We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
ReportsOpen Accesscc iconby iconnc iconnd icon

Direct-current electric field stimulation promotes proliferation and maintains stemness of mesenchymal stem cells

    Mengchang Liu‡

    Southwest Medical University, No. 1 Section 1, Xianglin Road, Luzhou City, Sichuan Province, 646000, China

    ‡Authors contributed equally

    Search for more papers by this author

    ,
    Defu Xie‡

    Southwest Medical University, No. 1 Section 1, Xianglin Road, Luzhou City, Sichuan Province, 646000, China

    ‡Authors contributed equally

    Search for more papers by this author

    ,
    Huizhen Zeng

    Southwest Medical University, No. 1 Section 1, Xianglin Road, Luzhou City, Sichuan Province, 646000, China

    ,
    Ning Zhai

    Southwest Medical University, No. 1 Section 1, Xianglin Road, Luzhou City, Sichuan Province, 646000, China

    ,
    Lan Liu

    Department of Plastic & Burn Surgery, Affiliated Hospital of Southwest Medical University, National Key Clinical Construction Specialty, Wound Repair & Regeneration Laboratory, No. 25 Taiping Street, Jiangyang District, Luzhou, Sichuan Province, 646000, China

    &
    Hong Yan

    *Author for correspondence: Tel.: +86 0830 3165405;

    E-mail Address: xnshaoshang123@163.com

    Department of Plastic & Burn Surgery, Affiliated Hospital of Southwest Medical University, National Key Clinical Construction Specialty, Wound Repair & Regeneration Laboratory, No. 25 Taiping Street, Jiangyang District, Luzhou, Sichuan Province, 646000, China

    Published Online:https://doi.org/10.2144/btn-2022-0112

    Abstract

    Mesenchymal stem cells are frequently utilized in the study of regenerative medicine. Electric fields (EFs) influence many biological processes, such as cell proliferation, migration and differentiation. In the present study, a novel device capable of delivering a direct current of EF stimulation to cells cultured in vitro is described. This bioreactor was customized to simultaneously apply a direct-current EF to six individual cell culture wells, which reduces the amount of experimental time and minimizes cost. In testing the device, adipose-derived mesenchymal stem cells stimulated with an EF in the bioreactor exhibited a greater cell proliferation rate while retaining stemness. The results provide a unique perspective on adipose-derived mesenchymal stem cell proliferation, which is needed for tissue engineering and regenerative medicine.

    METHOD SUMMARY

    A customized bioreactor was built to deliver electric field (EF) stimulation. Adipose-derived mesenchymal stem cells were exposed to a direct-current EF at 0 or 200 mV/mm for 1 h/d over the course of 3 days. A previously described in vitro electrical stimulation method was enhanced and optimized by exposing adipose-derived mesenchymal stem cells to EF signals to induce cell proliferation.

    Mesenchymal stem cells (MSCs) have gained more attention in regenerative medicine than embryonic stem cells and induced pluripotent stem cells, which are constrained by ethical concerns and potential tumorigenicity [1]. MSCs provide a plentiful source of adult stem cells with the capacity for self-renewal and multidirectional differentiation [2]. Because of their abundance [3], simplicity of separation [4,5], pluripotency [6], immunosuppressive effects [7], antifibrotic properties [8] and angiogenic potential [9,10], MSCs are the perfect cell type for regenerative medicine. The isolation of adipose-derived MSCs (Ad-MSCs) can be achieved by the enzymatic digestion of adipose tissue that has been removed by liposuction or lipectomy [4]. Harvesting MSCs from adipose tissue can provide a larger proportion of MSCs compared with other stem cell sources [5] and donor site problems are minimal. Furthermore, Ad-MSCs can be safely transplanted into autologous or allogeneic bodies, with few graft allografts and foreign body reactions [11,12]. Also, there are no ethical concerns because they can be derived from autologous fat. Based on these benefits, Ad-MSCs provide a more effective and practical treatment option for tissue and organ transplantation.

    Although current approaches to obtaining Ad-MSCs are reasonably effective, their therapeutic use is limited because of their limited available quantity [13] and the fact that cell therapy regimens normally require hundreds of millions of MSCs for each treatment. It requires approximately ten weeks to obtain enough cells by in vitro expansion before implantation. In addition, studies have shown that the ideal culture conditions to produce MSCs at a clinical scale vary with the age and characteristics of the patient and cellular senescence of MSCs in vitro, potentially leading to aging and age-related diseases [14,15]. Prolonged culture expansion of human MSCs (hMSCs) in vitro leads to a loss of differentiation potential [16]. In addition, the clinical outcomes of hMSCs transplantation remain variable [15], and the short survival rate of transplanted hMSCs and the regenerative potential of Ad-MSCs following transplantation are reduced [17,18]. The therapeutic efficacy of individual-derived MSCs must be enhanced due to their long expansion period, individual differences, low survival rate and limited regenerative capacity following transplantation, which is a challenge to their large-scale clinical use.

    Electric fields (EFs) are required for cells to maintain dynamic homeostasis and to engage in a variety of biological phenomena ranging from embryonic development to tissue regeneration [19]. Biological processes, such as long-distance protein interactions, electron transfers during chemical reactions [20], embryogenesis [21], cell differentiation [22], growth [23], wound healing, tissue repair and remodeling [24,25], are influenced by endogenous EF, which exist in the cytoplasm and extracellular space [26,27]. The duration, intensity and orientation of these fields can also affect cell proliferation. Khaw et al. [28] devised a capacitive electrical stimulation device to apply 100 and 200 mV/mm uniform EFs to MSCs daily for 14 days, which increased the cell proliferation rate by fivefold. Martin et al. [29] found that osteoprogenitor cells stimulated with 125–500 mV/mm direct current (DC) for 3 hours every day for 3 days exhibited a higher propensity to proliferate and differentiate. Panda et al. [30] stimulated hMSCs with 100 mv/mm DC for 10 min each day for 21 days and discovered that their proliferation rate decreased, but osteogenic differentiation capacity was enhanced. Notably, EF can have either a stimulatory or an inhibitory effect, depending on the exposure conditions and cell type [31,32]. Nonetheless, there is evidence that EF signals play an important role during the cellular regeneration process [23]. Clinically, EF has been used to treat several diseases, including osteoporosis, chronic skin ulcers, cardiac arrhythmias and traumatic injury [33–36].

    Based on these findings, EF stimulation can be used to stimulate cell regeneration. Because it does not require the addition of exogenous immunogenic biological substances or complex equipment, EF signaling stimulation offers many advantages [37,38]. Currently, in vitro electrical stimulation approaches, such as DCEF, are being used [39]. DCEF not only affects cell survival and proliferation, but also influences cell migration and morphology [40,41].

    Materials & methods

    Cell isolation, culture & passaging

    Ad-MSCs were harvested from adipose tissue following liposuction surgery. The principles outlined in the Declaration of Helsinki were followed. Human adipose tissues obtained from liposuction were soaked in ice-cold phosphate-buffered saline (PBS; Biological Industries, CT, USA), washed three times with PBS, shredded into a crumbly form and digested with 10 mg collagenase I (Gibco™, NY, USA) in DMEM/F12 (Gibco, NY, USA) (1:1) medium containing 2% double antibiotic (Gibco, NY, USA) and filter sterilized. Digestion was performed while shaking manually at 37°C for 1 hour. Tissue clumps were removed by filtration through a 70-μm filter membrane. The membrane was washed with PBS and the single nucleated cells were collected by centrifugation at 500 g for 5 min to yield Ad-MSCs. The cells (2000 cells/cm2) were seeded in DMEM (Gibco, NY, USA) in culture flasks containing 10% fetal bovine serum (FBS; Gibco, NY, USA). Cells were then suspended in 10 ml of growth medium (DMEM/F12, 10% CS, 2% double antibodies) and incubated in 10-cm dishes. After 12–24 hours, the medium was removed and the cells were washed with PBS to remove unadhered cells, tissue clumps and debris. The medium was changed every 3 days until 90% confluence of the cells. The Ad-MSCs were incubated with 0.25% trypsin-EDTA (Biological Industries, CT, USA) then passaged or seeded into 6-well plates at a density of 1 × 105/well and cultured with growth medium. Ad-MSCs from passages 3–5 were used for this research.

    Authentication of Ad-MSCs

    After DCEF stimulation, the MSCs phenotype was characterized by flow cytometry. Next, 1 × 105 Ad-MSCs were incubated with monoclonal antibodies CD90, CD105, CD45, CD73, CD14, CD31 and CD34 (Becton Dickinson, NJ, USA) for 40 min at 4°C protected from light. Flow cytometry (LSRF Ortessa, BD Biosciences, USA) was used to record and analyze the data. At least 1 × 104 cells per run were analyzed.

    Differentiation of MSCs

    For MSCs with or without EF treatment, adipogenic differentiation was induced in DMEM/F12 medium supplemented with 10% FBS, 1% penicillin-streptomycin, 500 μM 3-isobutyl-1-methylxanthine, 1 μM dexamethasone, 10 μM insulin and 200 μM indomethacin (Sigma, LA, USA). After 2 weeks, two groups of MSCs were fixed in 4% paraformaldehyde and stained with Oil Red O (Sigma, LA, USA) to observe lipid droplets. Cultured MSC cells in DMEM-HG supplemented with 10% FBS, 1% penicillin-streptomycin, 10 nM dexamethasone, 50 μM ascorbic acid 2-phosphate (Sigma, LA, USA) and 10 nM β-glycerophosphate (Sigma, LA, USA) were used to induce osteogenic differentiation. After 2 weeks, the MSCs were fixed in 4% paraformaldehyde and stained with Alizarin red (Sigma, LA, USA) to examine mineralized matrix apposition.

    DC electrical stimulation chamber

    The in vitro electrical stimulation method proposed by Mobini et al. [37,41] was enhanced and optimized by exposing Ad-MSCs to EF signals to induce cell proliferation. This in vitro electrical stimulation tool works with changes in the electrodes and the presence of an electric current generates electrothermal effects that evaporate the medium, which may have additional electrothermal effects on the cells. Therefore, the device was improved by using a DC and an electrode plate to generate a uniform EF to eliminate the current throughout the experiment. The device is suitable for various biological and biomedical studies requiring bioelectric field stimulation.

    The device consisted of gold-plated electrodes that are attached to the top cover of the cell culture chamber and fit standard 6-well cell culture plates (128 mm × 85 mm × 22 mm; inner diameter: 33.78 mm; growth area: 8.96 cm2) for easy manipulation and sterilization and to reduce evaporation of the medium. Each electrode plate group consisted of 13 gold-plated electrodes of 1-mm diameter running in parallel. To prevent harmful material released during electrical stimulation from interfering with the experimental results, each set of electrode plates was sealed with polymethylmethacrylate. The electrode points protruding from the lid were soldered to parallel circuits of silver-plated copper wires and connected to a standard power supply with a banana connection. By adjusting the power supply, the amount of electricity and its application pattern could be modified. Before the experiment, the device was sterilized in 75% ethanol for 2 h in a calcium-free solution, washed in DPBS (Gibco, USA), then exposed to UV radiation overnight.

    To demonstrate the effectiveness of the EF signal delivery device, the EF was applied to Ad-MSCs for 3 days and its effects on cell proliferation and stemness were assessed. All tests were performed in triplicate. Briefly, human Ad-MSCs were cultured at a density of 2 × 104 cells/cm2 to 80% confluence and then expanded for three generations. The fourth-generation cells were seeded into 6-well cell culture plates at a density of 5 × 103 cells/cm2 and incubated at 37°C in a 5% CO2/3% oxygen atmosphere. After inoculation, the cells were cultured in growth medium. After 1 day, the culture plate cover was changed to the EF device cover (with electrodes). A 4.4 V DC voltage was applied to generate a uniform DCEF of 200 mV/mm in the cell culture medium of the stimulation group. Cells were exposed to the DCEF for 1 hour/day for 3 days and subsequently compared with the control group. After completing the experiment, the equipment was cleaned and sterilized in preparation for usage the next day. The cells were washed twice with PBS 24 hours before the end of the experiment. Prior to the next EF stimulation, the medium was changed and the cells were photographed under a microscope.

    Protocol for electrical stimulation

    The magnitude of the EF strength in the cell surrounding is dependent upon the amount of DC voltage. According to the uniform EF formula: U = V/d (U: EF magnitude; V: DC voltage; d: electrode plate distance), the current electrical stimulation device may be considered a uniform EF. The current device stimulation mode (200 mV/mm, 1 hour) was selected as an efficient strategy for cell proliferation.

    Cell counting

    Cell counting was performed using an automated cell counter (RWD, Shenzhen, China). MSCs were digested with 0.25% trypsin from each well every day. The cells were resuspended in DMEM and the cell number for each group was counted.

    Cell cycle analysis

    Cell cycle analysis was performed by cell cycle and apoptosis detection kits (Beyotime Institute of Biotechnology, Shanghai, China). MSCs were digested with 0.25% trypsin and washed twice with PBS after treatment. The cells were resuspended in 75% cold ethanol and fixed overnight at 4°C with 75% ethanol. The following day, the cells were rinsed and disseminated with cold PBS to avoid aggregation. Propidium iodide (PI) staining solution was added to the cell pellet (0.5 ml/tube). After mixing, the cells were incubated at 37°C for 30 min in the dark. Subsequently, flow cytometry (Agilent Technologies, CA, USA) was used to examine the cell cycle pattern.

    Cell proliferation analysis

    Cell proliferation was determined using a cell counting kit (CCK-8; Beyotime, Shanghai, China) and Taipan Blue staining. Following treatment with or without EF, the cells were collected, seeded at a density of 2000 cells/well in 96-well plates and incubated in 5% CO2 at 37°C overnight. CCK-8 solution (10 μl/well) was added to each well and incubated in 5% CO2 at 37°C for 3 h. Finally, the optical density was measured at 450 nm using a microplate reader (Victor 3; Perkin Elmer, Inc).

    Quantitative real-time PCR

    After EF stimulation, the cells were harvested by TRIzol (Invitrogen, NY, USA) and disrupted in QIAshredder columns (Invitrogen, NY, USA). RNA was extracted using the RNeasy kit (Invitrogen, NY, USA) following the manufacturer's instructions. To produce cDNA, a first-strand cDNA synthesis kit (Invitrogen, NY, USA) was utilized. To validate the PCR assay, real-time PCR was performed with cDNA and repeated three times for each gene on SYBR Green (Roche, Basler, Switzerland). Primers for Bcl-2, BAX, cyclin D1, C-myc and β-actin (housekeeping gene) were provided by QuantiTectPrimer Assay (Qiagen, Duesseldorf,GER). The Step-One system (Applied Biosystems, NY, USA) was applied for real-time PCR.

    western blot analysis

    Protein expression of the Sox-2, Oct-4 and Nanog transcription factors was analyzed after EF stimulation. The cells were resuspended in cell lysis buffer (Thermo Scientific, USA) and sonicated. A BCA protein quantification kit (Thermo Scientific, USA) quantified protein in the supernatants after centrifugation. Proteins (90 μg) from each group were mixed with Laemmli sample buffer and boiled for 10 min at 100°C. SDS-PAGE and polyvinylidene difluoride membrane blotting separated proteins; western blot was performed using anti-Oct-4 (Abcam, UK), anti-Sox-2 (Abcam, UK), anti-Nanog (Abcam, UK) and anti-β-actin (Abcam, UK) antibodies. Primary antibodies were incubated overnight at 4°C on the membranes. The membranes were then incubated with horseradish peroxidase-conjugated secondary antibodies for 1 h after vigorous washing. The blots were developed by an enhanced chemiluminescence detection system (Siemens, Germany).

    Statistical analysis

    All experiments were repeated three times. A one-way analysis of variance was performed followed by Tukey's post-hoc test using Stats Direct software; p < 0.05 and p < 0.01 were considered significant and highly significant, respectively.

    Results & discussion

    Ad-MSCs maintain normal morphology after stimulation with DCEF

    The Ad-MSCs obtained from adipose tissue were adherent fibroblast-like cells that grew as elongated, adherent fibroblast-like cells even after three days of EF stimulation (Figure 1A). For cell surface antigen expression, the cells were assessed for MSC markers before and after EF treatment. Greater than 90% of the adipose MSCs were positive for CD73, CD90 and CD105 and negative for CD34, CD31 and CD45 in waveforms before EF stimulation and 3 days after treatment, which was similar to untreated control Ad-MSCs (Figure 1B).

    Figure 1. Phenotypic characterization of Ad-MSCs with and without EF stimulation.

    (A) Cell morphology with and without EF stimulation. EF treatment had a significant proliferative effect on cells after 3 days of stimulation; scale bar: 100 μm. (B) Expression level of Ad-MSC surface antigens determined by flow cytometry. Ad-MSCs were harvested from each group and analyzed for CD14, CD31, CD34, CD45, CD73, CD90 and CD105 expression. Greater than 90% of MSCs were positive for CD73, CD90 and CD105 and negative for CD14, CD31, CD34 and CD45. Treated MSCs exhibited same morphology as untreated MSCs and therefore, there was no effect of EF stimulation on MSC marker expression.

    Ad-MSC: Adipose-derived mesenchymal stem cell; EF: Electric fields; MSC: Mesenchymal stem cell.

    Ad-MSCs maintain ability to differentiate after EF stimulation & exhibit higher expression of stemness markers

    In vitro differentiation assays demonstrated the differentiation of Ad-MSCs into osteoblasts and adipocytes before and after electrical stimulation (Figure 2A). Western blot analysis revealed higher expression of Nanog, Oct-4 and Sox2 in Ad-MSCs with EF treatment compared with the untreated Ad-MSCs (Figure 2B).

    Figure 2. Differentiation of MSCs and expression of stemness markers.

    (A) Microscopic images of Ad-MSCs with and without EF stimulation (control) cultured in adipogenic and osteogenic induction media for 14 days. Cells were stained with Oil Red O for detection of adipogenesis and Alizarin red for detection of osteogenesis; scale bars: 200 um. (B) western blot analysis of Nanog, Oct-4 and Sox2. EF-treated Ad-MSCs exhibited higher expression of stemness markers compared with untreated Ad-MSCs. Maintenance of pluripotency in MSCs was attributed to several important transcription factors, including Oct-4, Sox-2 and Nanog.

    Ad-MSC: Adipose-derived mesenchymal stem cell; EF: Eelectric field; MSC: Mesenchymal stem cell.

    Compared with unstimulated Ad-MSCs, proliferation of Ad-MSCs significantly increased following electrical stimulation

    EF is expected to promote the proliferation of MSCs. Therefore, cell proliferation was measured on days 1, 2 and 3 after EF stimulation and the proliferation of Ad-MSCs after EF stimulation was significantly higher compared with that of unstimulated Ad-MSCs (p < 0.05; Figure 3A). On day 2, the proliferation of EF-stimulated Ad-MSCs was significantly higher compared with that of other EF-stimulated and unstimulated Ad-MSCs (p < 0.05).

    Figure 3. Proliferation phenotype of adipose-derived mesenchymal stem cells with and without EF stimulation.

    (A) Assessment of proliferation in adipose-derived mesenchymal stem cells with or without EF stimulation as measured by cell counting and CCK8 assay. (B) Cell cycle distribution of adipose-derived mesenchymal stem cells with and without EF stimulation. Cells were collected, adjusted to 1 × 106 cells/ml and fixed in 70% (v/v) ethanol. Propidium iodide was used to stain nucleus and red fluorescence at 488 nm was recorded. (C) Quantitative real-time PCR analysis of proliferation genes after 3 days of stimulation. EF stimulation significantly increased expression of Bcl-2, Bax, cyclin D1 and c-myc on day 3. Error bars represent the standard deviation of samples. *p < 0.05, **p < 0.01.

    EF: Electric field.

    Cell cycle analysis indicates more cells in S phase after EF compared with unstimulated cells

    To determine the effect of EF on the cell cycle, a PI-based cell cycle analysis was performed before EF and on days 1 and 2 following EF (Figure 3B). After EF stimulation, the numbers of cells in the S and G2 phases, but not G1, were markedly higher in Ad-MSCs on days 1 and 2 compared with unstimulated Ad-MSCs (p < 0.05). The fraction of cells in the S phase, which represents the DNA synthesis phase, was increased. Compared with unstimulated Ad-MSCs, there were more cells in the S and G2 phases and significantly fewer in G1.

    Quantitative PCR analysis reveals increased gene expression in EF-treated cells compared with unstimulated cells

    Changes in the expression of genes associated with cell proliferation were measured in EF-treated Ad-MSCs. Ad-MSCs in the EF-stimulated and control groups were collected and real-time PCR was used to measure the expression of Bcl-2, Bax, Cyclin D1 and C-myc (Figure 3C). The results indicated that Bcl-2, cyclin D1 and c-myc were upregulated, whereas Bax was downregulated, in the EF-stimulated group compared with the control group (p < 0.05).

    To our knowledge, this is the first study using a modified EF device to analyze the effect of a DCEF on Ad-MSCs in vitro. Ad-MSCs treated with a DCEF at an intensity of 200 mV/mm for 1 h/d for a total of 3 days had significantly higher proliferation rates compared with untreated cells; however, their morphology did not change considerably after EF stimulation (Figure 1A), whereas differentiation capacity remained intact. We used this device to expose Ad-MSCs to EF signals to induce cell proliferation. The in vitro electrical stimulation device was prepared without exterior changes near the electrode plate and without electrothermal effects that could evaporate the medium in the culture dish from the absence of current, thus preventing additional electrothermal effects that may affect the cells. A uniform EF was generated by means of a DC and electrode plates, thus eliminating the effect of the electric current on the whole experimental apparatus. This device is suitable for various biological and biomedical studies exploring bioelectric field stimulation effects and their applications.

    Ad-MSCs were selected to evaluate the impact of DC fields as they are one of the primary cell types engaged in wound healing. We examined Ad-MSCs, which were positive for CD73, CD90 and CD105, but negative for CD34, CD31, CD14 and CD45 expression (Figure 1B). We also examined the differentiation ability of the cells before and after EF treatment and discovered that they all developed into a distinct MSC lineage (Figure 2A). Pluripotency markers, such as Oct-4, Sox-2 and Nanog, are crucial for renewal and differentiation capabilities. western blot analysis revealed higher expression of Nanog, Oct-4 and Sox2 in EF-treated Ad-MSCs compared with the untreated cells (Figure 2B). In addition, the proliferation of Ad-MSCs after EF stimulation was significantly higher compared with that of unstimulated Ad-MSCs (p < 0.05; Figure 3A). After 3 days of EF treatment, more cells entered the G2 and S phases of the cell cycle, whereas fewer cells entered the G0/G1 phase. EF-stimulated cells exhibited a higher proliferation rate than unstimulated Ad-MSCs (Figure 3B) Finally, quantitative PCR revealed that Bcl-2, cyclin D1 and C-myc were upregulated, whereas Bax was downregulated, in the EF-stimulated group compared with the control group (p < 0.05; Figure 3C).

    Several EF stimulation treatments have been clinically utilized to treat skin wounds [42–44]. Many studies have claimed effectiveness and the results of these diverse experiments have been thoroughly reviewed elsewhere. However, many studies [45] in this area are replete with poor and uncontrolled experiments where different types of metal electrodes were directly inserted into the wound bed or cell culture medium. Secondary electrochemical effects of O2 or H+ released by metal ions or electrodes can affect the tissue pH within the wound, which are not separate from the primary effects of EF [46]. Unfortunately, our understanding of the cell biology of EF-induced wound healing remains limited. Thus, a more complete understanding of the effects of EF treatment is needed to identify the ideal stimulation conditions for applying a DCEF to heal skin wounds.

    A large number of in vitro studies are centered on the effects of EF stimulation on cells [47,48]. In the present study, the EF supplied by the developed device significantly increased the proliferation rate of MSCs compared with unstimulated cells [49]. Furthermore, the greater working surface of this design not only provides a larger sample volume but also enables studies that incorporate 3D cultures. This device may be a useful tool for examining the effects of EF on diverse cell types in vitro. Finally, exposing cells to EF using this device is simple, repeatable, adjustable, affordable and may accommodate various experimental methods.

    Conclusion

    Ad-MSCs treated with a DCEF at 0 or 200 mV/mm for 1 h/d for 3 days using this customized bioreactor exhibited a markedly higher proliferation rate and retained their stemness phenotype.

    Future perspective

    The expanded working surface in this design not only allows for a larger sample volume but also enables future studies involving 3D cultures. This device may be beneficial for exploring the effects of EF on various cell types in vitro. Finally, exposing cells to EF with this device is straightforward, repeatable, tunable, inexpensive and may be utilized for a variety of experiments. This study revealed a unique perspective on Ad-MSC proliferation, which is imperative for tissue engineering and regenerative medicine. Because this device does not require the addition of exogenous immunogenic biological substances or complex equipment, EF stimulation can be used to stimulate cell regeneration.

    Executive summary
    • To enhance adipose-derived mesenchymal stem cell (Ad-MSC) proliferation and stemness, a novel device capable of delivering direct-current electric field (DCEF) stimulation was applied to cells cultured in vitro.

    Experimental

    • DCEF stimulation was applied via a customized electrical stimulation chamber.

    • After DCEF stimulation, the Ad-MSCs phenotype was characterized and the differentiation ability was validated. The proliferation effect with or without DCEF was demonstrated.

    Results & discussion

    • Ad-MSCs stimulated with DCEF in this bioreactor exhibited a greater cell proliferation rate while retaining a normal phenotype and stemness.

    Conclusion

    • The results provide a unique perspective on Ad-MSC proliferation, which is needed for tissue engineering and regenerative medicine.

    Author contributions

    H Yan conceptualized and designed the study. M Liu and D Xie performed the experimental work, analyzed the associated data, wrote the draft of the manuscript and revised it critically for important intellectual content. M Liu and H Zeng contributed to the analysis and interpretation of data. N Zhai and D Xie contributed to the final approval of the version to be published. M Liu agreed to be 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. All authors approved the final manuscript.

    Acknowledgments

    The authors are grateful to the Affiliated Hospital of Southwest Medical University for supporting this research.

    Financial & competing interests disclosure

    This work was supported by the National Natural Science Foundation of China (grant no. 81571876) and the Joint Project of Southwest Medical University and Luzhou City (project no. 2019LZXNYDZ08). The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

    No writing assistance was utilized in the production of this manuscript.

    Open access

    This work is licensed under the Attribution-NonCommercial-NoDerivatives 4.0 Unported License. To view a copy of this license, visit http://creativecommons.org/licenses/by-nc-nd/4.0/

    Papers of special note have been highlighted as: • of interest; •• of considerable interest

    References

    • 1. Li P, Guo X. A review: therapeutic potential of adipose-derived stem cells in cutaneous wound healing and regeneration. Stem Cell Res. Ther. 9(1), 302 (2018).
    • 2. Yan W, Diao S, Fan Z. The role and mechanism of mitochondrial functions and energy metabolism in the function regulation of the mesenchymal stem cells. Stem Cell Res. Ther. 12(1), 140 (2021).
    • 3. Gentile P, Sterodimas A, Calabrese C, Garcovich S. Systematic review: advances of fat tissue engineering as bioactive scaffold, bioactive material, and source for adipose-derived mesenchymal stem cells in wound and scar treatment. Stem Cell Res. Ther. 12(1), 318 (2021).
    • 4. Ferroni L, De Francesco F, Pinton P, Gardin C, Zavan B. Methods to isolate adipose tissue-derived stem cells. Methods Cell Biol. 171, 215–228 (2022).
    • 5. Lisini D, Nava S, Pogliani S et al. Adipose tissue-derived mesenchymal stromal cells for clinical application: an efficient isolation approach. Curr. Res. Transl. Med. 67(1), 20–27 (2019).
    • 6. Vyas B, Shah A, Marathe A, Ansarullah, Vyas R, Bhonde R. Adipose tissue: a natural resource for multipotent mesenchymal stem cells with potential translation to trigerminal layers. Indian J. Plast. Surg. 51(2), 177–181 (2018).
    • 7. Fiori A, Uhlig S, Klüter H, Bieback K. Human adipose tissue-derived mesenchymal stromal cells inhibit CD4+ T cell proliferation and induce regulatory T cells as well as CD127 expression on CD4+CD25+ T cells. Cells 10(1), 58 (2021).
    • 8. Farge D, Loisel S, Lansiaux P, Tarte K. Mesenchymal stromal cells for systemic sclerosis treatment. Autoimmun. Rev. 20(3), 102755 (2021).
    • 9. Gentile P, Garcovich S. Systematic review: adipose-derived mesenchymal stem cells, platelet-rich plasma and biomaterials as new regenerative strategies in chronic skin wounds and soft tissue defects. Int. J. Mol. Sci. 22(4), (2021).
    • 10. Shin RL, Lee CW, Shen OY, Xu H, Lee OK. The crosstalk between mesenchymal stem cells and macrophages in bone regeneration: a systematic review. Stem Cells Int. 2021, 8835156 (2021).
    • 11. Sanabria-De La Torre R, Quiñones-Vico MI, Fernández-González A et al. Alloreactive immune response associated to human mesenchymal stromal cells treatment: a systematic review. J. Clin. Med. 10(13), 2991 (2021).
    • 12. Naji A, Eitoku M, Favier B, Deschaseaux F, Rouas-Freiss N, Suganuma N. Biological functions of mesenchymal stem cells and clinical implications. Cell. Mol. Life Sci. 76(17), 3323–3348 (2019).
    • 13. Nikolits I, Nebel S, Egger D, Kreß S, Kasper C. Towards physiologic culture approaches to improve standard cultivation of mesenchymal stem cells. Cells 10(4), 886 (2021).
    • 14. Thamm K, Möbus K, Towers R et al. A chemically defined biomimetic surface for enhanced isolation efficiency of high-quality human mesenchymal stromal cells under xenogeneic/serum-free conditions. Cytotherapy 24(10), 1049–1059 (2022).
    • 15. Zhai W, Yong D, El-Jawhari JJ et al. Identification of senescent cells in multipotent mesenchymal stromal cell cultures: current methods and future directions. Cytotherapy 21(8), 803–819 (2019).
    • 16. Banfi A, Muraglia A, Dozin B, Mastrogiacomo M, Cancedda R, Quarto R. Proliferation kinetics and differentiation potential of ex vivo expanded human bone marrow stromal cells: implications for their use in cell therapy. Exp. Hematol. 28(6), 707–715 (2000).
    • 17. Lin H, Chen H, Zhao X et al. Advances in mesenchymal stem cell conditioned medium-mediated periodontal tissue regeneration. J. Transl. Med. 19(1), 456 (2021).
    • 18. Liu H, Liu S, Qiu X et al. Donor MSCs release apoptotic bodies to improve myocardial infarction via autophagy regulation in recipient cells. Autophagy 16(12), 2140–2155 (2020).
    • 19. Kolosnjaj-Tabi J, Gibot L, Fourquaux I, Golzio M, Rols MP. Electric field-responsive nanoparticles and electric fields: physical, chemical, biological mechanisms and therapeutic prospects. Adv. Drug Deliv. Rev. 138, 56–67 (2019).
    • 20. Sun L, Li M, Zhang S, Bao Z, Lin S. Mechanism of Ser-Ala-Gly-Pro-Ala-Phe treatment with a pulsed electric field to improve ethanol-induced gastric mucosa injury in mice. Food Funct. 13(12), 6716–6725 (2022).
    • 21. Ryan CNM, Doulgkeroglou MN, Zeugolis DI. Electric field stimulation for tissue engineering applications. BMC Biomed. Eng. 3(1), 1 (2021).
    • 22. Fathi E, Farahzadi R. Enhancement of osteogenic differentiation of rat adipose tissue-derived mesenchymal stem cells by zinc sulphate under electromagnetic field via the PKA, ERK1/2 and Wnt/β-catenin signaling pathways. PLoS One 12(3), e0173877 (2017).
    • 23. Thrivikraman G, Boda SK, Basu B. Unraveling the mechanistic effects of electric field stimulation towards directing stem cell fate and function: a tissue engineering perspective. Biomaterials 150, 60–86 (2018).
    • 24. Luo R, Dai J, Zhang J, Li Z. Accelerated skin wound healing by electrical stimulation. Adv. Healthc. Mater. 10(16), e2100557 (2021).
    • 25. Tai G, Tai M, Zhao M. Electrically stimulated cell migration and its contribution to wound healing. Burns Trauma 6, 20 (2018).
    • 26. Aguilar AA, Ho MC, Chang E et al. Permeabilizing cell membranes with electric fields. Cancers (Basel) 13(9), 2283 (2021).
    • 27. Mclaughlin KA, Levin M. Bioelectric signaling in regeneration: mechanisms of ionic controls of growth and form. Dev. Biol. 433(2), 177–189 (2018).
    • 28. Khaw JS, Xue R, Cassidy NJ, Cartmell SH. Electrical stimulation of titanium to promote stem cell orientation, elongation and osteogenesis. Acta Biomater. 139, 204–217 (2022). • Describes a protocol of 100 and 200 mV/mm uniform electric field stimulation applied to mesenchymal stem cells.
    • 29. Martín D, Bocio-Nuñez J, Scagliusi SF et al. DC electrical stimulation enhances proliferation and differentiation on N2a and MC3T3 cell lines. J. Biol. Eng. 16(1), 27 (2022).
    • 30. Panda AK, Sitaramgupta VSNT, Pandya HJ, Basu B. Electrical stimulation waveform-dependent osteogenesis on PVDF/BaTiO(3) composite using a customized and programmable cell stimulator. Biotechnol. Bioeng. 119(6), 1578–1597 (2022).
    • 31. Fathi E, Farahzadi R. Zinc sulphate mediates the stimulation of cell proliferation of rat adipose tissue-derived mesenchymal stem cells under high intensity of EMF exposure. Biol. Trace Elem. Res. 184(2), 529–535 (2018). • Reports an association between electromagnetic field exposure and cell proliferation of mesenchymal stem cells.
    • 32. Zhang B, Xie Y, Ni Z, Chen L. Effects and mechanisms of exogenous electromagnetic field on bone cells: a review. Bioelectromagnetics 41(4), 263–278 (2020).
    • 33. Wang T, Yang L, Jiang J et al. Pulsed electromagnetic fields: promising treatment for osteoporosis. Osteoporos. Int. 30(2), 267–276 (2019).
    • 34. Bostan LE, Almqvist S, Pullar CE. A pulsed current electric field alters protein expression creating a wound healing phenotype in human skin cells. Regen. Med. 15(5), 1611–1623 (2020).
    • 35. Verma A, Asivatham SJ, Deneke T, Castellvi Q, Neal RE 2nd. Primer on pulsed electrical field ablation: understanding the benefits and limitations. Circ. Arrhythm. Electrophysiol. 14(9), e010086 (2021).
    • 36. Li J. Weak direct current (DC) electric fields as a therapy for spinal cord injuries: review and advancement of the oscillating field stimulator (OFS). Neurosurg. Rev. 42(4), 825–834 (2019).
    • 37. Mobini S, Leppik L, Barker JH. Direct current electrical stimulation chamber for treating cells in vitro. BioTechniques 60(2), 95–98 (2016). •• Describes a device delivering direct-current electrical stimulation.
    • 38. Zimmermann J, Budde K, Arbeiter N et al. Using a digital twin of an electrical stimulation device to monitor and control the electrical stimulation of cells in vitro. Front Bioeng. Biotechnol. 9, 765516 (2021).
    • 39. Ren X, Sun H, Liu J et al. Keratinocyte electrotaxis induced by physiological pulsed direct current electric fields. Bioelectrochemistry 127, 113–124 (2019).
    • 40. Yao L, Li Y. The role of direct current electric field-guided stem cell migration in neural regeneration. Stem Cell Rev. Rep. 12(3), 365–375 (2016).
    • 41. Mobini S, Leppik L, Thottakkattumana Parameswaran V, Barker JH. In vitro effect of direct current electrical stimulation on rat mesenchymal stem cells. PeerJ 5, e2821 (2017). •• Reports the in vitro effect of direct-current electrical stimulation on mesenchymal stem cells.
    • 42. Xu J, Jia Y, Huang W et al. Non-contact electrical stimulation as an effective means to promote wound healing. Bioelectrochemistry 146, 108108 (2022).
    • 43. Abedin-Do A, Zhang Z, Douville Y, Méthot M, Bernatchez J, Rouabhia M. Electrical stimulation promotes the wound-healing properties of diabetic human skin fibroblasts. J. Tissue Eng. Regen. Med. 16(7), 643–652 (2022).
    • 44. Kai H, Yamauchi T, Ogawa Y et al. Accelerated wound healing on skin by electrical stimulation with a bioelectric plaster. Adv. Healthc. Mater. 6(22), 1700645 (2017).
    • 45. Tandon N, Goh B, Marsano A et al. Alignment and elongation of human adipose-derived stem cells in response to direct-current electrical stimulation. Annu. Int. Conf. IEEE Eng. Med. Biol. Soc. 2009, 6517–6521 (2009).
    • 46. Lu C, Kolbenschlag J, Nüssler AK et al. Direct current electrical fields improve experimental wound healing by activation of cytokine secretion and Erk1/2 pathway stimulation. Life (Basel) 11(11), 1195 (2021).
    • 47. Hlavac N, Bousalis D, Ahmad RN et al. Effects of varied stimulation parameters on adipose-derived stem cell response to low-level electrical fields. Ann. Biomed. Eng. 49(12), 3401–3411 (2021).
    • 48. Ning T, Zhang K, Heng BC, Ge Z. Diverse effects of pulsed electrical stimulation on cells with a focus on chondrocytes and cartilage regeneration. Eur. Cell Mater. 38, 79–93 (2019).
    • 49. Chen C, Bai X, Ding Y, Lee IS. Electrical stimulation as a novel tool for regulating cell behavior in tissue engineering. Biomater. Res. 23, 25 (2019). •• Summarizes electrical stimulation devices and the effect of electrical stimulation on cell proliferation and differentiation.