Scholar 

  24. Larijani B, Arjmand B, Ahmadbeigi N, Falahzadeh K, Soleimani M, Sayahpour FA, et al. A simple and cost-effective method for isolation and expansion of human fetal pancreas derived mesenchymal stem cells. Arch Iran Med. 2015;18(11):770–5.

    PubMed  Google Scholar 

  25. Krampera M, Galipeau J, Shi Y, Tarte K, Sensebe L. Immunological characterization of multipotent mesenchymal stromal cells. The international society for cellular therapy (ISCT) working proposal. Cytotherapy. 2014;16(4):S81.

    Article  Google Scholar 

  26. González-González R, Molina-Frechero N, Carreón-Burciaga RG, López-Verdín S, Robles-Bonilla C, Pereira-Prado V, et al. Comparison between manual and automated methods for ki-67 immunoexpression quantification in ameloblastomas. Anal Cell Pathol. 2016;2016:7486989.

    Article  Google Scholar 

  27. WHO Expert Committee on Biological Standardization. WHO Technical Report Series 978 Sixty-First Report. Annex 3. Geneva, Switzerland: WHO; 2013. p. 131–9.

  28. Borowicz S, Van Scoyk M, Avasarala S, Rathinam MKK, Tauler J, Bikkavilli RK, et al. The soft agar colony formation assay. J Vis Exp. 2014. https://doi.org/10.3791/51998.

    Article  PubMed  PubMed Central  Google Scholar 

  29. Peloso A, Citro A, Zoro T, Cobianchi L, Kahler-Quesada A, Bianchi CM, et al. Regenerative medicine and diabetes: targeting the extracellular matrix beyond the stem cell approach and encapsulation technology. Front Endocrinol. 2018;9:445.

    Article  Google Scholar 

  30. Cianfarani F, Toietta G, Di Rocco G, Cesareo E, Zambruno G, Odorisio T. Diabetes impairs adipose tissue–derived stem cell function and efficiency in promoting wound healing. Wound Repair Regen. 2013;21(4):545–53.

    Article  Google Scholar 

  31. Rennert RC, Sorkin M, Januszyk M, Duscher D, Kosaraju R, Chung MT, et al. Diabetes impairs the angiogenic potential of adipose-derived stem cells by selectively depleting cellular subpopulations. Stem Cell Res Ther. 2014;5(3):1–12.

    Article  Google Scholar 

  32. Arjmand B, Aghayan HR. Cell manufacturing for clinical applications. Stem Cells. 2014;32(9):2557–8.

    Article  Google Scholar 

  33. Ball O, Zylberberg C. Towards a common framework for defining ancillary material quality across the development spectrum. Cytotherapy. 2019;21(12):1234–45.

    Article  Google Scholar 

  34. Inamdar AA, Inamdar AC. Culture conditions for growth of clinical grade human tissue derived mesenchymal stem cells: comparative study between commercial serum-free media and human product supplemented media. J Regen Med Tissue Eng. 2013;2:10.

    Article  Google Scholar 

  35. Shahdadfar A, Frønsdal K, Haug T, Reinholt FP, Brinchmann JE. In vitro expansion of human mesenchymal stem cells: choice of serum is a determinant of cell proliferation, differentiation, gene expression, and transcriptome stability. Stem cells. 2005;23(9):1357–66.

    Article  CAS  Google Scholar 

  36. Jung J, Moon N, Ahn J-Y, Oh E-J, Kim M, Cho C-S, et al. Mesenchymal stromal cells expanded in human allogenic cord blood serum display higher self-renewal and enhanced osteogenic potential. Stem cells Develop. 2009;18(4):559–72.

    Article  CAS  Google Scholar 

  37. de Paula ACC, Zonari AAC, Martins TMDM, Novikoff S, da Silva ARP, Correlo VM, et al. Human serum is a suitable supplement for the osteogenic differentiation of human adipose-derived stem cells seeded on poly-3-hydroxibutyrate-co-3-hydroxyvalerate scaffolds. Tissue Eng Part A. 2013;19(1–2):277–89.

    Article  Google Scholar 

  38. Gregory CA, Reyes E, Whitney MJ, Spees JL. Enhanced engraftment of mesenchymal stem cells in a cutaneous wound model by culture in allogenic species-specific serum and administration in fibrin constructs. Stem Cells. 2006;24(10):2232–43.

    Article  CAS  Google Scholar 

  39. Kuznetsov SA, Mankani MH, Robey PG. Effect of serum on human bone marrow stromal cells: ex vivo expansion and in vivo bone formation. Transplantation. 2000;70(12):1780–7.

    Article  CAS  Google Scholar 

  40. Wiese C, Rolletschek A, Kania G, Blyszczuk P, Tarasov K, Tarasova Y, et al. Nestin expression–a property of multi-lineage progenitor cells? Cell Mol Life Sci CMLS. 2004;61(19):2510–22.

    Article  CAS  Google Scholar 

  41. Chen S, Du K, Zou C. Current progress in stem cell therapy for type 1 diabetes mellitus. Stem Cell Res Ther. 2020;11(1):1–13.

    Article  Google Scholar 

  42. Joglekar MV, Joglekar VM, Joglekar SV, Hardikar AA. Human fetal pancreatic insulin-producing cells proliferate in vitro. J Endocrinol. 2009;201(1):27.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

The authors acknowledge Dr. Hossein Adibi and Dr. Mohsen Khorshidi for their administrative support and also for their valuable contribution to facilitating the fetal donation process.

Funding

This work was supported by the Endocrinology and Metabolism Research Institute, Tehran University of Medical Sciences, Tehran, Iran [grant number: 1392–02-107–1713].

Author information

Authors and Affiliations

Authors

Contributions

HRA contributed to the conception and design of the study. KF, SA, SAM, and ZJ contributed to material preparation, cell culture experiments, and laboratory testing. SA done the animal experiments. HRA, BA, and BL performed acquisition, analysis, and interpretation of data. The first draft of the manuscript was prepared by ZJ. HRA performed critical review and approval of the submitted manuscript. All authors read and approval the final manuscript.

Corresponding author

Correspondence to Hamid Reza Aghayan.

Ethics declarations

Ethics approval and consent to participate

This study was approved by the Ethics Committee of Endocrinology and Metabolism Research Institute, Tehran University of Medical Sciences, Tehran, Iran (IRB code: EC-00264). All studies were carried out in compliance with the ARRIVE guidelines (https:// arriveguidelines. org) for the reporting of animal experiments. All methods were performed in accordance with the relevant guidelines and regulations.

Consent for publication

Not applicable.

Competing interests

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

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Jabbarpour, Z., Aghayan, S., Arjmand, B. et al. Xeno-free protocol for GMP-compliant manufacturing of human fetal pancreas-derived mesenchymal stem cells. Stem Cell Res Ther 13, 268 (2022). https://doi.org/10.1186/s13287-022-02946-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13287-022-02946-5

Keywords