You are on page 1of 16

View Article Online

View Journal

Journal of
Materials Chemistry B
Materials for biology and medicine

Accepted Manuscript

This article can be cited before page numbers have been issued, to do this please use: Y. Chen, J. Shi, Y.
Zhang, J. Miao, Z. Zhao, X. Jin, L. Liu, L. Yu, C. Shen and J. Ding, J. Mater. Chem. B, 2020, DOI:
10.1039/C9TB02523E.
Volume 6
Number 3
21 January 2018
This is an Accepted Manuscript, which has been through the
Pages 341-528
Royal Society of Chemistry peer review process and has been
Journal of accepted for publication.
Materials Chemistry B
Materials for biology and medicine
rsc.li/materials-b
Accepted Manuscripts are published online shortly after acceptance,
before technical editing, formatting and proof reading. Using this free
service, authors can make their results available to the community, in
citable form, before we publish the edited article. We will replace this
Accepted Manuscript with the edited and formatted Advance Article as
soon as it is available.

You can find more information about Accepted Manuscripts in the


Information for Authors.

Please note that technical editing may introduce minor changes to the
text and/or graphics, which may alter content. The journal’s standard
ISSN 2050-750X Terms & Conditions and the Ethical guidelines still apply. In no event
PAPER
Wei Wei, Guanghui Ma et al.
Macrophage responses to the physical burden of cell-sized
shall the Royal Society of Chemistry be held responsible for any errors
particles

or omissions in this Accepted Manuscript or any consequences arising


from the use of any information it contains.

rsc.li/materials-b
Page 1 of 15 Journal of Materials Chemistry B View Article Online
DOI: 10.1039/C9TB02523E

Journal of Materials Chemistry B RSCPublishing

ARTICLE

Journal of Materials Chemistry B Accepted Manuscript


An injectable thermosensitive hydrogel loaded with an
Cite this: DOI: 10.1039/x0xx00000x
ancient natural drug colchicine for myocardial repair
after infarction
Yu Chena,‡, Jiayue Shib,‡, Yaping Zhanga, Jiajun Miaob, Zhe Zhaoa, Xian Jina,
Published on 31 December 2019. Downloaded on 1/3/2020 1:26:29 AM.

Received 00th January 20xx, Liang Liua, Lin Yub,c,*, Chengxing Shena,*, Jiandong Dingb,c
Accepted 00th January 20xx

DOI: 10.1039/x0xx00000x Localized administration of anti-inflammatory agents benefits patients after myocardial infarction (MI) by
repressing/modulating inflammatory response of the MI region and thus accelerating repair of the impaired
www.rsc.org/
tissues. Colchicine (Col), an ancient natural drug, has excellent anti-inflammatory effects; however, its
utilization is strictly limited due to its severe systemic toxicity and narrow therapeutic window. In this
study, we developed an intramyocardial delivery system of Col using an injectable, thermosensitive
poly(lactide-co-glycolide)-poly(ethylene glycol)-poly(lactide-co-glycolide) (PLGA-PEG-PLGA)
polymer hydrogel as the vehicle for the treatment of MI while minimizing its systemic toxicity. The
aqueous PLGA-PEG-PLGA solution loaded with Col (Col@Gel) underwent a sol-gel transition at 35 °C
and maintained a gel state at body temperature. Col was released from the Col@Gel in an initial burst
followed by a sustained release manner for over 8 days. The in vitro cell tests showed that the Col@Gel
system significantly inhibited macrophage proliferation and migration. In a mouse model of MI, a single
intramyocardial administration of the Col@Gel effectively alleviated cardiac inflammation, inhibited
myocardial apoptosis and fibrosis, improved cardiac function and structure, and increased mouse survival
without inducing severe systemic toxicity, which was observed following intraperitoneal administration
of Col solution. These results suggested that the Col@Gel system is a reliable drug delivery system for the
sustained local release of Col and has great potential as an anti-inflammatory therapy for the treat of MI.

Introduction promotes cardiomyocyte apoptosis and accelerates extracellular

Acute myocardial infarction (MI) and follow-up induced heart matrix synthesis, leading to myocardial fibrosis, adverse ventricular

failure are one of the leading causes of death in many countries remodeling, and cardiac dysfunction.2-4 In view of the important roles

and remain a therapeutic challenge worldwide.1, 2 In addition to of inflammation in myocardial injury, increasing attention has been

ischemia and hypoxia-induced myocardial damage, post-MI paid to the potential benefits of anti-inflammatory therapies to

injury is exacerbated by excessive local inflammation, which repress/modulate local inflammation post-MI, thereby accelerating
myocardial repair, inhibiting left ventricular remodeling, and
improving patient prognosis.2, 5, 6
aDepartment of Cardiology, Shanghai Jiao Tong University Affiliated Sixth
People’s Hospital, Shanghai 200233, China. Colchicine (Col), an ancient natural drug extracted from Colchicum
E-mail: shencx@sjtu.edu.cn
bState Key Laboratory of Molecular Engineering of Polymers, Department of autumnale, was first described and utilized in Egypt more than 3000
Macromolecular Science, Fudan University, Shanghai 200438, China.
E-mail: yu_lin@fudan.edu.cn
years ago.7 Col prevents mitotic spindle formation, arrests
cZhuhai Fudan Innovation Institute, Zhuhai, Guangdong 51900, China
microtubule polymerization and inhibits crucial inflammatory

This journal is © The Royal Society of Chemistry 2019 J. Mater. Chem. B, 2019, 00, 1-3 | 1
Journal of Materials Chemistry B Page 2 of 15
View Article Online
DOI: 10.1039/C9TB02523E
Paper Journal of Materials Chemistry B

signaling pathways, such as inflammasomes, proinflammatory convenient synthesis, good reproducibility and wide recognition of
cytokines, as well as expression of adhesion molecules.7, 8 Therefore, safety.38, 39 It has been developed into a platform for the delivery of
Col has potent anti-inflammatory effects. Nowadays, Col is various kinds of drugs to treat systemic or regional diseases, including
recognized as a wonder drug for the treatment of refractory gout and anti-cancer treatments,40-42 anti-diabetic treatments,43-45 anti-
familial Mediterranean fever.7, 9 Col has also been tried to prevent osteopenia treatments,39 and others.46-50 For example, the local

Journal of Materials Chemistry B Accepted Manuscript


cardiovascular disease in clinic.6 However, the therapeutic window of administration of ropivacaine hydrochloride-loaded PLGA-PEG-
Col is narrow. Col with excessive dose can cause severe, even lethal, PLGA hydrogel was able to prolong its analgesic efficacy and thus
systemic toxicity including gastrointestinal disturbance and alleviated postoperative pain.49 Zhu et al. found that the
multiorgan failure,7, 10, 11 which significantly limited its clinic use, subconjunctival co-delivery of metformin and levofloxacin
even if it has a very long application history and well-characterized hydrochloride using PLGA-PEG-PLGA hydrogel as the depot
effects. effectively inhibited corneal neovascularization.50 Also, Ding and
Published on 31 December 2019. Downloaded on 1/3/2020 1:26:29 AM.

Several studies found that the gout patients treated with Col exhibited Chen developed a tumor microenvironment-sensitive PLGA-PEG-
a reduced prevalence of MI.12, 13 Two recent clinic trials revealed that PLGA-doxorubicin conjugate hydrogel and a synergistic anticancer
the patients with acute MI receiving the systemic administration of effect was achieved after intratumoral injection of the conjugate
Col showed a decrease in left ventricular remodeling or infarct size.14, hydrogel containing docetaxel.51 Recently, the Hotta’s group revealed
15 More recently, in a mouse model of MI, oral ingestion or that the introduction of laponite clay nanoparticles to an aqueous
intraperitoneal injection of Col had beneficial effects on myocardial solution of PLGA-PEG-PLGA could facilely modulate the sol-gel
repair.8, 16 These findings opened up a new avenue for the old drug in transition temperature from 25 °C to 37 °C, while the degradation rate
new use and provided a promising strategy for the treatment of acute of composite hydrogel was mainly dependent on the LA/GA ratio.52
MI. Of note, the risk of systemic toxicity remains a considerable 53 It is worth pointing out that the application of PLGA-PEG-PLGA
hurdle for the application of Col. One valid alternative to avoid the hydrogel for MI treatment is rarely reported.
systemic toxicity of Col is to realize the regional sustained delivery of In this study, we developed a sustained delivery of Col based on an
the drug. Meanwhile, advances in echocardiography and injectable thermosensitive PLGA-PEG-PLGA hydrogel for
thoracoscopy technologies have made intramyocardial injection of myocardial repair after MI. First, a PLGA-PEG-PLGA triblock
drugs feasible. copolymer with rational molecular composition was designed and
Injectable hydrogels formed by in situ gelation have received synthesized to ensure its temperature-responsive sol-gel transition in
increasing attention as vehicles for drug delivery and as scaffolds for water below body temperature. The delivery system of Col (labeled
tissue repair due to the unique superiority of minimally invasive Col@Gel) was then fabricated using the PLGA-PEG-PLGA hydrogel
application of these systems.17-26 In particular, temperature-induced as the vehicle, as shown in Fig. 1A. Next, in vitro release behavior and
gelling materials composed of poly(ethylene glycol) effects of the Col@Gel on macrophage proliferation and migration
(PEG)/polyester,27-30 PEG/polypeptide,31-33 poly(N- were evaluated. Finally, a mouse model of MI was constructed and
isopropylacrylamide-co-2- hydroxyethyl methacrylate-co- the safety and efficacy of intramyocardial injection of Col@Gel for
methacrylate-polylactide) (poly(NIPAAm-co-HEMA- co- the post-MI treatment was confirmed by analysis of mouse weight,
MAPLA),34, 35 poly(phosphazene)s copolymers36, 37 are low-viscous survival, liver function, cardiac structure and function, and
liquids at ambient temperature, but can rapidly convert into standing myocardial inflammation, apoptosis, and fibrosis. Fig. 1B
in situ gels in response to physiological temperature. Therefore, schematically presents the creation of mouse model of MI and the use
various therapeutic agents can readily be loaded into these materials of Col@Gel after MI.
by blending them in the sol state without drug loss. Upon in vivo
injection, the in situ forming hydrogels can offer sustained release of Materials and Methods
the encapsulated drugs.
Materials
So far, the thermosensitive and biodegradable poly(lactide-co-
Poly(ethylene glycol) (PEG) with molecular weight (MW) 1500 and
glycolide)-poly(ethylene glycol)-poly(lactide-co-glycolide) (PLGA-
stannous octoate (Sn(Oct)2) were purchased from Sigma-Aldrich (St.
PEG-PLGA) hydrogel enjoys great popularity because of its

2 | J. Name., 2019, 00, 1-3 This journal is © The Royal Society of Chemistry 2019
Page 3 of 15 Journal of Materials Chemistry B View Article Online
DOI: 10.1039/C9TB02523E
Journal Name ARTICLE

Journal of Materials Chemistry B Accepted Manuscript


Published on 31 December 2019. Downloaded on 1/3/2020 1:26:29 AM.

Fig. 1 A) The fabrication of Col@Gel system. The Col@Gel system exhibited a free-flowing sol at ambient temperature (25 °C) and
spontaneously converted into a semi-solid hydrogel at body temperature. This sol-gel transition was reversible. B) The establishment of
mouse MI model and the subsequent administration of Col@Gel system into three sites around the infarct region after MI. MI was
induced by ligating the left anterior descending coronary artery (LAD).
Louis, MO, USA). DL-Lactide (LA) and glycolide (GA) were supplied 0.01 mol) was placed in a three-necked flask and dried under vacuum
by Hangzhou Mingzhong Biotechnology Co. Ltd. (China) and used as with stirring at 125 °C for 3 h to remove moisture. LA and GA at a
received. given amount were added to the flask and the mixture was incubated
All animal experimental procedures were approved by the at 80 °C for 30 min. After LA and GA had melted, 0.08 g of Sn(Oct)2
Institutional Authority for Laboratory Animal Care of Shanghai Jiao was added, and the reaction mixture was heated with stirring at 150
Tong University Affiliated Sixth People’s Hospital and conformed to °C for 12 h under an argon atmosphere. The temperature was then
the Guide for the Care and Use of Laboratory Animals (National cooled to 120 °C with stirring for 3 h under vacuum to eliminate the
Research Council, Eighth Edition, 2010) and the AVMA Guidelines unreacted monomers. Subsequently, the crude polymers were poured
for the Euthanasia of Animals (2013 Edition). Male C57BL/6 mice (6 into 80 °C water to remove water-soluble oligomers and unreacted
weeks of age) were supplied by SLAC Laboratory Animal Co. Ltd. monomers. This washing procedure was carried out for at least three
(Shanghai, China). Mice were housed under appropriate humidity and times. Afterwards, residual water was removed by freeze drying. The
temperature conditions and received fed standard chow. Tap water purified products were harvested and stored at −20 °C until use and
was available ad libitum. the yield was approximately 80%.
The structure and composition of PLGA-PEG-PLGA was analyzed
Synthesis and characterization of PLGA-PEG-PLGA via 1H-nuclear magnetic resonance (NMR) on a 500 MHz proton

A ring-opening copolymerization approach using LA and GA spectrometer (DMX500 spectrometer, Bruker). Deuterated

monomers was used to prepare PLGA-PEG-PLGA triblock chloroform with the internal standard tetramethylsilane was selected

copolymers and PEG 1500 and Sn(Oct)2 were selected as macro- as the solvent. The value of molar mass dispersity (ĐM) of the polymer
initiator and catalyst, respectively.39, 47 Briefly, PEG 1500 (15.0 g, was analyzed by gel permeation chromatography (GPC; Agilent

This journal is © The Royal Society of Chemistry 2019 J. Name., 2019, 00, 1-3 | 3
Journal of Materials Chemistry B Page 4 of 15
View Article Online
DOI: 10.1039/C9TB02523E
Paper Journal of Materials Chemistry B

1260). Tetrahydrofuran was selected as the eluent, the flow rate was monitored for up to 20 days. Male C57BL/6 mice were anesthetized
set at 1 mL/min and the measurement temperature was fixed at 35 °C. with chloral hydrate and then injected subcutaneously with 0.1 mL of
Monodisperse polystyrene microspheres were used as standards. aqueous polymer solution (25 wt %) into the dorsal region. 3 mice
were euthanized at each specified time point and the remaining
Measurement of sol-gel transition temperature hydrogels were dissected. The appearance of the injected gels was

Journal of Materials Chemistry B Accepted Manuscript


The tube-inverting method39 was used to determine the sol-gel recorded with a digital camera, mass was measured using an analytical
transition point of aqueous polymer solution. 0.5 mL of polymeric balance, and MW was analyzed by GPC after freeze drying. The
aqueous solution was added into a 2-mL vial and equilibrated at 4 °C tissues around the administration sites were collected and subjected to
for 12 h. Subsequently, the capped vial was submerged in a water bath hematoxylin-eosin (HE) staining to examine the tissue responses.
and the temperature of water bath was increased from 20 to 50 °C at
a rate of 1 °C/step. At each temperature, the vial was incubated for 10 Cellular experiments
Published on 31 December 2019. Downloaded on 1/3/2020 1:26:29 AM.

min and then inverted by 180°. If the sample showed no evidence of Cell culture. The mouse macrophage cell line Raw264.7 was
flow within 30 s, it was considered to be a gel. supplied by the Cell Bank of the Chinese Academy of Sciences
The temperature-induced gelation behaviors of the aqueous polymer (Shanghai, China). The cells were incubated in DMEM medium
solutions in the presence or absence of Col were further measured (Gibco) containing fetal bovine serum (FBS, 10%) and penicillin-
using a dynamic rotational rheometer (Kinexus PRO, Malvern) with streptomycin (100 U/mL) under a 5% CO2 atmosphere at body
a cone plate (1° steel cone, 60 mm diameter). The time sweep program temperature.
was set at an oscillatory frequency of 10 rad/s and the temperature was In vitro cytotoxicity assay. The cytotoxicity of blank hydrogel and
increased from 15 °C to 50 °C at a heating rate of 1 °C/min. Col@Gel system against Raw264.7 cells was evaluated by measuring
cell viability with a Cell Counting Kit-8 (CCK-8, Dojondo) assay.
In vitro Col release Transwell chambers and a cell culture plate are shown in Fig. S1A.
The aqueous polymer solutions containing Col (0.25, 0.5 or 1 mg/mL) As presented in Fig. S1B, aqueous polymer solutions (25 wt%) with
were readily prepared by blending the aqueous polymer solution (25 or without Col (1.0 or 0 mg/mL) were added to Transwell chambers
wt%) with Col at 4 °C to form a homogenous solution. The Col-loaded at 100 μL/chamber and incubated at 37 C for 10 min to form
aqueous polymer solution maintained a free-flowing sol state at room hydrogels in situ. Raw264.7 cells were seeded into 24-well culture
temperature while a gel state at body temperature, which was labeled plates at a density of 1×104 cells/well in DMEM growth medium and
as Col@Gel. The in vitro release of Col from Col@Gel was incubated for 24 h. The cells were then washed for three times and
investigated and the method was implemented following the previous incubated in fresh DMEM. Next, the Transwell chambers containing
protocols.18, 45 Briefly, the samples (0.5 mL) were injected into 10 mL the gel samples were placed into the culture wells and the DMEM
vials, equilibrated at 4 °C for at least 12 h, and then incubated in a could get touch with the gel through the permeable membrane of
shaking water bath (50 rpm) at 37 °C for 10 min to form gels. Transwell chamber. After co-culture for 48 h, the cells were washed
Phosphate-buffered solution (PBS, pH 7.4) adding 0.025 wt % NaN3 with PBS, and CCK-8 reagent was then added to the wells in
was introduced at 5 mL/vial. At various time points, 3 mL of PBS was accordance with the instructions provided by the manufacturer.
withdrawn from the vials and replaced with 3 mL of prewarmed (37 Control cells were incubated in the same manner in the absence of gel-
°C) fresh PBS. The Col contents in the PBS samples were detected containing Transwell inserts, and their viability was set at 100% for
using a double beam UV-vis spectrophotometer (TU-1950). The UV the purpose of calculating relative cell viability. Samples were
absorbance differences at 354 nm between the Col-containing analyzed in triplicate.
hydrogel samples and the Col-free blank hydrogel samples were In vitro cell migration assay. In vitro migration of Raw264.7
converted into the amounts of Col released at different time points by macrophages was measured using a Transwell assay. As presented in
comparison with the standard curve. Fig. S1C, aqueous polymer solutions (25 wt%) with or without Col
(1.0 or 0 mg/mL) were added to cell culture plates and then kept at 37
In vivo hydrogel degradation
℃ to form gels in situ. Each well with 1 mL of hydrogel was incubated
Changes in visual appearance, mass, and MW of the PLGA-PEG-
in DMEM (1 mL) at 37 C for 24 h and subsequently the DMEM was
PLGA hydrogel after subcutaneous injection into mice were

4 | J. Name., 2019, 00, 1-3 This journal is © The Royal Society of Chemistry 2019
Page 5 of 15 Journal of Materials Chemistry B View Article Online
DOI: 10.1039/C9TB02523E
Journal Name ARTICLE

collected. Raw264.7 cells (1×104 cells in the 100 μL of DMEM retro-orbitally and centrifuged to obtain serums. Serum
collected) were added to the Transwell chambers, the chambers were concentrations of BNP, ALT, and AST were measured using a BNP
placed in new cell culture plates, and an additional 600 μL of fresh ELISA Assay Kit (FineTest, Wuhan, China) or a Catalyst One®
DMEM was added per well. After incubation of 48 h, the chambers Chemistry Analyzer (IDEXX, Westbrook, USA) according to the
were taken out, washed using PBS, and the cells on the membrane of manufacturers’ instructions.

Journal of Materials Chemistry B Accepted Manuscript


chamber were fixed with 4% phosphate-buffered paraformaldehyde Myocardial tissue collection. Following collection of blood
for 1 h at ambient temperature and then crystal violet was used to stain samples, the anesthetized mice were euthanized and the hearts were
the cells. Cells on the upper surface of the membrane were eliminated collected. Samples of ventricular myocardium were excised, fixed in
with a cotton swab, and cells on the lower surface (migrated) were formalin, embedded in paraffin, cut into 5-μm-thick sections, and
observed under an OLYMPUS microscope (Japan). mounted on slides for analysis of collagen deposition, inflammation,
and apoptosis by Masson’s trichrome, F4/80 immunofluorescence,
Published on 31 December 2019. Downloaded on 1/3/2020 1:26:29 AM.

Animal experiments and terminal deoxynucleotidyl transferase dUTP nick end labeling
In vivo application of the Col@Gel in a mouse model of MI. (TUNEL) staining, respectively.
Male C57BL/6 mice were randomized into four groups (15 per Analysis of tissue fibrosis. Myocardial tissue sections were stained
group): Sham, MI control, MI + Col@Gel, and MI + Col (i.p.). MI with Masson’s trichrome. Sections were observed using an
was induced by ligating the LAD of mouse. The Sham group OLYMPUS microscope (Japan). The fibrotic area within each tissue
underwent surgery but the LAD was not ligated. The MI control group section was measured and analyzed via ImageJ software 1.48
underwent MI surgery but did not receive any treatment. The (National Institutes of Health, Bethesda, MD, USA).
Col@Gel group received intramyocardial injections of 1 mg/mL Col- TUNEL assay. A TUNEL assay was employed to detect apoptotic
loaded polymer solution (10 μL) into three sites around the infarct cells in myocardial tissues. The sections were probed using an
region immediately after LAD ligation. The Col (i.p.) group received TUNEL detection kit (Beyotime Biotechnology, Shanghai, China)
a single intraperitoneal injection of Col solution at the same dose. according to the manufacturer’s recommendations. Three images of
Electrocardiography was performed using a Biological Signal the infarct zone in each section were acquired using an OLYMPUS
Acquisition System (Techman, Chengdu, China) during LAD microscope (Japan). The number of TUNEL-positive and total
ligation. cardiomyocyte nuclei in each zone were counted using ImageJ
The ST-segment elevation of electrocardiogram suggested that MI software, and the percentage of apoptotic cardiomyocytes was
was successfully modeled, as illustrated in Fig. S2. The mice were calculated.
monitored three times per day during the 4-week follow-up period. Immunofluorescence staining of macrophages. Myocardial
The weights per week and the death of mice post-MI were recorded. inflammation was assessed by counting the number of F4/80-positive
Echocardiography measurement. At day 28 after MI, the survival macrophages in tissue sections. After deparaffinization, antigen
mice were shaved in the anterior and left lateral thoracic regions and unmasking, and permeabilization, the myocardial sections were
then placed in the supine position. Ultrasound transonic gel was incubated overnight at 4 °C with anti-F4/80 antibody (1:700 dilution,
smeared on the thoracic regions to optimize visibility. Two- CST, Danvers, USA), washed, and incubated with a goat anti-mouse
dimensional and m-mode echocardiographic images were acquired Alexa Fluor 488-labeled antibody (1:400 dilution, Jackson, West
using echocardiography. Left ventricular internal dimensions were Grove, USA) for 1 h at 37 C in the dark. Cell nuclei were
recorded during systole (LVIDs) and diastole (LVIDd). The ejection counterstained with 4',6-diamidino-2-phenylindole (DAPI) for 10 min
fraction (EF) and fractional shortening (FS) were calculated from the at room temperature. Three images of the infarct zone in each section
measured parameters. were acquired using an OLYMPUS microscope and the number of
Brain natriuretic peptide (BNP) and liver function tests. To F4/80-positive cells per mm2 were counted using ImageJ software.
assess serum levels of BNP, a biomarker of heart failure,54, 55 and Tumor necrosis factor-α (TNF-α) assay. The myocardial content
alanine transaminase (ALT) and aspartate aminotransferase (AST), of TNF-α was evaluated using a Mouse TNF-α ELISA kit (Sigma-
two biomarkers of liver function,56 the mice were anesthetized using Aldrich). Another gang of male C57BL/6 mice that underwent surgery
isoflurane at day 28 post-treatment and blood samples were sampled and then received different treatments were likewise divided into four

This journal is © The Royal Society of Chemistry 2019 J. Name., 2019, 00, 1-3 | 5
Journal of Materials Chemistry B Page 6 of 15
View Article Online
DOI: 10.1039/C9TB02523E
Paper Journal of Materials Chemistry B

groups: Sham, MI control, MI + Col@Gel, and MI+Col (i.p.). 3 mice undergoes deformation, while G″ denotes the viscous behavior. At
per group were euthanized each week, the hearts were removed, and low temperatures, G″ was higher than G′, suggesting a free-flowing
the myocardial tissues in the infarct zone were excised and stored at - liquid state and good injectability. As the temperature increased, both
80°C until analysis. Then, the stored myocardial tissues were of them increased, but G′ increased more rapidly. The sol-gel
homogenized five times in ice-cold lysis buffer for 1 min each time transition point was observed at 35 °C, when G′ was equal to G″.

Journal of Materials Chemistry B Accepted Manuscript


and centrifuged at 10000 g for 20 min at 4 °C. The supernatants were Meanwhile, the addition of 1.0 mg/mL Col had virtually no effect on
collected and TNF-α content was measured using the ELISA kit the rheological curves of the polymer/water system as a function of
according to the manufacturer’s instruction. The protein temperature.
concentrations in the supernatants were also measured, and the data
are presented as the TNF-α content in pg per 200 μg of protein.
Published on 31 December 2019. Downloaded on 1/3/2020 1:26:29 AM.

Statistical analysis
Data are presented as the mean ± standard deviation (SD). Group
means were compared via one-way analysis of variance or unpaired t-
test. A p value of <0.05 was considered statistically significant.

Results

Synthesis and characterization of the PLGA-PEG-PLGA triblock


copolymers
The PLGA-PEG-PLGA triblock copolymers were obtained through
ring-opening polymerization of LA and GA in the presence of PEG Fig. 2 Elastic modulus (G′) and viscous modulus (G″) of the
1500, which contained two hydroxyl end groups, with Sn (Oct)2 as the aqueous polymer solutions in the presence or absence of Col. The
catalyst. The structure and composition of PLGA-PEG-PLGA were concentration of the polymer solution was 25 wt % and the drug
assayed by 1H-NMR (Fig. S3). This analysis gave a number-average loading amount was 1.0 mg/mL.
molecular weight (Mn) of up to PLGA1850-PEG1500-PLGA1850 and a
molar ratio of LA:GA of 1:1. The ĐM value of PLGA-PEG-PLGA In vitro Col release
was determined to be 1.30 by GPC analysis using monodisperse The in vitro release rate of Col was tested using three Col@Gel
polystyrene microspheres as standards. These characterizations systems loaded with various amounts of Col. The results are shown in
confirmed the successful preparation of expected sample. Fig. 3. All of the three Col@Gel systems exhibited a burst in the first
12 h and about 21 wt% of loaded Col was released, suggesting that
Sol-gel transition of the PLGA-PEG-PLGA aqueous solution the loaded drug could be rapidly released and then delivered to the
The temperature-induced sol-gel transition of the PLGA-PEG-PLGA adjacent lesions after local injection in vivo. Thereafter, Col continued
aqueous solution was first investigated by the tube-inverting method to be released over the next week, and more than 90 wt % of the loaded
and the phase-transition diagram was obtained, as shown in Fig. S4. drug was released within 8 days. The Col release profiles of the three
Above a critical gel concentration (approximately 12 wt%), the Col@Gel systems were similar, indicating a concentration-
aqueous polymer solution underwent three physical states, namely, independent of release dynamics.
sol, gel, and sol (suspension), with an increase in temperature. In vivo degradation of the PLGA-PEG-PLGA hydrogel
The thermogelling properties of the 25 wt% PLGA-PEG-PLGA To determine how the in vivo environment affected hydrogel
aqueous solutions in the presence or absence of Col were further degradation, the 25 wt% PLGA-PEG-PLGA aqueous solution was
assessed using dynamic rheological analysis. As shown in Fig. 2, the injected into the subcutaneous layer of back of C57BL/6 mouse,
changes in the elastic modulus (G′) and viscous modulus (G″) of the which rapidly converted into a semi-solid hydrogel within 30 s at the
aqueous polymer solutions upon heating were monitored. Generally,
G′ represents the elastic behavior and energy storage when a sample

6 | J. Name., 2019, 00, 1-3 This journal is © The Royal Society of Chemistry 2019
Page 7 of 15 Journal of Materials Chemistry B View Article Online
DOI: 10.1039/C9TB02523E
Journal Name ARTICLE

administration site in response to the body heat. As displayed in Fig.


4A, the in situ-forming hydrogel showed an oval or flat shape and was
maintained for over 20 days, but its volume gradual decreased over
time. At predetermined time points, the remaining hydrogels were

Journal of Materials Chemistry B Accepted Manuscript


excised from the mice after euthanasia, and the mass was then
weighed. The mass percentage of remaining gel is shown in Fig. 4C.
This analysis showed a consistent decrease in mass as the time
prolonged, and only 20 % of the initial mass of the hydrogel was left
after 20 days of in vivo degradation. Meanwhile, the excised gels were
freeze-dried and then analyzed by GPC (Fig. 4D). The results revealed
Published on 31 December 2019. Downloaded on 1/3/2020 1:26:29 AM.

that the MWs of the recovered PLGA-PEG-PLGA polymers gradually


reduced, while the ĐMs continued to increase, confirming the in vivo
Fig. 3 Cumulative drug release curves of Col@Gel systems with degradation feature of PLGA-PEG-PLGA polymers.
different amounts of Col. The results were presented as mean ± SD Moreover, the tissues around the hydrogels were collected and
(n=3). The photographs of the Col@Gel system containing 1 subjected to HE staining to examine the tissue responses (Fig. 4B).
mg/mL Col were taken at preinstalled time points. No severe inflammatory response was observed at the various time

Fig. 4 In vivo degradation and biocompatibility of the thermosensitive PLGA-PEG-PLGA hydrogel. A) In vivo hydrogel maintenance.
The pictures were shoot at various time points after dorsal subcutaneous injection. B) Optical micrographs of HE-stained slices of
surrounding tissues at designated time points after subcutaneous administration of hydrogel. M: muscle; G: hydrogel; S: skin; F: fibrous
capsule. C) The mass percentage of remaining gel excised from C57BL/6 mice measured by the gravimetric method. D) GPC traces of
residual PLGA-PEG-PLGA polymers collected from the excised gels during the in vivo degradation.

This journal is © The Royal Society of Chemistry 2019 J. Name., 2019, 00, 1-3 | 7
Journal of Materials Chemistry B Page 8 of 15
View Article Online
DOI: 10.1039/C9TB02523E
Paper Journal of Materials Chemistry B

Journal of Materials Chemistry B Accepted Manuscript


Published on 31 December 2019. Downloaded on 1/3/2020 1:26:29 AM.

Fig. 5 A) The effect of Col@Gel on the proliferation of Raw264.7 macrophages in vitro after 48 h of incubation (n=3). The viability of
Raw264.7 cells that did not receive any treatment was defined as 100%. B) The effect of Col@Gel on the migration of Raw264.7 cells after 48
h of incubation (n=3). ***p<0.001. C) Representative micrographs of migrated Raw264.7 cells. Raw264.7 cells were stained purple using
crystal violet. The cells that received different treatments migrated from the upper surface of membrane of chamber to the lower surface of
membrane and then the lower surface of membrane was photographed after 48 h of incubation.
points. Apparently, the in vivo biodegradability and biocompatibility
of the PLGA-PEG-PLGA hydrogel supports the feasibility and safety
of clinical application as a long-acting drug release system.

Effects of the Col@Gel on macrophage proliferation and


migration in vitro
Macrophage is one of the most vital inflammatory cells involved in
MI.57, 58 To determine the cytotoxicity of blank hydrogel and
Col@Gel against macrophages, we first measured their effects on the
proliferation of Raw264.7 macrophages in vitro using the CCK-8
Fig. 6 The cumulative survival rate of mice that received the
assay. As shown in Fig. 5A, the viability of Raw264.7 cells that
different treatments post-MI.
received the treatment of the blank hydrogel for 48 h did not suffer
from any influence, indicating that the hydrogel itself was
biocompatible, which did not interfere with the proliferation of Effects of the Col@Gel on a mouse model of MI
Raw264.7 cells. However, incubation with the Col@Gel containing Post-operative survival
1.0 mg/mL Col for 48 h led to a remarkable decrease in the viability A mouse model of MI was constructed and then the mice received the
of Raw264.7 cells. different treatments. Post-operative survival of mice was observed for
Furthermore, the effect of the Col@Gel treatment on macrophage 4 weeks and the results are shown in Fig. 6. None of the mice in the
migration was assessed using the Transwell assay. As presented in Sham group died, whereas 7 mice in the MI group died (days 3, 9, 16,
Fig. 5B and 5C, the migration of Raw264.7 cells treated with the 20, 23, 23, and 26) and the overall survival rate was only 53.3%. The
Col@Gel system for 48 h was significantly inhibited compared with survival rate of the Col@Gel group was markedly higher than that of
the blank hydrogel-treated group. These outcomes affirmed that the the MI group (86.7% vs 53.3%, p<0.05), and just 2 mice died at the
Col released from the gel depot could remarkably suppress the late stage of the experiment (days 20 and 26). The number of mice
proliferation and migration of Raw264.7 macrophages. that survived to day 28 in the Col (i.p.) group (73.3%) was also lower

8 | J. Name., 2019, 00, 1-3 This journal is © The Royal Society of Chemistry 2019
Page 9 of 15 Journal of Materials Chemistry B View Article Online
DOI: 10.1039/C9TB02523E
Journal Name ARTICLE

Journal of Materials Chemistry B Accepted Manuscript


Fig. 7 The systemic toxicity of Col. A) Weight changes of mice receiving the indicated treatments at 4 weeks post-MI. B) Serum ALT
and AST levels of mice at 4 weeks post-MI. n=the survival mice number per group at 4 weeks post-MI, specially, nSham=15, nMI=8,
Published on 31 December 2019. Downloaded on 1/3/2020 1:26:29 AM.

nCol@Gel=13, nCol (i.p.)=11. *p<0.05, ***p<0.001.


than that in the Col@Gel group, although the difference between them
did not reach statistical significance.

In vivo toxicity
The systemic toxicity of Col in vivo was assessed by recording the
changes in body weights and serum ALT and AST levels of mice. As
shown in Fig. 7A, the mice in the Col@Gel group gained a mean
weight increase of 3.42±0.5 g after MI for 4 weeks, which was not
significantly different from the gain in either the Sham group or the
MI group. The Col (i.p.) group gained significantly less weight than
the other three groups (2.95±0.4, p<0.05), illustrating the
intraperitoneal administration of Col solution caused the remarkably
systemic toxicity compared with the intramyocardially injected
Col@Gel. In a similar way, serum AST and ALT levels in the Col
(i.p.) group were significantly higher than those in the other three
groups (p<0.001), suggesting that the systemic exposure to Col
induced severe liver injury (Fig. 7B). It is worth noting that the slight
increases in AST and ALT levels in the MI and Col@Gel groups
compared with the Sham group may be attributed to the MI surgery
itself.

Fig. 8 Myocardial inflammation of mice receiving the indicated Myocardial inflammation


treatments at 4 weeks post-MI. A) Immunofluorescence-stained As macrophages are key players in MI-associated inflammation,
myocardial tissues. F4/80-positive macrophages were stained F4/80, a macrophage marker in myocardial tissue, was stained with
green with Alexa Fluor 488-labeled antibody, and cell nuclei immunofluorescence. As shown in Fig. 8A, lots of macrophages were
were counterstained blue with DAPI. B) Statistical analysis of the detected in the myocardium of mice with MI compared with the Sham
abundance of F4/80-positive macrophages in the myocardium of group; however, the abundance of macrophages in mice treated with
each group. n=the survival mice number per group at 4 weeks Col@Gel after MI was significantly reduced. Fig. 8B further presents
post-MI, specially, nSham=15, nMI=8, nCol@Gel=13, nCol (i.p.)=11. the statistical results. The number of F4/80-positive cells per mm2 was
***p<0.001. 9±3 in the Sham group, 1031±133 in the MI group (p<0.001 vs Sham),
249±33 in the Col@Gel group (p<0.001 vs MI), and 448±59 in the

This journal is © The Royal Society of Chemistry 2019 J. Name., 2019, 00, 1-3 | 9
Journal of Materials Chemistry B Page 10 of 15
View Article Online
DOI: 10.1039/C9TB02523E
Paper Journal of Materials Chemistry B

Col (i.p.) group (p<0.001 vs MI). More importantly, the number of


macrophages in the myocardial sections of the Col@Gel-treated mice
was significantly lower than that in the Col (i.p.)-treated mice
(p<0.001).
To further confirm the aforementioned findings, we performed an

Journal of Materials Chemistry B Accepted Manuscript


additional experiment to measure the TNF-α content in myocardial
tissue extracts. TNF-α is produced by a variety of cells, including
neutrophils and macrophages, and it plays an important role not only
in MI-related inflammation response but also in myocardial apoptosis
and fibrosis.59, 60 As shown in Fig. 9, the TNF-α level in the MI group

Fig. 9 Changes of myocardial TNF-α levels of mice that contributed to the timely suppression of the overactive acute
Published on 31 December 2019. Downloaded on 1/3/2020 1:26:29 AM.

received different treatments after MI (n = 3 per time point). inflammatory response as well as the subsequent chronic
inflammation in the infarct region. In contrast, the anti-inflammation
effect was much weaker after a single intraperitoneal injection of Col
solution.

Myocardial fibrosis
peaked at 1 week post-MI and gradually declined thereafter. Both the
Col@Gel and Col (i.p.) treatments significantly reduced the
myocardial TNF-α content at each time point examined, and the use
of Col@Gel was far more effective in suppressing TNF-α production
than that of Col (i.p.) (p<0.05). Combined with the results presented
in Fig. 3, we believed that the sustained release of Col for up to 1 week
To detect MI-induced myocardial fibrosis, myocardial tissues were
stained with Masson’s trichrome to reveal collagen fibers. As
displayed in Fig. 10A, the fibrosis degree of myocardial tissue in the
infarction zone of mice in the MI group was significantly higher than
that in the Sham group, while that was significantly reduced in the
mice treated with Col@Gel. Specifically, the proportion of total tissue
area with evidence of fibrosis was 1.03±0.07% in the Sham group,
24.83±3.55% in the MI group (p<0.001 vs Sham), 11.53±2.50% in the
Col@Gel group (p<0.001 vs MI), and 19.47±2.93% in the Col (i.p.)
group (p<0.001 vs MI group) (Fig. 10B). Also, the fibrosis area of
myocardial tissue in the Col@Gel group was significantly lower than
that in the Col (i.p.) group, indicating that the intramyocardial
injection of Col@Gel and the subsequent sustained release of Col was
more effective in reducing myocardial fibrosis post-MI than the

Fig. 10 The myocardial fibrosis degree of mice that received intraperitoneal injection of Col solution.

different treatments at 4 weeks post-MI. A) Masson’s trichrome-


stained myocardial tissues. B) Statistical analysis of the
Myocardial apoptosis
myocardial fibrosis area of each group. n=the survival mice
Myocardial apoptosis caused by LAD ligation was assessed by the
number per group at 4 weeks post-MI, specially, nSham=15,
TUNEL assay. This analysis revealed that the MI operation
nMI=8, nCol@Gel=13, nCol (i.p.)=11. ***p <0.001.
significantly caused myocardial apoptosis in the MI group compared

10 | J. Name., 2019, 00, 1-3 This journal is © The Royal Society of Chemistry 2019
Page 11 of 15 Journal of Materials Chemistry B View Article Online
DOI: 10.1039/C9TB02523E
Journal Name ARTICLE

Journal of Materials Chemistry B Accepted Manuscript


Published on 31 December 2019. Downloaded on 1/3/2020 1:26:29 AM.

Fig. 11 The myocardial apoptosis of mice that received different


treatments at 4 weeks post-MI. A) TUNEL-stained myocardial
Fig. 12 The cardiac structure and function of mice that received
tissues. Apoptotic cell nuclei in the myocardium were stained
different treatments at 4 weeks post-MI. A) Representative
green (TUNEL-positive). The total cardiomyocyte nuclei were
echocardiographic images. B) Statistical analysis of the
stained blue with DAPI. B) Statistical analysis of the percentage of
echocardiographic parameters of each group. n=the survival mice
apoptotic cardiomyocytes (TUNEL-positive cells) of each group.
number per group at 4 weeks post-MI, specially, nSham=15, nMI=8,
n=the survival mice number per group at 4 weeks post-MI,
nCol@Gel=13, nCol (i.p.)=11. *p<0.05, ***p<0.001.
specially, nSham=15, nMI=8, nCol@Gel=13, nCol (i.p.)=11. ***p <0.001
echocardiographic images and Fig. 12B displays the statistical
with the Sham group, but here we observed that the Col@Gel outcomes. In the MI group, EF and FS were significantly decreased
treatment significantly alleviated apoptosis of cardiomyocytes (Fig. (p<0.001), while LVIDs and LVIDd were significantly increased
11A). As shown in Fig. 11B, the percentage of apoptotic (p<0.001) compared with the Sham group, indicating severe MI-
cardiomyocytes was 0.25±0.06% in the Sham group, 12.93±2.00% in induced structural and functional damage to the myocardium. In the
the MI group (p<0.001 vs Sham), 4.44±1.08% in the Col@Gel group Col@Gel group, all of these echocardiographic parameters were
(p<0.001 vs MI), and 9.04±1.44% in the Col (i.p.) group (p<0.001 vs significantly improved compared with the MI group (p<0.05 or
MI and Col@Gel). These findings mirrored the analysis of tissue p<0.001), whereas only EF and FS were improved in the Col (i.p.)
fibrosis, as presented in Fig. 10, and revealed that treatment with group (p<0.05 or p<0.001). Apparently, the Col@Gel treatment post-
Col@Gel provided more benefits in inhibiting apoptosis of MI had a strikingly beneficial effect on myocardial structure and
cardiomyocytes after MI than Col by intraperitoneal administration. function, whereas the response to Col (i.p.) treatment was much
Cardiac structure and function weaker.
Changes in cardiac structure and function following MI induction
Heart failure biomarker
were assessed by echocardiography. Fig. 12A shows representative

This journal is © The Royal Society of Chemistry 2019 J. Name., 2019, 00, 1-3 | 11
Journal of Materials Chemistry B Page 12 of 15
View Article Online
DOI: 10.1039/C9TB02523E
Paper Journal of Materials Chemistry B

Finally, serum concentrations of BNP, a biomarker of congestive confirmed that oral or intraperitoneal administration of Col had anti-
heart failure, were measured at 4 weeks post-MI (Fig. 13). Compared inflammatory and anti-fibrotic effects on experimentally induced MI
with the Sham group, the MI group showed a significantly higher BNP in mice, but their findings on improvement of cardiac function were
level (p<0.001), as expected. However, the serum BNP levels were inconsistent.8, 16 Moreover, they were unable to draw a clear
significantly lower in both the Col@Gel- and Col (i.p.)-treated groups conclusion about the systemic toxicity of Col in their studies.

Journal of Materials Chemistry B Accepted Manuscript


compared with the MI group (p<0.001). It was exciting that the BNP Considering that its systemic toxicity is the greatest obstacle for Col
level in the Col@Gel group was much lower than that in the Col (i.p.) use in clinic, we speculated that localized administration of Col maybe
group (p<0.001). This finding further confirmed the superiority of a provide better therapeutic efficacy while significantly alleviating the
locally deposited Col@Gel system in preventing heart failure side effects. With the development of echocardiographic and
compared with a systemically administered Col solution. thoracoscopic technologies, intramyocardial administration of anti-
inflammatory drugs has become a potentially feasible strategy for
Published on 31 December 2019. Downloaded on 1/3/2020 1:26:29 AM.

post-MI treatment.
In the current work, we constructed an injectable drug delivery
system, enabling the localized, sustained release of Col in the
myocardium. First, we prepared a thermosensitive hydrogel
composed of PLGA-PEG-PLGA polymers that was an injectable
liquid at room temperature and converted into a non-flowing gel
within 30 s when exposed to body temperature. Col was readily,
homogenously dispersed in the aqueous polymer solution by blending
them at low or ambient temperature. The loading of Col did not
influence the injectability and temperature-induced gelation of
Fig. 13 The serum BNP concentrations of mice that received
polymer/water system (Fig. 2). The acute stage in the first 7 days after
different treatments at 4 weeks post-operation. n=the survival mice
an MI is the crucial period for myocardial repair as well as the peak
number per group at 4 weeks post-MI, specially, nSham=15, nMI=8,
period of myocardial necrosis, apoptosis, and inflammation
nCol@Gel=13, nCol (i.p.)=11. ***p <0.001.
response.64, 65 The in vitro release of Col from the Col@Gel system
exhibited an initial burst followed by a sustained release pattern for
Discussion
over 8 days (Fig. 3), suggesting that the liberated Col could rapidly
Overactive, persistent tissue inflammatory response post-MI, which
exert the anti-inflammatory effect upon injection as well as
not only disturbs tissue repair but also induces myocardial apoptosis,
throughout the follow-up crucial acute stage post-MI. This feature was
necrosis, fibrosis and leads to ventricular remodeling and cardiac
well confirmed by sustained, effective inhibition of the expression of
insufficiency, is a major cause of myocardial injury.2-4 Therefore, the
the inflammatory cytokine TNF-α following the intramyocardial
timely suppression of excess early myocardial inflammatory response
administration of the Col@Gel system post-MI (Fig. 9).
is in favor of promoting and accelerating myocardial repair of patients
The in vitro cell tests showed that the Col released from the Col@Gel
after MI.2, 3, 61, 62 For example, some clinic studies have demonstrated
effectively inhibited the proliferation and migration of macrophages
that administration of anti-inflammatory drugs, such as canakinumab,
(Fig. 5), which make vital negative contributions to cardiac
an anti-IL-1β monoclonal antibody, has beneficial effects on the
remodeling post-MI.57, 58 On the basis of demonstrating the in vivo
prognosis of MI patients.2, 63 However, the studies on the anti-
biodegradability and biocompatibility of PLGA-PEG-PLGA hydrogel
inflammatory remedy for patients after MI are still limited compared
itself (Fig. 4), the effect of Col@Gel system on myocardial repair of
with the other therapies, such as antiplatelet and lipid-lowering
mice after infarction was evaluated. We found that a single
treatments.
intramyocardial administration of the Col@Gel post-MI and the
Col is a mitotic spindle poison utilized for more than 3000 years and
follow-up sustained release of Col significantly reduced the
has pleiotropic anti-inflammatory properties, but the widespread
accumulation of macrophages and inhibited myocardial apoptosis and
clinical use is precluded because of its severe toxicity profile and
fibrosis in the infarct region, lowered the expression of the heart
narrow therapeutic window.7, 10, 11 Recently, Fujisue and Akodad

12 | J. Name., 2019, 00, 1-3 This journal is © The Royal Society of Chemistry 2019
Page 13 of 15 Journal of Materials Chemistry B View Article Online
DOI: 10.1039/C9TB02523E
Journal Name ARTICLE

failure biomarker BNP, improved the cardiac structure and function, tests confirmed the safety and efficacy of Col@Gel for the treatment
and prolonged the survival of mice (Figs. 6, 8, 10-13). Apparently, of MI after intramyocardial injection were significantly superior to
intramyocardial injection of the Col@Gel imparted multiple benefits those of intraperitoneal injection of Col solution, including lower
at the most critical stage of treatment after MI. myocardial inflammation, apoptosis, and fibrosis, better heart
Furthermore, we compared the safety and efficacy of Col function and structure, higher animal survival and negligible systemic

Journal of Materials Chemistry B Accepted Manuscript


administered by intramyocardial injection and intraperitoneal side effects. Overall, the local sustained delivery of Col offers a new
injection. On the one hand, we found that a single intraperitoneal perspective on the old drug in new use and the Col@Gel system has
injection of the Col solution has some beneficial effects on anti- great potential for promoting tissue repair after MI.
inflammation and improvement of cardiac function, which was
consistent with the Fujisue’s study.16 However, the effects were less Conflicts of interest
striking than those obtained from intramyocardial injection of the The authors declare no competing financial interest.
Published on 31 December 2019. Downloaded on 1/3/2020 1:26:29 AM.

Col@Gel, possibly because of poor local uptake and accumulation of


Col at the infarct region after a single systemic administration. On the Acknowledgements
other hand, a single intraperitoneal injection of the Col solution still This work was supported by the Natural Science Foundation of China
caused systemic toxicity, such as liver damage and reduced weight (grant nos. 81603327, 81670321, 51773043 and 51533002), China
gain (Fig. 7), that were negligible in the mice treated with Col@Gel. Postdoctoral Science Foundation (grant no. 2018M632144), the
In addition, the survival rate of mice in the Col (i.p.) group after MI Research Fund of Shanghai Jiao Tong University Affiliated Sixth
was worse compared with the Col@Gel group, which also reflected People’s Hospital (No. 0004) and National Key R&D Program of
either inferior myocardial efficacy or systemic toxicity. In contrast, a China (grant no. 2016YFC1100300).
single intramyocardial administration of the Col@Gel and the follow-
up continuous liberation of Col ensured the high local drug References
concentration in the myocardium while minimizing the systemic 1. D. P. Zipes, P. Libby, R. O. Bonow, D. L. Mann and G. F.
Tomaselli, Braunwald's Heart Disease E-Book: A Textbook of
toxicity, both of which are beneficial to the patients post-MI. Cardiovascular Medicine, Elsevier Health Sciences, 2018.
2. S. B. Ong, S. Hernández-Reséndiz, G. E. Crespo-Avilan, R. T.
Consequently, the intramyocardial injection of Col@Gel has great Mukhametshina, X. Y. Kwek, H. A. Cabrera-Fuentes and D. J.
potential for myocardial repair after MI. Hausenloy, Pharmacol. Ther., 2018, 186, 73-87.
3. T. Anzai, Circ. J., 2013, 77, 580-587.
It is worth noting that some synthetic or natural derived hydrogels, 4. N. G. Frangogiannis, Nat. Rev. Cardiol., 2014, 11, 255.
5. I. M. Seropian, S. Toldo, B. W. Van Tassell and A. Abbate, J. Am.
such as poly(NIPAAm-co-HEMA-co-MAPLA) hydrogel or Coll. Cardiol., 2014, 63, 1593-1603.
hyaluronic acid-based hydrogel, without drug loading could also 6. P. M. Ridker and T. F. Lüscher, Eur. Heart J., 2014, 35, 1782-1791.
7. G. Nuki, Curr. Rheumatol. Rep., 2008, 10, 218.
improve the heart function post-MI by intramyocardial injection.34, 35, 8. M. Akodad, J. Fauconnier, P. Sicard, F. Huet, F. Blandel, A.
66
Bourret, P. de Santa Barbara, S. Aguilhon, M. LeGall, G. Hugon,
These hydrogels presented high gel moduli, and the researchers A. Lacampagne and F. Roubille, Int. J. Cardiol., 2017, 240, 347-
believed that their intramyocardial administration enhanced the 353.
9. A. Qaseem, R. P. Harris and M. A. Forciea, Ann. Intern. Med., 2017,
mechanical stiffness of infarct zone, thereby attenuating left 166, 58-68.
10. M. Mullins, A. A. Cannarozzi, T. C. Bailey and P. Ranganathan,
ventricular remodeling and improving cardiac function. Nevertheless, Clin. Toxicol., 2011, 49, 648-652.
11. Y. Finkelstein, S. E. Aks, J. R. Hutson, D. N. Juurlink, P. Nguyen,
our PLGA-PEG-PLGA hydrogel is much softer, whether such a soft
G. Dubnov-Raz, U. Pollak, G. Koren and Y. Bentur, Clin. Toxicol.,
hydrogel alone could enhance the mechanical stiffness of ventricular 2010, 48, 407-414.
12. D. B. Crittenden, R. A. Lehmann, L. Schneck, R. T. Keenan, B.
wall remains to be explored in future. Shah, J. D. Greenberg, B. N. Cronstein, S. P. Sedlis and M. H.
Pillinger, J. Rheumatol., 2012, 39, 1458-1464.
13. D. H. Solomon, C. C. Liu, I. H. Kuo, A. Zak and S. C. Kim, Ann.
Conclusions Rheum. Dis., 2016, 75, 1674-1679.
14. S. Deftereos, G. Giannopoulos, C. Angelidis, N. Alexopoulos, G.
Herein, we successfully developed an injectable PLGA-PEG-PLGA Filippatos, N. Papoutsidakis, G. Sianos, J. Goudevenos, D.
Alexopoulos, V. Pyrgakis, M. W. Cleman, A. S. Manolis, D.
hydrogel for the intramyocardial sustained delivery of Col to treat MI. Tousoulis and J. Lekakis, Circulation, 2015, 132, 1395-1403.
The prepared Col@Gel system maintained the temperature- 15. M. Akodad, B. Lattuca, N. Nagot, V. Georgescu, M. Buisson, J. P.
Cristol, F. Leclercq, J. C. Macia, R. Gervasoni, T. T. Cung, S. Cade,
responsive gelling feature, and the release of Col from the Col@Gel F. Cransac, J. Labour, A. M. Dupuy and F. Roubille, Arch.
Cardiovasc. Dis., 2017, 110, 395-402.
exhibited a sustained manner over an 8-day period. In vitro and in vivo 16. K. Fujisue, K. Sugamura, H. Kurokawa, J. Matsubara, M. Ishii, Y.

This journal is © The Royal Society of Chemistry 2019 J. Name., 2019, 00, 1-3 | 13
Journal of Materials Chemistry B Page 14 of 15
View Article Online
DOI: 10.1039/C9TB02523E
Paper Journal of Materials Chemistry B

Izumiya, K. Kaikita and S. Sugiyama, Circ. J., 2017, 81, 1174-1182. P. Liu, L. Yu, K. Y. Cai and J. D. Ding, Chin. J. Polym. Sci., 2019,
17. S. J. Buwalda, T. Vermonden and W. E. Hennink, 37, 548-559.
Biomacromolecules, 2017, 18, 316-330. 49. X. Fu, H. Zeng, J. Guo, H. Liu, Z. Shi, H. Chen, D. Li, X. Xie and
18. J. G. Sun, X. Liu, Y. Lei, M. Y. Tang, Z. X. Dai, X. W. Yang, X. C. Kuang, Chem. Pharm. Bull., 2017, 65, 229-235.
B. Yu, L. Yu, X. H. Sun and J. D. Ding, J. Mater. Chem. B, 2017, 50. D. Liu, Q. Wu, Y. Zhu, Y. Liu, X. Xie, S. Li, H. Lin, W. Chen and
5, 6400-6411. F. Zhu, Drug Deliv., 2019, 26, 522-531.
19. R. Bao, B. Y. Tan, S. Liang, N. Zhang, W. Wang and W. G. Liu, 51. Y. Zhang, J. Zhang, W. Xu, G. Xiao, J. X. Ding and X. S. Chen,
Biomaterials, 2017, 122, 63-71. Acta Biomater., 2018, 77, 63-73.

Journal of Materials Chemistry B Accepted Manuscript


20. D. N. Wang, X. W. Yang, Q. Liu, L. Yu and J. D. Ding, J. Mater. 52. M. Miyazaki, T. Maeda, K. Hirashima, N. Kurokawa, K.
Chem. B, 2018, 6, 6067-6079. Nagahama and A. Hotta, Polymer, 2017, 115, 246-254.
21. H. J. Song, P. X. Yang, P. S. Huang, C. N. Zhang, D. L. Kong and 53. M. Kitagawa, T. Maeda and A. Hotta, Polym. Degrad. Stabil., 2018,
W. W. Wang, Theranostics, 2019, 9, 2299-2314. 147, 222-228.
22. W. Wang, B. Y. Tan, J. R. Chen, R. Bao, X. R. Zhang, S. Liang, Y. 54. J. Koglin, S. Pehlivanli, M. Schwaiblmair, M. Vogeser and P.
Y. Shang, W. Liang, Y. L. Cui, G. W. Fan, H. Z. Jia and W. G. Liu, Cremer, J. Am. Coll. Cardiol., 2001, 38, 1934-1941.
Biomaterials, 2018, 160, 69-81. 55. R. W. Troughton, C. M. Frampton, T. G. Yandle, E. A. Espine, M.
23. Y. Han, W. Yang, W. Cui, K. Yang, X. Wang, Y. Chen, L. Deng, G. Nicholls and A. M. Richards, Lancet, 2000, 355, 1126-1130.
Y. Zhao and W. Jin, J. Mater. Chem. B, 2019, 7, 1563-1580. 56. J. Ozer, M. Ratner, M. Shaw, W. Bailey and S. Schomaker,
24. H. Chang, C. Li, R. Huang, R. Su, W. Qi and Z. He, J. Mater. Chem. Toxicology, 2008, 245, 194-205.
Published on 31 December 2019. Downloaded on 1/3/2020 1:26:29 AM.

B, 2019, 7, 2899-2910. 57. J. M. Lambert, E. F. Lopez and M. L. Lindsey, Int. J. Cardiol., 2008,
25. Q. Tang, D. Zhao, H. Yang, L. Wang and X. Zhang, J. Mater. Chem. 130, 147-158.
B, 2019, 7, 30-42. 58. T. Weinberger and C. Schulz, Front. Physiol., 2015, 6, 107.
26. K. Zhang, Z. Shi, J. Zhou, Q. Xing, S. Ma, Q. Li, Y. Zhang, M. 59. M. Tian, Y. C. Yuan, J. Y. Li, M. R. Gionfriddo and R. C. Huang,
Yao, X. Wang and Q. Li, J. Mater. Chem. B, 2018, 6, 2982-2992. Chronic. Dis. Transl. Med., 2015, 1, 18-26.
27. S. Q. Cui, L. Yu and J. D. Ding, Acta Polym. Sin., 2018, 8, 863- 60. D.-Y. Nah and M.-Y. Rhee, Korean Circ. J., 2009, 39, 393-398.
881. 61. N. G. Frangogiannis, Circ. Res., 2012, 110, 159-173.
28. H. J. Lee and B. Jeong, Small, 2019, DOI: 10.1002/smll.201903045, 62. C. X. Shen, H. Z. Chen and J. B. Ge, Chin. Med. J., 2004, 117, 133-
e1903045. 139.
29. S. Q. Cui, L. Yu and J. D. Ding, Macromolecules, 2019, 52, 3697- 63. P. M. Ridker, B. M. Everett, T. Thuren, J. G. MacFadyen, W. H.
3715. Chang, C. Ballantyne, F. Fonseca, J. Nicolau, W. Koenig and S. D.
30. Y. Qu, B. Y. Wang, B. Y. Chu, C. L. Liu, X. Rong, H. Chen, J. R. Anker, New Engl. J. Med., 2017, 377, 1119-1131.
Peng and Z. Y. Qian, ACS Appl. Mater. Interfaces, 2018, 10, 4462- 64. J. W. Holmes, T. K. Borg and J. W. J. A. R. B. E. Covell, Annu.
4470. Rev. Biomed. Eng., 2005, 7, 223-253.
31. M. Patel, H. J. Lee, S. Park, Y. Kim and B. Jeong, Biomaterials, 65. A. E. Arai, JACC. Cardiovasc. Imag., 2015, 8, 680-683.
2018, 159, 91-107. 66. S. M. Dorsey, J. R. McGarvey, H. Wang, A. Nikou, L. Arama, K.
32. S. S. Lee, G. E. Choi, H. J. Lee, Y. Kim, J. H. Choy and B. Jeong, J. Koomalsingh, N. Kondo, J. H. Gorman III, J. J. Pilla and R. C.
ACS Appl. Mater. Interfaces, 2017, 9, 42668-42675. Gorman, Biomaterials, 2015, 69, 65-75.
33. S. J. Yu, D. L. Zhang, C. L. He, W. J. Sun, R. J. Cao, S. S. Cui, M.
X. Deng, Z. Gu and X. S. Chen, Biomacromolecules, 2017, 18,
4341-4348.
34. T. Yoshizumi, Y. Zhu, H. Jiang, A. D'Amore, H. Sakaguchi, J.
Tchao, K. Tobita and W. R. Wagner, Biomaterials, 2016, 83, 182-
193.
35. Y. Matsumura, Y. Zhu, H. Jiang, A. D'Amore, S. K. Luketich, V.
Charwat, T. Yoshizumi, H. Sato, B. Yang, T. Uchibori, K. E. Healy
and W. R. Wagner, Biomaterials, 2019, 217, 119289.
36. Z. Q. Zhang and S. C. Song, Biomaterials, 2017, 132, 16-27.
37. B. B. Seo, H. I. Chang, H. Choi, J. T. Koh, K. D. Yun, J. Y. Lee
and S. C. Song, J. Biomed. Mater. Res. B, 2018, 106, 751-759.
38. L. Yu, Z. Zhang, H. Zhang and J. D. Ding, Biomacromolecules,
2010, 11, 2169-2178.
39. Y. P. Liu, X. B. Chen, S. Y. Li, Q. Guo, J. Xie, L. Yu, X. Y. Xu, C.
M. Ding, J. S. Li and J. D. Ding, ACS Appl. Mater. Interfaces, 2017,
9, 23428-23440.
40. D. H. Shin and G. S. Kwon, J. Control. Release, 2017, 268, 176-
183.
41. W. J. Shen, X. B. Chen, J. B. Luan, D. N. Wang, L. Yu and J. D.
Ding, ACS Appl. Mater. Interfaces, 2017, 9, 40031-40046.
42. X. B. Chen, M. L. Wang, X. W. Yang, Y. B. Wang, L. Yu, J. Sun
and J. D. Ding, Theranostics, 2019, 9, 6080-6098.
43. L. Yu, K. Li, X. J. Liu, C. Chen, Y. C. Bao, T. Y. Ci, Q. H. Chen
and J. D. Ding, J. Pharm. Sci., 2013, 102, 4140-4149.
44. Y. P. Chen, Y. Z. Li, W. J. Shen, K. Li, L. Yu, Q. H. Chen and J.
D. Ding, Sci. Rep., 2016, 6, 31593.
45. Y. P. Zhuang, X. W. Yang, Y. M. Li, Y. P. Chen, X. C. Peng, L.
Yu and J. D. Ding, ACS Appl. Mater. Interfaces, 2019, 11, 29604-
29618.
46. X. Z. Li, J. X. Ding, Z. Z. Zhang, M. D. Yang, J. K. Yu, J. C. Wang,
F. Chang and X. S. Chen, ACS Appl. Mater. Interfaces, 2016, 8,
5148-5159.
47. J. B. Luan, Z. Zhang, W. J. Shen, Y. P. Chen, X. W. Yang, X. B.
Chen, L. Yu, J. Sun and J. D. Ding, ACS Appl. Mater. Interfaces,
2018, 10, 30235-30246.
48. W. K. Xu, J. Y. Tang, Z. Yuan, C. Y. Cai, X. B. Chen, S. Q. Cui,

14 | J. Name., 2019, 00, 1-3 This journal is © The Royal Society of Chemistry 2019
Page 15 of 15 Journal of Materials Chemistry B

For Table of Contents use only View Article Online


DOI: 10.1039/C9TB02523E

Graphical and textual abstract for TOC

Journal of Materials Chemistry B Accepted Manuscript


An injectable thermosensitive hydrogel loaded with an

ancient natural drug colchicine for myocardial repair

after infarction

Yu Chen‡, Jiayue Shi‡, Yaping Zhang, Jiajun Miao, Zhe Zhao, Xian Jin, Liang Liu, Lin
Published on 31 December 2019. Downloaded on 1/3/2020 1:26:29 AM.

Yu*, Chengxing Shen*, Jiandong Ding

The intramyocardial injection of colchicine-loaded hydrogel system effectively

promoted myocardial repair after infarction while minimizing the systemic toxicity of

colchicine.

You might also like