You are on page 1of 17

Review Article

Najmeh Ahangari1
Curcumin in tissue engineering: A Saeid Kargozar1
traditional remedy for modern medicine Majid Ghayour-Mobarhan1,2,3
Francesco Baino4
Alireza Pasdar1,5
Amirhossein Sahebkar6,7
Gordon A. A. Ferns8
Hae-Won Kim9,10,11
Masoud Mozafari 12,13,14*

1
Department of Modern Sciences and Technologies, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
2
Metabolic Syndrome Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
3
Cardiovascular Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
4
Institute of Materials Physics and Engineering, Applied Science and Technology Department, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129,
Torino, Italy
5
Division of Applied Medicine, Medical School, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK
6
Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
7
Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; School of Pharmacy, Mashhad University of Medical
Sciences, Mashhad, Iran
8
Brighton and Sussex Medical School, Division of Medical Education, Rm 342, Mayfield House, University of Brighton, Brighton, UK
9
Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, South Korea
10
Department of Biomaterials Science, School of Dentistry, Dankook University, Cheonan, South Korea
11
Department of Nanobiomedical Science and BK21 PLUS NBM Global Research Center for Regenerative Medicine Research Center, Dankook University,
Cheonan, South Korea
12
Bioengineering Research Group, Nanotechnology and Advanced Materials Department, Materials and Energy Research Center (MERC), Tehran, Iran
13
Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
14
Department of Tissue Engineering and Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran

Abstract
Curcumin is the principal polyphenolic compound present in the potential to facilitate tissue healing, including anti-inflamma-
turmeric with broad applications in tissue engineering and regen- tory, anti-oxidant, and antibacterial activities. Therefore, curcumin
erative medicine. It has some important inherent properties with has been used for the treatment of various damaged tissues,

Abbreviation: ALP, alkaline phosphatase; α-SMA, alpha-smooth muscle actin; ANG, angiogenin; BMSCs, bone marrow mesenchymal stem cell; BMP-2,
bone morphogenetic protein 2; CaP, calcium phosphate; CS, calcium silicate; CMCS, carboxymethyl chitosan; CNCs, cellulose nanocrystals; Ce, cerium;
Cur, curcumin; DNA, deoxyribonucleic acid; EISA, evaporation-induced self-assembly; ECM, extracellular matrix; FDA, Food and Drug Administration; Ga,
gallium; GRAS, generally recognized as safe; GT, gum tragacanth; H&E, hematoxylin and eosin staining; HO-1, heme oxygenase 1; HFF-1, human foreskin
fibroblast cells; HA, hyaluronic acid; OH−, hydroxyl radicals; HIF-1, hypoxia-inducible factor-1; IFNγ, interferon gamma; IL-6, interleukin-6; MDA, malondialde-
hyde; mTOR, mammalian target of rapamycin; MMP-9, matrix metallopeptidase 9; MBGs, mesoporous bioactive glasses; MesoCS, mesoporous calcium sil-
icate; MRSA, methicillin-resistant Staphylococcus aureus; MAOA, monoamine oxidase A; n-GO, Nanographene oxide; NIPAAM, N-isopropylacrylamide; NO,
Nitric oxide; NSAID, nonsteroidal anti-inflammatory drug; VP, N-vinyl-2-pyrrolidone; ROO, peroxyl radicals; NOO, peroxynitrite; pDNA, plasmid DNA; PEG-A,
poly (ethyleneglycol) monoacrylate; PEG, poly(ethylene glycol); PCL, poly(ε-caprolactone); PCL-PEG-PCL, PCEC, Poly(ε-caprolactone)-poly(ethylene glycol)-
poly(ε-caprolactone); PLGA, poly(lactic-co-glycolic acid); PEI, polyethylenimine; ROS, reactive oxygen species; RANKL, receptor activator of nuclear factor
kappa-B ligand; RPE, Retinal pigment epithelium cells; RUNX2, Runt-related transcription factor 2; O−, Scavenging superoxide anion; SD, Sprague Dawley;
SD, Sprague–Dawley rat model; SDF1, stromal cell-derived factor 1; ESBL, the enzyme extended-spectrum beta-lactimases; TGF-β1, transforming growth
factor beta 1; TNF, tumor necrosis factor; VEGF, vascular endothelial growth factor; WHO, World Health Organization; Zn, Zinc
© 2018 International Union of Biochemistry and Molecular Biology
Volume 00, Number 00, 2018, Pages 1–17
*Address for correspondence: Masoud Mozafari, PhD, Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran.
E-mail: mozafari.masoud@gmail.com
Received 6 September 2018; accepted 12 October 2018
DOI 10.1002/biof.1474
Published online 00 Month 2018 in Wiley Online Library
(wileyonlinelibrary.com)

BioFactors 1
BioFactors

especially wound injuries. There are different forms of curcumin, reports on the advantages of this compound, further research is
among which nano-formulations are of a great importance in required to fully explore its clinical usage. The review describes
regenerative medicine. It is also important to design sophisti- the physicochemical and biological properties of curcumin and
cated delivery systems for controlled/localized delivery of curcu- the current state of the evidence on its applications in tissue engi-
min to the target tissues and organs. Although there are many neering. © 2018 BioFactors, 00(00):1–17, 2018

Keywords: curcumin; anti-inflammation; anti-oxidant; antibacterial;


wound healing; scaffold; tissue repair; tissue engineering

1. Introduction component. As the most commonly used approach, solvent extrac-


tion followed by column chromatography is reported for purifying
Research over the past four decades has described the various
curcumin from turmeric. However, other polar and non-polar
pharmacological effects of curcumin [1]. Curcumin has been
organic solvents are also applied including hexane, ethylacetate,
applied to chemoprevention as well as in eliciting a potential
acetone, and methanol [15]. Whilst chlorinated solvents extract cur-
therapeutic effect in several chronic diseases [2,3]. Hence, the
cumin very efficiently, they are not commonly employed due to their
extraction and synthesis of curcumin and new synthetic deriva-
non-acceptability in the food industry. Other approaches including
tives of curcumin is a major focus of research [4].
soxhlet extraction, ultrasonic extraction, microwave, zone-refining,
Curcumin is the principal curcuminoid of the turmeric rhi-
and dipping methods have also been tried. In general, the soxhlet,
zome (Curcuma longa). Curcuminoids comprise curcumin I
ultrasonic, and microwave extractions are the most widely used
(77%), curcumin II (17%), curcumin III (3%), and cyclocurcumin,
methods for curcumin extraction [16,17].
and were originally isolated by Vogel and Pelletier in 1815 [5].
The chemical structure and chemical synthesis of curcumin were
reported by Lampe and Miłobędzka in 1913 [6]. Curcumin can be
extracted as a yellow-orange dye from the powdered root of Cur-
3. Nanoformulation of curcumin
cuma longa, and has been widely used in Ayurveda, Siddha, and Nanotechnology-based delivery systems have gained wide-
traditional Chinese medicine for centuries, as it has several ther- spread use because of their promising potential and advantages
apeutic properties, including: analgesic, anti-inflammatory, anti- over the conventional approaches. These advantages include:
oxidant, antiseptic, and anti-carcinogenic properties [7,8]. It has (1) limiting biodegradation of the encapsulated cargo [18];
been shown that a dose of up to 12 g/day is safe for human con- (2) facilitation of the controlled/sustained release of the encap-
sumption over short periods in clinical trials [9]. sulated drugs; (3) enhancement of dissolution and tissue perme-
There has been increasing attention on the use of curcumin ability of the poorly water-soluble drugs [19]; (4) enhancement
for regenerative medicine applications. Most studies on curcu- of cellular uptake for effective targeting of bioactive molecules;
min have focused on the wound healing process [10]. However, (5) and optimization of target-specific delivery of drugs and
there has been a gradually broadening of applications to recon- superior drug retention into the target tissues [20,21]. Accord-
struct other tissues like tendon and bone [11,12]. Based on the ingly, recent studies have applied nano-technology-based
literature, the use of curcumin with appropriate biomaterials approaches for the formulation of curcumin to increase its bio-
scaffolds (e.g., polymeric matrices) has been strongly suggested availability. These approaches include encapsulation of curcu-
for tissue engineering applications. min in liposomes [22], chitosan [23], biodegradable
In this review article, we aim to introduce the potential of cur- microspheres [24], silk microspheres [25], complexed with
cumin as a promising biomaterial in the repair and regeneration of phospholipids [26] and polymerizing N-isopropylacrylamide
soft and hard tissues such as skin, tendon, bone, and cartilage. (NIPAAM) with N-vinyl-2-pyrrolidone (VP) and poly (ethylene-
Moreover, we highlight research gaps for the broader use of curcu- glycol) monoacrylate (PEG-A) (see Fig. 1) [27].
min for tissue engineering and regenerative medicine applications.

4. Physico-chemical properties of
2. Extraction of curcumin curcumin
Curcuma longa L (turmeric) is cultivated in tropical and sub- 4.1. Hydrophobicity
tropical regions. India is the main producer of turmeric, and Curcumin is practically insoluble in water at either acidic or
curcumin has been used as a home remedy for several ailments neutral pH. The compound is, however, soluble in alkaline
for many centuries [13,14]. media. The pKa values for the dissociation of the three acid
The word “curcuminoid” denotes a group of compounds includ- protons in curcumin have previously been determined to be
ing curcumin, demethoxycurcumin, bis-demethoxycurcumin, and 7.8, 8.5, and 9.0, respectively [28]. It has been shown that this
cyclic curcumin. Curcumin has been identified as the major substance can interacts with several biomolecules via non-

2 Curcumin in tissue engineering


covalent and covalent binding. The aromatic and tautomeric shown to be an effective scavenger of reactive oxygen species
structure as well as the flexibility of the linker group of curcu- (ROS) and to exert cytoprotective effects against H2O2-induced
min offers its hydrogen bonding and hydrophobicity that is oxidative stress in cultured retinal pigment epithelium (RPE)
responsible for the non-covalent interactions. cells [35]. Curcumin exerts an antioxidant effect by affecting
reactive species; scavenging superoxide anion (O−), peroxyni-
4.2. Stability trite (NOO), nitric oxide (NO), peroxyl radicals (ROO), and
The use of curcumin was previously limited due to low water hydroxyl (OH−) radicals and inducing an up-regulation of anti-
solubility under acidic or neutral conditions, high decomposi- oxidant proteins [36]. Curcumin has been reported to reverse
tion rate in alkaline media and photo-degradation in organic liver, brain and kidney damage, hyperglycemia, lipid oxidation,
solvents. Attempts to prepare water-soluble curcumin by com- inflammation, and oxidative stress [37].
plex formation or interaction with various macromolecules
(e.g., gelatin, polysaccharides) have been reported [29,30]. The 5.2. Anti-inflammatory functions
rapid degradation of curcumin clearly complicates the process. The roles of curcumin as an inhibitor of inflammatory cytokines
It is therefore of interest to study the possibility of forming a have been extensively studied [38–40]. Several mechanisms by
water-soluble curcumin complex at a pH where hydrolysis is at which curcumin can exert anti-inflammatory activity have been
a minimum and to investigate the stability of curcumin in such suggested. It was proposed that curcumin relieves oxidative
a complex [31]. Several recent studies reported different stress and inflammation in chronic diseases by its effects on the
methods to stabilize curcumin in biologic conditions as a thera- Nrf2-keap1 signaling pathway. Moreover, curcumin can sup-
peutic agent [31,32]. Microencapsulation of curcumin within press pro-inflammatory pathways associated with most chronic
hybrid structures, consisting of curcumin-loaded liposomes conditions and block both the production of tumor necrosis fac-
within a protein matrix, significantly increased its bioaccessibil- tor (TNF) and the cell signaling mediated through TNF in differ-
ity (1.seven-fold) compared to the free compound, and could ent cell types [41–43]. Because of the chemical structure of
successfully stabilize it against degradation in PBS (pH = 7.4). curcumin, it may act as a natural-free radical scavenger which
The proposed approach thus proved to be a promising alterna- can reduce the release of various interleukins over NF-κB. Cur-
tive to produce powder-like functional ingredients [33]. cumin may mimic the stress response that induces some com-
ponents of the protein homeostasis network [44].

5.3. Antibacterial activity


5. Biological characteristics The antibacterial effect of curcumin has been well documented
5.1. Antioxidant effects and the relevant mechanism involves the interaction with the
Several studies have shown that curcumin has antioxidant essential cell division initiating protein FtsZ [45]. The FtsZ pro-
properties [34]. The antioxidant activity of curcumin, dimethox- tein is involved in bacterial cell division and is the first protein
ycurcumin, and bisdemthoxycurcumin have been often studied to appear at the impending site of division. FtsZ is present in
with in vitro model systems. For example, curcumin has been almost all prokaryotic species and is also essential for the

Different formulations suggested for preparing nanoscale curcumin can increase its therapeutic effects. With permission from
FIG 1 Ref [10].

ahangari et al. 3
BioFactors

division of chloroplasts and mitochondria in some eukaryotes. 5.6. Toxicity and limitations
Immunolocalization experiments have shown that, in bacteria, Despite the potentially beneficial roles of curcumin, a few stud-
the FtsZ protein undergoes polymerization, forming a ring ies have suggested that high concentrations of curcumin trigger
structure known as the Z-ring at the site of imminent cell divi- nuclear DNA fragmentation in mammalian cell lines [63]. Balaji
sion. The ring structure indicates that FtsZ is a cytoskeletal pro- and Chempakam evaluated the toxicity of 200 chemical com-
tein, exhibiting functional homology with tubulin, a eukaryotic pounds from turmeric. To cross-validate the results, the
cytoskeleton protein. However, the structures of bacterial cyto- authors selected curcumin. The results indicated that, in con-
skeleton proteins are markedly different from their eukaryotic trast to curcumin and its derivatives, there are few other com-
homologs, making it possible to develop specific inhibitors for pounds in turmeric that are non-mutagenic, non-carcinogenic,
bacterial proteins [46,47]. Experimental data suggest that the non-hepatotoxic, and without side-effects [64]. Ganiger
methoxy and hydroxyl groups of curcumin are directly involved et al. [65] examined the reproductive toxicity of curcumin and
in the antimicrobial activity [48,49]. As an illustration, curcu- reported no adverse effect over two generations of rats. How-
min could strongly prohibit the formation of the cytokinetic Z- ever, these results were then reviewed by a committee of the
ring in Bacillus subtilis 168 without affecting the segregation World Health Organization (WHO) and the risk of curcumin-
and organization of the molecule. The authors of this study con- induced reproductive toxicity could not be completely excluded.
cluded that curcumin hinders bacterial cell division by prevent- The potential use of curcumin in chemopreventive or thera-
ing the assembly dynamics of FtsZ in the Z-ring [47]. peutic settings has raised the obvious issues of toxicity and tol-
erance [66]. Multiple studies have been done so far to establish
the safe dose of curcumin. It has been stated that an oral
5.4. Anti-apoptotic effects administration of 400 mg/kg of curcumin is required to obtain
It is commonly assumed that curcumin exerts its action through detectable tissue levels in rats [67]. Moreover, oral administra-
antioxidant and anti-apoptotic effects [50]. However, it has also tion of curcumin was safe in humans at doses between 36 and
been shown that pre-treatment with curcumin is able to reduce 180 mg/day even if taken for up to 4 months [68]. Phase I clini-
the levels of apoptosis in intestinal epithelial cells exposed to cal trials suggest that curcumin is well tolerated in human sub-
interferon gamma (IFNγ) [51]. However, it is not clear how cur- jects when taken at doses as high as 12,000 mg/day [69,70].
cumin acts on IFNγ-induced apoptosis and cytokine secretion. These results were confirmed by Lao and coworkers [71]. The
One of the known actions of curcumin regards iron chelation available experimental evidence clearly showed that, although
and reduction [52,53]. Other studies showed the anti-apoptotic the acceptable daily intake of curcumin as an additive had been
effect of curcumin in other conditions such as drug-induced defined by the WHO as 0–3 mg/kg body weight, it was well tol-
liver injury [54] and cadmium-induced apoptosis in rat testes erated in human subjects at a dose about 50 times higher [72].
[55]. Based on the available literature, it seems likely that cur- It is worth underlining that turmeric is currently listed as “gen-
cumin may impart global protection in vivo by opposing several erally recognized as safe” (GRAS) as a coloring and flavoring
crucial events that are instrumental to both apoptosis and agent in food by the US Food and Drug Administration
necrosis. (FDA) [73].

5.5. Stimulatory and inhibitory effects on angiogenesis 6. Curcumin in TE strategies


It has been demonstrated that a curcumin can inhibit invasion As mentioned in the literature, it is possible to use curcumin by
of cancer-associated fibroblast-driven prostate cancer cells by the topical administration [72]. This provides the feasibility of
regulating MAOA/mTOR/HIF-1α signaling [56]. Thus, curcumin preparing and fabricating a broad range of curcumin-loaded
appears to regulate tumor progression through multiple biolog- constructs, like nanofibrous scaffolds for reconstruction strate-
ical pathways [57]. Studies on curcumin have shown its antian- gies. Therefore, a large number of scaffolds with different
giogenic potential by suppressing the expression or synthesis of applications from hard to soft tissue repair and regeneration
HIF-1α and vascular endothelial growth factor (VEGF) in pitui- are being developed. These are summarized below.
tary adenomas, fibrotic liver, and diabetic kidneys [58,59]. On
the other hand, curcumin promotes healing of indomethacin- 6.1. Wound healing
induced gastric ulceration by stimulation of angiogenesis via Among various natural candidates, curcumin has been exten-
upregulation of VEGF and TGF-β1 [60] and accelerated healing sively used for wound healing applications [74]. Yen et al. have
in dexamethasone impaired wound via enhanced expression of recently shown that curcumin can accelerate cutaneous wound
TGF-β1 and its receptors [61]. It has also been reported that healing via the regulation of a series of molecules such as TNF-
curcumin stimulates the neovascularization at the diabetic α, MMP-9, α-SMA, and collagen [75]. The authors prepared
wound site directly, by the increased expression of antigenic Pluronic F127 gels containing 0.2 mg/mL of curcumin and used
factors such as VEGF, TGF-β1, and other factors such as HIF- them by topical administration for treating wounds in mice for
1α, SDF1a, and HO-1, as well as indirectly, by anti- 3, 5, 7, 9, and 12 days. The obtained results from histology and
inflammatory and antioxidant action [62]. immunohistochemistry analyses revealed the fast wound

4 Curcumin in tissue engineering


closure and well-formed granulation tissue in the mice treated nonsteroidal anti-inflammatory drugs (NSAIDs), antidepres-
with the curcumin-containing gels. They reported an increase sants, and anticonvulsants [79–81]. According to the previously
in the expression levels of collagen and α-SMA in the samples published reports, the analgesic effect of curcumin is related to
treated with curcumin-containing gels. Moreover, the expres- its anti-inflammatory properties [82].
sion of mRNA and protein of TNF-α and MMP-9 significantly There is a need to improve the delivery systems for curcu-
decreased in the experimental group as compared to the con- min because of its relatively low bioavailability. Two nano-
trol, confirming the positive effect on the acceleration of wound based approaches have been developed to overcome this prob-
healing. Mohanty et al. showed that curcumin improves healing lem. Krausz et al. prepared curcumin-encapsulated nanoparti-
in a Sprague–Dawley (SD) excisional full-thickness wound cles as an innovative antimicrobial and wound healing agent
model via the down-regulation of the NF-kB pathway that con- for managing burns. Nanoformulation of curcumin improves its
tributes to the depletion of inflammation and lipid peroxidation poor aqueous solubility and rapid degradation profile as well as
[76]. Taken together, these results provide evidence that curcu- enables its extended topical delivery [83]. The authors of this
min accelerates wound healing by reducing inflammation, study examined the efficacy of this system on the burn injuries
increasing cell proliferation and improving collagen deposition created by heat (160  C for 10 s) on the back of Balb/c mice.
in the injured sites. The experiment included different groups: untreated control,
Different topical formulations of curcumin have been used silver sulfadiazine, control nanoparticle, curcumin, and curcu-
for wound healing including films, fibers, hydrogels, emulsion, min nanoparticle. The mice treated with the sol–gel-prepared
and nano-sized formats (nanoparticles, nanovesicles, micelles, curcumin nanoparticles benefitted from a significant improve-
and nanofibers) (see Fig. 2). Curcumin is mainly used as an ment in the treatment of the burns with reduced bacterial load
additive in these natural (e.g., chitin, chitosan and alginate) or and enhanced wound healing (Fig. 3).
synthetic (e.g., PLGA and PCL) polymeric formulations for Mo et al. [84] prepared electrospun nanofibres containing
wound dressing to manage various wound complications curcumin and angiogenin (ANG) with controlled dual delivery
including burns, diabetics ulcers, and full-thickness excision capacity that can accelerate skin regeneration. They fabricated a
(see Table 1). novel drug and plasmid DNA (pDNA) dual delivery system by
electro-spinning the dispersion composed of PEI-CMCS/pDNA-
Burns. The therapeutic effects of the topical administration of ANG nanoparticles, curcumin, PLGA, and cellulose nanocrystals
curcumin on burn wounds have been previously evaluated by (CNCs). In vitro cell experiments confirmed the bioactivity of cur-
Kulac et al. [78]. The authors produced second-degree burn cumin and ANG in the nanofibers. Moreover, the release profile
wound (2.5 cm in diameter) on the back of the Wistar-albino showed 90% release of the curcumin for 6 days. In order to dem-
rats using aluminum branding irons. The experimental groups onstrate the in vivo angiogenesis and anti-infection properties,
received topical curcumin at a concentration of 100 mg/kg body the authors transplanted the PLGA/CNCs/Curcumin/pDNA-ANG
weight, once daily for 12 days. The results of histopathological composite nanofibres into infected full-thickness burn wounds in
and biochemical analyses showed faster wound healing in the mice. Based on a set of analyses including histology, immunohis-
rats treated with curcumin. The improved rates of inflamma- tochemistry, immunofluorescence, real-time quantitative PCR (rt-
tory cell migration, collagen deposition, angiogenesis, granula- qPCR), and western blotting the PLGA/CNCs/Curcumin/pDNA-ANG
tion tissue formation, and epithelialization were observed in the nanofibres were proven to prevent local infection as well as to
curcumin-treated injuries in comparison to the controls. It has promote skin regeneration.
been proposed that curcumin could reduce burn injury-
associated pain and wounds as an alternative to opioids, Diabetic ulcers. Curcumin has anti-oxidant and anti-
inflammatory properties and these have been used in tissue-
engineered constructs suitable for healing these chronic
wounds. Merrell et al. [85] examined the therapeutic effects of
curcumin-loaded poly(ε-caprolactone) (PCL) nanofibers for
treatment of a streptozotocin-induced diabetic mouse model.
They showed the feasibility and potential of PCL nanofibres as a
delivery system for controlled release of curcumin in wound
healing. The in vitro experiments revealed that this system had
no adverse effects on the human foreskin fibroblast cells (HFF-
1) and showed a good anti-oxidant effect while maintaining the
viability of the cells under conditions of oxidative stress. More-
over, the construct could reduce inflammation as demonstrated
by reduced levels of interleukin-6 (IL-6) release from mouse
Various topical formulations of curcumin for wound monocyte-macrophages seeded onto the nanofibres following
FIG 2 healing strategies. With permission from Ref [77]. stimulation by E-coli-derived lipopolysaccharide. The in vivo
results in mice confirmed the efficacy of curcumin loaded-PCL

ahangari et al. 5
BioFactors

Some of the most important studies on the use of curcumin for wound healing
TABLE 1

Composition Therapeutic Target Remarks Ref (s)


Curcumin (100 mg/kg, topically) Burns - Accelerating wound healing in Wistar– [78]
Albino rats through improving the rates
of inflammatory cells, collagen
deposition, angiogenesis, granulation
tissue formation, and epithelialization
Curcumin nanoparticles (curc-np) Burns - Inhibiting in vitro growth of [83]
methicillin-resistant Staphylococcus
aureus (MRSA) and Pseudomonas
aeruginosa in a dose-dependent fashion
- Improving the wound healing in an
in vivo murine wound model
Electrospun nanofibers composed of Burns - Releasing nearly 90% of curcumin during [84]
PEI-CMCS/pDNA-ANG nanoparticles, the first 6 days
curcumin, poly (D, L-lactic-co-glycolic - Releasing nearly 90% of ANG during the
acid) (PLGA) and cellulose nanocrystals first 20 days
(CNCs) - Exhibiting the excellent biocompatibility
- Preventing the local infection in the
injured site
- Promoting skin regeneration
Collagen-cellulose nanocrystal scaffolds Burns - Along and sustained curcumin release [95]
containing curcumin-loaded profile
microspheres - Exhibiting a significant antibacterial
activity
- Preventing local inflammation in vivo
- Accelerating dermis regeneration in
Male SD rats
Curcumin-loaded poly (ε-caprolactone) Diabetics - Showing a sustained release of curcumin [85]
nanofibers for 72 h
- Lack of cytotoxicity
- Exhibiting antioxidant,
anti-inflammation, and antibacterial
activities
- Increasing the rate of wound closure in a
diabetic mice model
Curcumin (0.3%) in Pluronic F-127 gel Diabetics - Improving the wound contraction [96]
- Decreased the expressions of
inflammatory cytokines/enzymes such
as TNF-α, IL-1β, and MMP-9
- Increasing the levels of
anti-inflammatory cytokine of IL-10
- Increasing the antioxidant enzymes of
superoxide dismutase, catalase, and
glutathione peroxidase
- Better granulation tissue dominated by
marked fibroblast proliferation and
collagen deposition, and wounds were
covered by a thick regenerated epithelial
layer

6 Curcumin in tissue engineering


(Continued)
TABLE 1

Composition Therapeutic Target Remarks Ref (s)


Curcumin-loaded gum tragacanth/poly Diabetics - Exhibiting antibacterial effects against [87]
(ε-caprolactone) electrospun nanofibers the MRSA
- Improving wound healing as a result of
better wound closure with well-formed
granulation tissue dominated by
fibroblast proliferation, collagen
deposition, complete early regenerated
epithelial layer and formation of sweat
glands and hair follicles
Curcumin conjugated to hyaluronic acid Diabetics - Enhancing the cell proliferation [88]
(HA) - Decreasing oxidative damage
- Improving the migration of cells in
scratch wound model in vitro
- Exhibiting antibacterial activity against
MRSA
- Better wound closure in rats received
25 mM of curcumin conjugated to
210 mg/ml of HA, topically
- Fewer inflammatory cells and increased
deposition of dermal layers in the
damaged site
- Formation of well-formed granulation
tissue rich in fibroblast, collagen, and
re-epithelialization
Curcumin loaded chitosan nanoparticles Diabetics - Showing good in vitro characteristics in [97]
impregnated into collagen-alginate terms of better water uptake,
scaffolds biocompatibility, and sustained drug
availability
- Improving wound contraction
- Formation of complete epithelialization
with thick granulation tissue formation
Curcumin-loaded Excisional full-thickness - Exhibiting significant anti-oxidant [93]
poly(e-caprolactone)-poly(ethylene efficacy and low cytotoxicity
glycol)-poly(e-caprolactone) fibrous - Sustained release of curcumin
mats - Regenerating wound defects of Wistar
rats to its normal condition after 21 days
Curcumin containing polymeric micelles Excisional full-thickness - Exhibiting well tissue adhesiveness [92]
(Cur-M) loaded thermosensitive - Releasing curcumin in an extended
hydrogels period
- higher tensile strength and thicker
epidermis in the injured site
- Better wound closure
- Higher collagen content, better
granulation, and higher wound maturity
- Decreasing superoxide dismutase, and
slight increasing in catalase
Curcumin loaded nanographene oxide Excisional full-thickness - Showing antibacterial activity against [98]
(n-GO) reinforced fish scale collagen both positive and negative strains

ahangari et al. 7
BioFactors

(Continued)
TABLE 1

Composition Therapeutic Target Remarks Ref (s)


- Faster wound healing efficiency of
curcumin-loaded constructs
Curcumin-Loaded Chitosan/Gelatin Excisional full-thickness - Improving water uptake ability and [99]
Composite Sponge antibacterial activity
- A greater wound closure in wounds
treated with curcumin-loaded constructs
Curcumin incorporatedcollagen films Excisional full-thickness - Showing an excellent anti-oxidant [91]
activity
- Increased wound reduction, enhancing
cell proliferation, and efficient free
radical scavenging
- Higher shrinkage temperature of the
curcumin containing films and thereby
increasing hydrothermal stability as
compared to normal collagen films

nanofibers improving wound closure in diabetic ulcers. In curcumin to repair diabetic wounds. The in vitro results showed
accordance to what already pointed out elsewhere, these that the nanofibrous membranes release curcumin for about
authors concluded that the biological activity of curcumin is 20 days. The antibacterial effects of the curcumin-containing scaf-
highly dependent on concentration, and the long-term release folds were analyzed against methicillin-resistant Staphylococcus
of curcumin from nanofibres and its subsequent biological aureus (MRSA) and the enzyme extended-spectrum beta-
activity should deserve further evaluation. lactimases (ESBL) bacteria revealed that PCL/GT/Cur nanofibers
Angiogenesis, a vital process for wound healing, gets com- were 99.9% antibacterial against MRSA and 85.14% against ESBL.
promised in diabetes resulting in a delay in wound healing pro- Moreover, the authors showed that the implantation of these scaf-
cess. Previously published reports showed that curcumin can folds with umbilical cord-derived mesenchymal stem cells in male
either stimulate or inhibit the angiogenesis process [62,86]. Wistar rats results in a fast wound closure with well-formed gran-
Kant et al. [62] attempted to exploit the angiogenic potential of ulation tissue dominated by fibroblast proliferation, collagen depo-
curcumin for accelerating wound healing in diabetic rats. They sition, complete early regenerated epithelial layer, and formation
treated open excisional diabetic wounds on the back of adult of sweat glands and hair follicles in the injured sites.
male Wistar rats by topical administration of Pluronic gel (25%) More recently, Sharma et al. examined wound healing
and curcumin (0.3%) for 19 days. The data obtained from dif- activity of curcumin-conjugated to hyaluronic acid (HA) in dia-
ferent analyses including histology, immunohistochemistry (for betic mice [88]. They used this system to overcome the hydro-
CD31 marker), real-time PCR (for VEGF, TGF-β1, HIF-1α, SDF- phobicity and lack of stability of curcumin. The in vitro data
1α, and HO-1), and western blotting (for VEGF and TGF-β1) revealed that HA-conjugated curcumin treatment leads to
confirmed the positive effect of curcumin in the treatment of improved cell proliferation, decreased oxidative damage, and
diabetics’ wounds. Rapid wound closure with well-formed gran- enhanced cell migration in the 2D scratch assay as compared
ulation tissue dominated by fibroblast proliferation, collagen to the treatment with native curcumin. According to the in vivo
deposition, and complete early regenerated epithelial layer was results, the authors reported a better wound healing in dia-
observed in rats treated with curcumin. Furthermore, the betics’ rats treated with HA-conjugated curcumin in compari-
expression of the angiogenic markers was significantly higher son to HA-free curcumin and HA alone. They suggested this
in the groups receiving curcumin as a therapeutic agent. In formulation as a promising candidate for enhancing wound
brief, the authors concluded that curcumin can enhance the healing in diabetic ulcers.
angiogenesis and thereby accelerate the wound healing in dia-
betic rats through up-regulation of different angiogenic factors. Full-thickness excisional wounds. Full-thickness excisional
In 2016, Ranjbar et al. [87] assessed the potential of wound models are commonly used for evaluating a plenty of
curcumin-loaded gum tragacanth/poly(ε-caprolactone) (Cur/PCL/ wound healing situations in vivo [89]. Curcumin has been previ-
GT) electrospun nanofibers for antibacterial performance and ously proposed as a therapeutic agent for the treatment of this
in vivo diabetic wound healing. In fact, the antibacterial PCL/GT/ type of skin injuries [90]. In a pioneering study, Gopinath
Cur membranes were proposed for sustainably delivering et al. [91] took benefit from the antioxidant effects of curcumin

8 Curcumin in tissue engineering


Effects of curcumin on burn wounds. Curcumin nanoparticle is able to enhance the formation of granulation tissue, collagen deposi-
FIG 3 tion, and neoangiogenesis. Hematoxylin and eosin (H&E) and Masson’s trichrome staining of wounds treated with different mate-
rials. Untreated: control, SS: silver sulfadiazine, np: wounds treated with nanoparticles, curcumin: wounds treated with curcumin,
and curcumin nanoparticle: wounds treated with curcumin nanoparticle. On H&E images, the magnification is 4×, bar = 500 μm;
10×, bar = 100 μm and on trichrome images, the magnification is 40×, bar = 100 μm. With permission from Ref [83].

embedded in collagen films to improve wound healing in a full- wounds. For instance, Gong et al. [92] prepared biodegradable
thickness model of male Wistar rats. Spectroscopic studies con- hydrogels containing curcumin encapsulated in micelles for
firmed the correct bonding of curcumin to the collagen films improving cutaneous wound healing. They aimed to take
without affecting its triple helicity. Also, an anti-oxidant assay advantage of the anti-oxidant and anti-inflammation properties
using 2, 20 -azobisisobutyronitrile proved the in vitro antioxidant of curcumin in the hydrogels. In order to overcome curcumin’s
activity of the constructs. The authors then implanted the high hydrophobicity, the authors encapsulated it in polymeric
curcumin-containing films into the animals and evaluated the micelles (Cur-M) with high drug loading and encapsulation
results by histological analysis. The obtained results showed a capability. Then, the Cur-M-loaded thermosensitive hydrogels
substantial increase in neutrophils along with proliferating (Cur-M-H) was prepared and used as wound dressing. The
fibroblasts and macrophages in the rats treated with curcumin- in vitro results showed the good tissue adhesiveness of Cur-M-H
doped collagen, while only a moderate increase in the animals as well as the sustained release of curcumin from the con-
treated with collagen films and control group was observed on structs. The in vivo data revealed the higher tensile strength
day 7 of wound healing. and thicker epidermis in the excisional injure treated with Cur-
Various porous polymeric scaffolds containing curcumin M-H as compared to other groups. Moreover, better wound clo-
have been fabricated to use in the management of full-thickness sure and granulation, as well as higher collagen content and

ahangari et al. 9
BioFactors

The schematic illustration of the study performed by Yang et al. regarding the efficacy of curcumin for wound healing process.
FIG 4 As shown, the authors firstly implanted the BMSC sheets into the skin wounds of the recipient mice. Then they added curcumin
to the wound site for inducing the secretion of various chemokines such as stromal cell-derived factor 1 (Sdf1) and subsequent
improved wound healing. With permission from Ref [94].

wound maturity, were observed in the groups treated with the fibroblast cells. The analysis of results of in vivo examination
Cur-M-H. showed a significant skin regeneration (93.3  6.6%) in the
Drug-loaded electrospun polymer mats are being used as groups treated with the curcumin-loaded PCEC mats. The
wound dressings for wound healing due to their similarity to imbalanced immune response is one of the main problems in
the three-dimensional porous structure of native ECM of skin. adult full-thickness cutaneous wound repair, which may result
In this regard, Fu et al. [93] successfully prepared curcumin in non-functional reconstructed tissue and fibrosis. For addres-
containing poly(ε-caprolactone)-poly(ethylene glycol)-poly sing this issue, Yang et al. recently investigated curcumin-
(ε-caprolactone) (PCL-PEG-PCL, PCEC) copolymer using co- mediated bone marrow mesenchymal stem cell (BMSCs) sheets
electrospinning technology. They evaluated the potential of the as an immune microenvironment for adult full-thickness cuta-
scaffolds regarding skin regeneration in full-thickness exci- neous wound healing (see Fig. 4) [94]. They found that curcu-
sional defects of Wistar rats. After confirming the incorporation min can promote the proliferation rate of BMSCs in vitro and
and subsequent sustained release of curcumin from the mats, modify the characteristics of produced ECM (secreted by
the authors showed the antioxidant activity of these membranes BMSCs), especially the contents of ECM structural proteins like
(with the curcumin content of 20 wt %) on primary mouse skin fibronectin and collagens I and III. The results obtained from

10 Curcumin in tissue engineering


the in vivo study revealed that the group of curcumin-induced the nanofibers at 1 and 5 wt% (CU1 and CU5, respectively), and
BMSCs provided a favorable immune microenvironment with observed the sustained release of curcumin from the fibrous
abundant amounts of SDF1, facilitating a mass migration of leu- scaffolds, with a higher release of drug release from CU5. As an
kocytes. In conclusion, the authors stated that the curcumin- important note, the curcumin-releasing scaffolds reduced cell
induced BMSCs group has better engraftment; meanwhile, cur- proliferation as compared with controls. However, the results
cumin can improve the delivery and efficacy of BMSCs to pro- of gene and protein expression of osteogenic markers (RUNX2
mote its pro-healing functions. and BMP-2) as well as quantification of mineral deposition (ALP
production and bone nodule formation) revealed a significant
6.2. Musculoskeletal regeneration
improvement regarding bone regeneration for the group CU1,
Anti-inflammatory and antineoplastic effects are two key
but not CU5.
properties of curcumin regarding its application in the repair
Calcium silicate (CS) cements are widely used for bone tis-
and regeneration of musculoskeletal disorders [100,101]. It
sue engineering, especially for dental applications due to their
has been well-documented that curcumin can improve multi-
excellent bioactivity and ability to induce the bone-like apatite
ple aspects of bone health in subjects with osteoporosis
formation [113]. However, there is a problem with the use of
through affecting mechanisms involved in the activation and
these materials, that is, their degradability and the subse-
differentiation of osteoclasts. These mechanisms include the
quent condition that can cause the inflammatory response at
inhibition of NF-κB, RANKL, NO production, ROS generation,
the early post-implantation stage [114]. Regarding this issue,
and inflammatory cytokine synthesis [102–109]. By affecting
Chen et al. [115] prepared and evaluated the anti-
these mechanisms, curcumin decreases the number of osteo-
inflammatory effects of curcumin-loaded mesoporous calcium
clasts as well as their differentiation and activation, leading to
silicate (MesoCS/Cur). They synthesized the MesoCS nanopar-
shifting the balance between osteoblast and osteoclast activity
ticles via a template method and add curcumin to this formu-
in favor of osteoblasts. Moon et al. [110] showed the effective-
lation at a concentration of 2, 5 mg/mL, and 10 mM (named as
ness of curcumin on other bone-related disorders by using
C0, C2, C5, and C10, respectively). The results showed good
intraperitoneal injection of curcumin in a mice model of oste-
biocompatibility of MesoCS/Cur samples without any adverse
oarthritis for 2 weeks. There was a proliferation of splenic T
effects on their apatite-formation ability. Moreover, these sys-
cells, the expression of tumor TNF-α, and IL-1β in the ankle
tems could reduce the expression of inflammatory molecules
joint as well as down-regulation of the serum immunoglobulin
including TNF-α and IL-1, confirming its anti-inflammatory
concentrations [110].
ability.
The role of curcumin in the treatment of other specific mus-
More recently, Sedghi et al. developed biocompatible
culoskeletal disorders has not been well documented. There
zinc/curcumin-loaded coaxial nanofibers for bone tissue
are few reports focused on bone/cartilage tissue engineering as
engineering application [116]. They showed that the
well as tendon repair and regeneration; in the following sec-
curcumin-containing scaffolds are cytocompatible and can
tions, we present the studies conducted on the reconstruction of
promote osteogenesis in vitro. Also, the scaffolds showed an
these tissues.
excellent antibacterial activity against E. coli and S. aureus
Bone and cartilage regeneration. In a pioneering study, strains. In another study, Bose et al. evaluated the effects of
Shruti et al. evaluated the potential of cerium-, gallium-, and different polymeric matrices (PCL, PEG, and PLGA) on cur-
zinc-substituted mesoporous bioactive glasses for the loading cumin release from calcium phosphate matrix for in vitro
and delivery of curcumin [111]. The authors firstly synthesized and in vivo bone regeneration [12]. The main object of this
the MBGs based on 80%SiO2–15%CaO–5%P2O5 (mol%) formula- study was the local delivery of curcumin via the polymers
tion using the evaporation-induced self-assembly (EISA) for an effectual treatment regarding improving its bioavail-
method. Then, they applied the impregnation method for incor- ability and providing a higher release. The in vitro results
porating curcumin into MBGs. The in vitro results revealed that revealed enhanced cell viability in the group of curcumin +
gallium- and cerium-containing MBGs-loaded curcumin more PCL-PEG coated scaffolds in comparison to control samples.
than un-substituted glasses as a result of the high affinity of Moreover, the authors developed 3D printed curcumin-
curcumin toward hard Lewis acids. However, all the prepared coated TCP scaffolds for non-load-bearing tissue engineer-
samples (high, low, and un-substituted MBGs) showed a release ing applications (see Fig. 5). The presence of curcumin in
profile of curcumin that is able to exert pharmacological activi- the scaffolds resulted in an increased bone formation after
ties. The authors concluded that MBGs doped with low amounts 6 weeks as compared to pure TCP scaffolds (control). The
of Ce, Ga, and Zn (1%, 1%, and 2%, respectively) can be consid- authors also suggested that local release of curcumin could
ered as proper drug release formulation for curcumin since be exploited in load-bearing implants with the aid of
these glasses showed the higher release of curcumin with quick strengthening polymeric coatings deposited on the brittle
in vitro bioactivity and the added values of the substituents. TCP structure.
Jain et al. [112] examined the effects of curcumin-loaded
PCL electrospun nanofibers on osteogenesis via phytochemical Cartilage reconstruction. The use of curcumin has also been
based bone tissue engineering. The authors loaded curcumin in proposed for cartilage repair and regeneration. In this regard,

ahangari et al. 11
BioFactors

Schematic illustration of experiments conducted by Bose et al. for preparing a curcumin-PCL/PEG-coated 3D printed CaP scaf-
FIG 5 folds, which implanted in the femoral defect of rats. With permission from Ref [12].

Kim et al. fabricated porous composite curcumin/silk scaffolds 5–20 μM can inhibit the inflammation process induced by IL-1
to find an appropriate clinical replacement for defected carti- in human tenocytes. Moreover, curcumin suppressed IL-
lage [117]. For this purpose, the authors fabricated silk-based 1-induced NF-κB activation via inhibiting phosphorylation and
scaffolds with different concentrations of curcumin (0.5–- degradation of inhibitor of κBα, inhibition of inhibitor of κB
2 mg/ml). The obtained data revealed higher cell viability rate kinase activity, and inhibition of nuclear translocation of NF-
and ECM formation for 1 mg/ml curcumin-containing silk κB. This study was the first report on the potential of curcu-
scaffolds as compared with the other groups. After implanta- min in treating tendon inflammation through modulation of
tion in the subcutaneous region of nude athymic mice, the his- NF-κB signaling.
tological evaluations confirmed the biocompatibility and Jiang et al. evaluated the therapeutic potential of curcumin
formation of a cartilaginous matrix in the chondrocytes regarding tendon healing in SD rats [11]. For this aim, they cre-
(1 × 104 cells/scaffold) loaded curcumin/silk scaffolds groups ated a patellar tendon window defect in the animals and
(see Fig. 6). Therefore, curcumin-containing silk scaffolds administered curcumin (100 mg/kg) via an oral gavage. The
show promise in providing clinical support for the patients results of histological and biochemical evaluations showed that
with different cartilage disorders. curcumin could improve the healing process through stimulat-
ing collagen deposition with well-organized structure in the
Tendon healing. Inflammation is one of the main processes damaged sites and decreasing malondialdehyde (MDA) level,
playing a role in the pathogenesis of tendon-related disorders respectively. Moreover, the biomechanical parameters
such as tendinitis and tendinopathy [118,119]. This inflamma- (e.g., tensile strength of tendon) were improved in the rats trea-
tion is controlled by several pro-inflammatory cytokines like ted with curcumin. Based on the results, the authors stated that
IL-1 and TNF-α. So, the use of materials that have anti- the use of curcumin could be an additional therapeutic agent
inflammation activity has been proposed as a potential way to for the management of damaged tendon tissue.
manage these diseases. Buhrmann et al. [120] showed that Cui et al. showed that the controlled release of curcumin
curcumin can modulate NF-κB-mediated inflammation in can prevent peritendinous adhesion during Achilles tendon heal-
human tenocytes in vitro. They also evaluated the mechanism ing in rats [121]. The authors loaded curcumin in nanomicelles
of curcumin action on IL-1-mediated inflammatory signaling. (gold nanorods [GNRs]-1/curcumin in polymeric nanomicelles
Their results revealed that curcumin at concentrations of [curc@PMs]) and then injected these nano-structures into the

12 Curcumin in tissue engineering


The results of histological evaluations (H&E, safranin-O, and alcian blue staining) of curcumin-containing silk scaffolds implanted
FIG 6 in the subcutaneous region of nude mice for 2 and 4 weeks. With permission from Ref [117].

surgical site (0.44 mg curcumin/kg) at weeks 1, 2, and 3, for microspheres in Achilles tendinopathy has been recently evalu-
three times 10 s each. In the same manner, the control groups ated by Kim et al. [122]. They prepared curcumin-loaded porous
received 0.44 mg curcumin/kg and 0.1 mL of saline, respectively. PLGA microspheres (Cur/PMSs) by a fluidic device. The cell cul-
The data obtained from biomechanical and histological evalua- ture study showed that the Cur/PMSs have anti-inflammatory
tions revealed the lowest grade of peritendinous adhesions in the effects on LPS-treated tenocytes in a dose-dependent manner.
animals treated with NRs-1/curc@PMs group in comparison to This effect was proven through significant down-regulation of
the other groups. The authors claimed that curcumin-loaded pro-inflammatory markers including MMP-3 and MMP-13, COX-
nanoparticles possess the great potential for preventing adhe- 2, ADAMTS-5, IL-6, and TNF-α. The in vivo results revealed the
sion in clinical patients. effectiveness of the local injection of Cur/PMSs in terms of ten-
In vitro and in vivo anti-inflammatory and tendon-healing don tissue healing of rats with collagenase-induced Achilles ten-
effects of an extended curcumin delivery with porous dinopathy (Fig. 7). Similar to the above-mentioned studies, an

The schematic illustration of curcumin loaded porous microspheres (Cur/PMSs) injected in the damaged Achilles (tendinitis) of
FIG 7 rats with the aim of reducing the inflammation and improving the healing process. With permission from Ref [122].

ahangari et al. 13
BioFactors

improvement in the biomechanical properties, that is, the incre- tau phosphorylation and, more generally, neuroinflammation
ment of the tensile strength of tendon tissue, was observed in or oxidative stress. In the field of neurosurgery, curcumin was
the animals receiving Cur/PMSs in a dose-dependent manner. also shown to be able to promote peripheral nerve regenera-
tion in rats under both normal [128] and diabetic condi-
tions [129].
7. Conclusions and outlook Potential applications of curcumin in the field of lung can-
cer treatment have been suggested through further biomolecu-
The literature clearly supports the application of curcumin in
lar and preclinical studies should be carried out to elucidate its
modern regenerative medicine. However, the insolubility in
effectiveness. In fact, if on one hand curcumin was reported to
water and poor bioavailability of unmodified curcumin has lim-
inhibit the migratory and invasive ability of A549 lung cancer
ited its therapeutic applications. The advent of new delivery
cells in human patients by blocking the adiponectin receptor
strategies and biocompatible carriers, curcumin has now been
1 [130], on the other hand a negative effect was observed in
employed in drug therapies based on implantable biomaterials
smokers and ex-smokers, where curcumin was reported to
rather than simple oral ingestion. The anti-inflammatory, anti-
stimulate ROSs production and, thus, have a potential carcino-
oxidant, and angiogenetic properties of curcumin have been
genic effect [131].
shown very helpfully in accelerating the regeneration of bone
In summary, there should be further studies testing the
tissue, cartilage, and injured tendon as well as in promoting the
effects of curcumin in the regeneration of complex tissues and
healing of chronic wounds.
organs such as nerve, liver, heart, and lung. For this purpose,
Looking at the future, new applications of curcumin to treat
reliable animal models should be used that take account the
other damaged tissues/organs and diseases deserve investiga-
best way for curcumin administration in the specific clinical
tion, also considering that its use is considered clinically safe
case considered. Local delivery instead of oral ingestion could
and, thus there appear to be limited barriers to testing and
be indeed preferred to maximize the efficacy and minimize any
experimentation from a regulatory viewpoint.
possible side effect. The selection of compatible biomaterials to
Cardio-protective effects of curcumin are particularly
use as carrier/scaffold for curcumin (e.g., soft polymeric matri-
attractive, as cardiovascular diseases are estimated by the
ces with tunable porosity degradation rates) will be the key to
WHO to be the first cause of death globally [123]. These benefi-
the success of these new therapeutic approaches.
cial effects have recently been reviewed by Wongcharoen and
Phrommintikul [124]. The antioxidant effects of curcumin have
been shown to attenuate adriamycin-induced cardiotoxicity and
to prevent cardiovascular complications caused by diabetes. Its
Conflict of interest
anti-thrombotic, antiproliferative, and anti-inflammatory effects The authors declare that there are no conflicts of interest
along its ability to decrease the serum cholesterol level may regarding the publication of this paper.
protect against the pathological changes associated with ath-
erosclerosis. Curcumin is also known to reduce the fatty acid
References
deposits on the walls of blood vessels. The inhibitory effect of
[1] Sahebkar, A. (2013) Why it is necessary to translate curcumin into clinical
curcumin on p300-HAT has been demonstrated to reduce the
practice for the prevention and treatment of metabolic syndrome? Biofac-
development of cardiac hypertrophy and heart failure in animal tors 39, 197–208.
models. The modulatory effect of curcumin on calcium homeo- [2] Wilken, R., Veena, M. S., Wang, M. B., and Srivatsan, E. S. (2011) Curcumin:
stasis may also play a role in the prevention of ventricular a review of anti-cancer properties and therapeutic activity in head and neck
arrhythmias. squamous cell carcinoma. Mol. Cancer 10, 12.

[3] Grynkiewicz, G., and Slifirski, P. (2012) Curcumin and curcuminoids in quest
Curcumin’s ability to inhibit several factors like NF-κB,
for medicinal status. Acta Biochim. Pol. 59, 201–212.
which modulates some pro-inflammatory and profibrotic cyto-
[4] Priyadarsini, K. I. (2014) The chemistry of curcumin: from extraction to ther-
kines, provide a rational molecular basis for its use in hepatic apeutic agent. Molecules 19, 20091–20112.
disorders [125]. Curcumin can attenuate liver injury induced by [5] Vogel, H., and Pelletier, J. (1815) Curcumin-biological and medicinal proper-
alcohol abuse, iron overdose, cholestasis and acute CCl4 intoxi- ties. J Pharma. 2, 24.
cation. Interestingly, curcumin can even reverse CCl4-induced [6] Lee, W.-H., Loo, C.-Y., Bebawy, M., Luk, F., Mason, R. S., et al. (2013) Curcu-
min and its derivatives: their application in neuropharmacology and neuro-
liver cirrhosis to some extent. The relatively limited number of
science in the 21st century. Current Neuropharmacol. 11, 338–378.
studies in this area should be dramatically expanded in the next [7] Zhou, Y., Xie, M., Song, Y., Wang, W., Zhao, H., et al. (2016) Two traditional
few years as hepatic diseases are one of the main causes of dis- Chinese medicines Curcumae radix and Curcumae rhizoma: an ethnophar-
ability and mortality worldwide. macology, phytochemistry, and pharmacology review. Evid. Based Comple-
Belcacemi et al. [126], has suggested that curcumin could ment. Alternat. Med. 2016, 1–30.
[8] Cikrikci, S., Mozioglu, E., and Yilmaz, H. (2008) Biological activity of curcumi-
also play a role in the treatment of Alzheimer disease; one of
noids isolated from Curcuma longa. Records Natural Prod. 2, 19.
the most important societal and medical challenges in the [9] Gupta, S. C., Patchva, S., and Aggarwal, B. B. (2013) Therapeutic roles of
next few years [127]. In fact, in vitro and in vivo studies have curcumin: lessons learned from clinical trials. AAPS J. 15, 195–218.
revealed that curcumin can target biomolecular pathways [10] Hussain, Z., Thu, H. E., Ng, S.-F., Khan, S., and Katas, H. (2017) Nanoencap-
involved in this type of dementia, such as β-amyloid cascade, sulation, an efficient and promising approach to maximize wound healing

14 Curcumin in tissue engineering


efficacy of curcumin: a review of new trends and state-of-the-art. Colloids in vivo pharmacokinetics and biodistribution. Nanomed. Nanotechnol. Biol.
Surf. B Biointerf. 150, 223–241. Med. 13, 943–953.
[11] Jiang, D., Gao, P., Lin, H., and Geng, H. (2016) Curcumin improves tendon [33] Gómez-Mascaraque, L. G., Sipoli, C. C., de La Torre, L. G., and López-
healing in rats: a histological, biochemical, and functional evaluation. Con- Rubio, A. (2017) Microencapsulation structures based on protein-coated
nect. Tissue Res. 57, 20–27. liposomes obtained through electrospraying for the stabilization and
[12] Bose, S., Sarkar, N., and Banerjee, D. (2018) Effects of PCL, PEG and PLGA improved bioaccessibility of curcumin. Food Chem. 233, 343–350.
polymers on curcumin release from calcium phosphate matrix for in vitro [34] Mozafari, M., Rajadas, J., & Kaplan, D. (Eds.). (2018). Nanoengineered Bio-
and in vivo bone regeneration. Mater. Today Chem. 8, 110–120. materials for Regenerative Medicine. Elsevier.
[13] Aggarwal, B. B., Kumar, A., and Bharti, A. C. (2003) Anticancer potential of [35] Woo, J. M., Shin, D.-Y., Lee, S. J., Joe, Y., Zheng, M., et al. (2012) Curcumin
curcumin: preclinical and clinical studies. Anticancer Res. 23, 363–398. protects retinal pigment epithelial cells against oxidative stress via induc-
[14] Esatbeyoglu, T., Huebbe, P., Ernst, I., Chin, D., Wagner, A. E., et al. (2012) tion of heme oxygenase-1 expression and reduction of reactive oxygen.
Curcumin—from molecule to biological function. Angew. Chem. Int. Ed. 51, Mol. Vis. 18, 901.
5308–5332. [36] Trujillo, J., Chirino, Y. I., Molina-Jijón, E., Andérica-Romero, A. C., Tapia, E.,
[15] Paulucci, V. P., Couto, R. O., Teixeira, C. C., and Freitas, L. A. P. (2013) Opti- et al. (2013) Renoprotective effect of the antioxidant curcumin: recent find-
mization of the extraction of curcumin from Curcuma longa rhizomes. Rev ings. Redox Biol. 1, 448–456.
Bras Farmacogn. 23, 94–100. [37] Xu, Y., Lin, D., Li, S., Li, G., Shyamala, S. G., et al. (2009) Curcumin reverses
[16] Lee, K.-J., Yang, H.-J., Jeong, S.-W., and Ma, J.-Y. (2012) Solid-phase extrac- impaired cognition and neuronal plasticity induced by chronic stress. Neu-
tion of curcuminoid from turmeric using physical process method. Korean ropharmacology 57, 463–471.
J. Pharmacogn. 43, 250–256. [38] Yadav, R., Jee, B., and Awasthi, S. K. (2015) Curcumin suppresses the pro-
[17] Lee, K.-J., Ma, J.-Y., Kim, Y.-S., Kim, D.-S., and Jin, Y. (2012) High purity duction of pro-inflammatory cytokine interleukin-18 in lipopolysaccharide
extraction and simultaneous high-performance liquid chromatography anal- stimulated murine macrophage-like cells. Indian J. Clin. Biochem. 30,
ysis of curcuminoids in turmeric. J Appl Biol Chem. 55, 61–65. 109–112.
[18] Sun, M., Su, X., Ding, B., He, X., Liu, X., et al. (2012) Advances in nanotechnology- [39] Mollazadeh, H., Cicero, A. F., Blesso, C. N., Pirro, M., Majeed, M., et al. (2017)
based delivery systems for curcumin. Nanomedicine 7, 1085–1100. Immune modulation by curcumin: the role of interleukin-10. Crit. Rev. Food
[19] Ahmad, M. Z., Alkahtani, S. A., Akhter, S., Ahmad, F. J., Ahmad, J., Sci. Nutr. 1–13. https://doi.org/10.1080/10408398.2017.1358139
et al. (2016) Progress in nanotechnology-based drug carrier in designing of [40] Ghandadi, M., and Sahebkar, A. (2017) Curcumin: an effective inhibitor of
curcumin nanomedicines for cancer therapy: current state-of-the-art. interleukin-6. Curr. Pharm. Des. 23, 921–931.
J. Drug Target. 24, 273–293. [41] Chang, N.-S., Joki, N., Mattison, J., Dinh, T., and John, S. (1997) Characteri-
[20] Hussain, Z., Katas, H., Amin, M. C. I. M., Kumolosasi, E., Buang, F., zation of serum adhesive proteins that block tumor necrosis factor-mediated
et al. (2013) Self-assembled polymeric nanoparticles for percutaneous co- cell death. Cell Death Differ. 4, 779–786.
delivery of hydrocortisone/hydroxytyrosol: an ex vivo and in vivo study [42] Gupta, S. C., Tyagi, A. K., Deshmukh-Taskar, P., Hinojosa, M., Prasad, S.,
using an NC/Nga mouse model. Int. J. Pharm. 444, 109–119. et al. (2014) Downregulation of tumor necrosis factor and other proinflam-
[21] Ghalandarlaki, N., Alizadeh, A. M., and Ashkani-Esfahani, S. (2014) Nano- matory biomarkers by polyphenols. Arch. Biochem. Biophys. 559, 91–99.
technology-applied curcumin for different diseases therapy. Biomed. Res. [43] Anthwal, A., Thakur, B. K., Rawat, M., Rawat, D., Tyagi, A. K., et al. (2014)
Int. 2014, 1–23. Synthesis, characterization and in vitro anticancer activity of C-5 curcumin
[22] Wang, D., Veena, M. S., Stevenson, K., Tang, C., Ho, B., et al. (2008) Lipo- analogues with potential to inhibit tnftnf-α-induced NF-κB activation.
some-encapsulated curcumin suppresses growth of head and neck squa- Biomed. Res. Int. 2014, 1–10.
mous cell carcinoma in vitro and in xenografts through the inhibition of [44] Giordano, S., Darley-Usmar, V., and Zhang, J. (2014) Autophagy as an
nuclear factor κB by an AKT-independent pathway. Clin. Cancer Res. 14, essential cellular antioxidant pathway in neurodegenerative disease. Redox
6228–6236. Biol. 2, 82–90.
[23] Das, R. K., Kasoju, N., and Bora, U. (2010) Encapsulation of curcumin in [45] Erickson, H. P., Taylor, D. W., Taylor, K. A., and Bramhill, D. (1996) Bacterial
alginate-chitosan-pluronic composite nanoparticles for delivery to cancer cell division protein FtsZ assembles into protofilament sheets and minir-
cells. Nanomed. Nanotechnol. Biol. Med. 6, 153–160. ings, structural homologs of tubulin polymers. Proc. Natl. Acad. Sci. USA
[24] Kumar, V., Lewis, S. A., Mutalik, S., Shenoy, D. B., and Udupa, N. (2002) Bio- 93, 519–523.
degradable microspheres of curcumin for treatment of inflammation. Indian [46] Kaur, S., Modi, N. H., Panda, D., and Roy, N. (2010) Probing the binding site
J. Physiol. Pharmacol. 46, 209–217. of curcumin in Escherichia coli and Bacillus subtilis FtsZ – a structural
[25] Gupta, V., Aseh, A., Ríos, C. N., Aggarwal, B. B., and Mathur, A. B. (2009) insight to unveil antibacterial activity of curcumin. Eur. J. Med. Chem. 45,
Fabrication and characterization of silk fibroin-derived curcumin nanoparti- 4209–4214.
cles for cancer therapy. Int. J. Nanomed. 4, 115. [47] Rai, D., Singh, J. K., Roy, N., and Panda, D. (2008) Curcumin inhibits FtsZ
[26] Maiti, K., Mukherjee, K., Gantait, A., Saha, B. P., and Mukherjee, P. K. (2007) assembly: an attractive mechanism for its antibacterial activity. Biochem. J.
Curcumin–phospholipid complex: preparation, therapeutic evaluation and 410, 147–155.
pharmacokinetic study in rats. Int. J. Pharm. 330, 155–163. [48] Han, S., and Yang, Y. (2005) Antimicrobial activity of wool fabric treated
[27] Bisht, S., Feldmann, G., Soni, S., Ravi, R., Karikar, C., et al. (2007) Polymeric with curcumin. Dyes Pigm 64, 157–161.
nanoparticle-encapsulated curcumin ("nanocurcumin"): a novel strategy for [49] Tesaki, S., Tanabe, S., Ono, H., Fukushi, E., Kawabata, J., et al. (1998)
human cancer therapy. J. Nanobiotechnol. 5, 3. 4-Hydroxy-3-nitrophenylacetic and sinapic acids as antibacterial compounds
[28] Tønnesen, H. H., and Karlsen, J. (1985) Studies on curcumin and curcumi- from mustard seeds. Biosci. Biotechnol. Biochem. 62, 998–1000.
noids. Z Lebensm Unters Forsch 180, 402–404. [50] Banerjee, S., Singh, S. K., Chowdhury, I., Lillard, J. W., Jr., and Singh, R.
[29] Maing, Y., and Miller, I. (1981) Curcumin-metal colour complexes and pro- (2017) Combinatorial effect of curcumin with docetaxel modulates apoptotic
cess for preparing them. Eur. Pat Appl. EP. 25, A1. and cell survival molecules in prostate cancer. Front. Biosci 9, 235.
[30] Leshik RR. Stabilized curcumin colorant. Google Patents; 1981. [51] Loganes, C., Lega, S., Bramuzzo, M., Vecchi Brumatti, L., Piscianz, E.,
[31] Tønnesen, H. H., Másson, M., and Loftsson, T. (2002) Studies of curcumin et al. (2017) Curcumin anti-apoptotic action in a model of intestinal epithelial
and curcuminoids. XXVII. Cyclodextrin complexation: solubility, chemical inflammatory damage. Nutrients 9, 578.
and photochemical stability. Int. J. Pharm. 244, 127–135. [52] Yang, C., Ma, X., Wang, Z., Zeng, X., Hu, Z., et al. (2017) Curcumin induces
[32] Bi, C., Miao, X. Q., Chow, S. F., Wu, W. J., Yan, R., et al. (2017) Particle size apoptosis and protective autophagy in castration-resistant prostate cancer
effect of curcumin nanosuspensions on cytotoxicity, cellular internalization, cells through iron chelation. Drug Design Dev. Ther. 11, 431.

ahangari et al. 15
BioFactors

[53] Galleggiante, V., De Santis, S., Cavalcanti, E., Scarano, A., De Benedictis, M., [76] Mohanty, C., Das, M., and Sahoo, S. K. (2012) Sustained wound healing
et al. (2017) Dendritic cells modulate iron homeostasis and inflammatory activity of curcumin loaded oleic acid based polymeric bandage in a rat
abilities following quercetin exposure. Curr. Pharm. Des. 23, 2139–2146. model. Mol. Pharm. 9, 2801–2811.
[54] Bulku, E. J., Stohs, S., Cicero, L., Brooks, T., Halley, H. D., et al. (2012) Curcu- [77] Mohanty, C., and Sahoo, S. K. (2017) Curcumin and its topical formulations
min exposure modulates multiple pro-apoptotic and anti-apoptotic signaling for wound healing applications. Drug Discov. Today 22, 1582–1592.
pathways to antagonize acetaminophen-induced toxicity. Curr. Neurovasc. [78] Kulac, M., Aktas, C., Tulubas, F., Uygur, R., Kanter, M., et al. (2013) The
Res. 9, 58–71. effects of topical treatment with curcumin on burn wound healing in rats.
[55] Aktas, C., Kanter, M., Erboga, M., and Ozturk, S. (2012) Anti-apoptotic effects J. Mol. Histol. 44, 83–90.
of curcumin on cadmium-induced apoptosis in rat testes. Toxicol. Ind. [79] Sharma, S., Kulkarni, S. K., Agrewala, J. N., and Chopra, K. (2006) Curcumin
Health 28, 122–130. attenuates thermal hyperalgesia in a diabetic mouse model of neuropathic
[56] Du, Y., Long, Q., Zhang, L., Shi, Y., Liu, X., et al. (2015) Curcumin inhibits pain. Eur. J. Pharmacol. 536, 256–261.
cancer-associated fibroblast-driven prostate cancer invasion through [80] Mittal, N., Joshi, R., Hota, D., and Chakrabarti, A. (2009) Evaluation of antihy-
MAOA/mTOR/HIF-1α signaling. Int. J. Oncol. 47, 2064–2072. peralgesic effect of curcumin on formalin-induced orofacial pain in rat. Phyt-
[57] Singh, S., and Khar, A. (2006) Biological effects of curcumin and its role in other. Res. 23, 507–512.
cancer chemoprevention and therapy. Anti-Cancer Agents Med. Chem. 6, [81] Agarwal, K. A., Tripathi, C., Agarwal, B. B., and Saluja, S. (2011) Efficacy of
259–270. turmeric (curcumin) in pain and postoperative fatigue after laparoscopic
[58] Shan, B., Schaaf, C., Schmidt, A., Lucia, K., Buchfelder, M., et al. (2012) Cur- cholecystectomy: a double-blind, randomized placebo-controlled study.
cumin suppresses HIF-1A synthesis and VEGFA release in pituitary adeno- Surg. Endosc. 25, 3805–3810.
mas. J. Endocrinol. 214, 389–398. [82] Cheppudira, B., Fowler, M., McGhee, L., Greer, A., Mares, A., et al. (2013)
[59] D-w, Z., Fu, M., Gao, S.-H., and Liu, J.-L. (2013) Curcumin and diabetes: a Curcumin: a novel therapeutic for burn pain and wound healing. Expert
systematic review. Evidence Based Complement Alternat. Med. 2013, 1–16. Opin. Investig. Drugs 22, 1295–1303.
[60] Sharma, A. V., Ganguly, K., Paul, S., Maulik, N., and Swarnakar, S. (2012) [83] Krausz, A. E., Adler, B. L., Cabral, V., Navati, M., Doerner, J., et al.
Curcumin heals indomethacin-induced gastric ulceration by stimulation of (2015) Curcumin-encapsulated nanoparticles as innovative antimicrobial
angiogenesis and restitution of collagen fibers via VEGF and MMP-2 medi- and wound healing agent. Nanomed. Nanotechnol. Biol. Med. 11,
ated signaling. Antioxid. Redox Signal. 16, 351–362. 195–206.
[61] Mani, H., Sidhu, G. S., Kumari, R., Gaddipati, J. P., Seth, P., et al. (2002) Cur- [84] Mo, Y., Guo, R., Zhang, Y., Xue, W., Cheng, B., et al. (2017) Controlled dual
cumin differentially regulates TGF-β1, its receptors and nitric oxide synthase delivery of angiogenin and curcumin by electrospun nanofibers for skin
during impaired wound healing. Biofactors 16, 29–43. regeneration. Tissue Eng. Part A 23, 597–608.
[62] Kant, V., Gopal, A., Kumar, D., Pathak, N. N., Ram, M., et al. (2015) Curcu- [85] Merrell, J. G., McLaughlin, S. W., Tie, L., Laurencin, C. T., Chen, A. F.,
min-induced angiogenesis hastens wound healing in diabetic rats. J. Surg. et al. (2009) Curcumin-loaded poly (ε-caprolactone) nanofibres: diabetic
Res. 193, 978–988. wound dressing with anti-oxidant and anti-inflammatory properties. Clin.
[63] Ting, C.-Y., Wang, H.-E., Yu, C.-C., Liu, H.-C., Liu, Y.-C., et al. (2015) Curcu- Exp. Pharmacol. Physiol. 36, 1149–1156.
min triggers DNA damage and inhibits expression of DNA repair proteins in [86] Bhandarkar, S. S., and Arbiser, J. L. (2007) Curcumin as an inhibitor of
human lung cancer cells. Anticancer Res 35, 3867–3873. angiogenesis. In The molecular targets and therapeutic uses of curcumin in
[64] Balaji, S., and Chempakam, B. (2010) Toxicity prediction of compounds from health and disease. pp. 185–195, Springer, New York City, USA.
turmeric (Curcuma longa L). Food Chem. Toxicol. 48, 2951–2959. [87] Ranjbar-Mohammadi, M., Rabbani, S., Bahrami, S. H., Joghataei, M., and
[65] Ganiger, S., Malleshappa, H., Krishnappa, H., Rajashekhar, G., Rao, V. R., Moayer, F. (2016) Antibacterial performance and in vivo diabetic wound
et al. (2007) A two generation reproductive toxicity study with curcumin, tur- healing of curcumin loaded gum tragacanth/poly (ε-caprolactone) electro-
meric yellow, in Wistar rats. Food Chem. Toxicol. 45, 64–69. spun nanofibers. Mater. Sci. Eng. C 69, 1183–1191.
[66] Soleimani, V., Sahebkar, A., and Hosseinzadeh, H. (2018) Turmeric (Cur- [88] Sharma, M., Sahu, K., Singh, S. P., and Jain, B. (2018) Wound healing activ-
cuma longa) and its major constituent (curcumin) as nontoxic and safe sub- ity of curcumin conjugated to hyaluronic acid: in vitro and in vivo evalua-
stances. Phytother. Res. 32, 985–995. tion. Artif Cells Nanomed Biotechnol. 46, 1009–1017. https://doi.org/10.1080/
[67] Ravindranath, V., and Chandrasekhara, N. (1980) Absorption and tissue dis- 21691401.2017.1358731
tribution of curcumin in rats. Toxicology 16, 259–265. [89] Wong, V. W., Sorkin, M., Glotzbach, J. P., Longaker, M. T., and
[68] Sharma, R. A., McLelland, H. R., Hill, K. A., Ireson, C. R., Euden, S. A., Gurtner, G. C. (2010) Surgical approaches to create murine models of
et al. (2001) Pharmacodynamic and pharmacokinetic study of oral curcuma human wound healing. Biomed. Res. Int. 2011, 1–8.
extract in patients with colorectal cancer. Clin. Cancer Res. 7, 1894–1900. [90] Jagetia, G. C., and Rajanikant, G. K. (2012) Acceleration of wound repair by
[69] Shoba, G., Joy, D., Joseph, T., Majeed, M., Rajendran, R., et al. (1998) Influ- curcumin in the excision wound of mice exposed to different doses of frac-
ence of piperine on the pharmacokinetics of curcumin in animals and tionated γ radiation. Int. Wound J. 9, 76–92.
human volunteers. Planta Med. 64, 353–356. [91] Gopinath, D., Ahmed, M. R., Gomathi, K., Chitra, K., Sehgal, P., et al. (2004)
[70] Hsieh, C.-Y. (2001) Phase I clinical trial of curcumin, a chemopreventive Dermal wound healing processes with curcumin incorporated collagen
agent, in patients with high-risk or pre-malignant lesions. Anticancer Res. films. Biomaterials 25, 1911–1917.
21, 2895–2900. [92] Gong, C., Wu, Q., Wang, Y., Zhang, D., Luo, F., et al. (2013) A biodegradable
[71] Lao, C. D., Ruffin, M. T., Normolle, D., Heath, D. D., Murray, S. I., et al. (2006) Dose hydrogel system containing curcumin encapsulated in micelles for cutane-
escalation of a curcuminoid formulation. BMC Complement. Altern. Med. 6, 10. ous wound healing. Biomaterials 34, 6377–6387.
[72] Agrawal, S., and Goel, R. K. (2016) Curcumin and its protective and thera- [93] Fu, S. Z., Meng, X. H., Fan, J., Yang, L. L., Wen, Q. L., et al. (2014) Accelera-
peutic uses. Natl. J. Physiol. Pharm. Pharmacol. 6, 1–8. tion of dermal wound healing by using electrospun curcumin-loaded poly
[73] Goel, A., Kunnumakkara, A. B., and Aggarwal, B. B. (2008) Curcumin as (ε-caprolactone)-poly (ethylene glycol)-poly (ε-caprolactone) fibrous mats.
“Curecumin”: from kitchen to clinic. Biochem. Pharmacol. 75, 787–809. J. Biomed. Mater. Res. B Appl. Biomater. 102, 533–542.
[74] Akbik, D., Ghadiri, M., Chrzanowski, W., and Rohanizadeh, R. (2014) Curcu- [94] Yang, Z., He, C., He, J., Chu, J., Liu, H., et al. (2018) Curcumin-mediated
min as a wound healing agent. Life Sci. 116, 1–7. bone marrow mesenchymal stem cell sheets create a favorable immune
[75] Yen, Y. H., Pu, C. M., Liu, C. W., Chen, Y. C., Chen, Y. C., et al. (2018) Curcu- microenvironment for adult full-thickness cutaneous wound healing. Stem
min accelerates cutaneous wound healing via multiple biological actions: Cell Res. Ther. 9, 21.
the involvement of TNF-α, MMP-9, α-SMA, and collagen. Int. Wound J. 15, [95] Guo, R., Lan, Y., Xue, W., Cheng, B., Zhang, Y., et al. (2017) Collagen-
605–617. cellulose nanocrystal scaffolds containing curcumin-loaded microspheres

16 Curcumin in tissue engineering


on infected full-thickness burns repair. J. Tissue Eng. Regen. Med. 11, [112] Jain, S., Meka, S. R. K., and Chatterjee, K. (2016) Curcumin eluting nanofi-
3544–3555. bers augment osteogenesis toward phytochemical based bone tissue engi-
[96] Kant, V., Gopal, A., Pathak, N. N., Kumar, P., Tandan, S. K., et al. (2014) Anti- neering. Biomed. Mater. 11, 055007.
oxidant and anti-inflammatory potential of curcumin accelerated the cutane- [113] Saghiri, M. A., Orangi, J., Asatourian, A., Gutmann, J. L., Garcia-Godoy, F.,
ous wound healing in streptozotocin-induced diabetic rats. Int. et al. (2017) Calcium silicate-based cements and functional impacts of vari-
Immunopharmacol. 20, 322–330. ous constituents. Dent. Mater. J. 36, 8–18.
[97] Karri, V. V. S. R., Kuppusamy, G., Talluri, S. V., Mannemala, S. S., [114] Ding, S.-J., Shie, M.-Y., Hoshiba, T., Kawazoe, N., Chen, G., et al. (2010)
Kollipara, R., et al. (2016) Curcumin loaded chitosan nanoparticles impreg- Osteogenic differentiation and immune response of human bone-marrow-
nated into collagen-alginate scaffolds for diabetic wound healing. Int. derived mesenchymal stem cells on injectable calcium-silicate-based bone
J. Biol. Macromol. 93, 1519–1529. grafts. Tissue Eng. Part A 16, 2343–2354.
[98] Mitra, T., Manna, P. J., Raja, S., Gnanamani, A., and Kundu, P. (2015) [115] Chen, Y.-C., Shie, M.-Y., Wu, Y.-H. A., Lee, K.-X. A., Wei, L.-J., et al. (2017)
Curcumin loaded nano graphene oxide reinforced fish scale collagen–a Anti-inflammation performance of curcumin-loaded mesoporous calcium
3D scaffold biomaterial for wound healing applications. RSC Adv. 5, silicate cement. J. Formos. Med. Assoc. 116, 679–688.
98653–98665. [116] Sedghi, R., Sayyari, N., Shaabani, A., Niknejad, H., and Tayebi, T. (2018)
[99] Nguyen, V. C., Nguyen, V. B., and Hsieh, M.-F. (2013) Curcumin-loaded chit- Novel biocompatible zinc-curcumin loaded coaxial nanofibers for bone tis-
osan/gelatin composite sponge for wound healing application. Int. sue engineering application. Polymer 142, 244–255.
J. Polymer Sci. 2013, 1–7. [117] Kim, D. K., Kim, J. I., Sim, B. R., and Khang, G. (2017) Bioengineered porous
[100] Peddada, K. V., Peddada, K. V., Shukla, S. K., Mishra, A., and Verma, V. composite curcumin/silk scaffolds for cartilage regeneration. Mater. Sci.
(2015) Role of curcumin in common musculoskeletal disorders: a review of Eng. C 78, 571–578.
current laboratory, translational, and clinical data. Orthopaedic Surg. 7, [118] Fredberg, U., and Stengaard-Pedersen, K. (2008) Chronic tendinopathy tis-
222–231. sue pathology, pain mechanisms, and etiology with a special focus on
[101] Sahebkar, A. H., Panahi, Y., Sabouri, A., Rahimnia, A., Sharafi, M., inflammation. Scand. J. Med. Sci. Sports 18, 3–15.
et al. (2015) Efficacy and safety of supplementation with curcuminoids in [119] Riley, G. (2003) The pathogenesis of tendinopathy. A molecular perspective.
the treatment of patients with osteoarthritis: a randomized controlled trial. Rheumatology 43, 131–142.
Avicenna J. Phytomed. 5, 16–17. [120] Buhrmann, C., Mobasheri, A., Busch, F., Aldinger, C., Stahlmann, R.,
[102] Oh, S., Kyung, T.-W., and Choi, H.-S. (2008) Curcumin inhibits Osteoclasto- et al. (2011) Curcumin modulates nuclear factor κB (NF-κB)-mediated inflam-
genesis by decreasing receptor activator of nuclear factor-κB ligand mation in human tenocytes in vitro role of the phosphatidylinositol
(RANKL) in bone marrow stromal cells. Mol Cells. 26, 486–489. 3-kinase/akt pathway. J. Biol. Chem. 286, 28556–28566.
[103] von Metzler, I., Krebbel, H., Kuckelkorn, U., Heider, U., Jakob, C., et al. (2009) [121] Zhang, W., Li, X., Franchini, M. C., Xu, K., Locatelli, E., et al. (2016) Con-
Curcumin diminishes human osteoclastogenesis by inhibition of the trolled release of curcumin from curcumin-loaded nanomicelles to prevent
signalosome-associated IκB kinase. J. Cancer Res. Clin. Oncol. 135, 173–179. peritendinous adhesion during achilles tendon healing in rats. Int.
[104] Ozaki, K., Kawata, Y., Amano, S., and Hanazawa, S. (2000) Stimulatory J. Nanomed 11, 2873.
effect of curcumin on osteoclast apoptosis. Biochem. Pharmacol. 59, [122] Kim, S. E., Yun, Y.-P., Shim, K.-S., Jeon, D. I., Park, K., et al. (2018) In vitro
1577–1581. and in vivo anti-inflammatory and tendon-healing effects in Achilles tendi-
[105] Bharti, A. C., Takada, Y., and Aggarwal, B. B. (2004) Curcumin (diferuloyl- nopathy of long-term curcumin delivery using porous microspheres. J. Ind.
methane) inhibits receptor activator of NF-κB ligand-induced NF-κB activa- Eng. Chem. 58, 123–130.
tion in osteoclast precursors and suppresses osteoclastogenesis. [123] Stewart, J., Manmathan, G., and Wilkinson, P. (2017) Primary prevention of
J. Immunol. 172, 5940–5947. cardiovascular disease: a review of contemporary guidance and literature.
[106] Moran, J. M., Roncero-Martin, R., Rodriguez-Velasco, F. J., Calderon- JRSM Cardiovasc. Dis. 6, 2048004016687211.
Garcia, J. F., Rey-Sanchez, P., et al. (2012) Effects of curcumin on the prolif- [124] Wongcharoen, W., and Phrommintikul, A. (2009) The protective role of cur-
eration and mineralization of human osteoblast-like cells: implications of cumin in cardiovascular diseases. Int. J. Cardiol. 133, 145–151.
nitric oxide. Int. J. Mol. Sci. 13, 16104–16118. [125] Rivera-Espinoza, Y., and Muriel, P. (2009) Pharmacological actions of curcu-
[107] Moon, H.-J., Ko, W.-K., Han, S. W., Kim, D.-S., Hwang, Y.-S., et al. (2012) min in liver diseases or damage. Liver Int. 29, 1457–1466.
Antioxidants, like coenzyme Q10, selenite, and curcumin, inhibited osteo- [126] Belkacemi, A., Doggui, S., Dao, L., and Ramassamy, C. (2011) Challenges
clast differentiation by suppressing reactive oxygen species generation. Bio- associated with curcumin therapy in Alzheimer disease. Expert Rev. Mol.
chem. Biophys. Res. Commun. 418, 247–253. Med. 13, e34.
[108] Kim, W., Ke, K., Sul, O., Kim, H., Kim, S., et al. (2011) Curcumin protects [127] Scheltens, P., Blennow, K., Breteler, M. M. B., de Strooper, B., Frisoni, G. B.,
against ovariectomy-induced bone loss and decreases osteoclastogenesis. et al. (2016) Alzheimer’s disease. Lancet 388, 505–517.
J. Cell. Biochem. 112, 3159–3166. [128] Ma, J., Liu, J., Yu, H., Wang, Q., Chen, Y., et al. (2013) Curcumin promotes
[109] Yang, M.-W., Wang, T.-H., Yan, P.-P., Chu, L.-W., Yu, J., et al. (2011) Curcu- nerve regeneration and functional recovery in rat model of nerve crush
min improves bone microarchitecture and enhances mineral density in injury. Neurosci. Lett. 547, 26–31.
APP/PS1 transgenic mice. Phytomedicine 18, 205–213. [129] Ma, J., Yu, H., Liu, J., Chen, Y., Wang, Q., et al. (2016) Curcumin promotes
[110] Moon, D.-O., Kim, M.-O., Choi, Y. H., Park, Y.-M., and Kim, G.-Y. (2010) Cur- nerve regeneration and functional recovery after sciatic nerve crush injury
cumin attenuates inflammatory response in IL-1β-induced human synovial in diabetic rats. Neurosci. Lett. 610, 139–143.
fibroblasts and collagen-induced arthritis in mouse model. Int. Immuno- [130] Tsai, J.-R., Liu, P.-L., Chen, Y.-H., Chou, S.-H., Cheng, Y.-J., et al. (2015) Cur-
pharmacol. 10, 605–610. cumin inhibits non-small cell lung cancer cells metastasis through the adi-
[111] Shruti, S., Salinas, A. J., Ferrari, E., Malavasi, G., Lusvardi, G., et al. (2013) ponectin/NF-κb/MMPs signaling pathway. PLoS One. 10, e0144462.
Curcumin release from cerium, gallium and zinc containing mesoporous [131] Dance-Barnes, S. T., Kock, N. D., Moore, J. E., Lin, E. Y., Mosley, L. J., et al.
bioactive glasses. Microporous Mesoporous Mater. 180, 92–101. (2009) Lung tumor promotion by curcumin. Carcinogenesis 30, 1016–1023.

ahangari et al. 17

You might also like