1. Introduction
Recombinant growth factors have been increasingly used throughout the years as signaling cues to optimize not only regular cell culture systems but more recently organoid biofabrication platforms [
[1]- Müller S.
- Lindemann S.
- Gigout A.
Effects of sprifermin, IGF1, IGF2, BMP7, or CNP on bovine chondrocytes in monolayer and 3D culture.
,
[2]- Yoon Y.J.
- Kim D.
- Tak K.Y.
- et al.
Salivary gland organoid culture maintains distinct glandular properties of murine and human major salivary glands.
]. These off-the-shelf growth factors are mainly produced in prokaryotic or eukaryotic systems such as bacteria, yeast, and mammalian cells [
[3]- Tripathi N.K.
- Shrivastava A.
Recent developments in Bioprocessing of recombinant proteins: expression hosts and process development.
]. Bacteria is a common host utilized to generate many commercial recombinant growth factors due to ease of growth and manipulation of this microorganism. However, bacterial expression systems have certain limitations including lack of post-translational modifications, formation of insoluble inclusion bodies, and potential contamination with toxins [
[4]Recombinant protein expression in bacteria.
]. To overcome these limitations and also the high production costs related to other systems, plant molecular farming has emerged as a novel, easy adaptable and feasible alternative. Advantages of plant molecular farming include low potential for human pathogen contamination, lower production costs, and the possibility for the inclusion of post-translational modifications in plant-derived protein products [
[5]- Obembe O.O.
- Popoola J.O.
- Leelavathi S.
- et al.
Advances in plant molecular farming.
].
Nicotiana benthamiana is a frequent host for synthesizing recombinant proteins since it can be easily modified via genetic engineering and it is a plant species that can be harvested all year [
[6]- Conley A.J.
- Zhu H.
- Le L.C.
- et al.
Recombinant protein production in a variety of Nicotiana hosts: a comparative analysis.
].
Epidermal growth factor (EGF) is a known signaling cue for exocrine gland organogenesis [
7- Oka T.
- Yoshimura M.
- Lavandero S.
- et al.
Control of growth and differentiation of the mammary gland by growth factors.
,
8- Karasawa Y.
- Shinomiya N.
- Takeuchi M.
- et al.
Growth factor dependence of the proliferation and survival of cultured lacrimal gland epithelial cells isolated from late-embryonic mice.
,
9- Häärä O.
- Koivisto T.
- Miettinen P.J.
EGF-receptor regulates salivary gland branching morphogenesis by supporting proliferation and maturation of epithelial cells and survival of mesenchymal cells.
]. EGF supports epithelial cell proliferation and/or branching morphogenesis of many exocrine glands including the mammary gland [
[10]Role of epidermal growth factor and transforming growth factors in mammary development and lactation.
] and salivary gland (SG) [
[11]Substitution for mesenchyme by basement-membrane-like substratum and epidermal growth factor in inducing branching morphogenesis of mouse salivary epithelium.
,
[12]EGF-dependent lobule formation and FGF7-dependent stalk elongation in branching morphogenesis of mouse salivary epithelium in vitro.
]. In addition, EGF is a critical component in culture media of epithelial organoids such as salivary [
[13]- Nanduri L.S.
- Baanstra M.
- Faber H.
- et al.
Purification and ex vivo expansion of fully functional salivary gland stem cells.
,
[14]- Lee H.W.
- Hsiao Y.C.
- Chen Y.C.
- et al.
Salispheres from different major salivary glands for glandular regeneration.
], mammary [
[15]- Mollica P.A.
- Booth-Creech E.N.
- Reid J.A.
- et al.
3D bioprinted mammary organoids and tumoroids in human mammary derived ECM hydrogels.
], lacrimal [
[16]- Hayashi R.
- Okubo T.
- Kudo Y.
- et al.
Generation of 3D lacrimal gland organoids from human pluripotent stem cells.
], and intestinal [
[17]- Sato T.
- Vries R.G.
- Snippert H.J.
- et al.
Single Lgr5 stem cells build crypt-villus structures in vitro without a mesenchymal niche.
] organoids. Recently, plant-derived recombinant human EGF (P-EGF) has been considered a promising alternative to the commercially available bacteria-derived recombinant human EGF (B-EGF) due to low-cost production and large scalability in the manufacturing process. One of our research teams has successfully produced P-EGF from
Nicotiana benthamiana host [
[18]- Hanittinan O.
- Oo Y.
- Chaotham C.
- et al.
Expression optimization, purification and in vitro characterization of human epidermal growth factor produced in Nicotiana benthamiana.
]. This product had shown a similar effect on skin epithelial cell proliferation when compared to commercial B-EGF in monolayer cultures; however, this P-EGF has not been tested on epithelial organoids and neither on exocrine gland organoid biofabrication.
Organoids are micro-scale three-dimensional (3D) constructs, which are considered as 'mini-organs' since they resemble the structure and function of
in vivo organs. Recently, organoids have been fabricated from various exocrine glands such as mammary [
[19]- Sumbal J.
- Chiche A.
- Charifou E.
- et al.
Primary mammary organoid model of lactation and involution.
], prostate [
[20]- Drost J.
- Karthaus W.R.
- Gao D.
- et al.
Organoid culture systems for prostate epithelial and cancer tissue.
], sweat [
[21]- Diao J.
- Liu J.
- Wang S.
- et al.
Sweat gland organoids contribute to cutaneous wound healing and sweat gland regeneration.
], lacrimal [
[22]- Bannier-Hélaouët M.
- Post Y.
- Korving J.
- et al.
Exploring the human lacrimal gland using organoids and single-cell sequencing.
], and salivary gland [
[23]- Maimets M.
- Rocchi C.
- Bron R.
- et al.
Long-term in vitro expansion of salivary gland stem cells driven by WNT signals.
]. For instance, salivary gland organoids can be established from embryonic stem cells [
[24]- Tanaka J.
- Mishima K.
- Kannan N.
- Beer P.
Generation of salivary gland organoids from mouse embryonic stem cells.
] and adult stem/progenitor cells [
[2]- Yoon Y.J.
- Kim D.
- Tak K.Y.
- et al.
Salivary gland organoid culture maintains distinct glandular properties of murine and human major salivary glands.
,
[25]- Xu Q.L.
- Furuhashi A.
- Zhang Q.Z.
- et al.
Induction of salivary gland-like cells from dental follicle epithelial cells.
], which may recapitulate
in vivo gland. Therefore, glandular epithelial organoids can be a promising model for studying organogenesis [
[26]- Hosseini Z.F.
- Nelson D.A.
- Moskwa N.
- et al.
Generating embryonic salivary gland organoids.
], personalized medicine [
[2]- Yoon Y.J.
- Kim D.
- Tak K.Y.
- et al.
Salivary gland organoid culture maintains distinct glandular properties of murine and human major salivary glands.
], and drug screening [
[27]- Rodboon T.
- Souza G.R.
- Mutirangura A.
- et al.
Magnetic bioassembly platforms for establishing craniofacial exocrine gland organoids as aging in vitro models.
]. Despite the potential of organoid technology, certain limitations linger. For example, organoid culture time usually lasts 8–12 days [
[2]- Yoon Y.J.
- Kim D.
- Tak K.Y.
- et al.
Salivary gland organoid culture maintains distinct glandular properties of murine and human major salivary glands.
,
[27]- Rodboon T.
- Souza G.R.
- Mutirangura A.
- et al.
Magnetic bioassembly platforms for establishing craniofacial exocrine gland organoids as aging in vitro models.
,
[28]- Adine C.
- Ng K.K.
- Rungarunlert S.
- et al.
Engineering innervated secretory epithelial organoids by magnetic three-dimensional bioprinting for stimulating epithelial growth in salivary glands.
], which requires additional and costly media supplements to maintain organoids, augments the potential for cellular genetic aberrations, and increases the waiting time for completion of drug screening assays in patient-derived organoids [
[29]- Karakasheva T.A.
- Kijima T.
- Shimonosono M.
- et al.
Generation and characterization of patient-derived head and neck, oral, and esophageal cancer organoids.
]. In addition, most reports on organoid biofabrication have used xeno-derived extracellular matrix (ECM) extracted from Engelbreth-Holm-Swarm mouse tumors, which generate lot-to-lot biological variations and possess poor human immune compatibility and translational potential [
[30]- Kleinman H.K.
- Martin G.R.
Matrigel: basement membrane matrix with biological activity.
]. Recently, a member of our research group has developed a hydrogel composed of hyaluronic acid (HA) and alginate (Alg) [
[31]- Thanh T.N.
- Laowattanatham N.
- Ratanavaraporn J.
- et al.
Hyaluronic acid crosslinked with alginate hydrogel: a versatile and biocompatible bioink platform for tissue engineering.
]. Both HA and Alg biomaterials are well-known to support
in vitro cell culture platforms [
[32]- Bhatt S.P.
- Harrington D.A.
- Duncan R.L.
- et al.
Implantable three-dimensional salivary spheroid assemblies demonstrate fluid and protein secretory responses to neurotransmitters.
,
[33]- Jorgensen M.
- Ramesh P.
- Toro M.
- et al.
Alginate hydrogel microtubes for salivary gland cell organization and cavitation.
] and have received approval from Food and Drug Administration (FDA) in the United States [
[34]- Cattelan G.
- Guerrero Gerbolés A.
- Foresti R.
- et al.
Alginate formulations: current developments in the race for hydrogel-based cardiac regeneration.
,
[35]- Woodward J.
- Ranjit-Reeves R.
- Katz D.F.
- et al.
Comparing water absorption of food and drug administration-approved hyaluronic acid fillers.
] due to their biocompatibility [
[36]- Ghosh K.
- Reis R.L.
- Neves N.M.
- Mano J.F.
- et al.
28 - Biocompatibility of hyaluronic acid: from cell recognition to therapeutic applications.
,
[37]Alginate: properties and biomedical applications.
]. In our previous report, prior to hydrogel preparation, HA and Alg were chemically modified to HA-NH2 and Alg-CHO, respectively, for tissue engineering purposes. This HA/Alg hydrogel supported
in vitro proliferation and expansion of stem cells [
[31]- Thanh T.N.
- Laowattanatham N.
- Ratanavaraporn J.
- et al.
Hyaluronic acid crosslinked with alginate hydrogel: a versatile and biocompatible bioink platform for tissue engineering.
]. Though, this hydrogel needs signaling cues to induce cellular differentiation because it does not provide in its core any relevant growth factors like EGF for optimal organogenesis and organoid biofabrication. Thus, combining this HA/Alg hydrogel with growth and differentiation cues derived from innovative
Nicotiana benthamiana-based molecular farming technologies requires further investigation to determine whether HA/Alg can support the delivery of such cues.
Herein, this in vitro study aimed to compare the biological effects of our in-house produced P-EGF with commercially available B-EGF during organoid growth and expansion conditions for future scalability of the biofabrication process involving glandular epithelial organoids. Primary submandibular gland (SMG) cells from porcine were utilized for this proof-of-concept investigation as these are similar to their human counterparts and they can be extracted in large amounts. A protein delivery system was developed where P-EGF was encapsulated in a HA/Alg hydrogel in vitro platform to improve the efficacy of glandular epithelial cell proliferation and organoid biofabrication. Both P-EGF and B-EGF supported glandular epithelial cell proliferation at a concentration ranging from 5 – 20 ng/mL. P-EGF-encapsulated HA/Alg hydrogel out-performed conventional P-EGF supplemented to the media in terms of organoid forming efficiency and cell growth. Furthermore, transcriptomic, proteomic, and functional assays supported the application of this P-EGF-encapsulated HA/Alg platform towards glandular epithelial organoid biofabrication processes as well as to provide functionality to such organoids. Thus, this P-EGF-encapsulated hydrogel can be integrated into in vitro culture platforms for exocrine gland organoid development and drug screening, potentially leading to novel pharmacological discoveries.
2. Materials and methods
2.1 Extraction and isolation of glandular epithelial cells
All animal procedures were performed according to the approval of the Institutional Animal Care and Use Committee (IACUC) at the Chulalongkorn University Laboratory Animal Center (protocol number 1,973,004). This study was approved by the Research Ethics Committee (HREC-DCU 2022–019) and the Institutional Biosafety Committee (DENT CU-IBC 026/2021) of the Faculty of Dentistry, Chulalongkorn University. Fresh SMG were collected from 3 to 5-month old
Sus Scrofa (pig). Glands were cut into small pieces (0.5 –1 cm
3) and placed into a 50-mL conical tube together with 30 mL of collection buffer including phosphate buffered saline (PBS) and 10% antibiotic-antimycotic (Gibco, ThermoFisher Scientific, Waltham, MA) at 4 ºC for 6 – 8 h. Connective tissues were removed from porcine tissue sections and primary cells were mechanically and enzymatically dissociated based on previous protocols for mouse and human SG biopsies [
[38]- Pringle S.
- Nanduri L.S.
- van der Zwaag M.
- et al.
Isolation of mouse salivary gland stem cells.
,
[39]- Pringle S.
- Maimets M.
- van der Zwaag M.
- et al.
Human salivary gland stem cells functionally restore radiation damaged salivary glands.
]. Briefly, 200 mg of minced tissue was digested for one hour with 2 mL of PBS containing 1% antibiotic-antimycotic, 1% bovine serum albumin (BSA; Capricorn Scientific GmbH, Ebsdorf, Germany), 1.25 mM calcium chloride (VWR chemicals, Radnor, PA), 0.4 mg/mL collagenase II (Gibco), and 0.7 mg/mL hyaluronidase (Sigma-Aldrich, St. Louis, MO). Digested cells were collected by filtering through 100 μm and 40 μm cell strainers (SPL Life Sciences, Bucheon, Korea), respectively. Cells that flowed through the cell strainers were cultured in growth media consisting of Dulbecco's Modified Eagle Medium/Nutrient Mixture F-12 (DMEM/F12; Sigma-Aldrich) supplemented with 10% fetal bovine serum (FBS; Hyclone, Logan, UT), 1% antibiotic-antimycotic, and 1x
l-glutamine (Hyclone).
2.2 Expansion of glandular epithelial cells
From passage 1 (P1) onwards, the concentration of FBS was decreased to 5% and maintained at this level until P7 to limit the potential fibroblast expansion. Culture media, comprising of DMEM/F12, 5% FBS, 1% antibiotic-antimycotic, and 1x
l-glutamine, were refreshed every 2–3 days and SMG cells were subcultured when reaching 80% confluency. For subculture purposes, cells were dissociated using TrypLE Select (Gibco). At each passage, cell viability was determined using the trypan blue exclusion method. Population doubling time (PDT) from P1 to P7 was calculated as previously described [
[39]- Pringle S.
- Maimets M.
- van der Zwaag M.
- et al.
Human salivary gland stem cells functionally restore radiation damaged salivary glands.
]. Size of suspended cells was measured with Countess 3 automated cell counter (ThermoFisher Scientific, Waltham, MA). Morphology of SMG cells through each passage was observed under phase-contrast light microscopy, and micrographs were taken at 10x magnification with an inverted light microscope (Leica DMi1, Leica Microsystems, Wetzlar, Germany).
2.3 Embryonic murine salivary gland ex vivo culture
Fetal SMG and sublingual glands (SLG) were dissected from ICR mice embryos at embryonic day 16 (E16) under a stereo microscope (SZH10, Olympus, Tokyo, Japan). Glands were placed on top of a porous polycarbonate membrane (Whatman Nucleopore, Sigma-Aldrich), which was floating on growth media composed of DMEM/F12 (Gibco), 1% antibiotic-antimycotic, 150 μg/mL ascorbic acid (Sigma-Aldrich), 100 μg/mL holo-transferrin (Sigma-Aldrich), and B-EGF (2–200 ng/mL) (Sigma-Aldrich). Growth media without B-EGF supplementation was used as a positive control. The protocol was described thoroughly in previous publications [
[40]- Sulistiyani E.
- Brimson J.M.
- Chansaenroj A.
- et al.
Epigallocatechin-3-gallate protects pro-acinar epithelia against salivary gland radiation injury.
,
[41]- Muthumariappan S.
- Ng W.C.
- Adine C.
- et al.
Localized delivery of pilocarpine to hypofunctional salivary glands through electrospun nanofiber mats: an ex vivo and in vivo study.
]. Glands were incubated for 24 h at 37 ºC, 5% CO
2, and were observed under brightfield and phase-contrast microscopy at baseline and 24 h. Number of epithelial buds was counted with ImageJ (version 1.53 s, NIH, Bethesda, MD) at 24 h and normalized to baseline numbers to calculate the Spooner's ratio, also named branching morphogenesis index (BMI).
2.4 Cell proliferation assays
To screen the concentration of B-EGF and P-EGF, an MTT [3-(4,5-dimethyl-2thiazolyl)−2,5-diphenyl-2H-tetrazolium bromide] assay (PanReac AppliChem, Darmstadt, Germany) was used. Briefly, SMG cells (P1 – P3) were seeded in 96-well plates with 1000 cells/well density. Each well contained 200 µL of growth media composed of DMEM/F12 (Sigma-Aldrich), 3% FBS, 1% antibiotic-antimycotic, and 1x l-glutamine. Culture plates were incubated at 37 ºC, 5% CO2 for 24 h to allow cells attach completely. Then, growth media was replaced at baseline by fresh growth media supplemented with B-EGF or P-EGF at concentration ranging from 5 ng/mL to 20 ng/mL. Growth media without B-EGF or P-EGF was used as control (CTL). Plates were incubated (37 ºC, 5% CO2) for 2 h, 3 days, and/or 6 days. MTT assay was done at each time point according to the manufacturer's protocol. Glomax Discover Microplate Reader (Promega, Madison, WI) was used to measure absorbance at 560 nm. Optical density (OD) values at 3 days and 6 days were normalized to OD values at 2 h to calculate the fold change of cell proliferation.
Luciferase-based ATP assay such as CellTiter Glo 3D assay (Promega) and ATPlite (PerkinElmer, Waltham, MA) was used to determine the optimal concentration of B-EGF and P-EGF in monolayer and 3D culture. For monolayer cell culture, the experimental design was similar to the above mentioned MTT assay. In 3D culture, 10,000 SMG cells/well were seeded in a 96-well plate. Each well was pre-coated with 20 µL of HA/Alg hydrogel. This assay was conducted following the manufacturer's protocol. Glomax Discover Microplate Reader was used to measure luminescence. Then, all bioluminescence values were subtracted to background values from media only wells to calculate the final bioluminescence. Fold change in cell proliferation was calculated by normalizing bioluminescence values of later time points to numbers at baseline.
2.5 HA/Alg hydrogel degradation
Hyaluronic acid sodium salt (Sigma-Aldrich) and sodium alginate (Sigma-Aldrich) were chemically modified to amine-hyaluronic acid (HA-NH
2) and aldehyde-alginate (Alg-CHO), respectively. Detailed protocol of HA and Alg modification was reported previously [
[31]- Thanh T.N.
- Laowattanatham N.
- Ratanavaraporn J.
- et al.
Hyaluronic acid crosslinked with alginate hydrogel: a versatile and biocompatible bioink platform for tissue engineering.
]. To determine the optimal formula for HA/Alg hydrogel, volume ratio of HA:Alg:PBS was varied as follows: 5:4:1, 6:3:1, 7:2:1, 8:1:1. Exact volume of each hydrogel's component was shown in table S2. Alginate 20 mg/mL and PBS were mixed thoroughly before adding to 18 mg/mL HA. This mixture was vortexed for 30 s and then transferred 50 µL to each well of 96 well-plate. All procedures were performed on ice to prolong the gelling time. Two hundred µL of DMEM/F12 were added to each well and refreshed every 3 days. To observe hydrogel degradation byproducts released to media, discarded media was placed in a microscope glass slide with a hydrophobic circle to let the media air-dry for three hours at room temperature. Glass slide was washed with 200 µL PBS three times to remove media. Next, the remained substances were stained with 100 µL rhodamine-labeled peanut agglutinin (Rho-PNA; Vector Laboratories, Burlingame, CA) 5 µg/mL within one hour. Glass slides were observed under a fluorescent microscope (Evos FL Auto II, ThermoFisher Scientific). This process was repeated at every time point, 3, 6, 9, 12, and 14 days.
2.6 Protein release profile
HA/Alg hydrogels (BSA loaded and unloaded) were used to determine the BSA release profile. Hydrogels were fabricated similarly to the experiment of hydrogel degradation for two ratios 5:4:1 and 6:3:1. BSA 40 mg/mL replaced PBS in the BSA-loaded group. At pre-determined time intervals (2 h, 1, 2, 3, 6, 9, 12, and 14 days), the supernatants (10 μL) were collected to measure the concentration of BSA released by Pierce™ BCA Protein Assay Kit (ThermoFisher Scientific). Fresh DMEM/F12 of 10 μL was added to the original sample well. Same procedure was repeated for all collection time points. Cumulative release of BSA was determined by the following equations [
[42]- Zhang Y.
- Li X.
- Zhong N.
- et al.
Injectable in situ dual-crosslinking hyaluronic acid and sodium alginate based hydrogels for drug release.
]:
mt: the total quantity of BSA released at time t, m0: the quantity of BSA loaded in hydrogel, and Ci: the concentration of BSA in the released media at time t.
To determine the P-EGF release profile with optimal volume ratio 5:4:1, BSA was replaced by 10 ng/mL P-EGF. PBS was used as blank control. Each replicate had 12 wells per group. At each time point (2 h, 1, 2, and 3 days), 10 µL/well of supernatants were collected from each group (12 wells). Mixture of supernatants was centrifuged in a Vivaspin 500 concentrator (GE Healthcare, Chicago, IL) to increase the concentration of P-EGF in the mixture. After such a step, the measurement of P-EGF concentration was similar to BSA release profile.
2.7 Cytotoxicity and viability assays
Firstly, 96-well plate was coated with 20 µL hydrogel. Non-coated wells were used as CTL. Growth media composed of DMEM/F12, 5% FBS, 1% antibiotic-antimycotic, and 1x l-glutamine was added to each well, and plate was incubated for 3 days. Conditioned media of both coated and non-coated groups was collected every 24 h. These conditioned media were stored at −20 ºC until usage. Next, SMG cells were seeded with a density of 5000 cells/well and cultured for 1, 2, and 3 days with respective conditioned media.
To determine cytotoxicity of hydrogel to SMG cells, a CellTox Green cytotoxicity assay kit (Promega) was used according to manufacturer's instructions. Briefly, 100 µL of CellTox Green reagent (2X) was added to 100 µL media with cells. Plate was incubated at room temperature for 15 min (protected from light). Solution in each well was further transferred to an opaque-walled 96-well plate. Glomax Discover Microplate Reader was used to measure fluorescence with excitation 475 nm and emission 500–550 nm.
To measure cell viability during culture, a ReadyProbes™ cell viability imaging kit (Blue/Green, Invitrogen, ThermoFisher Scientific) was used following instructions of manufacturer. Briefly, two drops of NucBlue™ Live and NucGreen™ Dead were added to 1 mL of media with cells. NucBlue™ reagent stained all nuclei, whereas NucGreen™ Dead stained nuclei of compromised cell membranes. Next, fluorescent micrographs were taken using Evos FL Auto II. These micrographs were analyzed by CellProfiler (version 4.2.1, Broad Institute, Cambridge, MA). Finally, cell viability was quantified by dividing the number of live cells by total cells.
2.8 Organoid forming efficiency
To assess organoid forming efficiency of SMG cells cultured over P-EGF-encapsulated HA/Alg
in vitro platforms, 10,000 cells were seeded in the well pre-coated with P-EGF-encapsulated hydrogel. HA/Alg hydrogel without P-EGF encapsulation was used as a CTL. In CTL group, P-EGF 20 ng/mL was supplemented in the growth media, which was equal amount as P-EGF-encapsulated hydrogel. Basic growth media used to culture SG organoids was composed of DMEM/F12, 3% FBS, 1% antibiotic-antimycotic, 1x
l-glutamine, 20 ng/mL FGF2 (ImmunoTools, Friesoythe, Germany), 1x NDiff Neuro-2 (Sigma-Aldrich), 10 µg/mL insulin (Sigma-Aldrich), 1 µM dexamethasone (Sigma-Aldrich) [
[39]- Pringle S.
- Maimets M.
- van der Zwaag M.
- et al.
Human salivary gland stem cells functionally restore radiation damaged salivary glands.
]. After 3 days of culture, the number of organoids larger than 50 µm was noted. Organoid forming efficiency was calculated as follows [
[43]- Rocchi C.
- Cinat D.
- Serrano Martinez P.
- et al.
The Hippo signaling pathway effector YAP promotes salivary gland regeneration after injury.
]:
Organoid forming efficiency of P-EGF-encapsulated hydrogel was normalized to P-EGF supplemented media group to calculate fold change.
2.9 Quantifying viable cell density in 3D culture
To access the expansion of glandular epithelial cells in the presence of PBS-, B-EGF-, and P-EGF-encapsulated hydrogels, 10,000 cells were cultured in each well of a 96-well plate that had been pre-coated with the respective hydrogels. In order to obtain sufficient cell numbers for the cytometric analysis, 10 wells were combined per experiment group as well as per time point and 3 runs per group were performed. Organoids were dissociated on days 1, 2, and 3 using TrypLE Select, and the number of viable cells was determined using the trypan blue exclusion method and Countess 3 automated cell counter. The viable cell density at each time point was divided by 10 to calculate the final cell density per well.
2.10 Gene expression arrays
Gene expression of glandular epithelial genes in SMG cells (P1 to P3) and epithelial organoids was assessed using quantitative polymerase chain reaction (qPCR). Firstly, total RNA was extracted from SMG cells and organoids using the Monarch® Total RNA Miniprep Kit (New England Biolabs, Ipswich, MA). Next, cDNA was synthesized from total RNA using reverse transcriptase enzyme SuperScriptTM III First-Strand Synthesis System (ThermoFisher Scientific). SYBR® green-based qPCR was performed using 1 ng of cDNA. Forward and reverse sequences of all oligonucleotide primers are listed in Table S3. Then, qPCR reaction was conducted with a mixture of 10 µL cDNA, 9.5 µL QuantiTect SYBR® Green PCR kit (QIAGEN, Hilden, Germany), and 0.5 µL of forward and reverse oligonucleotide primer using an Applied Biosystems QuantStudio 3 Real-Time PCR System (ThermoFisher Scientific). Data was analyzed using the 2−(ddCT) approach to calculate the relative expression of target genes of SMG cells and organoids after normalization with a housekeeping gene (S29) relative to control groups (SMG cells at P1 in monolayer culture or SMG cells at baseline for SG organoids).
2.11 Immunocytochemistry and whole-mount immunohistochemistry
To detect specific glandular epithelial markers, SMG cells in monolayer culture and organoids were fixed with 4% paraformaldehyde (Sigma-Aldrich) for 10 min, followed by permeabilization with 0.1% Triton X (Loba Chemie, Mumbai, India) for 15 min, and then washed with PBS. Next, monolayer cells or organoids were blocked in 10% horse serum (Gibco), 5% BSA, and 0.1% Tween 20 (Loba Chemie) in PBS for 2 h. After the blocking step, incubation with primary antibodies at 4 ºC was performed overnight. This was followed by three PBS washing steps and incubation with secondary antibodies for one hour. Primary antibodies, secondary antibodies, and their respective concentration are listed in table S4. Hoechst 33,342 (ThermoFisher Scientific) was used to counterstain nuclei. SG organoids were transferred to a glass slide and then mounted by resin mounting media, whereas monolayer cells were observed directly in a 96-well plate. Evos FL Auto II was used to take fluorescence micrographs of immunostained cells (in monolayer and organoids). These micrographs were analyzed by CellProfiler and ImageJ.
2.12 Intracellular calcium activity
A Fluo‐4 Direct Calcium assay kit (Invitrogen, ThermoFisher Scientific) was used according to the manufacturer procedure to assess the intracellular calcium influx in epithelial organoids. Organoids were stimulated with carbachol (an acetylcholine receptor agonist) at a dosage of 10 μM. Fluorescence signal was taken by Evos Auto FL II at before and after 1 h of carbachol treatment.
2.13 α-Amylase enzymatic activity
To assess the functional enzymatic activity of epithelial organoids, an α-amylase activity assay was performed using Enzcheck™ amylase kit (ThermoFisher Scientific) according to the manufacturer instructions. Epithelial organoids at day 3 were treated with 10 µM of carbachol overnight then culture media was collected from untreated and treated group. Glomax Discover Microplate Reader was used to measure fluorescence. Eliminating background fluorescence in wells containing only fresh medium enabled the calculation of relative fluorescence units (RFU). Fluorescence values of the treated group were normalized to the numbers of untreated group to calculate fold change of amylase activity.
2.14 Statistical analysis
Mean, standard deviation (SD) or standard error of the mean (SEM) were used to display all data as specified in figure captions. To compare two groups, Student's t-test (paired or unpaired) or multiple unpaired Student's t-tests were utilized. To compare three or more groups, we used a one-way ANOVA or two-way ANOVA with Dunnett or Turkey post-hoc analysis. Statistical significance was defined at 5%. GraphPad Prism (version 9, GraphPad, San Diego, CA) was used to conduct all of the statistical analyses.
4. Discussion
This is the first study to investigate whether a plant-derived EGF produced from Nicotiana benthamiana can replace B-EGF in the biofabrication of glandular epithelial organoids. Herein, an in vitro EGF delivery platform was successfully developed comprising of HA/Alg hydrogel and encapsulated P-EGF to efficiently generate mitotically active organoids that are amenable to expansion and possess specific glandular epithelial features.
The manufacturing process of recombinant proteins is deemed highly relevant in the pharmaceutical industry for producing complex therapeutic molecules, as well as in the biotechnology industry focused on the development of soluble protein cues for the
in vitro biofabrication of cellularized 3D constructs (a.k.a. organoids) and tissue engineering applications. Various cell lines were transformed and applied to biomanufacturing processes, including yeast, bacteria, insect, and mammalian cells. The ability of Chinese hamster ovary (CHO) cells to carry out post translational modifications makes these a very popular production system [
[47]- Gupta V.
- Sengupta M.
- Prakash J.
- Gupta V.
- Sengupta M.
- Prakash J.
- et al.
Production of recombinant pharmaceutical proteins.
]. However, its slow growth, relatively high production costs and long manufacturing times, is driving the field to relay on low-cost bacteria production systems, particularly the ones using endotoxin-free strains. The lack of intracellular key enzymes crucial for posttranslational modifications and proper protein folding of multi-domain proteins, and the need to increase the protein yields have turned the attention to plant molecular farming.
Nicotiana benthamiana or tobacco plant producing systems can result in high protein yields of up to 400 µg/mL [
[48]- Buntru M.
- Vogel S.
- Spiegel H.
- et al.
Tobacco BY-2 cell-free lysate: an alternative and highly-productive plant-based in vitro translation system.
]
. Hence, several companies based in North America, Germany and Thailand (Medicago Inc., Icon Genetics, Baiya Phytopharm Co. Ltd.), have set up current good manufacturing practices (cGMP) and regulatory approval processes to produce recombinant proteins in plants. This major step was boosted by promising pre-clinical trials with plant-produced vaccines [
[49]- Shanmugaraj B.
- Khorattanakulchai N.
- Paungpin W.
- et al.
Immunogenicity and efficacy of recombinant subunit SARS-CoV-2 vaccine candidate in the Syrian hamster model.
,
[50]- Shanmugaraj B.
- Khorattanakulchai N.
- Panapitakkul C.
- et al.
Preclinical evaluation of a plant-derived SARS-CoV-2 subunit vaccine: protective efficacy, immunogenicity, safety, and toxicity.
]. The major advantages of plant expression systems include low cost, high scalability, and these systems do generally offer a higher and more robust biosafety profile than other common expression systems (mammalian or bacteria).
In conventional monolayer culture systems, both P-EGF and B-EGF (5 – 20 ng/mL) supported epithelial cell proliferation and metabolism like in previous reports, where P-EGF was comparable to B-EGF for the proliferation of kidney epithelial cells [
[51]- Bai J.Y.
- Zeng L.
- Hu Y.L.
- et al.
Expression and characteristic of synthetic human epidermal growth factor (hEGF) in transgenic tobacco plants.
] and human keratinocytes [
[18]- Hanittinan O.
- Oo Y.
- Chaotham C.
- et al.
Expression optimization, purification and in vitro characterization of human epidermal growth factor produced in Nicotiana benthamiana.
]. Moreover, Wirth and colleagues reported that P-EGF can bind to EGF receptors with the same affinity as B-EGF does [
[52]- Wirth S.
- Calamante G.
- Mentaberry A.
- et al.
Expression of active human epidermal growth factor (hEGF) in tobacco plants by integrative and non-integrative systems.
]. Interestingly, P-EGF enhanced this epithelial proliferation and ATP-dependent metabolism earlier (day 3), whereas B-EGF did such later (day 6). In the 3D culture system, P-EGF-encapsulated HA/Alg platform but less noticeably the B-EGF one supported the differentiation of epithelial cell clusters as per upregulation of AQP5 and
Krt19 after short-term culture (3 days). This maybe possible due to the gain of function after proper folding of proteins when these are produced by plant expression systems; whereas bacteria-based systems lack enzymes responsible for post-translational modifications leading to misfolding of protein products [
[53]- Nosaki S.
- Hoshikawa K.
- Ezura H.
- et al.
Transient protein expression systems in plants and their applications.
]. Thus, P-EGF may be biologically more effective than B-EGF in terms of epithelial proliferation and pro-acinar differentiation.
The rationale for encapsulating P-EGF in the hydrogel is based on the natural development of the exocrine gland organ, marked by fluctuations in the expression of EGF through time [
]. Hence, the levels of EGF are critical for proper organoid development: if one is exposing primary SG cells to high levels of EGF at early stages, this may lead to a negative feedback loop, which downregulates EGFR-activated signaling [
[55]- Ma H.
- Zongyu Z.
- Tanjun T.
The effects of epidermal growth factor on gene expression in human fibroblasts.
]. Concerning the developed hydrogel platform for EGF
in vitro release, variation of HA and Alg volume ratio affected hydrogel degradation. The most stable volume ratio was 5:4:1, whereas 8:4:1 ratio degraded the fastest. This outcome may be due to a higher imine bond between HA and Alg in 5:4:1 ratio. These findings are supported by previous literature where 5:5 vol ratio of HA and Alg degraded slowly and was stable until day 50 in PBS solution [
[31]- Thanh T.N.
- Laowattanatham N.
- Ratanavaraporn J.
- et al.
Hyaluronic acid crosslinked with alginate hydrogel: a versatile and biocompatible bioink platform for tissue engineering.
]. Our hydrogel released BSA and P-EGF on a sustained manner within 3 days and was suitable for short-term culture strategies for prompt drug screening and testing. In addition, HA/Alg hydrogel platforms lacked cytotoxicity to glandular epithelial cells. Hyaluronic acid and alginate have been widely used to fabricate hydrogels for biotechnology platforms, and several reports have shown their biocompatibility features for cells [
[56]- Zhang Q.
- Wei X.
- Ji Y.
- et al.
Adjustable and ultrafast light-cured hyaluronic acid hydrogel: promoting biocompatibility and cell growth.
], organoids [
[33]- Jorgensen M.
- Ramesh P.
- Toro M.
- et al.
Alginate hydrogel microtubes for salivary gland cell organization and cavitation.
], and tissue culture systems [
[57]- Lee S.W.
- Ryu J.H.
- Do M.J.
- et al.
NiCHE platform: nature-inspired catechol-conjugated hyaluronic acid environment platform for salivary gland tissue engineering.
]. Interestingly, HA and Alg can be chemically modified by various methods for specific applications such as injectable drug delivery system [
[42]- Zhang Y.
- Li X.
- Zhong N.
- et al.
Injectable in situ dual-crosslinking hyaluronic acid and sodium alginate based hydrogels for drug release.
,
[58]- Li J.
- Sudiwala S.
- Berthoin L.
- et al.
Long-term functional regeneration of radiation-damaged salivary glands through delivery of a neurogenic hydrogel.
] and adipose tissue engineering [
[59]- Chen Y.S.
- Chen Y.Y.
- Hsueh Y.S.
- et al.
Modifying alginate with early embryonic extracellular matrix, laminin, and hyaluronic acid for adipose tissue engineering.
]. Multiple cues or growth factors have been encapsulated into HA/Alg hydrogels to generate protein delivery systems, such as hepatocyte growth factor [
[60]- Choi J.S.
- Heang Oh S.
- Kim Y.M.
- et al.
Hyaluronic acid/alginate hydrogel containing hepatocyte growth factor and promotion of vocal fold wound healing.
], bFGF [
[61]- Choi Y.H.
- Kim S.H.
- Kim I.G.
- et al.
Injectable basic fibroblast growth factor-loaded alginate/hyaluronic acid hydrogel for rejuvenation of geriatric larynx.
], vascular endothelial growth factor and keratinocyte growth factor [
[62]- Peattie R.A.
- Rieke E.R.
- Hewett E.M.
- et al.
Dual growth factor-induced angiogenesis in vivo using hyaluronan hydrogel implants.
]. Though, EGF has never been encapsulated in such hydrogels and our goal was to determine whether our hydrogel-based EGF delivery platform can optimize the biofabrication process of exocrine gland epithelial organoids. Indeed, our P-EGF encapsulated HA/Alg hydrogel
in vitro platform supported epithelial organoid formation by enhancing organoid forming efficiency and cellular viability, expansion and ATP-dependent metabolic activity in the organoid culture. This might be due to the sustainable release of P-EGF from the HA/Alg hydrogel, which may better mimic the release of soluble EGF to the microenvironment during exocrine gland epithelial development and morphogenesis [
]. The manufactured basal media with only 3% FBS (which was used across all experimental groups and controls) also carries certain soluble growth factors like EGF [
[63]- Lee D.Y.
- Lee S.Y.
- Yun S.H.
- et al.
Review of the current research on fetal bovine serum and the development of cultured meat.
], but when we compare PBS-encapsulated hydrogel with B-EGF- or P-EGF-encapsulated hydrogel, the sustained release of B-EGF or P-EGF to the media significantly promoted cellular viability and growth in the organoid after day 1 (
Fig. 5D). Hence, the media supplementation step with soluble factors (individually or using serum) can be minimized or perhaps completely eliminated as we move towards scalability and humanized models in organoid biofabrication and biomanufacturing processes. This means that our P-EGF encapsulation hydrogel is the way forward to optimize organoid biomanufacturing and reduce the exposure to growth factor or serum supplementation, which is well known to have lot-to-lot variations that can impair the consistency and reproducibility of organoid cultures [
[64]- Zhao Z.
- Chen X.
- Dowbaj A.M.
- et al.
Organoids.
].
Our P-EGF hydrogel platform supported epithelial viability within 3 days but not more than 5 days (Supplemental Fig. S9). Furthermore, these organoids expressed specific glandular epithelial markers and high mitotic activity (38–62%). Hydrogels derived from HA have been previously used to encapsulate human salivary gland (SG) stem and progenitor cells. However, the size of the SG spheroids was 57.1 ± 12.3 µm after 28 days of culture, indicating cells grew slowly inside this hydrogel [
[65]- Ozdemir T.
- Fowler E.W.
- Liu S.
- et al.
Tuning hydrogel properties to promote the assembly of salivary gland spheroids in 3D.
]. Similarly, our HA/Alg hydrogel was also used to encapsulate glandular epithelial cells; however, organoids did not grow well (data not shown). More interestingly, these glandular epithelial organoids were successfully generated when epithelial cells were cultured on top of the hydrogel platform. This may relate to the fact that P-EGF can support epithelial mitosis, self-renewal, and aggregation [
[18]- Hanittinan O.
- Oo Y.
- Chaotham C.
- et al.
Expression optimization, purification and in vitro characterization of human epidermal growth factor produced in Nicotiana benthamiana.
]. Our preliminary organoid functional outcomes are consistent with Pradhan and colleagues, in which human parotid gland cells were cultured on top of a HA-based hydrogel and such exhibited secretory functions similar to the ones observed herein [
[32]- Bhatt S.P.
- Harrington D.A.
- Duncan R.L.
- et al.
Implantable three-dimensional salivary spheroid assemblies demonstrate fluid and protein secretory responses to neurotransmitters.
]; however, our group achieved this organoid functionality in only 3 culture days (
versus 12 days with their previous report).
In conclusion, P-EGF-encapsulated HA/Alg in vitro platforms can shorten organoid culture time to 3 days and efficiently improve organoid formation and cellular viability, expansion and ATP-dependent metabolism when compared to B-EGF encapsulation or media supplementation. In addition, organoids fabricated on our P-EGF-encapsulated platforms displayed a robust mitotic activity, a large epithelial progenitor population and a wide variety of glandular epithelial cells (pro-acinar and ductal) amenable to cholinergic stimulation. Future studies will focus on applying this P-EGF-encapsulated HA/Alg hydrogel via an automated bioprinting apparatus into in vitro culture systems (culture plates/dishes) to optimize the consistency of organoid biofabrication. Combining humanized media components as well as glandular epithelial cells from human biopsies will be our main focus during the scalability of this in vitro platform. From a regenerative medicine stand point, our P-EGF-encapsulated hydrogel may have a gland regenerative potential as an injectable drug delivery system, but this needs to be investigated in upcoming in vivo studies.
Declaration of Competing Interest
Joao N. Ferreira reports financial support and equipment, drugs, or supplies were provided by National Research Council of Thailand. Joao N. Ferreira reports financial support, equipment, drugs, or supplies, and statistical analysis were provided by International Association for Dental Research. Waranyoo Phoolcharoen reports a relationship with Baiya Phytopharm Co., Ltd that includes: board membership, funding grants, and non-financial support. Supansa Yodmuang and Truc Thanh Nguyen has patent #63/240,262 pending to Chulalongkorn University. Waranyoo Phoolcharoen has patent issued to Chulalongkorn University.
The authors declare the following ownership, which may be considered as potential competing interests: Dr. Waranyoo Phoolcharoen is CTO and co-founder of Baiya Phytopharm Co., Ltd, a Chulalongkorn university-based spin-off company.
Article info
Publication history
Published online: March 24, 2023
Accepted:
March 21,
2023
Received in revised form:
February 24,
2023
Received:
August 30,
2022
Publication stage
In Press Corrected ProofCopyright
© 2023 The Author(s). Published by Elsevier Inc. on behalf of Society for Laboratory Automation and Screening.