You are on page 1of 15

pubs.acs.

org/journal/abseba Review

Trehalose in Biomedical Cryopreservation−Properties, Mechanisms,


Delivery Methods, Applications, Benefits, and Problems
Yuying Hu, Xiangjian Liu, Fenglin Liu, Jingxian Xie, Qubo Zhu, and Songwen Tan*

Cite This: ACS Biomater. Sci. Eng. 2023, 9, 1190−1204 Read Online

ACCESS Metrics & More Article Recommendations


See https://pubs.acs.org/sharingguidelines for options on how to legitimately share published articles.
Downloaded via CINVESTAV on March 6, 2024 at 20:30:16 (UTC).

ABSTRACT: Cells and tissues are the foundation of translational medicine. At present, one of the main technological obstacles is
their preservation for long-term usage while maintaining adequate viability and function. Optimized storage techniques must be
developed to make them safer to use in the clinic. Cryopreservation is the most common long-term preservation method to maintain
the vitality and function of cells and tissues. But, the formation of ice crystals in cells and tissues is considered to be the main
mechanism that could harm cells and tissues during freezing and thawing. To reduce the formation of ice crystals, cryoprotective
agents (CPAs) must be added to the cells and tissues to achieve the cryoprotective effect. However, conventional cryopreservation of
cells and tissues often needs to use toxic organic solvents as CPAs. As a result, cryopreserved cells and tissues may need to go
through a time-consuming washing process to remove CPAs for further applications in translational medicine, and multiple valuable
cells are potentially lost or killed. Currently, trehalose has been researched as a nontoxic CPA due to its cryoprotective ability and
stability during cryopreservation. Nevertheless, trehalose is a nonpermeable CPA, and the lack of an effective intracellular trehalose
delivery method has become the main obstacle to its use in cryopreservation. This article illustrated the properties, mechanisms,
delivery methods, and applications of trehalose, summarized the benefits and limits of trehalose, and summed up the findings and
research direction of trehalose in biomedical cryopreservation.
KEYWORDS: trehalose, cryopreservation, cryoprotective agents, nontoxic, nonpermeable

1. INTRODUCTION who need urgent treatment or who are ineligible for ovarian
Cells and tissues are essential in translational medicine. Due to stimulation.3 Cryopreservation is defined as the long-term
negative storage effects on morphological and biochemical preservation of individual living cells and biological tissues at
features, the shelf life of red blood cells (RBCs) for transfusion extremely low temperatures, often at −196 °C for liquid
is short (less than 42 days) after donation, resulting in an nitrogen. At this temperature, cells can remain genetically
unwanted waste of 1.7 million units of blood in the U.S.1,2 stable for a very long period until they are required because
Therefore, prolonging RBCs’ storage duration is essential to cellular processes are temporarily stopped. Meanwhile, it is the
their supply. The cryopreservation technology offers a new way best approach for preserving living tissues for a long time
for long-term storage of RBCs. Additionally, ovarian tissue because it is less expensive than other methods.4 However,
(OT) cryopreservation can effectively preserve hundreds of
primordial follicles simultaneously. OT is carefully and slowly Received: October 18, 2022
frozen to −140 °C and is then kept in liquid nitrogen at −196 Accepted: February 1, 2023
°C. During this process, no serious tissue deformation is Published: February 13, 2023
observed. Nowadays, ovarian tissue cryopreservation and
transplantation (OTC-T) offers a promising method for
preserving fertility, especially for postpubertal cancer patients

© 2023 American Chemical Society https://doi.org/10.1021/acsbiomaterials.2c01225


1190 ACS Biomater. Sci. Eng. 2023, 9, 1190−1204
ACS Biomaterials Science & Engineering pubs.acs.org/journal/abseba Review

Table 1. Structure, Mechanism, and Application of Typical CPAsa,b,15,16,32−63

a
The structure of α,α-1,1-trehalose.64 bThe diagram shows the schematic structure of HES.15

1191 https://doi.org/10.1021/acsbiomaterials.2c01225
ACS Biomater. Sci. Eng. 2023, 9, 1190−1204
ACS Biomaterials Science & Engineering pubs.acs.org/journal/abseba Review

cells and tissues are vulnerable to damage in the freezing Table 2. Properties of Trehalose
environment. Mazur et al.5 proposed that freezing of water
property result
damages cells in two different ways: Slow cooling rates will
expose the cells to high solute concentrations and cause chemical formula C12H22O11
osmotic damage. But, if the cooling rate is too fast, intracellular appearance white, odorless powder
ice induced by the trapped water will lead to mechanical sweetness (relative) 45% (as compared to that of sucrose)
damage to the cells. These two damages restrict the toxicity nontoxic
development of cryopreservation.6 Thus, cryoprotectants molecular weight 342.30 g/mol
(CPAs) must be added to cells and tissues to minimize relative density 1.22 g/cm3 (at 25 °C and weight fraction of
0.5)
freezing damage in cryopreservation.7 There are two kinds of
glass transition 117 °C
CPAs according to their functions and mechanisms: permeable temperature
and impermeable.8 The former are mostly small molecules, freezing temperature −197 °C (100 mg/mL water)
which penetrate into the cells to replace intracellular water and chemical nature nonreducing disaccharide
ultimately prevent cell damage caused by intracellular ice chemical nomenclature α-D-glucopyranosyl-α-D-glucopyranoside
formation (IIF) and excessive cell shrinkage.9 In addition, they three possible anomers α,β-trehalose, β,β-trehalose, α,α-trehalose
can get inside cells to adjust osmotic pressure and decrease
osmotic damage.10 Permeable glycerol,11 dimethyl sulfoxide
(DMSO),12 and ethylene glycol (EG)13 fall under this 3. MECHANISMS OF CRYOPROTECTIVE EFFECTS OF
category. The latter act in extracellular to work by promoting TREHALOSE
cell dehydration to inhibit the growth of intracellular ice and Trehalose is a nonpermeable CPA, whose high molecular
also protect cells from extracellular ice crystals’ damage,9 weight limits its permeability.72 Although the effects of
including trehalose,14 hydroxyethyl starch (HES),15 polyam- trehalose on biological molecules in cryopreservation have
pholytes,16 poly(vinyl)alcohol (PVA),17 and so forth. The been extensively studied, mechanisms of cryoprotective effects
properties, functions, and mechanisms of these typical CPAs of trehalose are extremely complex, and there are contradictory
are described in more detail (listed in Table 1). conclusions in different articles. The hypotheses about the
However, conventional cryopreservation of cells and tissues main cryopreservation mechanisms of trehalose are summar-
needs to utilize toxic organic solvents (such as DMSO18 and ized as follows:
glycerol19) as CPAs. Therefore, cryopreserved cells and tissues 3.1. Preferential Exclusion Theory. The cellular
have to withstand a cumbersome washing process to eliminate protective effect of trehalose is attributable to its preferential
them for further applications. Moreover, this process may repellency.73 According to this theory, trehalose does not
cause osmotic shock, resulting in cellular damage.20,21 directly interact with biological macromolecules. It means that
Conversely, trehalose, a nontoxic, nonpermeable CPA, can water is ordered around trehalose rather than included in the
replace the traditional CPAs that are highly toxic to cells and biomolecules’ solvation layer. Trehalose and biomolecules
tissues.22 Additionally, the trehalose-cryopreserved cells can be compete for the available water as the concentration of
employed without being washed, which should make cell-based trehalose in a large volume of water increases. This
treatment more widely available.23 At present, trehalose has competition leads to water molecules being destructured
been efficiently used for cryopreservation of murine spermato- surrounding biomolecules and “structured” around trehalose,
logical,24 adult hematopoietic,25,26 erythrocytes,27 human which makes it become a kosmotrope or water structure-
adipose tissue-derived mesenchymal stromal cells,28 and maker.74,75 This indicates that trehalose and water interact
human transplantable hepatocytes.29 Additionally, it has also considerably more strongly than water and water does.71
been applied to human pancreatic islets30 and fetal skin.31 Furthermore, the hydrated radius of trehalose is exceedingly
This article gives a thorough overview of the recent studies large, and nuclear magnetic resonance (NMR) relaxation time
on the use of trehalose in cryopreservation. It includes measurements indicate that trehalose has the highest hydration
probable mechanisms, delivery methods, applications, benefits capacity of all investigated saccharides.76,77 The water
and limits, and the related findings and research direction of molecules are not uniformly distributed around the trehalose,
trehalose in biomedical cryopreservation in detail. We hope but trehalose can manipulate the water structure to become
this review can help researchers fully understand the relevant organized around it in an oriented manner, with hydrogen
knowledge of trehalose in cryopreservation and promote the bonds in all orientations, which makes it possible for trehalose
development of translational medicine. to be involved in the bioprotection of cells. Moreover, a large
number of experiments, such as infrared and Raman spectros-
2. PROPERTIES OF TREHALOSE copies, NMR, ultrasound measurements,78,79 and inelastic
Trehalose is a nonreducing disaccharide that is found in plants, neutron scattering (INS) measurement,80 demonstrated the
animals, microcells, and tissues.65 But, it is not found in destruction of the tetrahedral network of water molecules after
mammals.66 Although there are three possible anomers of adding trehalose, thus hindering the crystallization of ice
trehalose, namely, α,β-1,1-, β,β-1,1-, and α,α-1,1-, only the α,α- efficiently.
trehalose, which is commonly known as trehalose, has been For the protection ability of proteins, the addition of
isolated from and biosynthesized in living cells and tissues.67 trehalose in solution keeps water molecules away from
Besides, trehalose comprises two glucopyranosyl units linked proteins, reducing their hydrated radius and improving their
by α-1,1-glycosidic bonds.68 And, the molecular formula of compactness and stability to effectively prevent protein
trehalose is C12H22O11.69 molecules from denaturation. These properties make trehalose
Trehalose can be cleaved into two constituent D-glucose become one of the best CPAs (Figure 1A).74 It is especially
units by trehalase, and its nature is very stable.70 Some noteworthy that trehalose stabilizes the protein’s partially
properties of it are listed in Table 2.67,71 unfolded state rather than its native structure.81
1192 https://doi.org/10.1021/acsbiomaterials.2c01225
ACS Biomater. Sci. Eng. 2023, 9, 1190−1204
ACS Biomaterials Science & Engineering pubs.acs.org/journal/abseba Review

Figure 2. Illustration of the mechanism of lipid bilayer’ stabilization in


the freezing state by trehalose.

3.4. Active Protein Stabilizer. Trehalose is an active


Figure 1. Illustration of the mechanisms of various theories of protein stabilizer.92,93 During cryopreservation, it is speculated
trehalose on proteins during cryopreservation. (A) Preferential that trehalose can prevent the inactivation and aggregation of
exclusion theory. (B) Vitrification theory. (C) Water replacement proteins at low temperatures.67,72 All the three hypotheses
theory. above seem to explain the mechanism of protein protection by
trehalose. At the same time, the biological protection provided
3.2. Vitrification Theory. Vitrification is the solidification by trehalose is not only to stabilize proteins but also to
process from liquid to glass.70 Trehalose has a strong capacity nonspecifically stabilize intracellular nucleic acid and other
for intermolecular interactions, making it easy to build cluster biological macromolecules, which leads to reducing the
structures that can contain glass phase and have a modest appearance of intracellular ice during freezing.71,94 Because
impact on neighboring and remote water molecules.82 trehalose acts on different types of biomolecular structures, the
Meanwhile, trehalose has excellent glass-forming properties protective mechanism of trehalose in cryopreservation is
because of its different polymorphic forms.71 Besides, trehalose described as a nonspecific process.
is superior to other sugars for preserving cells and tissues in 3.5. Increase of Membrane Permeability. Trehalose
certain circumstances because of its highest glass transition protects cells by increasing the permeability of membranes to
temperature among all the disaccharides.83−85 And, the glass water at subzero temperatures and decreasing the amount of
transition temperature of the freeze-concentrated trehalose cellular water before freezing, thus reducing the likelihood of
solution is in the vicinity of −30 °C.31 Therefore, the solution intracellular ice during freezing and preventing excessive
viscosity of trehalose rapidly increases during the freezing swelling and osmotic shock.31,72,95
process to form a stable glass matrix that acts like a cocoon, To sum up, trehalose works by modulating osmotic stress
which is another benefit as an increase in viscosity might lead and inhibiting ice formation.96 Various experimental and
to reduced crystal growth. These benefits do not damage the theoretical studies have revealed that the mechanisms of the
cell structure and protect the proteins and other biomolecules cryoprotective effects of trehalose on cells and tissues are
(Figure 1B).72,86 complicated, and none of them can be fully accepted since they
3.3. Water Replacement Theory. Water replacement sometimes lead to contradictory conclusions. At the same time,
theory refers to the direct interaction of trehalose molecules the stability of trehalose under various circumstances can be
with the protected biological structures via hydrogen bonds.87 explained by any one or all of these theories. They are not
This hypothesis could explain the protective effect of trehalose necessarily mutually exclusive and may be unified. Therefore,
on proteins and other bioactive macromolecules. The there is no doubt that we need to analyze the mechanism of
protective membrane formed by hydrogen bonding between trehalose under different conditions using experiments and
trehalose molecules and proteins replaces the necessary water theoretical foundations.
molecules to maintain the three-dimensional structure during
the freezing process, allowing the biomolecules to retain the 4. DELIVERY METHODS OF TREHALOSE DURING
integrity of cell membrane structure and function at very low CRYOPRESERVATION
moisture levels to avoid the loss of intracellular components Trehalose is a particularly attractive CPA because of its well-
(Figure 1C). Meanwhile, trehalose is a membrane stabilizer.88 known role in keeping cells’ structure and protecting cells from
During cryopreservation, trehalose and phospholipid polar various damages in cryopreservation.73 Trehalose must be
groups could replace the lost structural water by hydrogen present in both intracellular and extracellular cells to supply
bonding. By delaying the onset of the phase transition from a optimal protection against stress and damage during
liquid crystal to a gel state, the membranes are prevented from cryopreservation.97 Whereas, trehalose is difficult to transport
approaching each other, thus inhibiting membrane fusion to across the cell membrane, so its cryoprotective activity is
stabilize the cell membrane whose fluidity declines during limited.98,99 Consequently, the current research focuses on the
temperature downshift (Figure 2).74,85,89,90 However, some delivery methods of trehalose into cells. Janis et al.100 provided
previous studies have indicated that the water replacement evaluation criteria for new delivery methods: (1) the
theory is unable to explain the cryoprotective properties of efficiencies of loading trehalose to RBCs are comparable to
trehalose.47,91 classic methods, including but not limited to electroporation
1193 https://doi.org/10.1021/acsbiomaterials.2c01225
ACS Biomater. Sci. Eng. 2023, 9, 1190−1204
ACS Biomaterials Science & Engineering pubs.acs.org/journal/abseba Review

and fluid-phase endocytosis and (2) Avoiding cellular However, they are invasive delivery methods and may seriously
pressures that from high trehalose concentrations and long damage cells, causing further freezing damages during
incubation time. The criteria are important for us to find cryopreservation.22
promising new delivery methods for trehalose-loaded cells. 4.3. Microinjection. Exogenous trehalose can be injected
4.1. Phospholipid Phase Transition. One of the into the cytoplasm of mammalian cells by microinjection.
methods of introducing trehalose into mammalian cells is to Eroglu et al.106 showed that the microinjection technology
enhance the permeability of cell membranes by inducing allowed for the introduction of trehalose into oocytes in a
phospholipid phase changes.30 So far, thermal, osmotic, and controllable manner by the volumetric response. And,
electrical stress have been investigated as induction methods to trehalose was a unique CPA for oocytes, which quickly
improve membrane permeability and permit trehalose to disappeared during embryonic development.107 Obviously,
deliver into cells.101 trehalose has been transported into mammalian oocytes with
Beattie et al.30 used the lipid phase transition of islet cell the size of ∼100 μm in diameter and the number of dozens or
membranes to introduce trehalose into the cells before freezing hundreds by using this method. However, it is often hard to
with a high concentration of trehalose outside. Furthermore, apply for smaller cells (usually less than 20 μm in diameter) or
when islet cells underwent a thermogenic lipid phase massive needs (usually millions to billions) of mammalian
transition, membrane permeability would increase. It allowed cells.23,108,109
trehalose to diffuse from the surrounding environment along 4.4. TRET1 Transporter. Researchers have isolated the
its concentration gradient to control the load. trehalose transporter TRET1 from the anhydrobiotic larvae of
Zhang et al.95 have recently shown that endocytic and the African chironomid, which is called Polypedilum
freezing-induced trehalose uptake have been combined for vanderplanki. Uchida et al.110 conducted a comparative
cryopreservation of mammalian cells. The presence of ice cryopreservation experiment between the TRET1-expressing
affected the permeability of water and nonpermeable CHO-K1 cells (CHO-TRET1) and the CHO-K1 cells
molecules. During cryopreservation, membranes became transfected with the empty vector (CHO-vector). The
permeable as a result of a freeze-induced membrane phase conclusion showed that trehalose delivered into the cells
transition and an osmotic driving force, resulting in the with TRET1 dramatically increased the cryoprotective
delivery of trehalose into fibroblasts.102 function. In summary, TRET1 enabled cells to deliver
Satpathy et al.103 proposed a method for loading RBCs with trehalose specifically and effectively across the plasma
trehalose. Through the combination of osmotic imbalance and membrane, but the cells needed to be genetically modified first.
phospholipid phase transition, trehalose from the foreign 4.5. Polymers. Lynch et al.97 have investigated synthetic
medium may be loaded into RBCs, which led to an polymers to help deliver trehalose into the cytoplasm by
approximately 40 mM intracellular trehalose concentration. interacting with the cell membrane and transiently enhancing
And, trehalose was not likely to decompose in RBCs. its permeability. More than 250 mM trehalose was introduced
Therefore, it must be noted that RBCs were exposed to to human RBCs using the membrane-permeabilizing polymer
hypertonic trehalose solution during loading, causing morpho- PP-50, exceeding the estimated bioprotection threshold of
logical changes and possible damages. 100−200 mM intracellular trehalose. PP-50 is noncytotoxic
Electropermeabilization, which is also called electroporation and can be cleared from cell membranes through tiny changes
or electroinjection, has been taken to enhance membrane in pH. And, increased membrane permeability could be
permeability and deliver extracellular molecules into the reversed by washing in PBS.101 Sharp et al.111 and Slater et
cytoplasm.104 Dovgan et al.105 cryopreserved human adipose- al.112 expanded the application of PP-50 from RBCs to a
derived stem cells (hADSCs) by combining reversible nucleated cell line that originated from human osteosarcoma
electroporation with trehalose, which allowed for long-term (SAOS-2). Biopolymer-mediated trehalose uptake could trans-
cryopreservation without DMSO. However, this method is port high-concentration trehalose at relatively low osmolarities
highly invasive and may seriously damage cells, causing further but with a long incubation time.
freezing damages during cryopreservation.22,73 4.6. Liposomes. Liposomes are thought to be potential
The advantage of this method is that the concentration vesicles for delivering trehalose to mammalian cells.113 Stoll et
gradient can be manipulated to control the loading of al.102 found that liposomes could play a role in the
trehalose, and membrane permeability is increased. Never- cryopreservation of RBCs. It is thought that liposomes alter
theless, membrane permeability is nonspecifically permitting cell membranes by exchanging lipid and cholesterol, thereby
the entry of substances other than trehalose, and there are changing the physical properties and making the cells more or
problems about cytotoxicity and membrane stability caused by less stable at low temperatures. Nevertheless, the approach can
thermal, osmotic, and electric shock to cells.101 only load micromolar concentrations of trehalose within cells.
4.2. Engineered Pores. Trehalose can be loaded into Moreover, the release of trehalose in vesicles is inefficient.22,101
mammalian cytoplasm by producing pores on the membranes 4.7. Nanoparticles (NPs). Rao et al.23 synthesized pH
of cells.26 Eroglu et al.98 loaded low concentrations (0.2 M) of responsive genipin-cross-linked Pluronic F127-chitosan nano-
trehalose into 3T3 fibroblasts and human keratinocytes particles (GNPs) and encapsulated trehalose in GNPs to
through exploiting a genetically engineered mutant of Staph- obtain nanoparticles-encapsulated trehalose (nTre). Further-
ylococcus aureus α-hemolysin to create pores in cell membranes, more, GNPs, a vehicle, had superior biocompatibility and were
which could significantly improve the survival rate of cells in taken up by cells. The results proved that trehalose could be
cryopreservation. Moore et al.85 reported that trehalose could effectively encapsulated in GNPs for intracellular transport,
be delivered into an isolated mammalian mitochondrial matrix enabling primary hADSCs to be frozen. As a consequence, it
through the transient opening of the mitochondrial perme- was a noninvasive prospective strategy as it exploited a cell’s
ability transition pore (MPTP). The MPTP is a megapore of natural feeding process called endocytosis to absorb trehalose
the inner membrane composed of ATP synthase dimers. encapsulated in the NPs. Nevertheless, hADSCs were needed
1194 https://doi.org/10.1021/acsbiomaterials.2c01225
ACS Biomater. Sci. Eng. 2023, 9, 1190−1204
ACS Biomaterials Science & Engineering pubs.acs.org/journal/abseba Review

to be incubated with the trehalose-laden NPs for 1 day to Consequently, this method has advantages and disadvan-
slowly release trehalose from the NPs that were absorbed by tages. On the one hand, the method can specifically deliver
cells, which caused a prolonged and tedious cryopreservation large amounts of trehalose into mammalian cells, and no
process. modification to the cells is needed. On the other hand, the
Under normal conditions, trehalose has low permeability to disadvantage of this method is the lengthy incubation period.
RBCs. The mechanism by which apatite NP assists in the 4.8. Fluid-Phase Endocytosis. Fluid-phase endocytosis,
transport of bioactive molecules in cells is usually on the basis an inherent property of cells incorporating membrane-
of cytosolic-mediated translocation. Whereas, endocytosis is nonpermeable molecules, can also be exploited for the
not present in mature mammalian RBCs,114 which implies that introduction of trehalose into cells, resulting in increased
with the assistance of apatite NPs, the transfer of trehalose to intracellular trehalose contents.95 It is a nonspecific process.118
RBCs should occur through different mechanisms, such as This mechanism involves the uptake of some substances from
local modification of RBCs’ physical properties. Stefanic et the environment by the cells and their internalization through
al.115 reported for the first time a glycerol-free cryopreservation a vesicle pinched off from the plasma membrane. This cellular
method that used colloidal, bioinspired apatite NPs as process has been utilized to deliver trehalose into mammalian
bioactive promoters of RBCs’ cryopreservation mediated by cells by simply culturing the cells in the existence of high
trehalose. Instead of through the direct formation of transient extracellular concentrations of trehalose.101 Jong et al.119 used
membrane holes, the translocation of trehalose to RBCs was Paesun cells as experimental subjects. The researchers wanted
promoted by an indirect mechanism resulting from NP/bilayer to see how cryoprotective media affected the cryopreservation
interactions. The findings implied that the transport of of Paesun cells using loading trehalose. The cryoprotective
trehalose by apatite NPs was through the local modification media was a “Cryocool” solution, an intracellular-like cooling
of the three-dimensional structure of lipids in the interaction solution for the treatment of the molecular-based cellular
with NPs and the temporary breaching of the lipid bilayer so as damages (cryopreservation-induced delayed onset cell death)
to enhance the permeation of trehalose through the bilayers. In during cryopreservation. The efficiency of cryopreservation via
addition, apatite NPs did not insert themselves into the lipid loading trehalose was impacted by the cryoprotective media. In
bilayer core but rather adsorbed on it. Moreover, apatite NPs this process, trehalose was loaded through fluid-phase
were biocompatible with RBCs and suitable for blood endocytosis of cells at physiological temperature. However,
transfusion applications. The findings are in good agreement fluid-phase endocytosis does not deliver sufficient amounts
with recent studies that reported good hemocompatibility of (0.1 M or more) of trehalose to cells73 and a long incubation
apatite NPs, particularly in direct contact with RBCs.116,117 time is required.101
Zhang et al.22 utilized a cold-responsive polymer (poly(N- 4.9. Sonoporation. Janis et al.100 discovered that trehalose
isopropylacrylamide-co-butyl acrylate)) to synthesize NPs for may be effectively delivered into RBCs to promote long-term
the encapsulation and intracellular transport of trehalose. cryopreservation by the transitory pore formation induced by
Trehalose could effectively absorb cold-responsive NPs ultrasound and microbubbles (sonoporation). After freezing
through endocytosis (or other mechanisms). The rapid and and thawing, RBCs could maintain normal morphology.
irreversible disassembly of the NPs after cold treatment Sonoporation occurred when ultrasound drove the oscillation
allowed for the controlled and quick release of trehalose and collapse of microbubbles near the cell membrane, thereby
from intracellular NPs. Cells with intracellular trehalose inducing transient pores formation, which could absorb
conveyed using the NPs showed comparable survival after impermeant compounds including trehalose. This process
cryopreservation compared to cells treated with DMSO. In also contributed to the microstreaming in the surrounding
addition, the precipitated polymers resulting from the cooling fluid, which enhanced the transport of compounds via formed
and warming of intracellular NPs could be excreted from the pores that reseal in a few seconds in a calcium-dependent
cells via exocytosis if they were not degraded inside the cells. It pathway. Therefore, in contrast to the method that relies on
is also crucial to consider that the incubation time of the passive diffusion, sonoporation can actively transport trehalose
trehalose encapsulated in the PLGA−pNIPAM-B−PF127 NPs into cells with higher efficiency through the plasma membrane
(nTre) must be optimized. There are two reasons. The former of RBCs. The advantage of sonoporation is that the loading of
is that the reduction of intracellular trehalose might be trehalose can be completed in less than 1 min in combination
insufficient to prevent the cells from cryodamage because of with a fluidics device, and the process could be simply
the short incubation time with nTre NPs. The latter is that extended to the required yield. In addition, accurate loading of
excess of intracellular trehalose caused by excessive incubation the needed trehalose concentrations can be achieved by
time with nTre NPs might increase intracellular osmotic stress changing the dosage of microbubbles, which is essential to
and allow extracellular water to enter the cells, ultimately strike a balance between loading enough trehalose for efficient
leading to an expansion in cell volume and subsequent cell cryopreservation and lessening the risk of osmotically induced
rupture. Hence, the incubation period with nTre NPs has to be hemolysis during infusion. In summary, the use of sonopora-
optimized to provide a suitable amount of intracellular tion to enhance the load of CPAs in RBCs is a promising
trehalose to avoid any injury to the cells prior to method to increase the storage time in cryopreservation.
cryopreservation. As mentioned above, delivery of trehalose However, it is worth noting that the membrane permeability is
using GNPs requires 24 h of incubation to allow cells to take not specific for trehalose, and ultrasound might also impair
up and release trehalose encapsulated in NPs,108 whereas cells activity or alter the morphology of cells.101
can be cryopreserved after 4 h of incubation with cold- 4.10. Other Methods. Abazari et al.120 investigated the
responsive NPs loaded with trehalose because cold-triggered membrane permeability of engineered lipophilic derivatives of
trehalose can be rapidly released from nTre NPs (within trehalose. Trehalose conjugated with six acetyl groups
several minutes), greatly reducing the preparation time of (trehalose hexaacetate or 6-O-Ac-Tre) exhibited more
cryopreserved cells.22 excellent permeability in rat hepatocytes than regular trehalose.
1195 https://doi.org/10.1021/acsbiomaterials.2c01225
ACS Biomater. Sci. Eng. 2023, 9, 1190−1204
Table 3. Summary of Various Methods for Delivery of Trehalose into Cells and Tissues during Cryopreservationa
incubation conditions of the experiment survival rate of the incubation conditions of the control survival rate of the control
cryopreserved samples delivery method group experiment group group group references
islets thermal shock 300 mmol/L trehalose + DMSO 92% DMSO alone 58% 199730
mouse embryonic fibroblast freezing-induced, fluid-phase DMEM with trehalose ∼56% (membrane intact DMEM without trehalose ∼9.3% (membrane intact cells) 201695
(3T3) cells endocytosis cells)
hADSCs electroporation electroporation at 1.5 kV/cm + 250 mM ∼83.8% 10% DMSO in 90% fetal bovine serum ∼91.5% 2017105
trehalose
human keratinocytes engineered pores 0.2 M intra- and extracellular trehalose ∼71.9% 0.2 M extracellular trehalose ∼43.3% 200098
oocytes microinjection 0.5 M extracellular trehalose + 0.15 M 63% (−15 °C), 0.5 M extracellular trehalose 22% (−15 °C), 2002106
intracellular trehalose
53% (−30 °C), all oocytes degenerated (−30
°C and-60 °C)
66% (−60 °C)
CHO-K1 cells TRET1 transporter CHO-TRET1 cells + 400 mM trehalose ∼80.6% (maximum CHO-vector cells + 250 mM trehalose ∼9.9% (maximum viabilities) 2007110
viabilities)
ACS Biomaterials Science & Engineering

human RBCs polymers 250 mM trehalose (intracellular ∼75% extracellular trehalose alone ∼60% 201197
concentration)
SAOS-2 cells polymers 25 μg/mL PP-50 + 0.2 M trehalose ∼60% (cell viability) 0.2 M trehalose ∼44% (cell viability) 2013111
RBCs liposomes liposomes + trehalose + HES ∼97.4% trehalose + HES ∼94.5% 2012102
hADSCs NPs 6.2 ± 0.2 mM nanoparticle-encapsulated ∼91.2% (cell viability) 10% (v/v) DMSO ∼88.2% (cell viability) 201523
trehalose
RBCs NPs 2.25 mg/mL apatite NP (pH = 6.5) + 0.35 ∼90.7% trehalose alone (pH = 7.4) ∼51.8% 2017115
M trehalose

1196
hADSCs NPs nTre ∼82.9% (cell viability) 10% (v/v) DMSO 86.3% (cell viability) 201922
MDA-MB-231 cancer cells NPs nTre ∼85.3% (cell viability) 10% (v/v) DMSO 90.2% (cell viability) 201922
Paesun cells fluid-phase endocytosis “Cryocool” + 30 mM trehalose ∼89.2% 10% FBS’s MEM + 30 mM trehalose ∼33.1% 2020119
RBCs sonoporation 5% (v/v) MB concentration + PBS + 200 ∼90% 0% (v/v) MB concentration + PBS + ∼35% 2021100
mM trehalose 200 mM trehalose
a
Abbreviations: DMSO = dimethyl sulfoxide, DMEM = Dulbecco’s modified Eagle medium, hADSCs = human adipose-derived stem cells, CHO-TRET1 cells = TRET1-expressing CHO-K1 cells,
CHO-vector = CHO-K1 cells transfected with the empty vector, RBCs = red blood cells, HES = hydroxyethyl starch, NP = nanoparticles, nTre = trehalose encapsulated in PNP nanoparticles, MB =
microbubble, PBS = phosphate buffered saline.
pubs.acs.org/journal/abseba
Review

https://doi.org/10.1021/acsbiomaterials.2c01225
ACS Biomater. Sci. Eng. 2023, 9, 1190−1204
ACS Biomaterials Science & Engineering pubs.acs.org/journal/abseba Review

Table 4. Cells and Tissues Cryopreserved Using Trehalose Alonea


cryopreserved concentrations of trehalose used for
samples cryopreservation effect of cryopreservation with trehalose references
platelets 300 mM trehalose significantly higher cryosurvival in trehalose compared to 5% DMSO 201247
yeast cells 0.5% trehalose within 5 days, exogenous trehalose could greatly increase the survival of yeast cells during 201748
cryopreservation
mouse sperm 0.3 M trehalose in skim milk 79% post thaw fertility in trehalose compared to 10% fertility in 0.3 M glycerol 200149
adipose tissues 0.25 M trehalose trehalose can enhance the adipose tissues’ long-term preservation 200550
MVs and PBS supplemented with 25 mM the morphology, integrity, and HSC-supportive potential of MVs and exosomes are well 202188
exosomes trehalose preserved using pTRE at 80 °C
a
Abbreviations: DMSO = dimethyl sulfoxide, MVs = microvesicles, PBS = phosphate buffered saline, pTRE = PBS supplemented with trehalose,
HSC = hematopoietic stem cell.

Once inside the cell, intracellular esterases hydrolyzed the 6-O- methods appear to be highly efficient for loading non-
Ac-Tre molecule, releasing the free trehalose into the permeable trehalose into cells, including but not limited to
cytoplasm. The overall concentration of intracellular trehalose electroporation, TRET1 transporter, liposomes, NPs, fluid-
(plus acetylated variants) achieved a suitable concentration for phase endocytosis, sonoporation, and cryoprotective media,
applications in cryopreservation. This method of loading which may significantly facilitate the application of trehalose as
trehalose controlled only the chemical properties of the an alternative and sole CPA for cells and tissues.
trehalose, not the cell, to elevate the membrane permeability.
Trehalose loading rate by 6-O-Ac-Tre was comparable to fluid- 5. APPLICATIONS OF TREHALOSE IN
phase endocytosis of platelets and the engineered TRET1 CRYOPRESERVATION
transporter protein but was typically slower than most other Trehalose has been widely used for cells and tissues in
methods involving perforation of the membrane. Compared to cryopreservation. We will briefly discuss in this section the use
other methods, the delivery efficiency of this method is of trehalose alone and in combination with other CPAs.
unprecedented. Overall, the engineering of the chemical 5.1. Individual Use of Trehalose. Trehalose is commonly
structure of trehalose is a harmless and cell-friendly way of used as a CPA.122,123 Using it as the sole CPA could also
delivering trehalose. It has the potential to load trehalose in protect cells and tissues even when it is not present in the
various cell types and can promote the widespread use of cell.96,124
trehalose in cryopreservation. Gläfke et al.47 investigated the application of trehalose in the
Additionally, mammalian cells can endogenously synthesize preservation of human platelets. Trehalose uptake could be
trehalose by genetic engineering. Whereas, this method needs mediated by the phase transition of platelets’ membrane
the application of adenoviral vectors at high infection induced by freezing during cryopreservation, thereby increas-
multiplicities, which can lead to substantial cytotoxicity. And, ing the survival rate of cryopreserved platelets. Yin et al.48
it is mainly used in freeze-drying, so it may not be suitable for discovered that adding 0.5% trehalose preserved yeast cells in a
cryopreservation.23,121 short time but was ineffective for cryopreservation for longer
In conclusion, if trehalose is to be applied as an effective than 5 days. It still indicated that yeast cells could survive with
CPA for the preservation of cells and tissues, the method of a significantly higher survival rate in cryopreservation when
loading should not markedly change cell structural or exogenous trehalose was present. Trehalose has also been used
functional aspects or lead to cellular harm or death. Methods alone for cryopreservation of sperm with excellent results.
that permit selective access to trehalose are also the preferred Sztein et al.49 compared the cryoprotective ability of permeable
options since the influx or efflux of other small molecules that and nonpermeable compounds on freezing mouse sperm. Post-
may negatively affect cells is undesirable. In the meantime, freezing sperm viability studies showed that trehalose was more
methods that allow for rapid uptake of trehalose are preferred protective than permeable CPA (e.g., DMSO or glycerol) for
because incubation of cells typically needs to be completed mouse sperm.
within 24 h. NPs or other methods seem promising because For the long-term preservation of living tissues, cryopre-
they can specifically deliver trehalose without altering servation is also an efficient method. Pu et al.50 used 0.25 M
important cellular structures. Moreover, the survival of cells trehalose in the cryopreservation of adipose tissue. Compared
treated with trehalose-loaded NPs after cryopreservation is with the group without trehalose, in the trehalose-optimal
similar to that treated with DMSO. Changing the NP design to group, more viable adipocytes and greater cellular adipose
enable a faster and more efficient uptake of the cells is this tissue function were observed.
method’s future direction for improvement. All these methods Based on the results, trehalose can be used as a sole CPA to
have proven the success of trehalose in cryopreserving cells, effectively protect adipose tissue during cryopreservation.
but each method has its superiorities and limitations. In other aspects, Budgude et al.88 used PBS or PBS
Here, we present a summary of the various methods that supplemented with trehalose (pTRE) to cryopreserve extrac-
have been studied to deliver trehalose into cells and tissues for ellular vesicles (EVs)-microvesicles (MVs) and exosomes at
efficient cryopreservation, as listed in Table 3. It indicated that different temperatures, and they further investigated the impact
the uptake of trehalose during cryopreservation can enor- of cryopreserved EVs on the ex vivo expansion of
mously enhance the survival rates of cells and tissues by hematopoietic stem cells (HSCs). As a result, cryopreservation
comparing the survival rates of the experimental and control at 80 °C in pTRE retained the morphology, integrity, and
groups. Moreover, extracellular and intracellular trehalose can HSC-supportive potential of both MVs and exosomes.
significantly increase the viability of the cells and tissues In summary, trehalose can significantly improve the survival
compared to extracellular trehalose alone. Overall, these rate of cells and tissues compared with the group without
1197 https://doi.org/10.1021/acsbiomaterials.2c01225
ACS Biomater. Sci. Eng. 2023, 9, 1190−1204
ACS Biomaterials Science & Engineering pubs.acs.org/journal/abseba Review

Table 5. Cells and Tissues Cryopreserved Using Trehalose in Combination with Other CPAsa
cryopreserved incubation conditions of incubation conditions of trehalose and
samples trehalose alone survival rate other CPAs survival rate references
fetal skin 0.5 M Tre + 10% DMSO 65% (the membrane integrity, 200231
compared to 44% for 10% DMSO)
normal human 100 mM Tre 58.5% 10% (w/w) DMSO + 100 mM Tre 85.6% 201782
skin fibroblasts
ADSCs 1.0 M Tre 65 ± 1.10% 1.0 M Tre + 20% Gly 77 ± 1.72% 2020130
RBCs 0.2Tre ∼75% (hemolysis of 1.5Pro&0.2Tre 11.2 ± 2.73% 2019131
thawed RBCs)
(hemolysis of thawed RBCs)
RBCs 0.36 M Tre 78.3 ± 0.4% (pH 0.36 M Tre + 10 mg·mL−1 PKDF1T 89.2 ± 0.2% (pH 6.0) 2020132
6.0)
51.0 ± 5.1% (pH 81.9 ± 0.5% (pH 7.4)
7.4)
RBCs 290 mM Tre 77.8 ± 15.1% Tre + 24.5% (w/w) HES + 92.6−97.4% 2012102
0.5−7 mM liposomes
RBCs 100 mM Tre ∼40% 100 mM Tre, 100 mg·mL−1 97% 2021133
polyampholyte, 10% DMSO, in
water
RBCs 300 mM Tre + 100 μg/ 54.8 ± 1.7% 300 mM Tre +100 μg/mL polymer 61.2 ± 1.4% 2016135
mL polymer PP-50 PP-50 + 200 μM Sal
a
Abbreviations: DMSO = dimethyl sulfoxide, Tre = Trehalose, ADSCs = adipose-derived stem cells, Gly = glycerol, RBCs = red blood cells, 0.2Tre
= 0.2 mol of trehalose dissolve into 1 L of Dulbeco’s phosphate buffered saline (DPBS), 1.5Pro&0.2Tre = 1.5 mol of L-proline and 0.2 mol of
trehalose dissolve into 1 L of DPBS, PKDF1T = PEG-b-(Lys23-co-Asp45)-co-Phe6-g-Tre8, a kind of glycopeptide, HES = hydroxyethyl starch, Sal
= salidroside.

trehalose. And, the effects of cryopreservation with trehalose al.131 used natural CPA combinations of L-proline and
on the different cells and tissues are summarized in Table 4. trehalose to obtain a low level of hemolysis (11.2 ± 2.73%)
5.2. Combination. Combining one CPA with another is a after thawing. Moreover, Na+/K+-ATPase and Ca2+/Mg2+-
common way to enhance the performance of CPAs.125 ATPase activities and the morphology of RBCs were well-
Trehalose is a nonpermeable CPA that is receiving increasing maintained. A further mechanistic study showed that L-proline
concern owing to its capacity to protect against ice crystal penetrated into the cells during freezing, which helped reduce
formation and exert cryoprotective influences.126,127 Never- the freezing point and prevent the growth of ice crystals. In
theless, trehalose may not inhibit the formation of intracellular contrast, trehalose prevents the growth of ice during freezing
ice crystals because of its inability to pass through the cell and the recrystallization of ice during thawing. This simple
membrane. Consequently, using trehalose in combination with method offers a possible substitute for the current time-
other CPAs might be necessary to protect cells both inside and consuming and labor-intensive RBCs’ cryopreservation meth-
outside. Previous studies have shown that this type of od.
combined treatment greatly enhances the protection of cells Second, with the help of nonpermeable CPAs alone or the
or tissues during cryopreservation by a probable synergistic combination of nonpermeable and permeable CPAs, trehalose
mechanism.128,129 can produce more positive results for cells and tissues during
First, trehalose can be used with permeable CPAs for cryopreservation. Liu et al.132 proposed biocompatible
protecting cells and tissues during cryopreservation. For synthetic glycopeptides to protect RBCs during cryopreserva-
example, Erdag et al.31 found that trehalose and DMSO tion. By using poly(ethylene glycol)-NH2 to induce the ring-
working together as CPAs enhanced the cryopreservation of opening polymerization of N-carboxyanhydrides of lysine,
fetal skin. Additionally, Kratochvilová́ et al.82 found that the aspartic acid, and phenylalanine and then chemically anchoring
highest survival rate was obtained when DMSO was combined carboxylated trehalose to the pendant amino moieties, a series
with trehalose, which significantly altered the freezing process of glycopeptides have been produced. The specially designed
in normal human skin fibroblasts (NHDF). The results were glycopeptides with trehalose improved the cryopreservation
compared with the samples that included control, AFP, survival of sheep RBCs at pH 6.0 and pH 7.4 to 89.2 ± 0.3%
DMSO, and trehalose. and 81.9 ± 0.3%, respectively. Synergistic cryopreservation of
Trehalose and glycerol together are also a successful method RBCs was achieved through membrane stabilization of
for cell cryopreservation. Zhang et al.130 postulated that the glycopeptides by tuning water diffusion and inhibiting the ice
combination of glycerol and trehalose could protect adipose- recrystallization of trehalose during cryopreservation. This
derived stem cells (ADSCs) against cryodamage and preserve combination of synthetic glycopeptides and trehalose provides
the cells’ viability. In their study, they first determined the most a possible method for effective and glycerol-free cryopreserva-
ideal concentration of trehalose and glycerol when supplied tion of RBCs.
alone and then evaluated the impact of trehalose and glycerol Stoll et al.102 used liposomes, trehalose, and HES to protect
combined on maintaining ADSCs’ viability and cell function. RBCs synergistically during cryopreservation. After cryopre-
The findings showed that 1.0 M Tre + 20% Gly could servation, trehalose was delivered to cells by freezing induction,
cryopreserve ADSCs more effectively, making it potentially while liposomes could stabilize RBCs during cryopreservation.
useful for clinical applications. HES is also a nonpermeable CPA, which could effectively
Cryopreservation of RBCs has great potential benefits in inhibit the formation of intracellular ice crystals and improve
providing timely blood transfusions in emergencies. Dou et cell survival rate.15 When cells were incubated with trehalose
1198 https://doi.org/10.1021/acsbiomaterials.2c01225
ACS Biomater. Sci. Eng. 2023, 9, 1190−1204
ACS Biomaterials Science & Engineering pubs.acs.org/journal/abseba Review

and liposomes and frozen in HES, the recovery was between macrophages and fibroblasts in only a few hours at 37 °C.143
92.6% and 97.4%. In contrast, the recovery of RBCs incubated Meanwhile, Eroglu et al.106 proposed that the small number of
and frozen in trehalose medium was 77.8%. Therefore, the intracellular sugars could be metabolized after nonspecific
synergistic protective effect of trehalose, liposome, and HES cleavage of their backbones, excreted by exocytosis, or even
could significantly improve the survival rate of RBCs, which more diluted by cell division. And, mammalian cells tolerate
was expected to become the future direction for development intracellular trehalose of at least 0.2 M without damaging their
of new methods for cryopreservation of RBCs. The benefit of functions. Therefore, it does not have to be eliminated from
this method is that it does not require the removal of cryopreserved cells and tissues, and the effect of trehalose on
protective compounds before blood transfusion, so as to avoid cells may be negligible.110,144 It greatly facilitates the
a large number of time-consuming cleaning procedures.102 widespread use of emerging cellular medicine.23,50,102
Murray et al.133 showed that combining CPAs with different 6.2. Limits of Trehalose in Cryopreservation. It must
modes of action could result in a high post-thaw recovery of be noted that sugar also has the shortcomings of not being able
RBCs. In this study, the results demonstrated that the most to penetrate cell membranes. And, since mammalian cells do
effective formulation with a post-thaw recovery of 97% not metabolize trehalose, its use in mammalian cells is
contained 100 mg mL−1 polyampholyte, 10% DMSO, and limited.145 The nonpermeating trehalose provides the
100 mM trehalose in water. The exact mechanism of the cryoprotective ability, but to achieve the best protection for
cryoprotective ability of polyampholytes was not yet clear, but cells, trehalose is supposed to exist inside and outside the
the combination of intracellular (DMSO), extracellular cells.72 Trehalose must be delivered into cells using various
(trehalose), and polymer could lead to reduced hemolysis. In methods or it must be combined with other CPAs.
terms of the exact mechanism of cryoprotective ability by However, in order to obtain higher intracellular trehalose
polyampholytes, Matsumura et al.134 did relevant research by contents, cells must be incubated for a long time in the
using solid-state NMR spectroscopy. This technique revealed existence of trehalose or at a higher trehalose concentration.
the mechanism of the cryoprotective ability of carboxylated But, if the preloading trehalose concentration is too high, the
poly-L-lysines (COOH-PLLs), which could be summarized as cells will lose their viability due to osmotic shock.95 There are
follows. First, under mild conditions, cell dehydration from the two probabilities for cell responses to high osmotic pressure.
outside inhibited IIF. This was accomplished by regulating First, the cell membrane is unable to tolerate the movement of
osmotic pressure by encasing water and salt molecules in the the solution, leading to disruption of the cell membrane.
polymer matrix. Second, preserving a viscous condition at Second, when osmotic stress is imposed and the cell is overly
higher temperatures inhibited IIF caused by the influx of contracted, the deformation destroys the cell structure. An
extracellular ice. improvement in trehalose concentration is associated with an
Despite the protective effect of CPAs, freeze−thaw processes improvement in osmotic stress. Therefore, the incubation
can still alter cells and result in oxidative damage. This might period of trehalose has to be optimized to ensure a moderate
cause severe cellular damage by enhanced production of free concentration of intracellular trehalose and an ideal balance
radical oxygen species (ROS).135 Antioxidants are able to between trehalose’s protective and destructive impacts. To sum
counteract oxidative stress and lessen ROS-related damage. up, the optimal trehalose concentration is needed to produce
Consequently, the damage caused by cryopreservation can be finer ice crystals and reduce osmotic damage. Additionally, to
significantly reduced by using a combination of antioxidants decrease the possibility of osmotic shocks, a high-capacity
and CPAs.136 Alotaibi et al.135 evaluated the effects of a novel trehalose transporter can be used. It can relatively quickly
CPA and a strong antioxidant named salidroside (Sal) on reduce the trehalose gradient on the cell membrane.144
sheep RBC biological functions during cryopreservation,
combining glycerol and trehalose/PP-50. The addition of Sal 7. CONCLUSION
improved survival by 7.6 ± 0.3% as compared to using
As more and more translational medicine products enter the
trehalose alone. Here, Sal can reduce the oxidative damage of
clinic, the need for advanced storage of cells and tissues is
sheep RBCs during cryopreservation.
increasing. One of the many factors considered in emerging
In summary, trehalose can effectively improve the survival
translational medicine is the safety of cryopreserving cells and
rate of cells and tissues by combining with other CPAs and can
tissues. Therefore, the utility and demand for CPAs to prevent
be used as a new method in cryopreservation (Table 5).
or minimize damage from freezing are gradually improving.
6. BENEFITS AND LIMITS OF TREHALOSE IN Trehalose has gained interest in cryopreservation due to its
capacity to prevent ice crystal formation, protect proteins and
CRYOPRESERVATION
other biomolecules, stabilize cell membranes, and increase
6.1. Benefits of Trehalose in Cryopreservation. membrane permeability. In this article, the properties,
Trehalose is a particularly attractive biological CPA because mechanisms, delivery methods, applications, and benefits and
its role in protecting cells and tissues from freezing damages is limits of trehalose are described comprehensively. There are
well-known.67,137 Conventional cryopreservation of cells and the following findings and research direction of trehalose in
tissues needs the utilization of toxic organic solvents (such as biomedical cryopreservation.
DMSO) as CPAs, which cannot be used directly on
patients.138−140 As a result, cryopreserved cells have to (1) The effect of trehalose in cells is negligible. This
withstand cumbersome washing procedures to eliminate argument is based on relevant experiments with oocytes,
organic solvents, resulting in significant cell loss.141,142 where Kikawada et al.144 incubated oocytes in a 105 mM
Conversely, as a sugar, trehalose is not toxic to cells and trehalose solution for 3 h and then discovered that the
tissues. Besides, intracellular trehalose can be delivered to the elimination of intracellular trehalose from oocytes took
extracellular space via exocytosis, which has been demonstrated only about 2 h. Meanwhile, osmotically sensitive
to remove cytoplasmic sucrose (also a disaccharide) from mammalian oocytes were also capable of surviving

1199 https://doi.org/10.1021/acsbiomaterials.2c01225
ACS Biomater. Sci. Eng. 2023, 9, 1190−1204
ACS Biomaterials Science & Engineering pubs.acs.org/journal/abseba Review

after being microinjected with 150 mM trehalose. For Fenglin Liu − Xiangya School of Pharmaceutical Sciences,
oocytes, trehalose was a special CPA, which quickly Central South University, Changsha, Hunan 410013, China
disappeared during embryonic development. Similarly, Jingxian Xie − Xiangya School of Pharmaceutical Sciences,
although intracellular high trehalose might increase Central South University, Changsha, Hunan 410013, China
intracellular osmotic pressure, there was no significant Qubo Zhu − Xiangya School of Pharmaceutical Sciences,
effect on liver mononuclear cells (LMC) activity and Central South University, Changsha, Hunan 410013, China;
function after loading 240 mM trehalose. This might be orcid.org/0000-0001-5202-7887
due to the ability of various tissue cells, including Complete contact information is available at:
hepatocytes and kidney cells, to activate cell volume https://pubs.acs.org/10.1021/acsbiomaterials.2c01225
control mechanisms that adjusted cell volume according
to intraosmotic pressure over time.73 Consequently, the Author Contributions
effect of trehalose on cells during the incubation period All authors made contributions to the manuscript’s writing.
in a normal medium may be negligible. The published version of the manuscript has been read and
(2) The use of trehalose in RBCs needn’t remove steps. approved by all authors.
Janis et al. 100 delivered trehalose to RBCs by Funding
sonoporation without removal steps, and the thawed
There was no external funding.
RBCs did not cause any adverse transfusion reactions.
Namely, trehalose in cells may not cause adverse blood Notes
transfusion reactions and RBCs can be directly used for The authors declare no competing financial interest.
clinical applications.
(3) More researches are needed to determine whether
trehalose must be delivered into the cells. As mentioned
■ REFERENCES
(1) National Blood Collection & Utilization Survey; U.S. Department
above, trehalose must be present in both intracellular of Health and Human Services, 2011.
and extracellular cells to supply maximum protection (2) Janssen, S. J.; Braun, Y.; Wood, K. B.; Cha, T. D.; Schwab, J. H.
against stress and damage during cryopreservation. But, Allogeneic blood transfusions and postoperative infections after
lumbar spine surgery. The Spine Journal 2015, 15, 901.
Crowe et al.124 expected trehalose to stabilize the cell (3) Lee, S.; Ozkavukcu, S.; Ku, S.-Y. Current and Future
membrane through the pathway from extracellular to cell Perspectives for Improving Ovarian Tissue Cryopreservation and
membrane and membrane localization protein. This Transplantation Outcomes for Cancer Patients. Reproductive Sciences
study pointed out that, even when trehalose was not 2021, 28 (6), 1746−1758.
present in the cell, it could still protect cells and enhance (4) Afreen, M. Significance of cryopreservation biotechnology for
liver-specific functions. Additionally, each delivery protection of aquatic species. Eurasian Journal of Agricultural Research
method of trehalose has its superiorities and limitations. 2020, 4 (2), 64−71.
Therefore, it is hard to evaluate whether trehalose must (5) Mazur, P.; Leibo, S. P.; Chu, E. H. A two-factor hypothesis of
be delivered into the cells. freezing injury. Evidence from Chinese hamster tissue-culture cells.
Experimental cell research 1972, 71 (2), 345−55.
(4) Exploring optimal concentration and proportion. (6) Yeste, M. Sperm cryopreservation update: Cryodamage, markers,
Trehalose has poor cell permeability, so other and factors affecting the sperm freezability in pigs. Theriogenology
supplementary CPAs are always added to provide 2016, 85 (1), 47−64.
enough intracellular protection. When trehalose is (7) Gao, D.; Critser, J. K. Mechanisms of Cryoinjury in Living Cells.
combined with other CPAs, the optimal concentration ILAR Journal 2000, 41 (4), 187−196.
and proportion should be explored first. (8) Yurchuk, T.; Petrushko, M.; Fuller, B. Science of cryopreserva-
tion in reproductive medicine − Embryos and oocytes as exemplars.
Early Human Development 2018, 126, 6−9.
8. PROSPECT (9) Jahan, S.; Kaushal, R.; Pasha, R.; Pineault, N. Current and Future
Trehalose displays great promise as a nontoxic CPA that does Perspectives for the Cryopreservation of Cord Blood Stem Cells.
not require removal from cells and tissues before translational Transfusion Medicine Reviews 2021, 35 (2), 95−102.
medicine application, and much progress has been made in the (10) Yang, J.; Pan, C.; Zhang, J.; Sui, X.; Zhu, Y.; Wen, C.; Zhang, L.
delivery of trehalose through the membrane to cells. In the Exploring the Potential of Biocompatible Osmoprotectants as Highly
Efficient Cryoprotectants. ACS Appl. Mater. Interfaces 2017, 9 (49),
future, trehalose is hopeful to be applied in the cryopreserva- 42516−42524.
tion of human tissues and organs to advance the development (11) Paul, R. K.; Kumar, D.; Singh, R. Carboxymethyl cellulose and
of translational medicine. glycerol act synergistically as cryoprotectant during cryopreservation

■ AUTHOR INFORMATION
Corresponding Author
of ram semen. Cryobiology 2021, 101, 61−66.
(12) Stubbs, C.; Bailey, T. L.; Murray, K.; Gibson, M. I.
Polyampholytes as Emerging Macromolecular Cryoprotectants.
Songwen Tan − Xiangya School of Pharmaceutical Sciences, Biomacromolecules 2020, 21 (1), 7−17.
(13) Keskin, N.; Erdogan, C.; Bucak, M. N.; Ozturk, A. E.; Bodu,
Central South University, Changsha, Hunan 410013, China; M.; Ili, P.; Baspinar, N.; Dursun, S. Cryopreservation effects on ram
orcid.org/0000-0002-2138-7465; Email: songwen.tan@ sperm ultrastructure. Biopreservation and Biobanking 2020, 18 (5),
csu.edu.cn 441−448.
(14) Ntai, A.; La Spada, A.; De Blasio, P.; Biunno, I. Trehalose to
Authors cryopreserve human pluripotent stem cells. Stem Cell Research 2018,
Yuying Hu − Xiangya School of Pharmaceutical Sciences, 31, 102−112.
Central South University, Changsha, Hunan 410013, China (15) Stolzing, A.; Naaldijk, Y.; Fedorova, V.; Sethe, S. Hydrox-
Xiangjian Liu − Xiangya School of Pharmaceutical Sciences, yethylstarch in cryopreservation - mechanisms, benefits and problems.
Central South University, Changsha, Hunan 410013, China Transfus Apher Sci. 2012, 46 (2), 137−47.

1200 https://doi.org/10.1021/acsbiomaterials.2c01225
ACS Biomater. Sci. Eng. 2023, 9, 1190−1204
ACS Biomaterials Science & Engineering pubs.acs.org/journal/abseba Review

(16) Rajan, R.; Hayashi, F.; Nagashima, T.; Matsumura, K. Toward a testicular tissue: comparison of protocols with glycerol, propanediol
Molecular Understanding of the Mechanism of Cryopreservation by and dimethylsulfoxide as cryoprotectants. Hum. Reprod. 2005, 20 (6),
Polyampholytes: Cell Membrane Interactions and Hydrophobicity. 1676−1687.
Biomacromolecules 2016, 17 (5), 1882−1893. (35) Newton, H.; Aubard, Y.; Rutherford, A.; Sharma, V.; Gosden,
(17) Bachtiger, F.; Congdon, T. R.; Stubbs, C.; Gibson, M. I.; Sosso, R. Ovary and ovulation: Low temperature storage and grafting of
G. C. The atomistic details of the ice recrystallisation inhibition human ovarian tissue. Human reproduction 1996, 11 (7), 1487−1491.
activity of PVA. Nat. Commun. 2021, 12 (1), 1323. (36) Bakhach, J. The cryopreservation of composite tissues:
(18) Shaluei, F.; Imanpoor, M. R.; Shabani, A.; Nasr-Esfahani, M. H. principles and recent advancement on cryopreservation of different
Effect of different concentrations of permeable and non−permeable type of tissues. Organogenesis 2009, 5 (3), 119−126.
cryoprotectants on the hatching rate of goldfish (Carassius auratus) (37) Zhang, P.-Q.; Tan, P.-C.; Gao, Y.-M.; Zhang, X.-J.; Xie, Y.;
embryos. Asian Pacific Journal of Reproduction 2013, 2 (3), 185−188. Zheng, D.-N.; Zhou, S.-B.; Li, Q.-F. The effect of glycerol as a
(19) Best, B. P. Cryoprotectant Toxicity: Facts, Issues, and cryoprotective agent in the cryopreservation of adipose tissue. Stem
Questions. Rejuvenation Research 2015, 18 (5), 422. Cell Res. Ther. 2022, 13 (1), 152.
(20) Ataseven, E.; Tufekci, O.; Yilmaz, S.; Guleryuz, H.; Oren, H. (38) Lovelock, J. E.; Bishop, M. W. H. Prevention of Freezing
Neurotoxicity Associated With Dimethyl Sulfoxide Used in Allogeneic Damage to Living Cells by Dimethyl Sulfoxide. Nature 1959, 183
Stem Cell Transplantation. J. Pediatr. Hematol. Oncol. 2017, 39 (5), (4672), 1394−1395.
E297−E299. (39) Hornberger, K.; Yu, G.; McKenna, D.; Hubel, A.
(21) Bakaltcheva, I. B.; Odeyale, C. O.; Spargo, B. J. Effects of Cryopreservation of Hematopoietic Stem Cells: Emerging Assays,
alkanols, alkanediols and glycerol on red blood cell shape and Cryoprotectant Agents, and Technology to Improve Outcomes.
hemolysis. Biochimica et Biophysica Acta (BBA) - Biomembranes 1996, Transfusion Medicine and Hemotherapy 2019, 46 (3), 188−196.
1280 (1), 73−80. (40) Jia, X.; Yu, Y.; Li, D. Investigation on the effect of trehalose on
(22) Zhang, Y.; Wang, H.; Stewart, S.; Jiang, B.; Ou, W.; Zhao, G.; alpha-actinin in cryopreserved human skin. Zhongguo Xiu Fu Chong
He, X. Cold-Responsive Nanoparticle Enables Intracellular Delivery Jian Wai Ke Za Zhi 2008, 22 (4), 446−449.
and Rapid Release of Trehalose for Organic-Solvent-Free Cryopre- (41) Xia, Z.; Duan, X.; Murray, D.; Triffitt, J. T.; Price, A. J. A
servation. Nano Lett. 2019, 19 (12), 9051−9061. method of isolating viable chondrocytes with proliferative capacity
(23) Rao, W.; Huang, H.; Wang, H.; Zhao, S.; Dumbleton, J.; Zhao, from cryopreserved human articular cartilage. Cell and tissue banking
G.; He, X. Nanoparticle-Mediated Intracellular Delivery Enables 2013, 14 (2), 267−276.
Cryopreservation of Human Adipose-Derived Stem Cells Using (42) Keskin, N.; Erdogan, C.; Bucak, M. N.; Ozturk, A. E.; Bodu,
Trehalose as the Sole Cryoprotectant. ACS Appl. Mater. Interfaces M.; Ili, P.; Baspinar, N.; Dursun, S. Cryopreservation Effects on Ram
2015, 7 (8), 5017−5028. Sperm Ultrastructure. Biopreserv Biobank 2020, 18 (5), 441−448.
(24) Lee, Y. A.; Kim, Y. H.; Kim, B. J.; Kim, B. G.; Kim, K. J.; Auh, J. (43) Oldenhof, H.; Gojowsky, M.; Wang, S.; Henke, S.; Yu, C.;
H.; Schmidt, J. A.; Ryu, B. Y. Cryopreservation in trehalose preserves Rohn, K.; Wolkers, W. F.; Sieme, H. Osmotic Stress and Membrane
functional capacity of murine spermatogonial stem cells. PLoS One Phase Changes During Freezing of Stallion Sperm: Mode of Action of
2013, 8 (1), e54889. Cryoprotective Agents1. Biol. Reprod. 2013, 88 (3), 1−11.
(25) Rowley, S. D.; Anderson, G. L. Effect of DMSO exposure (44) Chen, S.-U.; Lien, Y.-R.; Chao, K.-H.; Lu, H.-F.; Ho, H.-N.;
without cryopreservation on hematopoietic progenitor cells. Bone Yang, Y.-S. Cryopreservation of mature human oocytes by vitrification
Marrow Transplant 1993, 11 (5), 389−393. with ethylene glycol in straws. Fertility and Sterility 2000, 74 (4),
(26) Buchanan, S. S.; Gross, S. A.; Acker, J. P.; Toner, M.; 804−808.
Carpenter, J. F.; Pyatt, D. W. Cryopreservation of stem cells using (45) von Bomhard, A.; Elsässer, A.; Ritschl, L. M.; Schwarz, S.;
trehalose: evaluation of the method using a human hematopoietic cell Rotter, N. Cryopreservation of Endothelial Cells in Various
line. Stem Cells Dev 2004, 13 (3), 295−305. Cryoprotective Agents and Media − Vitrification versus Slow
(27) Chen, S.; Wu, L.; Ren, J.; Bemmer, V.; Zajicek, R.; Chen, R. Freezing Methods. PLoS One 2016, 11 (2), No. e0149660.
Comb-like Pseudopeptides Enable Very Rapid and Efficient Intra- (46) Chi, H. J.; Koo, J. J.; Kim, M. Y.; Joo, J. Y.; Chang, S. S.; Chung,
cellular Trehalose Delivery for Enhanced Cryopreservation of K. S. Cryopreservation of human embryos using ethylene glycol in
Erythrocytes. ACS Appl. Mater. Interfaces 2020, 12 (26), 28941− controlled slow freezing. Hum. Reprod. 2002, 17 (8), 2146−51.
28951. (47) Gläfke, C.; Akhoondi, M.; Oldenhof, H.; Sieme, H.; Wolkers,
(28) Fujita, Y.; Nishimura, M.; Komori, N.; Sawamoto, O.; Kaneda, W. F. Cryopreservation of platelets using trehalose: the role of
S. Protein-free solution containing trehalose and dextran 40 for membrane phase behavior during freezing. Biotechnol. Prog. 2012, 28
cryopreservation of human adipose tissue-derived mesenchymal (5), 1347−54.
stromal cells. Cryobiology 2021, 100, 46. (48) Yin, H.; Wang, Y.; He, Y.; Xing, L.; Zhang, X.; Wang, S.; Qi, X.;
(29) Katenz, E.; Vondran, F. W.; Schwartlander, R.; Pless, G.; Gong, Zheng, Z.; Lu, J.; Miao, J. Cloning and expression analysis of tps, and
X.; Cheng, X.; Neuhaus, P.; Sauer, I. M. Cryopreservation of primary cryopreservation research of trehalose from Antarctic strain
human hepatocytes: the benefit of trehalose as an additional Pseudozyma sp. 3 Biotech 2017, 7, 343.
cryoprotective agent. Liver Transpl 2007, 13 (1), 38−45. (49) Sztein, J. M.; Noble, K.; Farley, J. S.; Mobraaten, L. E.
(30) Beattie, G. M.; Crowe, J. H.; Lopez, A. D.; Cirulli, V.; Ricordi, Comparison of permeating and nonpermeating cryoprotectants for
C.; Hayek, A. Trehalose: a cryoprotectant that enhances recovery and mouse sperm cryopreservation. Cryobiology 2001, 42 (1), 28−39.
preserves function of human pancreatic islets after long-term storage. (50) Pu, L. L.; Cui, X.; Fink, B. F.; Cibull, M. L.; Gao, D.
Diabetes 1997, 46 (3), 519−23. Cryopreservation of adipose tissues: the role of trehalose. Aesthet Surg
(31) Erdag, G.; Eroglu, A.; Morgan, J. R.; Toner, M. J. 2005, 25 (2), 126−31.
Cryopreservation of fetal skin is improved by extracellular trehalose. (51) Lionetti, F. J.; Hunt, S. M. Cryopreservation of human red cells
Cryobiology 2002, 44 (3), 218−228. in liquid nitrogen with hydroxyethyl starch. Cryobiology 1975, 12 (2),
(32) Zheng, X.; Liu, J.; Liu, Z.; Wang, J. Bio-inspired Ice-controlling 110−118.
Materials for Cryopreservation of Cells and Tissues. Acta Chimica (52) Lionetti, F. J.; Hunt, S. M.; Gore, J. M.; Curby, W. A.
Sinica 2021, 79 (6), 729. Cryopreservation of human granulocytes. Cryobiology 1975, 12 (3),
(33) Jesus, A. R.; Meneses, L.; Duarte, A. R. C.; Paiva, A. Natural 181−191.
deep eutectic systems, an emerging class of cryoprotectant agents. (53) Ashwood-Smith, M.; Warby, C.; Connor, K.; Becker, G. Low-
Cryobiology 2021, 101, 95−104. temperature preservation of mammalian cells in tissue culture with
(34) Keros, V.; Rosenlund, B.; Hultenby, K.; Aghajanova, L.; polyvinylpyrrolidone (PVP), dextrans, and hydroxyethyl starch
Levkov, L.; Hovatta, O. Optimizing cryopreservation of human (HES). Cryobiology 1972, 9 (5), 441−449.

1201 https://doi.org/10.1021/acsbiomaterials.2c01225
ACS Biomater. Sci. Eng. 2023, 9, 1190−1204
ACS Biomaterials Science & Engineering pubs.acs.org/journal/abseba Review

(54) Kenmochi, T.; Asano, T.; Maruyama, M.; Saigo, K.; Akutsu, N.; (72) Diaz-Dussan, D.; Peng, Y.-Y.; Sengupta, J.; Zabludowski, R.;
Iwashita, C.; Ohtsuki, K.; Suzuki, A.; Miyazaki, M. Cryopreservation Adam, M. K.; Acker, J. P.; Ben, R. N.; Kumar, P.; Narain, R.
of human pancreatic islets from non-heart-beating donors using Trehalose-Based Polyethers for Cryopreservation and Three-Dimen-
hydroxyethyl starch and dimethyl sulfoxide as cryoprotectants. Cell sional Cell Scaffolds. Biomacromolecules 2020, 21 (3), 1264−1273.
transplantation 2008, 17 (1−2), 61−67. (73) Wang, B.; Liu, G.; Balamurugan, V.; Sui, Y.; Wang, G.; Song, Y.;
(55) Luo, K.; Wu, G.; Wang, Q.; Sun, Y.; Liu, H. Effect of Chang, Q. Apatite nanoparticles mediate intracellular delivery of
dimethylsulfoxide and hydroxyethyl starch in the preservation of trehalose and increase survival of cryopreserved cells. Cryobiology
fractionated human marrow cells. Cryobiology 1994, 31 (4), 349−354. 2019, 86, 103−110.
(56) Neveux, N.; De Bandt, J.; Charrueau, C.; Savier, E.; Chaumeil, (74) Jain, N. K.; Roy, I. Effect of trehalose on protein structure.
J.-C.; Hannoun, L.; Giboudeau, J.; Cynober, L. A. Deletion of Protein Sci. 2008, 18 (1), 24−36.
hydroxyethylstarch from University of Wisconsin solution induces cell (75) Ajito, S.; Hirai, M.; Iwase, H.; Shimizu, N.; Igarashi, N.; Ohta,
shrinkage and proteolysis during and after cold storage of rat liver. N. Protective action of trehalose and glucose on protein hydration
Hepatology 1997, 25 (3), 678−682. shell clarified by using X-ray and neutron scattering. Physica B:
(57) Mabrut, J.; Adham, M.; Bourgeot, J.; Eljaafari, A.; DelaRoche, Condensed Matter 2018, 551, 249−255.
E.; Ducerf, C. Mechanical and histological characteristics of human (76) Kawai, H.; Sakurai, M.; Inoue, Y.; Chûjô, R.; Kobayashi, S.
trachea before and after cryopreservation: an opportunity for tracheal Hydration of oligosaccharides: Anomalous hydration ability of
tissue banking. Transplantation Proceedings 2001, 33, 609−611.
trehalose. Cryobiology 1992, 29 (5), 599−606.
(58) Watanabe, H.; Kohaya, N.; Kamoshita, M.; Fujiwara, K.;
(77) Crowe, J. H.; Crowe, L. M.; Oliver, A. E.; Tsvetkova, N.;
Matsumura, K.; Hyon, S.-H.; Ito, J.; Kashiwazaki, N. Efficient
Wolkers, W.; Tablin, F. The Trehalose Myth Revisited: Introduction
production of live offspring from mouse oocytes vitrified with a
novel cryoprotective agent, carboxylated ε-poly-L-lysine. PLoS One to a Symposium on Stabilization of Cells in the Dry State. Cryobiology
2013, 8 (12), e83613. 2001, 43 (2), 89−105.
(59) Tariq, A.; Ahmad, M.; Iqbal, S.; Riaz, M. I.; Tahir, M. Z.; (78) Lerbret, A.; Bordat, P.; Affouard, F.; Descamps, M.; Migliardo,
Ghafoor, A.; Riaz, A. Effect of carboxylated poly l-Lysine as a F. How Homogeneous Are the Trehalose, Maltose, and Sucrose
cryoprotectant on post-thaw quality and in vivo fertility of Nili Ravi Water Solutions? An Insight from Molecular Dynamics Simulations. J.
buffalo (Bubalus bubalis) bull semen. Theriogenology 2020, 144, 8− Phys. Chem. B 2005, 109 (21), 11046−11057.
15. (79) Lerbret, A.; Bordat, P.; Affouard, F.; Guinet, Y.; Hédoux, A.;
(60) Matsumura, K.; Hyon, S. H. Polyampholytes as low toxic Paccou, L.; Prévost, D.; Descamps, M. Influence of homologous
efficient cryoprotective agents with antifreeze protein properties. disaccharides on the hydrogen-bond network of water: complemen-
Biomaterials 2009, 30 (27), 4842−9. tary Raman scattering experiments and molecular dynamics
(61) Nakayama, K.; Yamanaka, T.; Tamada, Y.; Hirabayashi, M.; simulations. Carbohydr. Res. 2005, 340 (5), 881−887.
Hochi, S. Supplementary cryoprotective effect of carboxylated ε-poly- (80) Branca, C.; Maccarrone, S.; Magazu, S.; Maisano, G.;
l-lysine during vitrification of rat pancreatic islets. Cryobiology 2019, Bennington, S. M.; Taylor, J. Tetrahedral order in homologous
88, 70−74. disaccharide-water mixtures. J. Chem. Phys. 2005, 122 (17), 174513.
(62) Inada, T.; Lu, S.-S. Inhibition of Recrystallization of Ice Grains (81) Sola-Penna, M.; Ferreira-Pereira, A.; Lemos, A. d. P.; Meyer-
by Adsorption of Poly(Vinyl Alcohol) onto Ice Surfaces. Cryst. Ferwandes, J. R. Carbohydrate protection of enzyme structure and
Growth Des. 2003, 3 (5), 747−752. function against guanidinium chloride treatment depends on the
(63) Bachtiger, F.; Congdon, T. R.; Stubbs, C.; Gibson, M. I.; Sosso, nature of carbohydrate and enzyme. Eur. J. Biochem. 1997, 248 (1),
G. C. The atomistic details of the ice recrystallisation inhibition 24−29.
activity of PVA. Nat. Commun. 2021, 12 (1), 1323. (82) Kratochvílová, I.; Golan, M.; Pomeisl, K.; Richter, J.; Sedláková,
(64) Nunes, S. C. C.; Jesus, A. J. L.; Moreno, M. J.; Eusébio, M. E. S. S.; Š ebera, J.; Mičová, J.; Falk, M.; Falková, I.; Ř eha, D.; Elliott, K. W.;
Conformational preferences of α,α-trehalose in gas phase and aqueous Varga, K.; Follett, S. E.; Š imek, D. Theoretical and experimental study
solution. Carbohydr. Res. 2010, 345 (14), 2048−2059. of the antifreeze protein AFP752, trehalose and dimethyl sulfoxide
(65) Zhang, S.; Wang, H.; Luo, J.; Yu, W.; Xiao, Y.; Peng, F. Peach cryoprotection mechanism: correlation with cryopreserved cell
PpSnRK1α interacts with bZIP11 and maintains trehalose balance in viability. RSC Adv. 2017, 7 (1), 352−360.
plants. Plant Physiology and Biochemistry 2021, 160, 377−385. (83) Crowe, L. M.; Reid, D. S.; Crowe, J. H. Is trehalose special for
(66) Stolz, J.; Luyckx; Baudouin, C. Trehalose: an intriguing preserving dry biomaterials? Biophysical journal 1996, 71 (4), 2087−
disaccharide with potential for medical application in ophthalmology. 93.
Clin Ophthalmol 2011, 5, 577−581. (84) Green, J. L.; Angell, C. A. Phase relations and vitrification in
(67) Elbein, A. D.; Pan, Y. T.; Pastuszak, I.; Carroll, D. New insights
saccharide-water solutions and the trehalose anomaly. J. Phys. Chem.
on trehalose: a multifunctional molecule. Glycobiology 2003, 13 (4),
1989, 93 (8), 2880−2882.
17R−27R.
(85) Moore, D. S.; Hand, S. C. Cryopreservation of lipid bilayers by
(68) Furze, C. M.; Delso, I.; Casal, E.; Guy, C. S.; Seddon, C.;
Brown, C. M.; Parker, H. L.; Radhakrishnan, A.; Pacheco-Gomez, R.; LEA proteins from Artemia franciscana and trehalose. Cryobiology
Stansfeld, P. J.; Angulo, J.; Cameron, A. D.; Fullam, E. Structural basis 2016, 73 (2), 240−7.
of trehalose recognition by the mycobacterial LpqY-SugABC (86) Corradini, D.; Strekalova, E. G.; Stanley, H. E.; Gallo, P.
transporter. J. Biol. Chem. 2021, 296, 100307. Microscopic mechanism of protein cryopreservation in an aqueous
(69) Heczko, D.; Grelska, J.; Jurkiewicz, K.; Spychalska, P.; solution with trehalose. Sci. Rep. 2013, 3 (1), 1218.
Kasprzycka, A.; Kamiński, K.; Paluch, M.; Kamińska, E. Anomalous (87) Lins, R. D.; Pereira, C. S.; Hünenberger, P. H. Trehalose−
narrowing of the shape of the structural process in derivatives of protein interaction in aqueous solution. Proteins: Struct., Funct., Bioinf.
trehalose at high pressure. The role of the internal structure. J. Mol. 2004, 55 (1), 177−186.
Liq. 2021, 336, 116321. (88) Budgude, P.; Kale, V.; Vaidya, A. Cryopreservation of
(70) Richards, A. B.; Krakowka, S.; Dexter, L. B.; Schmid, H.; mesenchymal stromal cell-derived extracellular vesicles using trehalose
Wolterbeek, A. P. M.; Waalkens-Berendsen, D. H.; Shigoyuki, A.; maintains their ability to expand hematopoietic stem cells in vitro.
Kurimoto, M. Trehalose: a review of properties, history of use and Cryobiology 2021, 98, 152−163.
human tolerance, and results of multiple safety studies. Food Chem. (89) Ohtake, S.; Schebor, C.; de Pablo, J. J. Effects of trehalose on
Toxicol. 2002, 40 (7), 871−898. the phase behavior of DPPC−cholesterol unilamellar vesicles.
(71) Jain, N. K.; Roy, I. Trehalose and protein stability. Curr. Protoc Biochimica et Biophysica Acta (BBA) - Biomembranes 2006, 1758
Protein Sci. 2010, 59, 1−12. (1), 65−73.

1202 https://doi.org/10.1021/acsbiomaterials.2c01225
ACS Biomater. Sci. Eng. 2023, 9, 1190−1204
ACS Biomaterials Science & Engineering pubs.acs.org/journal/abseba Review

(90) Rudolph, A. S.; Crowe, J. H. Membrane stabilization during (109) Eroglu, A.; Lawitts, J. A.; Toner, M.; Toth, T. L. Quantitative
freezing: The role of two natural cryoprotectants, trehalose and microinjection of trehalose into mouse oocytes and zygotes, and its
proline. Cryobiology 1985, 22 (4), 367−377. effect on development. Cryobiology 2003, 46 (2), 121−34.
(91) Zhang, M.; Oldenhof, H.; Sieme, H.; Wolkers, W. F. Freezing- (110) Uchida, T.; Furukawa, M.; Kikawada, T.; Yamazaki, K.;
induced uptake of trehalose into mammalian cells facilitates Gohara, K. Intracellular trehalose via transporter TRET1 as a method
cryopreservation. Biochim. Biophys. Acta 2016, 1858 (6), 1400−9. to cryoprotect CHO-K1 cells. Cryobiology 2017, 77, 50−57.
(92) Mitchell, D. E.; Fayter, A. E.; Deller, R. C.; Hasan, M.; (111) Sharp, D. M. C.; Picken, A.; Morris, T. J.; Hewitt, C. J.;
Gutierrez-Marcos, J.; Gibson, M. I. Ice-recrystallization inhibiting Coopman, K.; Slater, N. K. H. Amphipathic polymer-mediated uptake
polymers protect proteins against freeze-stress and enable glycerol- of trehalose for dimethyl sulfoxide-free human cell cryopreservation.
free cryostorage. Materials horizons 2019, 6 (2), 364−368. Cryobiology 2013, 67 (3), 305−311.
(93) Lee, J.; Lin, E.-W.; Lau, U. Y.; Hedrick, J. L.; Bat, E.; Maynard, (112) Mercado, S.A.; Slater, N. K. H. Increased cryosurvival of
H. D. Trehalose Glycopolymers as Excipients for Protein Stabiliza- osteosarcoma cells using an amphipathic pH-responsive polymer for
tion. Biomacromolecules 2013, 14 (8), 2561−2569. trehalose uptake. Cryobiology 2016, 73, 175.
(94) Bosch, S.; de Beaurepaire, L.; Allard, M.; Mosser, M.; (113) Holovati, J. L.; Gyongyossy-Issa, M. I. C.; Acker, J. P. Effects
Heichette, C.; Chretien, D.; Jegou, D.; Bach, J. M. Trehalose prevents of trehalose-loaded liposomes on red blood cell response to freezing
aggregation of exosomes and cryodamage. Sci. Rep 2016, 6, 36162. and post-thaw membrane quality. Cryobiology 2009, 58 (1), 75−83.
(95) Zhang, M.; Oldenhof, H.; Sieme, H.; Wolkers, W. F. (114) Bonazzi, M.; Cossart, P. Bacterial entry into cells: A role for
Combining endocytic and freezing-induced trehalose uptake for the endocytic machinery. FEBS Lett. 2006, 580 (12), 2962−2967.
cryopreservation of mammalian cells. Biotechnol. Prog. 2017, 33 (1), (115) Stefanic, M.; Ward, K.; Tawfik, H.; Seemann, R.; Baulin, V.;
229−235. Guo, Y.; Fleury, J. B.; Drouet, C. Apatite nanoparticles strongly
(96) Yoshida, K.; Ono, F.; Chouno, T.; Perocho, B. R.; Ikegami, Y.; improve red blood cell cryopreservation by mediating trehalose
Shirakigawa, N.; Ijima, H. Cryoprotective enhancing effect of very low delivery via enhanced membrane permeation. Biomaterials 2017, 140,
concentration of trehalose on the functions of primary rat 138−149.
hepatocytes. Regenerative Therapy 2020, 15, 173−179. (116) Choimet, M.; Hyoung-Mi, K.; Jae-Min, O.; Tourrette, A.;
(97) Lynch, A. L.; Slater, N. K. H. Influence of intracellular trehalose Drouet, C. Nanomedicine: Interaction of biomimetic apatite colloidal
concentration and pre-freeze cell volume on the cryosurvival of nanoparticles with human blood components. Colloids Surf., B 2016,
rapidly frozen human erythrocytes. Cryobiology 2011, 63 (1), 26−31. 145, 87−94.
(98) Eroglu, A.; Russo, M. J.; Bieganski, R.; Fowler, A.; Cheley, S.; (117) Han, Y.; Wang, X.; Dai, H.; Li, S. Nanosize and Surface
Bayley, H.; Toner, M. Intracellular trehalose improves the survival of Charge Effects of Hydroxyapatite Nanoparticles on Red Blood Cell
cryopreserved mammalian cells. Nat. Biotechnol. 2000, 18 (2), 163− Suspensions. ACS Appl. Mater. Interfaces 2012, 4 (9), 4616−4622.
(118) Rauch, C.; Pluen, A.; Foster, N.; Loughna, P.; Mobasheri, A.;
167.
Lagadic-Gossmann, D.; Counillon, L. On Some Aspects of the
(99) Zhang, T. Y.; Tan, P. C.; Xie, Y.; Zhang, X. J.; Zhang, P. Q.;
Thermodynamic of Membrane Recycling Mediated by Fluid Phase
Gao, Y. M.; Zhou, S. B.; Li, Q. F. The combination of trehalose and
Endocytosis: Evaluation of Published Data and Perspectives. Cell
glycerol: an effective and non-toxic recipe for cryopreservation of
Biochem. Biophys. 2010, 56 (2), 73−90.
human adipose-derived stem cells. Stem Cell Res. Ther. 2020, 11 (1),
(119) Jong, K. S.; Hui, Y. L.; Yu, C. M.; Ki, S. Y.; Kim, S. H.; Pak, H.
460. H. The impact of cryoprotective media on cryopreservation of cells
(100) Janis, B. R.; Priddy, M. C.; Otto, M. R.; Kopechek, J. A.;
using loading trehalose. Cryobiology 2020, 92, 258−259.
Menze, M. A. Sonoporation enables high-throughput loading of (120) Abazari, A.; Meimetis, L. G.; Budin, G.; Bale, S. S.; Weissleder,
trehalose into red blood cells. Cryobiology 2021, 98, 73−79. R.; Toner, M. Engineered Trehalose Permeable to Mammalian Cells.
(101) Stewart, S.; He, X. Intracellular Delivery of Trehalose for Cell PLoS One 2015, 10 (6), No. e0130323.
Banking. Langmuir 2019, 35 (23), 7414−7422. (121) Guo, N.; Puhlev, I.; Brown, D. R.; Mansbridge, J.; Levine, F.
(102) Stoll, C.; Holovati, J. L.; Acker, J. P.; Wolkers, W. F. Trehalose expression confers desiccation tolerance on human cells.
Synergistic effects of liposomes, trehalose, and hydroxyethyl starch for Nat. Biotechnol. 2000, 18 (2), 168−71.
cryopreservation of human erythrocytes. Biotechnol. Prog. 2012, 28 (122) Shinde, P.; Khan, N.; Melinkeri, S.; Kale, V.; Limaye, L.
(2), 364−371. Freezing of dendritic cells with trehalose as an additive in the
(103) Satpathy, G. R.; Török, Z.; Bali, R.; Dwyre, D. M.; Little, E.; conventional freezing medium results in improved recovery after
Walker, N. J.; Tablin, F.; Crowe, J. H.; Tsvetkova, N. M. Loading red cryopreservation. Transfusion 2019, 59 (2), 686−696.
blood cells with trehalose: a step towards biostabilization. Cryobiology (123) Susa, F.; Bucca, G.; Limongi, T.; Cauda, V.; Pisano, R.
2004, 49 (2), 123−136. Enhancing the preservation of liposomes: The role of cryoprotectants,
(104) Shirakashi, R.; Köstner, C. M.; Müller, K. J.; Kürschner, M.; lipid formulations and freezing approaches. Cryobiology 2021, 98, 46−
Zimmermann, U.; Sukhorukov, V. L. Intracellular delivery of trehalose 56.
into mammalian cells by electropermeabilization. J. Membr. Biol. 2002, (124) Crowe, J. H.; Crowe, L. M.; Carpenter, J. F.; Rudolph, A. S.;
189 (1), 45−54. Wistrom, C. A.; Spargo, B. J.; Anchordoguy, T. J. Interactions of
(105) Dovgan, B.; Barlič, A.; Knežević, M.; Miklavčič, D. sugars with membranes. Biochim. Biophys. Acta 1988, 947 (2), 367−
Cryopreservation of Human Adipose-Derived Stem Cells in 84.
Combination with Trehalose and Reversible Electroporation. J. (125) Cai, L.; Nian, L.; Cao, A.; Zhang, Y.; Li, X. Effect of
Membr. Biol. 2017, 250 (1), 1−9. Carboxymethyl Chitosan Magnetic Nanoparticles Plus Herring
(106) Eroglu, A.; Toner, M.; Toth, T. L. Beneficial effect of Antifreeze Protein on Conformation and Oxidation of Myofibrillar
microinjected trehalose on the cryosurvival of human oocytes. Fertility Protein From Red Sea Bream (Pagrosomus major) After Freeze-Thaw
and Sterility 2002, 77 (1), 152−158. Treatment. Food Bioprocess Technol. 2020, 13, 355.
(107) Eroglu, A.; Elliott, G.; Wright, D. L.; Toner, M.; Toth, T. L. (126) Shen, L.; Guo, X.; Ouyang, X.; Huang, Y.; Gao, D.; Zhao, G.
Progressive elimination of microinjected trehalose during mouse Fine-tuned dehydration by trehalose enables the cryopreservation of
embryonic development. Reproductive BioMedicine Online 2005, 10 RBCs with unusually low concentrations of glycerol. J. Mater. Chem. B
(4), 503−510. 2021, 9 (2), 295−306.
(108) Bhowmick, P.; Eroglu, A.; Wright, D. L.; Toner, M.; Toth, T. (127) Kamalifar, S.; Azarpira, N.; Sadeghi, L.; Ghorbani-Dalini, S.;
L. Osmometric behavior of mouse oocytes in the presence of different Nekoei, S. M.; Aghdaie, M. H.; Esfandiari, E.; Azarpira, M. R. ROCK
intracellular sugars. Cryobiology 2002, 45 (2), 183−187. Y-27632 Inhibitor, Ascorbic Acid, and Trehalose Increase Survival of

1203 https://doi.org/10.1021/acsbiomaterials.2c01225
ACS Biomater. Sci. Eng. 2023, 9, 1190−1204
ACS Biomaterials Science & Engineering pubs.acs.org/journal/abseba Review

Human Wharton Jelly Mesenchymal Stem Cells After Cryopreserva- (145) Campbell, L. H.; Brockbank, K. G. M. Culturing with
tion. Exp Clin Transplant 2020, 18 (4), 505−511. trehalose produces viable endothelial cells after cryopreservation.
(128) Stokich, B.; Osgood, Q.; Grimm, D.; Moorthy, S.; Cryobiology 2012, 64 (3), 240−244.
Chakraborty, N.; Menze, M. A. Cryopreservation of hepatocyte
(HepG2) cell monolayers: Impact of trehalose. Cryobiology 2014, 69
(2), 281−290.
(129) Kang, X. L.; Shen, H. Pigmentation of skin graft is improved
by cryopreservation of human skin with trehalose. J. Oral Maxillofac
Surg 2012, 70 (6), 1464−72.
(130) Zhang, T.-Y.; Tan, P.-C.; Xie, Y.; Zhang, X.-J.; Zhang, P.-Q.;
Gao, Y.-M.; Zhou, S.-B.; Li, Q.-F. The combination of trehalose and
glycerol: an effective and non-toxic recipe for cryopreservation of
human adipose-derived stem cells. Stem Cell Res. Ther. 2020, 11 (1),
460.
(131) Dou, M.; Lu, C.; Sun, Z.; Rao, W. Natural cryoprotectants
combinations of l-proline and trehalose for red blood cells
cryopreservation. Cryobiology 2019, 91, 23−29.
(132) Liu, B.; Zhang, L.; Zhang, Q.; Gao, S.; Zhao, Y.; Ren, L.; Shi,
W.; Yuan, X. Membrane Stabilization of Poly(ethylene glycol)-b-
polypeptide-g-trehalose Assists Cryopreservation of Red Blood Cells.
ACS Appl. Bio Mater. 2020, 3 (5), 3294−3303.
(133) Murray, A.; Congdon, T. R.; Tomás, R. M. F.; Kilbride, P.;
Gibson, M. I. Red Blood Cell Cryopreservation with Minimal Post-
Thaw Lysis Enabled by a Synergistic Combination of a Cryoprotect-
ing Polyampholyte with DMSO/Trehalose. Biomacromolecules 2022,
23, 467.
(134) Matsumura, K.; Hayashi, F.; Nagashima, T.; Rajan, R.; Hyon,
S.-H. Molecular mechanisms of cell cryopreservation with poly-
ampholytes studied by solid-state NMR. Commun. Mater. 2021, 2 (1),
15.
(135) Alotaibi, N. A. S.; Slater, N. K. H.; Rahmoune, H. Salidroside
as a Novel Protective Agent to Improve Red Blood Cell
Cryopreservation. PLoS One 2016, 11 (9), No. e0162748.
(136) Liu, X.; Xu, Y.; Liu, F.; Pan, Y.; Miao, L.; Zhu, Q.; Tan, S. The
feasibility of antioxidants avoiding oxidative damages from reactive
oxygen species in cryopreservation. Front. Chem. 2021, 9, 64. Recommended by ACS
(137) Crowe, J. H.; Crowe, L. M. Preservation of mammalian cells -
Learning nature’s tricks. Nat. Biotechnol. 2000, 18 (2), 145−146. Human Natural Killer Cells Cryopreserved without DMSO
(138) Zambelli, A.; Poggi, G.; Da Prada, G.; Pedrazzoli, P.; Cuomo, Sustain Robust Effector Responses
A.; Miotti, D.; Perotti, C.; Preti, P.; Robustelli della Cuna, G. Clinical
Soumyajit Das, Sandro Matosevic, et al.
toxicity of cryopreserved circulating progenitor cells infusion.
JANUARY 17, 2024
Anticancer Res. 1998, 18 (6B), 4705.
MOLECULAR PHARMACEUTICS READ
(139) Ferrucci, P. F.; Martinoni, A.; Cocorocchio, E.; Civelli, M.;
Cinieri, S.; Cardinale, D.; Peccatori, F. A.; Lamantia, G.; Agazzi, A.;
Corsini, C.; Tealdo, F.; Fiorentini, C.; Cipolla, C. M.; Martinelli, G. Physicochemical Mechanisms of Protection Offered by
Evaluation of acute toxicities associated with autologous peripheral Agarose Encapsulation during Cryopreservation of
blood progenitor cell reinfusion in patients undergoing high-dose Mammalian Cells in the Absence of Membrane-Penetrati...
chemotherapy. Bone Marrow Transplant 2000, 25 (2), 173−7. Mian Wang, Alptekin Aksan, et al.
(140) Chen-Plotkin, A. S.; Vossel, K. A.; Samuels, M. A.; Chen, M. MAY 22, 2023
H. Encephalopathy, stroke, and myocardial infarction with DMSO use ACS APPLIED BIO MATERIALS READ
in stem cell transplantation. Neurology 2007, 68 (11), 859−61.
(141) Antonenas, V.; Shaw, P. J.; Bradstock, K. F. Infusion of Engineered Test Tissues: A Model for Quantifying the Effects
unwashed umbilical cord blood stem cells after thawing for allogeneic of Cryopreservation Parameters
transplantation. Bone Marrow Transplant 2004, 34 (8), 739.
Jonian Grosha, Marsha W. Rolle, et al.
(142) Perotti, C. G.; Fante, C. D.; Viarengo, G.; Papa, P.; Rocchi, L.;
OCTOBER 06, 2023
Bergamaschi, P.; Bellotti, L.; Marchesi, A.; Salvaneschi, L. A new ACS BIOMATERIALS SCIENCE & ENGINEERING READ
automated cell washer device for thawed cord blood units. Transfusion
2004, 44 (6), 900−906.
(143) Besterman, J. M.; Airhart, J. A.; Woodworth, R. C.; Low, R. B. Investigation of Rapid Rewarming Chips for
Exocytosis of pinocytosed fluid in cultured cells: kinetic evidence for Cryopreservation by Joule Heating
rapid turnover and compartmentation. J. Cell Biol. 1981, 91 (3), 716− Hengxin Han, Yi Xu, et al.
727. JULY 27, 2023
(144) Kikawada, T.; Saito, A.; Kanamori, Y.; Nakahara, Y.; Iwata, K.- LANGMUIR READ
i.; Tanaka, D.; Watanabe, M.; Okuda, T. Trehalose transporter 1, a
facilitated and high-capacity trehalose transporter, allows exogenous
Get More Suggestions >
trehalose uptake into cells. Proc. Natl. Acad. Sci. U. S. A. 2007, 104
(28), 11585.

1204 https://doi.org/10.1021/acsbiomaterials.2c01225
ACS Biomater. Sci. Eng. 2023, 9, 1190−1204

You might also like