ArticlePDF Available

Isolation, Culture, and Characterization of Primary Salivary Gland Cells

Authors:

Abstract

Primary cells are an essential tool for in vitro studies and are obtained directly from living tissues or organs. They closely mimic the physiological state and maintain in vivo functions for short periods of time under optimal conditions. Isolation and culture of salivary gland (SG) cells are useful to decipher the various mechanisms involved in salivary gland dysfunction. However, unlike some other primary cell cultures, SG cell cultures from patient‐derived tissues present several challenges. They are difficult to obtain, culture, expand, and characterize due to their sensitive heterogenous cell population and limited expansion potential. In addition, the majority of saliva‐secreting acinar cells fail to maintain a differentiated state ex vivo for long periods, and eventually succumb to an acinar‐to‐ductal metaplasia, losing their secretory phenotype and functions. Herein, we describe two detailed protocols for primary SG cell isolation, culture, and expansion from human (or mouse) salivary tissues using serum‐free culture media. We also describe the growth kinetics of these primary cells along with their immunocytochemical characterization. © 2022 Wiley Periodicals LLC. Basic Protocol 1 : Preparation of SG single‐cell culture from freshly obtained human or mouse SG tissues. Basic Protocol 2 : Preparation of SG explant culture from freshly obtained human or mouse SG tissues
Isolation, Culture, and Characterization
of Primary Salivary Gland Cells
Xinyun Su,1,2 Sangeeth Pillai,1Younan Liu,1and Simon D. Tran1,3
1McGill Craniofacial Tissue Engineering and Stem Cells Laboratory, Faculty of Dental
Medicine and Oral Health Sciences, McGill University, Montreal, Canada
2Guangdong Provincial Key Laboratory of Stomatology, Department of Operative Dentistry
and Endodontics, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou,
China
3Corresponding author: simon.tran@mcgill.ca
Published in the Cell Biology section
Primary cells are an essential tool for in vitro studies and are obtained directly
from living tissues or organs. They closely mimic the physiological state and
maintain in vivo functions for short periods of time under optimal conditions.
Isolation and culture of salivary gland (SG) cells are useful to decipher the
various mechanisms involved in salivary gland dysfunction. However, unlike
some other primary cell cultures, SG cell cultures from patient-derived tissues
present several challenges. They are difcult to obtain, culture, expand, and
characterize due to their sensitive heterogenous cell population and limited
expansion potential. In addition, the majority of saliva-secreting acinar cells
fail to maintain a differentiated state ex vivo for long periods, and eventually
succumb to an acinar-to-ductal metaplasia, losing their secretory phenotype
and functions. Herein, we describe two detailed protocols for primary SG cell
isolation, culture, and expansion from human (or mouse) salivary tissues us-
ing serum-free culture media. We also describe the growth kinetics of these
primary cells along with their immunocytochemical characterization. © 2022
Wiley Periodicals LLC.
Basic Protocol 1: Preparation of SG single-cell culture from freshly obtained
human or mouse SG tissues.
Basic Protocol 2: Preparation of SG explant culture from freshly obtained hu-
man or mouse SG tissues
Keywords: acinar cells rductal cells rexplant culture rprimary epithelial
cells rsalivary glands rsingle-cell culture
How to cite this article:
Su, X., Pillai, S., Liu, Y., & Tran, S. D. (2022). Isolation, culture,
and characterization of primary salivary gland cells. Current
Protocols,2, e479. doi: 10.1002/cpz1.479
INTRODUCTION
Salivary glands (SGs) are indispensable exocrine organs that produce saliva to maintain
homeostasis in the oral cavity. In mammals, there are three pairs of major salivary glands
(parotid, submandibular, and sublingual glands) and hundreds of minor salivary glands
(de Paula et al., 2017). Each of these major and minor SGs consist of acinar cells, my-
oepithelial cells, and ductal cells supported by a complex neural and vascular supply.
Major SGs are responsible for most of the salivary secretion, while minor SGs contribute
to lubricating the mucosa, as they are dispersed throughout the mucosa in palatal, buc-
cal, lingual, and labial tissues (Hand, Padmanabhan, & Field, 1999; Riva et al., 2000).
Current Protocols e479, Volume 2
Published in Wiley Online Library (wileyonlinelibrary.com).
doi: 10.1002/cpz1.479
© 2022 Wiley Periodicals LLC.
Su et al.
1of13
Tab le 1 Sequence of Events
Time Event
Day 0-1 Tissue digestion and single-cell seeding
Day 4-6 Re-seeding the oating cells into new plates (small colonies start forming)
Day 10-17 Colonies of cells start expanding and connecting
Day 17-21 Cell passaging and re-seeding
Saliva secretion is essential for health maintenance, for example, in digestion, lubrication,
taste, and protection against microorganisms (Pedersen, Bardow, Jensen, & Nauntofte,
2002). Reduced saliva secretion inevitably causes complications such as secondary ram-
pant caries, dysphagia, dysgeusia, and inammation of the oral mucosa (Vissink, Burlage,
Spijkervet, Jansma, & Coppes, 2003). As such, a better understanding of salivary glands
at the cellular level can enhance our success in restoring their secretory function and in
regenerating and bioengineering salivary tissues.
The ‘Human Salivary Gland’ (HSG) cell line has been commonly used in SG research
(Shirasuna, Sato, & Miyazaki, 1981). However, as an immortalized cell line, HSG cells
have limited potential for clinical applications (Tran et al., 2006). Additionally, HSG
was reported as being contaminated with HeLa cells (Lin et al., 2018). Given the lack of
availability of stable cell lines for SG disease modeling and in vitro drug testing, scien-
tists have alternatively used primary SG epithelial cells isolated and characterized from
murine salivary glands (Maimets et al., 2016; Pringle, Nanduri, van der Zwaag, van Os, &
Coppes, 2011; Varghese et al., 2019) and human salivary glands (Beucler & Miller, 2019;
Seo et al., 2019; Su et al., 2016; Tran et al., 2005). Herein, we describe two protocols that
are convenient and reproducible to isolate epithelial cells from human salivary glands:
rst, the single-cell culture method (Basic Protocol 1), and second, the explant culture
method. Single-cell culture is the preferred approach when larger tissues are available
from which to isolate cells, for example, a complete major SG. It allows for complete
utilization of glands and obtaining cell-type-specic cultures for various in vitro appli-
cations. Explant cultures on the other hand are efcient for culturing primary cells out of
smaller tissues, like human minor SGs or glands isolated from mice. This allows us to
maximize the number of cells from a small amount of tissue with high viability. The tech-
nique is less time consuming (Table 1) and more cost-efcient, as no additional enzyme
processing or mechanical dissociation is required to digest the tissues, and the culture can
be achieved using simple surgical instruments and culture reagents routinely available in
labs (Agnihotri, Gaur, & Albin, 2020). However, the type of cells released from the ex-
plants usually depends on the tissue site from which it is isolated. Sometimes, failure to
remove the brous capsule from the gland tissue may lead to limited or no cell release
from these small tissue pieces. Based on our experience, to get an increase in the total
output of cells, it is advisable to use a combination of both single-cell and explant culture
techniques, depending on the type and size of glands.
NOTE: All procedures are performed in a Class II biological hazard ow hood.
NOTE: All work with human samples should be covered under the Institutional Research
Ethics Board. The work presented here adheres to the regulations from the McGill Re-
search Ethics Board. All information on patients and the samples obtained are docu-
mented and are kept condential.
NOTE: All solutions and equipment used in the handling and processing of tissue and
cells must be sterile, and proper aseptic techniques should be used. All personnel han-
dling human samples should wear disposable gloves, disposable gowns, and protective
eyewear.
Su et al.
2of13
Current Protocols
NOTE: All incubation steps are performed in a humidied 37°C, 5% CO2incubator un-
less otherwise specied.
BASIC
PROTOCOL 1
PREPARATION OF SALIVARY GLAND (SG) SINGLE-CELL CULTURE
FROM FRESHLY OBTAINED HUMAN OR MOUSE SG TISSUES
In this protocol, we describe the use of Liberase enzymes to digest fresh human and
mouse SG tissues to obtain single cells, which can be used for downstream applications
such as radiation response, drug testing, or tissue engineering. This protocol is usually
applied when larger tissues (such as a complete human SGs) or several smaller tissues
(such as SGs from multiple mice) are available to isolate cells from.
The timeline from tissue harvesting to the culture of single cells (Basic Protocol 1) is
shown in Table 1.
Materials
Fresh human or mouse salivary gland tissues
Washing solution: phosphate-buffered saline (PBS), pH 7.4; or calcium-,
magnesium-, and phenol red–free Hank’s Balanced Salt Solution (HBSS; Gibco,
cat. no. 14175-095) with 2% antibiotic-antimycotic (Gibco, cat. no. 15240-062)
If freshly isolated tissues have bleeding spots or coagulated blood clots, add 1000
U heparin (Sandoz, cat no. 02303086) to washing solution (PBS or HBSS)
Digestion buffer: DMEM medium with 35 μg/L Liberase (Roche, cat. no.
05401119001) and 1% (v/v) antibiotic/antimycotic
Sterile 0.9% NaCl solution
Complete medium: Epi Max (Wisent, cat. no. 002-010-CL; https://www.
wisentbioproducts.com/en/ 113-special-media-systems) with 1%
antibiotic/antimycotic
100-mm plastic cell culture dishes
Ruler and electronic scale to calculate size/weight of sample
Ice or ice packs
Individually wrapped sterile pipettes (5 ml, 10 ml, and 25 ml)
15-ml and 50-ml sterile conical tubes
Surgical scissors (curved), scalpels (Fisher scientic, cat no. 36-101-5346),
tweezers
Micropipette and pipette tips (1000 μl)
GentleMACS tubes (Miltenyi Biotec, cat. no. 5150320302)
GentleMACS dissociator (Miltenyi Biotec)
70-μm cell strainers (Fisher Scientic, cat. no. 22363548)
Humidied, 37°C, 5% CO2cell culture incubator
37°C water bath
Additional reagents and equipment for cell culture techniques including counting
cells (see Current Protocols article: Phelan & May, 2015)
Tissue cleaning
1. Transfer the tissues into a pre-weighed 100-mm plastic dish and measure the size of
the gland by placing the dish on a white paper with a ruler (Fig. 1A).
Usually a single submadibular gland (described here) would weigh between 7 and 15 g
depending on the age of the patient and if they have any underlying disease. Sublingual
glands weigh 3-5 g.
We have processed two submandibular (left and right) at once and two sublingial glands
(left and right) at once to speed up the tissue digestion process.
Su et al.
3of13
Current Protocols
Figure 1 (A) One human submandibular gland (huSMG). (B) The gland is wetted with small vol-
ume of digestion buffer and mechanically cut with scissors into small pieces. (C) The small tissue
pieces are further minced finely using two scissors simultaneously. (D) The tissue is made into
a fine slurry paste. (E) The entire slurry is transferred into a 50-ml conical tube with the diges-
tion buffer (see materials for Basic Protocol 1). (F) After shaking, the sample is transferred into
gentleMACS c-tubes and processed with the gentleMACS dissociator.
2. Weigh the sample in the dish on the same scale and subtract the weight of the dish to
obtain sample weight. In case of mouse tissues, place two submandibular or several
parotid glands obtained from different mice in one 100-mm dish and prepare as
described below for processing. Keep on ice.
3. Once sample details are obtained, begin tissue cleaning by transferring all the tissue
or the whole SG from the 100-mm dish into a 50-ml conical tube. Add PBS or HBSS
with antibiotic-antimycotic (and optionally heparin), and gently shake the tube up
and down manually (using your hands) for about 45 s to wash the whole gland.
Remove the wash buffer and add fresh PBS/HBSS to wash again.
4. Repeat step 3 at least three times or until the washing solution is clear of blood or
oating brous debris.
Tissue preparation
5. Using ne tweezers and scalpels, remove the brous capsule, connective, and fat
tissues along with any cauterized tissue ends from the SG surface, and proceed to
next step.
6. Place the tissues on a 100-mm dish and add 0.3-0.5 ml of digestion buffer (depend-
ing upon tissue size). Use sharp curved scissors to rst cut the tissues into small
pieces (about 5 mm in size; Fig. 1B). Then, further mince these tissue pieces us-
ing two curved scissors to make into a ne slurry paste (residual tissue pieces are
approximately 1 mm or less; Fig. 1C and D).
Since the salivary gland tissues are soft, it is challenging to mince bigger tissue pieces.
Cutting the tissues rst into small pieces (5 mm in size) will improve efciency during
the mincing step, which is done using two sharp curved scissors simultaneously to facil-
itate tissue cutting. One scissor can be used if the operator is not comfortable with using
both hands, but it will take more time to obtain the ne slurry paste.
Su et al.
4of13
Current Protocols
7. Transfer this slurry using a 1000-μl micropipette tip (cut the pipette tip at 1cm
from the tip using a sterile scissor to widen the opening) into a 50-ml conical tube
and add the digestion buffer.
Volume of digestion buffer depends on the weight of the sample, roughly, 35 ml of diges-
tion buffer for 2-3 g of SG tissue (Fig. 1D and E).
8. Mix the sample with the digestion buffer by vigorous shaking for 30 s using a vortex.
9. Transfer the mixture into gentleMACS tubes (15 ml per tube) and use the appro-
priate program on the GentleMACS dissociator (C2, D2; 45-60 s) to vigorously mix
the slurry with the enzymes (Fig. 1F).
10. Transfer the sample back from the gentleMACS tubes to a 50-ml tube, or let it remain
in the same gentleMACS tube (depending on if you wish to repeat step 5 for further
digestion) on a plate shaker inside the cell culture incubator with 5% CO2at 37°C
for 1-3 hr.
Incubation time depends upon the size of tissue—more incubation time leads to better
digestion, but decreases the viability of cells.
11. Every 20-30 min, remove the tube from the incubator and shake it manually for
about 20 s (i.e., hold the tube in one hand and pivot the tube upside down and right
side up for 15-20 times).
This action will disperse the tissue pieces and avoid clumping. Also, this manual shaking
does not add bubbles to the solution.
12. Check the tube during and at end of the incubation time for any remaining undigested
tissue pieces. If the solution appears cloudy and completely homogenous (free of
tiny visible tissue pieces), then proceed to the ltration step.
13. Pass the digested tissue suspension through a 70-μm cell strainer and transfer the
owthrough to a new 50-ml conical tube. Centrifuged 5 min at 300 ×g,4°C.
14. Remove the supernatant carefully and leave about 5 ml of remaining supernatant in
the tube (to avoid sucking off any cells close to the pellet).
15. Add 10 ml of sterile 0.9% NaCl solution to the 50-ml tube and gently mix using
a 1000-μl pipette tip to wash the cells. Centrifuge 45 s at 60 ×gand remove the
supernatant. Repeat the washing and centrifugation steps with 10 ml of 0.9% NaCl
until the supernatant is clear and devoid of the pink color from the enzyme solution
as well as debris.
These steps will dilute and remove as much as possible the digestion enzymes (pink in
color) remaining in the 5 ml solution at the end of step 4. In addition, low centrifugation
speeds allow cells to pellet, while the cellular debris, which are lighter, remain in solution
and thus can be eliminated from the culture).
16. Resuspend the cell pellet in 1-2 ml of Epi Max serum-free medium (depending on
cell number), count the cell number (Phelan & May, 2015) , seed at a density of
1-2 ×106cells per 100-mm dish, and incubate at 37°C with 5% CO2.
Maintenance of the single-cell culture
Epi Max is a serum-free medium that comes with three supplements (contents are propri-
etary ) to which we only add 1% (v/v) antibiotic-antimycotic solution for all our cultures.
After passaging salivary gland primary cells, sometimes, if they fail to attach (from P0
to P1), we add 5% fetal bovine serum (see Basic Protocol 2 materials list for cat. no.)
to the Epi Max culture medium to promote cell attachment. After 48 hr, once cells are
attached, we replace with ‘serum free’ Epi Max medium.
Su et al.
5of13
Current Protocols
Figure 2 Phase-contrast images showing the morphology of salivary gland cells cultured using
Basic Protocol 1. (A) Epithelial cells on Day 7 at passage 0. (B) Epithelial cells on day 10 after
passage 1. Scale bar: 43 μm.
17. At day 0, when cells are seeded, add 4-5 ml culture medium (Epi Max) to the plate
and leave undisturbed for about 48 hr. After 2 days, add an additional 2-3 ml of
medium to the plates and leave undisturbed for the next 2 days.
18. At day 4-6, small, isolated cell colonies start to appear on the culture plate (Fig. 2).
On this day, collect all the medium from the plate and transfer into a 15-ml conical
tube. Next, wash the culture plate gently using a pipette with Ca-, Mg-, and phenol
red–free HBSS or PBS solution to remove any debris, and add 5-6 ml of fresh culture
medium. Replace with fresh medium every 3-4 days.
19. Centrifuge the medium collected in the 15-ml tubes from the plates for 5 min at 300
×g, 4°C. Remove most of the supernatant and keep 1 ml in the bottom of the tube
to avoid sucking up the cell pellet. Then, add 5-10 ml of 0.9% NaCl solution and
centrifuge at 45 s at 60 ×g, 4°C, to remove dead cells, debris, and lysed RBCs.
20. Resuspend the remaining cell pellet with 5-6 ml of fresh culture medium (for 1–2 ×
106cells) using a 1000-μl micropipette, and gently mix the cells before plating into
new 100-mm dishes. Repeat steps 17 and 18 for these re-plated cells (oating cells
in the parent plate at passage 0).
21. At day 4-6 for the new plates (which contain oating cells from the parent plates),
new cell colonies start appearing. On this day, discard all the medium with any re-
maining oating cells and debris, and replace with 5-6 ml fresh culture medium.
Subsequently, replace with fresh medium every 3-4 days.
The time taken for the entire process starting from tissue cleaning to cell seeding depends
upon the size and number of glands. For reference, to process an entire left and right
human submandibular gland, it takes 5-6 hr.
For cell passaging, see Basic Protocol 2, steps 14-22
BASIC
PROTOCOL 2
PREPARATION OF SG EXPLANT CULTURE FROM FRESHLY OBTAINED
HUMAN OR MOUSE SG TISSUES
In this protocol, we provide an alternative method to isolate primary cells from fresh
human or mouse samples by cutting the SG tissues into small pieces (1-2 mm) and
growing them in tissue culture plates for downstream applications. We also detail the
steps to successfully passage these isolated cells using trypsin-EDTA.
Materials
Mice (6- 8-week-old; e.g., C57BL/6)
70% ethanol in spray bottle
Su et al.
6of13
Current Protocols
Phosphate-buffered saline (PBS), pH 7.4; or calcium-, magnesium-, and phenol
red–free Hank’s Balanced Salt Solution (HBSS); Gibco, cat. no. 14175-095)
with 2% antibiotic-antimycotic (Gibco, cat. no. 15240-062)
Complete medium: Epi Max (Wisent, cat. no. 002-010-CL) with 1%
antibiotic/antimycotic
Trypsin-EDTA (1×), 0.25% (Gibco, cat. no. 15050-057)
Neutralizing medium: DMEM with 10% FBS (Gibco, cat. no. 26140-079) and 1%
antibiotic-antimycotic
Surgical instruments: scalpels, dissecting forceps, scissors
Tissue culture plates (100 mm, 60 mm, 6-well, 12-well; depending on your
application/expected number of cells)
Humidied, 37°C, 5% CO2cell culture incubator
Refrigerated centrifuge
96-well culture plates (optional)
Additional reagents and equipment for euthanasia of rodents (see Current Protocols
article: Donovan & Brown, 2006)
NOTE: All procedures involving live animals must be approved by the local animal care
and use committee and follow governmental and institutional regulations.
Isolation of mice salivary glands
1. Sacrice 6–8-week-old mice (e.g., C57BL/6).
Procedures for euthanasia of mice are described in Donovan & Brown (2006).
There is no specic method of mouse sacrice that is recommended. For isolation
of mouse salivary gland cells, it is best to sedate the mouse, then harvest the sali-
vary gland tissue and follow with CO2euthanasia. Details are described by McGill’s
animal SOP (https://www.mcgill.ca/research/les/research/301-_rodent_euthanasia_-
_july_2021_0.pdf ).
Number of mice to be sacriced depends on how many cells you need and the skill of the
researchers. For practice, 1-2 mice would be recommended to isolate the cells. Going
forward, researchers can sacrice mice based on their demands.
2. Spray 70% ethanol on to the head and neck regions of the mice to sterilize the skin
surface.
3. Use sterilized scalpels, dissecting forceps, and scissors to harvest the submandibular
and sublingual glands from mice.
4. Place the glands on a 100-mm dish and cover entirely with PBS (room temperature).
Human SG explant culture method
5. Follow the tissue cleaning (steps 1-4) and preparation steps (step 5) from Basic Pro-
tocol 1.
6. Mince the salivary gland tissues (human or mice) into 1-2 mm (diameter) pieces
with a sharp scalpel (Fig. 3A).
Always keep all the tissue wet with culture medium.
7. Prepare a 100-mm dish with 2 ml complete medium (Epi Max), and rinse the dish
until medium wets the entire surface of the dish.
8. Disperse the small pieces of sample into the pre-wet 100-mm dish (Fig. 3A).
Do not place the pieces at the edge of the dish. Keep enough distance between the tissue
pieces to provide enough area for the cells to expand.
9. Incubate the dish in the incubator overnight at 37°C with 5% CO2.Su et al.
7of13
Current Protocols
Figure 3 Explant culture of human salivary gland cells. (A) Small pieces (1-2 mm) of explant
tissue placed on a 100-mm dish. (B) Piece of minor labial gland attached to culture dish for 14
days; primary salivary cells have migrated to the periphery of the explant culture. (C) Example of
an explant culture from a human submandibular gland (day 18) and (D) of a human parotid gland
(day 21). Scale bar: 90 μm.
Figure 4 Primary SG cell growth kinetics measured using WST-8 cell metabolic assay. Primary
huSMG cells were seeded at a concentration of 2000 cells/well in a 96-well plate at day 0. Samples
were taken at days 0, 1, 3, 5, and 7, and absorbance was measured at 460 nm using a microplate
reader.Primary cells need 24 hr to attach to the culture plates.Once attached, there is a log phase
where the cell metabolism increases, which continues until day 3, suggesting cell proliferation.
After day 3, cells start showing a decrease in metabolic activity, which could be associated with
cell crowding and overlapping. (Standard curve is shown on the left).
10. For the rst 48 hr, keep the culture medium at 2-3 ml per 100-mm dish.
Medium should not cover the whole tissue; otherwise the tissue will fail to attach and
oat. However, if there is an insufcient volume of medium, the tissue pieces will dry and
become necrotic.
11. If needed, add an additional 1-2 ml medium during these 48-72 hr to keep the nal
volume around 3-5 ml in the 100-mm dish.
Su et al.
8of13
Current Protocols
Figure 5 (A) E-cadherin, salivary epithelial cell surface marker. (B) Aquaporin-5 (AQP-5), wa-
ter channel protein found in salivary acinar cell membrane. (C) Pan-cytokeratins (wide spectrum
cytokeratin) detect a wide range of cytokeratin filament proteins found in salivary gland epithelial,
and myoepithelial cells. (D) Zona occludens-1 (ZO-1), tight junction–associated protein found in
salivary cell membrane. Scale bar: 70 μm.
12. Change the medium every 2-3 days. Keep a constant check on the medium during
the rst 3-7 days to prevent the tissues from getting dry. When required, add fresh
complete medium gently from the side of the dish to prevent any tissue dislodge-
ment.
13. After 5-7 days, cells begin to grow out from the tissue piece (Fig. 3). During the
rst week, do not move the dish too frequently. When changing and adding culture
medium, move the dish gently. When cells begin to grow from most of the tissue,
change medium every 3 days and replace with 5 ml complete Epi Max medium.
Cell passaging (for Basic Protocols 1 and 2)
14. Timing for cell passage. Passage cells when one of these situations is observed:
a. Several large colonies/islands of cells in the culture plate Su et al.
9of13
Current Protocols
b. Two cell colonies merge to form larger colonies, or
c. Some of the cells begin to die and oat due to cell crowding within an individual
large cell colony/island, which usually takes around 3 weeks.
15. For the explant culture method (Basic Protocol 2), transfer the tissue pieces to a
new pre-wet dish using tweezers. Cells can continue to grow and migrate from these
pieces of tissues again by following the Basic Protocol 2, steps 1-9.
While re-plating these tissue pieces into new dishes, care should be taken with the tips of
the tweezer to avoid damaging the cells released around the tissue explant.
16. Aspirate culture medium in the cell culture dish.
From here, the steps for passaging cells are the same for both Basic Protocols 1 and 2.
17. Wash the dish with 5 ml PBS or HBSS. Aspirate the PBS/HBSS.
18. Add 3 ml 0.25% trypsin-EDTA (cell-detaching solution) to the 100-mm dish. Place
culture dish in a 37°C incubator for 3-5 min. Gently tap the sides of the dishes to
mechanically detach cells.
19. Add 3 ml of neutralizing solution (DMEM with 5%-10% FBS and 1% antibiotic-
antimycotic) to inhibit the trypsin activity. Detach cells by ushing the trypsin +
DMEM (6 ml) with a pipette.
Due to the adherent nature of salivary epithelial cells, the rst trypsinization might only
remove portions of the cell colonies within the 100-mm dish. When this happens, repeat
steps 5 and 6 again to detach the remaining adherent cells.
20. Collect all the detached cells (cells +trypsin +DMEM) into a 15- or 50-ml conical
tube. Centrifuge 5 min at 300 ×g,4°C.
21. Remove the supernatant and resuspend the cell pellet in the conical tube with com-
plete medium (Epi Max with 1% antibiotic-antimycotic). Gently pipette up and
down to obtain a single-cell suspension.
22. Transfer cell solution into two new 100-mm dishes (a 1:2 split ratio).
23. Optional: Cells can also be seeded at a concentration of 2000 cells/well in a 96-well
plate to measure cell proliferation (samples taken at 0, 1, 3, 5, and 7 days; Fig. 4)
or subjected to immunocytochemistry to look at the development of cell markers
(Fig. 5).
COMMENTARY
Background Information
Permanent loss of saliva-secreting acinar
cells due to radiation, Sjögren’s syndrome, or
aging, resulting in hyposalivation and xerosto-
mia, has a substantial negative impact on the
oral health and quality of life of patients. Cur-
rent treatment strategies using lifelong substi-
tutes or drugs to alleviate dry mouth symp-
toms are temporary approaches. An important
step in the development pipeline of treatments
aiming at SG regeneration is designing func-
tional ex vivo human models to study the ef-
fects of drugs, as well as cell and gene thera-
pies, on SGs. Cultured primary SG cells play
a crucial role in developing various in vitro
and ex vivo disease models, as these cells are
able to retain their native SG cell morphology
and thereby the localization of key proteins.
We have previously demonstrated the abil-
ity to culture polarized human primary sub-
mandibular cells with characteristics such as
proper localization of tight junction proteins,
ion channels, and co-transporters for para-
cellular uid movements (Tran et al., 2005).
Using analogous cell culture approaches, we
have developed additional in vitro and ex vivo
SG models over the years for SG researchers
(Maria, Liu, El-Hakim, Zeitouni, & Tran,
2017; Maria & Tran, 2011; Maria, Zeitouni,
Gologan, & Tran, 2011; Seo et al., 2019; Su
et al., 2020). Also during the past decade, we
have tested several culture conditions includ-
ing the use of serum-containing, serum-free,
and dened media to nd an optimal growth
condition for culturing salivary epithelial
cells (Charbonneau, Kinsella, & Tran, 2019;
Su et al.
10 of 13
Current Protocols
Charbonneau & Tran, 2020; Zhang, Pham,
Munguia-Lopez, Kinsella, & Tran, 2020). In
this protocol, we have described a simple and
reproducible method to obtain epithelial cells
from human SG tissues for different down-
stream applications. Our described protocols
have been previously used to culture monkey
salivary epithelial cells as well (Tran et al.,
2006).
Critical Parameters
Using this protocol, we have been able to
successfully isolate, culture, and expand dif-
ferent salivary gland cell types in vitro from
patient samples. Time is very crucial for an
optimal yield of cells after single-cell diges-
tion. Samples should be processed as soon as
they arrive at the lab from the hospital. All tis-
sue cleaning and cutting procedures, until in-
cubation, should be done on ice. Longer pro-
cessing and digestion time leads to lower cell
viability. However, the time needed for each
gland usually depends on the size and qual-
ity of the tissue obtained. Larger salivary tis-
sue with highly brous connective tissue takes
more time to prepare (to make the tissue free
of fat and connective tissues) and digest (more
tissue, longer incubation period). The entire
process must be optimized on the day of re-
ceipt of the glands, and the reagents and arma-
mentarium should be made ready a day before
to avoid delays on the day of processing.
Troubleshooting
Removing the fat and connective tissue is
crucial before cutting the tissues, as their im-
proper removal might necessitate longer incu-
bation time for cell digestion. After cell di-
gestion, ltration with a 70-μm strainer might
pose a challenge, as the strainers tend to clog
after 2-4 ml of the cell solution is passed.
In such situations, use 0.9% NaCl solution to
wash down some of the clogged cells by force
of liquid ow using a pipette or a 10-ml sy-
ringe. If the cell solution still fails to pass
through, change the lter immediately and use
a new lter. After ltration, make sure to wash
the cell pellet multiple times with 0.9% NaCl
to remove as much debris as possible. This will
lead to cleaner cultures when cells are seeded.
When seeding, cells tend to be in clumps. Gen-
tly pipetting the solution up and down will dis-
perse these cell clumps and result in a more
uniform distribution of cells throughout the
culture dish.
Understanding Results
The total yield of cells from Basic Proto-
cols 1 and 2 depends on the quality of tissue.
Glands harvested from older patients have less
lobular gland structure and more brotic tis-
sues, resulting in poor yield. As a reference,
a whole submandibular gland from a 37-year-
old patient can yield up to 25-30 ×106cells in
single-cell culture after processing. For larger
tissue samples, Basic Protocol 1 is preferred,
to make use of the entire gland. Alternatively,
the explant culture method (Basic Protocol 2)
could be used in conjunction with single-cell
digestion to increase total yields. Yields from
single cells are also dependent on how oat-
ing cells attach and expand after replating into
new dishes (see Basic Protocol 1, steps 17-
21). These oating cells, if carefully isolated
and replated in a timely fashion, can maxi-
mize the total number of cells obtained from
the single-cell digestion protocol. These cells
form colonies similar to the parent plate and
retain all the morphological characteristics of
primary SG epithelial cells. In addition, these
replated cell cultures are cleaner due to ab-
sence of any lysed RBCs or cell debris, which
are commonly seen in the parent plates after
initial seeding. For the explant culture method,
when cells are ready to be passaged, the small
pieces of tissue can be removed from the orig-
inal dish with a tweezer and transferred into
a new 100-mm dish pre-wet with medium. We
have successfully continued to grow cells from
these pieces of tissue up to three times.
Time Considerations
Preparation of SG single-cell culture (Basic
Protocol 1) usually takes about 5-6 hr depend-
ing on the size of the tissue sample. The ex-
plant culture (Basic Protocol 2) usually takes 1
hr for tissue washing and cleaning and around
1 hr to plate the small tissue pieces into 100-
mm dishes. For both single-cell and explant
culture (Basic Protocols 1 and 2), it takes
around 17-21 days for the cells to be ready.
The total duration of the primary cell culture
may be about 6-8 weeks, depending on the
number of passages the cells can survive (usu-
ally 2-3 passages).
Acknowledgments
This work was supported in part by
the Canadian Institutes of Health Research
(CIHR). We thank all the anonymous patients
for their salivary gland tissues that were used
in this work. We thank all current and past
members of our lab at McGill University who
contributed toward the development of this
protocol over the years. We also thank all
our collaborating surgeons from McGill Uni-
versity Health Center and Montreal General Su et al.
11 of 13
Current Protocols
Hospital for providing us the human salivary
gland samples.
Author Contributions
Xinyun Su: conceptualization, data cura-
tion, formal analysis, investigation, methodol-
ogy, project administration, validation, visual-
ization, writing original draft, writing review
and editing; Sangeeth Pillai: data curation,
formal analysis, investigation, methodology,
project administration, validation, visualiza-
tion, writing original draft, writing review and
editing; Younan Liu: data curation, formal
analysis, investigation, methodology, project
administration, resources, validation, visual-
ization, writing review and editing; Simon
D. Tran: conceptualization, data curation,
formal analysis, funding acquisition, investi-
gation, methodology, project administration,
resources, supervision, validation, visualiza-
tion, writing original draft, writing review and
editing.
Conict of Interest
The authors have no conicts to declare.
Data Availability Statement
Data openly available in a public repository
that issues datasets with DOIs.
Literature Cited
Agnihotri, R., Gaur, S., & Albin, S. (2020).
Nanometals in dentistry: Applications
and toxicological implications-a system-
atic review [Review]. Biological Trace
Element Research,197(1), 70–88. doi:
10.1007/s12011-019- 01986-y
Beucler, M. J., & Miller, W. E. (2019). Isolation
of salivary epithelial cells from human sali-
vary glands for in vitro growth as salispheres or
monolayers. Journal of Visualized Experiments:
JoVE, 149. doi: 10.3791/59868
Charbonneau, A. M., Kinsella, J. M., & Tran, S. D.
(2019). 3D cultures of salivary gland cells in na-
tive or gelled egg yolk plasma, combined with
egg white and 3D-printing of gelled egg yolk
plasma. Materials,12(21), 3480. doi: 10.3390/
ma12213480
Charbonneau, A. M., & Tran, S. D. (2020). 3D cell
culture of human salivary glands using nature-
inspired functional biomaterials: The egg yolk
plasma and egg white. Materials,13(21), 4807.
doi: 10.3390/ma13214807
de Paula, F., Teshima, T. H. N., Hsieh, R., Souza,
M. M., Nico, M. M. S., & Lourenco, S. V.
(2017). Overview of human salivary glands:
Highlights of morphology and developing pro-
cesses. Anatomical Record (Hoboken),300(7),
1180–1188. doi: 10.1002/ar.23569
Donovan, J., & Brown, P. (2006). Euthanasia. Cur-
rent Protocols in Immunology,73, 1.8.1–1.8.4.
doi: 10.1002/0471142735.im0108s73
Hand, A. R., Pathmanathan, D., & Field, R.
B. (1999). Morphological features of the mi-
nor salivary glands. Archives of Oral Biol-
ogy,44(Suppl 1), S3–S10. doi: 10.1016/S0003-
9969(99)90002-X
Lin, L. C., Elkashty, O., Ramamoorthi, M., Trinh,
N., Liu, Y., Sunavala-Dossabhoy, G., Tran,
S. D. (2018). Cross-contamination of the human
salivary gland HSG cell line with HeLa cells:
A STR analysis study. Oral Diseases,24(8),
1477–1483. doi: 10.1111/odi.12920
Maimets, M., Rocchi, C., Bron, R., Pringle, S.,
Kuipers, J., Giepmans, B. N., Coppes, R. P.
(2016). Long-term in vitro expansion of salivary
gland stem cells driven by wnt signals. Stem Cell
Reports,6(1), 150–162. doi: 10.1016/j.stemcr.
2015.11.009
Maria, O. M., Liu, Y., El-Hakim, M., Zeitouni,
A., & Tran, S. D. (2017). The role of hu-
man bronectin-or placenta basement mem-
brane extract-based gels in favouring the forma-
tion of polarized salivary acinar-like structures.
Journal of Tissue Engineering and Regenerative
Medicine,11(9), 2643–2657. doi: 10.1002/term.
2164
Maria, O. M., & Tran, S. D. (2011). Human mes-
enchymal stem cells cultured with salivary gland
biopsies adopt an epithelial phenotype. Stem
Cells and Development,20(6), 959–967. doi:
10.1089/scd.2010.0214
Maria, O. M., Zeitouni, A., Gologan, O., & Tran, S.
D. (2011). Matrigel improves functional proper-
ties of primary human salivary gland cells. Tis-
sue Engineering Part A,17(9–10), 1229–1238.
doi: 10.1089/ten.tea.2010.0297
Pedersen, A. M., Bardow, A., Jensen, S. B., &
Nauntofte, B. (2002). Saliva and gastrointestinal
functions of taste, mastication, swallowing and
digestion. Oral Diseases,8(3), 117–129. doi:
10.1034/j.1601-0825.2002.02851.x
Phelan, K., & May, K. M. (2015). Basic techniques
in mammalian cell tissue culture. Current Pro-
tocols in Cell Biology,66, 1.1.1–1.1.22. doi:
10.1002/0471143030.cb0101s66
Pringle, S., Nanduri, L. S., van der Zwaag, M., van
Os, R., & Coppes, R. P. (2011). Isolation of
mouse salivary gland stem cells. JoVE,48, doi:
10.3791/2484
Riva, A., Puxeddu, R., Uras, L., Loy, F., Serreli,
S., & Testa Riva, F. (2000). A high resolution
SEM study of human minor salivary glands. Eu-
ropean Journal of Morphology,38(4), 219–226.
doi: 10.1076/0924-3860(200010)38:4;1- O;FT
219
Seo, Y. J., Lilliu, M. A., Abu Elghanam, G.,
Nguyen, T. T., Liu, Y., Lee, J. C., Tran, S.
D. (2019). Cell culture of differentiated human
salivary epithelial cells in a serum-free and scal-
able suspension system: The salivary functional
units model. Journal of Tissue Engineering and
Regenerative Medicine,13(9), 1559–1570. doi:
10.1002/term.2908
Shirasuna, K., Sato, M., & Miyazaki, T.
(1981). A neoplastic epithelial duct cell
line established from an irradiated human
Su et al.
12 of 13
Current Protocols
salivary gland. Cancer,48(3), 745–752. doi:
10.1002/1097-0142(19810801)48:3<745::Aid-
cncr2820480314>3.0.co;2-7.
Su, X., Fang, D., Liu, Y., Ramamoorthi, M.,
Zeitouni, A., Chen, W., & Tran, S. (2016).
Three-dimensional organotypic culture of hu-
man salivary glands: The slice culture model.
Oral Diseases,22(7), 639–648. doi: 10.1111/
odi.12508
Su, X., Liu, Y., Bakkar, M., ElKashty, O., El-
Hakim, M., Seuntjens, J., & Tran, S. (2020).
Labial stem cell extract mitigates injury to
irradiated salivary glands. Journal of Den-
tal Research,99(3), 293–301. doi: 10.1177/
0022034519898138
Tran, S. D., Sugito, T., Dipasquale, G., Cotrim, A.
P., Bandyopadhyay, B. C., Riddle, K., Baum,
B. J. (2006). Re-engineering primary epithelial
cells from rhesus monkey parotid glands for use
in developing an articial salivary gland. Tissue
Engineering,12(10), 2939–2948. doi: 10.1089/
ten.2006.12.2939
Tran, S. D., Wang, J., Bandyopadhyay, B. C., Red-
man, R. S., Dutra, A., Pak, E., Baum, B. J.
(2005). Primary culture of polarized human sali-
vary epithelial cells for use in developing an ar-
ticial salivary gland. Tissue Engineering,11(1-
2), 172–181. doi: 10.1089/ten.2005.11.172
Varghese, J. J., Hansen, M. E., Sharipol, A., Ingalls,
M. H., Ormanoski, M. A., Newlands, S. D.,
Benoit, D. S. W. (2019). Salivary gland cell ag-
gregates are derived from self-organization of
acinar lineage cells. Archives of Oral Biology,
97, 122–130. doi: 10.1016/j.archoralbio.2018.
10.017
Vissink, A., Burlage, F. R., Spijkervet, F. K.,
Jansma, J., & Coppes, R. P. (2003). Prevention
and treatment of the consequences of head and
neck radiotherapy. Critical Reviews in Oral Bi-
ology & Medicine,14(3), 213–225.
Zhang, Y., Pham, H. M., Munguia-Lopez, J.
G., Kinsella, J. M., & Tran, S. D. (2020).
The optimization of a novel hydrogel—
Egg white-alginate for 2.5 D tissue engi-
neering of salivary spheroid-like structure.
Molecules (Basel, Switzerland),25(23),
5751. doi: 10.3390/molecules252357
51
Su et al.
13 of 13
Current Protocols
... Human primary SG cells can be obtained by either dissociating the major SG tissues using digestive enzymes or by culturing them as explant tissues. We recently published a detailed protocol for the isolation, culture, and characterization of primary SG cells using these methods from human and mice tissues (Su et al., 2022a). In addition to the three major cell types, adult human SGs also comprise of progenitor/stem cells that help maintain cell turnover and homeostasis in response to any injury. ...
... Our lab routinely uses the EPIMAX serum-free culture medium to culture primary human and mice SG epithelial cells (Su et al., 2022a). We have noted that supplementing the EPIMAX with 10% FBS significantly increases the proliferation potential in SG cell lines. ...
... While primary cells maintain a characteristic cobblestone phenotype with this medium, due to their sensitive nature, it is seen to be beneficial to supplement the medium with 5-10% FBS when passaging them. Especially, in SG single cell culture, this approach promotes the initial attachment of the cell to tissue culture plastic, and the medium can then be replaced with serum-free conditions to allow their proliferation (Su et al., 2022a). ...
Article
Full-text available
Salivary glands are specialized structures developed as an extensively compact, arborized design through classical embryogenesis, accompanied by a cascade of events channelized by numerous growth factors and genetic regulatory pathways. Salivary secretions maintain oral homeostasis and, when diminished in certain conditions, present as xerostomia or salivary hypofunction, adversely impacting the patient's quality of life. The current available treatments primarily aim at tackling the immediate symptoms providing temporary relief to the patient. Despite scientific efforts to develop permanent and effective solutions to restore salivation, a significant permanent treatment is yet to be established. Tissue engineering has proven as a promising remedial tool in several diseases, as well as in xerostomia, and aims to restore partial loss of organ function. Recapitulating the physiological cellular microenvironment to in vitro culture conditions is constantly evolving. Replicating the dynamic multicellular interactions, genetic pathways, and cytomorphogenic forces, as displayed during salivary gland development have experienced considerable barriers. Through this review, we endeavour to provide an outlook on the evolution of in vitro salivary gland research, highlighting the key bioengineering advances and the challenges faced with the current therapeutic strategies for salivary hypofunction, with an insight into our team's scientific contributions.
... 52 We recently published a comprehensive protocol for isolating and culturing single cells from human and mice SG tissues under serum-free conditions. 29 Acinar or ductal SG epithelial cells can be obtained either from small tissue explants (∼1−2 mm in size) or through digestion using Liberase enzymes and a gentleMACS dissociator, rendering them suitable for downstream applications. 29 Nonetheless, maintaining primary SG cells in a single-cell state is commonly quite challenging due to issues such as slow cell proliferation, a high rate of dedifferentiation, and alterations associated with epithelial-to-mesenchymal transitions (EMT) in glandular organs. ...
... 29 Acinar or ductal SG epithelial cells can be obtained either from small tissue explants (∼1−2 mm in size) or through digestion using Liberase enzymes and a gentleMACS dissociator, rendering them suitable for downstream applications. 29 Nonetheless, maintaining primary SG cells in a single-cell state is commonly quite challenging due to issues such as slow cell proliferation, a high rate of dedifferentiation, and alterations associated with epithelial-to-mesenchymal transitions (EMT) in glandular organs. 53−55 This has led to the development of alternative culture practices to maintain both progenitor and differentiated epithelial populations isolated from adult SG tissues. ...
... All cells were grown in the proper condition descibed in the Supplemental Material. For human salivary gland primary cells, the published protocol (Su et al. 2022b) was used with a minor modification. The biopsy tissues were obtained from patients at the Center for Orphaned Autoimmune Disorders at the University of Florida College of Dentistry. ...
Preprint
Full-text available
Viral and endogenous double-stranded RNAs (dsRNAs) are recognized by cytosolic sensors and initiate antiviral signaling as a part of innate responses. Recent studies reveal a close association between mitochondrial dsRNAs (mt-dsRNAs) and several immune-related diseases. However, the mechanism by which mt-dsRNAs stimulate immune responses remains poorly understood. Here, we discover SRA stem-loop interacting RNA binding protein (SLIRP) as a key amplifier of mt-dsRNA-triggered antiviral signals in the setting of an autoimmune condition and viral infection. We find that the activation of melanoma differentiation-associated gene 5 (MDA5) by dsRNAs upregulates SLIRP expression and facilitates its mitochondrial import. SLIRP then stabilizes mt-dsRNAs and promotes their cytosolic release where they further activate MDA5 to amplify the antiviral signaling, augmenting the interferon response. Interestingly, downregulation of cytosolic mt-dsRNAs by targeted knockdown of SLIRP or blocking mt-dsRNA release from mitochondria dampens the interferon response and increases cell vulnerability to certain viral infections. Furthermore, we find elevated SLIRP expression in monocytes of autoimmune patients, and downregulation of SLIRP partially rescues the abnormal interferon-stimulated gene expression in patients' primary cells. Our study unveils amplified positive feedback of antiviral signaling through the stabilization of mt-dsRNAs by SLIRP, which promotes a robust interferon response.
... This approach exhibited a therapeutic effect comparable to MSCs in NOD mice (Abughanam et al., 2019). Another study showed that this strategy can be used to treat irradiation-injured SGs, as cell extracts of adipose-and BM-derived MSCs or salivary gland stem cell containing numerous angiogenesis-related growth factors restored the function of radiogenic SGs (Fang et al., 2018;Su et al., 2020;Su, Pillai, et al., 2022). Additionally, our preliminary data demonstrated that cell extracts do not accelerate tumor growth. ...
Article
Full-text available
Objective: For functional restoration of salivary glands (SGs) injured by radiation therapy or Sjögren's syndrome (SS), various experimental approaches, such as gene therapy, tissue engineering, and cell-based therapy, have been proposed. This narrative review summarized recent progresses in research using cell-based therapies, including promising trials that could lead to bench-to-clinic applications. Methods: A literature review based on PubMed publications in the last two decades was performed to summarize progresses in cell-based therapies for SG dysfunction. Results: Over 100 experimental studies have shown the therapeutic potential of several types of cells, such as SG stem cells and mesenchymal stem cells, as well as effectively conditioned mononuclear cells, in both radiation injury and SS animal models. These therapies affect to slow fibrosis progression and stimulate tissue regeneration in atrophic glands. However, to date, only a total of seven studies have been developed to the stage of clinical study, showing the safety and preliminary efficacy. Conclusion: To lead the radical effectiveness expected in cell-based therapy, advances in reverse translational research and in innovative experimental research, based on the findings of recent clinical studies, will be critical in the next decade.
... In general, an ideal biomaterial used in SG tissue engineering should: (1) support the cell proliferation and migration within the matrix, (2) maintain the phenotypic characteristics of SG cells, (3) stimulate selective differentiation of SG stem/progenitor cells, (4) enable cell assembly and reorganization (cell polarization and lumen formation), (5) support matrix remodeling (permissible hydrogels), and (6) allow duct expansion (promote branching morphogenesis) ( Figure 2). For bioengineering an artificial SG, the biomaterial strategy depends on the type of cells used such as cell lines [22], primary single cells [11,23], cell clusters such as salivary functional units [14], acinar cell clusters and intercalated ducts [15], the tissue derivation (adult or embryonic-epithelium or mesenchyme) [24], and the expected functions of the model. The three main types of SG cells include the acinar, ductal, and myoepithelial cells and, so far, there is no fully characterized description of an ideal biomaterial that can foster all adult epithelial cells simultaneously to fulfil their function or to promote the selective differentiation of available stem/progenitor cells and their organization within a matrix. ...
Article
Full-text available
Mimicking the complex architecture of salivary glands (SGs) outside their native niche is challenging due their multicellular and highly branched organization. However, significant progress has been made to recapitulate the gland structure and function using several in vitro and ex vivo models. Hydrogels are polymers with the potential to retain a large volume of water inside their three-dimensional structure, thus simulating extracellular matrix properties that are essential for the cell and tissue integrity. Hydrogel-based culture of SG cells has seen a tremendous success in terms of developing platforms for cell expansion, building an artificial gland, and for use in transplantation to rescue loss of SG function. Both natural and synthetic hydrogels have been used widely in SG tissue engineering applications owing to their properties that support the proliferation, reorganization, and polarization of SG epithelial cells. While recent improvements in hydrogel properties are essential to establish more sophisticated models, the emphasis should still be made towards supporting factors such as mechanotransduction and associated signaling cues. In this concise review, we discuss considerations of an ideal hydrogel-based biomaterial for SG engineering and their associated signaling pathways. We also discuss the current advances made in natural and synthetic hydrogels for SG tissue engineering applications.
Chapter
An organotypic tissue culture model can maintain the cellular and molecular interactions, as well as the extracellular components of a tissue ex vivo. Thus, this 3D model biologically mimics in vivo conditions better than commonly used 2D culture in vitro models. Here, we provide a detailed workflow for generating live 3D organotypic tissue slices from patient-derived freshly resected salivary glandular tissues. We also cover the processing of these tissues for various downstream applications like live-dead viability/cytotoxicity assay, FFPE sectioning and immunostaining, and RNA and protein extraction with a focus on the salivary gland radiation injury model. These procedures can be applied extensively to various solid organs and used for disease modeling for cancer research, radiation biology, and regenerative medicine.
Article
Full-text available
The egg yolk plasma (EYP)-a translucent fraction of the egg yolk (EY) obtained by centrifugation-was tested as a developmentally encouraging, cost-effective, biomaterial for salivary gland (SG) tissue engineering. To find optimal incubating conditions for both the human NS-SV-AC SG acinar cell line and SG fibroblasts, cells were stained with Live/Dead®. The cellular contents of 96-well plates were analyzed by high content screening image analysis. Characteristically, the EYP biomaterial had lipid and protein content resembling the EY. On its own, the EYP was non-conducive to cell survival. EYP's pH of 6 mainly contributed to cell death. This was demonstrated by titrating EYP's pH with different concentrations of either commercial cell culture media, NaOH, or egg white (EW). These additives improved SG mesenchymal and epithelial cell survival. The best combinations were EYP diluted with (1) 70% commercial medium, (2) 0.02 M NaOH, or (3) 50% EW. Importantly, commercial medium-free growth was obtained with EYP + NaOH or EYP + EW. Furthermore, 3D cultures were obtained as a result of EW's gelatinous properties. Here, the isolation, characterization, and optimization of three EYP-based biomaterial combinations are shown; two were free of commercial medium or supplements and supported both SG cells' survival.
Article
Full-text available
Nanotechnology is a vital part of health care system, including the dentistry. This branch of technology has been incorporated into various fields of dentistry ranging from diagnosis to prevention and treatment. The latter involves application of numerous biomaterials that help in restoration of esthetic and functional dentition. Over the past decade, these materials were modified through the incorporation of metal nanoparticles (NP) like silver (Ag), gold (Au), titanium (Ti), zinc (Zn), copper (Cu), and zirconia (Zr). They enhanced antimicrobial, mechanical, and regenerative properties of these materials. However, lately, the toxicological implications of these nanometal particles have been realized. They were associated with cytotoxicity, genotoxicity altered inflammatory processes, and reticuloendothelial system toxicity. As dental biomaterials containing metal NPs remain functional in oral cavity over prolonged periods, it is important to know their toxicological effects in humans. With this background, the present systematic review is aimed to gain an insight into the plausible applications and toxic implications of nano-metal particles as related to dentistry.
Article
Full-text available
For salivary gland (SG) tissue engineering, we cultured acinar NS-SV-AC cell line or primary SG fibroblasts for 14 days in avian egg yolk plasma (EYP). Media or egg white (EW) supplemented the cultures as they grew in 3D-Cryo histology well inserts. In the second half of this manuscript, we measured EYP’s freeze-thaw gelation and freeze-thaw induced gelled EYP (GEYP), and designed and tested further GEYP tissue engineering applications. With a 3D-Cryo well insert, we tested GEYP as a structural support for 3D cell culture or as a bio-ink for 3D-Bioprinting fluorescent cells. In non-printed EYP + EW or GEYP + EW cultures, sagittal sections of the cultures showed cells remaining above the well’s base. Ki-67 expression was lacking for fibroblasts, contrasting NS-SV-AC’s constant expression. Rheological viscoelastic measurements of GEYP at 37 °C on seven different freezing periods showed constant increase from 0 in mean storage and loss moduli, to 320 Pa and 120 Pa, respectively, after 30 days. We successfully 3D-printed GEYP with controlled geometries. We manually extruded GEYP bio-ink with fluorescence cells into a 3D-Cryo well insert and showed cell positioning. The 3D-Cryo well inserts reveal information on cells in EYP and we demonstrated GEYP cell culture and 3D-printing applications.
Article
Full-text available
Objective: The objective of this study was to characterize the mechanism by which salivary gland cells (SGC) aggregate in vitro. Design: Timelapse microscopy was utilized to analyze the process of salivary gland aggregate formation using both primary murine and human salivary gland cells. The role of cell density, proliferation, extracellular calcium, and secretory acinar cells in aggregate formation was investigated. Finally, the ability of cells isolated from irradiated glands to form aggregates was also evaluated. Results: Salivary gland cell self-organization rather than proliferation was the predominant mechanism of aggregate formation in both primary mouse and human salivary gland cultures. Aggregation was found to require extracellular calcium while acinar lineage cells account for ∼80% of the total aggregate cell population. Finally, aggregation was not impaired by irradiation. Conclusions: The data reveal that aggregation occurs as a result of heterogeneous salivary gland cell self-organization rather than from stem cell proliferation and differentiation, contradicting previous dogma. These results suggest a re-evaluation of aggregate formation as a criterion defining salivary gland stem cells.
Article
Full-text available
Salivary glands are essential organs that produce and secrete saliva to the oral cavity. During gland morphogenesis, many developmental processes involve a series of coordinated movements and reciprocal interactions between the epithelium and mesenchyme that generate the ductal system and the secretory units. Recent studies have shown new findings about salivary gland development, particularly regarding lumen formation and expansion, with the involvement of apoptosis and cell polarization, respectively. Moreover, it has been observed that human minor salivary glands start forming earlier than previously published and that distinct apoptotic mediators can trigger duct lumen opening in humans. This review summarizes updated morphological and cellular features of human salivary glands and also explores new aspects of the human developmental process. This article is protected by copyright. All rights reserved.
Article
Hydrogels have been used for a variety of biomedical applications; in tissue engineering, they are commonly used as scaffolds to cultivate cells in a three-dimensional (3D) environment allowing the formation of organoids or cellular spheroids. Egg white-alginate (EWA) is a novel hydrogel which combines the advantages of both egg white and alginate; the egg white material provides extracellular matrix (ECM)-like proteins that can mimic the ECM microenvironment, while alginate can be tuned mechanically through its ionic crosslinking property to modify the scaffold’s porosity, strength, and stiffness. In this study, a frozen calcium chloride (CaCl2) disk technique to homogenously crosslink alginate and egg white hydrogel is presented for 2.5D culture of human salivary cells. Different EWA formulations were prepared and biologically evaluated as a spheroid-like structure platform. Although all five EWA hydrogels showed biocompatibility, the EWA with 1.5% alginate presented the highest cell viability, while EWA with 3% alginate promoted the formation of larger size salivary spheroid-like structures. Our EWA hydrogel has the potential to be an alternative 3D culture scaffold that can be used for studies on drug-screening, cell migration, or as an in vitro disease model. In addition, EWA can be used as a potential source for cell transplantation (i.e., using this platform as an ex vivo environment for cell expansion). The low cost of producing EWA is an added advantage.
Article
Stem cell–based therapies could provide a permanent treatment for salivary gland (SG) hypofunction caused by ionizing radiation (IR) injury. However, current challenges for SG stem cells to reach the clinic include surgical invasiveness, amount of tissue needed, cell delivery, and storage methods. The objective of this study was to develop a clinically less invasive method to isolate and expand human SG stem cells and then to obtain a cell-free extract to be used as a therapy for IR-injured SGs. Human labial glands were biopsied, and labial stem cells (LSCs) were expanded by explant culture. The LSC extract (LSCE) was obtained by releasing the cellular components after 3 freeze-thaw cycles and 17,000 g force centrifugation. LSCE was injected intravenously into mice that had their SGs injured with 13-Gy IR. Positive (non-IR) and negative (IR) control mice received injections of saline (vehicle control). Three pieces of labial glands (0.1 g weight) could expand 1 to 2 million cells. LSCs had a doubling time of 18.8 h; could differentiate into osteocytes, adipocytes, and chondrocytes; and were positive for mesenchymal stem cell markers. Both angiogenic (FGF-1, FGF-2, KGF, angiopoietin, uPA, VEGF) and antiangiogenic factors (PAI-1, TIMP-1, TSP-1, CD26) were detected in LSCE. In addition, some angiogenic factors (PEDF, PTX3, VEGF) possessed neurotrophic functions. Mice treated with LSCE had 50% to 60% higher salivary flow rate than saline-treated mice at 8 and 12 wk post-IR. Saliva lag time measurements also confirmed that LSCE restored SG function. Histologic analyses of parotids and submandibular glands reported comparable numbers of acinar cells, blood vessels, and parasympathetic nerves and cell proliferation rates in sham IR and LSCE-treated mice, though significantly lower in saline-treated mice. An explant culture method can harvest a large number of LSCs from small pieces of labial glands. LSCE showed clinical potential to mitigate IR-injured SGs.
Article
The salivary glands are a site of significant interest for researchers interested in multiple aspects of human disease. One goal of researchers is to restore function of glands damaged by radiation therapies or due to pathologies associated with Sjögren's syndrome. A second goal of researchers is to define the mechanisms by which viruses replicate within glandular tissue where they can then gain access to salivary fluids important for horizontal transmission. These goals highlight the need for a robust and accessible in vitro salivary gland model that can be utilized by researchers interested in the above mentioned as well as related research areas. Here we discuss a simple protocol to isolate epithelial cells from human salivary glands and propagate them in vitro. Our protocol can be further optimized to meet the needs of individual studies. Briefly, salivary tissue is mechanically and enzymatically separated to isolate single cells or small clusters of cells. Selection for epithelial cells occurs by plating onto a basement membrane matrix in the presence of media optimized to promote epithelial cell growth. These resulting cultures can be maintained as three-dimensional clusters, termed "salispheres", or grown as a monolayer on treated plastic tissue culture dishes. This protocol results in the outgrowth of a heterogenous population of mainly epithelial cells that can be propagated for 5-8 passages (15-20 population doublings) before undergoing cellular senescence.
Article
Saliva aids in digestion, lubrication, and protection of the oral cavity against dental caries and oropharyngeal infections. Reduced salivary secretion, below an adequate level to sustain normal oral functions, is unfortunately experienced by head and neck cancer patients treated with radiotherapy, and by patients with Sjögren's syndrome. No disease‐modifying therapies exist to date to address salivary gland hypofunction (xerostomia, dry mouth) because pharmacotherapies are limited by the need for residual secretory acinar cells, which are lost at the time of diagnosis, while novel platforms such as cell therapies are yet immature for clinical applications. Autologous salivary gland primary cells have clinical utility as personalized cell therapies, if they could be cultured to a therapeutically useful mass while maintaining their in vivo phenotype. Here, we devised a serum‐free scalable suspension culture system that grows partially‐digested human salivary tissue filtrates comprising of acinar and ductal cells attached to their native extracellular matrix components while retaining their 3D in vivo spatial organization; we have coined these salivary spheroids as “Salivary Functional Units (SFU)”. The proposed SFU culture system was sub‐optimal, but we have found that the cells could still survive and grow into larger salivary spheroids through cell proliferation and aggregation for 5 to 10 days within the oxygen diffusion rates in vitro. In summary, by using a less disruptive cell isolation procedure as the starting point for primary cell culture of human salivary epithelial cells, we demonstrated that aggregates of cells remained proliferative and continued to express acinar and ductal cell‐specific markers.
Article
Objectives The Human Salivary Gland cell line (HSG), labelled as a submandibular ductal cell line, is commonly used as in vitro models to study radiation therapy, Sjögren's syndrome, pleomorphic adenoma, mucocele, epithelial‐to‐mesenchymal transition, and epigenetics. However, the American Type Culture Collection (ATCC) has recently released a list of cross‐contaminated cell lines that included HSG. Despite this notice, some research labs still use HSG as a salivary cell model. Accordingly, this study examined the authenticity of HSG sampled from three different laboratories Methods DNA was extracted from HSG and additional salivary cell lines (NS‐SV‐AC, NS‐SV‐DC, A253, HSY), and submitted for cell line authentication with short tandem repeat (STR) analysis Results All HSG samples had STR profiles indicating > 80% match with HeLa in both the ATCC and Deutsche Sammlung von Mikroorganismen und Zellkulturen (DSMZ) databases. This confirmed that HSG sampled from three different laboratories and HSY shared a common ancestry (host) with HeLa, while NS‐SV‐AC, NS‐SV‐DC, and A253 had unique STR profiles Conclusion STR analysis revealed that HSG was contaminated by the HeLa cell line. Furthermore, because genotyping of the original HSG cell line was not performed during its establishment, it will be difficult to authenticate an uncontaminated sample of HSG. This article is protected by copyright. All rights reserved.