You are on page 1of 23

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/348568444

Integrating Biomaterials and Food Biopolymers for Cultured Meat Production

Article in Acta Biomaterialia · January 2021


DOI: 10.1016/j.actbio.2021.01.017

CITATIONS READS

64 3,446

2 authors, including:

Motoichi Kurisawa
Agency for Science, Technology and Research (A*STAR)
113 PUBLICATIONS 7,106 CITATIONS

SEE PROFILE

All content following this page was uploaded by Shengyong Ng on 15 August 2021.

The user has requested enhancement of the downloaded file.


JID: ACTBIO
ARTICLE IN PRESS [m5G;February 21, 2021;17:50]
Acta Biomaterialia xxx (xxxx) xxx

Contents lists available at ScienceDirect

Acta Biomaterialia
journal homepage: www.elsevier.com/locate/actbio

Review article

Integrating biomaterials and food biopolymers for cultured meat


production
Shengyong Ng∗, Motoichi Kurisawa∗
Institute of Bioengineering and Nanotechnology (A∗ STAR), 31 Biopolis Way, The Nanos, 138669, Singapore

a r t i c l e i n f o a b s t r a c t

Article history: Cultured meat has recently achieved mainstream prominence due to the emergence of societal and indus-
Received 2 September 2020 trial interest. In contrast to animal-based production of traditional meat, the cultured meat approach en-
Revised 18 December 2020
tails laboratory cultivation of engineered muscle tissue. However, bioengineers have hitherto engineered
Accepted 11 January 2021
tissues to fulfil biomedical endpoints, and have had limited experience in engineering muscle tissue for
Available online xxx
its post-mortem traits, which broadly govern consumer definitions of meat quality. Furthermore, existing
Keywords: tissue engineering approaches face fundamental challenges in technical feasibility and industrial scala-
Cultured meat bility for cultured meat production. This review discusses how animal-based meat production variables
Food biopolymers influence meat properties at both the molecular and functional level, and whether current cultured meat
Muscle approaches recapitulate these properties. In addition, this review considers how conventional meat pro-
Adipose ducers employ exogenous biopolymer-based meat ingredients and processing techniques to mimic desir-
Tissue engineering
able meat properties in meat products. Finally, current biomaterial strategies for engineering muscle and
adipose tissue are surveyed in the context of emerging constraints that pertain to cultured meat produc-
tion, such as edibility, sustainability and scalability, and potential areas for integrating biomaterials and
food biopolymer approaches to address these constraints are discussed.

Statement of Significance

Laboratory-grown or cultured meat has gained increasing interest from industry and the public, but cur-
rently faces significant impediment to market feasibility. This is due to fundamental knowledge gaps in
producing realistic meat tissues via conventional tissue engineering approaches, as well as translational
challenges in scaling up these approaches in an efficient, sustainable and high-volume manner. By defin-
ing the molecular basis for desirable meat quality attributes, such as taste and texture, and introducing
the fundamental roles of food biopolymers in mimicking these properties in conventional meat products,
this review aims to bridge the historically disparate fields of meat science and biomaterials engineering
in order to inspire potentially synergistic strategies that address some of these challenges.
© 2021 Acta Materialia Inc. Published by Elsevier Ltd. All rights reserved.

1. Introduction tured meat field to mature and demonstrate its commercial


feasibility.
Global trends in climate change and meat consumption high- Biomaterial-based scaffolds for engineering muscle or adipose
light an increasing need to complement animal-based meat pro- tissue are typically designed to fulfil biomedical endpoints, includ-
duction with alternative dietary protein sources. The emergence of ing efficient cellular differentiation, functional robustness and post-
cellular agriculture start-ups on the technological horizon, begin- transplantation viability. While these design principles fulfil neces-
ning with a focus on plant-derived proteins, is punctuated with sary intermediary requirements for cultured meat, they may not
a recent thrust towards cultured meat that comprises animal- suffice to meet its ultimate functional endpoint, which is to sat-
derived proteins [1]. This trend points to an increasing demand isfy consumer definitions of meat quality more cost-efficiently and
for bioengineering solutions that would allow the nascent cul- scalably than current cultured meat manufacturing models. It may
therefore be instructive for bioengineers to understand how meat
quality attributes like appearance, texture, flavour and nutritional

Corresponding authors. composition fundamentally relate to complex molecular processes
E-mail addresses: syng@ibn.a-star.edu.sg (S. Ng), mkurisawa@ibn.a-star.edu.sg that occur during meat production, ranging from in vivo muscle
(M. Kurisawa).

https://doi.org/10.1016/j.actbio.2021.01.017
1742-7061/© 2021 Acta Materialia Inc. Published by Elsevier Ltd. All rights reserved.

Please cite this article as: S. Ng and M. Kurisawa, Integrating biomaterials and food biopolymers for cultured meat production, Acta
Biomaterialia, https://doi.org/10.1016/j.actbio.2021.01.017
JID: ACTBIO
ARTICLE IN PRESS [m5G;February 21, 2021;17:50]

S. Ng and M. Kurisawa Acta Biomaterialia xxx (xxxx) xxx

development to its post-mortem transformation into meat, and its tion of intracellular glycogen triggers irreversible actomyosin com-
subsequent processing into meat products. A deeper insight into plex formation and permanent myofibre contraction (rigour mortis),
how the meat production industry augments the quality, value and whereas a steady state acidic pH that matches the isoelectric point
range of conventional meat products, using food-grade ingredi- of the actomyosin complex results in a decrease in water binding
ents and processing techniques in cost-effective and scalable ways, capacity [8,11–13].
could open up opportunities for combinatorial approaches that in- Current cultured meat technology entails the expansion and
tegrate food and biomedical materials to design practicable strate- subsequent differentiation of muscle precursor cells into fused
gies that address the above challenges. myotubes that comprise the fundamental building block of mus-
In this review, we first discuss how animal-based meat pro- cle tissue, which is indispensable for structured meat produc-
duction variables influence molecular-level material properties tion. However, unstructured meat, which may not preserve tissue-
of meat, how these molecular properties relate functionally to level structure, could require a simplified production process in-
macroscopic meat properties that the consumer experiences, and volving only cell expansion or even industrial fermentation for
whether current cell-based meat production recapitulates these recombinant meat protein production [6]. The analogous ver-
properties. We then illustrate how biopolymer-based meat ingre- sion of rigour mortis in cultured meat would theoretically oc-
dients contribute to desirable meat properties in semi-structured cur with a comparable time delay after perfusion within the dif-
and unstructured meat products, and explore how they may pro- ferentiation bioreactor with culture medium is terminated, like
mote consumer acceptance of future cultured meat products. Next, in animal-derived meat. In contrast to animal-based meat, post-
we survey conventional bioengineering strategies for promoting mortem metabolism of cultured meat is relatively uncharacter-
expansion and differentiation of muscle and adipose precursor ized, and basic studies to assess this process vis-à-vis animal-
cells and critically evaluate their feasibility for cultured meat pro- based meat are required given its significant contribution to meat
duction. Finally, we consider novel biomaterial constraints, such properties.
as the use of degradable, edible and animal-free scaffolds that
mitigate culture media costs and contribute to desirable meat 2.1. Appearance
quality attributes, and explore the integration of biomaterials and
food engineering approaches for cultured meat production. Al- The appearance of meat is a function of its colour and opac-
though this review focuses on specific scientific and technolog- ity, and greatly influences the consumer decision to purchase it
ical challenges that the cultured meat field faces, we recog- [14]. Myoglobin, a major determinant of meat colour, exists in a
nize that its long-term market feasibility also depends on ad- brownish, oxidized, non-oxygen-binding form (metmyoglobin), or a
dressing challenges posed by the environmental, socio-political purplish-red, reduced form (reduced myoglobin) that binds oxygen
and regulatory domains, as discussed in several recent reviews to form the attractive bright red oxymyoglobin that characterizes
[2–4]. fresh meat [8,11]. Meat oxygenation levels, which determine rela-
tive concentrations of myoglobin isoforms, is a complex function
2. How animal-based meat production influences meat of pre-slaughter animal stress levels and post-slaughter conditions
material properties and quality [8,11]. Furthermore, intrinsic myoglobin concentration varies with
animal species and tissue location, with beef being darker than
A fundamental motivation of the cultured meat industry is poultry, and chicken thigh tissues being darker than chicken breast
to complement traditional meat production to satisfy increas- tissue, for example [10,15].
ing consumer demand for sustainable meat products. However, In contrast to meat colour, the typical opaque appearance of
this demand is tempered by consumer unwillingness to com- meat compared with live muscle tissue is largely a function of the
promise on the organoleptic properties of meat [5]. Therefore, decreased myofibre water-binding capacity that results from post-
to increase consumer acceptance and ensure commercial viabil- mortem tissue acidification [8,11,12,16]. Pre-slaughter animal stress
ity, cultured meat should closely mimic conventional meat in may cause premature depletion of glycogen stores, decreased post-
terms of appearance, structure, texture, flavour and nutritional mortem anaerobic respiration, and a higher resultant pH than the
composition [6]. The wide variety of conventional meat prod- isoelectric point of actomyosin, leading to increased water binding,
ucts may facilitate earlier market entry of cultured meat prod- less light-scattering and a darker appearance that appears overly
ucts, as different products present different levels of technologi- dry and less fresh to consumers, thus illustrating the role of opac-
cal barriers to effective mimicry. For example, a full-cut steak re- ity in meat appearance [8,12].
quires a much higher similarity to freshly slaughtered meat than Conventional myogenic differentiation protocols culture muscle
a fine-ground sausage does. The latter demonstrates the degree precursor cells at ambient oxygen conditions (20–21% O2 ), under
to which conventional meat processing alters tissue and cellu- which myoglobin expression is suppressed and the cultured mus-
lar structures in the final product [6,7], suggesting that com- cle typically exhibits a pale appearance [6,17]. Possible approaches
plete replication of original muscle structure may not always be to increasing myoglobin expression include introducing hypoxia
necessary. [17–19] or supplementing iron, lipids or even myoglobin during
A complex interplay of pre- and post-mortem variables influ- culture [19,20]. The addition of myoglobin or iron-containing mim-
ence animal-based meat properties. Inter- or intra-species genetic ics, such as soy leghaemoglobin used in Impossible©, the plant-
variation due to natural or artificial selection, or individual vari- based meat analogue [21], or natural colorants such as beetroot
ation due to gender or age, rearing environment and diet rep- juice [22,23], offer alternative methods to replicate the reddish
resent diverse pre-mortem factors that meat scientists study to colour of meat post-culture. To ensure that cultured meat appears
optimize meat properties [8–11]. Concomitantly, the post-mortem similar to animal-based meat, the colour of the former should be
transformation of muscle tissue into meat is a sophisticated sci- quantified according to the three-dimensional colour space sys-
ence, with post-mortem processing, storage conditions and pre- tem developed by the Commission Internationale d’Léclairage (CIE),
slaughter animal conditioning designed to preserve desirable meat CIE L∗ a∗ b∗ , where L∗ indicates the brightness axis, a∗ indicates the
quality [8,11–13]. Post-mortem transformation of muscle tissue be- red-green axis and b∗ indicates the yellow-blue axis [8,24]. Both
gins with the development of tissue hypoxia after blood perfusion instrumental methods (e.g. colorimeters, spectrophotometers) and
ceases, which prompts a metabolic switch to anaerobic respiration trained visual appraisal panels may be used to assess meat colour
and results in decreased intracellular pH [8,11–13]. The exhaus- [8,24].

2
JID: ACTBIO
ARTICLE IN PRESS [m5G;February 21, 2021;17:50]

S. Ng and M. Kurisawa Acta Biomaterialia xxx (xxxx) xxx

2.2. Flavour expression [50], a lack of overall myotube alignment, and the ab-
sence of an appropriate connective tissue mimetic, any of which
Although the cooking processing generates much of meat could affect the various properties that underlie meat texture
flavour, various pre-cooking factors contribute significantly too. [47,51]. Appropriate scaffold design may enhance myofibre align-
Meat flavour intensity increases with animal age at slaughter, and ment and maturation, or promote cellular production of extracel-
free-range animals raised on curated or foraged diets generate lular matrix (ECM) molecules that comprise connective tissue, thus
terroir-specific flavours [8,25–27]. Fat is a major determinant of recapitulating their textural contributions in cultured meat (see
species-specific flavours, which are associated with distinct fatty Section 4.2.2). Additionally, gel-forming or texturized biopolymers
acid profiles and the varied diet-derived aromatic compounds that may independently substitute for meat texture, a common strat-
fat is a vehicle for [28,29]. Post-mortem ageing of meat further egy in unstructured meat and meat analogues (see Sections 3.2,
generates umami-generating amino acids and aromatic compounds 3.5) [52,53].
via proteolysis and lipolysis respectively [8,11]. Cooking intensi- Given the major role of post-mortem processes on meat texture,
fies meat flavour by generating volatile compounds from thermal their time- and environment-dependant effects on cultured meat
degradation of fat, and carbonyl compounds from the Maillard re- after the termination of culture medium perfusion should be char-
action associated with the browning flavour of well-cooked meat acterized. Well-established instrumental methods in the meat in-
[8,30–32]. dustry that directly quantify meat texture include the texture pro-
The specialization of several start-ups on cell-based fat produc- file analysis (a two-bite simulation compression test), which mea-
tion highlights the potential importance of fat in cultured meat sures firmness, springiness, cohesiveness, gumminess and chewi-
flavour [33,34]. While differentiation of fat precursor cells into ness [54], and the Warner-Bratzler shear force test, which mea-
adipocytes is technically feasible [35–39], certain limitations re- sures the perpendicular shear force needed to slice through mus-
main to be overcome, including the use of small molecules with cle fibres [42,55]. Other indirect quantitative methods measure wa-
uncharacterized food safety profiles and the generation of imma- ter holding capacity and myofibre anisotropy using various elec-
ture adipocytes [40,41]. Furthermore, the fabrication of structured tromagnetic modalities to infer meat texture [11,44,55,56]. As it is
meat containing both muscle and adipose cells requires complex non-trivial to correlate instrumental readouts with consumer ac-
co-culture or post-culture assembly methods that are currently ceptance of meat texture, standardized “descriptive attribute” sen-
lacking. In contrast, incorporating cultured fat into unstructured sory evaluations of meat juiciness, connective tissue amount and
meat products should be relatively facile and analogous to the mix- tenderness, performed by trained consumer panellists, are ulti-
ing of fat and lean meat from different sources in conventional mately required to capture the complexity of the consumer ex-
finely ground meat products [8]. perience and provide a more objective evaluation [42]. Due to an
Given the importance of post-mortem ageing and cooking in increasing appreciation of the importance of texture on poten-
generating flavour precursor molecules, it is critical to simulate tial consumer acceptance of cultured meat, recent studies have
both processes when comparing the flavour of cultured meat pro- performed texture analysis or sensory panel studies on cultured
totypes with animal-based meat [6]. The gold standard of meat meat prototypes in comparison with animal-based meat, and high-
flavour analysis involves the assessment of cooked meat flavour light the importance of scalable production methods for testing
by trained consumer sensory panels, using quantitative descriptive [6,57,58].
analysis of meat aroma, tastes and aftertastes [42], which requires
relatively large samples. In contrast, conventional electromagnetic 2.4. Nutrition
methods (e.g. ultrasound, light diffraction, optical impedance) mea-
sure meat composition in terms of fat and collagen content, Although consumers do not typically consider nutritional con-
thereby providing an estimate of meat flavour, whereas emerging tent when purchasing meat, meat is a nutrient-dense food that
molecular profiling methods (e.g. liquid chromatography – mass provides important amino acids, fatty acids, vitamins and other
spectrometry) directly quantify levels of meat flavour molecules minerals not readily found in other food classes. To fundamen-
using smaller sample sizes that current cultured meat methods can tally address the longer-term challenge of sustainably producing
produce [11,31,43,44]. alternative dietary protein, cultured meat producers should aim to
match the nutritional content of animal-based meat. Given that
2.3. Texture current in vitro methods for generating cultured muscle and adi-
pose tissue result in incomplete maturation [50], increasing cell
Meat texture, an interplay of tenderness, juiciness and lubric- differentiation efficiency and thereby cellular protein or lipid pro-
ity at the consumer level, is a complex function of molecular- duction is a subject of ongoing research in the biomedical field.
level properties such as firmness, and fat or water binding capacity The likely requirement for biomaterial-based scaffolds to
[45], which in turn are linked to the hierarchical organization of facilitate scalable cell expansion and cell differentiation (see
myofibers, connective tissue and intramuscular fat, and the effect Sections 4.1, 4.2) introduces both challenges and opportunities
of post-mortem ageing [6,8,12,46,47]. To ensure tender and juicy in designing the nutritional composition of cultured meat. One
meat, meat producers slaughter younger and fatter animals, hang technical challenge is the need to support high cell densities to
carcasses to minimize post-mortem muscle contraction prior to the match those in native tissue (108 – 109 cells/mL) [59]. However,
onset of rigour mortis, age meat to facilitate endogenous enzymatic most research into engineering muscle tissue has been performed
degradation, and control temperatures to avoid meat toughening at relatively low cell densities (107 cells/mL) [58,60–63], and the
[6,8,11,46,48]. Pre-cooking strategies, such as adding enzymes or need for high encapsulated cell densities may require scaffold re-
acid to degrade or solubilize connective tissue, and slow cook- formulation for ensuring fabrication feasibility and cellular mi-
ing methods for tougher cuts, further enhance meat tenderness croenvironmental optimization. Alternative approaches to maxi-
[8,11,48,49]. mize protein content in cultured meat prototypes include the use
The pioneer cultured meat prototype exhibited a dry texture, of edible, proteinaceous scaffolds, or biodegradable scaffolds that
which was attributed to it comprising only myotubes, and high- are replaced by cells and their secreted ECM proteins during cul-
lights the importance of fat to the texture of future cultured ture [6].
meat products [17]. Other limitations of current cultured meat ap- Not all nutritional components of meat are produced by en-
proaches include low levels of actin and myosin, immature isoform dogenous cells [6] and may require supplementation. For example,

3
JID: ACTBIO
ARTICLE IN PRESS [m5G;February 21, 2021;17:50]

S. Ng and M. Kurisawa Acta Biomaterialia xxx (xxxx) xxx

Molecular process Gelling Water binder Thickening Fat


agent (G) (W) agent (T) emulsifier (F)
Function in meat
Texture modulation G W T F G W T F G W T F
Flavour modulation G W T F G W T F G W T F
Bulk structural integrity G W T F G W T F G W T F
Water, brine retention G W T F G W T F G W T F
Spoilage delay G W T F G W T F G W T F
Fat replacement G W T F G W T F G W T F
Protein replacement G W T F G W T F G W T F
Emulsified/ Comminuted/
Structured
Meat product type unstructured chopped/
(steak, whole-
(Examples) (fine-grain semi-structured
muscle ham)
sausage) (burger, meatloaf)

Fig. 1. Role of ingredients added to conventional meat products.

fatty acids such as conjugated linoleic acid and docosahexaenoic flavour can be replicated by various biopolymers (Table 1), which
acid are synthesized by ruminant gut-specific biohydrogenation or were developed partly to mitigate the potentially negative health
algae respectively, and not by cultured adipocytes [29,64]. Other effects of excessive dietary phosphates and salt and are detailed in
micronutrients are either not produced by muscle or fat tissue, the following sections.
such as microbial-derived vitamin B12 [65], or are packaged in a
more easily absorbed form in meat, such as haem-bound iron and
zinc [6]. Increasing consumer preference for healthier diets also 3.1. Product stability
encourage meat producers to introduce healthier fatty acids into
their products [66]. Due to the loss of native tissue structure, restructured and un-
structured meat products often require the addition of biopoly-
mers that provide either thickening or gelling functions to enhance
3. Enhancing meat quality attributes with food-grade product formability and retain structural integrity under storage
biopolymers and cooking conditions. Hydrocolloids that swell and are typically
non-gelling at low temperatures, such as xanthan gum, guar gum,
Meat processing is integral to the production of various meat locust bean gum and pre-swollen starches, act as thickeners that
products, ranging from structured whole-muscle hams, semi- improve the formability of uncooked minced meat products and
structured burgers, to unstructured meat emulsions such as often suffice for maintaining product integrity (Table 1) [8,11,52]. In
sausages [8,11,52], and offer bioengineers practical lessons in the addition, their water binding capacity contributes to reduced water
use of food-safe biopolymers and industrial processing methods loss during storage and cooking.
to maximize the organoleptic and economic value of slaughtered Gel-forming materials are often employed to enhance product
meat without compromising consumer health. Ingredients added formability where thickeners alone do not suffice, such as in re-
to meat fulfil several broad functions that overlap, including tex- structured meat products comprising larger meat pieces (reformed
ture and flavour modulation, product stability, fat or protein re- steaks, surimi) [8,11,52]. In addition to their structural roles, gels
placement, and delivery of bioactive compounds (Fig. 1). These applied as films or coatings also reduce water loss during storage
functions define meat quality and are underpinned by molecular- and act as an oxidation barrier, thereby improving storage stabil-
level solubilisation of meat proteins, resulting in secondary molec- ity [66,67]. Typical examples of gelling biopolymers include hy-
ular processes such as water binding, thickening, fat emulsification drocolloids (e.g. alginate, κ -carrageenan, gellan gum, agar), pre-
and gelation [8,11,52]. swollen starches and proteins (e.g. gelatine, soy or pea isolates,
The use of phosphates and salt in the first step of traditional and whey) (Table 1) [8,11,52], the majority of which form phys-
meat processing illustrates several of these fundamental processes. ically crosslinked hydrogels in a thermo-responsive manner, with
Alkaline phosphates ionically disrupt the actin-myosin complexes the key exception of alginate which forms an ionically crosslinked,
formed during rigour mortis, thereby solubilizing this major source cold-set gel (Box 1). A common gelation mechanism involves the
of meat protein. Both phosphate and salt increase meat pH away spontaneous aggregation of biopolymer chains upon cooling a
from the isoelectric point of actin-myosin, thus increasing water heated biopolymer solution below its sol-gel transition temper-
binding capacity [8,11,12,16]. Protein solubilisation and subsequent ature, which characterizes κ -carrageenan, gellan gum, agar, pre-
water binding lead to an overall increase in meat tenderness and swollen starches and gelatine gels [8]. In contrast, physical gela-
juiciness. Additionally, solubilized meat proteins intrinsically emul- tion of soy, pea and whey protein isolates involve thermal denatu-
sify fat, which helps to retain its lubricity, and also undergo in- ration of the globular proteins and subsequent hydrophobic aggre-
termolecular binding and gel formation, which contribute to struc- gation in a pH- and salt-dependant manner [68,69]. The require-
tural integrity in less structured meat products [8,11]. Although the ment for high temperatures to solubilize gelling hydrocolloids or
protein solubilisation activity of phosphate is irreplaceable, the wa- denature gelling proteins limits the use of most physical crosslink-
ter binding capacity, fat emulsification and protein gelation that it ing methods to the production of cooked meat products. However,
engenders and their desirable contributions to meat texture and the development of enzymatic methods using microbial transglu-

4
JID: ACTBIO
ARTICLE IN PRESS [m5G;February 21, 2021;17:50]

S. Ng and M. Kurisawa Acta Biomaterialia xxx (xxxx) xxx

Table 1
Biopolymer ingredients in conventional meat products.

Biopolymer Gelling agents Water binder Thickener, stabilizer Fat emulsifier Fat replacer Protein replacer
√ √ √ √
Polysaccharides Alginate
√ √ √ √
κ -carrageenan
√ √ √ √
Gellan gum

Agar
√ √ √
Konjac ∼ 1
√2 √ √
Pectin
√ √ √ √
Xanthan gum ∼ 3
√ √ √
Locust bean ∼ 4
gum
√ √
Guar gum
√ √
Cellulose
√ √ √ √ √
Modified starch
√ √ √ √ √ √
proteins gelatine
√ √ √ √ √ √
Soy isolate
√ √ √ √ √
Pea isolate
√ √
Whey

Wheat gluten

Lipids Lecithin

Monoglycerides
& diglycerides
1. Forms thermoreversible gel when mixed with xanthan, κ -carrageenan or starch; also forms thermostable gel at pH > 9.5 and above 85°C but would
denature meat proteins and is not used in meat processing.
2. Low-methoxyl pectin forms thermoreversible, calcium-dependant ionic gels at pH < 6, unlike the 55% sugar and pH < 3.5 required for high-methoxyl
pectin to gel.
3. Forms gel when mixed with konjac or locust bean gum.
4. Forms gel when mixed with xanthan or κ -carrageenan. References: [8,11,52].

taminase and recombinant fibrinogen/thrombin allows the use of tributing to juiciness, lubricity and inhibiting the development of
gelling proteins for uncooked meat products by supporting the for- rancid flavours [8,52,73]. However, the degree of gelation should
mation of covalently crosslinked proteins gels at low temperatures be judiciously optimized as it may negatively affect the appearance
[70]. (unnatural, glassy, stretch marks) and flavour intensity of meat
[8,74,75].
3.2. Texture and flavour modulation
3.3. Fat and protein replacement
From a consumer perspective, desirable meat texture is per-
ceived as a complex interplay of tenderness, juiciness and lubricity Due to increasing consumer demand for healthier products,
[45], and biopolymer ingredients added to meat generally aim to meat producers have attempted to incorporate either healthier fat
replicate these properties by promoting water binding, gelation or or less fat in their products [66,76]. The first approach is to sim-
fat stabilization (Table 1) [8,11,52]. ply replace fat with combinations of water-binding starch, hydro-
As fat is a major source of meat flavour, effective fat stabiliza- colloids, or proteins [52,77,78], which contribute fewer calories
tion contributes significantly to flavour retention. Effective emulsi- and partially substitute fat-derived succulence with increased wa-
fiers reduce the separation of fat and water in meat products un- ter content. A second approach is to mimic fat textural properties
der both storage and cooking conditions, thus contributing to the using certain starches that exhibit a soft and creamy mouthfeel
retention of fat-associated flavour, juiciness and lubricity [8,11]. Ex- due to their rheological similarity to fat, such as oat-derived mal-
ogenous proteins from animal (e.g. caseinate, egg) and plant (e.g. todextrin and rice starch [8,52]. A third approach is to replace fat
gluten, soy, pea) sources are introduced to supplement the emul- with healthier versions like plant-based oils, which tend to exhibit
sifying properties of solubilised meat proteins [8,52]. Non-protein low melting points and require oil bulking agents such as alginate,
emulsifiers are commonly employed and include various lipids (e.g. konjac and cellulose or microencapsulation techniques to improve
lecithin, monoglycerides and diglycerides of fatty acids), modified their retention [66,79–81].
starches and more recently, pectin [52,71,72]. Meat proteins are often replaced with non-meat, non-protein
Non-gelling gum- and starch-based thickeners stabilize emulsi- extenders like plant-derived fibre and hydrocolloids for economic
fied fat primarily via their viscosity, whereas their high water bind- reasons [8,11,52], which merely substitute the mass, but not the
ing capacities efficiently retain injected brines in raw meat prod- nutritional value, of the replaced protein. Non-meat proteins may
ucts. These properties decrease fat and water loss during both stor- also replace meat proteins, and are used in meat analogue pro-
age and cooking, therefore enhancing the lubricity, juiciness and duction for vegetarians. In addition to being a nutritional re-
flavour of the cooked meat [8,52]. placement, these non-meat proteins should also fulfil the typical
Gel-forming biopolymers contribute significantly to the replica- molecular functions of meat proteins described above, including
tion of meat-like firmness in unstructured products that do not fat emulsification, gelation, and water binding. Soy and pea pro-
preserve native tissue structure, such as sausages. Even restruc- tein are major plant-based protein replacers, but tend to exhibit
tured whole-muscle ham is injected with gelling agents like car- favourable properties only in highly purified protein isolate forms
rageenan to compensate for the partial loss of original meat tex- [8,52]. For example, soy isolates containing 92% protein are in-
ture due to the protein solubilisation required for optimal meat jected into whole muscle hams to add firmness via its gelling be-
binding and fat emulsification [8]. In addition to providing firm- haviour, but soy concentrates containing 71% protein cannot form
ness, gelation facilitates the retention of moisture and fat, and en- a hydrogel due to the presence of insoluble carbohydrates [8].
ables the delivery of antioxidants in meat products, therefore con- Novel alternative protein sources include algae, fungi and insects,

5
JID: ACTBIO
ARTICLE IN PRESS [m5G;February 21, 2021;17:50]

S. Ng and M. Kurisawa Acta Biomaterialia xxx (xxxx) xxx

and currently exist as niche, standalone products that are not yet
widely used as protein replacers in conventional meat products scribed use of alginate in restructured meat utilized a com-
[82,83]. bination of alginate, calcium carbonate and an acid precur-
sor glucono-delta-lactone (GDL). GDL delays calcium ion re-
3.4. Relevance of food-grade biopolymers to cultured meat lease from calcium carbonate and enables proper mixing of
meat parts with alginate before ionic gelation [98]. The use
of alginate improves upon the use of phosphates and salt in
Table 1 summarizes major examples of biopolymers employed earlier versions of restructured meat, which solubilize meat
to replicate meat quality attributes during post-slaughter meat proteins and increase meat binding, but also introduce di-
processing or meat analogue production. In principle, similar etary concerns and require frozen storage to retain product
biopolymer ingredients and processes could be applied to cultured integrity. However, the use of calcium for alginate gelation
meat during post-culture processing to endow them with compa- imparts some bitterness to the product, which inevitably re-
rable properties to animal-based meat products. quires masking via sodium addition and partially negates the
The lower content of immature actin-myosin isoforms in my- envisioned role of alginate in reducing dietary sodium in pro-
otubes and lack of connective tissue in current cultured meat pro- cessed meats.
The calcium-induced gelation of alginate at low temper-
totypes [50] could lead to a smaller effect of rigour mortis, which
ature further enables its use as an oil bulking agent and
may decrease the need for phosphate-dependant protein solubili-
enables the replacement of animal fat in meat batters and
sation and suggest a supplementary role for protein-based ingredi- sausages with healthier plant-derived oils, which would oth-
ents in order to ensure nutritional similarity to slaughtered meat. erwise be poorly retained due to their relatively low melting
In addition, the lack of tissue-level structure in current cultured temperatures [79].
meat models may necessitate the use of gelling agents that provide Although the gelation mechanism of alginate limits it to
structural integrity and replicate meat-like texture. Gelling agents batch-type production processes, this is beneficial in certain
may continue to play major roles in future cultured meat products seafood analogues that replace animal proteins with non-
that are designed to comprise single cells, cell lysates or even re- animal soy protein isolates, where alginate layering tech-
combinant meat proteins [6]. niques are applied to mimic the flake-like structure of fish
fillets [67,85].
When future cultured meat development reaches the stage of
As a film coating, alginate helps to delay rancidity and
generating macroscale cultured meat units that resemble coarsely colour changes in frozen meat and fish [67,99]. Alginate
chopped meat, gelling agents could play novel roles in their post- coated meat patties also exhibit improved water retention,
culture modular assembly into even larger cultured meat con- which reduces product shrinkage during storage, and in-
structs using additive manufacturing techniques [84], analogous to creases cooking yields, tenderness and flavour retention dur-
their current role in restructured meat production (see Box 1). If ing cooking [100].
cultured meat technology attains the stage of one-step production
of cultured full-cut meat, water-binding agents like cold-swelling
gums or starches may become helpful to retain water and augment
product juiciness upon cooking. Biopolymer-based microencapsula- 4. Adapting biomaterial design strategies from biomedical
tion or coating strategies for the delivery of bioactive compounds engineering to the cultured meat process
can also be readily adapted to post-culture processing of cultured
meat at various stages of complexity. The original cultured meat prototype in 2013 comprised thou-
While not a typical component of conventional meat process- sands of self-assembled myotubes that were individually cultured
ing, the structuring processes employed to produce completely in manually fabricated chambers, and these myotubes represent
animal-free meat analogues may be instructive for cultured meat the most mature form of muscle cells currently achievable [101].
production. Such methods aim to mimic the fibrous texture of Although absent from this initial prototype, adipose tissue will be
structured meat and include both top-down (e.g. extrusion, shear considered an essential cell type in future cultured meat prod-
cell, or freeze structuring) and bottom-up (e.g. wet spinning, elec- ucts. Broadly, the generation of mature myotubes and adipose cells
trospinning) methods [53,85–94]. Although developed primarily comprises two main stages: 1) expansion of progenitor cells, and
for non-animal protein sources such as plants or fungi [95–97], 2) differentiation of expanded progenitor cells into myotubes and
such structuring methods could allow whole-cell cultured my- subsequently myofibers, or adipocytes. Suitable progenitors include
ocytes, cell lysates, or even recombinant animal protein, to be in- lineage-committed cells, such as myoblasts and muscle satellite
troduced seamlessly to form hybrid meat analogues comprising cells from skeletal muscle, or preadipocytes from adipose tissue.
a portion of culture-derived animal proteins, thus potentially ex- Alternatively, lineage-uncommitted cells such as bone marrow-
panding the range of cultured meat production methods. derived mesenchymal stem cells (BM-MSCs), adipose-derived stem
Beyond post-culture processing of cultured meat, some food- cells (ADSCs) or induced pluripotent stem cells (iPSCs) may be
grade biopolymers may provide potential sources of scaffold mate- considered. With the exception of immortal iPSCs, the other cell
rials for muscle and fat culture that are intrinsically cost-effective, sources exhibit limited and variable proliferation capacities, and
scalable, and sustainable, by virtue of having met these constraints would require either regular isolation from animal biopsies or im-
within the context of the meat industry (see Section 4.4). mortalization.
Bioengineers have elucidated various design criteria that bio-
Box 1: Alginate in animal-based meat products material scaffolds should fulfil for engineering tissue for biomed-
ical applications [102–107]. Nevertheless, both cell expansion and
differentiation stages face significant technological hurdles to in-
Alginate is a seaweed-derived hydrocolloid that forms a phys-
ically crosslinked gel in the presence of bivalent cations such dustrial translation. We first contextualize and evaluate the feasi-
as calcium. As it forms hydrogels at low but non-freezing bility of current biomaterials strategies for promoting efficient ex-
temperatures, alginate is particularly useful in the produc- pansion and differentiation of muscle and adipose precursor cells
tion of restructured raw meat products, such as meatloaves, within the setting of large-scale bioreactors, and identify possi-
meatballs, or seafood-based surimi, where a larger piece of ble avenues for improvement (Table 2A). In addition, we consider
raw meat is formed from several smaller pieces. The first de- novel constraints specific to cultured meat production, such as the
role for material degradability at different stages of cultured meat

6
JID: ACTBIO
ARTICLE IN PRESS [m5G;February 21, 2021;17:50]

S. Ng and M. Kurisawa Acta Biomaterialia xxx (xxxx) xxx

Table 2
Scaffold design principles for cultured meat production.

(A) Conventional design criteria for engineering muscle and adipose tissue

Cell expansion:
 Surface chemistry
 Cell adhesion
 Scaffold stiffness

Additional criteria for cell differentiation:


 Cell alignment
 Mechanical stimuli (passive & dynamic)
 Electrical stimuli

(B) Additional design criteria for adapting classical tissue engineering approaches for cultured meat production

Scaffold fabrication:
 Compatibility with bioreactors for cell expansion & differentiation
 Scalability of fabrication method for cultured meat production

Scaffold crosslinking: chemical (elasticity) versus physical (viscoelasticity)


 Mechanical stability during bioreactor culture
 Role of elasticity or viscoelasticity on cell expansion & differentiation
 Post-culture product stability & organoleptic properties (texture)

Scaffold degradability:
 Ease of harvesting expanded cells
 Responsiveness to cellular modelling during differentiation
 Organoleptic, stabilizing and nutritional functions post-differentiation

Scaffold edibility:
 Optional for cell expansion, necessary for cell differentiation
 Compatibility with food-safe cultured meat production

Scaffold sustainability:
 Animal-free, natural or recombinant sources

Cultured meat cost:


 Growth factor release, sequestration & mimicry to mitigate culture medium cost
Adoption of food biopolymers to mitigate scaffold costs

Product fulfils meat quality attributes: appearance, flavour, texture, nutrition


 Meat quality evaluation with conventional methods like textural profile analysis, human sensory testing

production, the potential for biomaterials to mitigate high growth aerated (bubble column, airlift) bioreactors that could support up
media costs, the need for edible, cost-effective, sustainable scaf- to 1 × 106 L (Fig. 2B) [110].
fold materials and their potential sources (Table 2B), and conclude Alternatively, hollow fibre bioreactors (HFBs) (Fig. 2C) have ex-
by exploring how bioengineers may repurpose common biopoly- hibited the potential for expanding bone marrow-derived MSCs
mer meat ingredients as novel multifunctional biomaterial scaf- [113–115], embryonic stem cells [116], myoblasts [117,118] and
folds that support engineered muscle and adipose tissue produc- adipose-derived stem cells [114,119,120]. Such systems provide ma-
tion and contribute to desirable meat quality attributes. jor advantages of higher cell densities (108 – 109 cells/mL) and vol-
ume efficiencies [108,111,121], which could provide a space-saving
conduit in the earlier part of a seed train process [109]. However,
4.1. Conventional biomaterial design strategies for cell expansion
high microfibre and cell densities may pose significant limitations
to cell harvesting efficiencies in the intermediate rounds of cell ex-
4.1.1. Bioreactor considerations
pansion [122], which suggests greater practicality of HFBs in the
Cell expansion currently appears to be less challenging to scale
cell differentiation stage of cultured meat production, where the
up than differentiation, given earlier efforts by the cell therapy in-
scaffold is designed to be edible without requiring separation from
dustry to expand adherent MSCs for allogeneic use on an indus-
differentiated cells.
trial scale using microcarriers in stirred tank bioreactors (STBs)
[102,108]. The possibility of bead-to-bead transfer as a mode of
cell passaging offers the attractive prospect of a semi-continuous 4.1.2. Scaffold design principles
or even continuous seed train process that minimizes cell han- Surface chemistry, particularly charge and hydrophilicity, pri-
dling and processing time, and therefore maximizes productivity marily promote initial cell attachment on microcarriers under dy-
of the cell expansion stage (Fig. 2A) [109]. Nevertheless, the re- namic seeding conditions [123]. Positively charged surfaces gen-
quired scale of cell expansion required for cultured meat produc- erally increase cell attachment efficiency due to the slightly neg-
tion is orders of magnitude larger than that for biomedical ap- atively charged cell surface [124–127], whereas moderately hy-
plications [1,110,111], and the relatively low working cell densities drophilic surfaces increase cell attachment indirectly by promot-
(105 – 106 cells/mL) and modest working volumes (50 L) of cur- ing surface protein adsorption, which mediates cell attachment
rently used bioreactors may pose significant scalability limitations [127–130]. However, microcarrier cell attachment is often a com-
[108]. In contrast to an estimated batch size of 5 × 1010 cells pro- plex, empirical function of inter-dependant factors including net
duced by this method, 1 kg of muscle cells contains approximately surface charge, charged group density, hydrophilicity, surface to-
3 × 1011 cells [111], and average daily global meat consumption is pography, curvature and shear rates [123]. A salient demonstration
approximately 8 × 108 kg [112]. Microcarriers may be compatible of this complexity is the observation that bovine myoblasts exhib-
with other bioreactor types that support higher cell densities, such ited similar cell attachment rates to net-positively charged Cytodex
as packed or fluidized bed bioreactors, or larger volumes, such as 1 microcarriers and net-negatively charged Synthemax microcar-

7
JID: ACTBIO
ARTICLE IN PRESS [m5G;February 21, 2021;17:50]

S. Ng and M. Kurisawa Acta Biomaterialia xxx (xxxx) xxx

Table 3
Common food biopolymers for tissue engineering.

Biopolymer Biocompatible scaffold fabrication strategies1 Applications2 Key review

Carrageenan Sol-gel transition + ionic gelation: composite hydrogel with alginate [307], hydrogel [314–316] Cartilage Yegappan et al.
Ionic gelation + polyelectrolyte complexation: composite microfibres with chitosan [313] [314,315] 2018 [311]
Acrylate-acrylate photopolymerization: methacrylated carrageenan hydrogels [308,310]
Sol-gel transition + freeze-drying: porous hydrogel [317]
Polyelectrolyte complexation + freeze-drying: composite porous hydrogel with chitosan [309]
Konjac Alkaline thermal gelation + freeze drying: porous hydrogel [355,357] Bone [357] Yang et al.
Electrospinning + Schiff-base formation: nanofibres [358] 2017 [384]
Water-in-oil emulsion: DEAE-modified microgel carriers [359]
Semi-interpenetrating network: composite hydrogel with polyphenol crosslinked fibrillar collagen
[356,360]
Pectin Ionic gelation: microgel carriers [318], interpenetrating networks with photopolymerized methacrylated Muscle [137] Noreen et al.
gelatine [321] Bone [318,322] 2017 [385]
Ionic gelation + freeze-drying: porous hydrogel [319] Skin [320]
Chemical gelation with (3-glycidyloxypropyl)trimethoxysilane (GPTMS) + freeze-drying: porous hydrogel Cartilage [325]
[323]
Electrospinning + Schiff-base formation: nanofibres [324]
Thiol-ene photopolymerization: norbornene-modified pectin hydrogel [320]
Disulphide bond formation: thiolated pectin hydrogel [137]
Acrylate-acrylate photopolymerization: methacrylated pectin hydrogels [322]
Schiff-base formation: composite hydrogel of aldehyde-modified HA and hydrazide-modified pectin
[325]
Polyelectrolyte complexation: composite hydrogel membranes with chitosan [326]
Gellan gum Sol-gel transition + ionic gelation: hydrogel [341,342,345], composite hydrogel with xanthan [346] Muscle [342] Stevens et al.
Sol-gel transition + ionic gelation + freeze-drying: porous hydrogel [347] Cartilage 2016 [339]
Acrylate-acrylate photopolymerization: methacrylated gellan gum microgels [340,348] [340,341,345]
Schiff-base formation: composite hydrogel of aldehyde-modified gellan gum and hydrazide-modified Neural [343]
gelatine [344] Cardiac [344]
Water-in-oil emulsion: microgel carriers [304] Bone [346]

Xanthan gum Ionic gelation: microcapsules of carboxymethylated xanthan [349] Bone [346] Kumar et al.
Sol-gel transition + ionic gelation: composite hydrogel with gellan [346] 2018 [351]
Borate-diol complexation: weak hydrogel or composite hydrogel with cellulose [350]
Hydrophobic-driven self-assembly: microcapsules of palmitoyl-modified xanthan [352] or
phospholipid-modified xanthan [353]
Thermal casting + chemical gelation with citric acid: hydrogel film [354]
Guar gum Acrylate-acrylate photopolymerization: methacrylated guar gum hydrogels [335] - George et al.
Borate-diol complexation + gas-foaming + freeze-drying: Porous composite hydrogel with polyvinyl 2019 [386]
alcohol [337]
Schiff-base formation: composite hydrogel of aldehyde-modified guar gum and collagen [336]
Chemical gelation with sodium trimetaphosphate + freeze-drying: porous hydrogel [338]
Cellulose Bacterial nanofibre self-assembly + freeze-drying: composite porous hydrogel with agarose/gelatine Muscle [178] Hickey et al.
[158,330,331] Skin [334] 2019 [159]
Decellularization: prevascularized plant scaffold [329,333] Cardiac [329]
Spin-coating of cellulose nanorods: nanofilms [178] Bone [330,331]
Electrospinning: nanofibres [328]
Rotary jet spinning: composite nanofibres with soy [334]
Chemical gelation + freeze-drying: ether-crosslinked composite porous hydrogels with soy isolate [332]
Chemical gelation + solvent casting: epoxide-crosslinked composite hydrogel films with soy [327]
Starch Melt spinning/fibre bonding: porous composite microfibre mesh with polycaprolactone (PCL) [374,375] Bone Hemamalini
Melt spinning/fibre bonding + electrospinning: composite microfibre mesh with PCL filled with [163,373,374,376] et al. 2018
nanofibres [373] Cartilage [377] [387]
Wet spinning: composite microfibres with polycaprolactone [163] Vascular [375]
Electrospinning: composite nanofibres with PCL [377] or with PEG [378]
Electrospinning + Schiff-base formation: nanofibres [358]
Thiol-ene + acrylate-acrylate photopolymerization: interpenetrating network of pentanoate-modified
starch with methacrylated gelatine [376]
Soy isolate Thermal gelation + freeze-drying: porous hydrogel [370] Muscle [58] Tansaz et al.
Chemical gelation + freeze-drying: porous hydrogel that is transglutaminase-crosslinked [367], Skin [334] 2016 [388]
glutaraldehyde-crosslinked [370], carbodiimide-crosslinked composites with gelatine/alginate/pectin Adipose [365]
[369] or ether-crosslinked composites with cellulose [332]
Electrospinning: nanofibres [365]
Rotary jet spinning: composite nanofibres with cellulose [334]
Wet spinning: microfibres [366]
Extrusion: texturized scaffold [58]
Chemical gelation + solvent casting: epoxide-crosslinked composite hydrogel films with cellulose [327]
Zein (corn) Solvent casting: nanoparticulate films [134], smooth hydrated films [389] Bone Paliwal et al.
Salt leaching + freeze-drying: Porous scaffold [362,364,383] [362,364,383] 2014 [390]
Electrospinning: nanofibres [361]
Thermal gelation + compression moudling; smooth composite film with pea [372]
1
Biocompatibility is defined as a quantitative or qualitative demonstration of cell viability by a conventional biochemical or microscopic method (e.g. redox assay or
calcein AM/propidium iodide labelling) over at least two time points (e.g. 1 and 3 days) after the seeding of any cell type in the fabricated biopolymer scaffold in either
two- or three-dimensions. As such, studies that perform indirect cytotoxicity tests involving the exposure of cells to culture medium supernatants from prior incubation
with the biopolymer scaffold are excluded.
2
A minimal tissue engineering application of a biopolymer scaffold involves the quantitative or qualitative demonstration of at least one relevant differentiated pheno-
type (e.g. biochemical assays, immunohistochemical labelling, or expression of genes or proteins associated with the differentiated phenotype) that typically characterizes
functional cells from the tissue of interest.

8
JID: ACTBIO
ARTICLE IN PRESS [m5G;February 21, 2021;17:50]

S. Ng and M. Kurisawa Acta Biomaterialia xxx (xxxx) xxx

Fig. 2. Bioreactors for cultured meat production. (A) A generic seed train process for expansion of precursor muscle or adipose cells. (B, C) Types of suspension and
perfusion bioreactors that are potentially suitable for scalable cell expansion and cell differentiation.

riers [131], as the enhanced protein adsorption conferred by the proliferated faster on hydrogels with an elastic modulus (E) of
hydrophilicity of the negatively charged Synthemax microcarriers 12 kPa, which mimic that of natural muscle, than on much stiffer
outweighed the adverse effect of its net negative charge on cell at- tissue-culture polystyrene [144]. Similarly, primary or immortal-
tachment [132]. ized murine myoblasts exhibited proliferation rates that increased
Cell adhesion moieties presented at optimal densities signal with substrate stiffness and plateaued above E = 12 kPa [104,145],
cell survival and proliferation after initial attachment, and are whereas increased muscle stiffness in aged muscle impaired en-
typically introduced by physically coating or covalently modify- dogenous myoblast proliferation [146]. In contrast to the well doc-
ing the microcarrier surface or biomaterial scaffold with ECM pro- umented increase in proliferation of BM-MSCs on stiffer substrates
teins (e.g. collagen, fibronectin) [126], or cell adhesion peptides [147–149], the relationship between substrate stiffness and ADSC
(e.g. fibronectin-derived RGD, laminin-derived YIGSR) [133]. If re- proliferation is currently less firmly established [105,150,151].
combinant proteins or synthesized peptides prove too costly for
cultured meat production, empirical screening of natural products
could reveal cheaper alternative sources of cell-adhesive proteins 4.2. Conventional biomaterial design strategies for cell
[134–136]. Whole proteins presenting cell adhesion domains may differentiation
also be incorporated into composite scaffolds [137].
Scaffold stiffness influences cell adhesion and proliferation in 4.2.1. Bioreactor considerations
multiple cell types [138–141], and these cell type-specific re- Myogenic differentiation of myoblasts/satellite cells is presum-
sponses may correlate with the rigidity of the in vivo cellular envi- ably more challenging to scale up than adipogenic differentiation
ronment [142,143]. For example, primary murine myosatellite cells of preadipocytes/ADSCs, because of a greater need to accommodate
intra-scaffold cellular self-assembly and provide dynamic differen-

9
JID: ACTBIO
ARTICLE IN PRESS [m5G;February 21, 2021;17:50]

S. Ng and M. Kurisawa Acta Biomaterialia xxx (xxxx) xxx

tiation stimuli in the former, whereas the latter may only require a alginate, starch, recombinant gelatine or soy protein [93,94,160–
switch in culture medium and static scaffold properties. The type 163], in conjunction with shear patterning to introduce micro-
of cultured meat product is an important consideration when de- striations that further guide cell alignment [164,165], and biofunc-
signing biomaterial scaffolds for the cell differentiation stage, as tionalisation strategies for cell adhesiveness. Given that the perfu-
the required degree of native muscle tissue mimicry depends on sion mode in HFBs can be intracapillary (intra-fibre) or extracap-
the structural complexity of the final product [6]. illary (between fibres) [121], direct fabrication of solid or tubu-
For unstructured meat applications that do not preserve tissue- lar cell-encapsulated microgel fibres via a single wet spinning step
level structure [6], replicating muscle tissue structure is unneces- prior to HFB incorporation is conceivable [166–170]. Such microfi-
sary. Cell differentiation could occur on edible microscale scaffolds bre cell encapsulation approaches could eliminate potential chal-
(μm – mm scale), which remain compatible with any suspension lenges in uniformly filling pre-fabricated hollow fibres with a cell-
bioreactor type used for cell expansion (Fig. 2B). However, this ap- encapsulated hydrogel, and offer a facile way to represent both
proach requires post-culture assembly of the microscale cultured muscle and adipose cells in the same HFB construct by combining
meat units into a macroscale product, perhaps leveraging conven- microfibres encapsulating different precursor cell types. The extra-
tional meat structuring processes (see Section 3) or newer additive capillary space could be filled with an edible hydrogel to obtain a
manufacturing technologies [84] and conventional food biopoly- monolithic construct suitable for structured meat applications, and
mers to substitute for meat texture and protein content [52,53]. could also encapsulate cells.
For structured meat applications, differentiation should ideally
occur in edible scaffolds with final macroscale dimensions (cm –
dm scale) that match the desired product (e.g. steaks or slices) to 4.2.2. Scaffold design principles
eliminate the need for secondary assembly of microscale cultured Myoblast alignment is a crucial step that precedes myotube for-
meat units. Both fixed bed bioreactors and HFBs can conceivably mation [101]. The provision of cellular alignment cues could occur
generate monolithic cultured tissue constructs with the requisite via the use of aligned fibrous scaffolds [57,169,171–175], microto-
macroscale dimensions and are scalable via bioreactor paralleliza- pographically patterned surfaces [164,165,173,176–181], anisotropic
tion (Fig. 2C) [1]. However, this necessitates the development of or directional pore formation [182,183], biochemically or mechan-
non-trivial methods for scaffold incorporation into the bioreactor, ically patterned materials [142,184], naturally aligned, decellular-
cell seeding, and harvesting of cultured tissues while minimizing ized plant-derived scaffolds [185] or dynamic mechanical stimula-
handling and retaining product integrity and sterility [1,152]. tion [186–190]. However, not all cell alignment strategies are in-
Fixed bed bioreactors are potentially feasible for scaffolds with dustrially scalable, and those that are implementable in macrop-
sufficiently large pores that balance the need to support high cell orous or fibrous formats for fixed bed bioreactors or HFBs, respec-
densities, which requires sufficiently high perfusion rates for ef- tively, are likely to be advantageous.
ficient intra-scaffold fluid transport, and maintain cell viability, The static mechanical environment, in particular substrate
which requires minimizing fluid shear stresses [1,110,153]. This stiffness, influences both myogenic and adipogenic differentia-
may limit the application of nanoporous hydrogels previously de- tion efficiency [104,142,145,191–197]. Mimicking the stiffness of
veloped for myogenesis or adipogenesis [154] due to the high fat tissue (E ≈ 1 – 3 kPa) [76,198,199] consistently enhances ADSC
shear stresses needed to ensure adequate perfusion, which could adipogenesis in vitro [194–197,200], highlighting the relevance
limit cell viability [110,153]. Current fixed bed bioreactors accom- of modulating scaffold stiffness for optimizing adipogenesis of
modate porous scaffolds with sizes ranging from millimetre to bovine adipose precursors. In contrast, myotube formation oc-
decimetre dimensions, but generally utilize inedible or animal- curs over a broad range of substrate rigidity [104,142,145,193],
derived scaffold materials such as glass, collagen and synthetic and late-stage myotube maturation in the form of actomyosin
polymers (e.g. polyesters and polystyrene) [152,155,156]. While striations exhibits a context-dependant relationship with stiff-
millimetre-sized scaffolds are limited to unstructured meat appli- ness that may involve other biochemical, topographical and cel-
cations, centimetre to decimetre-sized scaffolds may be suitable for lular variables [104,142,193]. For example, when cultured on
structured meat applications. The commercial availability of single- polyacrylamide gels presenting anisotropic collagen micropat-
use fixed bed bioreactors that are pre-loaded with centimetre- terns, C2C12 myoblasts exhibited maximal striation at moderate
sized polyester carriers, such as iCELLis R
[155], supports the fea- gel stiffnesses matching native mouse muscle (E ≈ 12 kPa) [142].
sibility of developing similar bioreactors that accommodate edible, In contrast, culturing C2C12 myoblasts on unpatterned poly-l-
porous macro-scaffolds with centimetre to decimetre dimensions lysine/hyaluronic acid polyelectrolyte films or primary myoblasts
for structured meat applications. Well-defined co-cultures of mul- on entactin/collagen/laminin-coated polyacrylamide gels monoton-
tiple cell types offer a potential strategy to represent both mus- ically increased myotube striation with increasing stiffness, with
cle and adipose cells in future structured meat products produced maximal striation on the stiffest control substrates tested [104,193].
by fixed bed bioreactors, as exemplified by the self-organization of Therefore, the feasibility of optimizing bovine myogenic differenti-
tri-cultures comprising myoblasts, fibroblasts and endothelial cells ation via scaffold stiffness modulation is currently unclear and re-
into engineered vascularized muscle [157], and the recent gener- quires further investigation. Importantly, increasing scaffold stiff-
ation of engineered bovine muscle for cultured meat applications ness often requires increasing material concentration or crosslink-
using a similar approach [58]. ing densities, which concomitantly decreases pore size, and may
HFBs may overcome the limited perfusion rates of fixed bed limit maximal perfusion rates and therefore engineered tissue
bioreactors by separating the media and cellular compartments, thickness in the context of fixed bed bioreactor-based production.
thereby protecting cells from shear stress, and the fibres may con- Encapsulating myoblasts in a hydrogel between two anchoring
veniently provide topographical cues for myoblast alignment [121]. posts and the subsequent passive development of static uniaxial
Existing HFBs are often designed for the production of secreted tension is a well-established method to generate relatively mature
proteins and viruses, and the entrapped cells are not the de- myofibers of macroscopic dimensions in vitro [101,201–204], and
sired end-product [121]. As such, current hollow fibres generally its limited industrial scalability may be circumvented by utilizing
comprise inedible synthetic polymers (e.g. polysulfones, polyethy- appropriate scaffold rigidity as a source of cellular tension [205].
lene, polyesters), with the exception of potentially edible cellulose Increased protein production due to static tension [204] increases
derivatives [158,159]. Wet spinning of hollow fibres, a common levels of texture-contributing actin/myosin proteins, as well as
fabrication method, could be adapted to edible materials such as flavour- and colour-contributing myoglobin protein in meat [206].

10
JID: ACTBIO
ARTICLE IN PRESS [m5G;February 21, 2021;17:50]

S. Ng and M. Kurisawa Acta Biomaterialia xxx (xxxx) xxx

In addition to static tension, dynamic mechanical stimuli [110,121,154]. Cells could conceivably be encapsulated either dur-
further increases myogenic maturation [187–190,207–209], but ing scaffold fabrication or after fabrication, given that biocompati-
scaling dynamic mechanical stimuli industrially is non-trivial ble crosslinking and fabrication strategies exist for both porous and
and would require substantial bioreactor development [210,211]. fibrous morphologies [238,239]. The choice of physical or cova-
Emerging advanced methods that remotely actuate intra-scaffold lent crosslinking approaches should be informed by whether clas-
mechanical stimuli could offer potential pathways towards large- sical scaffold design criteria for cell differentiation are fulfilled,
scale mechanical stimulation of myogenesis, and include the use and may require further studies on how their resultant viscoelas-
of moderate fluid shear stress [212,213], light-responsive nanopar- tic properties influence cell differentiation and post-culture proper-
ticle actuators of cell adhesion receptors [214], or magnetically ac- ties. For example, osteogenic differentiation of MSCs and chondro-
tuated biphasic ferrogels that undergo large, fatigue-resistant de- cyte production of cartilage matrix were both accelerated in vis-
formations [215]. coelastic compared with elastic alginate hydrogels [240,241], but
Exogenous electrical stimuli also promote various facets of my- the contribution of viscoelasticity to myogenesis and adipogene-
otube maturation, including cross-striation, MHC isoform expres- sis is currently unknown. The texture of cultured meat and its
sion, myofibrillar protein level, and contractility [216–222], but stability during storage and cooking are two post-culture prop-
also require substantial bioreactor development to ensure indus- erties that could be affected in opposite ways by scaffold vis-
trial scalability [210,211,223]. A unique approach to introduce scal- coelasticity. To illustrate this, gelling agents used in meat prod-
able electrical stimuli is by using electrically conductive bioma- ucts that undergo cooling-induced sol-gel transitions may lose
terials such as polyaniline, polypyrrole and polythiophenes [224], structural integrity and exhibit substantial water loss during cook-
which increase myogenesis when incorporated into scaffolds, even ing, which could be overcome by transglutaminase-mediated cova-
without exogenous electrical stimulation [225–228]. Such conduc- lent crosslinking between meat proteins. However, the viscoelas-
tive materials may synergize with other differentiation cues, such tic properties of animal-based meat [242,243] suggests that there
as cell alignment, to further promote myogenesis [229,230], but may be a threshold degree of covalent crosslinking in meat prod-
their suitability for human consumption is uncertain. ucts beyond which the increased elasticity may noticeably affect
meat texture, as exemplified by increased hardness and chewiness
4.3. Novel constraints on biomaterial design for cultured meat exhibited by transglutaminase-crosslinked restructured meat prod-
production ucts [244–246]. Given the potentially complex effects of scaffold
viscoelasticity on cultured meat properties, holistic characteriza-
4.3.1. Scaffold crosslinking and fabrication tion of its effects will be required.
Biomaterial scaffolds may be fabricated in various morpholo-
gies using physical or chemical crosslinking approaches. Natural
biopolymers often exhibit intrinsic gelation via physical crosslink- 4.3.2. Scaffold degradability
ing, most commonly thermal (collagen, gelatine, agar) or ionic Expanded cells would likely require multiple rounds of passag-
(alginate), and may naturally undergo enzyme-mediated covalent ing before final re-seeding in a different type of scaffold and biore-
crosslinking (thrombin/fibrin) [231–233]. In order to improve their actor for cell differentiation [109]. In this scenario, expanded cells
gelation kinetics and mechanical properties, biopolymers are of- have to be efficiently harvested from the scaffold, which could be
ten conjugated with reactive side-groups that enable chemical non-degradable or degradable.
crosslinking, such as acrylates, thiols or phenols [231–233], some Harvesting cells from non-degradable scaffolds would re-
of which undergo biocompatible crosslinking reactions that sup- quire efficient cellular dissociation and post-dissociation separa-
port viable cell encapsulation during scaffold fabrication. These tion strategies. Dissociation methods could include recombinant
include small molecule-based reactions such as azide-alkyne cy- enzymes [247,248], chemical mimetics of enzymes [123], mechan-
cloaddition, Michael-type addition, Schiff-base formation, photo- ical agitation [249–251], and thermo-responsive polymer coatings
initiated free radical reactions, and enzymatic crosslinking reac- based on poly(N-isopropylacrylamide) (pNIPAAm), methylcellulose,
tions such as phenol-phenol (peroxidase), phenol-amine (tyrosi- hydroxybutyl chitosan and xyloglucan [252–256]. Separation could
nase) and amine-amine (lysyl oxidase) reactions [231,232]. occur via filtration, centrifugal, inertial, or magnetic methods [257–
For cultured meat production, precursor cells will likely be 260], as long as the scaffold exhibits sufficiently different size, den-
seeded on pre-fabricated microcarriers for expansion and poten- sity or magnetism compared with the harvested cells. However,
tially for differentiation (for unstructured meat). In principle, ei- the accumulated inefficiency of both dissociation and separation
ther physical or chemical crosslinking approaches may be appli- steps, together with a desire for high cell purity to minimize the
cable, as long as the microcarriers fulfil critical design elements re-seeding of the non-degradable material in the subsequent differ-
that support cell expansion or differentiation. Certain physically entiation bioreactor, could result in significant cell loss [123,257].
crosslinked biomaterials that intrinsically lack mechanical stabil- The use of degradable scaffolds could simplify cell harvesting by
ity under dynamic culture conditions (e.g. collagen, fibrin) may eliminating the need for dissociation and separation, and such ma-
be unsuitable without secondary chemical crosslinking. Increasing terials may degrade hydrolytically, chemically, enzymatically, pho-
awareness that physically crosslinked hydrogels exhibit viscoelastic tolytically, or in response to pH or temperature changes [123].
properties (e.g. stress relaxation), whereas chemically crosslinked However, most biomaterials that undergo pH-dependant, thermal
hydrogels are purely elastic [234], coupled with knowledge of or photo-degradation are unlikely to be cell-compatible, whereas
the viscoelastic nature of native muscle and fat [234–236], has those that undergo hydrolysis, such as poly(lactic-co-glycolic acid)
helped to elucidate the independent effects of scaffold viscoelas- (PLGA) and polycaprolactone (PCL), may release cells prematurely.
ticity and elasticity (stiffness) on cell behaviour. For example, my- Therefore, only chemical, enzymatic and pH-dependant degrada-
oblasts exhibit increased cell spreading and proliferation on vis- tion remain viable options that are potentially fast, robust and
coelastic compared to elastic hydrogels of the same mechanical stimulus-specific. Chemically degradable biomaterials include ion-
stiffness [236,237], which suggests a potential role of scaffold vis- ically crosslinked polygalacturonic acid (PGA) and alginate, which
coelasticity in cell expansion optimization. degrade in response to sequestrants of bivalent cations such as
As discussed earlier, cell differentiation would likely require EDTA, citrate or phosphate. Enzymatically degradable biomateri-
the use of porous scaffolds or microfibres that are loaded post- als may include various polysaccharides, such as dextran, alginate,
fabrication into fixed bed or hollow fibre bioreactors respectively pectin and cellulose, and any non-animal-derived protein for which

11
JID: ACTBIO
ARTICLE IN PRESS [m5G;February 21, 2021;17:50]

S. Ng and M. Kurisawa Acta Biomaterialia xxx (xxxx) xxx

Fig. 3. Integrating biomaterials and food biopolymers for cultured meat production. This schematic provides a side-by-side comparison of conventional and cultured
meat production, and highlights the many steps other than the initial engineering of cultured muscle or adipose tissue in vitro that are involved in determining meat quality
attributes. Conventional food biopolymers may contribute to future cultured meat production by providing edible, sustainable and cost-efficient tissue engineering scaffold
materials, and supporting post-culture assembly and processing of cultured meat products.

recombinant enzymes that function under cell-compatible condi- Possible strategies to mitigate this significant cost include: (1)
tions are available [123]. reducing growth factor requirements by cell line adaptation, en-
The requirement for degradability of the differentiation scaffold gineering growth factors with improved stability or binding affin-
depends on a balance between the need to accommodate cellu- ity, or substituting growth factors with small molecule mimetics
lar remodelling during differentiation and the potential organolep- or plant/microbial hydrolysates, (2) reducing growth factor cost
tic or nutritional roles played by the scaffold in the final meat by producing them at industrial scale (e.g. for industrial enzymes
product. Differentiation scaffolds that comprise biopolymer-based like lipase and cellulase), (3) reducing basal medium cost by using
meat ingredients could conceivably fulfil different functions dur- food-grade rather than pharmaceutical-grade ingredients, or using
ing culture and post-culture, which could make degradability op- cheaper antioxidants or buffer systems [109,263]. However, these
tional. For example, wet-spun fibrous scaffolds comprising edible approaches do not address the fundamental inefficiency that arises
hydrocolloids may be designed to provide alignment and mechani- from the provision of soluble growth factors at supraphysiological
cal cues that support myogenesis during culture, and may also con- bulk concentrations in order to ensure sufficient concentrations at
tribute post-culture to structural and storage stability, and mod- the cellular level [264].
ulation of texture and flavour, in the final cultured meat prod- Engineered biomaterials may increase the efficiency of exoge-
uct via their gelation and water retention behaviour (see Table 1, nous growth factor utilization by localizing their sustained release
Sections 3.1, 3.2). From a nutritional perspective, scaffolds con- from the scaffold [264,265], or by controlling their spatial pre-
taining indigestible polysaccharides such as cellulose may supple- sentation with matrix-bound forms [264], which may exhibit pro-
ment dietary fibre, which is often underrepresented in modern di- longed biological activity [266] or enable multivalent interactions
ets [261], whereas those containing residual protein content post- that strengthen signal transduction [267]. Alternatively, biomate-
culture could supplement dietary protein. rials could mimic the non-covalent sequestration of endogenously
Scaffolds could even be molecularly designed to degrade into secreted growth factors by ECM via the incorporation of peptides
molecules with positive organoleptic or nutritional properties that mimic the binding domain of growth factor receptors or ap-
[154], such as flavour-generating aromatic compounds or essential tamers that specifically bind to growth factors [264,268], thereby
vitamins [65]. One possibility is the degradation-dependant release amplifying myogenic or adipogenic autocrine signals [269–271].
of specific fatty acids (e.g. omega-3) that act as maturation factors Biomaterials could even directly incorporate growth factor mimet-
during adipogenesis [262] and contribute flavour, mouthfeel and ics based on peptides, aptamers or small molecules as part of the
nutritional benefits post-culture. scaffold backbone [264]. While sustained local delivery approaches
could be conceivably adapted to various food-grade biopolymers
like lipids and hydrocolloids in various micro- or nano-scale forms
4.3.3. Culture medium cost mitigation with existing encapsulation technologies [272,273], engineered ma-
Culture medium is predicted to account for 55 – 95% of the terials that present new chemical entities such as molecular se-
marginal cost of industrial cultured meat production [109,263] due questrants or growth factor mimetics would require more exten-
to the need for chemically defined, animal-free medium, which sive food safety evaluation.
requires the replacement of bovine sera with relatively expen-
sive recombinant growth factors. A recent analysis estimated the 4.3.4. Scaffold edibility, sustainability and cost
marginal cost of culture medium alone for cultured meat produc- In principle, scaffolds for cell expansion could be either inedible
tion to range between $2100 – $15,0 0 0/kg cells produced at cur- or edible. In the likely scenario where expanded cells are harvested
rent medium component prices, in contrast to wholesale prices of for re-seeding in a differentiation scaffold, the use of an edible
conventional animal-based meat ($3.3 – $4.4/kg) [263]. Through scaffold could increase the tolerance level for remaining quantities
reductions in the cost of both growth factors and basal medium of the scaffold in the harvested cells, thus allowing for increased
via incremental methods that do not rely on fundamental new harvesting yields [123]. However, if a single scaffold is designed
technologies or innovations, the same analysis predicted that it is to support both cell expansion and subsequent cell differentiation
likely that cultivated meat can achieve price parity with conven- into the final product, scaffold edibility may become indispensable
tional meat once produced at industrial scale [263]. [123]. The scaffold used for cell differentiation must be edible as

12
JID: ACTBIO
ARTICLE IN PRESS [m5G;February 21, 2021;17:50]

S. Ng and M. Kurisawa Acta Biomaterialia xxx (xxxx) xxx

the final engineered muscle and fat tissues are likely to possess
intrinsically complex tissue-level structures that may be practically However, the marginal cost of microcarriers is indepen-
inseparable from the remaining portion of their original scaffold. dent of bioreactor type and size as long as microcarrier con-
Amongst current biomaterial scaffolds that support myo- centrations remain within reasonable limits compatible with
genic and adipogenic differentiation, including collagen or gela- the maintenance of cell viability, in contrast to the marginal
tine [202,203,274,275], hyaluronic acid (HA) [276–280], fibrin cost of culture medium, which varies with the maximum cell
density achievable in culture and is therefore dependant on
[281–283], alginate [278,284–287], chitosan [288], PEG [289,290],
bioreactor type.
PLGA [205,278,291,292], PCL [289,293] and decellularized tissues In the current analysis, we compare microporous Cytodex
[107,185,294–296], only alginate, gelatine and PEG are routinely 1/3 and macroporous Cytopore 2 microcarriers, and assumes
used in food applications. The use of decellularized tissues will a confluent cell density of 1 × 105 cells/cm2 [302] and a
probably be limited to plant or fungal sources and may face chal- generic cell mass of 3.4 × 10−12 kg/cell [109,111]. Microcarrier
lenges in both scalability and cost-efficiency. Although synthetic cost estimates are from the current retail prices of the largest
polymers like PEG, PCL and PLGA are approved for clinical appli- unit size of each microcarrier type at Cytiva Life Sciences
cations, such as drug delivery, sutures, and dermal fillers, and are (November 2020).
relatively cheaply produced, it is currently unclear if the larger
Cytodex 1 Cytodex 3 Cytopore 2
amounts of such polymers that may be regularly consumed as
part of cultured meat products will be considered food-safe. Al- Microcarrier cost ($/g 4.88 5.24 31.5
microcarrier)
though naturally derived biomaterials like HA, fibrin and chitosan
Microcarrier growth area 4400 2700 11,000
are likely also safe for consumption, they are conventionally de- (cm2 /g microcarrier)
rived from animal sources, which limits their use in cultured meat [302]
production to recombinant versions from transgenic plant, fun- Maximum cell productivity 4.4 × 108 2.7 × 108 1.1 × 109
(cells/g microcarrier)
gal or microbial sources [297–301]. Such recombinant sources are
Maximum cell mass 1.50 0.92 3.74
likely to remain prohibitively costly for cultured meat production productivity (kg cells/kg
until true economies of scale are realized with future increases in microcarrier)
demand. For example, microbial-derived HA is currently available Marginal microcarrier cost 3262 5708 8422
at a food-grade scale at $130/kg, in contrast to food-grade alginate, ($/kg cells produced)

which has a wholesale price of $10/kg. Assuming that every ex-


panded precursor cell is arbitrarily seeded at twice its own volume B) Scaffolds for differentiation:
in a 10 mg/mL (1% w/v) differentiation scaffold within a perfusion As scaffolds for differentiation may come in various forms
(macroporous hydrogels, microcarriers, fibres), this analysis
bioreactor, the marginal cost of raw materials for an alginate- or
assumes the use of a volume-averaged scaffold material con-
HA-based differentiation scaffold would be $0.23/kg or $2.95/kg of
centration of 10 mg/mL (1% w/v). We also arbitrarily assume
cells produced respectively (Box 2). Given that many food biopoly- that expanded precursor cells are re-seeded in a perfusion
mers used in conventional meat production (Table 1) are available bioreactor at twice the total volume of cells, to cater for the
at industrial scale and at prices that are competitive with alginate void space needed for the scaffold itself and its perfusion
(Box 2), the application of these biopolymers as potential differ- with culture medium [263]. Given an average meat density
entiation scaffold materials (see Section 4.4) could conceivably en- of 881 kg/m3 [263], which results in a volume of 1135 mL/kg
sure that the marginal cost of such scaffolds remain within a frac- of cells, the total mass of scaffold material required per kilo-
tion of the wholesale price of conventional meat. gram of cells seeded is:
In contrast to scaffolds for myogenic and adipogenic differentia- mL kg scaffold
tion, microcarriers for adherent cell expansion are widely available 1, 135 x 2 x 10−5
kg cells mL
commercially, and comprise a variety of materials such as dextran
= 0.023kg scaffold/kg cells
(Cytodex 1/3), gelatine (Cultispher), cellulose (Cytopore), polyethy-
lene (Cytoline), polystyrene (Synthemax II, SoloHill, Hillex), most Cost estimates for potential scaffold raw materials are de-
of which are either inedible or animal-derived. In addition to the rived from the median lower-bound prices of food-grade ma-
cell density at confluence, which is dependant on cell type, the terial, at the kilogram scale, from multiple suppliers at Al-
ibaba (November 2020). Importantly, this cost analysis con-
marginal cost of microcarriers for cell expansion also depends on
siders only the raw material cost and excludes processing
both the cost and the growth area per mass of microcarriers. As-
costs, such as those arising from chemical modification to in-
suming a cell density of 1 × 105 cells/cm2 at confluence [302], the troduce necessary biological functions (e.g. cell adhesiveness
marginal cost of microcarriers would be a prohibitively expensive or chemical crosslinking), or scaffold fabrication.
$320 0 – $840 0/kg of cells produced, at current retail prices (Box
2). The application of existing food biopolymers as materials for Food-grade material Marginal scaffold
cost ($/kg) cost ($/kg cells)
microcarrier fabrication, such as gellan gum and zein [303,304],
could potentially mitigate microcarrier costs and ensure microcar- Alginate 10 0.23
rier edibility. HA (recombinant, 130 2.95
microbial)
Carrageenan 5 0.11
Box 2: Marginal cost analysis of scaffolds for cultured meat produc- Pectin 11.5 0.26
tion Gellan gum 19 0.43
Xanthan gum 2 0.05
Cellulose 3 0.07
A) Microcarriers for cell expansion: Starch 0.5 0.01
The marginal cost of microcarriers for cell expansion is Soy isolate 2 0.05
dependant on: Zein (corn) 2 0.05
(1) Cell size, which is cell type-dependant and determines
the maximum achievable cell density at confluence,
(2) Microcarrier cost per mass, and
(3) Microcarrier growth area per mass

13
JID: ACTBIO
ARTICLE IN PRESS [m5G;February 21, 2021;17:50]

S. Ng and M. Kurisawa Acta Biomaterialia xxx (xxxx) xxx

4.4. Repurposing food biopolymers for cultured meat production The relative novelty of the cultured meat field, and the concen-
tration of cultured meat research in industrial start-ups as com-
As discussed in Section 3, a wide range of edible biopoly- pared with academia, are two reasons for the current dearth of
mers, from polysaccharides, proteins, lipids to synthetics have had data on the meat quality attributes of cultured meat prototypes.
long-standing applications as added ingredients in meat prod- To date, there are only two studies of engineered bovine muscle
ucts, but amongst these, only alginate and gelatine are widely tissue and one of engineered bovine adipose tissue for cultured
applied as tissue engineering scaffolds [305,306], and even so, meat applications [57,58,284]. Importantly, in addition to charac-
only recently have bioengineers started to apply them expressly terizing the phenotype of bovine cells during culture, these stud-
for cultured meat applications [57,284]. Due to an increasing in- ies utilized potentially scalable scaffold fabrication methods to fab-
terest in more sustainable and low-cost sources of tissue engi- ricate macroscale cultured meat prototypes. Moreover, with these
neering scaffolds, biomaterials engineers have started to fabri- macroscale prototypes, they demonstrated the application of con-
cate ECM-mimetic scaffolds with bioreactor-compatible morpholo- ventional methods for meat quality evaluation, such as textural
gies (porous hydrogels, fibres, microcarriers) using other edible profile analysis and human sensory panels, thereby paving the way
biopolymers and evaluating their biocompatibility, including car- for future integrative studies at the interface of tissue engineering
rageenan [307–317], pectin [318–326], cellulose [158,159,178,327– and meat science.
334], guar gum [335–338], gellan gum [304,339–348], xanthan
gum [346,349–354], konjac [355–360], protein isolates from soy or 5. Outlook and conclusion
corn (zein) [58,134,332,334,361–372], and starches [163,358,373–
378] (Table 3). Cultured meat production, while conceptually attractive, faces
Many of these edible biopolymers are useful as biomaterials fundamental challenges in technical feasibility and industrial scal-
due to their high water binding capacities, which enable them ability. Due to additional novel constraints on scaffold edibility,
to recapitulate the highly hydrated state of native ECM, or their sustainability and cost-efficiency, it is unlikely that a piecemeal
gelation behaviour, which could supplement the mechanical prop- merger of the meat science and tissue engineering fields, where
erties of softer materials [307,310,344,351,379]. However, biopoly- tissue engineers produce live muscle tissue and meat scientists
mer composites and chemical modifications that enable covalent characterize the post-mortem tissue via their respective conven-
crosslinking are often employed to enable gelation of non-gelling tional methods, will suffice to solve these non-trivial challenges.
agents, overcome harsh physical gelation conditions that are not Rather, bridging conceptual and knowledge divides between the
cytocompatible, and improve the range and control of gel physico- historically disparate fields of meat science and biomaterials engi-
chemical properties [158,311,380,381]. For example, unmodified κ - neering may help to illuminate novel strategies to address some of
carrageenan undergoes potassium-dependant ionic gelation upon these challenges (Fig. 3). A holistic consideration of the constraints
cooling from its heat-solubilized form, but starts gelling at 50 °C discussed in this review with an integrated perspective into bio-
to form a brittle, thermoreversible hydrogel that exhibits a non- materials and meat science earlier on in the cultured meat de-
linear variation of mechanical stiffness with cooling tempera- sign process could accelerate successful incorporation into existing
ture, and dissociates in physiological buffers [382]. In contrast, meat manufacturing pipelines and provide an advantage in even-
methacrylated κ -carrageenan forms photopolymerized, covalently tual market adoption.
crosslinked gels that are thermally and mechanically stable in
Declaration of Competing Interest
physiological buffers, and retain the viability of encapsulated cells
[308]. Other gelling biopolymers that undergo similar cooling-
There is no conflict of interest and disclosure associated with
dependant gelation (agar, gellan gum and gelatine) or conversely
the manuscript.
require high temperatures to induce thermal gelation driven by hy-
drophobic aggregation (konjac, methycellulose, soy and pea protein Acknowledgements
isolates), are also unable to support cell encapsulation and offer
limited control over hydrogel properties unless modified to sup- This work was funded by the Institute of Bioengineering and
port more biocompatible gelation mechanisms. Nanotechnology, Biomedical Research Council, Agency for Science,
The general lack of cell adhesiveness amongst edible biopoly- Technology and Research (A∗ STAR), Singapore.
mers, particularly polysaccharides, also requires direct chemical
modification with cell adhesion moieties or the development References
of composites that incorporate a cell-adhesive protein [158]. An
[1] E.A. Specht, D.R. Welch, E.M. Rees Clayton, C.D. Lagally, Opportunities for
example of industrial effort in this direction is the development applying biomedical production and manufacturing methods to the devel-
of pectin-thiopropionylamide conjugates by Modern Meadows, opment of the clean meat industry, Biochem. Eng. J. 132 (2018) 161–168,
which forms a covalently crosslinked hydrogel with thiol-modified doi:10.1016/j.bej.2018.01.015.
[2] N. Stephens, L. Di Silvio, I. Dunsford, M. Ellis, A. Glencross, A. Sexton, Bringing
proteins that naturally contain the RGD adhesive peptide domain, cultured meat to market: technical, socio-political, and regulatory challenges
such as cardosin at a cytocompatible pH [137], in contrast with in cellular agriculture, Trends Food Sci. Technol. (2018), doi:10.1016/j.tifs.2018.
the non-cytocompatible acidic and high sugar conditions required 04.010.
[3] N. Stephens, A.E. Sexton, C. Driessen, Making Sense of Making Meat: key Mo-
to form native pectin physical gels [52]. amongst the above
ments in the First 20 Years of Tissue Engineering Muscle to Make Food, Front.
biocompatible and edible biopolymers, carrageenan [314,315], Sustain. Food Syst. (2019), doi:10.3389/fsufs.2019.0 0 045.
pectin [318,320,322,325], cellulose [178,329–331,334], gellan gum [4] S. Chriki, J.F. Hocquette, The Myth of Cultured Meat: a Review, Front. Nutr. 7
(2020) 1–9, doi:10.3389/fnut.2020.0 0 0 07.
[340–346], konjac [357], starch [163,373–377], soy protein isolate
[5] W. Verbeke, A. Marcu, P. Rutsaert, R. Gaspar, B. Seibt, D. Fletcher, J. Barnett,
[58,334,365] and corn-derived zein protein [362,364,383] have Would you eat cultured meat?”: consumers’ reactions and attitude formation
demonstrated feasibility as non-composite or composite scaf- in Belgium, Portugal and the United Kingdom, Meat Sci (2015), doi:10.1016/j.
folds for various engineered tissues [304,314,318,320,322,329– meatsci.2014.11.013.
[6] I. Fraeye, M. Kratka, H. Vandenburgh, L. Thorrez, Sensorial and Nutritional
331,334,340–343,345,346,357,364,371], although to date, only Aspects of Cultured Meat in Comparison to Traditional Meat: much to Be In-
pectin, cellulose, gellan gum and soy protein isolate have been ferred, Front. Nutr. 7 (2020), doi:10.3389/fnut.2020.0 0 035.
investigated for their suitability in generating engineered muscle [7] S. Glorieux, L. Steen, D. Van de Walle, K. Dewettinck, I. Foubert, I. Fraeye, Ef-
fect of Meat Type, Animal Fat Type, and Cooking Temperature on Microstruc-
tissue [58,137,178,342] (Table 3). tural and Macroscopic Properties of Cooked Sausages, Food Bioprocess Tech-
nol. (2019), doi:10.1007/s11947- 018- 2190- 6.

14
JID: ACTBIO
ARTICLE IN PRESS [m5G;February 21, 2021;17:50]

S. Ng and M. Kurisawa Acta Biomaterialia xxx (xxxx) xxx

[8] G. Feiner, Meat Products Handbook, Practical Science and Technology, Wood- [37] S.B. Smith, H. Kawachi, C.B. Choi, C.W. Choi, G. Wu, J.E. Sawyer, Cellular regu-
head, 2006. lation of bovine intramuscular adipose tissue development and composition,
[9] M.V. Dodson, R.E. Allen, M. Du, W.G. Bergen, S.G. Velleman, S.P. Poulos, J. Anim. Sci. 87 (2009) 72–82, doi:10.2527/jas.2008-1340.
M. Fernyhough-Culver, M.B. Wheeler, S.K. Duckett, M.R.I. Young, B.H. Voy, [38] A.C. Grant, G. Ortiz-Colòn, M.E. Doumit, D.D. Buskirk, Optimization of in vitro
Evolution of meat animal growth research during the past 50 years, J. Anim. conditions for bovine subcutaneous and intramuscular preadipocyte differen-
Sci. 93 (2015) 457–481, doi:10.2527/jas2014-8221. tiation, J. Anim. Sci. 86 (2008) 73–82, doi:10.2527/jas.2007-0379.
[10] S. Chriki, B. Picard, Y. Faulconnier, D. Micol, J.P. Brun, M. Reichstadt, C. Jurie, [39] A.J. Lengi, B.A. Corl, Factors influencing the differentiation of bovine
D. Durand, G. Renand, L. Journaux, J.F. Hocquette, A data warehouse of muscle preadipocytes in vitro, J. Anim. Sci. 88 (2010) 1999–2008, doi:10.2527/jas.
characteristics and beef quality in France and a demonstration of potential 2009-2439.
applications, Ital. J. Anim. Sci. (2013), doi:10.4081/ijas.2013.e41. [40] K.D. Fish, N.R. Rubio, A.J. Stout, J.S.K. Yuen, D.L. Kaplan, Prospects and chal-
[11] Y.H. Hui, J.L. Aalhus, L. Cocolin, I. Guerrero-Legarreta, L.M. Nollet, R.W. Pur- lenges for cell-cultured fat as a novel food ingredient, Trends Food Sci. Tech-
chas, M.W. Schilling, P. Stanfield, Y.L. Xiong, Handbook of Meat and Meat Pro- nol. 98 (2020) 53–67, doi:10.1016/j.tifs.2020.02.005.
cessing, 2nd Edition, 2012. [41] E.D. Rosen, O.A. MacDougald, Adipocyte differentiation from the inside out,
[12] K.L. Pearce, K. Rosenvold, H.J. Andersen, D.L. Hopkins, Water distribution and Nat. Rev. Mol. Cell Biol. 7 (2006) 885–896, doi:10.1038/nrm2066.
mobility in meat during the conversion of muscle to meat and ageing and the [42] American Meat Science Association (AMSA), Research Guidelines for Cook-
impacts on fresh meat quality attributes - A review, Meat Sci (2011), doi:10. ery, Sensory Evaluation, and Instrumental Tenderness Measurements of Meat,
1016/j.meatsci.2011.04.007. 2016. http://www.meatscience.org.
[13] S.K. Matarneh, E.M. England, T.L. Scheffler, D.E. Gerrard, The Conver- [43] U. Andjelković, D. Josić, Mass spectrometry based proteomics as foodomics
sion of Muscle to Meat, Lawrie’s Meat Sci., 8th Ed., 2017, doi:10.1016/ tool in research and assurance of food quality and safety, Trends Food Sci.
B978- 0- 08- 100694- 8.00005- 4. Technol. (2018), doi:10.1016/j.tifs.2018.04.008.
[14] K.G. Grunert, L. Bredahl, K. Brunsø, Consumer perception of meat quality and [44] J.L. Damez, S. Clerjon, Meat quality assessment using biophysical methods re-
implications for product development in the meat sector - A review, Meat lated to meat structure, Meat Sci. (2008), doi:10.1016/j.meatsci.2008.05.039.
Sci. (2004), doi:10.1016/S0309-1740(03)00130-X. [45] J.X. Guinard, R. Mazzucchelli, The sensory perception of texture and mouth-
[15] M. Rødbotten, E. Kubberød, P. Lea, Ø. Ueland, A sensory map of the meat feel, Trends Food Sci. Technol. (1996), doi:10.1016/0924-2244(96)10025-X.
universe. Sensory profile of meat from 15 species, Meat Sci. (2004), doi:10. [46] E. Huff Lonergan, W. Zhang, S.M. Lonergan, Biochemistry of postmortem
1016/j.meatsci.2004.02.016. muscle - Lessons on mechanisms of meat tenderization, Meat Sci. (2010),
[16] G. Offer, P. Knight, R. Jeacocke, R. Almond, T. Cousins, J. Elsey, N. Parsons, doi:10.1016/j.meatsci.2010.05.004.
A. Sharp, R. Starr, P. Purslow, The structural basis of the water-holding, [47] P. Ertbjerg, E. Puolanne, Muscle structure, sarcomere length and influences on
appearance and toughness of meat and meat products, Food Microstruct. meat quality: a review, Meat Sci (2017), doi:10.1016/j.meatsci.2017.04.261.
(1989). [48] R.D. Warner, C.K. McDonnell, A.E.D. Bekhit, J. Claus, R. Vaskoska, A. Sikes,
[17] M.J. Post, J.F. Hocquette, New Sources of Animal Proteins, Vitro Meat, Elsevier F.R. Dunshea, M. Ha, Systematic review of emerging and innovative technolo-
Ltd„ 2017, doi:10.1016/B978- 0- 08- 100593- 4/00017- 5. gies for meat tenderisation, Meat Sci. (2017), doi:10.1016/j.meatsci.2017.04.
[18] S.B. Kanatous, P.P.A. Mammen, P.B. Rosenberg, C.M. Martin, M.D. White, 241.
J.M. DiMaio, G. Huang, S. Muallem, D.J. Garry, Hypoxia reprograms calcium [49] A.A. Bekhit, D.L. Hopkins, G. Geesink, A.A. Bekhit, P. Franks, Exogenous Pro-
signaling and regulates myoglobin expression, Am. J. Physiol. - Cell Physiol. teases for Meat Tenderization, Crit. Rev. Food Sci. Nutr. (2014), doi:10.1080/
(2009), doi:10.1152/ajpcell.00428.2008. 10408398.2011.623247.
[19] A.E. Schlater, M.A. De Miranda, M.A. Frye, S.J. Trumble, S.B. Kanatous, Chang- [50] L. Thorrez, H. Vandenburgh, Challenges in the quest for ‘clean meat, Nat.
ing the paradigm for myoglobin: a novel link between lipids and Myoglobin, Biotechnol. (2019), doi:10.1038/s41587- 019- 0043- 0.
J. Appl. Physiol. (2014), doi:10.1152/japplphysiol.00973.2013. [51] S.T. Joo, G.D. Kim, Y.H. Hwang, Y.C. Ryu, Control of fresh meat quality through
[20] R. Simsa, J. Yuen, A. Stout, N. Rubio, P. Fogelstrand, D.L. Kaplan, Extracellular manipulation of muscle fiber characteristics, Meat Sci. (2013), doi:10.1016/j.
heme proteins influence bovine myosatellite cell proliferation and the color meatsci.2013.04.044.
of cell-based meat, Foods. (2019), doi:10.3390/foods8100521. [52] R. Tarté, Ingredients in meat products: properties, functionality and applica-
[21] Y. Jin, X. He, K. Andoh-Kumi, R.Z. Fraser, M. Lu, R.E. Goodman, Evaluating Po- tions, 2009. 10.1007/978-0-387-71327-4.
tential Risks of Food Allergy and Toxicity of Soy Leghemoglobin Expressed in [53] B.L. Dekkers, R.M. Boom, A.J. van der Goot, Structuring processes for meat
Pichia pastoris, Mol. Nutr. Food Res. (2018), doi:10.10 02/mnfr.20170 0297. analogues, Trends Food Sci. Technol. (2018), doi:10.1016/j.tifs.2018.08.011.
[22] M.N. Hamilton, C.E. Ewing, Food coloring composition, CA2314727C, 2005. [54] M.C. Bourne, Texture Profile Analysis, Food Technol. 32 (1978) 62–66.
[23] P. Eisner, R. Osen, Extruded vegetable protein product with coloring vegetable [55] J.R. Claus, Methods for the Objective Measurement of Meat Product Texture,
ingredients and method of preparation, DE102015203653A1, 2016. in: Reciprocal Meat Conf. Proceedings, Am. Meat Sci. Assoc., 1995, pp. 96–101.
[24] A.M.S. AssociationAMSA Meat Color Measurement Guidelines, AMSA, 2012. [56] K.O. Honikel, Reference methods for the assessment of physical character-
[25] M.I. Khan, C. Jo, M.R. Tariq, Meat flavor precursors and factors influencing istics of meat, Meat Sci. 49 (1998) 447–457, doi:10.1016/S0309-1740(98)
flavor precursors-A systematic review, Meat Sci (2015), doi:10.1016/j.meatsci. 0 0 034-5.
2015.08.002. [57] L.A. MacQueen, C.G. Alver, C.O. Chantre, S. Ahn, L. Cera, G.M. Gonza-
[26] C.R. Calkins, J.M. Hodgen, A fresh look at meat flavor, Meat Sci (2007), doi:10. lez, B.B. O’Connor, D.J. Drennan, M.M. Peters, S.E. Motta, J.F. Zimmerman,
1016/j.meatsci.2007.04.016. K.K. Parker, Muscle tissue engineering in fibrous gelatin: implications for
[27] J.D. Wood, G.R. Nute, R.I. Richardson, F.M. Whittington, O. Southwood, G. Plas- meat analogs, Npj Sci. Food. (2019), doi:10.1038/s41538- 019- 0054- 8.
tow, R. Mansbridge, N. Da Costa, K.C. Chang, Effects of breed, diet and muscle [58] T. Ben-Arye, Y. Shandalov, S. Ben-Shaul, S. Landau, Y. Zagury, I. Ianovici,
on fat deposition and eating quality in pigs, Meat Sci. (2004), doi:10.1016/j. N. Lavon, S. Levenberg, Textured soy protein scaffolds enable the generation
meatsci.20 04.01.0 07. of three-dimensional bovine skeletal muscle tissue for cell-based meat, Nat.
[28] M.S. Arshad, M. Sohaib, R.S. Ahmad, M.T. Nadeem, A. Imran, M.U. Arshad, Food. (2020), doi:10.1038/s43016- 020- 0046- 5.
J.H. Kwon, Z. Amjad, Ruminant meat flavor influenced by different factors [59] L.B. Kleinert, J.B. Hoying, S.K. Williams, The neointima formed in endothelial
with special reference to fatty acids, Lipids Health Dis. 17 (2018) 1–13, cell sodded ePTFE vascular grafts results from both cellular-hyperplasia and
doi:10.1186/s12944-018-0860-z. extracellular-hypertrophy, Cell Transplant. (1996), doi:10.1016/0963-6897(96)
[29] J.D. Wood, M. Enser, A.V. Fisher, G.R. Nute, P.R. Sheard, R.I. Richardson, 0 0 024-3.
S.I. Hughes, F.M. Whittington, Fat deposition, fatty acid composition and meat [60] V. Mudera, A.S.T. Smith, M.A. Brady, M.P. Lewis, The effect of cell density on
quality: a review, Meat Sci. (2008), doi:10.1016/j.meatsci.2007.07.019. the maturation and contractile ability of muscle derived cells in a 3D tissue-
[30] D.S. Mottram, Flavour formation in meat and meat products: a review, Food engineered skeletal muscle model and determination of the cellular and me-
Chem. (1998), doi:10.1016/S0308-8146(98)0 0 076-4. chanical stimuli required for the synthesis of a postural phenotype, J. Cell.
[31] F. Shahidi, L.J. Rubin, L.A. D’Souza, Meat Flavor Volatiles: a Review of the Physiol. (2010), doi:10.1002/jcp.22271.
Composition, Techniques of Analysis, and Sensory Evaluation, Crit. Rev. Food [61] C.S. Cheng, B.N.J. Davis, L. Madden, N. Bursac, G.A. Truskey, Physiology and
Sci. Nutr. (1986), doi:10.1080/10408398609527435. metabolism of tissue-engineered skeletal muscle, Exp. Biol. Med. (2014),
[32] M. Kosowska, M.A. Majcher, T. Fortuna, Volatile compounds in meat and meat doi:10.1177/1535370214538589.
products, Food Sci. Technol. (2017), doi:10.1590/1678-457X.08416. [62] L. Madden, M. Juhas, W.E. Kraus, G.A. Truskey, N. Bursac, Bioengineered hu-
[33] C. Lamb, Barcelona’s Cubiq Foods Raises €5 M to Produce Better-for- man myobundles mimic clinical responses of skeletal muscle to drugs, Elife.
You Cultured Fat, (2020). https://thespoon.tech/barcelonas- cubiq- foods- (2015), doi:10.7554/eLife.04885.
raises- e5m- to- produce- better- for- you- cultured- fat/ (accessed July 27, 2020). [63] N.M. Wragg, D.J. Player, N.R.W. Martin, Y. Liu, M.P. Lewis, Development of
[34] O. Morrison, Coronavirus will boost clean meat trend, says supplier, tissue-engineered skeletal muscle manufacturing variables, Biotechnol. Bio-
(2020). https://www.foodnavigator.com/Article/2020/04/16/Coronavirus-will- eng. (2019), doi:10.1002/bit.27074.
boost- clean- meat- trend- says- supplier (accessed July 27, 2020). [64] J. Bradbury, Docosahexaenoic acid (DHA): an ancient nutrient for the modern
[35] P. Wu, K. Sato, F. Suzuta, Y. Hikasa, K. Kagota, Effects of lipid-related factors human brain, Nutrients (2011), doi:10.3390/nu3050529.
on adipocyte differentiation of bovine stromal-vascular cells in primary cul- [65] R. Obeid, S.G. Heil, M.M.A. Verhoeven, E.G.H.M. van den Heuvel, L.C.P.G.M. de
ture, J. Vet. Med. Sci. 62 (20 0 0) 933–939. Groot, S.J.P.M. Eussen, Vitamin B12 intake from animal foods, biomarkers, and
[36] K. Sato, N. Nakanishi, M. Mitsumoto, Culture conditions supporting adipose health aspects, Front. Nutr. (2019), doi:10.3389/fnut.2019.0 0 093.
conversion of stromal-vascular cells from bovine intramuscular adipose [66] F. Jimenez-Colmenero, L. Salcedo-Sandoval, R. Bou, S. Cofrades, A.M. Herrero,
tissues, J. Vet. Med. Sci. 58 (1996) 1073–1078 http://www.mendeley. C. Ruiz-Capillas, Novel applications of oil-structuring methods as a strategy to
com/research/geology- volcanic- history-eruptive-style-yakedake-volcano- improve the fat content of meat products, Trends Food Sci. Technol. (2015),
group- central- japan/. doi:10.1016/j.tifs.2015.04.011.

15
JID: ACTBIO
ARTICLE IN PRESS [m5G;February 21, 2021;17:50]

S. Ng and M. Kurisawa Acta Biomaterialia xxx (xxxx) xxx

[67] Y. Qin, J. Jiang, L. Zhao, J. Zhang, F. Wang, Chapter 13: Applications of flour-based nanofibres by electrospinning method, Int. J. Food Sci. Technol. 53
Alginate as a Functional Food Ingredient, Elsevier Inc., 2018, doi:10.1016/ (2018) 1269–1277, doi:10.1111/ijfs.13707.
B978- 0- 12- 811449- 0.0 0 013-X. [97] M. Wiebe, Myco-protein from fusarium venenatum: a well-established prod-
[68] X.D. Sun, S.D. Arntfield, Gelation properties of salt-extracted pea protein iso- uct for human consumption, Appl. Microbiol. Biotechnol. (2002), doi:10.1007/
late induced by heat treatment: effect of heating and cooling rate, Food s0 0253-0 02-0931-x.
Chem. (2011), doi:10.1016/j.foodchem.2010.07.063. [98] W.J. Means, G.R. Schmidt, Algin/Calcium Gel as a Raw and Cooked Binder
[69] S. Comfort, N.K. Howell, Gelation properties of soya and whey protein isolate in Structured Beef Steaks, J. Food Sci. (1986), doi:10.1111/j.1365-2621.1986.
mixtures, Food Hydrocoll. (2002), doi:10.1016/S0268- 005X(02)00033- 4. tb10836.x.
[70] D. Santhi, A. Kalaikannan, P. Malairaj, S. Arun Prabhu, Application of microbial [99] S.K. WILLIAMS, J.L. OBLINGER, R.L. WEST, EVALUATION OF A CALCIUM AL-
transglutaminase in meat foods: a review, Crit. Rev. Food Sci. Nutr. (2017), GINATE FILM FOR USE ON BEEF CUTS, J. Food Sci. (1978), doi:10.1111/j.
doi:10.1080/10408398.2014.945990. 1365-2621.1978.tb02288.x.
[71] M.M. Kasprzak, W. Macnaughtan, S. Harding, P. Wilde, B. Wolf, Stabilisation of [100] K.G. WANSTEDT, S.C. SEIDEMAN, L.S. DONNELLY, N.M. QUENZER, Sensory At-
oil-in-water emulsions with non-chemical modified gelatinised starch, Food tributes of Precooked, Calcium Alginate-Coated Pork Patties, J. Food Prot. 44
Hydrocoll. (2018), doi:10.1016/j.foodhyd.2018.03.002. (1981) 732–735, doi:10.4315/0362- 028x- 44.10.732.
[72] E.D. Ngouémazong, S. Christiaens, A. Shpigelman, A. Van Loey, M. Hendrickx, [101] M.E. Danoviz, Z. Yablonka-Reuveni, Skeletal muscle satellite cells: background
The Emulsifying and Emulsion-Stabilizing Properties of Pectin: a Review, and methods for isolation and analysis in a primary culture system, Methods
Compr. Rev. Food Sci. Food Saf. (2015), doi:10.1111/1541-4337.12160. Mol. Biol. (2012), doi:10.1007/978- 1- 61779- 343- 1_2.
[73] B. Gómez, F.J. Barba, R. Domínguez, P. Putnik, D. Bursać Kovačević, M. Pateiro, [102] R. Chen, L. Li, L. Feng, Y. Luo, M. Xu, K.W. Leong, R. Yao, Biomaterial-assisted
F. Toldrá, J.M. Lorenzo, Microencapsulation of antioxidant compounds through scalable cell production for cell therapy, Biomaterials 230 (2020) 119627,
innovative technologies and its specific application in meat processing, doi:10.1016/j.biomaterials.2019.119627.
Trends Food Sci. Technol. (2018), doi:10.1016/j.tifs.2018.10.006. [103] G. Molnar, N.A. Schroedl, S.R. Gonda, C.R. Hartzell, Skeletal muscle satellite
[74] K.G.C. Weel, A.E.M. Boelrijk, A.C. Alting, P.J.J.M. Van Mil, J.J. Burger, H. Grup- cells cultured in simulated microgravity, Vitr. Cell. Dev. Biol. - Anim. (1997),
pen, A.G.J. Voragen, G. Smit, Flavor release and perception of flavored whey doi:10.1007/s11626- 997- 0010- 9.
protein gels: perception is determined by texture rather than by release, J. [104] K.J.M. Boonen, K.Y. Rosaria-Chak, F.P.T. Baaijens, D.W.J. Van Der Schaft,
Agric. Food Chem. (2002), doi:10.1021/jf0202786. M.J. Post, Essential environmental cues from the satellite cell niche: opti-
[75] D. Renard, F. Van De Velde, R.W. Visschers, The gap between food gel struc- mizing proliferation and differentiation, Am. J. Physiol. - Cell Physiol. (2009),
ture, texture and perception, Food Hydrocoll. (2006), doi:10.1016/j.foodhyd. doi:10.1152/ajpcell.0 0 015.20 09.
2005.10.014. [105] K. Gwon, E. Kim, G. Tae, Heparin-hyaluronic acid hydrogel in support of cel-
[76] A.R. Patel, R.A. Nicholson, A.G. Marangoni, Applications of fat mimetics for lular activities of 3D encapsulated adipose derived stem cells, Acta Biomater.
the replacement of saturated and hydrogenated fat in food products, Curr. 49 (2017) 284–295, doi:10.1016/j.actbio.2016.12.001.
Opin. Food Sci. 33 (2020) 61–68, doi:10.1016/j.cofs.2019.12.008. [106] L.E. Flynn, G.D. Prestwich, J.L. Semple, K.A. Woodhouse, Proliferation and dif-
[77] W.T. Kao, K.W. Lin, Quality of reduced-fat frankfurter modified by konjac- ferentiation of adipose-derived stem cells on naturally derived scaffolds, Bio-
starch mixed gels, J. Food Sci. (2006), doi:10.1111/j.1750-3841.2006.00003.x. materials 29 (2008) 1862–1871, doi:10.1016/j.biomaterials.2007.12.028.
[78] L.C. Hoffman, F.D. Mellett, Quality characteristics of low fat ostrich meat pat- [107] C. Yu, A. Kornmuller, C. Brown, T. Hoare, L.E. Flynn, Decellularized adipose tis-
ties formulated with either pork lard or modified corn starch, soya isolate sue microcarriers as a dynamic culture platform for human adipose-derived
and water, Meat Sci. (2003), doi:10.1016/S0309- 1740(02)00293- 0. stem/stromal cell expansion, Biomaterials (2017), doi:10.1016/j.biomaterials.
[79] A.M. Herrero, P. Carmona, F. Jiménez-Colmenero, C. Ruiz-Capillas, Polysaccha- 2016.12.017.
ride gels as oil bulking agents: technological and structural properties, Food [108] V. Jossen, C. van den Bos, R. Eibl, D. Eibl, Manufacturing human mesenchymal
Hydrocoll. (2014), doi:10.1016/j.foodhyd.2013.08.008. stem cells at clinical scale: process and regulatory challenges, Appl. Microbiol.
[80] G.R. Krawczyk, A. Venables, D. Tuason, Microcrystalline cellulose, Handb. Hy- Biotechnol. (2018), doi:10.10 07/s0 0253- 018- 8912- x.
drocoll, 2nd Ed., 2009, doi:10.1533/9781845695873.740. [109] E. Swartz, Meeting the Needs of the Cell-Based Meat Industry, Chem. Eng.
[81] R.T. Heck, J.M. Lorenzo, B.A. Dos Santos, A.J. Cichoski, C.R. de Menezes, (CEP), AIChE Mag. (2019) 41–45 https://ezproxy.usim.edu.my:2056/docview/
P.C.B. Campagnol, Microencapsulation of healthier oils: an efficient strategy 2310613647/fulltextPDF/AF713AB89EE54383PQ/1?accountid=33993.
to improve the lipid profile of meat products, Curr. Opin. Food Sci. (2020), [110] G. Zhang, X. Zhao, X. Li, G. Du, J. Zhou, J. Chen, Challenges and possibilities for
doi:10.1016/j.cofs.2020.04.010. bio-manufacturing cultured meat, Trends Food Sci. Technol. 97 (2020) 443–
[82] M.A. Asgar, A. Fazilah, N. Huda, R. Bhat, A.A. Karim, Nonmeat protein alter- 450, doi:10.1016/j.tifs.2020.01.026.
natives as meat extenders and meat analogs, Compr. Rev. Food Sci. Food Saf. [111] S.J. Allan, P.A. De Bank, M.J. Ellis, Bioprocess Design Considerations for Cul-
(2010), doi:10.1111/j.1541-4337.2010.00124.x. tured Meat Production With a Focus on the Expansion Bioreactor, Front. Sus-
[83] N.R. Rubio, K.D. Fish, B.A. Trimmer, D.L. Kaplan, Possibilities for Engineered tain. Food Syst. 3 (2019), doi:10.3389/fsufs.2019.0 0 044.
Insect Tissue as a Food Source, Front. Sustain. Food Syst. (2019), doi:10.3389/ [112] OECDOECD Environmental Outlook to 2030, OECD, 2008, doi:10.1787/
fsufs.2019.0 0 024. 9789264040519-en.
[84] G. Forgacs, F. Marga, K.R. Jakab, ENGINEERED COMESTIBLE MEAT, 2012. [113] P. Nold, C. Brendel, A. Neubauer, G. Bein, H. Hackstein, Good manufac-
[85] R. Osen, U. Schweiggert-Weisz, High-Moisture Extrusion : meat Analogues, turing practice-compliant animal-free expansion of human bone marrow
Refence Modul. Food Sci. 1 (2016) 1–7, doi:10.1016/j.elspec.20 09.07.0 01. derived mesenchymal stroma cells in a closed hollow-fiber-based biore-
[86] G.A. Krintiras, J. Gadea Diaz, A.J. Van Der Goot, A.I. Stankiewicz, G.D. Stefani- actor, Biochem. Biophys. Res. Commun. (2013), doi:10.1016/j.bbrc.2012.11.
dis, On the use of the Couette Cell technology for large scale production of 001.
textured soy-based meat replacers, J. Food Eng. (2016), doi:10.1016/j.jfoodeng. [114] S.A. Tuin, B. Pourdeyhimi, E.G. Loboa, Fabrication of novel high surface area
2015.08.021. mushroom gilled fibers and their effects on human adipose derived stem
[87] Y. Ren, Texturized Vegetable Proteins: the Challenges and Solutions to Mim- cells under pulsatile fluid flow for tissue engineering applications, Acta Bio-
icking Meat Texture from Plant Proteins, 2011. mater. (2016), doi:10.1016/j.actbio.2016.03.025.
[88] S. Samard, B.Y. Gu, G.H. Ryu, Effects of extrusion types, screw speed and addi- [115] S. Khan, L. Davila, M. Salkhordeh, D.J. Stewart, S.H. Mei, L. McIntyre,
tion of wheat gluten on physicochemical characteristics and cooking stability D.W. Courtman, cGMP-compatible large-scale production of mesenchymal
of meat analogues, J. Sci. Food Agric. (2019), doi:10.1002/jsfa.9722. stem cells (MSCs) in Xeno- and serum-free media for allogeneic cell thera-
[89] K.S. Liu, F.H. Hsieh, Protein-protein interactions during high-moisture extru- pies, Cytotherapy (2018), doi:10.1016/j.jcyt.2018.02.107.
sion for fibrous meat analogues and comparison of protein solubility meth- [116] I. Roberts, S. Baila, R.B. Rice, M.E. Janssens, K. Nguyen, N. Moens, L. Ruban,
ods using different solvent systems, J. Agric. Food Chem. (2008), doi:10.1021/ D. Hernandez, P. Coffey, C. Mason, Scale-up of human embryonic stem cell
jf073343q. culture using a hollow fibre bioreactor, Biotechnol. Lett. (2012), doi:10.1007/
[90] V.L. Pietsch, M.A. Emin, H.P. Schuchmann, Process conditions influencing s10529- 012- 1033- 1.
wheat gluten polymerization during high moisture extrusion of meat analog [117] N.M.S. Bettahalli, J. Vicente, L. Moroni, G.A. Higuera, C.A. Van Blitterswijk,
products, J. Food Eng. (2017), doi:10.1016/j.jfoodeng.2016.10.027. M. Wessling, D.F. Stamatialis, Integration of hollow fiber membranes im-
[91] V. Rampon, P. Robert, N. Nicolas, E. Dufour, Protein structure and network proves nutrient supply in three-dimensional tissue constructs, Acta Biomater.
orientation in edible films prepared by spinning process, J. Food Sci. (1999), (2011), doi:10.1016/j.actbio.2011.06.012.
doi:10.1111/j.1365-2621.1999.tb15890.x. [118] Y. Yamamoto, A. Ito, H. Jitsunobu, K. Yamaguchi, Y. Kawabe, H. Mizumoto,
[92] M. Nieuwland, P. Geerdink, P. Brier, P. Van Den Eijnden, J.T.M.M. Henket, M. Kamihira, Hollow fiber bioreactor perfusion culture system for magnetic
M.L.P. Langelaan, N. Stroeks, H.C. Van Deventer, A.H. Martin, Food-grade elec- force-based skeletal muscle tissue engineering, J. Chem. Eng. Japan. (2012),
trospinning of proteins, Innov. Food Sci. Emerg. Technol. (2013), doi:10.1016/ doi:10.1252/jcej.11we237.
j.ifset.2013.09.004. [119] J.C. Gerlach, Y.C. Lin, C.A. Brayfield, D.M. Minteer, H. Li, J.P. Rubin, K.G. Marra,
[93] R. Boyer, High protein food product and process for its preparation, Adipogenesis of human adipose-derived stem cells within three-dimensional
US2682466A, 1954. hollow fiber-based bioreactors, Tissue Eng. - Part C Methods. (2012), doi:10.
[94] M.M. Sternberg, C.Y. Kim, Process for preparing wet spun proteinaceous fila- 1089/ten.tec.2011.0216.
ments, US3956514A, 1976. [120] B. Cunha, T. Aguiar, S.B. Carvalho, M.M. Silva, R.A. Gomes, M.J.T. Carrondo,
[95] F.K.G. Schreuders, B.L. Dekkers, I. Bodnár, P. Erni, R.M. Boom, A.J. van der Goot, P. Gomes-Alves, C. Peixoto, M. Serra, P.M. Alves, Bioprocess integration for hu-
Comparing structuring potential of pea and soy protein with gluten for meat man mesenchymal stem cells: from up to downstream processing scale-up to
analogue preparation, J. Food Eng. (2019), doi:10.1016/j.jfoodeng.2019.04. cell proteome characterization, J. Biotechnol. (2017), doi:10.1016/j.jbiotec.2017.
022. 01.014.
[96] S. Oguz, N. Tam, A. Aydogdu, G. Sumnu, S. Sahin, Development of novel pea [121] N. Wung, S.M. Acott, D. Tosh, M.J. Ellis, Hollow fibre membrane bioreac-

16
JID: ACTBIO
ARTICLE IN PRESS [m5G;February 21, 2021;17:50]

S. Ng and M. Kurisawa Acta Biomaterialia xxx (xxxx) xxx

tors for tissue engineering applications, Biotechnol. Lett. (2014), doi:10.1007/ neurogenic differentiation of human mesenchymal stem cells in 3D cul-
s10529- 014- 1619- x. ture, Biomaterials 31 (2010) 1148–1157, doi:10.1016/j.biomaterials.2009.10.
[122] S.B.M. Usuludin, X. Cao, M. Lim, Co-culture of stromal and erythroleukemia 042.
cells in a perfused hollow fiber bioreactor system as an in vitro bone mar- [148] Z. Li, Y. Gong, S. Sun, Y. Du, D. Lü, X. Liu, M. Long, Differential regulation of
row model for myeloid leukemia, Biotechnol. Bioeng. (2012), doi:10.1002/bit. stiffness, topography, and dimension of substrates in rat mesenchymal stem
24400. cells, Biomaterials (2013), doi:10.1016/j.biomaterials.2013.06.059.
[123] V. Bodiou, P. Moutsatsou, M.J. Post, Microcarriers for Upscaling Cultured Meat [149] J.S. Park, J.S. Chu, A.D. Tsou, R. Diop, Z. Tang, A. Wang, S. Li, The effect of
Production, Front. Nutr. 7 (2020) 1–16, doi:10.3389/fnut.2020.0 0 010. matrix stiffness on the differentiation of mesenchymal stem cells in response
[124] J.H. Lee, H.W. Jung, I.K. Kang, H.B. Lee, Cell behaviour on polymer sur- to TGF-β , Biomaterials (2011), doi:10.1016/j.biomaterials.2011.02.019.
faces with different functional groups, Biomaterials (1994), doi:10.1016/ [150] M. Kim, Y.H. Kim, G. Tae, Human mesenchymal stem cell culture on heparin-
0142-9612(94)90169-4. based hydrogels and the modulation of interactions by gel elasticity and hep-
[125] G.B. Schneider, A. English, M. Abraham, R. Zaharias, C. Stanford, J. Keller, The arin amount, Acta Biomater. 9 (2013) 7833–7844, doi:10.1016/j.actbio.2013.04.
effect of hydrogel charge density on cell attachment, Biomaterials (2004), 041.
doi:10.1016/j.biomaterials.2003.09.084. [151] J.M. Banks, L.C. Mozdzen, B.A.C. Harley, R.C. Bailey, The combined effects of
[126] A.K.L. Chen, X. Chen, A.B.H. Choo, S. Reuveny, S.K.W. Oh, Critical microcarrier matrix stiffness and growth factor immobilization on the bioactivity and dif-
properties affecting the expansion of undifferentiated human embryonic stem ferentiation capabilities of adipose-derived stem cells, Biomaterials 35 (2014)
cells, Stem Cell Res. (2011), doi:10.1016/j.scr.2011.04.007. 8951–8959, doi:10.1016/j.biomaterials.2014.07.012.
[127] S. Guo, X. Zhu, M. Li, L. Shi, J.L.T. Ong, D. Jańczewski, K.G. Neoh, Parallel Con- [152] M.J. Osiecki, T.D. Michl, B.K. Babur, M. Kabiri, K. Atkinson, W.B. Lott,
trol over Surface Charge and Wettability Using Polyelectrolyte Architecture: H.J. Griesser, M.R. Doran, Packed bed bioreactor for the isolation and expan-
effect on Protein Adsorption and Cell Adhesion, ACS Appl. Mater. Interfaces. sion of placental-derived mesenchymal stromal cells, PLoS ONE 10 (2015) 1–
(2016), doi:10.1021/acsami.6b09481. 18, doi:10.1371/journal.pone.0144941.
[128] L.C. Xu, C.A. Siedlecki, Effects of surface wettability and contact time on [153] D.W. Hutmacher, H. Singh, Computational fluid dynamics for improved biore-
protein adhesion to biomaterial surfaces, Biomaterials (2007), doi:10.1016/j. actor design and 3D culture, Trends Biotechnol. (2008), doi:10.1016/j.tibtech.
biomaterials.2007.03.032. 2007.11.012.
[129] C.J. Wilson, R.E. Clegg, D.I. Leavesley, M.J. Pearcy, Mediation of biomaterial- [154] T. Ben-Arye, S. Levenberg, Tissue Engineering for Clean Meat Production,
cell interactions by adsorbed proteins: a review, Tissue Eng. (2005), doi:10. Front. Sustain. Food Syst. 3 (2019) 1–19, doi:10.3389/fsufs.2019.0 0 046.
1089/ten.2005.11.1. [155] H.M. Leinonen, S. Lepola, E.M. Lipponen, T. Heikura, T. Koponen, N. Parker,
[130] M.C. Morán, G. Ruano, F. Cirisano, M. Ferrari, Mammalian cell viability on hy- S. Ylä-Herttuala, H.P. Lesch, Benchmarking of Scale-X Bioreactor System in
drophobic and superhydrophobic fabrics, Mater. Sci. Eng. C. (2019), doi:10. Lentiviral and Adenoviral Vector Production, Hum. Gene Ther. 31 (2020) 376–
1016/j.msec.2019.01.088. 384, doi:10.1089/hum.2019.247.
[131] S. Verbruggen, D. Luining, A. van Essen, M.J. Post, Bovine myoblast cell pro- [156] R. Portner, O.B. Platas, D. Fassnacht, D. Nehring, P. Czermak, H. Markl, Fixed
duction in a microcarriers-based system, Cytotechnology 70 (2018) 503–512, Bed Reactors for the Cultivation of Mammalian Cells: design, Performance
doi:10.1007/s10616- 017- 0101- 8. and Scale-Up, Open Biotechnol. J. (2007), doi:10.2174/187407070701014107.
[132] M. Hervy, J.L. Weber, M. Pecheul, P. Dolley-Sonneville, D. Henry, Y. Zhou, [157] S. Levenberg, J. Rouwkema, M. Macdonald, E.S. Garfein, D.S. Kohane, D.C. Dar-
Z. Melkoumian, Long term expansion of bone marrow-derived hMSCs on land, R. Marini, C.A. Van Blitterswijk, R.C. Mulligan, P.A. D’Amore, R. Langer,
novel synthetic microcarriers in xeno-free, defined conditions, PLoS ONE Engineering vascularized skeletal muscle tissue, Nat. Biotechnol. (2005),
(2014), doi:10.1371/journal.pone.0092120. doi:10.1038/nbt1109.
[133] S. Derakhti, S.H. Safiabadi-Tali, G. Amoabediny, M. Sheikhpour, Attachment [158] S. Torgbo, P. Sukyai, Bacterial cellulose-based scaffold materials for bone tis-
and detachment strategies in microcarrier-based cell culture technology: a sue engineering, Appl. Mater. Today. 11 (2018) 34–49, doi:10.1016/j.apmt.
comprehensive review, Mater. Sci. Eng. C. (2019), doi:10.1016/j.msec.2019. 2018.01.004.
109782. [159] R.J. Hickey, A.E. Pelling, Cellulose biomaterials for tissue engineering, Front.
[134] J. Dong, Q. Sun, J.Y. Wang, Basic study of corn protein, zein, as a biomaterial Bioeng. Biotechnol. (2019), doi:10.3389/fbioe.2019.0 0 045.
in tissue engineering, surface morphology and biocompatibility, Biomaterials [160] M. Hu, R. Deng, K.M. Schumacher, M. Kurisawa, H. Ye, K. Purnamawati,
(2004), doi:10.1016/j.biomaterials.2003.10.084. J.Y. Ying, Hydrodynamic spinning of hydrogel fibers, Biomaterials. 31 (2010)
[135] S. Tansaz, A.K. Durmann, R. Detsch, A.R. Boccaccini, Hydrogel films and micro- 863–869, doi:10.1016/j.biomaterials.2009.10.002.
capsules based on soy protein isolate combined with alginate, J. Appl. Polym. [161] Z.J. Meng, W. Wang, R. Xie, X.J. Ju, Z. Liu, L.Y. Chu, Microfluidic generation of
Sci. (2017), doi:10.1002/app.44358. hollow Ca-alginate microfibers, Lab Chip. (2016), doi:10.1039/c6lc00640j.
[136] M.A. Benjaminson, J.A. Gilchriest, M. Lorenz, In vitro edible muscle protein [162] U.H.T. Pham, M. Hanif, A. Asthana, S.M. Iqbal, A microfluidic device approach
production system (MPPS): stage 1, fish, Acta Astronaut. 51 (2002) 879–889, to generate hollow alginate microfibers with controlled wall thickness and
doi:10.1016/S0 094-5765(02)0 0 033-4. inner diameter, J. Appl. Phys. (2015), doi:10.1063/1.4919361.
[137] F.S. Marga, B.P. Purcell, G. Forgacs, A. Forgacs, Edible and animal-product-free [163] K. Tuzlakoglu, I. Pashkuleva, M.T. Rodrigues, M.E. Gomes, G.H. Van Lenthe,
microcarriers for engineered meat, US 9,752,122 B2, 2017. R. Müller, R.L. Reis, A new route to produce starch-based fiber mesh scaffolds
[138] D. Choquet, D.P. Felsenfeld, M.P. Sheetz, Extracellular matrix rigidity causes by wet spinning and subsequent surface modification as a way to improve
strengthening of integrin- cytoskeleton linkages, Cell (1997), doi:10.1016/ cell attachment and proliferation, J. Biomed. Mater. Res. - Part A. (2010),
S0 092-8674(0 0)81856-5. doi:10.1002/jbm.a.32358.
[139] K. Ghosh, Z. Pan, E. Guan, S. Ge, Y. Liu, T. Nakamura, X.D. Ren, M. Rafailovich, [164] X. Shi, S. Ostrovidov, Y. Zhao, X. Liang, M. Kasuya, K. Kurihara, K. Nakajima,
R.A.F. Clark, Cell adaptation to a physiologically relevant ECM mimic with dif- H. Bae, H. Wu, A. Khademhosseini, Microfluidic spinning of cell-responsive
ferent viscoelastic properties, Biomaterials (2007), doi:10.1016/j.biomaterials. grooved microfibers, Adv. Funct. Mater. (2015), doi:10.1002/adfm.201404531.
2006.09.038. [165] Y. Yang, J. Sun, X. Liu, Z. Guo, Y. He, D. Wei, M. Zhong, L. Guo, H. Fan,
[140] E.A. Klein, L. Yin, D. Kothapalli, P. Castagnino, F.J. Byfield, T. Xu, I. Levental, X. Zhang, Wet-spinning fabrication of shear-patterned alginate hydrogel mi-
E. Hawthorne, P.A. Janmey, R.K. Assoian, Cell-Cycle Control by Physiological crofibers and the guidance of cell alignment, Regen. Biomater. 4 (2017) 299–
Matrix Elasticity and In Vivo Tissue Stiffening, Curr. Biol. (2009), doi:10.1016/ 307, doi:10.1093/rb/rbx017.
j.cub.2009.07.069. [166] Y. Wang, R.K. Kankala, K. Zhu, S. Bin Wang, Y.S. Zhang, A.Z. Chen, Coaxial
[141] Y.H. Bae, K.L. Mui, B.Y. Hsu, S.L. Liu, A. Cretu, Z. Razinia, T. Xu, E. Puré, Extrusion of Tubular Tissue Constructs Using a Gelatin/GelMA Blend Bioink,
R.K. Assoian, A FAK-Cas-Rac-lamellipodin signaling module transduces extra- ACS Biomater. Sci. Eng. (2019), doi:10.1021/acsbiomaterials.9b00926.
cellular matrix stiffness into mechanosensitive cell cycling, Sci. Signal. (2014), [167] Y. Yu, L. Shang, J. Guo, J. Wang, Y. Zhao, Design of capillary microflu-
doi:10.1126/scisignal.2004838. idics for spinning cell-laden microfibers, Nat. Protoc. (2018), doi:10.1038/
[142] A.J. Engler, M.A. Griffin, S. Sen, C.G. Bönnemann, H.L. Sweeney, D.E. Dis- s41596- 018- 0051- 4.
cher, Myotubes differentiate optimally on substrates with tissue-like stiff- [168] D. Wei, J. Sun, J. Bolderson, M. Zhong, M.J. Dalby, M. Cusack, H. Yin, H. Fan,
ness: pathological implications for soft or stiff microenvironments, J. Cell Biol. X. Zhang, Continuous Fabrication and Assembly of Spatial Cell-Laden Fibers
(2004), doi:10.1083/jcb.200405004. for a Tissue-Like Construct via a Photolithographic-Based Microfluidic Chip,
[143] P.C. Georges, P.A. Janmey, Cell type-specific response to growth on soft mate- ACS Appl. Mater. Interfaces. (2017), doi:10.1021/acsami.7b0 0 078.
rials, J. Appl. Physiol. (2005), doi:10.1152/japplphysiol.01121.2004. [169] M. Hu, M. Kurisawa, R. Deng, C.M. Teo, A. Schumacher, Y.X. Thong,
[144] P.M. Gilbert, K.L. Havenstrite, K.E.G. Magnusson, A. Sacco, N.A. Leonardi, L. Wang, K.M. Schumacher, J.Y. Ying, Cell immobilization in gelatin-
P. Kraft, N.K. Nguyen, S. Thrun, M.P. Lutolf, H.M. Blau, Substrate elasticity reg- hydroxyphenylpropionic acid hydrogel fibers, Biomaterials. 30 (2009) 3523–
ulates skeletal muscle stem cell self-renewal in culture, Science (80-.) (2010), 3531, doi:10.1016/j.biomaterials.20 09.03.0 04.
doi:10.1126/science.1191035. [170] Y. Cheng, Y. Yu, F. Fu, J. Wang, L. Shang, Z. Gu, Y. Zhao, Controlled Fabrica-
[145] T. Boontheekul, E.E. Hill, H.J. Kong, D.J. Mooney, Regulating myoblast phe- tion of Bioactive Microfibers for Creating Tissue Constructs Using Microfluidic
notype through controlled gel stiffness and degradation, Tissue Eng. (2007), Techniques, ACS Appl. Mater. Interfaces. (2016), doi:10.1021/acsami.5b11445.
doi:10.1089/ten.2006.0356. [171] S. Jana, S.K.L. Levengood, M. Zhang, Anisotropic Materials for Skeletal-Muscle-
[146] G. Lacraz, A.J. Rouleau, V. Couture, T. Söllrald, G. Drouin, N. Veillette, Tissue Engineering, Adv. Mater. (2016), doi:10.10 02/adma.20160 0240.
M. Grandbois, G. Grenier, Increased stiffness in aged skeletal muscle impairs [172] M.F. Leong, J.K.C. Toh, C. Du, K. Narayanan, H.F. Lu, T.C. Lim, A.C.A. Wan,
muscle progenitor cell proliferative activity, PLoS ONE (2015), doi:10.1371/ J.Y. Ying, Patterned prevascularised tissue constructs by assembly of polyelec-
journal.pone.0136217. trolyte hydrogel fibres, Nat. Commun. (2013), doi:10.1038/ncomms3353.
[147] L.-.S. Wang, J.E. Chung, P.P.-Y. Chan, M. Kurisawa, Injectable biodegrad- [173] N.F. Huang, S. Patel, R.G. Thakar, J. Wu, B.S. Hsiao, B. Chu, R.J. Lee, S. Li,
able hydrogels with tunable mechanical properties for the stimulation of

17
JID: ACTBIO
ARTICLE IN PRESS [m5G;February 21, 2021;17:50]

S. Ng and M. Kurisawa Acta Biomaterialia xxx (xxxx) xxx

Myotube assembly on nanofibrous and micropatterned polymers, Nano Lett. [198] K. Comley, N.A. Fleck, A micromechanical model for the Young’s modulus of
(2006), doi:10.1021/nl060060o. adipose tissue, Int. J. Solids Struct. 47 (2010) 2982–2990, doi:10.1016/j.ijsolstr.
[174] J.S. Choi, S.J. Lee, G.J. Christ, A. Atala, J.J. Yoo, The influence of electro- 2010.07.001.
spun aligned poly(ε -caprolactone)/collagen nanofiber meshes on the forma- [199] A. Samani, J. Zubovits, D. Plewes, Elastic moduli of normal and patho-
tion of self-aligned skeletal muscle myotubes, Biomaterials (2008), doi:10. logical human breast tissues: an inversion-technique-based investigation
1016/j.biomaterials.2008.03.031. of 169 samples, Phys. Med. Biol. (2007), doi:10.1088/0031-9155/52/6/
[175] K.J. Aviss, J.E. Gough, S. Downes, Aligned electrospun polymer fibres for skele- 002.
tal muscle regeneration, Eur. Cells Mater. (2010), doi:10.22203/eCM.v019a19. [200] E.M. Chandler, C.M. Berglund, J.S. Lee, W.J. Polacheck, J.P. Gleghorn, B.J. Kirby,
[176] M.T. Lam, Y.C. Huang, R.K. Birla, S. Takayama, Microfeature guided skeletal C. Fischbach, Stiffness of photocrosslinked RGD-alginate gels regulates adi-
muscle tissue engineering for highly organized 3-dimensional free-standing pose progenitor cell behavior, Biotechnol. Bioeng. 108 (2011) 1683–1692,
constructs, Biomaterials 30 (2009) 1150–1155, doi:10.1016/j.biomaterials. doi:10.1002/bit.23079.
2008.11.014. [201] M.J. Post, Cultured beef: medical technology to produce food, J. Sci. Food
[177] S. Chen, T. Nakamoto, N. Kawazoe, G. Chen, Engineering multi-layered skele- Agric. (2014), doi:10.1002/jsfa.6474.
tal muscle tissue by using 3D microgrooved collagen scaffolds, Biomaterials [202] U. Cheema, S.Y. Yang, V. Mudera, G.G. Goldspink, R.A. Brown, 3-D in vitro
(2015), doi:10.1016/j.biomaterials.2015.09.010. model of early skeletal muscle development, Cell Motil. Cytoskeleton. (2003),
[178] J.M. Dugan, R.F. Collins, J.E. Gough, S.J. Eichhorn, Oriented surfaces of ad- doi:10.1002/cm.10095.
sorbed cellulose nanowhiskers promote skeletal muscle myogenesis, Acta Bio- [203] A.S.T. Smith, S. Passey, L. Greensmith, V. Mudera, M.P. Lewis, Characterization
mater. (2013), doi:10.1016/j.actbio.2012.08.050. and optimization of a simple, repeatable system for the long term in vitro
[179] H. Takahashi, T. Shimizu, M. Nakayama, M. Yamato, T. Okano, The use of culture of aligned myotubes in 3D, J. Cell. Biochem. (2012), doi:10.1002/jcb.
anisotropic cell sheets to control orientation during the self-organization of 23437.
3D muscle tissue, Biomaterials (2013), doi:10.1016/j.biomaterials.2013.06.033. [204] H. Vandenburgh, J. Shansky, M. Del Tatto, J. Chromiak, Organogenesis of
[180] V. Hosseini, S. Ahadian, S. Ostrovidov, G. Camci-Unal, S. Chen, H. Kaji, M. Ra- Skeletal Muscle in Tissue Culture, Tissue Eng. Methods Protoc. (1999) 217–
malingam, A. Khademhosseini, Engineered contractile skeletal muscle tissue 225, doi:10.1385/0- 89603- 516- 6:217.
on a microgrooved methacrylated gelatin substrate, Tissue Eng. - Part A. [205] L. Thorrez, J. Shansky, L. Wang, L. Fast, T. VandenDriessche, M. Chuah,
(2012), doi:10.1089/ten.tea.2012.0181. D. Mooney, H. Vandenburgh, Growth, differentiation, transplantation and sur-
[181] H.S. Yang, N. Ieronimakis, J.H. Tsui, H.N. Kim, K.Y. Suh, M. Reyes, D.H. Kim, vival of human skeletal myofibers on biodegradable scaffolds, Biomaterials
Nanopatterned muscle cell patches for enhanced myogenesis and dystrophin (2008), doi:10.1016/j.biomaterials.2007.09.014.
expression in a mouse model of muscular dystrophy, Biomaterials (2014), [206] M. Post, C. van der Weele, Principles of Tissue Engineering for Food, Princ.
doi:10.1016/j.biomaterials.2013.10.067. Tissue Eng, 4th Ed., 2013, doi:10.1016/B978- 0- 12- 398358- 9.0 0 078-1.
[182] S. Jana, A. Cooper, M. Zhang, Chitosan scaffolds with unidirectional micro- [207] H. Vandenburgh, S. Kaufman, In vitro model for stretch-induced hypertrophy
tubular pores for large skeletal myotube generation, Adv. Healthc. Mater. of skeletal muscle, Science (80-.) (1979), doi:10.1126/science.569901.
(2013), doi:10.10 02/adhm.20120 0177. [208] C.A. Powell, B.L. Smiley, J. Mills, H.H. Vandenburgh, Mechanical stimulation
[183] V. Kroehne, I. Heschel, F. Schügner, D. Lasrich, J.W. Bartsch, H. Jockusch, improves tissue-engineered human skeletal muscle, Am. J. Physiol. - Cell
Use of a novel collagen matrix with oriented pore structure for muscle Physiol. (2002), doi:10.1152/ajpcell.00595.2001.
cell differentiation in cell culture and in grafts, J. Cell. Mol. Med. (2008), [209] D.G. Moon, G. Christ, J.D. Stitzel, A. Atala, J.J. Yoo, Cyclic mechanical precondi-
doi:10.1111/j.1582-4934.20 08.0 0238.x. tioning improves engineered muscle contraction, Tissue Eng. - Part A. (2008),
[184] Y.S. Choi, L.G. Vincent, A.R. Lee, K.C. Kretchmer, S. Chirasatitsin, M.K. Dobke, doi:10.1089/tea.2007.0104.
A.J. Engler, The alignment and fusion assembly of adipose-derived stem [210] S. Rangarajan, L. Madden, N. Bursac, Use of flow, electrical, and mechanical
cells on mechanically patterned matrices, Biomaterials (2012), doi:10.1016/j. stimulation to promote engineering of striated muscles, Ann. Biomed. Eng.
biomaterials.2012.06.057. (2014), doi:10.1007/s10439- 013- 0966- 4.
[185] Y.-.W. Cheng, D.J. Shiwarski, R.L. Ball, K.A. Whitehead, A.W. Feinberg, [211] D.C. Turner, A.M. Kasper, R.A. Seaborne, A.D. Brown, G.L. Close, M. Murphy,
Engineering Aligned Skeletal Muscle Tissue Using Decellularized Plant- C.E. Stewart, N.R.W. Martin, A.P. Sharples, Exercising bioengineered skeletal
Derived Scaffolds, ACS Biomater. Sci. Eng. 6 (2020) 3046–3054, doi:10.1021/ muscle in vitro: biopsy to bioreactor, Methods Mol. Biol. (2019), doi:10.1007/
acsbiomaterials.0c0 0 058. 978- 1- 4939- 8897- 6_5.
[186] T. Matsumoto, J.I. Sasaki, E. Alsberg, H. Egusa, H. Yatani, T. Sohmura, Three- [212] P. Juffer, A.D. Bakker, J. Klein-Nulend, R.T. Jaspers, Mechanical Loading by
dimensional cell and tissue patterning in a strained fibrin gel system, PLoS Fluid Shear Stress of Myotube Glycocalyx Stimulates Growth Factor Expres-
ONE (2007), doi:10.1371/journal.pone.0001211. sion and Nitric Oxide Production, Cell Biochem. Biophys. (2014), doi:10.1007/
[187] H.H. Vandenburgh, P. Karlisch, Longitudinal growth of skeletal myotubes in s12013-013- 9812- 4.
vitro in a new horizontal mechanical cell stimulator, Vitr. Cell. Dev. Biol. [213] S. Naskar, V. Kumaran, B. Basu, On the Origin of Shear Stress Induced Myo-
(1989), doi:10.1007/BF02623630. genesis Using PMMA Based Lab-on-Chip, ACS Biomater. Sci. Eng. (2017),
[188] T. Okano, T. Matsuda, Tissue engineered skeletal muscle: preparation of doi:10.1021/acsbiomaterials.7b00206.
highly dense, highly oriented hybrid muscular tissues, Cell Transplant. (1998), [214] A. Ramey-Ward, H. Su, K. Salaita, Mechanical Stimulation of Adhesion Re-
doi:10.1016/S0963-6897(97)0 0 067-5. ceptors Using Light-Responsive Nanoparticle Actuators Enhances Myogenesis,
[189] P. Yilgor Huri, C.A. Cook, D.L. Hutton, B.C. Goh, J.M. Gimble, D.J. DiGiro- ACS Appl. Mater. Interfaces. (2020), doi:10.1021/acsami.0c08871.
lamo, W.L. Grayson, Biophysical cues enhance myogenesis of human adi- [215] C.A. Cezar, E.T. Roche, H.H. Vandenburgh, G.N. Duda, C.J. Walsh, D.J. Mooney,
pose derived stem/stromal cells, Biochem. Biophys. Res. Commun. (2013), Biologic-free mechanically induced muscle regeneration, Proc. Natl. Acad. Sci.
doi:10.1016/j.bbrc.2013.07.049. U. S. A. (2016), doi:10.1073/pnas.1517517113.
[190] H. Egusa, M. Kobayashi, T. Matsumoto, J.I. Sasaki, S. Uraguchi, H. Yatani, Ap- [216] M.L.P. Langelaan, K.J.M. Boonen, K.Y. Rosaria-Chak, D.W.J. van der Schaft,
plication of cyclic strain for accelerated skeletal myogenic differentiation of M.J. Post, F.P.T. Baaijens, Advanced maturation by electrical stimulation: dif-
mouse bone marrow-derived mesenchymal stromal cells with cell alignment, ferences in response between C2C12 and primary muscle progenitor cells, J.
Tissue Eng. - Part A. (2013), doi:10.1089/ten.tea.2012.0164. Tissue Eng. Regen. Med. (2011), doi:10.1002/term.345.
[191] M. Levy-Mishali, J. Zoldan, S. Levenberg, Effect of scaffold stiffness on my- [217] I.-.C. Liao, J.B. Liu, N. Bursac, K.W. Leong, Effect of Electromechanical Stimula-
oblast differentiation, Tissue Eng. - Part A. (2009), doi:10.1089/ten.tea.2008. tion on the Maturation of Myotubes on Aligned Electrospun Fibers, Cell. Mol.
0111. Bioeng. (2008), doi:10.1007/s12195- 008- 0021- y.
[192] A.J. Engler, S. Sen, H.L. Sweeney, D.E. Discher, Matrix Elasticity Directs Stem [218] S. Ahadian, J. Ramón-Azcón, S. Ostrovidov, G. Camci-Unal, V. Hosseini, H. Kaji,
Cell Lineage Specification, Cell. (2006), doi:10.1016/j.cell.2006.06.044. K. Ino, H. Shiku, A. Khademhosseini, T. Matsue, Interdigitated array of Pt elec-
[193] K. Ren, T. Crouzier, C. Roy, C. Picart, Polyelectrolyte multilayer films of con- trodes for electrical stimulation and engineering of aligned muscle tissue, Lab
trolled stiffness modulate myoblast cell differentiation, Adv. Funct. Mater. Chip. (2012), doi:10.1039/c2lc40479f.
(2008), doi:10.1002/adfm.200701297. [219] H. Kaji, T. Ishibashi, K. Nagamine, M. Kanzaki, M. Nishizawa, Electrically in-
[194] T. Zhang, S. Lin, X. Shao, S. Shi, Q. Zhang, C. Xue, Y. Lin, B. Zhu, X. Cai, Reg- duced contraction of C2C12 myotubes cultured on a porous membrane-based
ulating osteogenesis and adipogenesis in adipose-derived stem cells by con- substrate with muscle tissue-like stiffness, Biomaterials (2010), doi:10.1016/j.
trolling underlying substrate stiffness, J. Cell. Physiol. (2018), doi:10.1002/jcp. biomaterials.2010.05.071.
26193. [220] H. Park, R. Bhalla, R. Saigal, M. Radisic, N. Watson, R. Langer, G. Vunjak-
[195] J. Oliver-De La Cruz, G. Nardone, J. Vrbsky, A. Pompeiano, A.R. Pere- Novakovic, Effects of electrical stimulation in C2C12 muscle constructs, J. Tis-
strelo, F. Capradossi, K. Melajová, P. Filipensky, G. Forte, Substrate mechan- sue Eng. Regen. Med. (2008), doi:10.1002/term.93.
ics controls adipogenesis through YAP phosphorylation by dictating cell [221] E. Serena, M. Flaibani, S. Carnio, L. Boldrin, L. Vitiello, P. De Coppi, N. El-
spreading, Biomaterials 205 (2019) 64–80, doi:10.1016/j.biomaterials.2019.03. vassore, Electrophysiologic stimulation improves myogenic potential of mus-
009. cle precursor cells grown in a 3D collagen scaffold, Neurol. Res. (2008),
[196] J. Xie, D. Zhang, C. Zhou, Q. Yuan, L. Ye, X. Zhou, Substrate elasticity reg- doi:10.1179/174313208X281109.
ulates adipose-derived stromal cell differentiation towards osteogenesis and [222] A. Khodabukus, L. Madden, N.K. Prabhu, T.R. Koves, C.P. Jackman, D.M. Muoio,
adipogenesis through β -catenin transduction, Acta Biomater. 79 (2018) 83– N. Bursac, Electrical stimulation increases hypertrophy and metabolic flux in
95, doi:10.1016/j.actbio.2018.08.018. tissue-engineered human skeletal muscle, Biomaterials (2019), doi:10.1016/j.
[197] D.A. Young, Y.S. Choi, A.J. Engler, K.L. Christman, Stimulation of adipoge- biomaterials.2018.08.058.
nesis of adult adipose-derived stem cells using substrates that mimic the [223] K. Donnelly, A. Khodabukus, A. Philp, L. Deldicque, R.G. Dennis, K. Baar, A
stiffness of adipose tissue, Biomaterials 34 (2013) 8581–8588, doi:10.1016/j. Novel bioreactor for stimulating skeletal muscle in vitro, Tissue Eng. - Part C
biomaterials.2013.07.103. Methods. (2010), doi:10.1089/ten.tec.2009.0125.

18
JID: ACTBIO
ARTICLE IN PRESS [m5G;February 21, 2021;17:50]

S. Ng and M. Kurisawa Acta Biomaterialia xxx (xxxx) xxx

[224] R. Dong, P.X. Ma, B. Guo, Conductive biomaterials for muscle tissue engineer- detachment of hMSCs from microcarriers in multiple bioreactor platforms,
ing, Biomaterials (2020), doi:10.1016/j.biomaterials.2019.119584. Biochem. Eng. J. (2016), doi:10.1016/j.bej.2015.08.003.
[225] I. Jun, S. Jeong, H. Shin, The stimulation of myoblast differentiation by [252] M. Sponchioni, U. Capasso Palmiero, D. Moscatelli, Thermo-responsive poly-
electrically conductive sub-micron fibers, Biomaterials (2009), doi:10.1016/j. mers: applications of smart materials in drug delivery and tissue engineering,
biomaterials.2008.12.063. Mater. Sci. Eng. C. (2019), doi:10.1016/j.msec.2019.04.069.
[226] K.J. Gilmore, M. Kita, Y. Han, A. Gelmi, M.J. Higgins, S.E. Moulton, G.M. Clark, [253] B. Chen, J. Dang, T.L. Tan, N. Fang, W.N. Chen, K.W. Leong, V. Chan, Dynam-
R. Kapsa, G.G. Wallace, Skeletal muscle cell proliferation and differentiation ics of smooth muscle cell deadhesion from thermosensitive hydroxybutyl chi-
on polypyrrole substrates doped with extracellular matrix components, Bio- tosan, Biomaterials (2007), doi:10.1016/j.biomaterials.2006.11.027.
materials (2009), doi:10.1016/j.biomaterials.2009.06.059. [254] A. Tamura, M. Nishi, J. Kobayashi, K. Nagase, H. Yajima, M. Yamato, T. Okano,
[227] R.D. Breukers, K.J. Gilmore, M. Kita, K.K. Wagner, M.J. Higgins, S.E. Moulton, Simultaneous enhancement of cell proliferation and thermally induced har-
G.M. Clark, D.L. Officer, R.M.I. Kapsa, G.G. Wallace, Creating conductive struc- vest efficiency based on temperature-responsive cationic copolymer-grafted
tures for cell growth: growth and alignment of myogenic cell types on poly- microcarriers, Biomacromolecules (2012), doi:10.1021/bm300256e.
thiophenes, J. Biomed. Mater. Res. - Part A. (2010), doi:10.1002/jbm.a.32822. [255] C.H. Chen, C.C. Tsai, W. Chen, F.L. Mi, H.F. Liang, S.C. Chen, H.W. Sung, Novel
[228] S. Ribeiro, A.C. Gomes, I. Etxebarria, S. Lanceros-Méndez, C. Ribeiro, Electroac- living cell sheet harvest system composed of thermoreversible methylcellu-
tive biomaterial surface engineering effects on muscle cells differentiation, lose hydrogels, Biomacromolecules (2006), doi:10.1021/bm0506400.
Mater. Sci. Eng. C. (2018), doi:10.1016/j.msec.2018.07.044. [256] A.K.A. Silva, C. Richard, G. Ducouret, M. Bessodes, D. Scherman, O.W. Merten,
[229] M.C. Chen, Y.C. Sun, Y.H. Chen, Electrically conductive nanofibers with highly Xyloglucan-derivatized films for the culture of adherent cells and their ther-
oriented structures and their potential application in skeletal muscle tissue mocontrolled detachment: a promising alternative to cells sensitive to pro-
engineering, Acta Biomater (2013), doi:10.1016/j.actbio.2012.10.024. tease treatment, Biomacromolecules (2013), doi:10.1021/bm3017737.
[230] S.H. Ku, S.H. Lee, C.B. Park, Synergic effects of nanofiber alignment and [257] D. Billig, J.M. Clark, A.J. Ewell, C.M. Carter, C. Gebb, The separation of har-
electroactivity on myoblast differentiation, Biomaterials (2012), doi:10.1016/ vested cells from microcarriers: a comparison of methods, Dev. Biol. Stand.
j.biomaterials.2012.05.018. (1983).
[231] L.S. Moreira Teixeira, J. Feijen, C.A. van Blitterswijk, P.J. Dijkstra, M. Karperien, [258] R. Moloudi, S. Oh, C. Yang, K.L. Teo, A.T.L. Lam, M.E. Warkiani,
Enzyme-catalyzed crosslinkable hydrogels: emerging strategies for tissue en- M.W. Naing, Inertial-Based Filtration Method for Removal of Micro-
gineering, Biomaterials (2012), doi:10.1016/j.biomaterials.2011.10.067. carriers from Mesenchymal Stem Cell Suspensions, Sci. Rep. (2018),
[232] P. Bajaj, R.M. Schweller, A. Khademhosseini, J.L. West, R. Bashir, 3D biofabri- doi:10.1038/s41598- 018- 31019- y.
cation strategies for tissue engineering and regenerative medicine, Annu. Rev. [259] B. Cunha, C. Peixoto, M.M. Silva, M.J.T. Carrondo, M. Serra, P.M. Alves, Fil-
Biomed. Eng. (2014), doi:10.1146/annurev- bioeng- 071813- 105155. tration methodologies for the clarification and concentration of human mes-
[233] R.J. Wade, J.A. Burdick, Engineering ECM signals into biomaterials, Mater. To- enchymal stem cells, J. Memb. Sci. (2015), doi:10.1016/j.memsci.2014.12.041.
day. (2012), doi:10.1016/S1369-7021(12)70197-9. [260] C.Y. Lin, C.H. Huang, Y.K. Wu, N.C. Cheng, J. Yu, Maintenance of human adi-
[234] O. Chaudhuri, Viscoelastic hydrogels for 3D cell culture, Biomater. Sci. (2017), pose derived stem cell (hASC) differentiation capabilities using a 3D culture,
doi:10.1039/c7bm00261k. Biotechnol. Lett. (2014), doi:10.1007/s10529- 014- 1500- y.
[235] M. Geerligs, G.W.M. Peters, P.A.J. Ackermans, C.W.J. Oomens, F.P.T. Baai- [261] H.A. Grabitske, J.L. Slavin, Low-Digestible Carbohydrates in Practice, J. Am.
jens, Linear viscoelastic behavior of subcutaneous adipose tissue, Biorheology Diet. Assoc. (2008), doi:10.1016/j.jada.2008.07.010.
(20 08), doi:10.3233/BIR-20 08-0517. [262] K.I. Hilgendorf, C.T. Johnson, A. Mezger, S.L. Rice, A.M. Norris, J. Demeter,
[236] D.D. McKinnon, D.W. Domaille, J.N. Cha, K.S. Anseth, Biophysically defined W.J. Greenleaf, J.F. Reiter, D. Kopinke, P.K. Jackson, Omega-3 Fatty Acids Ac-
and cytocompatible covalently adaptable networks as viscoelastic 3d cell cul- tivate Ciliary FFAR4 to Control Adipogenesis, Cell 179 (2019) 1289–1305 e21,
ture systems, Adv. Mater. (2014), doi:10.1002/adma.201303680. doi:10.1016/j.cell.2019.11.005.
[237] A. Bauer, L. Gu, B. Kwee, W.A. Li, M. Dellacherie, A.D. Celiz, D.J. Mooney, Hy- [263] L. Specht, An Analysis of Culture Medium Costs and Production Volumes For
drogel substrate stress-relaxation regulates the spreading and proliferation of Cultivated Meat, Good Food Inst., Washington, DC, USA, 2020.
mouse myoblasts, Acta Biomater. (2017), doi:10.1016/j.actbio.2017.08.041. [264] A.W. Xie, W.L. Murphy, Engineered biomaterials to mitigate growth fac-
[238] C. Fan, D.A. Wang, Macroporous Hydrogel Scaffolds for Three-Dimensional tor cost in cell biomanufacturing, Curr. Opin. Biomed. Eng. 10 (2019) 1–10,
Cell Culture and Tissue Engineering, Tissue Eng. - Part B Rev. (2017), doi:10. doi:10.1016/j.cobme.2018.12.004.
1089/ten.teb.2016.0465. [265] C. Borselli, H. Storrie, F. Benesch-Lee, D. Shvartsman, C. Cezar, J.W. Lichtman,
[239] A.R. Kang, J.S. Park, J. Ju, G.S. Jeong, S.H. Lee, Cell encapsulation via microtech- H.H. Vandenburgh, D.J. Mooney, Functional muscle regeneration with com-
nologies, Biomaterials (2014), doi:10.1016/j.biomaterials.2013.12.073. bined delivery of angiogenesis and myogenesis factors, Proc. Natl. Acad. Sci.
[240] H.P. Lee, L. Gu, D.J. Mooney, M.E. Levenston, O. Chaudhuri, Mechanical con- U. S. A. (2010), doi:10.1073/pnas.0903875106.
finement regulates cartilage matrix formation by chondrocytes, Nat. Mater. [266] M.J. Worrallo, R.L.L. Moore, K.E. Glen, R.J. Thomas, Immobilized hematopoietic
(2017), doi:10.1038/nmat4993. growth factors onto magnetic particles offer a scalable strategy for cell ther-
[241] O. Chaudhuri, L. Gu, D. Klumpers, M. Darnell, S.A. Bencherif, J.C. Weaver, apy manufacturing in suspension cultures, Biotechnol. J. (2017), doi:10.1002/
N. Huebsch, H.P. Lee, E. Lippens, G.N. Duda, D.J. Mooney, Hydrogels with tun- biot.201600493.
able stress relaxation regulate stem cell fate and activity, Nat. Mater. (2016), [267] S. Rinker, Y. Ke, Y. Liu, R. Chhabra, H. Yan, Self-assembled DNA nanostructures
doi:10.1038/nmat4489. for distance-dependent multivalent ligand-protein binding, Nat. Nanotechnol.
[242] R. Myhan, M. Markowski, T. Daszkiewicz, P. Zapotoczny, P. Sadowski, Non- (2008), doi:10.1038/nnano.2008.164.
linear stress relaxation model as a tool for evaluating the viscoelastic proper- [268] D.G. Belair, N.N. Le, W.L. Murphy, Design of growth factor sequestering bio-
ties of meat products, J. Food Eng. (2015), doi:10.1016/j.jfoodeng.2014.09.006. materials, Chem. Commun. (2014), doi:10.1039/c4cc04317k.
[243] M.M. Fitzgerald, K. Bootsma, J.A. Berberich, J.L. Sparks, Tunable stress re- [269] S. Anastasi, S. Giordano, O. Sthandier, G. Gambarotta, R. Maione, P. Comoglio,
laxation behavior of an alginate-polyacrylamide hydrogel: comparison with P. Amati, A natural hepatocyte growth factor/scatter factor autocrine loop in
muscle tissue, Biomacromolecules (2015), doi:10.1021/bm501845j. myoblast cells and the effect of the constitutive met kinase activation on
[244] J. Carballo, J. Ayo, F.J. Colmenero, Microbial transglutaminase and caseinate as myogenic differentiation, J. Cell Biol. (1997), doi:10.1083/jcb.137.5.1057.
cold set binders: influence of meat species and chilling storage, LWT - Food [270] G.R. Adams, Invited Review: autocrine/paracrine IGF-I and skeletal muscle
Sci. Technol. (2006), doi:10.1016/j.lwt.2005.03.020. adaptation, J. Appl. Physiol. (2002), doi:10.1152/japplphysiol.01264.2001.
[245] C.B.B. Cazarin, G.C. Lima, J.K. da Silva, M.R. Maróstica Jr, Enzymes in Meat [271] M. Hemmingsen, S. Vedel, P. Skafte-Pedersen, D. Sabourin, P. Collas, H. Bruus,
Processing, in: M. Chandrasekaran (Ed.), Enzym. Food Beverage Process, 2015, M. Dufva, The Role of Paracrine and Autocrine Signaling in the Early Phase of
p. 337, doi:10.21048/ijnd.2019.56.2.23505. Adipogenic Differentiation of Adipose-derived Stem Cells, PLoS ONE (2013),
[246] R. Lantto, K. Kruus, E. Puolanne, K. Honkapaa, K. Roininen, J. Buchert, En- doi:10.1371/journal.pone.0063638.
zymes in Meat Processing, in: R.J. Whitehurst, M. van Oort (Eds.), Enzym. [272] M.R.I. Shishir, L. Xie, C. Sun, X. Zheng, W. Chen, Advances in micro and
Food Technol, 2009, p. 264, doi:10.1002/9781444309935. Second Ed.. nano-encapsulation of bioactive compounds using biopolymer and lipid-
[247] S. Rourou, N. Riahi, S. Majoul, K. Trabelsi, H. Kallel, Development of an in based transporters, Trends Food Sci. Technol. 78 (2018) 34–60, doi:10.1016/
situ detachment protocol of Vero cells grown on Cytodex1 microcarriers un- j.tifs.2018.05.018.
der animal component-free conditions in stirred bioreactor, Appl. Biochem. [273] B. Ghorani, N. Tucker, Fundamentals of Electrospinning As a Novel Delivery
Biotechnol. (2013), doi:10.1007/s12010- 013- 0307- y. Vehicle For Bioactive Compounds in Food Nanotechnology, Food Hydrocoll.,
[248] S.R. Caruso, M.D. Orellana, A. Mizukami, T.R. Fernandes, A.M. Fontes, 2015, doi:10.1016/j.foodhyd.2015.05.024.
C.A.T. Suazo, V.C. Oliveira, D.T. Covas, K. Swiech, Growth and functional har- [274] G. Liu, X. Wang, X. Sun, C. Deng, A. Atala, Y. Zhang, The effect of urine-derived
vesting of human mesenchymal stromal cells cultured on a microcarrier- stem cells expressing VEGF loaded in collagen hydrogels on myogenesis and
based system, Biotechnol. Prog. (2014), doi:10.1002/btpr.1886. innervation following after subcutaneous implantation in nude mice, Bioma-
[249] Q.A. Rafiq, K. Coopman, A.W. Nienow, C.J. Hewitt, Systematic microcar- terials (2013), doi:10.1016/j.biomaterials.2013.07.077.
rier screening and agitated culture conditions improves human mesenchy- [275] J. Zou, W. Wang, A.T. Neffe, X. Xu, Z. Li, Z. Deng, X. Sun, N. Ma, A. Lendlein,
mal stem cell yield in bioreactors, Biotechnol. J. (2016), doi:10.1002/biot. Adipogenic differentiation of human adipose derived mesenchymal stem cells
201400862. in 3D architectured gelatin based hydrogels (ArcGel), Clin. Hemorheol. Micro-
[250] Q.A. Rafiq, M.P. Hanga, T.R.J. Heathman, K. Coopman, A.W. Nienow, circ. (2017), doi:10.3233/CH-179210.
D.J. Williams, C.J. Hewitt, Process development of human multipotent stromal [276] C.A. Rossi, M. Flaibani, B. Blaauw, M. Pozzobon, E. Figallo, C. Reggiani, L. Vi-
cell microcarrier culture using an automated high-throughput microbioreac- tiello, N. Elvassore, P. De Coppi, In vivo tissue engineering of functional skele-
tor, Biotechnol. Bioeng. (2017), doi:10.1002/bit.26359. tal muscle by freshly isolated satellite cells embedded in a photopolymeriz-
[251] A.W. Nienow, C.J. Hewitt, T.R.J. Heathman, V.A.M. Glyn, G.N. Fonte, able hydrogel, FASEB J (2011), doi:10.1096/fj.10-174755.
M.P. Hanga, K. Coopman, Q.A. Rafiq, Agitation conditions for the culture and [277] W. Wang, M. Fan, L. Zhang, S.H. Liu, L. Sun, C.Y. Wang, Compatibility of

19
JID: ACTBIO
ARTICLE IN PRESS [m5G;February 21, 2021;17:50]

S. Ng and M. Kurisawa Acta Biomaterialia xxx (xxxx) xxx

hyaluronic acid hydrogel and skeletal muscle myoblasts, Biomed. Mater. [302] G. Blüml, Microcarrier Cell Culture Principles and Methods, in: Handbooks
(2009), doi:10.1088/1748-6041/4/2/025011. from GE Healthc, 2021, doi:10.1007/978- 1- 59745- 399- 8_5.
[278] F.S. Kamelger, R. Marksteiner, E. Margreiter, G. Klima, G. Wechselberger, [303] W. Li, Y. Han, H. Yang, G. Wang, R. Lan, J.Y. Wang, Preparation of microcarriers
S. Hering, H. Piza, A comparative study of three different biomaterials in the based on zein and their application in cell culture, Mater. Sci. Eng. C. (2016),
engineering of skeletal muscle using a rat animal model, Biomaterials (2004), doi:10.1016/j.msec.2015.09.045.
doi:10.1016/S0142- 9612(03)00520- 9. [304] C. Wang, Y. Gong, Y. Lin, J. Shen, D.A. Wang, A novel gellan gel-based micro-
[279] M. Halbleib, T. Skurk, C. De Luca, D. Von Heimburg, H. Hauner, Tissue engi- carrier for anchorage-dependent cell delivery, Acta Biomater. (2008), doi:10.
neering of white adipose tissue using hyaluronic acid-based scaffolds. I: in 1016/j.actbio.20 08.03.0 08.
vitro differentiation of human adipocyte precursor cells on scaffolds, Bioma- [305] B. Sarker, A.R. Boccaccini, Alginate Utilization in Tissue Engineering and Cell
terials 24 (2003) 3125–3132, doi:10.1016/S0142-9612(03)00156-X. Therapy, in: B.H.A. Rehm, M.F. Moradali (Eds.), Alginates Their Biomed, Appl.,
[280] L. Flynn, G.D. Prestwich, J.L. Semple, K.A. Woodhouse, Adipose tissue engi- 2018, pp. 121–155, doi:10.1007/978- 981- 10- 6910- 9_5.
neering with naturally derived scaffolds and adipose-derived stem cells, Bio- [306] A.B. Bello, D. Kim, D. Kim, H. Park, S.H. Lee, Engineering and functionalization
materials 28 (2007) 3834–3842, doi:10.1016/j.biomaterials.20 07.05.0 02. of gelatin biomaterials: from cell culture to medical applications, Tissue Eng.
[281] J.P. Beier, J. Stern-Straeter, V.T. Foerster, U. Kneser, G.B. Stark, A.D. Bach, Tissue - Part B Rev. (2020), doi:10.1089/ten.teb.2019.0256.
engineering of injectable muscle: three-dimensional myoblast-fibrin injection [307] E.G. Popa, M.E. Gomes, R.L. Reis, Cell delivery systems using alginate-
in the syngeneic rat animal model, Plast. Reconstr. Surg. (2006), doi:10.1097/ carrageenan hydrogel beads and fibers for regenerative medicine applications,
01.prs.0 0 0 02210 07.97115.1d. Biomacromolecules (2011), doi:10.1021/bm200965x.
[282] G.H. Borschel, D.E. Dow, R.G. Dennis, D.L. Brown, Tissue-engineered axially [308] S.M. Mihaila, A.K. Gaharwar, R.L. Reis, A.P. Marques, M.E. Gomes,
vascularized contractile skeletal muscle, Plast. Reconstr. Surg. (2006), doi:10. A. Khademhosseini, Photocrosslinkable kappa-carrageenan hydrogels for
1097/01.prs.0 0 0 0224295.54073.49. tissue engineering applications, Adv. Healthc. Mater. 2 (2013) 895–907,
[283] J. Liu, H.H.K. Xu, H. Zhou, M.D. Weir, Q. Chen, C.A. Trotman, Human umbilical doi:10.10 02/adhm.20120 0317.
cord stem cell encapsulation in novel macroporous and injectable fibrin for [309] J.V. Araujo, N. Davidenko, M. Danner, R.E. Cameron, S.M. Best, Novel porous
muscle tissue engineering, Acta Biomater (2013), doi:10.1016/j.actbio.2012.08. scaffolds of pH responsive chitosan/carrageenan-based polyelectrolyte com-
009. plexes for tissue engineering, J. Biomed. Mater. Res. - Part A. (2014), doi:10.
[284] F. Mehta, R. Theunissen, M.J. Post, Adipogenesis from bovine precursors, 1002/jbm.a.35128.
Methods Mol. Biol. (2019), doi:10.1007/978- 1- 4939- 8897- 6_8. [310] L. Tytgat, M. Vagenende, H. Declercq, J.C. Martins, H. Thienpont, H. Ottevaere,
[285] V. Guneta, Q.L. Loh, C. Choong, Cell-secreted extracellular matrix formation P. Dubruel, S. Van Vlierberghe, Synergistic effect of κ -carrageenan and gelatin
and differentiation of adipose-derived stem cells in 3D alginate scaffolds with blends towards adipose tissue engineering, Carbohydr. Polym. 189 (2018) 1–9,
tunable properties, J. Biomed. Mater. Res. - Part A. 104 (2016) 1090–1101, doi:10.1016/j.carbpol.2018.02.002.
doi:10.1002/jbm.a.35644. [311] R. Yegappan, V. Selvaprithiviraj, S. Amirthalingam, R. Jayakumar, Carrageenan
[286] S.W. Kang, B.H. Cha, H. Park, K.S. Park, K.Y. Lee, S.H. Lee, The Effect of Con- based hydrogels for drug delivery, tissue engineering and wound healing, Car-
jugating RGD into 3D Alginate Hydrogels on Adipogenic Differentiation of bohydr. Polym. 198 (2018) 385–400, doi:10.1016/j.carbpol.2018.06.086.
Human Adipose-Derived Stromal Cells, Macromol. Biosci. 11 (2011) 673–679, [312] R. Yegappan, V. Selvaprithiviraj, S. Amirthalingam, A. Mohandas, N.S. Hwang,
doi:10.1002/mabi.201000479. R. Jayakumar, Injectable angiogenic and osteogenic carrageenan nanocompos-
[287] W.S. Kim, D.J. Mooney, P.R. Arany, K. Lee, N. Huebsch, J. Kim, Adipose tissue ite hydrogel for bone tissue engineering, Int. J. Biol. Macromol. 122 (2019)
engineering using injectable, oxidized alginate hydrogels, Tissue Eng. - Part 320–328, doi:10.1016/j.ijbiomac.2018.10.182.
A. (2012), doi:10.1089/ten.tea.2011.0250. [313] S.M. Mihaila, E.G. Popa, R.L. Reis, A.P. Marques, M.E. Gomes, Fabrication of
[288] E. Zakhem, S. Raghavan, R.R. Gilmont, K.N. Bitar, Chitosan-based scaffolds for endothelial cell-laden carrageenan microfibers for microvascularized bone
the support of smooth muscle constructs in intestinal tissue engineering, Bio- tissue engineering applications, Biomacromolecules 15 (2014) 2849–2860,
materials (2012), doi:10.1016/j.biomaterials.2012.03.051. doi:10.1021/bm50 0 036a.
[289] M.H. Kim, H.N. Hong, J.P. Hong, C.J. Park, S.W. Kwon, S.H. Kim, G. Kang, [314] E.G. Popa, S.G. Caridade, J.F. Mano, R.L. Reis, M.E. Gomes, Chondrogenic po-
M.J. Kim, The effect of VEGF on the myogenic differentiation of adipose tis- tential of injectableκ -carrageenan hydrogel with encapsulated adipose stem
sue derived stem cells within thermosensitive hydrogel matrices, Biomateri- cells for cartilage tissue-engineering applications, J. Tissue Eng. Regen. Med.
als (2010), doi:10.1016/j.biomaterials.2009.10.057. (2015), doi:10.1002/term.1683.
[290] J.H. Elisseeff, A.T. Hillel, S. Unterman, Z. Nahas, B. Reid, J.M. Coburn, J. Axel- [315] P.M. Rocha, V.E. Santo, M.E. Gomes, R.L. Reis, J.F. Mano, Encapsula-
man, J.J. Chae, Q.Y. Guo, R. Trow, A. Thomas, Z.P. Hou, S. Lichtsteiner, D. Sut- tion of adipose-derived stem cells and transforming growth factor-β 1 in
ton, C. Matheson, P. Walker, N. David, S. Mori, J.M. Taube, Photoactivated carrageenan-based hydrogels for cartilage tissue engineering, J. Bioact. Com-
Composite Biomaterial for Soft Tissue Restoration in Rodents and in Humans, pat. Polym. (2011), doi:10.1177/0883911511420700.
Sci. Transl. Med. (2011). [316] Y. Deng, M. Huang, D. Sun, Y. Hou, Y. Li, T. Dong, X. Wang, L. Zhang, W. Yang,
[291] E.M. Cronin, F.A. Thurmond, R. Bassel-Duby, R.S. Williams, W.E. Wright, Dual Physically Cross-Linked κ -Carrageenan-Based Double Network Hydro-
K.D. Nelson, H.R. Garner, Protein-coated poly(L-lactic acid) fibers provide a gels with Superior Self-Healing Performance for Biomedical Application, ACS
substrate for differentiation of human skeletal muscle cells, J. Biomed. Mater. Appl. Mater. Interfaces. (2018), doi:10.1021/acsami.8b15385.
Res. - Part A. (2004), doi:10.1002/jbm.a.30009. [317] X. Qi, T. Su, M. Zhang, X. Tong, W. Pan, Q. Zeng, Z. Zhou, L. Shen, X. He,
[292] C.W. Patrick, P.B. Chauvin, J. Hobley, G.P. Reece, Preadipocyte seeded PLGA J. Shen, Macroporous Hydrogel Scaffolds with Tunable Physicochemical Prop-
scaffolds for adipose tissue engineering, Tissue Eng (1999), doi:10.1089/ten. erties for Tissue Engineering Constructed Using Renewable Polysaccharides,
1999.5.139. ACS Appl. Mater. Interfaces. (2020), doi:10.1021/acsami.9b20794.
[293] C. Brännmark, A. Paul, D. Ribeiro, B. Magnusson, G. Brolén, A. Enejder, [318] F. Munarin, S.G. Guerreiro, M.A. Grellier, M.C. Tanzi, M.A. Barbosa, P. Petrini,
A. Forslöw, Increased adipogenesis of human adipose-derived stem cells on P.L. Granja, Pectin-based injectable biomaterials for bone tissue engineering,
polycaprolactone fiber matrices, PLoS ONE 9 (2014) 1–27, doi:10.1371/journal. Biomacromolecules (2011), doi:10.1021/bm101110x.
pone.0113620. [319] N. Ninan, M. Muthiah, I.K. Park, A. Elain, S. Thomas, Y. Grohens,
[294] B. Perniconi, A. Costa, P. Aulino, L. Teodori, S. Adamo, D. Coletti, The pro- Pectin/carboxymethyl cellulose/microfibrillated cellulose composite scaffolds
myogenic environment provided by whole organ scale acellular scaffolds for tissue engineering, Carbohydr. Polym. 98 (2013) 877–885, doi:10.1016/j.
from skeletal muscle, Biomaterials (2011), doi:10.1016/j.biomaterials.2011.07. carbpol.2013.06.067.
016. [320] R.F. Pereira, C.C. Barrias, P.J. Bártolo, P.L. Granja, Cell-instructive pectin hy-
[295] M.T. Wolf, K.A. Daly, J.E. Reing, S.F. Badylak, Biologic scaffold composed drogels crosslinked via thiol-norbornene photo-click chemistry for skin tis-
of skeletal muscle extracellular matrix, Biomaterials (2012), doi:10.1016/j. sue engineering, Acta Biomater 66 (2018) 282–293, doi:10.1016/j.actbio.2017.
biomaterials.2011.12.055. 11.016.
[296] B.M. Sicari, J. Peter Rubin, C.L. Dearth, M.T. Wolf, F. Ambrosio, M. Boninger, [321] M.M. Fares, E. Shirzaei Sani, R. Portillo Lara, R.B. Oliveira, A. Khademhos-
N.J. Turner, D.J. Weber, T.W. Simpson, A. Wyse, E.H.P. Brown, J.L. Dziki, seini, N. Annabi, Interpenetrating network gelatin methacryloyl (GelMA) and
L.E. Fisher, S. Brown, S.F. Badylak, An acellular biologic scaffold promotes pectin-g-PCL hydrogels with tunable properties for tissue engineering, Bio-
skeletal muscle formation in mice and humans with volumetric muscle loss, mater. Sci. (2018), doi:10.1039/c8bm00474a.
Sci. Transl. Med. (2014), doi:10.1126/scitranslmed.3008085. [322] M. Mehrali, A. Thakur, F.B. Kadumudi, M.K. Pierchala, J.A.V. Cordova,
[297] J.A. Werkmeister, J.A.M. Ramshaw, Recombinant protein scaffolds for tissue M.A. Shahbazi, M. Mehrali, C.P. Pennisi, G. Orive, A.K. Gaharwar,
engineering, Biomed. Mater. (2012), doi:10.1088/1748-6041/7/1/012002. A. Dolatshahi-Pirouz, Pectin Methacrylate (PEMA) and Gelatin-Based Hy-
[298] Q. Wang, C. Zhong, H. Xiao, Genetic Engineering of Filamentous Fungi for Ef- drogels for Cell Delivery: converting Waste Materials into Biomaterials, ACS
ficient Protein Expression and Secretion, Front. Bioeng. Biotechnol. 8 (2020) Appl. Mater. Interfaces. 11 (2019) 12283–12297, doi:10.1021/acsami.9b00154.
1–8, doi:10.3389/fbioe.2020.00293. [323] A. Lapomarda, A. De Acutis, I. Chiesa, G.M. Fortunato, F. Montemurro, C. De
[299] T. Wang, J. Lew, J. Premkumar, C.L. Poh, M.Win Naing, Production of recom- Maria, M. Mattioli Belmonte, R. Gottardi, G. Vozzi, Pectin-GPTMS-Based
binant collagen: state of the art and challenges, Eng. Biol. 1 (2017) 18–23, Biomaterial: toward a Sustainable Bioprinting of 3D scaffolds for Tissue
doi:10.1049/enb.2017.0 0 03. Engineering Application, Biomacromolecules (2020), doi:10.1021/acs.biomac.
[300] S. Gomes, I.B. Leonor, J.F. Mano, R.L. Reis, D.L. Kaplan, Natural and genetically 9b01332.
engineered proteins for tissue engineering, Prog. Polym. Sci. (2012), doi:10. [324] S. Chen, S. Cui, H. Zhang, X. Pei, J. Hu, Y. Zhou, Y. Liu, Cross-Linked Pectin
1016/j.progpolymsci.2011.07.003. Nanofibers with Enhanced Cell Adhesion, Biomacromolecules (2018), doi:10.
[301] W. Suntornsuk, P. Pochanavanich, L. Suntornsuk, Fungal chitosan produc- 1021/acs.biomac.7b01605.
tion on food processing by-products, Process Biochem. (2002), doi:10.1016/ [325] F. Chen, Y. Ni, B. Liu, T. Zhou, C. Yu, Y. Su, X. Zhu, X. Yu, Y. Zhou, Self-
S0 032-9592(01)0 0265-5. crosslinking and injectable hyaluronic acid/RGD-functionalized pectin hydro-

20
JID: ACTBIO
ARTICLE IN PRESS [m5G;February 21, 2021;17:50]

S. Ng and M. Kurisawa Acta Biomaterialia xxx (xxxx) xxx

gel for cartilage tissue engineering, Carbohydr. Polym. (2017), doi:10.1016/j. A. Khademhosseini, An automated two-phase system for hydrogel microbead
carbpol.2017.02.059. production, Biofabrication (2012), doi:10.1088/1758-5082/4/3/035003.
[326] J.G. Martins, S.E.A. Camargo, T.T. Bishop, K.C. Popat, M.J. Kipper, A.F. Martins, [349] A.C. Mendes, E.T. Baran, R.C. Pereira, H.S. Azevedo, R.L. Reis, Encapsulation
Pectin-chitosan membrane scaffold imparts controlled stem cell adhesion and and Survival of a Chondrocyte Cell Line within Xanthan Gum Derivative,
proliferation, Carbohydr. Polym. (2018), doi:10.1016/j.carbpol.2018.05.062. Macromol. Biosci. (2012), doi:10.1002/mabi.201100304.
[327] Y. Zhao, M. He, L. Zhao, S. Wang, Y. Li, L. Gan, M. Li, L. Xu, P.R. Chang, [350] A. Kumar, K.M. Rao, S.E. Kwon, Y.N. Lee, S.S. Han, Xanthan gum/bioactive
D.P. Anderson, Y. Chen, Epichlorohydrin-Cross-linked Hydroxyethyl Cellu- silica glass hybrid scaffolds reinforced with cellulose nanocrystals: morpho-
lose/Soy Protein Isolate Composite Films as Biocompatible and Biodegrad- logical, mechanical and in vitro cytocompatibility study, Mater. Lett. (2017),
able Implants for Tissue Engineering, ACS Appl. Mater. Interfaces. (2016), doi:10.1016/j.matlet.2017.01.143.
doi:10.1021/acsami.5b11152. [351] A. Kumar, K.M. Rao, S.S. Han, Application of xanthan gum as polysaccha-
[328] K. Rodríguez, J. Sundberg, P. Gatenholm, S. Renneckar, Electrospun nanofi- ride in tissue engineering: a review, Carbohydr. Polym. (2018), doi:10.1016/
brous cellulose scaffolds with controlled microarchitecture, Carbohydr. Polym. j.carbpol.2017.10.009.
100 (2014) 143–149, doi:10.1016/j.carbpol.2012.12.037. [352] A.C. Mendes, E.T. Baran, C. Nunes, M.A. Coimbra, H.S. Azevedo, R.L. Reis,
[329] J.R. Gershlak, S. Hernandez, G. Fontana, L.R. Perreault, K.J. Hansen, S.A. Lar- Palmitoylation of xanthan polysaccharide for self-assembly microcapsule for-
son, B.Y.K. Binder, D.M. Dolivo, T. Yang, T. Dominko, M.W. Rolle, P.J. Weath- mation and encapsulation of cells in physiological conditions, Soft Matter
ers, F. Medina-Bolivar, C.L. Cramer, W.L. Murphy, G.R. Gaudette, Crossing king- (2011), doi:10.1039/c1sm05594a.
doms: using decellularized plants as perfusable tissue engineering scaffolds, [353] A.C. Mendes, E.T. Baran, R.L. Reis, H.S. Azevedo, Fabrication of phospholipid-
Biomaterials. 125 (2017) 13–22, doi:10.1016/j.biomaterials.2017.02.011. xanthan microcapsules by combining microfluidics with self-assembly, Acta
[330] Y. Huang, J. Wang, F. Yang, Y. Shao, X. Zhang, K. Dai, Modification and eval- Biomater. (2013), doi:10.1016/j.actbio.2013.01.035.
uation of micro-nano structured porous bacterial cellulose scaffold for bone [354] V.B. Bueno, R. Bentini, L.H. Catalani, L.R.S. Barbosa, D.F.S. Petri, Synthesis and
tissue engineering, Mater. Sci. Eng. C. (2017), doi:10.1016/j.msec.2017.02.174. characterization of xanthan-hydroxyapatite nanocomposites for cellular up-
[331] J. Ran, P. Jiang, S. Liu, G. Sun, P. Yan, X. Shen, H. Tong, Con- take, Mater. Sci. Eng. C. (2014), doi:10.1016/j.msec.2014.01.002.
structing multi-component organic/inorganic composite bacterial cellulose- [355] Z. Li, Y. Su, B. Xie, X. Liu, X. Gao, D. Wang, A novel biocompatible double
gelatin/hydroxyapatite double-network scaffold platform for stem cell- network hydrogel consisting of konjac glucomannan with high mechanical
mediated bone tissue engineering, Mater. Sci. Eng. C. (2017), doi:10.1016/j. strength and ability to be freely shaped, J. Mater. Chem. B. (2015), doi:10.
msec.2017.04.062. 1039/c4tb01653j.
[332] Y. Zhao, M. He, H. Jin, L. Zhao, Q. Du, H. Deng, W. Tian, Y. Li, X. Lv, Y. Chen, [356] J. Tang, J. Chen, J. Guo, Q. Wei, H. Fan, Construction and evaluation of fibrillar
Construction of highly biocompatible hydroxyethyl cellulose/soy protein iso- composite hydrogel of collagen/konjac glucomannan for potential biomedical
late composite sponges for tissue engineering, Chem. Eng. J. (2018), doi:10. applications, Regen. Biomater. (2018), doi:10.1093/rb/rby018.
1016/j.cej.2018.02.046. [357] H. Kanniyappan, P. Thangavel, S. Chakraborty, V. Arige, V. Muthuvijayan, De-
[333] R.J. Hickey, D.J. Modulevsky, C.M. Cuerrier, A.E. Pelling, Customizing the sign and evaluation of Konjac glucomannan-based bioactive interpenetrating
Shape and Microenvironment Biochemistry of Biocompatible Macroscopic network (IPN) scaffolds for engineering vascularized bone tissues, Int. J. Biol.
Plant-Derived Cellulose Scaffolds, ACS Biomater. Sci. Eng. (2018), doi:10.1021/ Macromol. (2020), doi:10.1016/j.ijbiomac.2019.12.012.
acsbiomaterials.8b00178. [358] H. Zhang, S. Cui, H. Lv, X. Pei, M. Gao, S. Chen, J. Hu, Y. Zhou, Y. Liu, A
[334] S. Ahn, C.O. Chantre, A.R. Gannon, J.U. Lind, P.H. Campbell, T. Grevesse, crosslinking strategy to make neutral polysaccharide nanofibers robust and
B.B. O’Connor, K.K. Parker, Soy Protein/Cellulose Nanofiber Scaffolds Mim- biocompatible: with konjac glucomannan as an example, Carbohydr. Polym.
icking Skin Extracellular Matrix for Enhanced Wound Healing, Adv. Healthc. (2019), doi:10.1016/j.carbpol.2019.03.075.
Mater. (2018), doi:10.1002/adhm.201701175. [359] L. Sun, Z. Xiong, W. Zhou, R. Liu, X. Yan, J. Li, W. An, G. Yuan, G. Ma, Z. Su,
[335] A. Tiwari, J.J. Grailer, S. Pilla, D.A. Steeber, S. Gong, Biodegradable hydrogels Novel konjac glucomannan microcarriers for anchorage-dependent animal
based on novel photopolymerizable guar gum-methacrylate macromonomers cell culture, Biochem. Eng. J. (2015), doi:10.1016/j.bej.2014.12.012.
for in situ fabrication of tissue engineering scaffolds, Acta Biomater. (2009), [360] J. Chen, Z. Cai, Q. Wei, D. Wang, J. Wu, Y. Tan, J. Lu, H. Ai, Proanthocyanidin-
doi:10.1016/j.actbio.20 09.06.0 01. crosslinked collagen/konjac glucomannan hydrogel with improved mechani-
[336] M. Ragothaman, T. Palanisamy, C. Kalirajan, Collagen-poly(dialdehyde) guar cal properties and MRI trackable biodegradation for potential tissue engineer-
gum based porous 3D scaffolds immobilized with growth factor for tissue en- ing scaffolds, J. Mater. Chem. B. (2020), doi:10.1039/c9tb02053e.
gineering applications, Carbohydr. Polym. (2014), doi:10.1016/j.carbpol.2014. [361] Q. Jiang, N. Reddy, Y. Yang, Cytocompatible cross-linking of electrospun zein
08.045. fibers for the development of water-stable tissue engineering scaffolds, Acta
[337] S. Kundu, A. Das, A. Basu, D. Ghosh, P. Datta, A. Mukherjee, Carboxymethyl Biomater (2010), doi:10.1016/j.actbio.2010.04.024.
guar gum synthesis in homogeneous phase and macroporous 3D scaffolds [362] S. Gong, H. Wang, Q. Sun, S.T. Xue, J.Y. Wang, Mechanical properties and in
design for tissue engineering, Carbohydr. Polym. (2018), doi:10.1016/j.carbpol. vitro biocompatibility of porous zein scaffolds, Biomaterials (2006), doi:10.
2018.03.007. 1016/j.biomaterials.2006.02.019.
[338] D. Anandan, G. Madhumathi, N.A. Nambiraj, A.K. Jaiswal, Gum based 3D com- [363] D. Dippold, M. Tallawi, S. Tansaz, J.A. Roether, A.R. Boccaccini, Novel electro-
posite scaffolds for bone tissue engineering applications, Carbohydr. Polym. spun poly(glycerol sebacate)-zein fiber mats as candidate materials for car-
214 (2019) 62–70, doi:10.1016/j.carbpol.2019.03.020. diac tissue engineering, Eur. Polym. J. (2016), doi:10.1016/j.eurpolymj.2015.12.
[339] L.R. Stevens, K.J. Gilmore, G.G. Wallace, M. In het Panhuis, Tissue engi- 030.
neering with gellan gum, Biomater. Sci. 4 (2016) 1276–1290, doi:10.1039/ [364] J. Tu, H. Wang, H. Li, K. Dai, J. Wang, X. Zhang, The in vivo bone formation by
c6bm00322b. mesenchymal stem cells in zein scaffolds, Biomaterials (2009), doi:10.1016/j.
[340] C.A. Vilela, C. Correia, A. da Silva Morais, T.C. Santos, A.C. Gertrudes, biomaterials.2009.04.054.
E.S. Moreira, A.M. Frias, D.A. Learmonth, P. Oliveira, J.M. Oliveira, R.A. Sousa, [365] H. Xu, S. Cai, A. Sellers, Y. Yang, Intrinsically water-stable electrospun three-
J.D. Espregueira-Mendes, R.L. Reis, In vitro and in vivo performance of dimensional ultrafine fibrous soy protein scaffolds for soft tissue engineering
methacrylated gellan gum hydrogel formulations for cartilage repair∗ , J. using adipose derived mesenchymal stem cells, RSC Adv. (2014), doi:10.1039/
Biomed. Mater. Res. - Part A. (2018), doi:10.1002/jbm.a.36406. c3ra47547f.
[341] Y. Gong, C. Wang, R.C. Lai, K. Su, F. Zhang, D.A. Wang, An improved injectable [366] N. Reddy, Y. Yang, Soyprotein fibers with high strength and water stability for
polysaccharide hydrogel: modified gellan gum for long-term cartilage regen- potential medical applications, Biotechnol. Prog. (2009), doi:10.1002/btpr.244.
eration in vitro, J. Mater. Chem. 19 (2009) 1968–1977, doi:10.1039/b818090c. [367] K.B. Chien, R.N. Shah, Novel soy protein scaffolds for tissue regeneration: ma-
[342] C.J. Ferris, L.R. Stevens, K.J. Gilmore, E. Mume, I. Greguric, D.M. Kirchmajer, terial characterization and interaction with human mesenchymal stem cells,
G.G. Wallace, M. In Het Panhuis, Peptide modification of purified gellan gum, Acta Biomater. (2012), doi:10.1016/j.actbio.2011.09.036.
J. Mater. Chem. B. (2015), doi:10.1039/c4tb01727g. [368] K.B. Chien, E. Makridakis, R.N. Shah, Three-dimensional printing of soy pro-
[343] N.A. Silva, M.J. Cooke, R.Y. Tam, N. Sousa, A.J. Salgado, R.L. Reis, M.S. Shoichet, tein scaffolds for tissue regeneration, Tissue Eng. - Part C Methods. (2013),
The effects of peptide modified gellan gum and olfactory ensheathing glia doi:10.1089/ten.tec.2012.0383.
cells on neural stem/progenitor cell fate, Biomaterials (2012), doi:10.1016/j. [369] H. Olami, M. Zilberman, Microstructure and in vitro cellular response to novel
biomaterials.2012.05.050. soy protein-based porous structures for tissue regeneration applications, J.
[344] J.T. Koivisto, C. Gering, J. Karvinen, R. Maria Cherian, B. Belay, J. Hyttinen, Biomater. Appl. (2016), doi:10.1177/0885328215614713.
K. Aalto-Setälä, M. Kellomäki, J. Parraga, Mechanically Biomimetic Gelatin- [370] J. Huang, K. Huang, X. You, G. Liu, G. Hollett, Y. Kang, Z. Gu, J. Wu, Evaluation
Gellan Gum Hydrogels for 3D Culture of Beating Human Cardiomyocytes, ACS of tofu as a potential tissue engineering scaffold, J. Mater. Chem. B. (2018),
Appl. Mater. Interfaces. (2019), doi:10.1021/acsami.8b22343. doi:10.1039/c7tb02852k.
[345] J.T. Oliveira, L. Martins, R. Picciochi, P.B. Malafaya, R.A. Sousa, N.M. Neves, [371] K. Huang, Z. Gu, J. Wu, Tofu-Incorporated Hydrogels for Potential Bone
J.F. Mano, R.L. Reis, Gellan gum: a new biomaterial for cartilage tissue en- Regeneration, ACS Biomater. Sci. Eng. (2020), doi:10.1021/acsbiomaterials.
gineering applications, J. Biomed. Mater. Res. - Part A. 93 (2010) 852–863, 9b01997.
doi:10.1002/jbm.a.32574. [372] C. Liu, Y. Chen, X. Wang, J. Huang, P.R. Chang, D.P. Anderson, Improvement
[346] D. Dyondi, T.J. Webster, R. Banerjee, A nanoparticulate injectable hydrogel as in physical properties and cytocompatibility of zein by incorporation of pea
a tissue engineering scaffold for multiple growth factor delivery for bone re- protein isolate, J. Mater. Sci. (2010), doi:10.1007/s10853-010-4774-z.
generation, Int. J. Nanomedicine. (2012), doi:10.2147/IJN.S37953. [373] K. Tuzlakoglu, N. Bolgen, A.J. Salgado, M.E. Gomes, E. Piskin, R.L. Reis, Nano-
[347] L.P. Da Silva, M.T. Cerqueira, R.A. Sousa, R.L. Reis, V.M. Correlo, A.P. Marques, and micro-fiber combined scaffolds: a new architecture for bone tissue engi-
Engineering cell-adhesive gellan gum spongy-like hydrogels for regenerative neering, J. Mater. Sci. Mater. Med. (2005), doi:10.1007/s10856- 005- 4713- 8.
medicine purposes, Acta Biomater (2014), doi:10.1016/j.actbio.2014.07.009. [374] M.E. Gomes, H.L. Holtorf, R.L. Reis, A.G. Mikos, Influence of the porosity of
[348] D.F. Coutinho, A.F. Ahari, N.N. Kachouie, M.E. Gomes, N.M. Neves, R.L. Reis, starch-based fiber mesh scaffolds on the proliferation and osteogenic differ-

21
JID: ACTBIO
ARTICLE IN PRESS [m5G;February 21, 2021;17:50]

S. Ng and M. Kurisawa Acta Biomaterialia xxx (xxxx) xxx

entiation of bone marrow stromal cells cultured in a flow perfusion bioreac- [382] A.M. Hermansson, E. Eriksson, E. Jordansson, Effects of potassium, sodium
tor, Tissue Eng. (2006), doi:10.1089/ten.2006.12.801. and calcium on the microstructure and rheological behaviour of kappa-
[375] M.I. Santos, S. Fuchs, M.E. Gomes, R.E. Unger, R.L. Reis, C.J. Kirkpatrick, carrageenan gels, Carbohydr. Polym. (1991), doi:10.1016/0144-8617(91)
Response of micro- and macrovascular endothelial cells to starch-based 90115-S.
fiber meshes for bone tissue engineering, Biomaterials (2007), doi:10.1016/ [383] Z.H. Qu, H.J. Wang, T.T. Tang, X.L. Zhang, J.Y. Wang, K.R. Dai, Evaluation of
j.biomaterials.20 06.08.0 06. the zein/inorganics composite on biocompatibility and osteoblastic differen-
[376] I. Van Nieuwenhove, A. Salamon, S. Adam, P. Dubruel, S. Van Vlierberghe, tiation, Acta Biomater. 4 (2008) 1360–1368, doi:10.1016/j.actbio.2008.03.006.
K. Peters, Gelatin- and starch-based hydrogels. Part B: in vitro mesenchymal [384] D. Yang, Y. Yuan, L. Wang, X. Wang, R. Mu, J. Pang, J. Xiao, Y. Zheng, A review
stem cell behavior on the hydrogels, Carbohydr. Polym. (2017), doi:10.1016/j. on konjac glucomannan gels: microstructure and application, Int. J. Mol. Sci.
carbpol.2017.01.010. 18 (2017), doi:10.3390/ijms18112250.
[377] M.A. Da Silva, A. Crawford, J. Mundy, A. Martins, J.V. Araújo, P.V. Hat- [385] A. Noreen, Z. i. H. Nazli, J. Akram, I. Rasul, A. Mansha, N. Yaqoob, R. Iqbal,
ton, R.L. Reis, N.M. Neves, Evaluation of extracellular matrix formation in S. Tabasum, M. Zuber, K.M. Zia, Pectins functionalized biomaterials; a new
polycaprolactone and starch-compounded polycaprolactone nanofiber meshes viable approach for biomedical applications: a review, Int. J. Biol. Macromol.
when seeded with bovine articular chondrocytes, Tissue Eng. - Part A. (2009), (2017), doi:10.1016/j.ijbiomac.2017.03.029.
doi:10.1089/ten.tea.2007.0327. [386] A. George, P.A. Shah, P.S. Shrivastav, Guar gum: versatile natural polymer for
[378] I. Silva, M. Gurruchaga, I. Goñi, M. Fernández-Gutiérrez, B. Vázquez, drug delivery applications, Eur. Polym. J. (2019), doi:10.1016/j.eurpolymj.2018.
J.S. Román, Scaffolds based on hydroxypropyl starch: processing, morphol- 10.042.
ogy, characterization, and biological behavior, J. Appl. Polym. Sci. (2013), [387] T. Hemamalini, V.R. Giri Dev, Comprehensive review on electrospinning of
doi:10.1002/app.37551. starch polymer for biomedical applications, Int. J. Biol. Macromol. (2018),
[379] D. Anandan, G. Madhumathi, N.A. Nambiraj, A.K. Jaiswal, Gum based 3D com- doi:10.1016/j.ijbiomac.2017.08.079.
posite scaffolds for bone tissue engineering applications, Carbohydr. Polym. [388] S. Tansaz, A.R. Boccaccini, Biomedical applications of soy protein: a brief
(2019), doi:10.1016/j.carbpol.2019.03.020. overview, J. Biomed. Mater. Res. - Part A. (2016), doi:10.1002/jbm.a.35569.
[380] N. Pettinelli, S. Rodríguez-Llamazares, V. Abella, L. Barral, R. Bouza, Y. Farrag, [389] H.J. Wang, J.X. Fu, J.Y. Wang, Effect of water vapor on the surface character-
F. Lago, Entrapment of chitosan, pectin or κ -carrageenan within methacry- istics and cell compatibility of zein films, Colloids Surfaces B Biointerfaces
late based hydrogels: effect on swelling and mechanical properties, Mater. (2009), doi:10.1016/j.colsurfb.2008.11.015.
Sci. Eng. C. (2019), doi:10.1016/j.msec.2018.11.071. [390] R. Paliwal, S. Palakurthi, Zein in controlled drug delivery and tissue engineer-
[381] A. Ali, S. Ahmed, Recent Advances in Edible Polymer Based Hydrogels as a ing, J. Control. Release. 189 (2014) 108–122, doi:10.1016/j.jconrel.2014.06.036.
Sustainable Alternative to Conventional Polymers, J. Agric. Food Chem. (2018),
doi:10.1021/acs.jafc.8b01052.

22

View publication stats

You might also like