You are on page 1of 17

Engineered Regeneration 3 (2022) 24–40

Contents lists available at ScienceDirect

Engineered Regeneration
journal homepage: http://www.keaipublishing.com/en/journals/engineered-regeneration/

Polyhydroxyalkanoates in tissue repair and regeneration


Wentai Guo a,b,c, Keli Yang a,b, Xiusen Qin a,b, Rui Luo a,b, Hui Wang a,b,∗, Rongkang Huang a,b,∗
a
Department of Colorectal Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen
University, Guangzhou 510655, China
b
Institute of Biomedical Innovation and Laboratory of Regenerative Medicine and Biomaterials, Biomedical Material Conversion and Evaluation Engineering Technology
Research Center of Guangdong Province, Guangzhou 510655, China
c
School of Materials Science and Engineering, National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology,
Guangzhou 510006, China

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

Keywords: Developing advanced biocompatibility materials is of critical importance in biomedical engineering. Polyhydrox-
Polyhydroxyalkanoates yalkanoates (PHA), being famous for its flexible mechanical properties, thermal properties, biocompatibility, and
Biomedical engineering biodegradability, has been widely used in wound dressings, artificial blood vessels, heart valves, nerve conduits,
Tissue repair
bone and cartilage scaffolds, surgical sutures, and other fields. However, reports on the application of PHA in
Tissue regeneration
tissue repair and regeneration are often lacking in systematics. Here, a comprehensive and in-depth perception of
Biomaterial
the performance advantages and application value of PHA is provided. In this review, the following applications
of PHA in biomedical engineering are covered: i) soft tissue, ii) organ tissue, iii) vascular tissue, iv) heart valve
tissue, v) nerve conduit tissue, vi) bone tissue, vii) cartilage tissue and viii) others. Finally, an outlook on the
future research directions and challenges of PHA is presented.

1. Introduction the hydroxyalkanoate (HA) monomer level [25–27]. Fig. 1 illustrates the
typical chemical structural of PHA, where each HA monomer has a side
Compared to traditional petrochemical-based polymers, bio-sourced chain attached to the main chain. It can be substituted by various func-
polymers show many advantages and are often fermented from cheap tional groups, including methyl (C1) to tridecyl (C13), aromatic, etc.
renewable resources. Moreover, biopolymers’ good biological proper- This variation makes PHA easily modified to synthesize polymers with
ties and degradability make them widely used in biomedical engineer- different properties [28,29]. According to the number of HA monomers
ing [1–3]. Among the many biopolymers, polyhydroxyalkanoates (PHA) integrated into the polymerization chain, PHA can be divided into three
are linear polyesters synthesized by microorganisms and prototyped categories: the first category consists of short chain length PHA (short-
as metabolic particles accumulated in the cytoplasm of prokaryotic chain PHA) with 3 to 5 carbons, such as polyhydroxyvalerate (PHV)
cells [4,5]. It is usually an energy and carbon storage material pro- and polyhydroxybutyrate (PHB); the second category consists of MCL-
duced by bacterial microbiomes under the conditions of sufficient car- PHA (medium chain length PHA) with 6 to 13 carbons, such as poly-
bon sources but lacks nitrogen and phosphorus nutrients [6,7]. PHA hydroxyhexanoates (PHHx) and polyhydroxyoctanoates (PHO); and the
can be synthesized by most Gram-positive bacteria (Bacillus [8,9]), third category of LCL-PHA (long chain length PHA) consisting of more
Gram-negative bacteria (Pseudomonas [10,11], Alcaligenes [12,13], and than 14 carbon units, such as 3-hydroxyoctadecanoic acid (3HOD) and
Aeromonas Bacillus [14,15]), anaerobic [16,17] and aerobic bacteria 3-hydroxyhexadecanoic acid (3HHD) [30]. Among them, SCL-PHA has
[18,19], photosynthetic bacteria [20,21], and archaea [22,23]. These typical thermoplastic properties with a high glass transition tempera-
microbiomes achieve PHA accumulation through a variety of carbon- ture (Tg); while MCL-PHA is similar to latex-like elastomers with a low
rich substrates. In addition, some bacteria have been shown to produce molecular weight.
PHA without any nutrient limitation, such as Alcaligenes latus strain IAM Over the past decade, about 150 monomers of PHA poly-
12,664 T [24]. mers have been identified, but only a few of them are in-
Currently, the PHA members produced by bacterial fermentation volved in the formation of PHA, including PHB, PHV, poly-4-
vary considerably in structure and properties. This mainly depends on hydroxybutyrate (P4HB), PHO, poly-hydroxybutyrate-hydroxyvalerate
microbial species, biosynthetic conditions, and carbon source substrate (PHBV), poly-hydroxybutyrate-co-hydroxyhexanoate (PHBHHx), and
in the production process, thereby changing the composition of PHA at poly-3-hydroxybutyrate-4-hydroxybutyrate (P3HB4HB) [31]. Due to its


Corresponding author.
E-mail addresses: wang89@mail.sysu.edu.cn (H. Wang), huangrk3@mail.sysu.edu.cn (R. Huang).

https://doi.org/10.1016/j.engreg.2022.01.003
Received 30 December 2021; Received in revised form 18 January 2022; Accepted 19 January 2022
Available online 21 January 2022
2666-1381/© 2022 The Authors. Publishing Services by Elsevier B.V. on behalf of KeAi Communications Co. Ltd. This is an open access article under the CC
BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/)
W. Guo, K. Yang, X. Qin et al. Engineered Regeneration 3 (2022) 24–40

organ, vascular, nerve, bone, cartilage, drug carrier


skin, organ, vascular, nerve, bone, cartilage

skin, organ, vascular, nerve, bone, cartilage


skin, organ, vascular, bone, cartilage
heart valve, suture
Application

heart valve

Young’s modulus (GPa)
Fig. 1. The typical chemical structural of PHA. (a) PHB. (b) P4HB. (c) PHV. (d)

0.149 ∼ 0.180
PHO. (e) P3HB4HB. (f) PHBV. (g) PHBHHx.

1.1 ∼ 3.5

0.2 ∼ 2.9
0.0199

0.0236
biocompatibility and adaptability, these representative PHA has been


widely used in wound dressings [32], artificial blood vessels [33], heart

Elongation at break (%)


valves [34], bone [35] and cartilage scaffolds [36], nerve conduits [37],
and surgical sutures [38]. However, reports on the application of PHA
in tissue repair and regeneration are often lacking in systematics [39].

696.6 ∼ 1014
Therefore, in this review, we review the achievements of PHA in tissue

380 ∼ 850
268 ∼626
repair and regeneration over the past decade or so, and provide an out-

312.9
5∼6

look on the future application trends of PHA. The medical-engineering 14

50
multidisciplinary intersection has strongly promoted the progress of
PHA in personalized precision tissue repair and regeneration, while pro-
Tensile strength (MPa)

viding a new strategy for the development of biomedical engineering.

2. Properties of PHA
13.8 ∼ 104
40 ∼ 62

20 ∼ 38
9 ∼ 21
3 ∼16
31.2

PHA has good biodegradability, biocompatibility, non-


12

carcinogenicity, thermal and mechanical properties. Due to the


abundance of monomers, PHA also exhibits a high degree of structural
−50 ∼ −47

−45 ∼ −7
−8 ∼ −1
−2 ∼ −1

diversity. Based on these properties, this type of polymeric material


Characteristics of different types of PHA.

Tg (°C)

−16.5
−37.2

has high application value in tissue repair and regeneration. As shown


4

in Table 1, we have summarized the properties and advantages of


common kinds of PHA.
104.2 ∼ 109.7
177 ∼ 180

152 ∼ 164
133 ∼ 170
120 ∼ 127
53 ∼ 61

2.1. Biodegradability
Tm (°C)

72.2

One of the unique biological properties of PHA is its degradability


in a variety of environments [40,41]. PHA can be degraded by mixed
Type of PHA

microbial populations existing in rivers, seawater, soil, and other en-


P3HB4HB

PHBHHx
Table 1

vironments under aerobic or anoxic conditions, and decomposed into


PHBV
P4HB

PHO
PHV
PHB

water and carbon dioxide [42,43]. These microorganisms degrade PHA


into low molecular weight monomers or oligomers by secreting PHA
depolymerase (hydrolases). The above oligomer and monomer compo-
nents are subsequently taken up by the cells as nutrients [44]. Qu et al.
tested the biodegradation of PHA in different environments and found
that PHB and PHBV can be wholly degraded under most conditions [45].
PHA is the currently known degradable biomaterial with a minor degra-
dation restriction. Its biodegradation rate mainly depends on the affinity
of PHA depolymerase for ester bonds in PHA polymers, which is influ-

25
W. Guo, K. Yang, X. Qin et al. Engineered Regeneration 3 (2022) 24–40

enced by polymer conformation, surface area, monomer type, monomer crystallinity of MCL-PHA is relatively low, typically 40%, with Tg range
composition, molecular weight, crystallinity, and environmental factors from −52°C to −25°C at room temperature and Tm range from 38°C to
(pH, humidity, and temperature) [46]. In general, the degradation rate 80°C [61]. Since the Tg of MCL-PHA is lower than room temperature, it
of polymers increases faster with decreasing crystallinity and melting can exhibit elastic properties similar to rubber at appropriate tempera-
temperature (Tm). Therefore, MCL-PHA with low crystallinity and low tures [40]; when the external temperature is higher than Tm, MCL-PHA
Tm degrades faster than SCL-PHA with high crystallinity and high Tm. will transform into an amorphous form with viscous.
For example, P3HB4HB containing 4-hydroxybutyrate (4HB) and PHBV
copolymer containing 3-hydroxyvalerate (3HV) have lower crystallinity 2.5. Mechanical properties
and Tm than PHB homopolymer, so P3HB4HB and PHBV degrade faster
than PHB, and the degradation accelerates with increasing side-chain The mechanical strength of PHA mainly depends on the carbon-chain
monomer content [47]. length, structure, and composition. PHB is the most rigid PHA family
member with a strength of over 45 MPa, and its physicochemical fea-
2.2. Biocompatibility tures are similar to those of traditional plastics [29]. However, it has
poor processing performance, rapid aging speed, and low elongation at
Another unique biological property of PHA is biocompatibility, break. It usually needs to be combined with other monomers to reduce
which has attracted particular attention. In vitro experiments have crystallinity, stiffness, and aging (4HB, 3HP, and 3HV). Another rep-
shown that PHA can support the growth of most tissue cells, including resentative member of SCL-PHA, P4HB, is a strongly thermoplastic, ex-
epithelial cells, chondrocytes, and fibroblasts [48,49]. PHA will not trig- tensible ductile material with elongation at break up to 1,000%, making
ger a robust immune response when implanted in living tissues such as it the most elastic PHA homopolymer produced to date. In 2007, P4HB
rats, rabbits, and humans. This series of studies proved that PHA has ex- was first approved by the Food and Drugs Administration (FDA) to man-
cellent biocompatibility and can even promote cellular tissue adhesion ufacture surgical sutures [62,63]. Furthermore, in contrast to SCL-PHA,
and proliferation [50,51]. Moreover, the biocompatibility of PHA can be MCL-PHA is more elastic, as well as a higher elongation at break and
reflected in their immunological and physiological response after degra- lower tensile strength. MCL-PHA is a class of copolymer composed of
dation. Some monomers of the PHA polymerization chain, such as the 3HHx, 3H2O, 3HD, and 3HDD, with tensile strength between 5 MPa
3-hydroxybutyric acid (3-HB), are natural metabolites produced by the −16.3 MPa and elongation at break of 88% - 356% [61]. Therefore, com-
oxidation of fatty acids in the human liver [52]. It is present in human pared with poly(𝜀-caprolactone) (PCL), PLA, PLGA, and other polymers,
blood at concentrations of 30–100 mg/L. 3-HB can reduce inflammation, PHA has wider applicability and the mechanical strength of PHA could
promote tissue regeneration [53], and act as an energy substance for the be adjusted by compounding or synthetic copolymerization to simulate
body when necessary (starvation), eventually being excreted as carbon the final target environment [64,65].
dioxide [54,55]. In contrast, degradable polymers such as polylactic acid
(PLA) and polylactic-co-glycolic acid (PLGA) will cause accumulation of 3. Applications of PHA
lactic acid products after being implanted in the host, while PHA main-
tains pH stability during the degradation process, which undoubtedly Due to its structural diversity and favorable physical-chemical prop-
makes PHA more advantageous in terms of biocompatibility. erties, PHA has received extensive attention in biomedical tissue engi-
neering. To date, many studies have reported partial application of PHA
2.3. Non-carcinogenicity in tissue repair and regeneration, including soft tissue engineering, or-
gan tissue engineering, vascular tissue engineering, heart valve tissue
Non-carcinogenicity is one of the critical properties of biopolymers engineering, nerve tissue engineering, bone tissue engineering, and car-
used in humans. Notably, few studies have reported whether PHA stim- tilage tissue engineering (Fig. 2).
ulates cancer cell development while promoting cell proliferation [29].
In this case, Peng et al. found that rat fibroblasts cultured with PHB, 3.1. Soft tissue engineering
PHBV, P3HB4HB, and PHBHHx did not induce carcinogenesis, suggest-
ing that cells proliferating rapidly on PHA are regulated by the normal Soft tissue engineering covers many tissues, including skin, tendons,
cell cycle. In addition, primary rat osteosarcoma cells cultured in PHA blood vessels, nerves, and other tissues. As many researchers have re-
maintained normal morphology and did not transform into tumor mor- ported PHA applications in this area, a detailed description will be pre-
phology [56]. The mechanism by which PHA promotes tissue regener- sented in the following chapters. In this section, we only introduce the
ation may be related to the tissue cell response to 3-HB and microRNA- application of PHA in the repair of superficial soft tissues such as skin
mediated material-cell interactions [57]. Nevertheless, there is still no and tendons.
clear evidence to confirm the non-oncogenic effect of PHA on all cell In wound healing, basal cells surrounding the wound usually pro-
lines. Promoting and enhancing the research of non-carcinogenicity of liferate and migrate to the center of the defect to form a single epithe-
PHA on whole-cell lines will undoubtedly be an important part of future lium layer and then differentiate into the squamous epithelium. Wound
PHA applications research. dressings are the most effective way to accelerate wound healing, espe-
cially in large areas of skin tissue defects or infective wounds [66,67].
2.4. Thermal properties As shown in Table 2, the PHA engineering scaffolds prepared by tis-
sue engineering technology have obvious advantages in skin tissue re-
PHA is a semi-crystalline polymer, and its thermodynamic proper- pair. This is mainly owing to the acceptable mechanical, biocompati-
ties are usually expressed by Tm of the crystalline phase and Tg of ble, and repair-promoting properties of PHA. For example, Volova et al.
the amorphous phase. Several studies have shown that Tm decreases prepared P3HB4HB fibrous membranes by solvent casting and electro-
with increasing side chain carbon length, while Tg increases accord- static spinning techniques as wound dressings. Their results showed
ingly [58,59]. Therefore, SCL-PHA and MCL-PHA often exhibit differ- that P3HB4HB electrospun membranes were more conducive to stem
ent thermodynamic properties. For example, the crystallinity of PHB is cell proliferation and differentiation. In contrast, these cells could pro-
relatively high, about 60–80%, Tm is about 170°C, and the decomposi- mote the migration of peri‑wound epidermal cells to adjacent tissues
tion temperature (Td) is about 200°C. The close temperature threshold by producing an extracellular matrix (ECM). Therefore, PHA enhanced
leads to the poor thermal stability of PHB processing, which has to be angiogenesis, reduced inflammation, and ultimately promoted wound
improved by adding second monomers such as 5-hydroxyvaleric acid healing [68,69]. Amirul et al. incorporated collagen peptides to fabri-
(5HV) and 3-hydroxypropionic acid (3HP) [60]. On the other hand, the cate P3HB4HB nanofibrous membranes as a wound dressing to improve

26
W. Guo, K. Yang, X. Qin et al. Engineered Regeneration 3 (2022) 24–40

Fig. 2. Application of PHA in tissue repair and regeneration.

Table 2
PHA in soft tissue engineering.

Type of PHA Combination Model Advantage Year Reference

PHB gelatin a diabetic rat wound model faster wound healing with higher content of hair follicles and sweat 2021 [72]
glands
PHBV GelMA/EGF a diabetic rat wound model rapid diabetic wound healing, favourable angiogenesis and cellular 2021 [74]
response
PHBV HMK/AgNPs a rat wound healing model better wound healing ability, good biocompatibility, and favorable 2020 [156]
antibacterial properties (Fig. 3b)
PHBV – a full-thickness mouse wound model mitigated excessive scar formation by regulating myofibroblast 2020 [71]
formation
PHBV nCeO2 – promoted the healing of diabetic wounds, enhanced vascularization 2020 [73]
and cell proliferation (Fig. 3c)
P3HB4HB BC third-degree skin burn model in rats promoted healing more effectively than VoskoPran (a commercial 2019 [69]
wound dressing)
PHBV AMPs/Tac a deep-wound mouse model accelerated wound healing by reducing the number of bacteria 2018 [78]
PHB DE a NOD/SCID mouse xenograft model greater angiogenic and M2 macrophage polarization properties 2018 [165]
P3HB4HB fibroblasts model skin defects in Wistar rats promoted wound healing, enhanced vascularization, and reduced 2016 [68]
inflammation
P3HB4HB FSCPs a rat wound healing model significant promotion on wound contractions (61%) 2016 [166]
PHB PCL/PEGMA – significant collagen deposition, enhanced vascularization and 2015 [167]
fibrosis
PHBV ASCs a rat wound healing model improved wound healing ability, reduced scarring, 2015 [168]
necessary bioactive
PHBV CS/USSCs – significantly promoted wound healing 2014 [169]
PHBV collagen/gelatin a rat wound healing model effective in promoting epidermal regeneration in the early stages of 2007 [170]
wound healing
HMK: high molecular keratin; AgNPs: Ag nanoparticles; DE: dermo-epidermal skin equivalents; FSCPs: fish-scale collagen peptides; USSCs: unrestricted somatic
stem cells.

surface wettability further while possessing biodegradability. Hence, in On the other hand, the long degradation cycle of PHA helps to de-
vivo study using a Sprague Dawley (SD) rat model showed a positive ef- velop functional wound dressings with drug delivery capability. For
fect of the P3HB4HB/collagen peptides composite membrane on wound example, Marian et al. prepared chitosan (CS)/PHB composite mem-
contraction with a maximum percentage of 79% [70]. To inhibit exces- branes loaded with kaempferol nanocrystals for infected wounds. The
sive scarring in wound healing, Kim et al. prepared three kinds of PHBV membrane demonstrated sustained antibacterial properties with power-
electrospun scaffolds with different 3HV contents (10, 30, and 60 mol%, ful killing effects against multi-drug resistant bacteria at low concentra-
Fig. 3a). As the 3HV content increased, the crystallinity of PHBV poly- tions, making it highly suitable for critically infected wounds healing
mer decreased and the scaffolds became more elastic. The electrospun [75]. Marcano et al. obtained asymmetrically structured PHA fibrous
scaffold group had thin granulation tissue generation in mouse full-skin scaffolds containing anti-biofilm protein Dispersin B (DB) through phase
wound models without dehydration or nodular scar formation. These separation techniques. By optimizing the micro-nano structure of the
results suggest that the higher 3HV content in PHBV makes the polymer PHA substrate, the PHA/DB could be captured more effectively to in-
more elastic and maintains minimal dermal fibroblast/myofibroblast hibit the tissue surface biofilms or disrupt the formed biofilms [76,77].
transition to alleviate excessive scar proliferation, demonstrating its po- Modification of implants with antimicrobial peptides (AMPs) could re-
tential as an ideal wound dressing [71]. For complex skin wounds, San- duce antibiotic resistance and prevent infection, so Qiong et al. labeled
hueza et al. prepared dual-size gelatin/PHB electrospun fibers for regen- the AMPs, tachyplesin I (Tac) with PHA particle binding protein (PhaP)
eration of diabetic wounds. The fibers treatment group exhibited higher and immobilized them on the PHBV scaffolds (Fig. 3d). The Tac-PhaP
hair follicles, higher sweat gland content, and lower fibroblast content in coating enhanced the hydrophilicity of the PHBV scaffold and efficiently
the wounds [72]. Robin and colleagues blended cerium oxide nanopar- inhibited the growth of harmful bacteria. Further experiments on deep
ticles (nCeO2 ) into PHBV electrospun fiber membranes. In their study, wound healing in mice verified the effect of the scaffold in inhibiting
the PHBV membranes incorporated with nCeO2 had a strong potential wound bacterial population and accelerating wound healing in vivo [78].
to enhance cell proliferation and vascularization, which could promote In the field of tendon repair, Webb et al. investigated the practi-
diabetic wound healing [73,74]. cal repair effect of the PHBHHx/gel porous fiber scaffold in rat Achilles

27
W. Guo, K. Yang, X. Qin et al. Engineered Regeneration 3 (2022) 24–40

Fig. 3. Examples of PHA in skin tissue engineering applications.


(a) PHBV fibrous meshes for preventing excessive scar formation
[71]. (b) Fabrication of PHBV/keratin composite membranes for
wound healing [156]. (c) GelMA/PHBV hybrid meshes loaded
with growth factors for diabetic wound healing [74]. (d) A PhaP-
Tac modified PHBV scaffold as anti-infective wound dressings
[78].

Fig. 4. Examples of PHA in organ tissue engineering applica-


tions. (a) A PHBHHx scaffold for tendon repair in rats [79]. (b)
PHBVHHx scaffolds applied to a mouse liver-injured model [83].
The scale bars are 5 mm, respectively. (c) The morphology of
PLA/PHB scaffolds [86]. (d) In vitro differentiation experiments
of PHB/CS/nBG scaffolds [87]. (e) CS/PHBV composite hydro-
gels for nucleus pulposus tissue engineering [157].

tendon injury models (Fig. 4a). The composite scaffold showed excellent tion of PHA in organs such as liver, gastrointestinal tract, urinary tract,
tensile strength that matched the rat tendons, with full weight-bearing teeth, and pancreatic islets (Table 3). For example, Su and colleagues
and functional recovery at 20 days postoperatively. Moreover, no late- developed a PHBVHHx-based bionic scaffold loaded with mesenchymal
onset inflammatory reaction was observed at 40 days after implanta- stem cells (MSCs); and then transplanted the scaffold into liver-injured
tion, indicating the suitability of the PHBHHx scaffold for tendon repair mice (Fig. 4b). The damaged liver showed a similar structure to normal
in vivo [79]. Next, Chen et al. prepared PHB membranes modified with liver tissue after 28 days, with a significant decrease in total bilirubin
glutaraldehyde (GA), CS, dopamine (DA), and hydrogen peroxide. In (TB) and alanine aminotransferase (ALT), indicating that the PHBVHHx
the rabbit anterior cruciate ligament (ACL) model, the functionalized scaffold could promote the regeneration of damaged liver [83]. In the
PHB membrane could enhance fixation and healing between tendon field of gastrointestinal tract repair, Fan et al. constructed a composite
and bone, providing a new approach to ACL reconstruction [80]. Be- esophageal tissue engineering scaffold by using PHBHHx/poly(lactic-
sides, Tashjian et al. utilized PHA scaffolds to enhance suture interface co-glycolic acid) (PLGA) combined with decellularized porcine small
tension in rotator cuff repair by investigating the performance of PHA intestine submucosa (SIS). The mixture was successfully applied to re-
scaffolds on cyclic and fracture strength, which ultimately resulted in construct a half circumferential esophagus defect, suggesting that PHB-
more optimal mechanical support than the control group [81]. HHx is an ideal artificial esophageal substrate [84]. In addition, El-
Khordagui et al. compared the anti-adhesion potential of PHB electro-
spun nanofibrous with methylene blue (MB) cast film. This PHB/MB
3.2. Organ tissue engineering composite film effectively prevented visceral adhesions and enhanced
cecum wall healing without causing a significant inflammatory response
In organ replacement repair, biomedical engineering techniques are [85]. In the field of urethral repair, Findrik et al. successfully utilized a
used to construct bionic scaffolds that simulate the function and struc- 3D printed PLA/ PHB composite scaffold as a tubular substitute for ure-
ture of organs. Subsequently, such scaffolds are implanted in the body thral replacement (Fig. 4c). The scaffold provided stable support condi-
to replace original dysfunctional organs, potentially providing solutions tions for urethral repair in dogs by adjusting the ratio between PLA and
to some intractable diseases [82]. Recent studies reported the applica- PHB [86].

28
W. Guo, K. Yang, X. Qin et al. Engineered Regeneration 3 (2022) 24–40

Table 3
PHA in organ tissue engineering.

Type of PHA Combination Model Advantage Year Reference

PHBV iPSCs – insulin, insulin protein, Pdx-1 and Glut-2 genes were 2021 [171]
significantly improved
PHB CNTs – induced only a mild foreign body reaction and 2020 [88]
improved vascularization
PHB CS/nBG – increased expression of ALP, collagen type-I and DSPP 2018 [87]
genes
PHB PLA a dog urethral defect model a tubular urethral substitute with 2018 [86]
significant stability
PHB MB a rat cecum model prevented visceral adhesions and enhanced cecum 2016 [85]
wall healing without causing a significant
inflammatory response
PHBV gelatin – phenotypic marker proteins and ECM proteins 2014 [172]
expressions were significantly improved
PHBVHHx MSCs liver-injured mouse models similar structure of normal livers; TB and ALT 2014 [83]
significantly decreased, and ALB significantly
increased
PHBHHx PLGA/SIS full circumferential esophageal defects in rats successfully used for reconstructing esophagus 2014 [84]
PHBHHx CS/insulin a diabetic rat model the bioavailability of insulin was significantly 2013 [90]
enhanced; novel ultralong-acting insulin injections
PHBHHx/P3HB4HB PLA – construct spherical islet-like structures with 2010 [91]
significantly upregulated gene expression and
extracellular secretion of insulin
iPSCs: induced pluripotent stem cells.

In some other works, Forughi and colleagues investigated the effect well withstand radial compressive stress, radial compliance, and circum-
of PHB/CS/ nano-bioactive glass (nBG) composite fiber scaffolds on the ferential tensile stress, as well as strength maintenance during suturing.
proliferation of exfoliated milk tooth stem cells (Fig. 4d). Their results Next, cytotoxicity and hemolysis tests compared the in vitro biocompat-
indicated that the composite scaffold increased cell proliferation rates ibility of PHBHHx vascular grafts and commercial artificial vessels. The
as well as calcium and phosphorus ion uptake rates. In addition, the ex- developed scaffold could maintain the proliferation of human umbilical
pression of type I collagen and dentin sialic acid phosphoprotein (DSPP) vein endothelial cells (HUVECs) for two weeks; the scaffold could effec-
increased 6-fold, making it suitable for treating dental diseases [87]. tively prevent thrombosis directly in contact with human blood [93].
Zarei et al. prepared PHB/carbon nanotubes (CNTs) scaffolds using elec- Besides, Jayakumar and colleagues fabricated a tri-layer fiber scaffold
trospinning technology, which showed that the scaffold induced only a based on elastin nanofibers, polyvinyl alcohol (PVA) and PHBV by elec-
mild foreign body reaction and improved vascularization [88]. trostatic spinning technique (Fig. 5b and c). The in vitro biocompatibil-
In the studies of islet transplantation for diabetes, insulin delivery ity shows that MSCs, smooth muscle cells (SMCs), and HUVECs exhib-
systems mixed with phospholipids were successfully prepared based on ited high growth rates on the composite scaffolds above. These results
PHBHHx nanoparticles, which significantly enhanced the controlled re- emphasize the importance of fiber arrangement on a triple electrospun
lease of insulin and the therapeutic effect [89,90]. More interestingly, scaffold, which is essential to mimic the multilayer structure of natural
Yang et al. used stem cell technology to construct islet-like spherical vessels [94].
structures on PHBHHx membranes, with significantly upregulated gene For tissue-engineered vascular applications, hemocompatibility is
expression and extracellular secretion of insulin [91]. In summary, PHB- another important biological feature. Qu et al. compared the platelet
HHx proved to be an excellent fabrication material for islet transplan- adhesion of PHB, PHBV, and PHBHHx. Among them, PHBHHx signif-
tation. icantly reduced hemolytic activity in vitro test contacted with erythro-
cytes and significantly reduced the number of adherent platelets com-
pared with other groups [95]. Li et al. investigated the platelet adhesion
3.3. Vascular tissue engineering
of P3HB4HB and poly (propylene glycol)-poly (ethylene glycol)-poly
(propylene glycol) (PPG-PEG-PPG). The platelet adhesion number of the
Vascular grafts are in high demand for cardiovascular surgery such as
composites could be changed by modifying the ratio of P3HB4HB to
aortic replacement and coronary artery bypass graft surgery. Through
PPG-PEG-PG [96]. These studies showed good biocompatibility of PHA
the division and proliferation of endothelial cells on both sides of the
polymers in prolonged contact with blood. Besides, Arg-Gly-Asp (RGD)
anastomosis, they are interconnected with the help of vascular grafts to
peptides coating has been reported to improve the biocompatibility of
restore the original endothelial structure. However, synthetic polymer
artificial vascular patches. Thus, Viktoriia et al. evaluated the hemo-
vessels lack growth potential and have a high risk of thromboembolism,
compatibility of RGD peptide modified PCL/PHBV patches and found
stenosis, and infection [92]. In recent years, PHA has become a sub-
that the composite scaffolds could significantly promote the formation of
strate material for developing functional tissue-engineered blood ves-
new vascular systems in vivo. RGD-modified PCL/PHBV patches showed
sels (TEBV) based on its modifiable elasticity, hemocompatibility, and
milder platelet aggregation, and fewer lysed red blood cells than com-
induction of elastin formation (Table 4). For example, Antonova et al.
mercially available patches [97]. In another study, Larissa et al. incor-
tested the effect of PHBV/PCL vascular grafts containing basic fibrob-
porated VEGF and RGD peptides into small-caliber vascular grafts of
last growth factor (bFGF) and vascular endothelial growth factor (VEGF)
PHBV/PCL, which improved the graft compression strength and accel-
in a sheep carotid artery interposition model, compared with expanded
erated the formation of functional monolayer endothelial cells (Fig. 5d)
polytetrafluoroethylene (ePTFE) artificial vessels (Fig. 5a). PHBV/PCL
[98]. In addition, Cheng and colleagues further validated the induction
vascular grafts showed a more stable long-term effect than ePTFE and
of P3HB4HB films on the elastin formation of intravascular RaSMCs.
could effectively prevent aneurysm formation after 18 months [33].
Fastin elastin assay showed that 4HB monomer promoted the formation
Puppi et al. prepared PHBHHx vascular grafts with an inner diameter of
and accumulation of elastin. Benefiting from its modifiable strength,
6 mm by wet electrostatic spinning to develop small-diameter polymeric
elasticity, and ability of elastin formation, P3HB4HB could be con-
biodegradable vessels. Their results confirmed that these vessels could

29
W. Guo, K. Yang, X. Qin et al. Engineered Regeneration 3 (2022) 24–40

Table 4
PHA in vascular tissue engineering.

Type of PHA Combination Model Advantage Year Reference

PHBV PCL/VEGF/bFGF/SDF a sheep carotid artery interposition model higher primary patency, promoted vascular tissue 2021 [33]
regeneration
PHBV PCL/RGD peptides a rat abdominal aortas model facilitated the formation of neovasculature, resistance 2020 [97]
to calcification
PHBV PVA/elastin – mimicked natural arterial vascular structure 2018 [94]
PHBHHx PCL – good antithrombotic properties, sustained proliferation 2017 [93]
of HUVECs
PHBV PCL/VEGF a rat abdominal aortas model improved graft patency and accelerated monolayer 2016 [173]
endothelial cell formation
P3HB4HB PPG-PEG-PPG – controllable amount of platelet adhesion 2011 [96]
PHB collagen/CS a nude rat subcutaneous model promoted osteogenesis and vascularization 2010 [174]
P3HB4HB SMCs – ability to induce elastin formation, adjustable 2008 [99]
elasticity and strength
PHBHHx HUVECs/SMCs – enhanced vascular-related cellular affinity 2005 [95]

SDF: stromal cell-derived factor.

Fig. 5. Examples of PHA in vascular and heart valve tissue engineering appli-
cations. (a) A PHBV/PCL tissue-engineered scaffold for carotid artery repair
[33]. (b-c) The construction process of engineered vascular scaffolds based on
PHBV/PVA [94]. (d) The process of implanting PHBV/PCL grafts containing
RGD peptides into rat abdominal aortas [98]. (e) A tri-leaflet heart valve scaf-
fold based on P4HB polymer [158].

sidered as a valuable material for artificial vascular engineering [99]. could survive and produce ECM efficiently after pulsatile flow exposure
Similarly, P4HB has also been studied for the manufacture of vascular [102,103]. Subsequently, Sodian et al. inoculated sheep carotid artery
scaffolds. For example, sheep autologous pulmonary artery epithelial vascular cells (ScAVCs) onto the P4HB heart valve scaffold, which was
cells were inoculated onto a P4HB/polyglycolic acid (PGA) scaffold to implanted and replaced the original pulmonary valve in lamb models.
strengthen pulmonary arteries in vivo. The up-regulation of matrix met- The results demonstrated that the P4HB heart valve scaffold was covered
alloproteinases (MMPs) expression demonstrated the effectiveness of the by normal tissue without thrombosis, and the valve showed no signifi-
P4HB scaffold in pulmonary artery remodeling [100]. cant regurgitation up to 120 days after the operation. The pathological
data suggested that the surrounding area of the scaffold was mainly lam-
3.4. Heart valve tissue engineering inar fibrous tissue with glycosaminoglycans as ECM [104].
In addition, PHA polymers can be used as heart valve coatings or
Surgical replacement of artificial biological or mechanical valves is patches. For example, the decellularized porcine aortic valves coated
the most effective therapeutic modality for heart valve disease. How- with PHBHHx showed better resistance, increasing tensile strength and
ever, these valves lack growth and durability property, leading to throm- reducing calcification in vivo [105]. Another example is a high porosity
botic complications in patients due to irregular blood contact surfaces. tri-leaflet heart valve prepared using polyglycolic acid (PGA) and coated
Therefore, heart valve tissue engineering (HVTE) based on dynamic con- with P4HB. In this study, Perry et al. isolated and extracted BMSCs from
ditions has emerged as an important strategy for treating heart valve the sternal bone marrow of adult sheep. The cells (2 × 106 cells/mL)
diseases [101]. Recent studies have shown that PHA is an excellent sub- were delivered onto the PGA/P4HB composite scaffold to form a "tis-
strate for HVTE (Table 5). Sodian et al. fabricated a tri-leaflet heart valve sue" with biomechanical properties similar to those of the natural heart
scaffold based on P3HB4HB and inoculated it with ovarian arterial vas- valve. After implantation in sheep for 20 weeks, the diameter of the
cular cells. As made of thermoplastic PHA polymers, these valve scaf- valve scaffold increased from 19 mm to 23 mm as sheep grew up [106].
folds could be molded into the desired valve shape using stereolithogra- Their results suggest that the P4HB composite valve may have growth
phy and ensure synchronized beating of the tri-leaflet valves. The cells potential. On the one hand, due to the inherent elasticity and ductility of

30
W. Guo, K. Yang, X. Qin et al. Engineered Regeneration 3 (2022) 24–40

Table 5
PHA in heart valve tissue engineering.

Type of PHA Combination Model Advantage Year Reference

MCL-PHA PCL/CPCs the postmortem murine heart models maximize regeneration of myocardial 2018 [34]
infarction, controlled CPCs delivery
PHO NVRMs – no negative effect on cardiomyocyte 2018 [107]
contraction, enhanced cell proliferation
and adhesion
PHB PCL/PLA/PA an infarcted rat heart model induced significant angiogenesis in 2014 [108]
cardiac tissue, attenuated the
inflammatory expression of M2
macrophage phenotype
PHBV PLLA/PGS – formation of myocardial patches with a 2010 [175]
structure similar to that of native tissue
PHBHHx decellularized porcine aortic valve pulmonary position in sheep without promoted the proliferation of cells 2007 [105]
cardiopulmonary bypass associated with valve tissue and inhibited
calcification
MCL-PHA/P4HB PGA/vascular cells 2 cm defect in the pulmonary artery of provided proper lung function in lambs 2000 [104]
sheep within 120 days
CPCs: cardiac progenitor cells; NVRMs: neonatal ventricular rat myocytes; PGS: poly(glycerol sebacate); PLLA: poly(L-lactide).

P4HB could adapt to tissue growth; on the other hand, stem cells prolif- functional recovery could be observed in the host, with more axons con-
erate and differentiate on the scaffold surface, forming a homogeneous necting the neural distal stump and a reduction in muscle atrophy [113].
layer of fused cells and extracellular matrix. This newly formed valve However, the hydrophobicity, brittleness, and crystallization ten-
tissue thickens the PGA/P4HB scaffold to gradually accommodate the dency of PHB may hinder the ductility and flexibility of the conduit. To
development of cardiovascular system defects in children with congeni- eliminate these effects, Yu et al. prepared porous nerve conduits with
tal heart disease. For cardiac tissue patches, Baghdadi et al. fabricated a PHBHHx and successfully applied them to repair sciatic nerve defects
multifunctional cardiac patch of PHO, whose mechanical strength was in rats (Fig. 6d) [114]. In addition, PHBHHx showed better biocom-
close to myocardial tissue [107]. Castellano et al. compared PHB with patibility with neuronal progenitor cells (NPCs) and stem cells (NSCs);
PCL, PLA, and polyamide (PA) scaffolds for cardiac repair in infarcted and thus promoted neuronal differentiation and synaptogenesis of these
rat hearts. The PHB composite scaffold could attenuate the inflamma- cells in vitro. Wang et al. implanted NSCs-loaded PHBHHx films into a rat
tory response in cardiac tissue. Although PHB and PCL materials could traumatic brain injury (TBI) model. Their results revealed that PHBHHx
reduce negative ventricular remodeling, only PHB could induce signif- films were able to support the long-term survival of transplanted NSCs
icant angiogenesis [108]. These results suggested that PHB was more in vivo; subsequently, NSCs could migrate into damaged brain areas, dif-
beneficial for repairing and remodeling cardiac tissue. Besides, Duver- ferentiate into astrocytes and neurons without additional reactive glial
noy et al. used the PHB cardiac patch for the first time in human patients. proliferation [37]. On the other hand, Wang et al. evaluated the repair
Computerized tomography (CT) showed a 27% reduction in adhesions capacity of PHBHHX, P3HB4HB, and PHB for the central nervous sys-
between PHB patch and heart surface compared to the non-patch group tem (CNS) and their differentiation on 3D or 2D matrices. In particular,
[109]. the 3D nanofiber structure of PHBHHX could facilitate CNS synaptogen-
esis and has great potential in promoting CNS differentiation into bene-
3.5. Nerve conduit tissue engineering ficial neurons [115,116]. Sakar et al. showed that nerve grafts made
of PHBHHx with 3D oriented nanofibers significantly strengthen the
The recovery of motor, sensory and autonomic function caused by nerve regeneration, either alone or in conjunction with hMSCs [117].
peripheral nerve injury is difficult in nerve repair. Traumatic neuromas Zhang et al. incorporated poly(ethylene oxide) (PEO) into the PHBV
will form if the nerve is severed too far apart (more than 2.5 cm) or scar- nerve conduits and found that the aligned nanofiber scaffolds provided
ring between the severed ends. Currently, nerve catheter implantation topographic guidance for directed cell growth and extension, and suc-
surgery is often performed clinically for nerve repair and functional re- cessfully repairing sciatic nerve defects in rats. [118]. Their results sug-
construction [110]. The nerve sheath cells in the injured area proliferate gested that PHBV was a promising manufacturing material for neural
to form ribbon-shaped syncytial cells, and then the proximal axon grad- repair.
ually grows at a rate of about 1 mm per day through the nerve conduit.
After reaching the terminal axon, the sheath cells produce myelin to en- 3.6. Bone tissue engineering
circle the axon to form a myelin sheath. This regenerative process often
takes more than several months to complete. PHA has been extensively Bone repair is a progressive and intersecting process, usually divided
employed for nerve repair due to its biocompatibility and piezoelectric into three phases: the hematoma mechanization phase, the primary bone
properties (Table 6). For example, PHB could promote differentiation scab formation phase, the bone scab remodeling and shaping phase. Al-
of Schwarm cells (SCs) and support axonal regeneration within the host though bone tissue can regenerate naturally, large bone defects (usually
sciatic nerve (Fig. 6c) [111]. Mohanna et al. tested the function of the >2 cm) are difficult to self-healing. Bone tissue engineering provides me-
PHB/glial growth factor (GGF) conduits in bridging long nerves using chanical support to promote new bone regeneration by inducing bone-
a rabbit gastrocnemius nerve defect model. At 60 days postoperatively, associated cell growth and differentiation [119]. Table 7 summarizes
the PHB composite conduits significantly accelerated nerve regeneration the applications of PHA in bone tissue engineering in recent years. Doyle
in the defect and reinformed the motor organ. At 120 days postopera- et al. reported that PHB implants could rapidly form bone scabs around
tively, the number of SCs and axonal regeneration were higher in the bone defects, which subsequently shaped into ordered new bone tissue
PHB composite conduits group than in the blank control group [112]. In connected to the surrounding normal bone tissue. No inflammation was
another study, Schaakxs et al. inoculated PHB catheters with differenti- found at 12 months after implantation [120]. However, compared to
ated adipose-derived stem cells (dASCs) and primary SCs. Their results the natural tissue of the host, PHA lacks the sufficient bioactive func-
found that the catheter promoted rapid nerve regeneration during the tion to generate a physiological apatite coating around the material sur-
early stage of sciatic nerve repair in rats. In the later stage, a superior face. Therefore, to improve the osteogenic properties of PHA, some inor-

31
W. Guo, K. Yang, X. Qin et al. Engineered Regeneration 3 (2022) 24–40

Table 6
PHA in nerve conduit tissue engineering.

Type of PHA Combination Model Advantage Year Reference

MCL-PHA PCL 10 mm transected sciatic nerve rat model excellent neuroregenerative properties and 2021 [160]
bioresorption rates (Fig. 6b)
PHBHHx NSCs a rat traumatic brain injury model promoted the survival and proliferation of 2019 [37]
transplanted NSCs in TBI rats
PHB chitosan/hMSCs a rat sciatic nerve injury model guided damaged axons across the injured region 2018 [176]
PHBV PEO/laminin 12 mm transected sciatic nerve rat model promoted peripheral nerve regeneration 2018 [118]
PHB SCs/dASCs – greater axon myelination and less muscle atrophy 2017 [113]
PHBHHx hMSCs 10 mm nerve gaps in rats the nerve grafts with 3D oriented nanofibers guided 2014 [117]
nerve regeneration
PHBV SCs 30 mm transected sciatic nerve rat model restored nerve continuity and myelination of nerve 2014 [177]
fibers
PHB – – facilitated recovery of motor and sensory functions of 2009 [178]
peripheral nerves
PHBHHx – 10 mm transected sciatic nerve rat model rapid recovery of severed nerve function 2009 [114]
PHB SCs a rat cervical spinal cord injury model significantly promoted the survival of SCs and axonal 2008 [179]
regeneration
PHB GGF 2–4 cm peroneal nerve gaps in rabbits stimulated regeneration of long interstitial nerves 2003 [180]

Fig. 6. Examples of PHA in nerve tissue engineering applica-


tions. (a) A PHBV electrospun fiber conduit for sciatic nerve re-
pair in rats [159]. (b) Nerve guidance conduits based on MCL-
PHA/PCL blend [160]. The scale bar is 100 μm. (c) Peptide func-
tionalized PHB nanofibrous scaffolds with enhanced activity of
SCs [111]. (d) In vivo morphological changes of PHBHHx nerve
conduits [114].

ganic/organic biological materials such as hydroxyapatite (HA), bioac- vided space for the growth of MSCs and induced osteogenic differentia-
tive glass (BG), and alginate (ALG) were incorporated with PHA. As HA tion of the cells. On day 28, the hydrogel-based PHB/HA composite scaf-
contributes up to 70% of the bone matrix, incorporation of HA could folds loaded with MSCs achieved a very high regeneration rate (94%)
improve the osteoinductivity and osteoconductivity of PHB composites of radial bone defects in rat parietal bone, demonstrating the potential
[121,122]. The PHB/HA composite scaffolds could induce a stable bone of bioactive polymer scaffolds in bone tissue engineering [35].
tissue adaptation response without foreign body inflammation. In vitro PHBV, another commonly used PHA polymer, was widely mixed
studies have also shown that the PHB/HA scaffold supports the adhesion with bioactive compounds for bone tissue engineering. PHBV exhib-
and differentiation of hMSCs and human maxillary osteoblasts (HOBs). ited comparable mechanical compressive strength as human bone and
In particular, PHB/HA scaffolds coated with type I collagen effectively improved the in vitro bioactivity of the PHA scaffold. For instance, an
induced ectopic bone formation in nude mice models. After a period of animal experiment reported that the PHBV/HA composite was acceler-
implantation, high expression of osteocalcin confirmed the osteogenic ated the thickness of the new bone increasing from the initial 130 𝜇m
characteristics of the cell-seeded PHB/HA scaffolds. In contrast, no sig- to 770 𝜇m within six months, indicating this scaffold had the supe-
nificant osteocalcin expression or osteoinduction was found in the con- rior property of promoting bone structural reconstruction [127]. Adeno-
trol group [123]. On the other hand, the addition of a specific concentra- sine, another bioactive compound mixed with PHBV, can promote os-
tion of nBG to the PHB scaffold significantly improved human osteosar- teogenic differentiation of MSCs and osteoprogenitor cell. Zhong and
coma cells (MG63) proliferation, induced better osteoconductivity and colleagues systematically investigated the osteogenic ability of the com-
alkaline phosphatase (ALP) activity [124,125]. Hajiali and colleagues posite nanofibers by doping adenosine molecules in PHBV electrospun
evaluated the feasibility of using PHB/nBG nanocomposites as bone tis- nanofibers in vitro and in vivo. The adenosine-loaded PHBV electrospun
sue engineering scaffolds. Immersion of PHB/nBG scaffolds in simulated fibers exhibited strong bone regenerative potential in rabbit skull de-
body fluid (SBF) for one month could form a thin layer of HA mineraliza- fect repair [128]. Furthermore, Cool et al. compared the osteogenic
tion on its surface, which provided augmented repairability for osseoin- properties of PHBV mixed with calcined hydroxyapatite (CHA), and
tegration and bone reconstruction [126]. Bioactive osteogenic scaffolds 𝛽-tricalcium phosphate (𝛽-TCP), respectively. It was shown that the
can be prepared by mixing PHB with hydrogels; therefore, Volkov et al. nanoscale reinforced phase incorporated in PHBV improved osteogenic
fabricated ALG hydrogel-based PHB/HA scaffold by salt leaching tech- properties while decreasing the inflammatory response [129]. In ad-
nique using three-dimensional printing (Fig. 7d). The produced scaffolds dition, Chernozem et al. proposed a new scaffold that combined PHB
provided high strength support for the hydrogels; on the other hand, the and PHBV with calcium carbonate mineralized piezoelectric bodies.
large number of interconnected pore structures inside the scaffolds pro- The presence of minerals could significantly promote apatite formation

32
W. Guo, K. Yang, X. Qin et al. Engineered Regeneration 3 (2022) 24–40

Table 7
PHA in bone tissue engineering.

Type of PHA Combination Model Advantage Year Reference

PHB HA/ALG/MSCs critical size parietal bone defect in rats enhanced regeneration of critical-sized bone defects 2020 [35]
PHBV aloe vera-derived gel – higher expression of ALP, mineralized bone-related 2020 [181]
genes and proteins
PHBHHx rhBMP-2 critical sized bone defect (15 mm) in rabbits enhanced bone regeneration and facilitated 2020 [133]
vascularization
PHB BC critical-size mouse calvaria defects support preadipocytes proliferation and osteoblast 2020 [182]
differentiation, consequently inducing new bone
formation
P3HB4HB – a calvarial defect model in rats excellent biocompatibility and bone regeneration 2019 [183]
ability
PHBV adenosine critical-sized rabbit cranial defects excellent biocompatibility and osteogenic capacity 2019 [128]
PHBV PEG – activation of the ALP activity, increased gene 2019 [184]
expression of bone specific markers, and mineral
nodules formation
P3HB4HB GO critical-sized calvarial defect of rats enhanced biomechanical properties, and fast 2017 [163]
osteogenic capability (Fig. 7c)
PHBV 𝜅-CG – significant bioactive and osteogenic properties 2017 [185]
PHB – critical size parietal bone defect in rats good biocompatibility and an osteoconductive 2015 [186]
properties
PHBHHx SMC critical defect of 15 mm in length in rabbits excellent biodegradability and biocompatibility for 2014 [187]
more effective osteogenesis in vivo
PHBHHx MBG critical-size rat calvarial defects stimulated bone regeneration, improved the 2014 [132]
osteogenic activity and bone defect restoration
PHBV HA critical-sized rabbit radius defects significant repair effects on bone defects in critical 2013 [164]
areas
PHBV 𝛽-Ca2 SiO4 – stimulated the transcription of TGF-𝛽1 and BMP-7 2013 [188]
genes, promoted the activity of ALP
PHB HAP/HOBs athymic rnu/run rats induced ectopic bone formation in nude rats 2006 [123]
PHBV CaP-Gelfix femoral defect model in rats increased bone mineral density 2004 [189]

𝜅-CG: 𝜅-carrageenan; HAP: hydroxyapatite; CaP: calcium phosphate.

Fig. 7. Examples of PHA in bone tissue engineering applications. (a) Optical


image of 3D PHBV/CS porous scaffolds fabricated by laser sintering [161].
The scale bar is 500 μm. (b) A novel PHBV composite for infection treat-
ment and bone defect filling [162]. (c) Electrospun P3HB4HB/graphene oxide
(GO) scaffolds for promoting bone repair in rats [163]. (d) PHB/HA/ALG scaf-
folds seeded with MSCs enhanced the regeneration of bone defects [35]. (e)
SEM morphology and micro-CT of PHBHHx scaffolds [133]. The scale bars
are 1 mm, respectively. (f) Procedure for the preparation of implantable 3D
HA/PHBV scaffolds [164].

around PHB/PHBV scaffolds, thus stimulating bone tissue growth [130]. fect repair. Hence, Zhao et al. prepared PHBHHx/mesoporous bioactive
These studies provided a new therapeutic strategy for PHBV polymers glass (MBG) composite porous scaffolds with high compressive strength
for bone tissue regeneration. through 3D printing technology. Compared with MBG scaffolds, PHB-
Apart from PHB and PHBV, PHBHHx has been used for bone tis- HHx exhibited enhanced bioactivity and osteogenic properties, includ-
sue engineering due to its biological properties, including increased ing hBMSCs proliferation, ALP elevation, rapid apatite formation, and
ALP activity and calcium deposition. Even under in vitro culture con- enhanced expression of bone-related genes. As a result, subsequent im-
ditions, hMSCs could maintain good morphology and differentiate into plantation of PHBHHx/MBG porous scaffolds into critical-sized rat cra-
osteoblasts on PHBHHx membranes [131]. The porous structure of the nial defects could greatly promote bone regeneration [132]. Moreover,
scaffold provided sufficient space for vascular growth during bone de- Yang et al. designed three kinds of PHBHHx-based porous structure scaf-

33
W. Guo, K. Yang, X. Qin et al. Engineered Regeneration 3 (2022) 24–40

Table 8
PHA in cartilage tissue engineering.

Type of PHA Combination Model Advantage Year Reference

PHBV quercetin a cartilage defect nude mice model reduced oxidative stress in chondrocytes and inhibited 2021 [138]
apoptosis
PHB chitosan a sheep knee cartilage defect model reconstructed subchondral calcification layer and 2021 [36]
improved bone tissue integration
PHB/PHBHHx SVFCs/hASCs cartilage defects in rabbit knee joints promoted cell proliferation and migration for more 2019 [190]
desirable cartilage healing
P3HB4HB ASCs/chondrocytes cartilage defects in rabbit knee joints significant improvement in cartilage damage repair 2015 [191]
PHBHHx PhaP-RGD – promoted chondrogenic differentiation of MSCs seeded 2015 [139]
on PHBHHx films
PHBV BG cartilage defects in rabbits higher cartilage matrix content and enhanced 2014 [137]
biomechanical properties
PHBV PLCL a cartilage defect nude mice model enhanced the compressive modulus of scaffolds and 2013 [192]
cartilaginous tissue formation
PHBV meniscal cells meniscal defects in rabbit knee joints regenerative tissue with structure similar to normal 2011 [193]
meniscal fibrocartilage
PHB/PHBHHx hASCs a cartilage defect nude mice model formation of cartilage-like tissue with positive 2009 [136]
toluidine blue staining, safranin O, and collagen type-II
PHBHHx chondrocytes cartilage defects in rabbits good subchondral bone connection with surrounding 2008 [135]
chondrocyte filling
PHBHHx/PHB chondrocytes – chondrocytes were able to proliferate and maintain 2002 [194]
their phenotype within 28 days
SVFCs: stromal vascular fraction cells; PLCL: poly (l-lactide-co-caprolactone).

Fig. 8. Examples of PHA in cartilage tissue engineering applications. (a) Eval-


uation of 3D scaffolds prepared from PHBHHx for cartilage repair in rab-
bits [135]. The scale bars are 4 mm, respectively. (b) The morphology of
hASCs/PHB/PHBHHx constructs after subcutaneous implantation [136]. The
scale bars are 1 cm, respectively. (c) A CHIT/PHB scaffold for treating osteo-
chondral defects [36].

folds and investigated the effect of hierarchical porous scaffolds on HU- cally created on the femoral patellar groove of the knee joints and filled
VECs (Fig. 7e). The PHBHHx scaffolds with large micropores were ver- with a PHBHHx cartilage scaffold in the lower defect area. Due to the
ified to promote vascularization and bone regeneration through rabbit pre-seeded chondrocytes in the scaffold, the engineered cartilage tis-
bone defects [133]. sue achieved better surface integrity and more ECM accumulation than
the control group, which demonstrates that PHBHHx could be an ex-
3.7. Cartilage tissue engineering cellent material for cartilage tissue engineering [135]. To investigate
the potential value of PHBHHx/PHB, Ye et al. inoculated hASCs onto
Unlike bone tissue, cartilage is poorly self-repairing because of the PHBHHx/PHB porous scaffolds and implanted them into subcutaneous
lack of vascular and lymphatic distribution, and fibrous tissue is of- layers of nude mice. Six months after implantation, significant cartilage-
ten involved in repairing larger defects. Traditional surgical procedures like tissue could be observed in the composite scaffold group, with pos-
such as joint lavage cleanup, microfracture, and cartilage grafting can- itive staining for type II collagen, saffron O, and toluidine blue (Fig. 8b)
not overcome the functional deficits caused by cartilage tissue injury. [136]. Zhou et al. inoculated rabbit chondrocytes on the PHBV/BG com-
In recent years, cartilage tissue engineering techniques have provided posite scaffold and could observe the same repair effect in a rabbit car-
new ideas for cartilage repair and regeneration. Table 8 summarizes the tilage defect repair model [137].
applications of PHA polymers in cartilage tissue engineering. Deng et al. On the other hand, to address the poor bioactivity of PHBV, Chen
inoculated rabbit articular chondrocytes on a PHBHHx/PHB porous scaf- et al. prepared a PHBV fibrous scaffold grafted with quercetin (QUE)
fold, which showed better growth than pure PHB. The level of type II by surface modification method. This composite scaffold significantly
collagen anchored on the scaffold was significantly increased through- promoted cartilage regeneration by regulating relevant gene transcrip-
out the culture cycle (collagen II can be used as a marker of chon- tion, reducing chondrocyte oxidative stress, and inhibiting chondrocyte
drocyte differentiation and maturation), indicating that PHBHHx/PHB apoptosis [138]. Li et al. coated the surface of PHBHHx with arginine
obviously promoted chondrocytes differentiation [134]. Subsequently, glycosyl aspartate RGD peptides as well as PhaP. In vitro cellular exper-
PHBHHx/PHB/hASCs composite scaffolds were implanted into a rabbit iments demonstrated that the PHBHHx scaffold coated with PhaP-RGD
cartilage defect model (Fig. 8a). The arrows point to the defects surgi- could promote adhesion, proliferation, and chondrogenic differentiation

34
W. Guo, K. Yang, X. Qin et al. Engineered Regeneration 3 (2022) 24–40

of hMSCs [139]. In addition, biopolymers based on decellularized chi- of PHA fermentation, the instruments for fermentation can only pro-
tosan (CHIT) have been effective in treating osteochondral defects and vided by a few manufactures and require high production conditions.
traumatic cartilage. A sheep knee cartilage defect repair model con- In addition, the technical barriers to PHA fermentation are high and
firmed the superior repair effect of porous CHIT/PHB scaffolds (Fig. 8c) usually rely on professionals for production and monitoring. From an
[36]. Ching et al. prepared PHB/PHO composite scaffolds with different economic point of view, the market competitiveness of PHA is insuf-
mixing ratios. When the ratio of PHB to PHO was 1:0.25, the composite ficient. Currently, numerous studies are attempting to reduce the cost
scaffolds matched the stiffness of natural articular cartilage, and the ori- of PHA fermentation processes and to improve the extraction of indus-
ented fibrous structure mimicked the collagen fiber network in cartilage trial processes by appropriate methods. More importantly, the physi-
tissue. Their results suggested that these scaffolds were expected to be cal properties of PHA have not exceeded or even reached the level of
used to repair cartilage tissue in clinical practice and reduce the risk of existing plastic products. The number of PHA monomers exceeds 150,
secondary osteoarthritis [140]. making it difficult for fermentation products to maintain complete con-
sistency with each other. The inherent physical aging, embrittlement,
slow crystallization, and low nucleation density limit its extension in
3.8. Others
many practical applications. Therefore, it is necessary to further con-
trol the monomer structure and properties of PHA through metabolic
In other fields, PHA has been used to manufacture medical sutures
engineering to improve the applicability of PHA and its derivative ma-
with sufficient strength to make them suitable for hemostasis, wound
terials. These fermentation and modification technologies are technical
healing, and ligature tissue attachment. After being implanted into ex-
hurdles that must be overcome in the future development of PHA. In
perimental animals, monofilament sutures made of PHB and PHBV
addition, there are no standards or policy support for technical review
showed significantly lower inflammatory and other reactions than those
guidelines of PHA production and application. In the future, with the
of conventional intestinal sutures. Shishatskaya et al. compared the re-
growing problem of "white plastic" and the promotion of the concept of
sponse of PHB and PHBV sutures to surrounding tissues, and found that
sustainable development, the acceptance of bioplastics by society will
both had enough required strength for suturing myofascial wounds; no
increase, which is conducive to the market of PHA and biodegradable
adverse effects such as necrosis, septic inflammation, or fibrocystic cal-
bioplastics industry.
cification were observed during up to one year of implantation [38,141].
In recent years, PHA has been applied in more and more fields, in-
In addition, PHBHHx exhibited better biocompatibility and hemocom-
cluding biological medicine, daily chemical, food packaging, agricul-
patibility compared with PHB and PHBV. Its excellent fracture strength
ture, and other areas. Among them, the biomedical field is undoubt-
and flexibility made the use of PHBHHx as a medical suture more fa-
edly the focal point for the future production and application of PHA.
vorable. He et al. prepared two types of fibers: a multifilament made
Therefore, innovative purification technologies such as synthetic bi-
of PHBV/PLA and a monofilament made of PHBHHx. The mechani-
ology and bioreactor system design, are urgently needed to remove
cal properties, biocompatibility, and degradability of these two fibers
lipopolysaccharides (LPS), proteins and other bacterial impurities re-
were evaluated. It was found that these fibers maintained a high ten-
maining from the fermentation process; and to ensure the stability of
sile strength even after one year of implantation [142]. PHA fibers have
molecular weight, composition, and purity of PHA polymers. Over-
great potential for tissue suturing because they can degrade into bene-
all, the unique properties such as elastic mechanical properties, ther-
ficial 3-HB monomers and reduce scar tissue formation.
mal properties, biocompatibility, biodegradability, nontoxic degrada-
Moreover, highly open porous microspheres with PHBHHx could
tion products, and non-carcinogenic effects make PHA more attractive
also be synthesized as injectable cell-laden scaffolds to protect cells from
and applicable than other biopolymers. In addition, PHA can be modi-
stress during injection and safely transport cells to defect tissues [143].
fied by blending with other organic or inorganic materials to meet the
PHA can also be used as carriers for drug delivery, such as antibiotics
needs of various applications. Therefore, PHA is widely used in biomed-
[144,145], anticancer drugs [146,147] and hormones [148,149]. Zhu
ical engineering, especially in tissue repair and regeneration, includ-
et al. prepared composite scaffolds of MBG and PHBHHx for the treat-
ing wound dressings, artificial blood vessels, heart valves, nerve con-
ment of osteoarthritic tuberculosis by three-dimensional printing tech-
duits, bone and cartilage scaffolds, surgical sutures, etc. However, only
nology. High doses of isoniazid (INH)/rifampicin (RFP) antituberculosis
an exceptionally few commercialized PHA products have been applied
drugs were preloaded into the chemically modified MBG. The compos-
to clinics, and the vast majority of research is still at the experimental
ite scaffold showed long-lasting drug release in vitro. In a rabbit femoral
stage with many technical hurdles. In the short-term future, given the
defect model, the antitubercular drug loading scaffold could achieve in-
high production cost of PHA, the direction of product development will
tratubular new bone production and long-lasting antitubercular effects
mainly focus on high-value medical consumables such as interventional
[150]. In the treatment of chronic osteomyelitis, PHBV and P3HB4HB
valves, artificial blood vessels, and artificial bone joints. By capturing
biopolymer rods loaded with antibiotics (Duocid, Sulfadiazine) were
the high-end medical market, it will catch up with other degradable
manufactured to prevent and treat associated infections [151]. More-
polymer materials in terms of research and development technologies.
over, 3HB and its derivative methyl 3-hydroxybutyrate (3HBME) could
In the long-term development trend, as the production cost of PHA de-
promote osteoblast growth and differentiation by regulating rapid cal-
creases, the product development direction will be extended to low-
cium deposition and serum ALP activity, leading to the prevention of os-
value medical consumables such as wound dressings and surgical su-
teoporosis [152,153]. Similarly, the PHBV/PLGA nanoparticles loaded
tures. With the performance advantages of PHA, it is further expected
with the recombinant protein teriparatide exhibit a stable and syner-
to achieve comprehensive coverage of the medical device market. In the
gistic effect on promoting bone resorption and have been used in the
future, with the benefits of medical-engineering multidisciplinary inter-
treatment of osteoporosis [154,155].
section, promoting and strengthening PHA research in personalized and
precise tissue repair and regeneration could provide new strategies for
4. Prospects and conclusion developing high-performance biomaterials.

PHA is a highly promising alternative to petroleum-derived plastics Author contributions


as a degradable biopolymer. Since PHA fermenting bacteria were first
identified, significant breakthroughs have been made in applying PHA, W.T.G. conducted conceptualization, data curation, and original
but its high production costs and poor processability hinder the commer- draft; K.L.Y. conducted data curation and formal analysis; X.S.Q. con-
cialization. According to statistics, the production cost of PHA is about ducted software and visualization; L.R. conducted investigation and
ten times higher than that of petroleum-derived plastics. In the process validation; H.W. conducted supervision, funding acquisition, and re-

35
W. Guo, K. Yang, X. Qin et al. Engineered Regeneration 3 (2022) 24–40

sources; R.K.H. conducted project administration, methodology, and re- [18] A.K. Singh, N. Mallick, Advances in cyanobacterial polyhydroxyalkanoates produc-
view & editing. tion, FEMS Microbiol. Lett. 364 (20) (2017) fnx189.
[19] M. Koller, Cyanobacterial polyhydroxyalkanoate production: status quo and quo
vadis? CBIOT 4 (4) (2016) 464–480.
Declaration of Competing Interest [20] M. Higuchi-Takeuchi, K. Morisaki, K. Toyooka, K. Numata, Synthesis of high–
molecular-weight polyhydroxyalkanoates by marine Photosynthetic Purple Bacte-
ria, PLoS One 11 (8) (2016) e0160981.
We declare that we have no financial and personal relationships with [21] B. Wang, S. Pugh, D.R. Nielsen, W. Zhang, D.R. Meldrum, Engineering cyanobacte-
other people or organizations that can inappropriately influence our ria for photosynthetic production of 3-hydroxybutyrate directly from CO2, Metab.
Eng. 16 (2013) 68–77.
work, there is no professional or other personal interest of any nature [22] M. Koller, Polyhydroxyalkanoate biosynthesis at the edge of water Activitiy-Haloar-
or kind in any product, service, and/or company that could be con- chaea as biopolyester factories, Bioengineering 6 (2) (2019) E34.
strued as influencing the position presented in, or the review of, the [23] C. Hermann-Krauss, M. Koller, A. Muhr, H. Fasl, F. Stelzer, G. Braunegg, Archaeal
production of polyhydroxyalkanoate (PHA) co- and terpolyesters from biodiesel
manuscript entitled, “Polyhydroxyalkanoates in tissue repair and re- industry-derived by-products, Archaea 2013 (2013) 129268.
generation”. [24] V. Martínez, C. Herencias, E. Jurkevitch, M.A. Prieto, Engineering a predatory bac-
terium as a proficient killer agent for intracellular bio-products recovery: the case
of the polyhydroxyalkanoates, Sci. Rep. 6 (2016) 24381.
Acknowledgments [25] S.J. Park, T.W. Kim, M.K. Kim, S.Y. Lee, S.C. Lim, Advanced bacterial polyhydrox-
yalkanoates: towards a versatile and sustainable platform for unnatural tailor-made
polyesters, Biotechnol. Adv. 30 (6) (2012) 1196–1206.
This work was supported by the National Key Research and
[26] A. Rodriguez-Contreras, Recent advances in the use of polyhydroyalkanoates in
Development Program of China (2021YFC2101700), and the Sun biomedicine, Bioengineering 6 (3) (2019) E82.
Yat-sen University Clinical Research 5010 Program (No. 2017008). [27] R. Durner, M. Zinn, B. Witholt, T. Egli, Accumulation of
poly[(R)-3-hydroxyalkanoates] in Pseudomonas oleovorans during growth in
Part of the materials for Fig. 2 were required from Servier
batch and chemostat culture with different carbon sources, Biotechnol. Bioeng. 72
Medical Art (http://smart.servier.com/), licensed under a Creative (3) (2001) 278–288.
Common Attribution 3.0 Generic License (https://creativecommons. [28] H.Y. Sagong, H.F. Son, S.Y. Choi, S.Y. Lee, K.J. Kim, Structural insights into poly-
org/licenses/by/3.0/). hydroxyalkanoates biosynthesis, Trends Biochem. Sci 43 (10) (2018) 790–805.
[29] A.K. Singh, J.K. Srivastava, A.K. Chandel, L. Sharma, N. Mallick, S.P. Singh,
Biomedical applications of microbially engineered polyhydroxyalkanoates: an in-
References sight into recent advances, bottlenecks, and solutions, Appl. Microbiol. Biotechnol.
103 (5) (2019) 2007–2032.
[1] V. Tournier, C.M. Topham, A. Gilles, B. David, C. Folgoas, E. Moya-Leclair, [30] M. Koller, Chemical and biochemical engineering approaches in manufacturing
E. Kamionka, M.L. Desrousseaux, H. Texier, S. Gavalda, M. Cot, E. Guémard, M. Dal- polyhydroxyalkanoate (PHA) biopolyesters of tailored structure with focus on the
ibey, J. Nomme, G. Cioci, S. Barbe, M. Chateau, I. André, S. Duquesne, A. Marty, diversity of building blocks, Chem. Biochem. Eng. Q. 32 (4) (2019) 413–438.
An engineered PET depolymerase to break down and recycle plastic bottles, Nature [31] G.Q. Chen, I. Hajnal, The “PHAome, Trends Biotechnol. 33 (10) (2015) 559–564.
580 (7802) (2020) 216–219. [32] A.S. Arampatzis, K. Giannakoula, K.N. Kontogiannopoulos, K. Theodoridis,
[2] S.A. Park, H. Jeon, H. Kim, S.H. Shin, S. Choy, D.S. Hwang, J.M. Koo, J. Jegal, E. Aggelidou, A. Rat, E. Kampasakali, A. Willems, D. Christofilos, A. Kritis, V.P. Pa-
S.Y. Hwang, J. Park, D.X. Oh, Sustainable and recyclable super engineering ther- pageorgiou, I. Tsivintzelis, A.N. Assimopoulou, Novel electrospun poly-hydroxybu-
moplastic from biorenewable monomer, Nat. Commun. 10 (1) (2019) 2601. tyrate scaffolds as carriers for the wound healing agents alkannins and shikonins,
[3] Y. Yu, Q. Wang, C. Wang, L. Shang, Living materials for regenerative medicine, Regen. Biomater. 8 (3) (2021) rbab011.
Eng. Regener. 2 (2021) 96–104. [33] L.V. Antonova, E.O. Krivkina, V.V. Sevostianova, A.V. Mironov, M.A. Rezvova,
[4] S.Y. Choi, I.J. Cho, Y. Lee, Y.J. Kim, K.J. Kim, S.Y. Lee, Microbial polyhydroxyalka- A.R. Shabaev, V.O. Tkachenko, S.S. Krutitskiy, M.Y. Khanova, T.Y. Sergeeva,
noates and nonnatural polyesters, Adv. Mater. 32 (35) (2020) e1907138. V.G. Matveeva, T.V. Glushkova, A.G. Kutikhin, R.A. Mukhamadiyarov, N.S. Deeva,
[5] A. Surendran, M. Lakshmanan, J.Y. Chee, A.M. Sulaiman, D.V. Thuoc, K. Sudesh, T.N. Akentieva, M.Y. Sinitsky, E.A. Velikanova, L.S. Barbarash, Tissue-engineered
Can polyhydroxyalkanoates be produced efficiently from waste plant and animal carotid artery interposition grafts demonstrate high primary patency and promote
oils? Front. Bioeng. Biotechnol. 8 (2020) 169. vascular tissue regeneration in the ovine model, Polymers (Basel) 13 (16) (2021)
[6] S. Shahid, S. Razzaq, R. Farooq, Z.I.H. Nazli, Polyhydroxyalkanoates: next gener- 2637.
ation natural biomolecules and a solution for the world’s future economy, Int. J. [34] C. Constantinides, P. Basnett, B. Lukasiewicz, R. Carnicer, E. Swider, Q.A. Majid,
Biol. Macromol. 166 (2021) 297–321. M. Srinivas, C.A. Carr, I. Roy, In vivo tracking and 1H/19F magnetic resonance
[7] S. Ansari, N. Sami, D. Yasin, N. Ahmad, T. Fatma, Biomedical applications of en- imaging of biodegradable polyhydroxyalkanoate/polycaprolactone blend scaffolds
vironmental friendly poly-hydroxyalkanoates, Int. J. Biol. Macromol. 183 (2021) seeded with labeled cardiac stem cells, ACS Appl. Mater. Interfaces 10 (30) (2018)
549–563. 25056–25068.
[8] S. Pradhan, P.K. Dikshit, V.S. Moholkar, Production, ultrasonic extraction, and [35] A.V. Volkov, A.A. Muraev, I.I. Zharkova, V.V. Voinova, E.A. Akoulina,
characterization of poly (3-hydroxybutyrate) (PHB) using Bacillus megaterium and V.A. Zhuikov, D.D. Khaydapova, D.V. Chesnokova, K.A. Menshikh, A.A. Dudun,
Cupriavidus necator, Polym. Adv. Technol. 29 (8) (2018) 2392–2400. T.K. Makhina, G.A. Bonartseva, T.F. Asfarov, I.A. Stamboliev, Y.V. Gazhva,
[9] B.P. Aneesh, J.K. Arjun, T. Kavitha, K. Harikrishnan, Production of short chain V.M. Ryabova, L.H. Zlatev, S.Y. Ivanov, K.V. Shaitan, A.P. Bonartsev,
length polyhydroxyalkanoates by Bacillus megaterium PHB29 from starch feed Poly(3-hydroxybutyrate)/hydroxyapatite/alginate scaffolds seeded with mes-
stock, Int. J. Curr. Microbiol. App. Sci. 5 (7) (2016) 816–823. enchymal stem cells enhance the regeneration of critical-sized bone defect, Mater.
[10] S.Y. Li, C.L. Dong, S.Y. Wang, H.M. Ye, G.Q. Chen, Microbial production of polyhy- Sci. Eng. C Mater. Biol. Appl. 114 (2020) 110991.
droxyalkanoate block copolymer by recombinant Pseudomonas putida, Appl. Mi- [36] E. Petrovova, M. Tomco, K. Holovska, J. Danko, L. Kresakova, K. Vdoviakova,
crobiol. Biotechnol. 90 (2) (2011) 659–669. V. Simaiova, F. Kolvek, P. Hornakova, T. Toth, J. Zivcak, P. Gal, D. Sedmera, L. Lup-
[11] V. Martínez, P. García, J.L. García, M.A. Prieto, Controlled autolysis facilitates the takova, L. Medvecky, PHB/CHIT scaffold as a promising biopolymer in the treat-
polyhydroxyalkanoate recovery in Pseudomonas putida KT2440, Microb. Biotech- ment of osteochondral defects-an experimental animal study, Polymers (Basel) 13
nol. 4 (4) (2011) 533–547. (8) (2021) 1232.
[12] K.H. Berwig, C. Baldasso, A. Dettmer, Production and characterization of [37] L. Wang, Z. Yang, F. Fan, S. Sun, X. Wu, H. Lu, X. Lu, PHBHHx facilitated the res-
poly(3-hydroxybutyrate) generated by Alcaligenes latus using lactose and whey idence, survival and stemness maintain of transplanted neural stem cells in trau-
after acid protein precipitation process, Bioresour. Technol. 218 (2016) 31–37. matic brain injury rats, Biomacromolecules 20 (9) (2019) 3294–3302.
[13] G.G. Choi, H.W. Kim, Y.H. Rhee, Enzymatic and non-enzymatic degradation of [38] E.I. Shishatskaya, T.G. Volova, A.P. Puzyr, O.A. Mogilnaya, S.N. Efremov, Tissue
poly (3-hydroxybutyrate-co-3-hydroxyvalerate) copolyesters produced by Alcali- response to the implantation of biodegradable polyhydroxyalkanoate sutures, J.
genes sp. MT-16, J. Microbiol. 42 (4) (2004) 346–352. Mater. Sci. Mater. Med. 15 (6) (2004) 719–728.
[14] L.F. Qin, X. Gao, Q. Liu, Q. Wu, G.Q. Chen, Biosynthesis of polyhydroxyalkanoate [39] A. Pryadko, M.A. Surmeneva, R.A. Surmenev, Review of hybrid materials based
copolyesters by Aeromonas hydrophila mutant expressing a low-substrate-speci- on polyhydroxyalkanoates for tissue engineering applications, Polymers (Basel) 13
ficity PHA synthase PhaC2Ps, Biochem. Eng. J. 37 (2) (2007) 144–150. (11) (2021) 1738.
[15] T. Fukui, N. Shiomi, Y. Doi, Expression and characterization of (R)-specific [40] M.E. Grigore, R.M. Grigorescu, L. Iancu, R.M. Ion, C. Zaharia, E.R. Andrei, Methods
enoyl coenzyme A hydratase involved in polyhydroxyalkanoate biosynthesis by of synthesis, properties and biomedical applications of polyhydroxyalkanoates: a
Aeromonas caviae, J. Bacteriol. 180 (3) (1998) 667–673. review, J. Biomater. Sci. Polym. Ed. 30 (9) (2019) 695–712.
[16] E.F. Gómez, F.C. Michel, Biodegradability of conventional and bio-based plastics [41] K. Numata, H. Abe, T. Iwata, Biodegradability of poly(hydroxyalkanoate) materi-
and natural fiber composites during composting, anaerobic digestion and long-term als, Materials (Basel) 2 (3) (2009) 1104–1126.
soil incubation, Polym. Degrad. Stab. 98 (12) (2013) 2583–2591. [42] D. Jendrossek, R. Handrick, Microbial degradation of polyhydroxyalkanoates,
[17] F. Cerrone, S.K. Choudhari, R. Davis, D. Cysneiros, V. O’Flaherty, G. Duane, Annu. Rev. Microbiol. 56 (2002) 403–432.
E. Casey, M.W. Guzik, S.T. Kenny, R.P. Babu, K. O’Connor, Medium chain length [43] T.G. Volova, A.N. Boyandin, A.D. Vasiliev, V.A. Karpov, S.V. Prudnikova,
polyhydroxyalkanoate (mcl-PHA) production from volatile fatty acids derived O.V. Mishukova, U.A. Boyarskikh, M.L. Filipenko, V.P. Rudnev, B. Bá Xuân, V. Việt
from the anaerobic digestion of grass, Appl. Microbiol. Biotechnol. 98 (2) (2014) Dũng, I.I. Gitelson, Biodegradation of polyhydroxyalkanoates (PHAs) in tropical
611–620.

36
W. Guo, K. Yang, X. Qin et al. Engineered Regeneration 3 (2022) 24–40

coastal waters and identification of PHA-degrading bacteria, Polym. Degrad. Stab. brous P(3HB-co-4HB)/collagen peptides construct as potential leave-on wound
95 (12) (2010) 2350–2359. dressing, Mater. Sci. Eng. C Mater. Biol. Appl. 66 (2016) 147–155.
[44] K. Elbanna, T. Lütke-Eversloh, D. Jendrossek, H. Luftmann, A. Steinbüchel, Studies [71] H.S. Kim, J. Chen, L.P. Wu, J. Wu, H. Xiang, K.W. Leong, J. Han, Prevention of
on the biodegradability of polythioester copolymers and homopolymers by poly- excessive scar formation using nanofibrous meshes made of biodegradable elas-
hydroxyalkanoate (PHA)-degrading bacteria and PHA depolymerases, Arch. Micro- tomer poly(3-hydroxybutyrate-co-3-hydroxyvalerate), J. Tissue Eng. 11 (2020)
biol. 182 (2–3) (2004) 212–225. 2041731420949332.
[45] X.H. Qu, Q. Wu, K.Y. Zhang, G.Q. Chen, In vivo studies of [72] C. Sanhueza, J. Hermosilla, A. Bugallo-Casal, A. Da Silva-Candal, C. Taboada,
poly(3-hydroxybutyrate-co-3-hydroxyhexanoate) based polymers: biodegradation R. Millán, A. Concheiro, C. Alvarez-Lorenzo, F. Acevedo, One-step electrospun scaf-
and tissue reactions, Biomaterials 27 (19) (2006) 3540–3548. fold of dual-sized gelatin/poly-3-hydroxybutyrate nano/microfibers for skin regen-
[46] X. Tang, A.H. Westlie, L. Caporaso, L. Cavallo, L. Falivene, E.Y.X. Chen, Biodegrad- eration in diabetic wound, Mater. Sci. Eng. C Mater. Biol. Appl. 119 (2021) 111602.
able polyhydroxyalkanoates by stereoselective copolymerization of racemic di- [73] R. Augustine, A. Hasan, N.K. Patan, Y.B. Dalvi, R. Varghese, A. Antony, R.N. Unni,
olides: stereocontrol and polyolefin-like properties, Angew. Chem. Int. Ed Engl. N. Sandhyarani, A.E.A. Moustafa, Cerium oxide nanoparticle incorporated elec-
59 (20) (2020) 7881–7890. trospun poly(3-hydroxybutyrate-co-3-hydroxyvalerate) membranes for diabetic
[47] Q. Wu, Y. Wang, G.Q. Chen, Medical application of microbial biopolyesters poly- wound healing applications, ACS Biomater. Sci. Eng. 6 (1) (2020) 58–70.
hydroxyalkanoates, Artif. Cells Blood. Substit. Immobil. Biotechnol. 37 (1) (2009) [74] R. Augustine, A. Hasan, Y.B. Dalvi, S.R.U. Rehman, R. Varghese, R.N. Unni,
1–12. H.C. Yalcin, R. Alfkey, S. Thomas, A.E. Al Moustafa, Growth factor loaded
[48] C.S. Wu, Mechanical properties, biocompatibility, and biodegradation of in situ photocrosslinkable poly(3-hydroxybutyrate-co-3-hydroxyvalerate)/gelatin
cross-linked cellulose acetate-reinforced polyester composites, Carbohydr. Polym. methacryloyl hybrid patch for diabetic wound healing, Mater. Sci. Eng. C Mater.
105 (2014) 41–48. Biol. Appl. 118 (2021) 111519.
[49] Y.J. Hu, X. Wei, W. Zhao, Y.S. Liu, G.Q. Chen, Biocompatibility of [75] M. Rofeal, F.A. El-Malek, X. Qi, In vitroassessment of green polyhydroxybu-
poly(3-hydroxybutyrate-co-3-hydroxyvalerate-co-3-hydroxyhexanoate) with tyrate/chitosan blend loaded with kaempferol nanocrystals as a potential dressing
bone marrow mesenchymal stem cells, Acta Biomater. 5 (4) (2009) 1115–1125. for infected wounds, Nanotechnology 32 (37) (2021) abf7ee.
[50] S.L. Ang, B. Shaharuddin, J.A. Chuah, K. Sudesh, Electrospun [76] A. Marcano, N. Bou Haidar, S. Marais, J.M. Valleton, A.C. Duncan, Designing
poly(3-hydroxybutyrate-co-3-hydroxyhexanoate)/silk fibroin film is a promising biodegradable PHA-based 3D scaffolds with antibiofilm properties for wound dress-
scaffold for bone tissue engineering, Int. J. Biol. Macromol. 145 (2020) 173–188. ings: optimization of the microstructure/nanostructure, ACS Biomater. Sci. Eng. 3
[51] S. Rathbone, P. Furrer, J. Lübben, M. Zinn, S. Cartmell, Biocompatibility of polyhy- (12) (2017) 3654–3661.
droxyalkanoate as a potential material for ligament and tendon scaffold material, [77] K. Ariga, Nanoarchitectonics revolution and evolution: from small science to big
J. Biomed. Mater. Res. A 93 (4) (2010) 1391–1403. technology, Small Sci. 1 (1) (2021) 2000032.
[52] B. Egan, D.P. D’Agostino, Fueling performance: ketones enter the mix, Cell Metab. [78] Q. Xue, X.B. Liu, Y.H. Lao, L.P. Wu, D. Wang, Z.Q. Zuo, J.Y. Chen, J. Hou, Y.Y. Bei,
24 (3) (2016) 373–375. X.F. Wu, K.W. Leong, H. Xiang, J. Han, Anti-infective biomaterials with surface-dec-
[53] T. Shimazu, M.D. Hirschey, J. Newman, W. He, K. Shirakawa, N. Le Moan, orated tachyplesin I, Biomaterials 178 (2018) 351–362.
C.A. Grueter, H. Lim, L.R. Saunders, R.D. Stevens, C.B. Newgard, R.V. Farese, [79] W.R. Webb, T.P. Dale, A.J. Lomas, G. Zeng, I. Wimpenny, A.J. El Haj, N.R. Forsyth,
R. de Cabo, S. Ulrich, K. Akassoglou, E. Verdin, Suppression of oxidative stress G.Q. Chen, The application of poly(3-hydroxybutyrate-co-3-hydroxyhexanoate)
by 𝛽-hydroxybutyrate, an endogenous histone deacetylase inhibitor, Science 339 scaffolds for tendon repair in the rat model, Biomaterials 34 (28) (2013)
(6116) (2013) 211–214. 6683–6694.
[54] X.Q. Xiao, Y. Zhao, G.Q. Chen, The effect of 3-hydroxybutyrate and its derivatives [80] W.C. Chen, C.H. Chen, H.W. Tseng, Y.W. Liu, Y.P. Chen, C.H. Lee,
on the growth of glial cells, Biomaterials 28 (25) (2007) 3608–3616. Y.J. Kuo, C.H. Hsu, Y.M. Sun, Surface functionalized electrospun fibrous
[55] X.H. Zou, H.M. Li, S. Wang, M. Leski, Y.C. Yao, X.D. Yang, Q.J. Huang, G.Q. Chen, poly(3-hydroxybutyrate) membranes and sleeves: a novel approach for fixation
The effect of 3-hydroxybutyrate methyl ester on learning and memory in mice, in anterior cruciate ligament reconstruction, J. Mater. Chem. B 5 (3) (2017)
Biomaterials 30 (8) (2009) 1532–1541. 553–564.
[56] S.W. Peng, X.Y. Guo, G.G. Shang, J. Li, X.Y. Xu, M.L. You, P. Li, G.Q. Chen, An [81] R.Z. Tashjian, C.W. Kolz, T. Suter, H.B. Henninger, Biomechanics of polyhydrox-
assessment of the risks of carcinogenicity associated with polyhydroxyalkanoates yalkanoate mesh-augmented single-row rotator cuff repairs, Am. J. Orthop. 45 (7)
through an analysis of DNA aneuploid and telomerase activity, Biomaterials 32 (2016) E527–E533.
(10) (2011) 2546–2555. [82] F. Zheng, F. Fu, Y. Cheng, C. Wang, Y. Zhao, Z. Gu, Organ-on-a-chip systems: mi-
[57] C. Yan, Y. Wang, X.Y. Shen, G. Yang, J. Jian, H.S. Wang, G.Q. Chen, Q. Wu, Mi- croengineering to biomimic living systems, Small 12 (17) (2016) 2253–2282.
croRNA regulation associated chondrogenesis of mouse MSCs grown on polyhy- [83] Z. Su, P. Li, B. Wu, H. Ma, Y. Wang, G. Liu, H. Zeng, Z. Li, X. Wei, PHBVHHx
droxyalkanoates, Biomaterials 32 (27) (2011) 6435–6444. scaffolds loaded with umbilical cord-derived mesenchymal stem cells or hepato-
[58] J. Tao, C. Song, M. Cao, D. Hu, L. Liu, N. Liu, S. Wang, Thermal prop- cyte-like cells differentiated from these cells for liver tissue engineering, Mater.
erties and degradability of poly(propylene carbonate)/poly(𝛽-hydroxybutyrate- Sci. Eng. C Mater. Biol. Appl. 45 (2014) 374–382.
co-𝛽-hydroxyvalerate) (PPC/PHBV) blends, Polym. Degrad. Stab. 94 (4) (2009) [84] M.R. Fan, M. Gong, L.C. Da, L. Bai, X.Q. Li, K.F. Chen, J.L. L.ing, Z.M. Yang,
575–583. H.Q. Xie, Tissue engineered esophagus scaffold constructed with porcine small in-
[59] K. Nagase, R. Shukuwa, H. Takahashi, N. Takeda, T. Okano, Enhanced mechani- testinal submucosa and synthetic polymers, Biomed. Mater. 9 (1) (2014) 015012.
cal properties and cell separation with thermal control of PIPAAm-brushed poly- [85] N. El-Sayed, S. Galal, H. El-Gowelli, L. El-Khordagui, Inhibition of postsurgical ad-
mer-blend microfibers, J. Mater. Chem. B 8 (28) (2020) 6017–6026. hesions by methylene blue-loaded nanofibers versus cast film matrices, J. Biomater.
[60] J.A. Chuah, M. Yamada, S. Taguchi, K. Sudesh, Y. Doi, K. Numata, Biosynthesis and Sci. Polym. Ed. 27 (10) (2016) 1029–1044.
characterization of polyhydroxyalkanoate containing 5-hydroxyvalerate units: ef- [86] A. Findrik Balogová, R. Hudák, T. Tóth, M. Schnitzer, J. Feranc, D. Bakoš, J. Živčák,
fects of 5HV units on biodegradability, cytotoxicity, mechanical and thermal prop- Determination of geometrical and viscoelastic properties of PLA/PHB samples
erties, Polym. Degrad. Stab. 98 (1) (2013) 331–338. made by additive manufacturing for urethral substitution, J. Biotechnol. 284
[61] S. Gopi, M. Kontopoulou, B.A. Ramsay, J.A. Ramsay, Manipulating the structure of (2018) 123–130.
medium-chain-length polyhydroxyalkanoate (MCL-PHA) to enhance thermal prop- [87] M. Khoroushi, M.R. Foroughi, S. Karbasi, B. Hashemibeni, A.A. Khademi, Effect
erties and crystallization kinetics, Int. J. Biol. Macromol. 119 (2018) 1248–1255. of Polyhydroxybutyrate/Chitosan/Bioglass nanofiber scaffold on proliferation and
[62] S.F. Williams, D.P. Martin, A.C. Moses, The history of GalaFLEX P4HB scaffold, differentiation of stem cells from human exfoliated deciduous teeth into odonto-
Aesthet. Surg. J. 36 (2016) S33–S42. blast-like cells, Mater. Sci. Eng. C Mater. Biol. Appl. 89 (2018) 128–139.
[63] S.F. Williams, S. Rizk, D.P. Martin, Poly-4-hydroxybutyrate (P4HB): a new gen- [88] M. Zarei, S. Karbasi, F.Sari Aslani, S. Zare, O. Koohi-Hosseinabad, N. Tanideh, In
eration of resorbable medical devices for tissue repair and regeneration, Biomed. vitro and in vivo evaluation of poly (3-hydroxybutyrate)/carbon nanotubes electro-
Tech. 58 (5) (2013) 439–452. spun scaffolds for periodontal ligament tissue engineering, J. Dent. 21 (1) (2020)
[64] L.F. Boesel, S.Le Meur, L. Thöny-Meyer, Q. Ren, The effect of molecular weight 18–30.
on the material properties of biosynthesized poly(4-hydroxybutyrate), Int. J. Biol. [89] Q. Peng, Z.R. Zhang, T. Gong, G.Q. Chen, X. Sun, A rapid-acting, long-acting in-
Macromol. 71 (2014) 124–130. sulin formulation based on a phospholipid complex loaded PHBHHx nanoparticles,
[65] G. Yun, S.Y. Tang, H. Lu, S. Zhang, M.D. Dickey, W. Li, Hybrid-filler stretchable Biomaterials 33 (5) (2012) 1583–1588.
conductive composites: from fabrication to application, Small Sci. 1 (6) (2021) [90] Q. Peng, X. Sun, T. Gong, C.Y. Wu, T. Zhang, J. Tan, Z.R. Zhang, Injectable
2000080. and biodegradable thermosensitive hydrogels loaded with PHBHHx nanoparticles
[66] X. Zhang, G. Chen, Y. Liu, L. Sun, L. Sun, Y. Zhao, Black phosphorus-loaded sepa- for the sustained and controlled release of insulin, Acta Biomater. 9 (2) (2013)
rable microneedles as responsive oxygen delivery carriers for wound healing, ACS 5063–5069.
Nano 14 (5) (2020) 5901–5908. [91] X.D. Yang, H.M. Li, M. Chen, X.H. Zou, L.Y. Zhu, C. Wei, G.Q. Chen,
[67] J. Chi, X. Zhang, C. Chen, C. Shao, Y. Zhao, Y. Wang, Antibacterial and angiogenic Enhanced insulin production from murine islet beta cells incubated on
chitosan microneedle array patch for promoting wound healing, Bioact. Mater. 5 poly(3-hydroxybutyrate-co-3-hydroxyhexanoate), J. Biomed. Mater. Res. A 92 (2)
(2) (2020) 253–259. (2010) 548–555.
[68] E.I. Shishatskaya, E.D. Nikolaeva, O.N. Vinogradova, T.G. Volova, Experimental [92] Z. Zhao, J. Wang, J. Lu, Y. Yu, F. Fu, H. Wang, Y. Liu, Y. Zhao, Z. Gu, Tubular inverse
wound dressings of degradable PHA for skin defect repair, J. Mater. Sci. Mater. opal scaffolds for biomimetic vessels, Nanoscale 8 (28) (2016) 13574–13580.
Med. 27 (11) (2016) 165. [93] D. Puppi, A. Pirosa, G. Lupi, P.A. Erba, G. Giachi, F. Chiellini, Design and fabri-
[69] T.G. Volova, A.A. Shumilova, E.D. Nikolaeva, A.K. Kirichenko, E.I. Shishatskaya, cation of novel polymeric biodegradable stents for small caliber blood vessels by
Biotechnological wound dressings based on bacterial cellulose and degradable computer-aided wet-spinning, Biomed. Mater. 12 (3) (2017) 035011.
copolymer P(3HB/4HB), Int. J. Biol. Macromol. 131 (2019) 230–240. [94] S. Deepthi, M. Nivedhitha Sundaram, P. Vijayan, S.V. Nair, R. Jayakumar, Engi-
[70] S. Vigneswari, V. Murugaiyah, G. Kaur, H.P.S. Abdul Khalil, A.A. Amirul, Simulta- neering poly(hydroxy butyrate-co-hydroxy valerate) based vascular scaffolds to
neous dual syringe electrospinning system using benign solvent to fabricate nanofi- mimic native artery, Int. J. Biol. Macromol. 109 (2018) 85–98.

37
W. Guo, K. Yang, X. Qin et al. Engineered Regeneration 3 (2022) 24–40

[95] X.H. Qu, Q. Wu, J. Liang, X. Qu, S.G. Wang, G.Q. Chen, Enhanced vascular-re- [120] C. Doyle, E.T. Tanner, W. Bonfield, In vitro and in vivo evaluation of polyhydroxy-
lated cellular affinity on surface modified copolyesters of 3-hydroxybutyrate and butyrate and of polyhydroxybutyrate reinforced with hydroxyapatite, Biomaterials
3-hydroxyhexanoate (PHBHHx), Biomaterials 26 (34) (2005) 6991–7001. 12 (9) (1991) 841–847.
[96] G. Li, P. Li, H. Qiu, D. Li, M. Su, K. Xu, Synthesis, characterizations and biocom- [121] S. Zhang, G. Jiang, M.P. Prabhakaran, X. Qin, S. Ramakrishna, Evaluation of elec-
patibility of alternating block polyurethanes based on P3/4HB and PPG-PEG-PPG, trospun biomimetic substrate surface-decorated with nanohydroxyapatite precip-
J. Biomed. Mater. Res. A 98 (1) (2011) 88–99. itation for osteoblasts behavior, Mater. Sci. Eng. C Mater. Biol. Appl. 79 (2017)
[97] V.V. Sevostianova, L.V. Antonova, A.V. Mironov, A.E. Yuzhalin, V.N. Silnikov, 687–696.
T.V. Glushkova, T.S. Godovikova, E.O. Krivkina, E. Bolbasov, T.N. Akentyeva, [122] Z. Chen, Y. Song, J. Zhang, W. Liu, J. Cui, H. Li, F. Chen, Laminated electrospun
M.Y. Khanova, V.G. Matveeva, E.A. Velikanova, R.S. Tarasov, L.S. Barbarash, nHA/PHB-composite scaffolds mimicking bone extracellular matrix for bone tissue
Biodegradable patches for arterial reconstruction modified with RGD peptides: re- engineering, Mater. Sci. Eng. C Mater. Biol. Appl. 72 (2017) 341–351.
sults of an experimental study, ACS Omega 5 (34) (2020) 21700–21711. [123] R. Mai, M.G. Hagedorn, M. Gelinsky, C. Werner, D. Turhani, H. Späth, T. Gedrange,
[98] L.V. Antonova, A.M. Seifalian, A.G. Kutikhin, V.V. Sevostyanova, V.G. Matveeva, G. Lauer, Ectopic bone formation in nude rats using human osteoblasts seeded
E.A. Velikanova, A.V. Mironov, A.R. Shabaev, T.V. Glushkova, E.A. Senokosova, poly(3)hydroxybutyrate embroidery and hydroxyapatite-collagen tapes constructs,
G.Y. Vasyukov, E.O. Krivkina, A.Y. Burago, Y.A. Kudryavtseva, O.L. Barbarash, J. Craniomaxillofac. Surg. 34 (Suppl 2) (2006) 101–109.
L.S. Barbarash, Conjugation with RGD peptides and incorporation of vascular en- [124] M. Parvizifard, S. Karbasi, Physical, mechanical and biological performance of PH-
dothelial growth factor are equally efficient for biofunctionalization of tissue-engi- B-Chitosan/MWCNTs nanocomposite coating deposited on bioglass based scaffold:
neered vascular grafts, Int. J. Mol. Sci. 17 (11) (2016) E1920. potential application in bone tissue engineering, Int. J. Biol. Macromol. 152 (2020)
[99] S.T. Cheng, Z.F. Chen, G.Q. Chen, The expression of cross-linked elastin by rab- 645–662.
bit blood vessel smooth muscle cells cultured in polyhydroxyalkanoate scaffolds, [125] Y. Ding, W. Li, T. Müller, D.W. Schubert, A.R. Boccaccini, Q. Yao, J.A. Roether,
Biomaterials 29 (31) (2008) 4187–4194. Electrospun polyhydroxybutyrate/poly(𝜀-caprolactone)/sol-gel bioactive glass hy-
[100] I. Cummings, S. George, J. Kelm, D. Schmidt, M.Y. Emmert, B. Weber, G. Zünd, brid scaffolds with highly improved osteogenic potential for bone tissue engineer-
S.P. Hoerstrup, Tissue-engineered vascular graft remodeling in a growing lamb ing, ACS Appl. Mater. Interfaces 8 (27) (2016) 17098–17108 58 s.
model: expression of matrix metalloproteinases, Eur. J. Cardiothorac. Surg. 41 (1) [126] H. Hajiali, M. Hosseinalipour, S. Karbasi, M.A. Shokrgozar, The influence of bio-
(2012) 167–172. glass nanoparticles on the biodegradation and biocompatibility of poly (3-hydrox-
[101] Z. Chen, F. Fu, Y. Yu, H. Wang, Y. Shang, Y. Zhao, Cardiomyocytes-actuated morpho ybutyrate) scaffolds, Int. J. Artif. Organs 35 (11) (2012) 1015–1024.
butterfly Wings, Adv. Mater. 31 (8) (2019) e1805431. [127] Z.B. Luklinska, H. Schluckwerder, In vivo response to HA-polyhydroxybu-
[102] R. Sodian, S.P. Hoerstrup, J.S. Sperling, S.H. Daebritz, D.P. Martin, F.J. Schoen, tyrate/polyhydroxyvalerate composite, J. Microsc. 211 (2) (2003) 121–129.
J.P. Vacanti, J.E. Mayer, Tissue engineering of heart valves: in vitro experiences, [128] L. Zhong, D. Hu, Y. Qu, J. Peng, K. Huang, M. Lei, T. Wu, Y. Xiao, Y. Gu, Z. Qian,
Ann. Thorac. Surg. 70 (1) (2000) 140–144. Preparation of adenosine-loaded electrospun nanofibers and their application in
[103] R. Sodian, J.S. Sperling, D.P. Martin, A. Egozy, U. Stock, J.E. Mayer, J.P. Va- bone regeneration, J. Biomed. Nanotechnol. 15 (5) (2019) 857–877.
canti, Fabrication of a trileaflet heart valve scaffold from a polyhydroxyalkanoate [129] S.M. Cool, B. Kenny, A. Wu, V. Nurcombe, M. Trau, A.I. Cassady, L. Grøndahl,
biopolyester for use in tissue engineering, Tissue Eng. 6 (2) (2000) 183–188. Poly(3-hydroxybutyrate-co-3-hydroxyvalerate) composite biomaterials for bone
[104] R. Sodian, S.P. Hoerstrup, J.S. Sperling, S. Daebritz, D.P. Martin, A.M. Moran, tissue regeneration: in vitro performance assessed by osteoblast proliferation, os-
B.S. Kim, F.J. Schoen, J.P. Vacanti, J.E. Mayer, Early in vivo experience with teoclast adhesion and resorption, and macrophage proinflammatory response, J.
tissue-engineered trileaflet heart valves, Circulation 102 (19 Suppl 3) (2000) Biomed. Mater. Res. A 82 (3) (2007) 599–610.
III22–III29. [130] R.V. Chernozem, M.A. Surmeneva, S.N. Shkarina, K. Loza, M. Epple, M. Ulbricht,
[105] S. Wu, Y.L. Liu, B. Cui, X.H. Qu, G.Q. Chen, Study on decellularized porcine aor- A. Cecilia, B. Krause, T. Baumbach, A.A. Abalymov, B.V. Parakhonskiy, A.G. Skir-
tic valve/poly (3-hydroxybutyrate-co-3-hydroxyhexanoate) hybrid heart valve in tach, R.A. Surmenev, Piezoelectric 3D fibrous poly(3-hydroxybutyrate)-based scaf-
sheep model, Artif. Organs 31 (9) (2007) 689–697. folds ultrasound-mineralized with calcium carbonate for bone tissue engineering:
[106] M.M.C.P. Brugmans, R.S. Soekhradj-Soechit, D. van Geemen, M. Cox, inorganic phase formation, osteoblast cell adhesion, and proliferation, ACS Appl.
C.V.C. Bouten, F.P.T. Baaijens, A. Driessen-Mol, Superior tissue evolution in Mater. Interfaces 11 (21) (2019) 19522–19533.
slow-degrading scaffolds for valvular tissue engineering, Tissue Eng. Part A 22 [131] Y.W. Wang, Q. Wu, G.Q. Chen, Attachment, proliferation and differentiation of os-
(1–2) (2016) 123–132. teoblasts on random biopolyester poly(3-hydroxybutyrate-co-3-hydroxyhexanoate)
[107] A.V. Bagdadi, M. Safari, P. Dubey, P. Basnett, P. Sofokleous, E. Humphrey, I. Locke, scaffolds, Biomaterials 25 (4) (2004) 669–675.
M. Edirisinghe, C. Terracciano, A.R. Boccaccini, J.C. Knowles, S.E. Harding, I. Roy, [132] S. Zhao, M. Zhu, J. Zhang, Y. Zhang, Z. Liu, Y. Zhu, C. Zhang, Three
Poly(3-hydroxyoctanoate), a promising new material for cardiac tissue engineer- dimensionally printed mesoporous bioactive glass and poly(3-hydroxybuty-
ing, J. Tissue Eng. Regen. Med. 12 (1) (2018) e495–e512. rate-co-3-hydroxyhexanoate) composite scaffolds for bone regeneration, J. Mater.
[108] D. Castellano, M. Blanes, B. Marco, I. Cerrada, A. Ruiz-Saurí, B. Pelacho, M. Araña, Chem. B 2 (36) (2014) 6106–6118.
J.A. Montero, V. Cambra, F. Prosper, P. Sepúlveda, A comparison of electrospun [133] Y. Liu, S. Yang, L. Cao, X. Zhang, J. Wang, C. Liu, Facilitated vascularization and
polymers reveals poly(3-hydroxybutyrate) fiber as a superior scaffold for cardiac enhanced bone regeneration by manipulation hierarchical pore structure of scaf-
repair, Stem Cells Dev. 23 (13) (2014) 1479–1490. folds, Mater. Sci. Eng. C Mater. Biol. Appl. 110 (2020) 110622.
[109] O. Duvernoy, T. Malm, J. Ramström, S. Bowald, A biodegradable patch used as a [134] Y. Deng, X. Lin, Z. Zheng, J.G. Deng, J.C. Chen, H. Ma, G.Q. Chen,
pericardial substitute after cardiac surgery: 6- and 24-month evaluation with CT, Poly(hydroxybutyrate-co-hydroxyhexanoate) promoted production of extracellu-
Thorac. Cardiovasc. Surg. 43 (5) (1995) 271–274. lar matrix of articular cartilage chondrocytes in vitro, Biomaterials 24 (23) (2003)
[110] H. Wei, Z. Chen, Y. Hu, W. Cao, X. Ma, C. Zhang, X. Gao, X. Qian, Y. Zhao, R. Chai, 4273–4281.
Topographically conductive butterfly wing substrates for directed spiral ganglion [135] Y. Wang, Y.Z. Bian, Q. Wu, G.Q. Chen, Evaluation of three-dimensional scaffolds
neuron growth, Small 17 (38) (2021) e2102062. prepared from poly(3-hydroxybutyrate-co-3-hydroxyhexanoate) for growth of al-
[111] E. Masaeli, P.A. Wieringa, M. Morshed, M.H. Nasr-Esfahani, S. Sadri, C.A. van Blit- logeneic chondrocytes for cartilage repair in rabbits, Biomaterials 29 (19) (2008)
terswijk, L. Moroni, Peptide functionalized polyhydroxyalkanoate nanofibrous scaf- 2858–2868.
folds enhance Schwann cells activity, Nanomedicine 10 (7) (2014) 1559–1569. [136] C. Ye, P. Hu, M.X. Ma, Y. Xiang, R.G. Liu, X.W. Shang, PHB/PHBHHx scaffolds and
[112] P.N. Mohanna, G. Terenghi, M. Wiberg, Composite PHB-GGF conduit for long nerve human adipose-derived stem cells for cartilage tissue engineering, Biomaterials 30
gap repair: a long-term evaluation, Scand. J. Plast. Reconstr. Surg. Hand Surg. 39 (26) (2009) 4401–4406.
(3) (2005) 129–137. [137] M. Zhou, D. Yu, Cartilage tissue engineering using PHBV and PHBV/Bioglass scaf-
[113] D. Schaakxs, D.F. Kalbermatten, E. Pralong, W. Raffoul, M. Wiberg, P.J. Kingham, folds, Mol. Med. Rep. 10 (1) (2014) 508–514.
Poly-3-hydroxybutyrate strips seeded with regenerative cells are effective promot- [138] W. Chen, Y. Li, Y. Huang, Y. Dai, T. Xi, Z. Zhou, H. Liu, Quercetin modified electro-
ers of peripheral nerve repair, J. Tissue Eng. Regen. Med. 11 (3) (2017) 812–821. spun PHBV fibrous scaffold enhances cartilage regeneration, J. Mater. Sci. Mater.
[114] Y.Z. Bian, Y. Wang, G. Aibaidoula, G.Q. Chen, Q. Wu, Evaluation of Med. 32 (8) (2021) 92.
poly(3-hydroxybutyrate-co-3-hydroxyhexanoate) conduits for peripheral nerve re- [139] X. Li, H. Chang, H. Luo, Z. Wang, G. Zheng, X. Lu, X. He, F. Chen, T. Wang,
generation, Biomaterials 30 (2) (2009) 217–225. J. Liang, M. Xu, Poly (3-hydroxybutyrate-co-3-hydroxyhexanoate) scaffolds coated
[115] L. Wang, Z.H. Wang, C.Y. Shen, M.L. You, J.F. Xiao, G.Q. Chen, Differentiation of with PhaP-RGD fusion protein promotes the proliferation and chondrogenic dif-
human bone marrow mesenchymal stem cells grown in terpolyesters of 3-hydrox- ferentiation of human umbilical cord mesenchymal stem cells in vitro, J. Biomed.
yalkanoates scaffolds into nerve cells, Biomaterials 31 (7) (2010) 1691–1698. Mater. Res. A 103 (3) (2015) 1169–1175.
[116] Y. Su, M.S. Toftdal, A. Le Friec, M. Dong, X. Han, M. Chen, 3D electrospun synthetic [140] K.Y. Ching, O.G. Andriotis, S. Li, P. Basnett, B. Su, I. Roy, R.S. Tare, B.G. Sen-
extracellular matrix for tissue regeneration, Small Sci. 1 (7) (2021) 2100003. gers, M. Stolz, Nanofibrous poly(3-hydroxybutyrate)/poly(3-hydroxyoctanoate)
[117] M. Sakar, P. Korkusuz, M. Demirbilek, D.U. Cetinkaya, S. Arslan, scaffolds provide a functional microenvironment for cartilage repair, J. Biomater.
E.B. Denkbas, C.M. Temucin, E. Bilgic, D.B. Hazer, G. Bozkurt, The effect of Appl. 31 (1) (2016) 77–91.
poly(3-hydroxybutyrate-co-3- hydroxyhexanoate) (PHBHHx) and human mes- [141] M.S. Sajid, L. Craciunas, P. Sains, K.K. Singh, M.K. Baig, Use of antibacterial su-
enchymal stem cell (hMSC) on axonal regeneration in experimental sciatic nerve tures for skin closure in controlling surgical site infections: a systematic review of
damage, Int. J. Neurosci. 124 (9) (2014) 685–696. published randomized, controlled trials, Gastroenterol. Rep. 1 (1) (2013) 42–50.
[118] X.F. Zhang, H.X. Liu, L.S. Ortiz, Z.D. Xiao, N.P. Huang, Laminin-modified and [142] Y. He, Z. Hu, M. Ren, C. Ding, P. Chen, Q. Gu, Q. Wu, Evaluation of PHBHHx and
aligned poly(3-hydroxybutyrate-co-3-hydroxyvalerate)/polyethylene oxide nanofi- PHBV/PLA fibers used as medical sutures, J. Mater. Sci. Mater. Med. 25 (2) (2014)
brous nerve conduits promote peripheral nerve regeneration, J. Tissue Eng. Regen. 561–571.
Med. 12 (1) (2018) e627–e636. [143] D.X. Wei, J.W. Dao, G.Q. Chen, A micro-ark for cells: highly open porous polyhy-
[119] L. Yang, Y. Liu, L. Sun, C. Zhao, G. Chen, Y. Zhao, Biomass microcapsules with stem droxyalkanoate microspheres as injectable scaffolds for tissue regeneration, Adv.
cell encapsulation for bone repair, Nanomicro Lett. 14 (1) (2021) 4. Mater. 30 (31) (2018) e1802273.

38
W. Guo, K. Yang, X. Qin et al. Engineered Regeneration 3 (2022) 24–40

[144] K. Perveen, F. Masood, A. Hameed, Preparation, characterization and evaluation P(3HB-co-4HB) scaffolds as a potential wound dressing, Biomed. Mater. 11 (5)
of antibacterial properties of epirubicin loaded PHB and PHBV nanoparticles, Int. (2016) 055009.
J. Biol. Macromol. 144 (2020) 259–266. [167] A.M. Gumel, M.R.M. Razaif-Mazinah, S.N.S. Anis, M.S.M. Annuar, Poly (3-hydrox-
[145] C.S. Wu, Y.C. Hsu, H.T. Liao, Y.X. Cai, Antibacterial activity and in vitro eval- yalkanoates)-co-(6-hydroxyhexanoate) hydrogel promotes angiogenesis and colla-
uation of the biocompatibility of chitosan-based polysaccharide/polyester mem- gen deposition during cutaneous wound healing in rats, Biomed. Mater. 10 (4)
branes, Carbohydr. Polym. 134 (2015) 438–447. (2015) 045001.
[146] G. Babos, J. Rydz, M. Kawalec, M. Klim, A. Fodor-Kardos, L. Trif, T. Feczkó, [168] A. Zonari, T.M.M. Martins, A.C.C. Paula, J.N. Boeloni, S. Novikoff, A.P. Marques,
Poly(3-Hydroxybutyrate)-based nanoparticles for Sorafenib and Doxorubicin an- V.M. Correlo, R.L. Reis, A.M. Goes, Polyhydroxybutyrate-co-hydroxyvalerate struc-
ticancer drug delivery, Int. J. Mol. Sci. 21 (19) (2020) E7312. tures loaded with adipose stem cells promote skin healing with reduced scarring,
[147] F. Masood, P. Chen, T. Yasin, N. Fatima, F. Hasan, A. Hameed, Encapsulation of Acta Biomater. 17 (2015) 170–181.
Ellipticine in poly-(3-hydroxybutyrate-co-3-hydroxyvalerate) based nanoparticles [169] S.H. Keshel, E. Biazar, M. Rezaei Tavirani, M. Rahmati Roodsari, A. Ronaghi,
and its in vitro application, Mater. Sci. Eng. C Mater. Biol. Appl. 33 (3) (2013) M. Ebrahimi, H. Rad, A. Sahebalzamani, A. Rakhshan, K. Afsordeh, The healing
1054–1060. effect of unrestricted somatic stem cells loaded in collagen-modified nanofibrous
[148] Z. Mohtashami, H.A. Javar, M.R. Tehrani, M.R. Esfahani, R. Roohipour, L. Aghajan- PHBV scaffold on full-thickness skin defects, Artif. Cells Nanomed. Biotechnol. 42
pour, F.A. Amoli, M.A. Vakilinezhad, F.A. Dorkoosh, Fabrication, optimization, and (3) (2014) 210–216.
in vitro and in vivo characterization of intra-vitreal implant of budesonide generally [170] I. Han, K.J. Shim, J.Y. Kim, S.U. Im, Y.K. Sung, M. Kim, I.K. Kang, J.C. Kim, Effect of
made of PHBV, AAPS PharmSciTech. 21 (8) (2020) 314. poly(3-hydroxybutyrate-co-3-hydroxyvalerate) nanofiber matrices cocultured with
[149] C. Wu, M. Zhang, Z. Zhang, K.W. Wan, W. Ahmed, D.A. Phoenix, A.M.A. Elhissi, hair follicular epithelial and dermal cells for biological wound dressing, Artif. Or-
X. Sun, Thymopentin nanoparticles engineered with high loading efficiency, im- gans 31 (11) (2007) 801–808.
proved pharmacokinetic properties, and enhanced immunostimulating effect us- [171] M.F. Abazari, S. Zare Karizi, N. Hajati-Birgani, S. Norouzi, Z. Khazeni, J. Hashemi,
ing soybean phospholipid and PHBHHx polymer, Mol. Pharm. 11 (10) (2014) L. Shafaghi, F. Soleimanifar, R.N. Mansour, S.E. Enderami, PHBV nanofibers pro-
3371–3377. motes insulin-producing cells differentiation of human induced pluripotent stem
[150] M. Zhu, K. Li, Y. Zhu, J. Zhang, X. Ye, 3D-printed hierarchical scaffold for local- cells, Gene 768 (2021) 145333.
ized isoniazid/rifampin drug delivery and osteoarticular tuberculosis therapy, Acta [172] P. Kuppan, S. Sethuraman, U.M. Krishnan, Poly(3-hydroxybutyrate-co-3-hydroxy-
Biomater. 16 (2015) 145–155. valerate)-based nanofibrous scaffolds to support functional esophageal epithelial
[151] F. Türesin, I. Gürsel, V. Hasirci, Biodegradable polyhydroxyalkanoate implants for cells towards engineering the esophagus, J. Biomater. Sci. Polym. Ed. 25 (6) (2014)
osteomyelitis therapy: in vitro antibiotic release, J. Biomater. Sci. Polym. Ed. 12 (2) 574–593.
(2001) 195–207. [173] L.V. Antonova, V.V. Sevostyanova, A.G. Kutikhin, A.V. Mironov, E.O. Krivkina,
[152] Q. Cao, J. Zhang, H. Liu, Q. Wu, J. Chen, G.Q. Chen, The mechanism of anti-osteo- A.R. Shabaev, V.G. Matveeva, E.A. Velikanova, E.A. Sergeeva, A.Y. Burago,
porosis effects of 3-hydroxybutyrate and derivatives under simulated microgravity, G.Y. Vasyukov, T.V. Glushkova, Y.A. Kudryavtseva, O.L. Barbarash, L.S. Bar-
Biomaterials 35 (28) (2014) 8273–8283. barash, Vascular endothelial growth factor improves physico-mechani-
[153] S. Gupta, B. Shen, Bone loss in patients with the ileostomy and ileal pouch for cal properties and enhances endothelialization of poly(3-hydroxybutyrate-
inflammatory bowel disease, Gastroenterol. Rep. 1 (3) (2013) 159–165. co-3-hydroxyvalerate)/poly(𝜀-caprolactone) small-diameter vascular grafts in vivo,
[154] N. Bahari Javan, H. Montazeri, L. Rezaie Shirmard, N. Jafary Omid, G.R. Barbari, Front. Pharmacol. 7 (2016) 230.
M. Amini, M.H. Ghahremani, M. Rafiee-Tehrani, F. Abedin Dorkoosh, Preparation, [174] C. Rentsch, B. Rentsch, A. Breier, A. Hofmann, S. Manthey, D. Scharnweber,
characterization and in vivo evaluation of a combination delivery system based on A. Biewener, H. Zwipp, Evaluation of the osteogenic potential and vascularization
hyaluronic acid/jeffamine hydrogel loaded with PHBV/PLGA blend nanoparticles of 3D poly(3)hydroxybutyrate scaffolds subcutaneously implanted in nude rats, J.
for prolonged delivery of Teriparatide, Eur. J. Pharm. Sci. 101 (2017) 167–181. Biomed. Mater. Res. A 92 (1) (2010) 185–195.
[155] N. Bahari Javan, L. Rezaie Shirmard, N. Jafary Omid, H. Akbari Javar, M. Rafiee [175] H. Kenar, G.T. Kose, V. Hasirci, Design of a 3D aligned myocardial tissue construct
Tehrani, F. Abedin Dorkoosh, Preparation, statistical optimisation and in vitro char- from biodegradable polyesters, J. Mater. Sci. Mater. Med. 21 (3) (2010) 989–997.
acterisation of poly (3-hydroxybutyrate-co-3-hydroxyvalerate)/poly (lactic-co-gly- [176] H. Ozer, H. Bozkurt, G. Bozkurt, M. Demirbilek, Regenerative potential of chitosan–
colic acid) blend nanoparticles for prolonged delivery of teriparatide, J. Microen- coated poly-3-hydroxybutyrate conduits seeded with mesenchymal stem cells in a
capsul. 33 (5) (2016) 460–474. rat sciatic nerve injury model, Int. J. Neurosci. 128 (9) (2018) 828–834.
[156] J.P. Ye, J.S. Gong, C. Su, Y.G. Liu, M. Jiang, H. Pan, R.Y. Li, Y. Geng, Z.H. Xu, [177] M. Karimi, E. Biazar, S.H. Keshel, A. Ronaghi, J. Doostmohamadpour, A. Janfada,
J.S. Shi, Fabrication and characterization of high molecular keratin based nanofi- A. Montazeri, Rat sciatic nerve reconstruction across a 30mm defect bridged by an
brous membranes for wound healing, Colloids Surf. B Biointerfaces 194 (2020) oriented porous PHBV tube with Schwann cell as artificial nerve graft, ASAIO. J.
111158. 60 (2) (2014) 224–233.
[157] M.B. Nair, G. Baranwal, P. Vijayan, K.S. Keyan, R. Jayakumar, Composite hydrogel [178] M. Aberg, C. Ljungberg, E. Edin, H. Millqvist, E. Nordh, A. Theorin, G. Terenghi,
of chitosan-poly(hydroxybutyrate-co-valerate) with chondroitin sulfate nanopar- M. Wiberg, Clinical evaluation of a resorbable wrap-around implant as an alter-
ticles for nucleus pulposus tissue engineering, Colloids Surf. B Biointerfaces 136 native to nerve repair: a prospective, assessor-blinded, randomised clinical study
(2015) 84–92. of sensory, motor and functional recovery after peripheral nerve repair, J. Plast.
[158] R. Sodian, S.P. Hoerstrup, J.S. Sperling, D.P. Martin, S. Daebritz, J.E. Mayer, Reconstr. Aesthet. Surg. 62 (11) (2009) 1503–1509.
J.P. Vacanti, Evaluation of biodegradable, three-dimensional matrices for tissue [179] L.N. Novikova, J. Pettersson, M. Brohlin, M. Wiberg, L.N. Novikov, Biodegradable
engineering of heart valves, ASAIO. J. 46 (1) (2000) 107–110. poly-beta-hydroxybutyrate scaffold seeded with Schwann cells to promote spinal
[159] E. Biazar, S.Heidari Keshel, A nanofibrous PHBV tube with Schwann cell as artificial cord repair, Biomaterials 29 (9) (2008) 1198–1206.
nerve graft contributing to rat sciatic nerve regeneration across a 30-mm defect [180] P.N. Mohanna, R.C. Young, M. Wiberg, G. Terenghi, A composite poly-hydroxy-
bridge, Cell Commun. Adhes. 20 (1–2) (2013) 41–49. butyrate-glial growth factor conduit for long nerve gap repairs, J. Anat. 203 (6)
[160] X. Mendibil, F. González-Pérez, X. Bazan, R. Díez-Ahedo, I. Quintana, F.J. Ro- (2003) 553–565.
dríguez, P. Basnett, R. Nigmatullin, B. Lukasiewicz, I. Roy, C.S. Taylor, A. Glen, [181] A. Tahmasebi, A. Shapouri Moghadam, S.E. Enderami, M. Islami, M. Kaabi,
F. Claeyssens, J.W. Haycock, W. Schaafsma, E. González, B. Castro, P. Duffy, E. Saburi, A. Daei Farshchi, F. Soleimanifar, V. Mansouri, Aloe Vera-derived
S. Merino, Bioresorbable and mechanically optimized nerve guidance conduit gel-blended PHBV nanofibrous scaffold for bone tissue engineering, ASAIO. J. 66
based on a naturally derived medium chain length polyhydroxyalkanoate and (8) (2020) 966–973.
poly(𝜀-caprolactone) blend, ACS Biomater. Sci. Eng. 7 (2) (2021) 672–689. [182] A. Codreanu, C. Balta, H. Herman, C. Cotoraci, C.V. Mihali, N. Zurbau, C. Zaharia,
[161] C. Shuai, W. Guo, C. Gao, Y. Yang, Y. Xu, L. Liu, T. Qin, H. Sun, S. Yang, M. Rapa, P. Stanescu, I.C. Radu, E. Vasile, G. Lupu, B. Galateanu, A. Hermenean,
P. Feng, P. Wu, Calcium silicate improved bioactivity and mechanical properties Bacterial cellulose-modified polyhydroxyalkanoates scaffolds promotes bone for-
of poly(3-hydroxybutyrate-co-3-hydroxyvalerate) scaffolds, Polymers (Basel) 9 (5) mation in critical size calvarial defects in mice, Materials (Basel) 13 (6) (2020)
(2017) E175. E1433.
[162] G.R. de Almeida Neto, M.V. Barcelos, M.E.A. Ribeiro, M.M. Folly, R.J.S. Rodríguez, [183] N. Fu, Z. Meng, T. Jiao, X. Luo, Z. Tang, B. Zhu, L. Sui, X. Cai, P34HB electrospun
Formulation and characterization of a novel PHBV nanocomposite for bone defect fibres promote bone regeneration in vivo, Cell Prolif. 52 (3) (2019) e12601.
filling and infection treatment, Mater. Sci. Eng. C Mater. Biol. Appl. 104 (2019) [184] Z. Wang, R. Liang, X. Cheng, Q. Lan, J. Xie, M. He, Y. Pang,
110004. F. Xiong, D. Lei, L. Zheng, J. Zhao, Osteogenic potential of electrospun
[163] T. Zhou, G. Li, S. Lin, T. Tian, Q. Ma, Q. Zhang, S. Shi, C. Xue, W. Ma, X. Cai, poly(3-hydroxybutyrate-co-4-hydroxybutyrate)/poly(ethylene glycol) nanofiber
Y. Lin, Electrospun poly(3-hydroxybutyrate-co-4-hydroxybutyrate)/graphene ox- membranes, J. Biomed. Nanotechnol. 15 (6) (2019) 1280–1289.
ide scaffold: enhanced properties and promoted in vivo bone repair in rats, ACS [185] N. Goonoo, B. Khanbabaee, M. Steuber, A. Bhaw-Luximon, U. Jonas, U. Pietsch,
Appl. Mater. Interfaces 9 (49) (2017) 42589–42600. D. Jhurry, H. Schönherr, 𝜅-Carrageenan enhances the biomineralization and os-
[164] L.X. Lü, X.F. Zhang, Y.Y. Wang, L. Ortiz, X. Mao, Z.L. Jiang, Z.D. Xiao, N.P. Huang, teogenic differentiation of electrospun polyhydroxybutyrate and polyhydroxybu-
Effects of hydroxyapatite-containing composite nanofibers on osteogenesis of mes- tyrate valerate fibers, Biomacromolecules 18 (5) (2017) 1563–1573.
enchymal stem cells in vitro and bone regeneration in vivo, ACS Appl. Mater. Inter- [186] T. Gredes, T. Gedrange, C. Hinüber, M. Gelinsky, C. Kunert-Keil, Histological and
faces 5 (2) (2013) 319–330. molecular-biological analyses of poly(3-hydroxybutyrate) (PHB) patches for en-
[165] D. Castellano, A. Sanchis, M. Blanes, M.D. Pérez Del Caz, A. Ruiz-Saurí, M. Pi- hancement of bone regeneration, Ann. Anat. 199 (2015) 36–42.
quer-Gil, B. Pelacho, B. Marco, N. Garcia, I. Ontoria-Oviedo, V. Cambra, F. Prosper, [187] S. Yang, S. Xu, P. Zhou, J. Wang, H. Tan, Y. Liu, T. Tang, C. Liu, Siliceous mesostruc-
P. Sepúlveda, Electrospun poly(hydroxybutyrate) scaffolds promote engraftment of tured cellular foams/poly(3-hydroxybutyrate-co-3-hydroxyhexanoate) composite
human skin equivalents via macrophage M2 polarization and angiogenesis, J. Tis- biomaterials for bone regeneration, Int. J. Nanomed. 9 (2014) 4795–4807.
sue Eng. Regen. Med. 12 (2) (2018) e983–e994. [188] N. Wang, Z. Zhou, L. Xia, Y. Dai, H. Liu, Fabrication and characterization of bioac-
[166] S. Vigneswari, V. Murugaiyah, G. Kaur, H.P.S. Abdul Khalil, A.A. Amirul, Biomacro- tive 𝛽-Ca2SiO4/PHBV composite scaffolds, Mater. Sci. Eng. C Mater. Biol. Appl. 33
molecule immobilization: grafting of fish-scale collagen peptides onto aminolyzed (4) (2013) 2294–2301.

39
W. Guo, K. Yang, X. Qin et al. Engineered Regeneration 3 (2022) 24–40

[189] G.T. Köse, F. Korkusuz, P. Korkusuz, V. Hasirci, In vivo tissue engineering of bone [192] C. Li, J. Zhang, Y. Li, S. Moran, G. Khang, Z. Ge, Poly (l-lactide-co-caprolac-
using poly(3-hydroxybutyric acid-co-3-hydroxyvaleric acid) and collagen scaffolds, tone) scaffolds enhanced with poly (𝛽-hydroxybutyrate-co-𝛽-hydroxyvalerate) mi-
Tissue Eng. 10 (7–8) (2004) 1234–1250. crospheres for cartilage regeneration, Biomed. Mater. 8 (2) (2013) 025005.
[190] K. Ba, X. Wei, D. Ni, N. Li, T. Du, X. Wang, W. Pan, Chondrocyte co-cul- [193] H.D. Lu, D.Z. Cai, G. Wu, K. Wang, D.H. Shi, Whole meniscus regeneration using
tures with the stromal vascular fraction of adipose tissue in polyhydroxybu- polymer scaffolds loaded with fibrochondrocytes, Chin. J. Traumatol. 14 (4) (2011)
tyrate/poly-(hydroxybutyrate-co-hydroxyhexanoate) scaffolds: evaluation of car- 195–204.
tilage repair in rabbit, Cell Transplant. 28 (11) (2019) 1432–1438. [194] Y. Deng, K. Zhao, X.F. Zhang, P. Hu, G.Q. Chen, Study on the three-dimensional pro-
[191] G. Li, N. Fu, J. Xie, Y. Fu, S. Deng, X. Cun, X. Wei, Q. Peng, X. Cai, Y. Lin, liferation of rabbit articular cartilage-derived chondrocytes on polyhydroxyalka-
Poly(3-hydroxybutyrate-co-4-hydroxybutyrate) based electrospun 3D scaffolds for noate scaffolds, Biomaterials 23 (20) (2002) 4049–4056.
delivery of Autogeneic Chondrocytes and adipose-derived stem cells: evaluation of
cartilage defects in rabbit, J. Biomed. Nanotechnol. 11 (1) (2015) 105–116.

40

You might also like