ArticlePDF Available

Advanced Biotechnology for Cell Cryopreservation

Authors:

Abstract and Figures

Cell cryopreservation has evolved as an important technology required for supporting various cell-based applications, such as stem cell therapy, tissue engineering, and assisted reproduction. Recent times have witnessed an increase in the clinical demand of these applications, requiring urgent improvements in cell cryopreservation. However, cryopreservation technology suffers from the issues of low cryopreservation efficiency and cryoprotectant (CPA) toxicity. Application of advanced biotechnology tools can significantly improve post-thaw cell survival and reduce or even eliminate the use of organic solvent CPAs, thus promoting the development of cryopreservation. Herein, based on the different cryopreservation mechanisms available, we provide an overview of the applications and achievements of various biotechnology tools used in cell cryopreservation, including trehalose delivery, hydrogel-based cell encapsulation technique, droplet-based cell printing, and nanowarming, and also discuss the associated challenges and perspectives for future development.
Content may be subject to copyright.
Vol.:(0123456789)
1 3
Transactions of Tianjin University
https://doi.org/10.1007/s12209-019-00227-6
REVIEW
Advanced Biotechnology forCell Cryopreservation
JingYang1,2,3· LeiGao1,2,3· MinLiu1,2,3· XiaojieSui1,2,3· YingnanZhu1,2,3· ChiyuWen1,2,3· LeiZhang1,2,3
Received: 29 October 2019 / Revised: 25 November 2019 / Accepted: 27 November 2019
© The Author(s) 2019
Abstract
Cell cryopreservation has evolved as an important technology required for supporting various cell-based applications, such
as stem cell therapy, tissue engineering, and assisted reproduction. Recent times have witnessed an increase in the clinical
demand of these applications, requiring urgent improvements in cell cryopreservation. However, cryopreservation technol-
ogy suffers from the issues of low cryopreservation efficiency and cryoprotectant (CPA) toxicity. Application of advanced
biotechnology tools can significantly improve post-thaw cell survival and reduce or even eliminate the use of organic solvent
CPAs, thus promoting the development of cryopreservation. Herein, based on the different cryopreservation mechanisms
available, we provide an overview of the applications and achievements of various biotechnology tools used in cell cryo-
preservation, including trehalose delivery, hydrogel-based cell encapsulation technique, droplet-based cell printing, and
nanowarming, and also discuss the associated challenges and perspectives for future development.
Keywords Cell cryopreservation· Biotechnology· Trehalose delivery· Hydrogel-based cell encapsulation· Droplet-based
cell printing· Nanowarming
Introduction
Cell cryopreservation is a technology used to preserve living
cells, while maintaining their cellular viability and func-
tions even at cryogenic temperatures (usually at − 80°C
or − 196°C). At such ultra-low temperatures, the chemi-
cal, biological, and physical processes normally occurring
at cellular level can remain suspended for a long time. In
recent times, cell cryopreservation has become an important
supporting technology for various cell-based applications
such as stem cell therapy, tissue engineering, assisted human
reproduction, and transfusion medicine [1]. The importance
of cryopreservation technology is correctly reflected by the
burgeoning demand of stem cell therapy, owing to which
approximately 400,000 units of umbilical cord blood have
been stored worldwide for public use and 900,000 units for
private use [2]. As per the report of Stem Cell Banking Mar-
ket, the global stem cell banking market had a current value
of ~ 18.2 billion dollars in 2017, and this will reach ~ 54.1
billion dollars by 2024. Red blood cell cryopreservation has
also gained importance in the past few years. Cryopreserva-
tion of red blood cells (RBCs) can extend the storage time
from 42 d (hypothermic preservation) to 10years, which will
ease the burden of short blood supply, especially in remote
areas [3].
However, during freezing–thawing cycles, cells inevitably
suffer from cryoinjuries, including solution injury and ice
injury. The freeze concentration-induced excessive dehydra-
tion can damage cells resulting in solution injury. Besides
this, ice formation and growth during cryopreservation can
mechanically damage the biological structure of cells result-
ing in ice injury [4] (Fig.1a). Cryoprotectants (CPAs) play
a pivotal role in protecting cells against these cryoinjuries
and allow their successful storage at cryogenic temperatures.
CPAs can be broadly classified into two main categories on
the basis of the permeability or non-permeability of CPAs
into the cellular membrane [5]. Permeating CPAs mainly
Jing Yang and Lei Gao have contributed equally to this work.
* Lei Zhang
lei_zhang@tju.edu.cn
1 Department ofBiochemical Engineering, School
ofChemical Engineering andTechnology, Tianjin
University, Tianjin300350, China
2 Frontier Science Center forSynthetic Biology andKey
Laboratory ofSystems Bioengineering (MOE), School
ofChemical Engineering andTechnology, Tianjin University,
Tianjin300350, China
3 Qingdao Institute forMarine Technology ofTianjin
University, Qingdao266235, China
J.Yang et al.
1 3
include organic solvents, such as glycerol and DMSO, which
can permeate phosphate bilayers. This permeability is usu-
ally driven by a concentration gradient and confers intracel-
lular protection to cells [6]. However, most of the organic
solvent CPAs exhibit toxicity or poor biocompatibility that
can cause serious side effects in patients like hemolysis,
neurotoxicity, cardiovascular failure, respiratory arrest, and
fatal arrhythmias [7, 8]. In comparison with this, non-per-
meating CPAs provide extracellular protection only. These
include natural non-toxic carbohydrates (such as trehalose
and sucrose) and biomacromolecules (such as proteins and
polymers) [6]. Generally, non-permeating CPAs are com-
bined with permeating CPAs to ensure both extracellular
and intracellular protection, where the latter is required for
the critical protection of cells from inside. This results in a
compromise between high cryopreservation efficiency and
CPA toxicity [9].
For most cell types, conventional cryopreservation pro-
tocol involves stepwise freezing of sample at slow cooling
rates using 10–20% DMSO solution. In order to improve cell
cryopreservation efficiency, previous studies have mainly
focused on the optimization of CPA formulation, CPA intro-
duction, and freezing–thawing protocol suitable for different
cell types [10, 11]. Pollock etal. [12] reported the use of a
differential evolution algorithm to optimize cryopreserva-
tion protocols for Jurkat cells (300mmol/L trehalose, 10%
glycerol, and 0.01% ectoine at 10°C/min) and mesenchymal
stem cells (300mmol/L ethylene glycol, 1mmol/L taurine,
and 1% ectoine at 1°C/min), which resulted in post-thawing
cell viabilities of 95% and 96%, respectively. However, the
optimization of cryopreservation protocol still suffers from
two major challenges: (1) unfavorable post-thaw cell viabil-
ity or functions and (2) safety concerns induced by CPA
toxicity. For cryopreservation of hepatocytes, Mahler and
co-workers [13] reported only 61–75% survival of isolated
cells post-cryopreservation, and post-thawing cell attach-
ment efficiency of 30–39%. Besides this, there are reports
where the critical functions in some of the therapeutic cells
such as mesenchymal stem cells (MSCs), natural killer cells,
and dendritic cells (DCs) were compromised after use of
conventional cryopreservation protocol [1417]. Organic
solvents glycerol and DMSO are widely used in intracellu-
lar protection; however, both lack biocompatibility. Glycerol
can induce severe hemolysis [18], while the use of DMSO
is found to be associated with many side effects in patients,
like neurotoxicity, cardiovascular failure, respiratory arrest,
fatal arrhythmias, and others [19]. Fetal bovine serum (FBS)
is frequently combined with organic solvents to supplement
Fig. 1 a Schematic diagram for
two types of cryoinjuries occur-
ring during cryopreservation
of cells; b schematic diagram
for various biotechnology tech-
niques used in cryopreservation
and their method of protection
Advanced Biotechnology forCell Cryopreservation
1 3
extracellular cryopreservation. However, FBS is derived
from animals and has risk of inducing viral infections and
immunogenicity inpatients [20, 21].
In recent years, the developments of biotechnology pro-
vided an opportunity to improve the final outcome of cell
cryopreservation. Generally, biotechnology refers to use
of scientific techniques to study and address the problems
associated with living organisms. In this review, we summa-
rize the applications and achievements of various advanced
biotechnology tools used in cell cryopreservation. These
include trehalose delivery, hydrogel-based cell encapsulation
technique, droplet-based cell printing, and nanowarming. All
these techniques broadly aim to enhance cell cryopreser-
vation and reduce or eliminate CPA toxicity. As shown in
Fig.1b, trehalose delivery is proposed to deliver non-per-
meable but biocompatible trehalose into cells as an alterna-
tive to organic solvents, with the aim to provide intracellular
cryopreservation. The other three techniques mainly enhance
extracellular protection during freezing or warming process
and thus aim to increase cryopreservation efficiency and
significantly reduce the concentration of organic solvents.
In addition to this, we discuss the challenges and provide
future perspectives for the development of biotechnology
tools used in cell cryopreservation.
Trehalose Delivery
Trehalose is a non-permeating disaccharide, which is used
as a bio-inspired CPA to protect cells or organisms against
cryoinjuries [22]. It facilitates the formation of a stable
glassy matrix and promotes preferential hydration in cellu-
lar biomolecules, stabilizing their functional conformations
[2325]. However, trehalose generally provides extracellular
protection only owing to its lower permeability. For intracel-
lular cryopreservation, it is used in combination with organic
solvent CPAs (glycerol or DMSO) [2628]. In order to avoid
the toxicity of organic solvents, several advanced biotechnol-
ogy tools have been used to deliver trehalose into cells to
provide both intracellular and extracellular cryopreservation.
These techniques help to achieve organic solvent-free cell
cryopreservation and high post-thaw cell survival efficiency
[29]. These biotechnology tools, including both physical and
chemical methods, can increase cellular membrane perme-
ability and thus transport non-permeable trehalose into cells
(Table1).
Physical Delivery Method
The physical delivery methods, including freezing-induced
membrane phase transition [3032] and electroporation
technology [3335], have been used to promote the per-
meation of trehalose into cells. These methods help to
achieve organic solvent-free cell cryopreservation. The
loading of trehalose can be easily controlled by manipu-
lating the concentration gradient. However, the increased
membrane permeability achieved by these methods suffers
from the issues of non-specificity, which results in uncon-
trolled influx and outflux of other molecules.
The use of freezing-induced membrane phase transition
method for intracellular delivery of trehalose into cells was
reported for the first time by Beattie etal. [36] in 1997, for
cryopreservation of pancreatic islet cells. During the cool-
ing process, changes occurring in the fluid-to-gel phase
transition result in reorganization of membrane lipid com-
ponents which increases the membrane permeability. A
concentration gradient then drives the intracellular move-
ment of trehalose to provide intracellular cryopreservation.
Gläfke etal. [30] reported the use of high extracellular
concentrations of trehalose for freezing platelets. This
method resulted in 98% membrane intact platelets, 76%
of which were in non-activated resting state. This platelet
cryopreservation protocol avoided any use of DMSO.
For more than 40years, electroporation technology
has been widely used for intracellular delivery of xeno-
molecules such as saccharides, drugs, plasmids, DNA
vaccine, siRNA, and proteins. It offers several advantages
like controllability, reproducibility, and high efficiency.
Application of an external electric pulse assists in the for-
mation of hydrophilic pores on the membrane resulting
in an increase in membrane permeability. This pore for-
mation can be reversible or irreversible depending on the
electric pulse conditions [3741]. In two separate studies,
Dovgan etal. [33] reported the use of electroporation for
efficient loading of trehalose into human adipose-derived
stem cells (hADSCs) and umbilical cord mesenchymal
stem cells (UC-MSCs). For electroporation in hADSCs,
the cells were incubated in 250mmol/L trehalose and
electroporated at the optimal conditions of 1.5kV/cm2,
at 8 pulses, 100μs, and 1Hz, prior to programmable
slow freezing. After thawing the cells, 83.8 ± 1.8% cell
recovery rate was observed, which was similar to that of
hADSCs (91.5 ± 1.6%) obtained using standard freezing
protocol (10% DMSO in 90% FBS). In comparison with
this, the electroporation of UC-MSCs under the optimal
electroporation conditions (430V, 8 pulses, 100μs, and
1Hz) resulted in 61% cell viability [34]. It has been pre-
viously shown that high voltage can result in great loss
of cell viability, while insufficient voltage compromises
the delivery of trehalose into cells. Therefore, to ensure
efficient trehalose delivery and favorable post-thaw cell
viability, it is important to optimize suitable voltage condi-
tions for different cell types.
J.Yang et al.
1 3
Table 1 Different trehalose delivery methods used for cell cryopreservation
Delivery methods Cell type Main results Advantages Disadvantages
Physical delivery Freezing-induced lipid-phase
transition
Platelets Post-thawing, 98% platelets
showed/were having intact
membrane
Organic solvent-free cell
cryopreservation based on
trehalose
1. Trehalose loading into
cells easily controlled by
concentration gradient
2. Reversible based on exter-
nal stimuli
3. Simple processing
1. Non-specific membrane
permeability
2. Membrane injury concerns
induced by thermal and
electric shock
Electroporation hADSCs 1. 84% cell survival rate
post-thawing
2. Maintenance of normal
cell proliferation and dif-
ferentiation potential
UC-MSCs 61% cell survival rate post-
thawing
Chemical delivery PP50 Erythrocyte 1. 123mmol/L of intracellu-
lar trehalose was achieved
in erythrocytes
2. 83% post-thaw erythrocyte
survival
Loading of high amounts of
intracellular trehalose.
1. Non-specific membrane
permeability
2. Long incubation time
3. Cytotoxicity concerns
4. Tedious washing step
5. Complex material prepara-
tion
SAOS-2 The number of metabolically
active cells at 24h post-
thaw was between 91% and
103%
α-Hemolysin Fibroblasts
and
keratino-
cytes
Long-term post-thaw
survival rate was 80% for
fibroblasts and 70% for
keratinocytes
1. Reversible based on exter-
nal stimuli.
2. Trehalose loading into
cells easily controlled by
concentration gradient
Genipin-cross-linked
Pluronic-F127 nanoparti-
cles (GNPs)
hADSCs Approximately 90% cell
viability and normal dif-
ferentiation potentials, and
distinctive markers expres-
sion was maintained
1. Utilize natural endocytosis
process
2. Specific transport of high
amounts of trehalose
1. Long incubation time
2. Complex material prepara-
tion
Apatite nanoparticles RBCs Increasing RBC cryosurvival
up to 91%, which is com-
parable to FDA-approved
cryopreservation protocol
employing glycerol
Advanced Biotechnology forCell Cryopreservation
1 3
Chemical Delivery Method
Chemical delivery methods utilize cell membrane perforat-
ing agents like synthetic polymers [42, 43], α-hemolysin
[4446], and nanoparticles [4749] to achieve intracellu-
lar delivery of trehalose. These chemical materials usually
involve a complex preparation process. They can interact
with cell membranes and increase membrane permeability
temporarily. Trehalose has been used as sole CPA in combi-
nation with these chemical delivery methods to successfully
cryopreserve RBCs, fibroblasts, keratinocytes, and human
mesenchymal stem cells (hMSCs).
Mercado etal. [43] designed and synthesized a series of
biomimetic derivatives of PLP polymer to facilitate intra-
cellular delivery of trehalose. Particularly, co-incubation
of PP50 (composed of PLP grafted with l-phenylalanine)
with erythrocytes and trehalose suspension increased the
intracellular concentration of trehalose to 123 ± 16mmol/L.
After cryopreservation of erythrocytes loaded with treha-
lose, the erythrocyte survival rate was 82.6±3.4%, which
was 20.4 ± 5.6% higher as compared to the unloaded eryth-
rocytes. PP50-mediated trehalose delivery method has been
also used in organic solvent-free cryopreservation of nucle-
ated human cell line SAOS-2. Although the post-thaw cell
viability was only 60 ± 2%, the number of metabolically
active cells at 24h post-thaw was in the range of 103±4 to
91±5%. This was comparable to the results observed for
cells frozen using DMSO [42]. Mechanically, PP50 adsorp-
tion onto the membrane contributed by its amphipathic char-
acteristic induced the appearance of thinner phospholipid
bilayer, which resulted in an increase in trehalose uptake
[42]. However, longer incubation times in cryopreserva-
tion of erythrocytes might result in undesirable hemoly-
sis of cells. Thus, PP50 must be removed to overcome the
safety issues, which generally involves a tedious washing
procedure.
α-Hemolysin is a genetically engineered endotoxin
derived from Staphylococcus aureus. It has been shown to
work via generation of pores in the lipid bilayers for both
fibroblasts and keratinocytes, allowing an influx of trehalose
[46, 50]. Buchanan etal. [46] used α-hemolysin to achieve
intracellular trehalose concentrations of up to 0.5mol/L.
Trehalose at concentration of only 0.2mol/L provided
cryopreservation in fibroblasts and keratinocytes, with tre-
halose as sole CPA. After long-term cryopreservation, the
post-thaw survival rates were 80% and 70% for fibroblasts
and keratinocytes, respectively. Despite being so promis-
ing, α-hemolysin being a bacteria-derived pore protein may
induce undesirable immune responses in patients. Therefore,
it must be removed from cells prior to their use in clinical
therapy. The safety concerns and necessary removal steps
associated with the use of α-hemolysin limit its clinical
application.
Nanomaterials are widely used in medical field espe-
cially as vehicles for the delivery of drugs, such as chemi-
cal molecules, DNA vaccine, and protein or peptide drugs,
to therapeutic target [5154]. In recent years, several stud-
ies have reported efficient delivery of trehalose into cells
using nanomaterials. The nanoparticles generally utilize the
natural process of endocytosis to specifically deliver treha-
lose into cells without any harmful effects [29]. The use
of nanoparticles for trehalose delivery and cryopreserva-
tion of cells with trehalose as sole CPA has been reported
to maintain cell viability and functions. Rao etal. [48]
developed a pH-responsive genipin-cross-linked Pluronic
F127–chitosan nanoparticle (GNP), which efficiently encap-
sulated trehalose for intracellular delivery (Fig.2a). For
cryopreservation of hADSCs, the cells were incubated with
trehalose-loaded GNPs (nTre) for 24h and cryopreserved
in culture medium containing 200mmol/L free trehalose.
After rewarming, pre-incubation with nTre resulted in 90%
cell viability which was comparable to the cell viability
obtained post-cryopreservation with DMSO. Besides this,
the differentiation potential and the expression of distinctive
markers in hADSCs remained unchanged upon cryopreser-
vation. Another nano-vehicle used for delivery of trehalose
is a type of biomimetic (bone-like) apatite nanoparticle.
These apatite nanoparticles have been shown to efficiently
deliver drugs and nucleic acids into various types of cells.
Stefanic etal. [49] utilized colloidal bio-inspired apatite
nanoparticles to mediate intracellular delivery of trehalose
into RBCs. The local interactions between apatite NPs and
the bilayer enhanced the translocation of trehalose into the
cells (Fig.2b). Cryopreservation of trehalose-laden RBCs
demonstrated that the use of this glycerol-free cryopreser-
vation protocol tremendously increased survival of RBCs
to 91%, which was 42% higher as compared to the control
without NP treatment. These results were comparable to the
FDA-approved cryopreservation protocol that utilized glyc-
erol as CPA. Nanoparticles-mediated intracellular delivery
of trehalose has shown great potential to achieve clinical
cryopreservation of therapeutic cells without any use of
organic solvents. A possible limitation of this method could
be requirement of long incubation time to achieve sufficient
intracellular trehalose concentration. The incubation time
required for trehalose delivery was 24h and 7h for GNPs
and apatite nanoparticles, respectively.
Hydrogel‑Based Cell Encapsulation
Technology
Hydrogel-based cell encapsulation technology refers to
the encapsulation of living cells within semipermeable
capsules prepared using hydrogel materials. This technol-
ogy has been found to be highly promising for various
J.Yang et al.
1 3
cell-based studies and applications. This technology pro-
vides suitable3D microenvironment similar to the extra-
cellular matrix, blocks the immunogenicity of encapsu-
lated cells, and directs the differentiation of stem cells
[5557]. In recent years, hydrogel-based cell encapsula-
tion technology has been widely used in cell cryopreser-
vation. The capsules not only protect the inner cells from
mechanical and osmotic stress during the freezing and
warming process, but also allow the bidirectional diffu-
sion of nutrients, oxygen, and waste products. Numerous
studies have established its positive effects on post-thaw
cell viability and functions [5860].
The method used for cell encapsulation greatly influences
the final outcome of cell cryopreservation. These methods
can be broadly divided into three categories, namely emul-
sion/thermal gelation, extrusion (electrostatic spray, air flow
nozzle, and vibrating nozzle), and microfluidic method [58].
Choice of method depends on two main factors: capability
to maintain high cell viability/function and ability to control
the capsule phenotypic characteristic such as size, shape,
strength, and permeability [61].
Emulsion Method
Emulsion is a generic method used to encapsulate cells
within bulk hydrogels. In this method, pre-gel solutions are
first prepared by suspending cells and gel materials. When
the dispersion reaches an equilibrium state, gel formation
is triggered by adding an initiator or changing the physical
conditions, such as UV light. Although emulsion method
is simple and easy to scale up, the process of gelation may
result in cell death and loss of functions owing to the toxicity
of initiator, chemical cross-linking, and unfavorable reaction
conditions [58, 62].
In recent years, numerous novel cross-linking approaches
have been used in the preparation of cell-loaded hydrogels
to avoid the negative effects of conventional methods.
PVA-based hydrogel including PVA–gelatin cryogels and
Fig. 2 Nanomaterials-mediated
trehalose transportation into
cells. a The encapsulation of
trehalose in genipin-cross-
linked Pluronic F127–chitosan
nanoparticles (GNPs) to pro-
duce nanoparticle-encapsulated
trehalose (nTre). b Interactions
of biomimetic (bone-like) apa-
tite nanoparticles with the lipid
bilayer and enhanced delivery
of trehalose into RBCs
Advanced Biotechnology forCell Cryopreservation
1 3
PVA–carrageenan (Car) scaffold was prepared via freeze-
gelation technique. The encapsulation process only involved
a freezing step without requirement of any external cross-
linking agents. After cryopreservation, cell viability and
functions were observed to be unaffected [6366]. Zeng
etal. [67] developed a supramolecular gel that also involved
a cooling process to trigger the gel formation. For cryo-
preservation of encapsulated PC12 cells and RSC96 cells, a
mixture of cell suspension, gelator, and DMSO was prepared
and cooled in ice-water bath. Gelator self-assembled to form
supramolecular gel at 8.2°C and then programmed freez-
ing of the system was carried out at − 80°C (Fig.3a). The
post-thaw viability of PC12 and RSC96 cells increased sig-
nificantly owing to the protection provided by the hydrogel
during the freezing and thawing process. In addition to this,
the thermo-reversible supramolecular gel could be removed
easily by centrifugation. Jain etal. [68] reported a two-
component molecular recognition gelation method that was
Fig. 3 Schematic diagram for cell encapsulation in hydrogel cap-
sules by different methods. Emulsion method: a cooling and thaw-
ing process for cells encapsulated in supramolecular gel, b insitu
hydrogelation via SPAAC click chemistry for cell encapsulation and
cryopreservation, c preparation and cryopreservation process for
hASCs–K-carrageenan hydrogel construct. Extrusion method: d a
two-fluidic electrospraying method for encapsulation of cells in core–
shell capsules. Microfluidic method: e electrohydrodynamic atomi-
zation (EHDA) method for fabrication of cell-laden microcapsules
with uniform size, f encapsulation of individual rat islets into alginate
hydrogel using a droplet microfluidic device at room temperature,
and g preparation, vitrification and warming process for cell-laden
alginate-based core–shell hydrogel produced using a double emulsion
flow-focusing tube-in-tube capillary microfluidic device
J.Yang et al.
1 3
adopted to develop a dextran-based polyampholyte hydrogel
having cryoprotective properties. The gelation process was
initiated by mixing of azide-Dex-PA and DBCO-Dex, and
this gelation mainly depended on the reactant concentration
(Fig.3b). In the absence of any CPA, L929 cells encapsu-
lated within these hydrogels showed a recovery rate of > 90%
(in optimum gelation condition) after thawing. As a natural
thermo-sensitive polymer, K-carrageenan could form stable
hydrogel via ionic gelation process. Potassium chloride was
used for the cross-linking of K-carrageenan, allowing further
stabilization of the hydrogels (Fig.3c). HMSCs encapsulated
with K-carrageenan hydrogel showed improved proliferation
and chondrogenic potential post-cryopreservation [69].
Extrusion Method
Extrusion methods majorly include two methods: electro-
static spray and air-jet encapsulation technology. These are
commonly used for cell encapsulation owing to their high
throughput and production of evenly sized beads [61].
Wolters etal. [70] developed an air-jet droplet generator
and used it to produce small, uniform, and smooth alginate
beads while maintaining high throughput. In 2010, Mal-
pique etal. [71] investigated for the first time cryopreserva-
tion of brain cell neurospheres by encapsulating the cells
within alginate hydrogel using the air-jet two-channel drop-
let generator. The results showed that the cell viability and
metabolic activity were significantly higher in encapsulated
neurospheres as compared to the non-encapsulated ones. It
might be contributed by reduced fragmentation and better
maintenance of spherical shape of aggregates upon encap-
sulation in alginate hydrogels. However, air-jet technology
applied during microcapsule formation has several limita-
tions such as use of harsh shearing forces and formation of
air bubbles and “tails.”
Cell encapsulation using electrostatic spray method
involves generation of droplets containing cells and poly-
mers from the nozzle, followed by spraying into a container
with gelling bath to form hydrogel beads. The hydrogel
beads formation is assisted by the electrostatic force between
the gelling bath and the nozzle [71]. Zhang etal. [72] encap-
sulated mouse MSCs into small (~ 100μm) Ca-alginate
microcapsules generated by electrostatic spray method. The
vitrification of cell-loaded microcapsules with low concen-
tration of DMSO maintained high post-thaw cell viability
in encapsulated cells. The Ca-alginate microcapsules pro-
vided great protection to the cells during cryopreservation.
Two-fluidic electro-co-spraying technique was developed
and adopted to continuously produce core–shell alginate
capsules, which had better mass transfer and were used to
encapsulate organoids (Fig.3d). The core–shell structure
of the capsules provided better cell recovery after cryo-
preservation of organoids, probably through prevention of
intracellular ice formation [7375]. Electrohydrodynamic
atomization (EHDA) is an attractive approach that immobi-
lizes living cells into biomaterials permitting localized and
minimally invasive delivery. It minimizes cell leakage and
maintains viability during the delivery process. Naqvi etal.
[76] combined alginate and EHDA technique to fabricate
bone marrow stomatal cells (BMSCs)-encapsulated micro-
capsules (Fig.3e). The results of cryopreservation showed
that micro-encapsulation of BMSCs within alginate main-
tained their cell viability and potential to synthesize sGAG
and collagen. Electrostatic spray method offers several
advantages including cytocompatibility, ease of operation,
and high efficiency. Besides this, the manufacturing process
of capsules could be performed in a sterile environment.
Therefore, the electrostatic spray method is promising and
suitable to encapsulate cell resources or CPTs for long-term
storage purpose.
Microuidic Method
Rapid development of micro- and nanotechnologies has
allowed operation of cell encapsulation procedures on-chip
[1]. Built on flow focusing, miniaturized devices are to
encapsulate cells into capsules. Microfluidic methods permit
a high degree of control over the morphological and dimen-
sional properties. The experimental platforms are physically
smaller than the macro-encapsulation systems. In addition
to these, microfluidic method offers several advantages over
macro-encapsulation systems, such as low cost, ease to scale
up, disposability, specific designs, and rapid implementation
[57, 76].
Many groups investigated the cryopreservation of cells
encapsulated within hydrogel beads produced by microflu-
idic methods. In a droplet microfluidic platform, individual
rat pancreatic islets were encapsulated with FOSD function-
alized hydrogel microcapsules. This study aimed to establish
single-islet-based quality control assay for assessing qual-
ity and functionality of individually cryopreserved islets
(Fig.3f). Hydrogel membrane surrounding the encapsulated
islet effectively enhanced the insulin secretion after thawing.
The unique microstructure of the hydrogel was character-
ized by the presence of a compact 3D porous network and
considerable amount of non-freezable bound water, which
may alleviate the cryoinjury to the cells, playing a role simi-
lar to the CPAs [77]. Large-volume low-CPA cell vitrifica-
tion was achieved by microfluidic-based alginate hydrogel
micro-encapsulation system (Fig.3g) [78, 79]. HASCs in
low concentration of CPA medium (2mol/L) were encap-
sulated into core–shell microcapsules using an elaborate
microfluidic system and then loaded into 0.25mL conven-
tional plastic straw (PS). These were directly plunged into
liquid nitrogen to realize/induce vitrification. After thawing,
hASCs liberated from the microcapsules showed no changes
Advanced Biotechnology forCell Cryopreservation
1 3
in viability and differential capacity. In terms of mechanism,
the microcapsule effectively inhibited the ice formation and
further propagation during cooling and warming process.
The IRI activity of microcapsules especially protected the
cells against severe mechanical injuries.
Droplet‑Based Cell Printing
Droplet-based techniques find wide applications in various
fields, such as inkjet printing, emulsion polymerization, and
DNA arraying, owing to their high efficiency and low cost
[8084]. Introduction of this advanced technology into cell
cryopreservation created a series of novel vitrification proto-
cols that are characterized by lower CPA concentration and
higher cooling and warming rates. This is mainly applied
to smaller volumes of cryo-system [85]. Droplet-based cell
vitrification process not only solved the problem of high
CPAs concentration required for conventional vitrification
procedure, but also conferred significant protection to cells
by reducing the time required for ice crystal formation and
alleviating the osmotic shock. Besides this, the whole pro-
cess is easy and quick, allowing a possibility of large-scale
automation [8688]. Therefore, this innovative approach
may contribute significantly in the development of cell
cryopreservation.
Fig. 4 Schematic diagram for preparation of cell-laden droplets using
different devices. a Cell-CPA solution was loaded into valve-based
droplet injector, and the resulting droplets were directly injected into
liquid nitrogen. The ejected cells were collected in a cell strainer and
were rapidly transferred (in nitrogen vapor) to the thawing media, fol-
lowed by a step-by-step thawing process. b RBC-CPA droplets were
printed onto a cryo-paper as nitrogen gas flowed through a droplet
ejector, which transformed the bulk of the RBC-CPA mixture into
nanoliter droplets. Vitrification was achieved by submerging the cryo-
paper into liquid nitrogen. Warming process was performed by thaw-
ing the cells on a cryo-paper in phosphate-buffered saline at 37°C.
c Cell-laden droplets were rapidly ejected onto a freezing film using
a cell printer with high throughput and precise spatial controllabil-
ity. Vitrification/thawing process was achieved by pouring the liquid
nitrogen/warm water onto the other side via boiling heat transfer that
helped to maintain high cooling/warming rate
J.Yang et al.
1 3
Demirci etal. [84] reported successful vitrification of
many cell types by means of optimal droplet-based proce-
dure. This technique involved generation of droplets contain-
ing cell and CPAs solution using a modified jet device, which
were further received/transferred in a container filled with
liquid nitrogen and warmed in 37°C water bath (Fig.4a).
Before vitrification, droplet generation process did not affect
the call viability and cell survival rate was maintained to
approximately 90%. Propanediol and trehalose were used as
CPAs instead of toxic DMSO for cell vitrification. Even after
thawing, the viability of cells was maintained well.
Assal etal. [89] developed a novel cryo-printer that could
transform a bulk volume of human blood into nanoliter cryo-
inks on a cryo-paper, which was immersed into liquid nitro-
gen for rapid vitrification (Fig.4b). After rewarming, the
recovered human RBCs showed normal characteristic fea-
tures. In addition to this, there was no effect on the essential
functions of recovered RBCs, including phosphorylation of
band 3 protein, expression of complement receptor 1, and
maintenance of intracellular nitric oxide and reactive oxygen
species levels. Besides these intrinsic advantages offered by
droplet-based vitrification, the cryo-ink containing CPAs
medium such as ectoine, trehalose, and PEG also reduced
the injuries suffered by RBCs during the cooling and warm-
ing processes.
High-throughput non-contact vitrification of cell-laden
droplets was reported by Shi etal. [90] in 2015. Cell printing
generated droplets containing cell CPAs onto an ultra-thin
freezing film. Vitrification/thawing process was operated by
pouring liquid nitrogen/warm water onto the other side via
boiling heat transfer. This ensured maintenance of high cool-
ing/warming rate and avoided direct contact between cells
and liquid nitrogen/water, preventing chances of potential
contamination (Fig.4c). The use of this novel method pro-
vided successful vitrification in NIH 3T3 cells and hASCs.
After thawing, both cell viability and differentiation poten-
tial remained unaffected.
Recently, a CPA-free cryopreservation method-based
inkjet cell printing technology was developed by Akiyama
etal. [91]. It was successfully used for the vitrification of
several mammalian cell types such as 3T3 cells, C2C12
cells, and rat MSCs at ultra-rapid cooling rates. The drop-
lets containing cells and culture medium were printed onto a
glass substrate cooled with liquid nitrogen to realize/induce
solid-surface vitrification. Immediately after thawing, the
viability of 3T3 cells for 40-pL droplets on thick substrates
(thickness: 150μm) was comparable to the cell viability
obtained using conventional freezing method. The ultra-
rapid cooling and warming rates significantly inhibited ice
formation and ice recrystallization and also protected the
cells against cryoinjuries during the freezing and warming
processes.
Nanowarming
Conventional warming method (37°C water bath) fails to
provide sufficient warming rates and uniform warming effect
with the increasing scale of bio-specimen. During rewarm-
ing process, once the warming rates go below the critical
warming rates, ice recrystallization/devitrification occurs,
which is one of the major causes of cell suffered injuries.
However, advances in bio-specimen cooling for cryopreser-
vation have not been matched well by similar developments
in rewarming procedure [92]. Nanomaterials-mediated
nanowarming technology has a potential to be established as
a new approach to allow ultra-rapid and uniform rewarming
[93]. Nanowarming generally involves use of some nano-
materials such as Fe3O4 nanoparticles (Fe3O4 NPs) or gold
nanorods (GNRs) that can rapidly convert electromagnetic
or light energy into heat energy. Thus, rapid and uniform
rewarming of bio-specimens can be realized/achieved by
incorporating these nanomaterials into CPA solution and
heating with external electromagnetic, radiofrequency (RF)
or laser fields [9496]. Numerous studies have affirmed that
nanowarming technology is significantly effective and can
be used for improving cryopreservation of cells, tissues, and
organs.
RF Inductive Warming Process
Magnetic iron oxide nanoparticles are capable of transform-
ing external electromagnetic energy into heat energy rapidly.
Modified iron oxide nanoparticles (msIONPs) character-
ized by compatibility, colloidal stability, and capability to
remain in solution at higher concentration were synthesized
by Manuchehrabadi etal. [93]. Human dermal fibroblasts
(HDFs) were vitrified using VS55 cryo-solution loaded with
msIONPs and warmed by different methods. In the 1mL
system, the viability of nanowarmed HDFs was statistically
similar to the fresh control sample and higher than the via-
bility of slow-warmed sample.
Liu etal. [97] successfully achieved low-CPA vitrifica-
tion of stem cell-alginate hydrogel constructs by combining
nanowarming and micro-encapsulation technology (Fig.5a).
Fe3O4 NPs were mixed with low-CPA solution for RF induc-
tive warming process. After nanowarming, porcine adipose-
derived stem cells (pADSCs) showed viability > 80%, while
the attachment efficiency improved by three times as com-
pared to the pADSCs treated with slow warming process.
Besides these, the expression of surface markers and multi-
lineage potentials of pADSCs after nanowarming remained
unaffected. Mechanically, in addition to the cryopreservation
provided by cryo-solution and alginate hydrogel, Fe3O4 NPs
uniformly present outside of hydrogel further suppressed
devitrification and recrystallization during nanowarming
Advanced Biotechnology forCell Cryopreservation
1 3
process. Nanowarming was the primary reason for promoted
attachment of thawed pADSCs [97]. This technology was
also found to be efficient in human UC-MSCs and resulted
in an improved vitrification outcome [98] (Fig.5b).
Laser Radioactive Warming Process
Gold nanoparticle-based laser warming has the potential to
provide a platform for both extra- and intracellular heating
of vitrified biomaterials, ranging in size from nm to mm
in µL-sized droplets. Khosla etal. [99] mixed GNRs with
cryo-solution for rapid cooling and nanowarming of HDFs
(Dcell = 10µm) in droplet volume. The method of nanowarm-
ing used was laser warming technology irradiated with a
1064nm laser pulse for 1ms (Fig.5c). A cell viability of >
90% was maintained in HDF cells post-laser warming. Simi-
lar warming method was used to rewarm vitrified zebrafish
embryos. Before vitrification, biocompatible PEGylated
GNRs were microinjected directly into zebrafish embryos
with 2.3mol/L PG, thereby helping to distribute the laser
energy throughout the embryo during warming (Fig.5d). As
compared to the conventionally warmed control group, the
GNRs-mediated laser warming of embryos resulted in 31%
viable embryos with consistent structure at 1h, 17% viable
embryos continuing development at 3h, and 10% viable
embryos showing movement at 24h post-warming [100].
Two-dimensional (2D) graphene oxide (GO) and molyb-
denum disulfide (MoS2) nanosheets (NSs) were used to
improve warming process of bio-samples owing to their
photothermal effects. Human umbilical vein endothelial cells
Fig. 5 Schematic diagram for different cell vitrification techniques
and nanoparticles-mediated nanowarming process. RF field-mediated
warming process: a Nanowarming procedure for vitrified PS loaded
with cell-alginate hydrogel constructs and NPs in CPA solution. b
Vitrification and nanowarming of hUCM-MSC-laden PS with mag-
netic induction heating. Laser field-mediated warming process: c
Sample droplet consisting of biomaterial(s), CPA, and gold nanorods
(GNRs) with a maximum volume of 1.8 µL was loaded onto a cus-
tomized cryotop. For rapid cooling, cryotop was directly immersed
into liquid nitrogen. Laser warming was achieved by pulsed laser irra-
diation yielding ultra-rapid rewarming at rates up to 2×107 °C/min.
d Overview of zebrafish embryo cryopreservation and laser GNRs
rewarming. Micro-injection consisting of 1064 nm resonant GNRs
and PG was injected into the space of the embryo, between yolk and
chorionic. Rapid cooling was achieved by immersing the modified
cryotop into liquid nitrogen. Laser irradiation of the embryo taken
from liquid nitrogen was used to achieve nanowarming. e Multistep
addition of vitrification solution and loading in PS, followed by rapid
cooling and photothermal rewarming with GO and MoS2/NSs at
37°C water bath
J.Yang et al.
1 3
(HUVECs) were chosen to study this novel NSs-mediated
spatial heating approach. For rewarming, cryopreserved
HUVECs were placed into warming solution (37°C) under
a near-infrared laser field and photothermal effect was
achieved at 5000mW/cm2 for 8–10s (Fig.5e). This warm-
ing technology significantly improved the cell viability as
compared to the conventional rewarming method and also
maintained normal cell function and subcellular ultrastruc-
ture. Further investigation showed that near-infrared laser
irradiation effectively decreased ice formation and restricted
recrystallization growth via micro- and macro-effects during
rewarming [101].
Conclusions andFuture Perspectives
With the advancement of cell-based applications, conven-
tional cell cryopreservation clearly failed to keep pace with
current and emerging needs. This review summarized the
recent advances in biotechnology tools, including trehalose
delivery, hydrogel-based cell encapsulation, droplet-based
cell printing, and nanowarming technology, used in cell
cryopreservation. Trehalose delivery technology helped to
overcome the major limitation of ultra-low permeability
of trehalose, and its application resulted in favorable post-
thaw cell survival rates without need of any organic solvent.
Trehalose delivery method provided both extracellular and
intracellular cryopreservation with trehalose as sole CPA
[29]. Hydrogel-based cell encapsulation technology created
a new platform for efficient cell transportation and preserva-
tion. It has promoted long-term storage of cell resources and
banking “off-the-shelf” cell-based therapy products at large
scale. Since hydrogel capsules supplement excellent cryo-
preservation, satisfactory outcomes of cell cryopreservation
can be also achieved at reduced DMSO concentrations [59,
102, 103]. Droplet-based cell printing can improve cell vitri-
fication and can be helpful to achieve high efficiency at low-
CPA concentration and reduce CPA toxicity and osmotic
stress during CPA loading and unloading process [86].
Nanowarming technique has revolutionized the progress in
warming method. Its use can achieve ultra-fast and uniform
rewarming, while avoiding the adverse effects of devitrifica-
tion on cells [93].
Remarkable progress has been made in the application of
advanced biotechnology tools to improve cell cryopreser-
vation. However, a serious challenge associated with cell
cryopreservation has been identified that needs to addressed.
Cryopreservation-induced delayed onset cell death has
been reported to result in significant loss (> 50%) in the
total cell population and compromises cellular functional-
ity [104106]. In order to further explore the true poten-
tial of biotechnology for cell cryopreservation, future work
should focus not only on the advancement of the existing
applications, but also discover new “binding domain” to
introduce other innovative biotechnology techniques.
Open Access This article is licensed under a Creative Commons Attri-
bution 4.0 International License, which permits use, sharing, adapta-
tion, distribution and reproduction in any medium or format, as long
as you give appropriate credit to the original author(s) and the source,
provide a link to the Creative Commons licence, and indicate if changes
were made. The images or other third party material in this article are
included in the article’s Creative Commons licence, unless indicated
otherwise in a credit line to the material. If material is not included in
the article’s Creative Commons licence and your intended use is not
permitted by statutory regulation or exceeds the permitted use, you will
need to obtain permission directly from the copyright holder. To view a
copy of this licence, visit http://creat iveco mmons .org/licen ses/by/4.0/.
References
1. Zhao G, Fu JP (2017) Microfluidics for cryopreservation. Bio-
technol Adv 35(2):323–336
2. Ballen K (2010) Challenges in umbilical cord blood stem cell
banking for stem cell reviews and reports. Stem Cell Rev Rep
6(1):8–14
3. Chang A, Kim Y, Hoehn R etal (2016) Cryopreserved packed red
blood cells in surgical patients: past, present, and future. Blood
Transfus 15(4):341–347
4. Yang J, Pan C, Zhang JM etal (2017) Exploring the potential of
biocompatible osmoprotectants as highly efficient cryoprotect-
ants. ACS Appl Mater Interfaces 9(49):42516–42524
5. Gurruchaga H, Saenz del Burgo L, Hernandez RM etal (2018)
Advances in the slow freezing cryopreservation of microencap-
sulated cells. J Control Release 281:119–138
6. Fuller B, Gonzalez-Molina J, Erro E etal (2017) Applications
and optimization of cryopreservation technologies to cellular
therapeutics. Cell Gene Therapy Insights 3(5):359–378
7. Pal R, Mamidi MK, Das AK etal (2012) Diverse effects of dime-
thyl sulfoxide (DMSO) on the differentiation potential of human
embryonic stem cells. Arch Toxicol 86(4):651–661
8. Shu Z, Heimfeld S, Gao D (2014) Hematopoietic SCT with
cryopreserved grafts: adverse reactions after transplantation and
cryoprotectant removal before infusion. Bone Marrow Transplant
49(4):469–476
9. Elliott GD, Wang SP, Fuller BJ (2017) Cryoprotectants: a review
of the actions and applications of cryoprotective solutes that
modulate cell recovery from ultra-low temperatures. Cryobiol-
ogy 76:74–91
10. Best BP (2015) Cryoprotectant toxicity: facts, issues, and ques-
tions. Rejuvenation Res 18(5):422–436
11. Lauterboeck L, Wolkers WF, Glasmacher B (2017) Cryobiologi-
cal parameters of multipotent stromal cells obtained from differ-
ent sources. Cryobiology 74:93–102
12. Pollock K, Budenske JW, McKenna DH etal (2017) Algorithm-
driven optimization of cryopreservation protocols for transfusion
model cell types including Jurkat cells and mesenchymal stem
cells. J Tissue Eng Regen Med 11(10):2806–2815
13. Mahler S, Desille M, Frémond B etal (2003) Hypothermic stor-
age and cryopreservation of hepatocytes: the protective effect of
alginate gel against cell damages. Cell Transplant 12(6):579–592
14. Chinnadurai R, Copland IB, Garcia MA etal (2016) Cryopre-
served mesenchymal stromal cells are susceptible to T-cell medi-
ated apoptosis which is partly rescued by IFNγ licensing. Stem
Cells 34(9):2429–2442
Advanced Biotechnology forCell Cryopreservation
1 3
15. Moll G, Alm JJ, Davies LC etal (2014) Do cryopreserved mes-
enchymal stromal cells display impaired immunomodulatory and
therapeutic properties? Stem Cells 32(9):2430–2442
16. Shaik S, Wu XY, Gimble J etal (2018) Effects of decade long
freezing storage on adipose derived stem cells functionality.
Sci Rep 8:8162
17. Zhou QQ, Zhang YL, Zhao M etal (2016) Mature dendritic
cell derived from cryopreserved immature dendritic cell shows
impaired homing ability and reduced anti-Viral therapeutic
effects. Sci Rep 6:39071
18. Sui XJ, Wen CY, Yang J etal (2019) Betaine combined with
membrane stabilizers enables solvent-free whole blood cryo-
preservation and one-step cryoprotectant removal. ACS Bio-
mater Sci Eng 5(2):1083–1091
19. Yang J, Cai NN, Zhai HW etal (2016) Natural zwitterionic
betaine enables cells to survive ultrarapid cryopreservation.
Sci Rep 6:37458
20. Cao TT, Zhang YQ (2017) The potential of silk sericin protein
as a serum substitute or an additive in cell culture and cryo-
preservation. Amino Acids 49(6):1029–1039
21. Kim SM, Yun CK, Park JH etal (2017) Efficient cryopreser-
vation of human mesenchymal stem cells using silkworm
hemolymph-derived proteins. J Tissue Eng Regen Med
11(8):2181–2192
22. Tsai S, Chong G, Meng PJ etal (2018) Sugars as supplemental
cryoprotectants for marine organisms. Rev Aquac 10:703–715
23. Cottone G (2007) A comparative study of carboxy myoglobin in
saccharide–water systems by molecular dynamics simulation. J
Phys Chem B 111(13):3563–3569
24. He XM, Fowler A, Toner M (2006) Water activity and mobility in
solutions of glycerol and small molecular weight sugars: implica-
tion for cryo- and lyopreservation. J Appl Phys 100(7):074702
25. Sun WQ, Leopold AC, Crowe LM etal (1996) Stability of dry
liposomes in sugar glasses. Biophys J 70(4):1769–1776
26. Chen FF, Zhang WJ, Wu W etal (2011) Cryopreservation of
tissue-engineered epithelial sheets in trehalose. Biomaterials
32(33):8426–8435
27. Ntai A, La Spada A, de Blasio P et al (2018) Trehalose to
cryopreserve human pluripotent stem cells. Stem Cell Res
31:102–112
28. Shinde P, Khan N, Melinkeri S etal (2019) Freezing of dendritic
cells with trehalose as an additive in the conventional freezing
medium results in improved recovery after cryopreservation.
Transfusion 59(2):686–696
29. Stewart S, He XM (2019) Intracellular delivery of trehalose for
cell banking. Langmuir 35(23):7414–7422
30. Gläfke C, Akhoondi M, Oldenhof H etal (2012) Cryopreserva-
tion of platelets using trehalose: the role of membrane phase
behavior during freezing. Biotechnol Progress 28(5):1347–1354
31. Zhang M, Oldenhof H, Sieme H etal (2016) Combining endo-
cytic and freezing-induced trehalose uptake for cryopreservation
of mammalian cells. Cryobiology 73(3):412
32. Zhang M, Oldenhof H, Sieme H etal (2016) Freezing-induced
uptake of trehalose into mammalian cells facilitates cryo-
preservation. Biochimica Et Biophys Acta Bba Biomembr
1858(6):1400–1409
33. Dovgan B, Barlič A, Knežević M etal (2017) Cryopreservation
of human adipose-derived stem cells in combination with treha-
lose and reversible electroporation. J Membrane Biol 250(1):1–9
34. Dovgan B, Dermol J, Barlič A etal (2016) Cryopreservation of
human umbilical stem cells in combination with trehalose and
reversible electroporation. 1st World congress on electropora-
tion and pulsed electric fields in biology, medicine and food &
environmental technologies. Springer, Singapore, pp 307–310
35. Shirakashi R, Köstner CM, Müller KJ etal (2002) Intracellular
delivery of trehalose into mammalian cells by electropermeabi-
lization. J Membr Biol 189(1):45–54
36. Beattie GM, Crowe JH, Lopez AD etal (1997) Trehalose: a
cryoprotectant that enhances recovery and preserves function
of human pancreatic islets after long-term storage. Diabetes
46(3):519–523
37. Batista Napotnik T, Miklavčič D (2018) Invitro electropora-
tion detection methods—an overview. Bioelectrochemistry
120:166–182
38. Cao YH, Ma EB, Cestellos-Blanco S etal (2019) Reply to
Nathamgari etal. Nanopore electroporation for intracellular
delivery of biological macromolecules. Proc Natl Acad Sci USA
116(46):22911
39. Rems L, Miklavčič D (2016) Tutorial: electroporation of cells in
complex materials and tissue. J Appl Phys 119(20):201101
40. Šatkauskas S, Ruzgys P, Venslauskas MS (2012) Towards
the mechanisms for efficient gene transfer into cells and tis-
sues by means of cell electroporation. Expert Opin Biol Ther
12(3):275–286
41. Yao CG, Liu HM, Zhao YJ etal (2017) Analysis of dynamic pro-
cesses in single-cell electroporation and their effects on param-
eter selection based on the finite-element model. IEEE Trans
Plasma Sci 45(5):889–900
42. Lynch AL, Chen RJ, Slater NKH (2011) PH-responsive polymers
for trehalose loading and desiccation protection of human red
blood cells. Biomaterials 32(19):4443–4449
43. Mercado SA, Slater NKH (2016) Increased cryosurvival of osteo-
sarcoma cells using an amphipathic pH-responsive polymer for
trehalose uptake. Cryobiology 73(2):175–180
44. Lynch AL, Chen RJ, Dominowski PJ etal (2010) Biopolymer
mediated trehalose uptake for enhanced erythrocyte cryosurvival.
Biomaterials 31(23):6096–6103
45. Acker JP, Lu XM, Young V etal (2003) Measurement of treha-
lose loading of mammalian cells porated with a metal-actuated
switchable pore. Biotechnol Bioeng 82(5):525–532
46. Buchanan SS, Gross SA, Acker JP etal (2004) Cryopreservation
of stem cells using trehalose: evaluation of the method using a
human hematopoietic cell line. Stem Cells Dev 13(3):295–305
47. Eroglu A, Russo MJ, Bieganski R etal (2000) Intracellular tre-
halose improves the survival of cryopreserved mammalian cells.
Nat Biotechnol 18(2):163–167
48. Rao W, Huang HS, Wang H etal (2015) Nanoparticle-mediated
intracellular delivery enables cryopreservation of human adi-
pose-derived stem cells using trehalose as the sole cryoprotect-
ant. ACS Appl Mater Interfaces 7(8):5017–5028
49. Stefanic M, Ward K, Tawfik H etal (2017) Apatite nanoparticles
strongly improve red blood cell cryopreservation by mediating
trehalose delivery via enhanced membrane permeation. Bioma-
terials 140:138–149
50. Zhang WJ, Rong JH, Wang Q etal (2009) The encapsulation and
intracellular delivery of trehalose using a thermally responsive
nanocapsule. Nanotechnology 20(27):275101
51. Russo MJ, Bayley H, Toner M (1997) Reversible permeabiliza-
tion of plasma membranes with an engineered switchable pore.
Nat Biotechnol 15(3):278–282
52. Solanki A, Kim JD, Lee KB (2008) Nanotechnology for regen-
erative medicine: nanomaterials for stem cell imaging. Nano-
medicine 3(4):567–578
53. Vernekar VN, James R, Smith KJ etal (2016) Nanotechnology
applications in stem cell science for regenerative engineering. J
Nanosci Nanotechnol 16(9):8953–8965
54. Watermann A, Brieger J (2017) Mesoporous silica nanoparticles
as drug delivery vehicles in cancer. Nanomaterials 7(7):189
J.Yang et al.
1 3
55. Qian HQ, Liu BR, Jiang XQ (2018) Application of nanomaterials
in cancer immunotherapy. Mater Today Chem 7:53–64
56. Orive G, Hernández RM, Gascón AR etal (2003) Cell encapsula-
tion: promise and progress. Nat Med 9(1):104–107
57. Zimmermann H, Ehrhart F, Zimmermann D et al (2007)
Hydrogel-based encapsulation of biological, functional tis-
sue: fundamentals, technologies and applications. Appl Phys A
89(4):909–922
58. Kang A, Park J, Ju J etal (2014) Cell encapsulation via micro-
technologies. Biomaterials 35(9):2651–2663
59. Zhang W, He X (2011) Microencapsulating and banking living
cells for cell-based medicine. J Healthc Eng 2(4):427–446
60. Huang HS, He XM (2016) Microscale materials and devices for
cell cryopreservation by vitrification. Multiscale Technologies
for Cryomedicine, World Scientific, Singapore, pp 101–124
61. Dluska E, Cui ZF, Markowska-Radomska A etal (2017) Cryo-
protection and banking of living cells in a 3D multiple emulsion-
based carrier. Biotechnol J 12(8):1600692
62. Majewski RL, Zhang WJ, Ma XJ etal (2016) Bioencapsulation
technologies in tissue engineering. J Appl Biomater Funct Mater
14(4):e395–e403
63. Rabanel JM, Banquy X, Zouaoui H etal (2009) Progress technol-
ogy in microencapsulation methods for cell therapy. Biotechnol
Prog 25(4):946–963
64. Chopra P, Nayak D, Nanda A etal (2016) Fabrication of
poly(vinyl alcohol)-carrageenan scaffolds for cryopreserva-
tion: effect of composition on cell viability. Carbohydr Polym
147:509–516
65. Vrana NE, Matsumura K, Hyon SH etal (2011) Cell encapsu-
lation and cryostorage in PVA–gelatin cryogels: incorporation
of carboxylated ε-poly-l-lysine as cryoprotectant. J Tissue Eng
Regen Med 6(4):280–290
66. Vrana NE, O’Grady A, Kay E etal (2009) Cell encapsulation
within PVA-based hydrogels via freeze-thawing: a one-step scaf-
fold formation and cell storage technique. J Tissue Eng Regen
Med 3(7):567–572
67. Zeng J, Yin YX, Zhang L etal (2016) A supramolecular gel
approach to minimize the neural cell damage during cryopreser-
vation process. Macromol Biosci 16(3):363–370
68. Jain M, Rajan R, Hyon SH etal (2014) Hydrogelation of dextran-
based polyampholytes with cryoprotective properties via click
chemistry. Biomater Sci 2(3):308–317
69. Popa EG, Rodrigues MT, Coutinho DF etal (2013) Cryopreser-
vation of cell laden natural origin hydrogels for cartilage regen-
eration strategies. Soft Matter 9(3):875–885
70. Wolters GHJ, Fritschy WM, Gerrits D etal (1992) A versatile
alginate droplet generator applicable for microencapsulation of
pancreatic islets. J Appl Biomater 3(4):281–286
71. Malpique R, Osório LM, Ferreira DS etal (2010) Alginate
encapsulation as a novel strategy for the cryopreservation of
neurospheres. Tissue Eng Part C Methods 16(5):965–977
72. Zhang WJ, He XM (2009) Encapsulation of living cells in small
(~ 100 μm) alginate microcapsules by electrostatic spraying: a
parametric study. J Biomech Eng 131(7):074515
73. Zhang WJ, Yang GE, Zhang AL etal (2010) Preferential vitri-
fication of water in small alginate microcapsules significantly
augments cell cryopreservation by vitrification. Biomed Micro-
devices 12(1):89–96
74. Zhao ST, Agarwal P, Rao W etal (2014) Coaxial electrospray of
liquid core–hydrogel shell microcapsules for encapsulation and
miniaturized 3D culture of pluripotent stem cells. Integr Biol
6(9):874–884
75. Lu YC, Fu DJ, An D etal (2017) Scalable production and cry-
ostorage of organoids using core-shell decoupled hydrogel cap-
sules. Adv Biosyst 1(12):1700165
76. Naqvi SM, Gansau J, Buckley CT (2018) Priming and cryo-
preservation of microencapsulated marrow stromal cells as a
strategy for intervertebral disc regeneration. Biomed Mater
13(3):034106
77. Kashaninejad N, Shiddiky MJA, Nguyen NT (2018) Advances
in microfluidics-based assisted reproductive technology: from
sperm sorter to reproductive system-on-a-chip. Adv Biosyst
2(3):1700197
78. Chen WY, Shu ZQ, Gao DY etal (2016) Sensing and sensibility:
single-islet-based quality control assay of cryopreserved pan-
creatic islets with functionalized hydrogel microcapsules. Adv
Healthcare Mater 5(2):223–231
79. Huang HS, Choi JK, Rao W etal (2015) Alginate hydrogel micro-
encapsulation inhibits devitrification and enables large-volume
low-CPA cell vitrification. Adv Funct Mater 25(44):6839–6850
80. Zhao G, Liu XL, Zhu KX etal (2017) Hydrogel encapsulation
facilitates rapid-cooling cryopreservation of stem cell-laden core-
shell microcapsules as cell-biomaterial constructs. Adv Health-
care Mater 6(23):1700988
81. Nir R, Lamed R, Gueta L etal (1990) Single-cell entrapment and
microcolony development within uniform microspheres amena-
ble to flow cytometry. Appl Environ Microbiol 56:2870–2875
82. Perçin G, Yaralioglu GG, Khuri-Yakub BT (2002) Microma-
chined droplet ejector arrays. Rev Sci Instrum 73(12):4385–4389
83. Blossey R (2003) Self-cleaning surfaces: virtual realities. Nat
Mater 2(5):301–306
84. Demirci U (2006) Acoustic picoliter droplets for emerging appli-
cations in semiconductor industry and biotechnology. J Micro-
electromech Syst 15(4):957–966
85. Reis N, Ainsley C, Derby B (2005) Ink-jet delivery of parti-
cle suspensions by piezoelectric droplet ejectors. J Appl Phys
97(9):094903
86. de Vries RJ, Banik PD, Nagpal S etal (2019) Bulk droplet vit-
rification: an approach to improve large-scale hepatocyte cryo-
preservation outcome. Langmuir 35(23):7354–7363
87. Youn JR, Song YS (2012) Cell-encapsulating droplet formation
and freezing. Appl Phys Lett 101(13):133701
88. Demirci U, Montesano G (2007) Cell encapsulating droplet vit-
rification. Lab Chip 7(11):1428
89. El Assal R, Guven S, Gurkan UA etal (2014) Bio-inspired cryo-
ink preserves red blood cell phenotype and function during nano-
liter vitrification. Adv Mater 26(33):5815–5822
90. Shi M, Ling K, Yong KW etal (2015) High-throughput non-
contact vitrification of cell-laden droplets based on cell printing.
Sci Rep 5:17928
91. Akiyama Y, Shinose M, Watanabe H etal (2019) Cryoprotectant-
free cryopreservation of mammalian cells by superflash freezing.
Proc Natl Acad Sci USA 116(16):7738–7743
92. Deller RC, Pessin JE, Vatish M etal (2016) Enhanced non-
vitreous cryopreservation of immortalized and primary cells by
ice-growth inhibiting polymers. Biomater Sci 4(7):1079–1084
93. Manuchehrabadi N, Gao Z, Zhang JJ etal (2017) Improved tissue
cryopreservation using inductive heating of magnetic nanoparti-
cles. Sci Transl Med 9(379):eaah4586
94. Czajka C (2017) Nanowarming improves cryopreservation. Sci-
ence 355(6328):920–921
95. Etheridge ML, Xu Y, Rott L etal (2014) RF heating of magnetic
nanoparticles improves the thawing of cryopreserved biomateri-
als. Technology 2(3):229–242
96. Wang T, Zhao G, Deng ZS etal (2015) Theoretical investigation
of a novel microwave antenna aided cryovial for rapid and uni-
form rewarming of frozen cryoprotective agent solutions. Appl
Therm Eng 89:968–977
97. Liu XL, Zhao G, Chen ZR etal (2018) Dual suppression effect
of magnetic induction heating and microencapsulation on ice
crystallization enables low-cryoprotectant vitrification of stem
Advanced Biotechnology forCell Cryopreservation
1 3
cell–Alginate hydrogel constructs. ACS Appl Mater Interfaces
10(19):16822–16835
98. Pan JJ, Shu ZQ, Zhao G etal (2018) Towards uniform and fast
rewarming for cryopreservation with electromagnetic resonance
cavity: numerical simulation and experimental investigation.
Appl Therm Eng 140:787–798
99. Khosla K, Wang YR, Hagedorn M etal (2017) Gold nanorod
induced warming of embryos from the cryogenic state enhances
viability. ACS Nano 11(8):7869–7878
100. Wang JY, Zhao G, Zhang ZL etal (2016) Magnetic induction
heating of superparamagnetic nanoparticles during rewarming
augments the recovery of hUCM-MSCs cryopreserved by vitri-
fication. Acta Biomater 33:264–274
101. Khosla K, Zhan L, Bhati A etal (2019) Characterization of laser
gold nanowarming: a platform for millimeter-scale cryopreserva-
tion. Langmuir 35(23):7364–7375
102. Panhwar F, Chen ZR, Hossain SMC etal (2018) Near-infra-
red laser mediated modulation of ice crystallization by two-
dimensional nanosheets enables high-survival recovery of
biological cells from cryogenic temperatures. Nanoscale
10(25):11760–11774
103. Swioklo S, Constantinescu A, Connon CJ (2016) Alginate-encap-
sulation for the improved hypothermic preservation of human
adipose-derived stem cells. Stem Cells Transl Med 5(3):339–349
104. Swioklo S, Ding P, Pacek AW etal (2017) Process parameters
for the high-scale production of alginate-encapsulated stem cells
for storage and distribution throughout the cell therapy supply
chain. Process Biochem 59:289–296
105. Bissoyi A, Nayak B, Pramanik K etal (2014) Targeting cry-
opreservation-induced cell death: a review. Biopreservation
Biobanking 12(1):23–34
106. Baust JG, Snyder KK, van Buskirk R etal (2017) Integrating
molecular control to improve cryopreservation outcome. Bio-
preservation Biobanking 15(2):134–141
Dr. Lei Zhang is a professor in
Department of Biochemical
Engineering, School of Chemical
Engineering and Technology,
Tianjin University. He received
his Ph.D. degree in chemical
engineering from University of
Washington in 2012. He was a
postdoctoral fellow at the Univer-
sity of Washington between 2012
and 2013. His research interests
are mainly focused on the devel-
opment of polymer/protein based
functional biomaterials, preser-
vation of living cells/tissues, and
marine anti-biofouling coatings.
... 4 At present, dimethyl sulfoxide (DMSO) and glycerol are the most widely used cell permeable, small-molecule cryoprotectant agents (CPAs) that decrease the freezing point and reduce intracellular ice formation by hydrogen bonding. 3,4 However, these small molecules interfere with cellular functions, are often toxic at the concentrations required for the cryoprotection, and must be fully removed before cell propagation and for their applications in tissue engineering and regenerative medicine. 1−6 To address these issues, various polyols 1−6 such as, trehalose, 2,9,10 dextran, 4 poly(vinyl alcohol) (PVA), 6,10,11 poly(ethylene glycol) (PEG), 7 and ionic liquids 12 are tested for their IRI activities and cryopreservation efficacies. ...
... However, most polyols are cell impermeable and require supplementation with low concentrations of membranepermeating agents to achieve good post-thaw cell survival. 2,3 Furthermore, extracellular cryoprotectants typically work at high concentrations (up to 20 wt %), leading to viscous solutions that are not easy to process and must be supplemented with other IRI-active cryoprotectants such as DMSO, glycerol, mannitol, or trehalose to maintain their antifreeze activities. 13 In an effort to overcome these issues, modification of polyols with cell permeating agents or their intracellular accumulation by physical methods have been found to improve the cell survival post-freeze−thaw (FT) process. ...
... 19−21 Nanocellulose, PEG-functionalized self-assembled structures, and PVA-functionalized nanoparticles are a handful examples of nanostructures that are recently explored for their IRI activities; however, their cryopreservation efficacies are not always studied. 3,19,22 In comparison to linear polymers, new platforms of nanodimensions are interesting for improved IRI activities and have the potential to overcome the issues of high viscosities associated with linear polymers during the cryopreservation process. In this study, we explore the synthesis, IRI activities, and cryopreservation efficacies of vitamin B5-analogous methacrylamide (B5AMA) nanogels, and their cryogenic behavior is compared with the linear poly(B5AMA). ...
... Our analyses showed a close phylosymbiosis of M. silvanus and A. flavithermus with their hosts strongly suggesting the development of a stable and intimate and mutualistic relationship, which allowed the endemic Antarctic polychaetes and their microbiome to better adapt to the freezing temperatures (Fig. 6). The findings of this study open wider perspectives on the comprehension of the mechanisms of adaptation and cryo-resistance of marine invertebrates mediated by associated bacteria and pave the way for the development of biotechnological applications of the enzymes and molecules produced by Meiothermus and Anoxybacillus for cryo-preservations tools (73). ...
Article
Full-text available
The microbiome plays a key role in the health of all metazoans. Whether and how the microbiome favors the adaptation processes of organisms to extreme conditions, such as those of Antarctica, which are incompatible with most metazoans, is still unknown. We investigated the microbiome of three endemic and widespread species of Antarctic polychaetes: Leitoscoloplos geminus , Aphelochaeta palmeri , and Aglaophamus trissophyllus . We report here that these invertebrates contain a stable bacterial core dominated by Meiothermus and Anoxybacillus , equipped with a versatile genetic makeup and a unique portfolio of proteins useful for coping with extremely cold conditions as revealed by pangenomic and metaproteomic analyses. The close phylosymbiosis between Meiothermus and Anoxybacillus and these Antarctic polychaetes indicates a connection with their hosts that started in the past to support holobiont adaptation to the Antarctic Ocean. The wide suite of bacterial cryoprotective proteins found in Antarctic polychaetes may be useful for the development of nature-based biotechnological applications.
... Second, the ice can directly cause mechanical injury to cells. 12,13 Furthermore, the overproduction of reactive oxygen species (ROS) induced by low temperature can cause lipid peroxidation, DNA damage, and protein oxidation. [14][15][16][17] To decrease cryoinjuries, cryoprotectants (CPAs) should be used in the cryopreservation process. ...
Article
Full-text available
The transfusion of red blood cells (RBCs) is crucial in current medicinal research. The shelf‐life of donated RBCs preserved by the normal method is very short, limiting their clinical application. Cryopreservation is a reliable and effective technology for the long‐term storage of RBCs. During the process, ice formation will cause irreversible injuries to RBCs. Glycerol is used as a cryoprotectant (CPA) for RBCs to mitigate cryoinjuries. But it severely induces RBC hemolysis and deformation. This review introduces some biocompatible and effective CPAs used for RBC cryopreservation, outlining recent advances. Currently, there is no uniform standard to determine whether a CPA is suitable for RBCs. According to previous studies, we summarize for the first time comprehensive methods to evaluate the performance of CPAs by ensuring their safety and effectiveness. The safety of CPAs is defined as the degree of damage to RBCs, while the effectiveness is demonstrated by the properties of thawed RBCs, including membrane properties, protein activities, rheological properties, and metabolites levels. A novel CPA that is confirmed suitable for RBCs by these methods may be applied to other cells. We believe comprehensive methods can thoroughly evaluate the performance of CPAs and promote the development of transfusion therapy in clinic.
Article
The degradation of scaffolds is a critical aspect in tissue engineering and regenerative medicine. Various factors that influence the degradation of scaffold are viscosity, density, hydrophilicity, temperature, cells and degradation environment. In present study, the degradation of scaffold were evaluated in presence of perfused media, Red blood cells (RBC), and cryogenic conditions. Different properties such as density, viscosity and contact angle of bio-ink used for fabrication of scaffold were also evaluated and their correlation to desired environment conditions were also evaluated. A poly electrolyte complex scaffolds (chitosan: alginate, chitosan: gelatine), and explicit scaffolds were considered for degradation study. Explicit scaffold and PEC scaffolds were fabricated form chitosan, alginate and gelatine at mixing ratio of 20:80, 50:50, and 80:20 concentration by solvent casting and etching method. The scaffold degradation studies in perfused media were performed at flow rate of 0.5 ml/min, 1 ml/min and 2 ml/min. The parameters related to degradation of scaffold such as weight and pH of discharge media were also evaluated. Results indicated that degradation of chitosan: alginate at 20:80 concentration had shown maximum degradation in presence of RBCs. Similarly, degradation pattern was equal to linear patten for all the scaffolds except 20:80 and 50:50 concentration of chitosan-alginate in cryogenic conditions. Result of scaffolds degradation in perfused media had shown that scaffolds were degraded maximal at a flow rate of 1 ml/min as compare to other flow rates. Contact angle measurement had proved that all PEC scaffold belongs to the category of hydrophilic family. The scaffold fabricated from higher concentration of chitosan had revealed slower degradation as compare to others configuration. The acceleration of scaffold degradation was tiggered by enzymatic and cellular activities in the presence of RBCs. Perfused media had caused scaffold degradation due to presence of mechanical shear stress.
Article
Assisted reproductive technology (ART) is a commonly used treatment for infertile couples. However, the suboptimal success rate of ART highlights the urgent need for optimization. One promising strategy to significantly improve ART outcomes is to standardize and automate all steps involved, ranging from high-quality gamete selection to in vitro fertilization, embryo culture, embryo examination/selection, and gamete/embryo cryopreservation. Cutting-edge technologies, such as microfluidics and artificial intelligence (AI), have been developed for this purpose and have demonstrated remarkable potential in this area. Microfluidic systems have the potential to integrate ART steps into a miniaturized and automated device, which can minimize human error, save time and costs, enhance reproducibility and consistency, and improve accuracy. Additionally, AI is playing an increasingly important role in automating key aspects of ART. Leveraging its powerful abilities in rapid, accurate, and intelligent analysis of large biomedical datasets, AI assists doctors and embryologists in making more informed and effective decisions. In summary, these bioengineering technologies have the potential to revolutionize ART, making it faster, more efficient, and ultimately more effective for couples seeking to conceive. This article provides a comprehensive overview of how microfluidics and AI are being applied in ART to advance the field and benefit patients.
Article
Full-text available
Conventional hydrogel microcapsules often suffer from inadequate mechanical stability, hindering their use. Here, water‐cored double‐network (DN) hydrogel shells are designed, formed by polyacrylamide and calcium alginate networks using triple‐emulsion templates. These DN hydrogel shells offer robust mechanical stability, optical transparency, and a precisely‐defined cut‐off threshold. The feasibility of this platform is demonstrated through the development of a fluorometric glucose sensor. Glucose oxidase is enclosed within the water core, while a pH‐responsive fluorescent dye is incorporated into the DN shells. Glucose diffuses into the core through the DN shells, where the glucose oxidase converts glucose into gluconic acid, leading to pH reduction and a subsequent decrease in fluorescence intensity of DN shells. Additionally, the pH‐sensitive colorant dissolved in the medium enables visual pH assessment. Thus, glucose levels can be determined using both fluorometric and colorimetric methods. Notably, the DN shells exhibit exceptional stability, enduring intense mechanical stress and cycles of drying and rehydration without leakage. Moreover, the DN shells act as effective barriers, safeguarding glucose oxidase against proteolysis by large disruptive proteins, like pancreatin. This versatile DN shell platform extends beyond glucose oxidase encapsulation, serving as a foundation for various capsule sensors utilizing enzymes and heterogeneous catalysts.
Article
Beyond single cell two‐dimensional (2D) culture, research on organoids that can mimic human organs is rapidly developing. However, there are still problems in commercialization and joint research using organoids due to the lack of technology to safely store organoids. Since organoids are 3D complex structures with a certain size (0.1–5 mm) beyond the size of cells, the conventional cell‐level cryopreservation method using cryoprotectant (CPA) cannot overcome the damage caused by volume change due to osmotic pressure difference and ice nucleation. Herein, we attempted to solve such limitations by applying a nanowarming system using CPA with high cell permeability and Fe 3 O 4 nanoparticles. By performing beat rate measurement, histological analysis, contractility analysis, and multi‐electrode array, it was verified that the developed method could significantly improve functional recovery and survival of heart organoids after freezing and thawing. In this study, we demonstrated a successful organoid cryopreservation method based on a Fe 3 O 4 nanowarming system. The developed technology will provide clues to the field of tissue cryopreservation and spur the application of organoids.
Article
Full-text available
Developing cell cryo-preservation methods on chips is not only crucial for biomedical science but also represents an innovative approach for preserving traditional cell samples. This study presents a simple method for direct cell freezing and thawing on chip, allowing for long-term storage of cells. During the freezing process, cells were injected into the microchannel along with a conventional cell cryopreservation solution, and the chip was packed using a self-sealing bag containing isopropyl alcohol and then stored in a -80 °C refrigerator until needed. During the thawing process, microcolumn arrays with a spacing of 8 μm were strategically incorporated into the microfluidic chip design to effectively inhibit cells from the channel. The breast cancer cell lines MDA-MB-231 and B47D demonstrated successful thawing and growth after cryopreservation for one month to one year. These findings offer a direct cell freezing and thawing method on a microfluidic chip for subsequent experiments.
Article
Full-text available
The successful exploitation of human pluripotent stem cells (hPSCs) for research, translational or commercial reasons requires the implementation of a simple and efficient cryopreservation method. Cryopreservation is usually performed with dimethylsulphoxide (DMSO), in addition to animal proteins. However, even at sub-toxic levels, DMSO diminishes the pluripotency capacity of hPSCs and affects their epigenetic system by acting on the three DNA methyltransferases (Dnmts) and histone modification enzymes. Our study aimed to test trehalose-based cryosolutions containing ethylene glycol (EG) or glycerol (GLY) on hESCs RC17, hiPSCs CTR2#6 and long-term neuroepithelial-like stem cells (lt-NES) AF22. Here, we demostrate the effectiveness of these cryosolutions in hPSCs by showing an acceptable rate of cell viability and high stability compared to standard 10% DMSO freezing medium (CS10). All cell lines retained their morphology, self renewal potential and pluripotency, and none of the cryosolutions affected their differentiation potential. Genotoxicity varied among different stem cells types, while trehalose-based cryopreservation did not sensibly alter the homeostasis of endoplasmic reticulum (ER). This study provides evidence that pluripotent and neural stem cells stored in trehalose alone or with other cryoprotectants (CPAs) maintain their functional properties, indicating their potential use in cell therapies if produced in good manufacturing practice (GMP) facility.
Article
Full-text available
Over the last decade and half, the optimization of cryopreservation for adipose tissue derived stromal/stem cells (ASCs) especially in determining the optimal combination of cryoprotectant type, cooling rate, and thawing rate have been extensively studied. In this study, we examined the functionality of ASCs that have been frozen-stored for more than 10 years denoted as long-term freezing, frozen within the last 3 to 7 years denoted as short-term freezing and compared their response with fresh ASCs. The mean post-thaw viability for long-term frozen group was 78% whereas for short-term frozen group 79% with no significant differences between the two groups. The flow cytometry evaluation of stromal surface markers, CD29, CD90, CD105, CD44, and CD73 indicated the expression (above 95%) in passages P1-P4 in all of the frozen-thawed ASC groups and fresh ASCs whereas the hematopoietic markers CD31, CD34, CD45, and CD146 were expressed extremely low (below 2%) within both the frozen-thawed and fresh cell groups. Quantitative real time polymerase chain reaction (qPCR) analysis revealed some differences between the osteogenic gene expression of long-term frozen group in comparison to fresh ASCs. Intriguingly, one group of cells from the short-term frozen group exhibited remarkably higher expression of osteogenic genes in comparison to fresh ASCs. The adipogenic differentiation potential remained virtually unchanged between all of the frozen-thawed groups and the fresh ASCs. Long-term cryopreservation of ASCs, in general, has a somewhat negative impact on the osteogenic potential of ASCs, especially as it relates to the decrease in osteopontin gene expression but not significantly so with respect to RUNX2 and osteonectin gene expressions. However, the adipogenic potential, post thaw viability, and immunophenotype characteristics remain relatively intact between all the groups.
Article
Significance Cryopreservation is routinely used for the long-term storage of cells in various areas of academic, industrial, and clinical research. To keep frozen cells alive, it is necessary to vitrify (or nanocrystallize) water on the inside and outside of the cells. Vitrification is conventionally achieved by adding at least one cryoprotective agent (CPA) to the medium. However, CPAs should ideally be avoided due to their cytotoxicity and potential side effects on the cells. Herein, we demonstrate a method of CPA-free cryopreservation, in which cells are almost vitrified by ultrarapid cooling using inkjet printing. The freezing method should be generally suitable for all kinds of cells that are susceptible to CPAs, including stem cells.
Article
Cryopreservation of red blood cells (RBCs) is fundamentally important to modern transfusion medicine. Currently, organic solvent glycerol is utilized as the state-of-the-art cryoprotectant (CPA) for RBC cryopreservation. However, glycerol must be removed before RBC transfusion to avoid intravascular hemolysis, via a time-consuming deglycerolization process with a specialized equipment (e.g. ACP215), thus limiting the clinical use of frozen RBCs. Herein, we report novel biocompatible CPA formulations combining zwitterionic betaine with membrane stabilizers (disaccharides or amino acids), which can achieve outstanding efficiency for RBC cryopreservation directly using whole blood without any separation process. Most importantly, due to the osmotic regulation capacity of betaine, a simple and fast one-step method can be used for CPA removal, which is significantly superior to current multi-step deglycerolization process. This work offers a promising solution for highly efficient and solvent-free RBC cryopreservation, and holds great potential to improve the long-term storage and long-distance distribution of RBCs.
Article
Loss of hepatocyte viability and metabolic function after cryopreservation is still a major issue. Although vitrification is a promising alternative, it has generally been proven to be unsuitable for vitrification of large cell volumes which is required for clinical applications. Here we propose a novel bulk droplet (3 to 5 mm diameter) vitrification method which allows high throughput volumes (4 ml/min), while using a low pre-incubated CPA concentration (15% v/v) to minimize toxicity and loss of cell viability and function. We used rapid (1.25 s) osmotic dehydration in order to concentrate a low pre-incubated intracellular CPA concentration ahead of vitrification, without the need of fully equilibrating toxic CPA concentrations. We compared direct post-preservation viability, long-term viability and metabolic function of bulk droplet vitrified, cryopreserved and fresh hepatocytes. Simulations and cooling rate measurements confirmed an adequate concentration of the intracellular CPA concentration (up to 8.53 M) after dehydration in combination with high cooling rates (960 to 1320°C/min) for successful vitrification. Compared to cryopreserved hepatocytes, bulk droplet vitrified hepatocytes had a significantly higher viability, directly after preservation and after one day in culture. Moreover, bulk droplet vitrified hepatocytes had evidently better morphology and showed significantly higher metabolic activity than cryopreserved hepatocytes in long term collagen sandwich cultures. In conclusion, we developed a novel bulk droplet vitrification method of which we validated the theoretical background and demonstrated the feasibility to use this method to vitrify large cell volumes. Moreover, we showed that this method results in improved hepatocyte viability and metabolic function as compared to cryopreservation.
Article
BACKGROUND Dendritic cell (DC) vaccination involves administration of multiple doses. Cryopreservation of tumor antigen–pulsed DCs can provide a ready to use vaccine source and eliminate the need of frequent withdrawal of the patient's blood for vaccine preparation. The aim of this study was to assess the effect of addition of trehalose in the freezing medium on the recovery of DCs after cryopreservation. STUDY DESIGN AND METHODS DCs were generated from mononuclear cells from apheresis samples of healthy donors. For long‐term storage of 6 months, cells were frozen with a rate‐controlled programmable freezer and stored in liquid nitrogen. For short‐term storage of 1 month, cells were frozen and stored at −80°C. DCs frozen with Iscove's Modified Dulbecco's Medium + 10% dimethyl sulfoxide + 20% fetal bovine serum served as the control group, while the test group was additionally supplemented with 50 μg/mL of trehalose. After revival of control and test DCs, they were assessed for viability, morphology, phenotype, and functions. RESULTS The addition of trehalose to the conventional freezing medium helped to preserve the viability and functionality of DCs better than dimethyl sulfoxide alone in both long‐ and short‐term cryopreservation. Trehalose also protected the mitochondrial membrane potential and cytoskeleton integrity of DCs, which are necessary for their functionality. Mediators of the intrinsic apoptotic pathway like Caspase‐9 and Bim‐1 were found to be low in the test. CONCLUSION Supplementation of conventional freezing medium with trehalose results in better quality of DCs revived after cryopreservation. This finding could help improve DC vaccine preparation for cancer immunotherapy.
Article
Preventing ice formation during cryopreservation by vitrification has led to the successful storage and banking of numerous cellular and tissue-based biomaterials. In their breakthrough work, Peter Mazur’s group achieved over 90% survival by using a laser warming technique for 100 µm mice oocytes that were cooled in 0.1 µL droplets with 2.3M CPA and extracellularly loaded India Ink (laser absorber). Laser warming can provide rapid and uniform warming rates to “outrun” damaging ice crystal growth. Here we generalize Mazur’s technique for microliter sized droplets using laser nanowarming to rewarm millimeter-scale biomaterials when loaded extracellularly and/or intracellularly with biocompatible 1064 nm resonant gold nanoparticles. We first show that droplets containing low concentration cryoprotectants (such as 2M Propylene Glycol +/- 1M Trehalose) can be rapidly cooled at rates up to 90,000 °C/min by plunging into liquid nitrogen to achieve either a visually transparent (i.e. vitrified) or a cloudy with ice (i.e. non-vitrified) state. Both modeling and experiments were then used to characterize the process for different laser energy (2-6J), pulse length (1-20ms), droplet volume (0.2-1.8µL), cryoprotectant (2-3M) and gold concentration (0.77-4.8 x1017 nps/m3) to assess physical and biological success. Physical success was achieved by finding conditions that minimize cloudiness and white spots within the droplets during cooling and warming as signs of damaging ice formation and ice crystallization respectively. Biological success was achieved using human dermal fibroblasts to find conditions that achieve ≥ 90 % cell viability normalized to controls post warming. Thus, physical and biological success can be achieved using this platform cryopreservation approach of cryotop cooling and laser gold nanowarming in mm scale systems.
Article
Advances in stem cell technology and regenerative medicine have underscored the need for effective baking of living cells. Cryopreservation, using very low temperatures to achieve suspended animation, is widely used to store or bank cells for later use. This process requires the use of cryoprotective agents (CPAs) to protect cells against damage caused by the freezing and thawing process. However, current popular CPAs like DMSO can be toxic to cells and must be thoroughly removed from cells before they can be used for research or clinical applications. Trehalose, a nontoxic sugar found in organisms capable of withstanding extreme cold or desiccation, has been explored as an alternative CPA. The disaccharide must be present on both sides of the cellular membrane to provide freezing protection. However, trehalose is not synthesized by mammalian cells nor has the capability to diffuse through their plasma membranes. Therefore, it is crucial to achieve intracellular delivery of trehalose for utilizing the full potential of the sugar for cell banking. In this review, various methods that have been explored to deliver trehalose into mammalian cells for their banking at both cryogenic and ambient temperatures are surveyed. Among them, the nanoparticle-mediated approach is particularly exciting. Collectively, studies in the literature demonstrate the great potential of using trehalose as the sole CPA for cell banking, to facilitate the widespread use of living cells in modern medicine.
Article
Over the past few decades, the use of cell microencapsulation technology has been promoted for a wide range of applications as sustained drug delivery systems or as cells containing biosystems for regenerative medicine. However, difficulty in their preservation and storage has limited their availability to healthcare centers. Because the preservation in cryogenic temperatures poses many biological and biophysical challenges and that the technology has not been well understood, the slow cooling cryopreservation, which is the most used technique worldwide, has not given full measure of its full potential application yet. This review will discuss the different steps that should be understood and taken into account to preserve microencapsulated cells by slow freezing in a successful and simple manner. Moreover, it will review the slow freezing preservation of alginate-based microencapsulated cells and discuss some recommendations that the research community may pursue to optimize the preservation of microencapsulated cells, enabling the therapy translate from bench to the clinic.