Skip to main content

Efficient generation of functional pancreatic β cells from dental-derived stem cells via laminin-induced differentiation

Abstract

Background

This study was designed to generate functional insulin-producing cells (IPCs) from dental-derived mesenchymal stem cells (MSCs) and further explore their therapeutic potential against diabetes mellitus in vivo. MSCs were isolated from human dental pulp and periodontal ligament and were induced to differentiate into insulin-producing cells (IPCs) using laminin-based differentiation protocol for 14 days. Confirmation of IPCs was performed through real-time PCR analysis and insulin release assay. Then, the generated IPCs were labeled with PKH26 dye prior to transplantation in experimental animals. Twenty-eight days later, blood glucose, serum insulin (INS), c-peptide (CP), and visfatin (VF) levels and pancreatic glucagon (GC) level were estimated. Pancreatic forkhead box protein A2 (Foxa2) and SRY-box transcription factor 17 (Sox17), insulin-like growth factor I (IGF-1), and fibroblast growth factor10 (FGF 10) gene expression levels were analyzed.

Results

Dental stem cells were successfully differentiated into IPCs that demonstrated increased expression of pancreatic endocrine genes. IPCs released insulin after being subjected to high levels of glucose. In vivo findings uncovered that the implanted IPCs triggered significant decrease in blood glucose, serum VF, and pancreatic GC levels with significant increase in serum INS and CP levels. Furthermore, the implanted IPCs provoked significant upregulation in the expression level of pancreatic genes. Histopathological description of the pancreas tissues revealed that transplantation of IPCs ameliorated the destabilization of pancreas tissue architecture.

Conclusion

This study demonstrates the significant role of the implantation of IPCs generated from dental-derived stem cells in treatment of diabetes mellitus.

Background

Diabetes mellitus (DM) is a debilitating disease that affects millions of people and results in serious complications that could lead to systemic impairment and even mortality. Currently, available therapies, vary from insulin-based treatments to beta cell replacement altogether either via the pancreas or pancreatic islet transplantation. However, such interventions are limited by the availability of the organs. Stem cell-based therapies offer an alternative treatment option for diabetes through sequential differentiation of stem cells into beta cells that are able to react to blood glucose and consequently synthesize and secrete insulin [6].

β cells differentiated from mesenchymal stem cells are considered the most auspicious source of cells for diabetic cell based therapies [34, 55]. Dental-derived stem cells in particular are undoubtedly considered an attractive source of MSCs [1]. They are easy to obtain through non-invasive procedures where the donors suffer no discomfort. Furthermore, there is no ethical controversy involved in acquiring the teeth, since dental stem cells are usually isolated from the teeth that are originally indicated for extraction whether due to impaction or for orthodontic treatment purposes [27, 46, 59].

Several differentiation protocols have been devised, which lead to differentiation of MSCs into insulin-producing cells (IPCs). These differentiation protocols work either through genetic manipulation of the cells or through the epigenetic effects elicited by the factors comprising the differentiation media. However, it should be noted that differentiation of stem cells into β cells in vitro is not a single step procedure, but rather a sequential series of steps that mimics pancreatic development [40].

There is compelling evidence that indigenous extracellular matrix (ECM) promotes stem cell survival and to regulate stem cell differentiation through maintaining its inherent structural, biochemical, and biomechanical properties [28, 38, 50]. Laminin specifically, which is a major constituent of the basement membrane, plays a role in supporting cell proliferation and differentiation [22]. During organ development, laminins interplay with several other ECM molecules and connect cells through cell surface receptors, which results in modulating cellular phenotypes and differentiation [24, 51, 53]. Recent studies reported that, laminin-411 promoted islet function in the developing pancreas. Moreover, it proved to be found crucial for beta cell proliferation and insulin maintenance [43]. Laminins also induce expression of islet-specific factors like pancreatic and duodenal homeobox 1 (PDX1), insulin 1, insulin 2, glucagon, somatostatin, and GLUT-2. They also activate protein kinase B and extracellular signal-regulated kinase, which are major factors in cell metabolism that are capable of inducing differentiation of beta cells [60].

In this study, we investigated the effect of Laminin 411 on the differentiation of human dental pulp stem cells (hDPSCs) and human periodontal ligament stem cells (hPDLSCs) into IPCs through applying a previously published protocol intended for human umbilical cord stem cells. Moreover, the therapeutic effect of the acquired IPCs was also validated on DM animal model.

Methods

Isolation and propagation of mesenchymal stem cells

Isolation and preparation of human dental pulp stem cells (hDPSCs)

Human third molar teeth indicated for extraction were collected from patients aged (16-24) after obtaining an informed consent. Extraction of teeth (n=3) was carried out at the National Research Centre’s clinics under the approval of the Centre's ethical committee. The extracted teeth were kept in Dulbecco modified Eagle’s media (Lonza) to which penicillin/streptomycin (Lonza), and 10% of FBS (Life Science Group) were added. The teeth were split open, and the pulp was gently removed using sterile dental probe. The pulp tissue was then carefully dissected into small pieces using sterile scissors. 0.2% collagenase II (Serva Electrophores, Germany) solution was then added to the dissected pulp tissue and shaken in a water bath shaker at 37°C for 30 min. The isolated dental pulp cells were cultured in fresh DMEM media supplemented with 15% FBS and incubated at 37°C and 5% CO2. Cells were selected on the basis of their ability to adhere to the tissue culture plastic; non adherent cells were removed during medium replacement. The medium was changed twice per week thereafter. Dental pulp cells were subsequently be detached using 0.25% trypsin/1-mM ethylene diaminetetraacetic acid (EDTA) re-plated at 5 × 103 cells/cm2 and cultured then passaged at 70 to 80% confluency.

Isolation of human periodontal ligament stem cells (hPDLSCs) cells

Healthy human impacted third molars (n=3) (free of inflammation) were placed in 50-ml sterile polypropylene tube containing alpha modified Eagle’s medium (alpha-MEM) supplemented with antibiotics (100 U/ml penicillin and 100 mg/ml streptomycin) immediately after extraction. The periodontal ligament tissue was removed from the surface of the root by sterile lancet, rinsed five times by PBS supplemented with penicillin and streptomycin. Periodontal ligament pieces were minced into tiny pieces, then digested in a solution of 2 mg/ml collagenase type II (Serva Electrophores, Germany) for 30 min at 37°C in a water bath. Single-cell suspensions were seeded into a T-25 flask with D-MEM (Lonza), supplemented with 15% fetal bovine serum (Lonza), 20,000 U/ml penicillin and 20,000 ng/ml streptomycin and then incubated in of 5% CO2 at 37°C. The flasks were periodically monitored by the phase contrast-inverted microscopy and the culture medium was changed 3-times per week, and the cells were sub-cultured when about 80% confluency was reached using 0.25% trypsin EDTA.

hDPSC and hPDLSC characterization

Morphological follow up by inverted microscope examination

Morphological changes were observed via an inverted light microscope (Leica, 6000B-4) using Suite V3 (Leica). Cells were regularly monitored and images were captured.

Flow cytometry analysis

To ensure that the cells in cultures were MSCs that maintained their phenotype after propagation in culture [12], the isolated cells were characterized by flow cytometry analysis for MSC-specific markers (CD73, CD105) in addition to hematopoietic markers (CD34 and CD45). The PE conjugated-CD 73 and FITC conjugated-CD 105 antibodies were procured from R&D Systems (UK). While the PE conjugated-CD 34 as well as FITC-conjugated CD 45 antibodies were purchased from Beckman Coulter Co. (USA). The cells were incubated with the antibody against each of the surface markers for 20 min at 25°C followed by flow cytometry analysis using Beckman Coulter Elite XL, USA, instrument.

Tri-lineage mesodermal differentiation

Both hDPSCs and hPDLSCs were differentiated into adipocytes upon culturing them with StemPro® adipogenesis differentiation medium provided by Gibco, Thermo Fisher Scientific Inc. (USA), for 14 days. Thereafter, adipogenic cultures were processed for Oil Red O staining (Sigma, USA) to detect the presence of highly refractive intracellular lipid vacuoles containing accumulated lipid droplets. Chondrocyte differentiation was carried out in turn by culturing the isolated MSCs with StemPro® chondrogenesis differentiation medium provided by Gibco, Thermo Fisher Scientific Inc. (USA). After 14 days under differentiating conditions, the cells were fixed with 4% formaldehyde solution for 30 min. After fixation, the wells were rinsed with PBS and stained with 1% Alcian Blue (Sigma, USA) solution prepared in 0.1 N HCL for 30 min. The wells were visualized under light microscope. Blue staining indicated synthesis of proteoglycans by chondrocytes. As for the osteogenic differentiation potential, the isolated MSCs were induced into the osteogenic pathway through culturing in StemPro® osteogenesis differentiation medium provided by Gibco, Thermo Fisher Scientific Inc. (USA). After 21 days, the cells were fixed with 4% formaldehyde solution for 30 min. The wells were then rinsed with PBS, and the cells were stained with 2% Alizarin Red S (Sigma, USA) solution (pH 4.2) for 2–3 min. The wells were visualized under light microscope for presence of calcium-rich mineralized nodules.

Differentiation of hDPSCs & hPDLSCs into insulin-producing cells (IPCs)

After the third passage, both hDPSCs and hPDLSCs were induced to differentiate into IPCs (Fig. 1). The protocol for differentiation was implemented as described by Qu et al. [43]. Both hDPSCs and hPDLSCs were suspended in complete culture media, and aliquots of 2.5 × 105 cells were placed in 6-well plates pre-coated with 5 μg/mL laminin (Gibco) overnight. The medium was replaced with HG-DMEM containing 10% FBS for 3 d (stage I). At stage II, the media were refreshed with DMEM/F-12 (Lonza) medium containing 2% FBS and 1% Insulin-Transferin-Selenium-A (ITS-A, Gibco) for another 4 days. At stage III, 10-mM nicotinamide was added into the media described in stage II, and the culture lasted for 3 days. At stage IV, the medium was changed with the same supplements as at stage III, but 1% N2 and 1% B27 supplements (STEMCELL Technologies Inc.) were added and incubated for 4 days.

Fig. 1
figure 1

Schematic diagram outlining the differentiation protocol for dental derived stem cells (hDPSCs & hPDLSCs) into insulin-producing cells (IPC). Durations and required factors for each differentiation step are outlined

Determination of the efficiency of differentiation of hDPSCs and hPDLSCs into insulin-producing cells (IPCs)

Quantitative real-time PCR analysis of IPCs

The total RNA was isolated from the IPCs generated from hDPSCs and hPDLSCs using Trizol (Invitrogen) according to the manufacturer’s instructions. The first-strand cDNA was synthesized with 50 ng of the total RNA by random hexamer priming using high-capacity cDNA synthesis kit (Qiagen) at 42°C for 60 min and at 70°C for 5 min. The quantitative real-time PCR was performed in triplicates using SYBR-green kit (Qiagen) according to the manufacturer’s instructions and using the Roche LC480 in a total reaction volume of 20 μl. Results were normalized to GAPDH (internal control) to correct RNA input in reactions. All reactions were performed by annealing at 58–60°C for 40 cycles, and the melt curve analysis was achieved at the end of each reaction. The primer sequences of the target genes are listed in (Table 1).

Table 1 List of human gene-specific primers in RT-PCR

Insulin release assay

The generated IPCs from both hDPSCs and hPDLSCs were gently washed twice with phosphate buffer saline (PBS, Biowest, France). Then, the cells were pre-incubated in culture media containing glucose (5.5 mM) at 37°C for 24 h [52], and supernatants were then collected for insulin quantification via ELISA technique using the kit procured from Epitope Diagnostics, Inc. (USA), according to the manufacturer’s instructions.

Labeling of the generated IPCs with PKH26 dye

The generated IPCs were harvested and labeled with PKH26 (Sigma-Aldrich, USA) fluorescent linker dye according to the manufacturer’s instructions prior to transplantation in experimental animals. The pancreatic tissues were examined using fluorescence microscope to detect and trace the cells. PKH26-derived fluorescence was detected by inverted fluorescent microscope (Olympus, CKX41, Japan).

Induction of DM and treatment of diabetic rats with IPCs

Animals

In total, 40 adult male rats (Sprague Dawley) weighing 150–170 g were supplied from a breading stock maintained in the Animal House of the  National Research Centre, Giza, Egypt. The animal house was ventilated with a 12-h light/dark cycle at the ambient temperature of 25–30°C throughout the experimental period with free access to tap water and a standard rodent chow (Meladco Co.). Rats were allowed to adapt to their environment for at least 2 weeks before the initiation of the experiment. Housing and management of animals and experimental protocol followed the guidelines of animal experiments and was approved by the Ethical Committee of the  Medical Research of the National Research Centre, Egypt (approval no. 19 109).

Study protocol

Ten rats served as the negative control group and received no drug (Group 1), while other rats were included in the induction of diabetes mellitus. Briefly, diabetes was induced via a single subcutaneous injection of streptozotocin (STZ, Sigma, USA) (50 mg/kg) following an overnight fast. Streptozotocin was dissolved in a 50-mM sodium citrate solution (pH 4.5) containing 150 mM NaCl. Fasting blood sugar was estimated after 3 days to confirm the development of diabetes mellitus using the kit supplied from Chemelex S.A. (Spain) [19]. Animals showing blood glucose levels above 250 mg/dL were included in the study.

Thereafter, diabetic animals were randomly divided into 3 experimental groups and were left without treatment (Group 2, n =10, DM untreated), transfused via tail vein with 5 × 106 cells/rat of IPCs generated from human DPSC differentiation (Group 3, n = 10, DM + hDPSCs), and finally transfused via tail vein with 5 × 106 cells/rat of IPCs generated from human PDLSCs differentiation (Group 4, n = 10, DM + hPDLSCs).

Twenty-eight days following the infusion of IPCs, the diets were withheld from the experimental rats for 12 h and then blood samples were collected, under diethyl ether anesthesia, from the retro-orbital venous plexus in clean centrifuge tubes and allowed to coagulate at room temperature. Serum samples were separated by centrifugation at 1800×g for 15 min at 4°C using cooling centrifuge for biochemical measurements and retained at −20°C till analysis. After the collection of blood samples, the rats were euthanized by cervical dislocation and a middle abdominal incision was performed and the pancreas of each rat were quickly dissected out and washed in ice cold saline. Some of pancreas tissues were immediately weighed and homogenized in ice-cold medium containing 50-mM Tris-HCl (pH 7.4) to give 10% homogenate (W/V) [19]. While others were immediately frozen in liquid nitrogen and kept at −80°C preceding the molecular genetics investigation. Lastly, the rest of the pancreas was placed in neutral-buffered formalin (10%) to be used for histopathological investigations.

Evaluation of IPC therapeutic efficacy of the generated IPCs

Biochemical analysis

Serum glucose level was estimated using the kit supplied from Chemelex S.A. (Spain). While serum insulin (INS), c-peptide (CP), and visfatin (VF) levels were determined by ELISA using the specific kits for rat supplied from SinGeneClon Biotech Co., Ltd, (China) according to the manufacturer’s manuals. Pancreatic glucagon (GC) level was determined by ELISA using the rat specific kit procured from SinGeneClon Biotech Co., Ltd, (China) following the manufacturer’s instructions.

Molecular genetic analysis

The total RNA was isolated from the pancreatic tissue of rats of different experimental groups using Trizol reagent (Invitrogen, USA. Cat #15596-018) in combination with RNeasy mini kit for total RNA purification from animal cells (Cat#74104, Qiagen, Germany) according to the manufacturer’s instruction. The RNA integrity was evaluated using Nano Drop 2000 (Thermo Fisher Scientific, Rockford, IL, USA) using 260/280 nm ratio. Then, the cDNA synthesis was performed using Revert Aid first-strand cDNA synthesis kit (Cat# K1621, Thermo Fisher Scientific, Inc., Lithuania) according to the manufacturer’s instruction. Gene expression analysis of forkhead box protein A2 (Foxa2) and SRY-box transcription factor 17 (Sox17), insulin-like growth factor I (IGF-1), and fibroblast growth factor (FGF) 10 was carried out in rat pancreatic tissues using DNA-Technology Real-Time PCR device (DT lite 4, Russia). The reaction mixture (25 μl volume) included 12.5 μl of QuantiTect SYBR Green master mix (Cat# 204141, Qiagen, Germany), 0.75 μl of forward and reverse primer of target gene (Invitrogen, USA), cDNA template (100 ng), and RNase free water. GAPDH was used as a housekeeping gene. Relative mRNA expression versus control value was assessed using the 2-ΔΔCt comparative method after normalization with GAPDH gene. The PCR cycling was set as follows: initial denaturation step at 94°C for 15 min, followed by 40 cycles of denaturation at 94°C for 15 s, annealing at 60°C for 30 s, and extension at 72°C for 30 s for 5 min. The primer sequences of each target gene are delineated in Table 2. The data were presented as the fold change in gene expression level in the diabetic group relative to the control group. While the data were represented as the fold change in gene expression as compared to diabetic group in case of treated groups.

Table 2 List of gene-specific primers in RT-PCR

Histopathological procedure

The pancreas tissues were fixed in 10% neutral-buffered formalin for 24 h, washed with tap water, and prepared and stained for light microscopy. For dehydration, serial dilutions of alcohol were applied, and thereafter, the specimens were cleared in xylene and embedded in paraffin wax in hot air oven at 56°C for 6 h. The paraffin wax tissue blocks were sectioned by using microtome at 5–6 micron thickness. Then, sections were collected on glass slides and de-paraffinized. They were stained for routine histological examination using hematoxylin and eosin stain [11].

Statistical analysis

The experimental results were represented as arithmetic means with their standard deviation (SD). Data were statistically analyzed by one-way analysis of variance (ANOVA) test using the Statistical Package for the Social Sciences (SPSS) 14 followed by determining the least significant difference (LSD) to evaluate the significance between groups. P < 0.05 was considered significant.

Results

Morphological appearance of isolated mesenchymal stem cells hDPSCs & hPDLSCs

The photomicrographs presented in Fig. 2 illustrate the morphological appearance of cells after isolation. Images indicated that MSCs derived from human dental pulp and periodontal ligament exhibited spindle-shaped, fibroblastic-like morphology which is characteristic to MSCs.

Fig. 2
figure 2

Photomicrograph illustrating the morphological changes of hDPSCs & hPDLSCs following isolation. In both cultures of hDPSCs and hPDLSCs after approximately 3 days of culture, scattered single cells started to attach to the culture plate assuming a spindle-shaped appearance (A). Gradually, these spindle-shaped cells started to increase in number, and colonies were evident by day 5 of culture (B). These colonies increased in size and reached 80% confluence by the 10th day of culture in hDPSCs (C) and by day 15 in hPDLSCs (D) (scale bar 100μm)

Mesenchymal stem cell surface markers

Both human dental pulp and human periodontal ligament-derived MSCs characterized by flow cytometry illustrated positive expression of mesenchymal stem cell markers CD 73 and CD 105 and negative expression of non-mesenchymal stem cell markers CD 34 and CD 45 (Fig. 3).

Fig. 3
figure 3

Flow cytometry analysis of cell surface markers. Mesenchymal stem cell surface markers; CD 73 and CD 105 were positively expressed in both hDPSCs and hPDLSCs whereas hematopoietic markers CD 34 and CD 45 were negatively expressed in both cell types

Tri-lineage mesodermal differentiation

Following in vitro differentiation, both hDPSCs and hPDLSCs were successfully differentiated into adipocytes. Intracellular lipid droplet characteristic of adipocytes was stained with Oil red O. Similarly, both hDPSCs and hPDLSCs were successfully differentiated into chondrocytes as indicated by Alcian blue staining of the deposited sulfated proteoglycans. Also, successful differentiation into osteoblasts confirmed by the presence of calcific deposits was illustrated by Alizarin red S (Fig. 4).

Fig. 4
figure 4

Multi-lineage differentiation of hDPSCs (A) and hPDLSCs (B). In order to confirm the stem cell identity of the isolated cells from dental pulp and periodontal ligament, multi-lineage differentiation potential of those cells into adipogenic, chondrogenic, and osteogenic lineages was assessed. Osteogenic differentiation of hDPSCs & hPDLSCs was evaluated by Alizarin red staining. Cells assumed a polygonal shape indicating successful differentiation and also dark orange stains indicating nodules of calcification were demonstrated in both groups (left). Chondrogenic differentiation of DPSCs & PDLSCs was assessed by Alcian Blue staining (middle). Cells demonstrated morphology similar to chondrocytes rather than the typical spindle-shaped appearance of stem cells. Blue staining was demonstrated indicating the presence of sulfated glycosaminoglycans by differentiated chondrocytes. Adipogenic differentiation of hDPSCs and hPDLSCs was confirmed by Oil Red O staining (right) and demonstrated lipid droplets in differentiated cells. Accumulation of intracellular lipid droplets can be seen scattered denoting successful differentiation (scale bar 100μm)

Confirmation of IPC generation

Gene expressions of IPCs generated through differentiation of hDPSCs and hPDLSCs

Following the induction of both hDPSCs and hPDLSCs with the Laminin 411-enriched differentiation protocol for 14 days, the derived IPCs from hDPSCs demonstrated increased expression pancreatic endocrine genes (Foxa-2, Sox-17, PDX-1, Ngn-3, insulin, and glucagon) in comparison with undifferentiated hDPSCs as illustrated in Fig. 5a. Moreover, the generated cells from human PDLSC differentiation exhibited significant (P < 0.05) upregulation in the expression level of PDX-1 and insulin genes when compared with the undifferentiated hPDLSCs (Fig. 5b).

Fig. 5
figure 5

Expression of pancreatic endocrine genes in generated IPCs from hDPSCs (A) and hPDLSCs (B). Data are expressed as means ± SD. *Significant change at P < 0.05 in comparison with the corresponding undifferentiated cells

In vitro response to glucose stimulation

Data in Fig. 6 illustrated that the generated IPCs from hDPSCs and hPDLSCs secreted compelling quantities of insulin in response to escalated glucose concentration (P < 0.05) as to undifferentiated cells.

Fig. 6
figure 6

Insulin release of generated IPCs from hDPSCs and hPDLSCs. Data are expressed as means ± SD. *Significant change at P < 0.05 in comparison with the corresponding undifferentiated cells

Homing of transplanted IPCs into the pancreatic tissues

Successful homing of the injected IPCs in the pancreas of treated groups was confirmed by tracking cells labeled by PKH-26 dye. Figure 7 illustrates multiple PKH-26-labeled cells in pancreatic sections.

Fig. 7
figure 7

Homing of systemically administered IPCs in pancreas sections of different treated groups. A IPCs generated from hDPSCS. B IPCs generated from hPDLCSs (scale bar 100μm)

Therapeutic efficacy of IPCs against DM in experimental animals

Influence of IPCs on diabetic indices

Figure 8 represented the effect of the transplanted IPCs on the blood glucose, serum INS, CP, and VF as well as pancreatic GC levels in STZ-induced diabetes. The diabetic rats that were not treated exhibited significant (P > 0.05) increase in fasting blood glucose, serum VF, and pancreatic GC levels associated with significant (P > 0.05) decline in serum INS and CP levels as to the negative control. While transplantation of the IPCs in diabetic rats elicited significant (P > 0.05) decrease in the blood glucose, serum VF, and pancreatic GC levels in concomitant with significant (P > 0.05) increase in serum INS and CP levels versus the untreated diabetic rats.

Fig. 8
figure 8

Effect of IPC implantation on (i) blood glucose, serum (ii) INS, (iii) CP, and (iv) VF as well as (v) pancreatic GC levels. Data are expressed as means ± SD of 8 rats/group. Significant change at P < 0.05 in comparison with the negative control group denoted by (a). Significant change at P < 0.05 in comparison with the untreated DM group denoted by (b)

Influence of IPCs on the expression level of pancreatic Foxa2, Sox17, IGF-1, and FGF10

Figure 9 demonstrates the effect of transplanted IPCs on the expression level of pancreatic Foxa2, Sox 17, IGF-1, and FGF10 genes in diabetic rats. The diabetic rats that received no treatment demonstrated significant (P> 0.05) decrease in the expression level of pancreatic Foxa2, Sox 17, IGF-1, and FGF 10 genes in comparison with the control group, while transplanting IPCs in diabetic rats resulted in a significant (P> 0.05) increase in the expression level of pancreatic Foxa2, Sox 17, IGF-1, and FGF 10 genes when compared with the untreated DM rats.

Fig. 9
figure 9

Effect of transplanted IPCs on the expression level of (i) Foxa2, (ii) Sox 17, (iii) IGF-1, and (iv) FGF 10 genes in diabetic rats. Data are expressed as means ± SD of 8 rats/group. Significant change at P > 0.05 denoted by (a) in comparison with the negative control group. Significant change at P > 0.05 in comparison with the untreated DM group

Influence on histological architecture of the pancreatic tissues of diabetic rats

Histological sections of the pancreas tissues of the control rats illustrated no histopathological alterations in the endocrine compartment islands of Langerhans cells which presented well-defined margins and plump islet cells. The exocrine acini together with acinar cells stained blue at their base due to the existence of a high content of RNA. Their apices stained pink where there was high content of digestive enzymes, in addition to the duct system of the exocrine portion (Fig. 10A). While the histological sections of the pancreas tissues of diabetic rats that received no treatment showed widespread destruction of Langerhans islets, the nuclei were small condensed and darkly stained, along with congested capillaries. Moreover, severe infiltration of inflammatory cells were also apparent (Fig. 10B). Histological sections of the pancreatic tissues of diabetic rats infused with IPCs generated from differentiation of hDPSCs presented normal pancreatic parenchyma with few inflammatory cell infiltration (Fig. 10C). Finally, histological examination of the pancreatic tissues of the diabetic rats treated with IPCs generated from differentiation of hPDLSCs showed moderate-sized islets of Langerhans with well-defined edges (Fig. 10D).

Fig. 10
figure 10

Histological section of the pancreas tissue. A Normal control rat showed intact pancreatic parenchyma (black star) and normal presentation of islets of Langerhans (yellow star). B The pancreatic tissue of untreated diabetic rat showed degenerated and disintegrated islets of Langerhans (black arrow) and heavy inflammatory cell infiltrations (green arrow). C The pancreatic tissue of diabetic rat infused with hDPSCs showed normal pancreatic parenchyma (yellow star) with few inflammatory cell infiltration (green arrow). D The pancreatic tissue of diabetic rat infused with hPDLSCs showed moderate sized islets of Langerhans (yellow star) and some inflammatory cell infiltration (scale bar 200μm)

Discussion

In this approach, we investigated the ability of dental-derived stem cells to differentiate into pancreatic β cells through implementing a sequential differentiation protocol originally designed for human umbilical cord mesenchymal stem cell [43]. The utilization of dental derived stem cells in this study was due to the remarkable ability of these cells to proliferate and differentiate into multiple cell types. Dental-derived stem cells are easily accessible by routine dental treatments with minimal invasive procedures. That is mainly what renders dental-derived stem cells, the cells of choice for the future of cell-based therapies. Teeth are usually extracted using local anesthesia, contrary to other sources of stem cells, which necessitates general anesthesia. Both the third molars and exfoliated deciduous teeth are considered a common source of DPSCs. Wisdom molar extraction is a frequent procedure in dental clinics. Moreover, it is the final tooth to form and is considered a valuable repository of immature cells in the former stages of odontogenesis [8, 9]. These cells are of tremendous therapeutic potential owing to their neural crest origin; additionally, they can be differentiated into various cell types, like hepatic cells and pancreatic β cells [32]. Similarly, exfoliated deciduous teeth are easily obtained from children whose age ranges from 6 to 16 years and share the same properties as those of third molars [45].

Our microscopic observation of the isolated cells indicated that MSCs derived from human dental pulp and periodontal ligament exhibited clear spindle-shaped, fibroblastic-like morphology. Such findings are supported by those of Miletić et al. [33] and Patil et al. [42] [33, 42]. In addition, both the human dental pulp and human periodontal ligament-derived MSCs are characterized by flow cytometry demonstrated positive expression of CD 73 (94.2%), CD 90 (75.8%), and CD 105 (81.9%), but were negative for CD 14 (5.56%), CD 34 (3.38%), and CD 45 (10.6%).

Pancreatic organogenesis spans a distinct interplay of factors that, together with other processes, directs stem cells to differentiate into pre-pancreatic cells and subsequently the development of a mature and functional organ. Thus, most stem cell-based differentiation strategies are mainly directed at producing mature, insulin-expressing β cells, that are reactive to glucose through mimicking the stages of pancreatic development [30, 35].

It is believed that the selection of a suitable protocol to differentiate cells is critical to the success of the differentiation [36, 60]. Additionally, the type of inductive factors used can affect the efficiency of differentiation via different pathways [3]. Differentiation can be improved through eliciting the expression of pancreatic genes such as Pdx1 and insulin which in turn results in increased insulin secretion in response to glucose stimulation [15, 44, 54]. We implemented an established endodermal lineage differentiation protocol that is based on the use of Laminin 411. In this protocol, laminin 411-induced differentiation of IPCs from umbilical cord-derived stem cells by way of both Pdx1 and Ngn3 pathways [43]. Results reported by this group indicated marked improvement in the differentiation ability of umbilical cord MSCs into IPCs. Also, improvement of diabetic symptoms and overall survival of diabetic rats were described.

Naturally, multiple extracellular matrix ECM proteins exist in the basement membranes of cells; however, the most eminent of which are collagen, laminins, and vitronectin. They were reported to contribute to β cell growth and viability by interacting with integrin receptors on cell surfaces. Such interactions facilitate cell migration and guide primitive streak formation and gastrulation [7]. Several research groups investigated the impact of various extracellular matrix proteins on enhancing the ability of stem cells to differentiate [13, 14, 57, 60, 61]. In fact, our group previously studied the role of recombinant human Vitronectin-N (VN) as an extracellular matrix protein on the induction of endodermal lineage differentiation from human-induced pluripotent stem cells (iPSCs). VN efficiently promoted differentiation of iPSCs into pancreatic-committed gut endoderm [5]. Similarly, Hsiao-Yun et al. reported that IPC differentiation by MSCs was remarkably improved when extracellular matrix proteins were added to the differentiation media.

In the current study, our results revealed that upon culturing dental-derived stem cells using the laminin 411-based differentiation protocol for 14 days, IPCs were successfully generated as evidenced by the upregulation in the expression of pancreatic endocrine genes (Foxa-2, Sox-17, PDX-1, Ngn-3, insulin, and glucagon). Additionally, when those cells where subjected to increasing amounts of glucose, they reacted by releasing significant quantities of insulin which further proved the successful derivation of functional pancreatic β cell.

To further assess the efficiency of the generated IPCs, we designed an experimental animal model where diabetes was induced via Streptozotocin (STZ). STZ is a diabetogenic agent, which is well known for diabetes induction in animal studies [23]. STZ induces the uptake of glucose transporter GLUT2 by pancreatic ß cells which causes further blockage of glucose-induced insulin secretion while ameliorating oxidative stress of ß cells [31]. Moreover, STZ selectively destroys β cells of the islets of Langerhans which results in the hindrance of insulin synthesis resulting in an increase in blood glucose level [18].

In this experimental setting, injection of streptozotocin in rats in the current study triggered significant elevation in blood glucose, serum VF, and pancreatic glucagon levels associated with significant reduction in serum INS and CP levels. Florence et al. [20] confirmed that administration of STZ in rats caused pancreatic swelling and degeneration of ß cells and resulted in eliciting experimental diabetes mellitus in 2 to 4 days [20]. From the molecular point of view, our data confirmed that injection of STZ caused significant downregulation in the expression levels of pancreatic Foxa2, Sox17, IGF-1, and FGF 10 genes.

At the same time, the ability of the generated IPCs to home to the impaired pancreatic tissues of diabetic rats was confirmed by PKH26 dye in concomitant with previous studies [4, 58]. Through analyzing the pancreatic tissue for positive tracing of PKH26 dye, we found that in all groups transplanted with IPCs; successful homing to the diabetic pancreas was achieved.

Our data additionally documented that the systemically administered IPCs contributed to blood glucose control in the diabetic rats through eliciting significant increase in serum INS and CP levels concomitant with significant decrease in serum VF and pancreatic GC levels. Visfatin is a unique adipo-cytokine, known as pre-B cell colony-enhancing factor or nicotinamide phosphoribosyl transferase, and plays a prominent role in insulin signal transduction, glucose homeostasis, and insulin-like effects [17, 29]. It has been suggested that elevated expression of VF is compensatory for the increased blood glucose and decreased insulin levels [37, 39, 49].

Our results also demonstrated a remarkably improved blood glucose level in diabetic rats treated with the generated IPCs. This is mainly due to the successful and constant secretion of insulin by the injected IPCs that homed in the pancreas of diabetic rats. This confirmed that the generated IPCs effectively secreted insulin which in turn succeeded in reversing the hyperglycemic state caused by diabetes. Similar research efforts have indicated that transplanting products of stem cells can reduce the symptoms of diabetes [10]. In a previous study, [62] found that insulin-secreting cells that are generated from human Warton jelly-derived mesenchymal cells successfully reversed the hyperglycemic state in diabetic rats [47]. Hsiao et al. [26] also reported that both serum CP and INS levels were significantly higher in rats after IPC transplantation [26].

Additionally, the implanted IPCs in diabetic rats resulted in an upregulated expression of pancreatic Foxa2, Sox17, IGF-1, and FGF 10. Both Foxa2 and Sox17 are considered critical regulators of definitive endoderm required for pancreas development and successful differentiation of all types of stem cells; mesenchymal, embryonic, or even induced pluripotent stem cells into insulin-producing-like cells. Jonatan et al. [29] reported the unique function of Sox17 in controlling insulin release by β cells both in normal and diabetic states [29]. As for IGF-1, several studies reported its positive influence on glucose homeostasis [21, 25, 56]. IGF-I has been reported to enhance insulin sensitivity through increasing glucose uptake peripherally and reducing hepatic glucose synthesis. It has been cited that, it is responsible for glucose metabolisms, in addition to its important role in triggering insulin sensitivity and regulating glucose uptake [2, 16, 48]. Although insulin and IGF share similar modes of action regarding glucose uptake stimulation, some studies reported that IGF is superior to insulin [41]. On the other hand, Palsgaard et al. [41] concluded that insulin resistance and hyperglycemia often witnessed in diabetic patients is due to the downregulation of IGF-1 [41]. Thus, taken together, the upregulated expression level of pancreatic Foxa2, Sox17, IGF-1, and FGF 10 reported in our study confirms the pancreatic beta cell identity of the derived IPCs.

Conclusions

Overall, the outcomes of this research work validated that laminin 411-based differentiation protocol substantially improved the generation of functional IPCs from dental derived stem cells; nevertheless, the generated IPCs released essential levels of insulin. Furthermore, our in vivo study demonstrated that intravenous administration of the IPCs derived by this protocol effectively diminished blood glucose levels, with significant prolongation of the post-transplanted survival and recovery of the DM–relevant markers. Thus, it could be established that the generated IPCs by this differentiation protocol efficiently functioned like an undamaged pancreas both in vitro and in vivo. In conclusion, selecting the suitable protocol for IPC differentiation is crucial, together with taking into consideration the original source of stem cells since each cell type possesses distinct characteristics. Finally, IPCs generated from dental-derived stem cells in this attempt, through mimicking the in vivo microenvironment, proved to be an efficient and feasible approach in cell replacement therapy of diabetes.

Recommendations

Although the generation of physiologically functioning beta cells from stem cells is rapidly advancing, there are yet some issues that need to be addressed in order to translate this strategy into clinical application. Future efforts need to be directed on acquiring a pure and homogenous functional population of endocrine cells free of undifferentiated cells. This could be achieved through genetically manipulating the differentiation signals or via precise gene editing. In addition, methods for scaling up the procedures while at the same time ensuring minimum variation in the final cell product need to be carefully designed. Finally, developing xeno-free strategies and tackling immune issues in addition to optimizing transplantation procedures shall eventually render this stem cell based therapy a clinical reality.

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Abbreviations

IPCs:

Insulin-producing cells

MSCs:

Mesenchymal stem cells

INS:

Serum insulin

CP:

C-Peptide

VF:

Visfatin

GC:

Pancreatic glucagon

Foxa2:

Forkhead box protein A2

Sox17:

SRY-box transcription factor 17

IGF-1:

Insulin-like growth factor I

FGF 10:

Fibroblast growth factor10

DM:

Diabetes mellitus

ECM:

Extracellular matrix

hDPSCs:

Human dental pulp stem cells

hPDLSCs:

Human periodontal ligament stem cells

STZ:

Streptozotocin

VN:

Vitronectin-N

iPSCs:

Induced pluripotent stem cells

GLUT2:

Glucose transporter

References

  1. Abdel Moniem EM, MM EL-B, Halawa AM, Gomaa DH, Eldeen GN, Aly RM (2019) Optimizing a serum-free/xeno-free culture medium for culturing and promoting the proliferation of human dental pulp stem cells. Stem Cell Investig 6:1–10. https://doi.org/10.21037/sci.2019.06.05

    Article  Google Scholar 

  2. Adamek A, Kasprzak A (2018) Insulin-like growth factor (IGF) system in liver diseases. Int J Mol Sci 19:1308

  3. Ahmed HH, Aglan HA, Mahmoud NS, Aly RM (2021) Preconditioned human dental pulp stem cells with cerium and yttrium oxide nanoparticles effectively ameliorate diabetic hyperglycemia while combatting hypoxia. Tissue Cell 73. https://doi.org/10.1016/j.tice.2021.101661

  4. Ali M, Mehmood A, Anjum MS, Tarrar MN, Khan SN, Riazuddin S (2015) Diazoxide preconditioning of endothelial progenitor cells from streptozotocin-induced type 1 diabetic rats improves their ability to repair diabetic cardiomyopathy. Mol Cell Biochem 410. https://doi.org/10.1007/s11010-015-2560-6

  5. Aly R, Yamamoto T, Takenaka C, Murakami E, Arita M, Nakamura M, Kawamata S (2018) Effective endodermal induction via vitronectin-coated feeder-free culture. J Stem Cells 13:114–125

    Google Scholar 

  6. Aly RM (2020) Current state of stem cell-based therapies: an overview. Stem Cell Investig 7. https://doi.org/10.21037/sci-2020-001

  7. Ameri J, Borup R, Prawiro C, Ramond C, Schachter KA, Scharfmann R, Semb H (2017) Efficient generation of glucose-responsive beta cells from isolated GP2+human pancreatic progenitors. Cell Rep 19:36–49. https://doi.org/10.1016/j.celrep.2017.03.032

    Article  Google Scholar 

  8. Atari M, Barajas M, Hernández-Alfaro F, Gil C, Fabregat M, Ferrés Padró E, Giner L, Casals N (2011) Isolation of pluripotent stem cells from human third molar dental pulp. Histol Histopathol 26:1057–1070. https://doi.org/10.14670/HH-26.1057

    Article  Google Scholar 

  9. Atari M, Gil-Recio C, Fabregat M, García-Fernández D, Barajas M, Carrasco MA, Jung HS, Alfaro FH, Casals N, Prosper F, Ferrés-Padró E, Giner L (2012) Dental pulp of the third molar: a new source of pluripotent-like stem cells. J Cell Sci 125:3343–3356. https://doi.org/10.1242/JCS.096537

    Article  Google Scholar 

  10. Bai L, Meredith G, Tuch BE (2005) Glucagon-like peptide-1 enhances production of insulin in insulin-producing cells derived from mouse embryonic stem cells. J Endocrinol 186:343–352. https://doi.org/10.1677/joe.1.06078

    Article  Google Scholar 

  11. Bancroft J, Layton C (2018) Theory and practice of histological techniques - John D. Bancroft - Google Books. In: Bancroft’s theory and practice of histological techniques, pp 126–176

    Google Scholar 

  12. Black IB, Woodbury D (2001) Adult rat and human bone marrow stromal stem cells differentiate into neurons. Blood Cells Mol Dis 27:632–636. https://doi.org/10.1006/bcmd.2001.0423

    Article  Google Scholar 

  13. Braam SR, Zeinstra L, Litjens S, Ward-van Oostwaard D, van den Brink S, van Laake L, Lebrin F, Kats P, Hochstenbach R, Passier R, Sonnenberg A, Mummery CL (2008) Recombinant vitronectin is a functionally defined substrate that supports human embryonic stem cell self-renewal via αVβ5 integrin. Stem Cells 26:2257–2265. https://doi.org/10.1634/stemcells.2008-0291

    Article  Google Scholar 

  14. Brafman DA, Phung C, Kumar N, Willert K (2013) Regulation of endodermal differentiation of human embryonic stem cells through integrin-ECM interactions. Cell Death Differ 20:369–381. https://doi.org/10.1038/cdd.2012.138

    Article  Google Scholar 

  15. Choi Y, Ta M, Atouf F, Lumelsky N (2004) Adult pancreas generates multipotent stem cells and pancreatic and nonpancreatic progeny. Stem Cells 22:1070–1084. https://doi.org/10.1634/STEMCELLS.22-6-1070

    Article  Google Scholar 

  16. Clemmons DR (2004) The relative roles of growth hormone and IGF-1 in controlling insulin sensitivity. J Clin Invest 113:25. https://doi.org/10.1172/JCI200420660

    Article  Google Scholar 

  17. Dahl TB, Holm S, Aukrust P, Halvorsen B (2012) Visfatin/NAMPT: a multifaceted molecule with diverse roles in physiology and pathophysiology. Annu Rev Nutr 32:229–243. https://doi.org/10.1146/ANNUREV-NUTR-071811-150746

    Article  Google Scholar 

  18. Daisy P, Balasubramanian K, Rajalakshmi M, Eliza J, Selvaraj J (2010) Insulin mimetic impact of Catechin isolated from Cassia fistula on the glucose oxidation and molecular mechanisms of glucose uptake on Streptozotocin-induced diabetic Wistar rats. Phytomedicine 17:28–36. https://doi.org/10.1016/j.phymed.2009.10.018

    Article  Google Scholar 

  19. El-Borady OM, Othman MS, Atallah HH, Abdel Moneim AE (2020) Hypoglycemic potential of selenium nanoparticles capped with polyvinyl-pyrrolidone in streptozotocin-induced experimental diabetes in rats. Heliyon 6:e04045. https://doi.org/10.1016/J.HELIYON.2020.E04045

    Article  Google Scholar 

  20. Florence NT, Benoit MZ, Jonas K, Alexandra T, Désiré DDP, Pierre K, Théophile D (2014) Antidiabetic and antioxidant effects of Annona muricata (Annonaceae), aqueous extract on streptozotocin-induced diabetic rats. J Ethnopharmacol 151:784–790. https://doi.org/10.1016/j.jep.2013.09.021

    Article  Google Scholar 

  21. Friedrich N, Thuesen B, Jrøgensen T, Juul A, Spielhagen C, Wallaschofksi H, Linneberg A (2012) The association between IGF-I and insulin resistance: a general population study in Danish adults. Diabetes Care 35:768. https://doi.org/10.2337/DC11-1833

    Article  Google Scholar 

  22. Fu J, Chen J, Li W, Yang X, Yang J, Quan H, Huang H, Chen G (2021) Laminin-modified dental pulp extracellular matrix for dental pulp regeneration. Front Bioeng Biotechnol 8:1504. https://doi.org/10.3389/FBIOE.2020.595096/BIBTEX

    Article  Google Scholar 

  23. Graham ML, Janecek JL, Kittredge JA, Hering BJ, Schuurman HJ (2011) The streptozotocin-induced diabetic nude mouse model: differences between animals from different sources. Comp Med 61:356–360

    Google Scholar 

  24. Hirsch T, Rothoeft T, Teig N, Bauer JW, Pellegrini G, De Rosa L, Scaglione D, Reichelt J, Klausegger A, Kneisz D, Romano O, Seconetti AS, Contin R, Enzo E, Jurman I, Carulli S, Jacobsen F, Luecke T, Lehnhardt M, Fischer M, Kueckelhaus M, Quaglino D, Morgante M, Bicciato S, Bondanza S, De Luca M (2017) Regeneration of the entire human epidermis using transgenic stem cells. Nature 551:327–332. https://doi.org/10.1038/nature24487

    Article  Google Scholar 

  25. Hong H, Cui ZZ, Zhu L, Fu SP, Rossi M, Cui YH (2017) Zhu BM (2017) Central IGF1 improves glucose tolerance and insulin sensitivity in mice. Nutr Diabetes 712(7):1–10. https://doi.org/10.1038/s41387-017-0002-0

    Article  Google Scholar 

  26. Hsiao CY, Chen TH, Chen PH, Tsai PJ, Huang BS, Su CH, Shyu JF (2020) Comparison between the therapeutic effects of differentiated and undifferentiated Wharton’s jelly mesenchymal stem cells in rats with streptozotocin-induced diabetes. World J Stem Cells 12:139–151. https://doi.org/10.4252/wjsc.v12.i2.139

    Article  Google Scholar 

  27. Huang GT-J, Gronthos S, Shi S (2009) Mesenchymal stem cells derived from dental tissues vs. those from other sources: their biology and role in regenerative medicine. J Dent Res 88:792–806. https://doi.org/10.1177/0022034509340867

    Article  Google Scholar 

  28. Hussey GS, Dziki JL, Badylak SF (2018) Extracellular matrix-based materials for regenerative medicine. Nat Rev Mater 37(3):159–173. https://doi.org/10.1038/s41578-018-0023-x

    Article  Google Scholar 

  29. Jonatan D, Spence JR, Method AM, Kofron M, Sinagoga K, Haataja L, Arvan P, Deutsch GH, Wells JM (2014) Sox17 regulates insulin secretion in the normal and pathologic mouse β cell. PLoS One 9. https://doi.org/10.1371/journal.pone.0104675

  30. Jørgensen MC, Ahnfelt-Rønne J, Hald J, Madsen OD, Serup P, Hecksher-Sørensen J (2007) An illustrated review of early pancreas development in the mouse. Endocr Rev 28:685–705. https://doi.org/10.1210/er.2007-0016

    Article  Google Scholar 

  31. Koroglu P, Senturk GE, Yucel D, Ozakpinar OB, Uras F, Arbak S (2015) The effect of exogenous oxytocin on streptozotocin (STZ)-induced diabetic adult rat testes. Peptides 63:47–54. https://doi.org/10.1016/j.peptides.2014.10.012

    Article  Google Scholar 

  32. Matei IV, Ii H, Yaegaki K (2017) Hydrogen sulfide enhances pancreatic β-cell differentiation from human tooth under normal and glucotoxic conditions. Regen Med 12:125–141. https://doi.org/10.2217/rme-2016-0142

    Article  Google Scholar 

  33. Miletić M, Mojsilović S, Okić-Dordević I, Kukolj T, Jauković A, Santibañez JF, Jovčić G, Bugarski D (2014) Mesenchymal stem cells isolated from human periodontal ligament. Arch Biol Sci 66:261–271. https://doi.org/10.2298/ABS1401261M

    Article  Google Scholar 

  34. Millman JR, Xie C, Van Dervort A, Gürtler M, Pagliuca FW, Melton DA (2016) Generation of stem cell-derived β-cells from patients with type 1 diabetes. Nat Commun 7:11463. https://doi.org/10.1038/ncomms11463

    Article  Google Scholar 

  35. Murtaugh LC (2007) Pancreas and beta-cell development: from the actual to the possible. Development 134:427–438

    Article  Google Scholar 

  36. Nemati M, Ranjbar Omrani G, Ebrahimi B, Alizadeh A (2021) Efficiency of stem cell (SC) differentiation into insulin-producing cells for treating diabetes: a systematic review. Stem Cells Int 2021. https://doi.org/10.1155/2021/6652915

  37. Nicholson T, Church C, Baker DJ (2018) Jones SW (2018) The role of adipokines in skeletal muscle inflammation and insulin sensitivity. J Inflamm 151(15):1–11. https://doi.org/10.1186/S12950-018-0185-8

    Article  Google Scholar 

  38. Nie Y, Zhang S, Liu N, Li Z (2016) Extracellular matrix enhances therapeutic effects of stem cells in regenerative medicine. In: Composition and Function of the Extracellular Matrix in the Human Body. IntechOpen. p. 323–40. https://www.intechopen.com/chapters/50099

  39. Oki K, Yamane K, Kamei N, Nojima H, Kohno N (2007) Circulating visfatin level is correlated with inflammation, but not with insulin resistance. Clin Endocrinol (Oxf) 67:796–800. https://doi.org/10.1111/j.1365-2265.2007.02966.x

    Article  Google Scholar 

  40. Pagliuca FW, Melton DA (2013) How to make a functional β-cell. Development 140:2472–2483. https://doi.org/10.1242/dev.093187

    Article  Google Scholar 

  41. Palsgaard J, Brown AE, Jensen M, Borup R, Walker M, De Meyts P (2009) Insulin-like growth factor I (IGF-I) is a more potent regulator of gene expression than insulin in primary human myoblasts and myotubes. Growth Horm IGF Res 19:168–178. https://doi.org/10.1016/J.GHIR.2008.09.004

    Article  Google Scholar 

  42. Patil R, Kumar BM, Lee WJ, Jeon RH, Jang SJ, Lee YM, Park BW, Byun JH, Ahn CS, Kim JW, Rho GJ (2014) Multilineage potential and proteomic profiling of human dental stem cells derived from a single donor. Exp Cell Res 320:92–107. https://doi.org/10.1016/j.yexcr.2013.10.005

    Article  Google Scholar 

  43. Qu H, Liu X, Ni Y, Jiang Y, Feng X, Xiao J, Guo Y, Kong D, Li A, Li X, Zhuang X, Wang Z, Wang Y, Chang Y, Chen S, Kong F, Zhang X, Zhao S, Sun Y, Xu D, Wang D, Zheng C (2014) Laminin 411 acts as a potent inducer of umbilical cord mesenchymal stem cell differentiation into insulin-producing cells. J Transl Med 12:135. https://doi.org/10.1186/1479-5876-12-135

    Article  Google Scholar 

  44. Ren J, Jin P, Wang E, Liu E, Harlan DM, Li X, Stroncek DF (2007) Pancreatic islet cell therapy for type I diabetes: understanding the effects of glucose stimulation on islets in order to produce better islets for transplantation. J Transl Med 5:1–15. https://doi.org/10.1186/1479-5876-5-1/FIGURES/5

    Article  Google Scholar 

  45. Saez DM, Sasaki RT, Da Costa NA, Da Silva MCP (2016) Stem cells from human exfoliated deciduous teeth: a growing literature. Cells Tissues Organs 202:269–280

    Article  Google Scholar 

  46. Seo BM, Miura M, Gronthos S, Bartold PM, Batouli S, Brahim J, Young M, Robey PG, Wang CY, Shi S (2004) Investigation of multipotent postnatal stem cells from human periodontal ligament. Lancet 364:149–155. https://doi.org/10.1016/S0140-6736(04)16627-0

    Article  Google Scholar 

  47. Seo MJ, Suh SY, Bae YC, Jung JS (2005) Differentiation of human adipose stromal cells into hepatic lineage in vitro and in vivo. Biochem Biophys Res Commun 328:258–264. https://doi.org/10.1016/j.bbrc.2004.12.158

    Article  Google Scholar 

  48. Shin M, Kang HS, Park JH, Bae JH, Song DK, Im SS (2017) Recent insights into insulin-like growth factor binding protein 2 transcriptional regulation. Endocrinol Metab 32:11–17. https://doi.org/10.3803/ENM.2017.32.1.11

    Article  Google Scholar 

  49. Stofkova A (2010) Resistin and visfatin: regulators of insulin sensitivity, inflammation and immunity. Endocr Regul 44:25–36. https://doi.org/10.4149/ENDO_2010_01_25

    Article  Google Scholar 

  50. Stowers RS (2021) Advances in extracellular matrix-mimetic hydrogels to guide stem cell fate. Cells Tissues Organs:1–18. https://doi.org/10.1159/000514851

  51. Sundberg M, Hyysalo A, Skottman H, Shin S, Vemuri M, Suuronen R, Narkilahti S (2011) A xeno-free culturing protocol for pluripotent stem cell-derived oligodendrocyte precursor cell production. Regen Med 6:1–12. https://doi.org/10.2217/RME.11.36

    Article  Google Scholar 

  52. Tariq M, Masoud MS, Mehmood A, Khan SN, Riazuddin S (2013) Stromal cell derived factor-1alpha protects stem cell derived insulin-producing cells from glucotoxicity under high glucose conditions in-vitro and ameliorates drug induced diabetes in rats. J Transl Med 11:115. https://doi.org/10.1186/1479-5876-11-115

    Article  Google Scholar 

  53. Tate CC, Shear DA, Tate MC, Archer DR, Stein DG, LaPlaca MC (2009) Laminin and fibronectin scaffolds enhance neural stem cell transplantation into the injured brain. J Tissue Eng Regen Med 3:208–217. https://doi.org/10.1002/term.154

    Article  Google Scholar 

  54. Trucco M (2005) Regeneration of the pancreatic β cell. J Clin Invest 115:5–12. https://doi.org/10.1172/JCI200523935

    Article  Google Scholar 

  55. Vegas AJ, Veiseh O, Gürtler M, Millman JR, Pagliuca FW, Bader AR, Doloff JC, Li J, Chen M, Olejnik K, Tam HH, Jhunjhunwala S, Langan E, Aresta-Dasilva S, Gandham S, McGarrigle JJ, Bochenek MA, Hollister-Lock J, Oberholzer J, Greiner DL, Weir GC, Melton DA, Langer R, Anderson DG (2016) Long-term glycemic control using polymer-encapsulated human stem cell-derived beta cells in immune-competent mice. Nat Med 22:306–311. https://doi.org/10.1038/nm.4030

    Article  Google Scholar 

  56. White MF (2014) IRS2 integrates insulin/IGF1 signalling with metabolism, neurodegeneration and longevity. Diabetes Obes Metab 16:4–15. https://doi.org/10.1111/dom.12347

    Article  Google Scholar 

  57. Wong JCY, Gao SY, Lees JG, Best MB, Wang R, Tuch BE (2010) Definitive endoderm derived from human embryonic stem cells highly express the integrin receptors alphaV and beta5. Cell Adh Migr 4:39–45

    Article  Google Scholar 

  58. Xin Y, Jiang X, Wang Y, Su X, Sun M, Zhang L, Tan Y, Wintergerst KA, Li Y, Li Y (2016) Insulin-producing cells differentiated from human bone marrow mesenchymal stem cells in vitro ameliorate streptozotocin-induced diabetic hyperglycemia. PLoS One 11. https://doi.org/10.1371/journal.pone.0145838

  59. Yamaza T, Kentaro A, Chen C, Liu Y, Shi Y, Gronthos S, Wang S, Shi S (2010) Immunomodulatory properties of stem cells from human exfoliated deciduous teeth. Stem Cell Res Ther 1:5. https://doi.org/10.1186/scrt5

    Article  Google Scholar 

  60. Yap L, Tay HG, Nguyen MTX, Tjin MS, Tryggvason K (2019) Laminins in cellular differentiation. Trends Cell Biol 29:987–1000. https://doi.org/10.1016/J.TCB.2019.10.001

    Article  Google Scholar 

  61. Zamir EA, Rongish BJ, Little CD (2008) The ECM moves during primitive streak formation—computation of ECM versus cellular motion. PLoS Biol 6:e247. https://doi.org/10.1371/journal.pbio.0060247

    Article  Google Scholar 

  62. Yi Kao S (2015) Transplantation of Hepatocytelike Cells Derived from Umbilical Cord Stromal Mesenchymal Stem Cells to Treat Acute Liver Failure Rat. J Biomed Sci 4:1. https://doi.org/10.4172/2254-609x.100002

Download references

Acknowledgements

Not applicable.

Funding

The authors acknowledge the financial support of the Science, Technology and Innovation Funding Authority, (STIFA) formally known as Science and Technology Development Fund (STDF) Egypt, (Special targeted call (Health) Grant number: 22943).

Author information

Authors and Affiliations

Authors

Contributions

(I) Conception and design: H.H. and E.A. (II) Administrative support: H.H. (III) Provision of study materials or patients: H.A and R.A. (IV) Collection and assembly of data: H.A and R.A. (V) In vitro molecular assessment studies: G.N. (VI) In vivo molecular assessment studies: N.M. (VII) Data analysis and interpretation: H.H, H.A, and R.A. (VIII) Manuscript writing: All authors. (IX) Final approval of manuscript: The authors read and approved the final manuscript.

Corresponding author

Correspondence to Riham M. Aly.

Ethics declarations

Ethics approval and consent to participate

The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved. Housing and management of animals and experimental protocol followed the guidelines of the National Institutes of Health guide for the care and use of Laboratory animals (NIH Publications No. 8023, revised 1978) and was approved by the Ethical Committee of the Medical Research of the National Research Centre, Egypt (approval no. 19 109).

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

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/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Aly, R.M., Aglan, H.A., Eldeen, G.N. et al. Efficient generation of functional pancreatic β cells from dental-derived stem cells via laminin-induced differentiation. J Genet Eng Biotechnol 20, 85 (2022). https://doi.org/10.1186/s43141-022-00369-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s43141-022-00369-6

Keywords