Satellite Nanoparticles for Anti

Publication Date (Web): February 3, 2017. Copyright © 2017 American Chemical Society. *E-mail: [email protected]., *E-mail: [email protected]...
0 downloads 0 Views 3MB Size
Subscriber access provided by Caltech Library

Article

Self-assembled raspberry-like core/satellite nanoparticles for anti-inflammatory protein delivery Tingting Wang, Yaqin Tang, Xiao He, Ju Yan, Xuli Feng, and Chenhui Wang ACS Appl. Mater. Interfaces, Just Accepted Manuscript • DOI: 10.1021/acsami.6b16277 • Publication Date (Web): 03 Feb 2017 Downloaded from http://pubs.acs.org on February 7, 2017

Just Accepted “Just Accepted” manuscripts have been peer-reviewed and accepted for publication. They are posted online prior to technical editing, formatting for publication and author proofing. The American Chemical Society provides “Just Accepted” as a free service to the research community to expedite the dissemination of scientific material as soon as possible after acceptance. “Just Accepted” manuscripts appear in full in PDF format accompanied by an HTML abstract. “Just Accepted” manuscripts have been fully peer reviewed, but should not be considered the official version of record. They are accessible to all readers and citable by the Digital Object Identifier (DOI®). “Just Accepted” is an optional service offered to authors. Therefore, the “Just Accepted” Web site may not include all articles that will be published in the journal. After a manuscript is technically edited and formatted, it will be removed from the “Just Accepted” Web site and published as an ASAP article. Note that technical editing may introduce minor changes to the manuscript text and/or graphics which could affect content, and all legal disclaimers and ethical guidelines that apply to the journal pertain. ACS cannot be held responsible for errors or consequences arising from the use of information contained in these “Just Accepted” manuscripts.

ACS Applied Materials & Interfaces is published by the American Chemical Society. 1155 Sixteenth Street N.W., Washington, DC 20036 Published by American Chemical Society. Copyright © American Chemical Society. However, no copyright claim is made to original U.S. Government works, or works produced by employees of any Commonwealth realm Crown government in the course of their duties.

Page 1 of 20

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Applied Materials & Interfaces

Self-assembled raspberry-like core/satellite nanoparticles for anti-inflammatory protein delivery Tingting Wang, Yaqin Tang, Xiao He, Ju Yan, Chenhui Wang∗ and Xuli Feng∗ Innovative Drug Research Centre and School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China. ABSTRACT: Functional proteins are very promising for protein therapeutics, however, effective delivery of therapeutic proteins remains challenging. Herein, we developed a novel core/satellite nanoparticles by tethering therapeutic proteins to the core/shell polymeric particle surface through cucurbit[8]uril (CB[8]) mediated host-guest interactions. The effectiveness of the core/satellite nanoparticles as protein carrier was demonstrated through the intra-articular delivery of interleukin-1 receptor antagonist (IL-1Ra). We showed that IL-1Ra can effectively self assemble onto the surface of the polymeric nanoparticles and maintained good protein bioactivity by inhibiting IL-1 mediated signalling. More importantly, in vivo results revealed that IL-1Ra bounded core/satellite nanoparticles could significantly increase the retention time of IL1Ra in the rat stifle joint comparing to soluble IL-1Ra, which could greatly improve the efficacy of IL-1Ra. These results indicate that the facile host guest self assembly can be exploited as an effective approach for realizing the therapeutic potential of proteins. KEYWORDS: Protein delivery, Self assembly, host guest interaction, core/satellite structure, IL-1Ra

ACS Paragon Plus Environment

1

ACS Applied Materials & Interfaces

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 2 of 20

INTRODUCTION Therapeutic proteins have gained increasing attention due to their great potential in combating various diseases.1-3 Despite many potentially therapeutic proteins including cytokines, antibodies, transcription factors and so on, have been discovered for decades,4 their further clinical application have been severely restricted and hindered due to short biological half-life and insufficient accumulation in the necessary site of action, such as IL-1Ra.5 IL-1Ra is an endogenous anti-inflammatory antagonist that could effectively block the signaling of proinflammatory cytokines such as interleukin-1 (IL-1) which are widely involved in the pathogenesis of some chronic diseases such as arthritis and type 2 diabetes mellitus.6-10 Currently, IL-1Ra has been explored as a therapy for the treatment of rheumatoid arthritis (RA).Through intra-articular injections of IL-1Ra for the treatment of osteoarthritis (OA) was found to be safe, however, the anti-inflammatory clinical efficacy is unsatisfactory due to its rapid clearance with retention time as short as 1-2 h.11 Thus several strategies have been explored to prolong the biological half-life and enhance the therapeutic effect of IL-1Ra.5,12 PLGA microspheres have been applied for encapsulating IL-1Ra in order to increase its biological half-life, however, the bioactivity of IL-1Ra has been reduced due to the hydrophobic nature of PLGA polymer.13,14 Polyethylene glycol (PEG) is a material approved by FDA, which is widely used in therapeutic protein modification. Indeed, PEGylated IL-1Ra has been proven to have longer circulation time, but its bioactivity has been greatly destroyed due to pegylation.15 Recombinant IL-1Ra variant modified with amyloidogenic motif and IL-1Ra tethered nanopartilces based on block copolymer have also been made to improve the half-life and bioactivity of IL-1Ra,16,17 however, better and simple delivery system for IL-1Ra is still needed due to the complex preparation process of these methods.

ACS Paragon Plus Environment

2

Page 3 of 20

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Applied Materials & Interfaces

Core/satellite nanostructures have drawn increasing interest in materials science due to their potential applications in many fields, such as optical sensing, surface-enhanced Raman spectroscopy (SERS) based sensor systems, drug delivery and so on.18-21 The typical compositions of core/satellite nanoparticles include a larger core particle (polystyrene, viruses, SiO2, metals, etc.) and smaller corona particles such as gold, silver, or semiconductor nanoparticles.22-24 Electrostatic interactions, α-ω functionalized alkane linkers, DNA and protein have been employed to adhere the smaller corona particles to the core.25-31 However, these methods suffer from poor stability and cumbersome preparation. In addition, DNA or protein assisted assembly methods are also restricted in terms of scalability and high cost. Host-guest interactions based on macrocyclic hosts, including crown ethers, cycylodextrins, cucurbiturils and pillararenes, have been used in the preparation of various supramolecular structures.32-35 Among them, Cucurbit[8]uril (CB[8]) has recently been widely employed to prepare stimuliresponsive micelles,hydrogels and polymeric colloids due to its capability of encapsulating two guest molecules inside its cavity simultaneously.36-39 This kind of binding is simple and stable, thus it would be meaningful to prepare core/satellite nanoparticle for protein delivery based on this efficient host-guest complexation method.

Herein, we present a facile route to prepare IL-1Ra core/satellite nanoparticles based on the host-guest interaction mediated by CB[8]. As shown in Figure 1, methyl viologen (MV) functionalized polymeric nanoparticles (MV-NP) were employed as the core, and the trans isomer of azobenzene (trans-Azo) modified IL-1Ra was chosen as the corona nanoparticles. MV and trans-Azo are good guest molecules for CB[8], thus upon the addition of CB[8], trans-Azo modified IL-1Ra could automatically adhere to the surface of MV-NP core through host-guest recognition. The formed IL-1Ra tethered core/satellite nanoparticles can competitively bind to

ACS Paragon Plus Environment

3

ACS Applied Materials & Interfaces

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 4 of 20

the IL-1R1 receptor, thus blocking the IL-1 signaling pathway. This simple and facile self assembly approach provides a platform to prepare protein delivery system under mild conditions. Therefore, the present system could be considered as a promising strategy for IL-1Ra based protein therapy.

Figure 1. Schematic illustration of the formation of IL-1Ra tethered core/satellite nanoparticle and its function as competitive blocker of IL-1 signaling pathway.

EXPERIMENTAL SECTION Materials and Reagents. IL-1Ra and IL-1β were purchased from Sino Biological Inc. (Beijing, China). Bovine serum albumin (BSA) was purchased from Sangon Biotech (Shanghai) Co., Ltd. Fluorescein-5-maleimide and Cyanine7 maleimide was obtained from Tianjin Heowns

ACS Paragon Plus Environment

4

Page 5 of 20

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Applied Materials & Interfaces

Biochemical Technology Co., Ltd. and Lumiprobe Corporation (Florida, USA) respectively. MTT(3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide), Hoechst33342 and DAPI

(4’,6-diamidino-2-phenylindole)

were

purchased

from

Beyotime

Institute

of

Biotechnology (Haimen, China). Ready Set Go® ELISA kits for the detection of TNF-α was gained from eBioscience (San Diego, CA, USA). RAW 264.7 macrophage cells and SW-982 cells (human synovial cell lines) were obtained from Cell Bank of the Chinese Academy of Sciences (Shanghai, China). All other chemicals used were of analytical reagent grade. Preparation of Azo-protein corona and tethering protein to the surface of MV-NP core. BSA or IL-1Ra protein was mixed with compound 4 (shown in Figure S2 in supporting information) in aqueous solution (pH=8). The mixture solution was placed at 4 oC and allowed to react overnight to form trans-Azo modified BSA (Azo-BSA) or (Azo- IL-1Ra) respectively. The trans-Azo modified protein was then mixed with MV-NP in the presence of CB[8] to form the corresponding BSA or IL-1Ra tethered core/satellite nanoparticles. Transmission electron microscopy (TEM) characterization. For TEM observation, a portion of the resultant core/satellite nanoparticles was diluted with water and then a drop of the diluted particle dispersion was deposited onto a piece of copper EM grid. After drying at room temperature, TEM measurements were conducted by using a Hitachi H-7500 electron microscope at an acceleration voltage of 100 kV. Cytotoxicity assay. A MTT reduction assay was used to perform the measurement of the cytotoxicity of MV-NP nanoparticles. The RAW 264.7 macrophage cells (1x105cells/well, 96well tissue culture plate) were incubated with the MV-NP nanoparticles at various concentrations (0.125–0.5mg/ml) in serum-free media for 24 h. After pipetting out the media, the cells were then treated with 100 µL of MTT substrate (5 mg/mL in PBS) and incubated at 37℃for another 4

ACS Paragon Plus Environment

5

ACS Applied Materials & Interfaces

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 6 of 20

h. After removing MTT, 100µL DMSO was added to each well to dissolve the purple formazan and then detecting the absorbance at 490 nm using plate reader (Berthold TriStar LB 941, Germany). Percentage cell viability was calculated by comparing the absorbance of MV-NP nanoparticles treated cells to that of the control cells. Synoviocyte binding experiments. SW-982 cells were seeded in a 24-well plate. After overnight incubation, 0.5mLofeither fluorescein-IL-1Ratethered or fluorescein-BSA tethered core/satellite particles was added to each well and treated with cells for24h. After washing the cells with 1xPBSthree times to remove the unbound particles, 500µL 4%paraformaldehyde was added to fix the cells for 10 min. Following rinsing three times, the cell nuclei were stained with 5 µg/ml Hoechst 33342 for20 min at room temperature. After washing all wells again, the cells were directlyanalysed for particle binding on fluorescence microscopy (Olympus IX 51, Japan). Inhibition of IL-1β β induced signalling. SW-982 cells (1x105 cells/well) were plated in a 24-well plate. After 48h incubation, cells were thoroughly rinsed with PBS and treated with 0.5µg/ml of soluble IL-1Ra, IL-1Ra tethered or BSA tethered core/satellite particles respectively. 1h later, 5 ng/ml of human IL-1β was added to each well. After incubation for another 24h, cell culture supernatants were analysed for TNF-α levels using Ready Set Go® ELISA kits according to manufacturer's instructions. IVIS imaging to evaluate protein retention. Male SD rats (8-10 week old) were anesthetized with 7% chloralhydrate deeply. Then 5 µgCy7 labelledIL-1Ra or IL-1Ratethered core/satellite particles was injected into the right stifle joint. The left stifle joint received saline at the same time as control. An imaging system (700 Series, Caliper Xenogen IVIS Lumina, Caliper Life Sciences, USA) was applied to take images. After setting the excitation or emission

ACS Paragon Plus Environment

6

Page 7 of 20

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Applied Materials & Interfaces

wavelengthat 745 nm or 780 nm respectively, a fixed region centered on the stifle was scanned and captured. Immunostaining. As described above, rats injected with 5µg Fluorescein-5-maleimide labelled IL-1Ra or IL-1Ratethered core/satellite particles for 3 days were sacrificed. The synovial membrane was harvested, embedded and kept in -80℃ refrigerator. 10µm thick sections were sectioned using a cryosectioner. After staining with DAPI and rinsing with PBS, the sections were imaged using fluorescence microscopy.

RESULTS AND DISCUSSION Preparation

and

characterization

of

protein

tethered

core/satellite

nanoparticles. To prepare protein tethered core/satellite nanoparticles, the choice of core is very important. Various organic and inorganic particles have been selected as the core, among them, core-shell polymeric particles as the core has been mostly common reported due to the ease of preparation and modification.37 Herein, in this research, soap-free emulsion polymerization was employed to synthesize MV functionalized core-shell polymeric nanoparticles.40 The synthetic route is illustrated in the supporting information (Figure S1). The biggest advantage of this method is that no surfactant is involved, and the formation of nanoparticles is simply based on the coagglutination of the hydrophilic sphere to the hydrophobic core. Hence, we can easily get the MV modified core/shell nanoparticles simply by washing with water after polymerization. TEM was performed to examine the morphology and size of MV-NP. Figure 2a demonstrates that MV-NP has well dispersed core/shell morphology with an average size of 200 nm. In order to make core/satellite nanoparticles for protein delivery, proteins were modified with trans-Azo to

ACS Paragon Plus Environment

7

ACS Applied Materials & Interfaces

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 8 of 20

act as the satellite particles (the synthetic route is described in Figure S2 in supporting information). The core/satellite nanoparticles were then prepared simply through hostguest recognition by adding an aqueous dispersion of MV-NP core into an aqueous dispersion of trans-Azo tethered protein corona in the presence of CB[8] at room temperature. From Figure 2b and c, we can clearly see that both BSA and IL-1Ra can be adhered to the surface of MV-NP core to form well-defined core/satellite nanoparticles. And the tethering density of IL-1Ra is higher than that of BSA, which is good for enhancing the retention time of IL-1Ra. To be noted, most reported core/satellite nanoparticles chose smaller inorganic nanoprticles as the corona, using protein as the corona is rarely studied. Therefore, using protein as the corona reported here provides new potential applications of core/satellite nanoparticles in biomedical research.

Figure 2. TEM images of (a) MV-NP, (b) BSA and (c) IL-1Ra tethered core/satellite particles. Insert is the enlargement image of the particles.

Target specificity of IL-1Ra tethered core/satellite nanoparticles. The above results demonstrated that protein can be easily tethered to the surface of MV-NP and form welldefined core/satellite nanocomplexes. We next examined whether IL-1Ra tethered

ACS Paragon Plus Environment

8

Page 9 of 20

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Applied Materials & Interfaces

core/satellite nanoparticles can specifically bind to SW-982 synoviocytes. Synoviocytes, in which nucleus was stained with Hoechst 33342, were incubated with fluorescein labeled IL-1Ra or BSA tethered nanoparticles. The specific binding of IL-1Ra tethered nanoparticles were investigated by fluorescent microscopy. As shown in Figure 3, when compared to synoviocytes incubated with BSA tethered nanoparticles, synoviocytes treated with IL-1Ra tethered nanoparticles exhibited great stronger fluorescence intensity, indicating the specific binding of IL-1Ra tethered core/satellite nanoparticles to synoviocyte cells.

Figure 3. Fluorescence images showing the binding of (a) IL-1Ra and (b) BSA tethered core/satellite particles to the synoviocytes. Scale bar is 40 µm.

Particle cytotoxicity. Since the toxicity of the delivery material is one of the major hurdles that hinder the use of protein as therapeutics, MTT assay was employed to test the cytotoxicity of different concentrations of MV-NP in macrophage cells (RAW 264.7). As shown in Figure 4, MV-NP showed minimum cytotoxicity even at the highest concentration of 500 µg mL−1, suggesting that MV-NP not only can be used as the core

ACS Paragon Plus Environment

9

ACS Applied Materials & Interfaces

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 10 of 20

for self assembly core/satellite nanostructure, but also can serve as a safe and effective vector for protein delivery.

Figure 4. Cell viability results after incubation of macrophage cells with various concentrations of MV-NP.

Effect of IL-1Ra tethered nanoparticles on IL-1β β -induced signaling. The bioactivity of IL-1Ra tethered nanoparticles was further evaluated by assessing their ability to block IL1β induced production of pro-inflammatory cytokines (such as TNF-α). SW982 synovial cells were pre-treated for 1 h with different formulations of IL-1Ra at a concentration equivalent to 0.5µg/ml of soluble IL-1Ra, including recombinant IL-1Ra itself, IL-1Ra tethered core/satellite nanoparticles and BSA tethered particles as a negative control. And then, all cell groups were treated with 5 ng/mL of IL-1β for 24 h to stimulate TNF-α production, which was quantified using an enzyme linked immunosorbent assay (ELISA). Figure 5 demonstrates that IL-1Ra tethered particles can efficiently inhibit the production of IL-1β stimulated pro-inflammatory cytokine in synoviocytes. The expression level of TNF-α in synoviocytes pre-treated with IL-1Ra tethered particles or soluble IL-1Ra is comparable to that of cells without IL-1β stimulation. Meanwhile, BSA particles

ACS Paragon Plus Environment

10

Page 11 of 20

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Applied Materials & Interfaces

pretreated cells, as a negative control, did not show any decrease of amounts of TNF-α expression. The results confirm that IL-1Ra as the corona of core/satellite nanostructure effectively retain the activity of IL-Ra itself, and this nanostructure can serve as a potential material for protein delivery.

Figure 5. IL-1Ra tethered nanoparticles inhibit IL-1β induced TNF-α production. n=3, *p < 0.01.

In vivo protein retention time of IL-1Ra tethered core/satellite nanoparticles. To achieve the therapeutic applications of IL-1Ra tethered core/satellite nanoparticles, the delivery systems not only need to keep good bioactivity of IL-1Ra, but also need to prolong the retention time of IL-1Ra in the necessary site. Therefore, we investigated the retention of IL-Ra tethered particles in vivo. Cy7 is one of the near infrared dyes which are widely used for in vivo imaging due to their strong tissue penetration and small damage. Hence, Cy7 labelling was used here for monitoring the retention of IL1Ratethered particles and soluble protein. Rats were intra-articular injected with either 5 µg of soluble protein or equivalent amounts of IL-1Ra tethered particles in the stifle joint and scanned in an in vivo imaging system. The entire imaging process continues for 14

ACS Paragon Plus Environment

11

ACS Applied Materials & Interfaces

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 12 of 20

days.From Figure 6a we can clearly see that strong fluorescent signals are still exhibited in rats received IL-1Ra tethered core/satellite nanoparticles even up to 14 days, while rapid loss of fluorescent signal was observed in those receiving soluble protein. The results showed that IL-1Ra tethered naoparticles could significantly prolong the half-life time of IL-Ra in the joint compared to the soluble form. Moreover, immunostainning was employed to further confirm the retention of IL-Ra tethered core/satellite particles in the joint tissues. Fluorescein labeledIL-1Ra or IL-1Ratethered core/satellite particles was injected into the right stifle joint of rats while the left stifle joints received saline as control. Frozen sectionswere counterstained with DAPI and imaged using microscopy. As shown in Figure 6b that, large numbers of fluorescein labeled IL-1Ra nanoparticles were retained in the joint space, while fluorescein tagged protein itself was cleared from the joint rapidly. All these results suggest that using IL-1Ra as the corona of core/satellite nanoparticles could be a potential strategy for enhancing the therapeutic efficacy of IL1Ra.

ACS Paragon Plus Environment

12

Page 13 of 20

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Applied Materials & Interfaces

Figure 6. (a) Representative IVIS images of the retention of Cy7 labeled IL-1Ra in knee joints. (b) Immunostaining to evaluate fluorescein labeled IL-1Ra retention in the intra-articular joint space. Cryosectioned samples was counterstained with DAPI to localize the synoviocytes. Scale bar is 40 µm.

CONCLUSIONS In summary, we have developed a simple and facile self-assembly method for preparing core/satellite nanostructures with polymeric nanoparticles as the core and protein as the corona. CB[8] was employed as the linking agent for tethering IL-1Ra protein to the surface of MV-NP nanoparticle via host-guest interaction in water. The formed well defined IL-1Ra tethered core/satellite nanoparticles kept the target specificity of IL-1Ra by specifically binding to the IL-1 receptors in cell surface. Moreover, the bioactivity of IL-1Ra itself has also been well retained by inhibiting the IL-1β stimulated pro-inflammatory production when treated synovial cells with IL-1Ra tethered core/satellite nanoparticles. More importantly, in vivo imaging and immunostainning further demonstrated that IL-1Ratethered nanoparticles could significantly enhance IL1Ra retention the joints of rats. Future studies will focus on evaluating the therapeutic

ACS Paragon Plus Environment

13

ACS Applied Materials & Interfaces

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 14 of 20

efficacy of these IL-1Ra tethered particles in mitigating cartilage destruction. We believe that the facile host guest self assembly can be exploited as an effective approach for realizing the therapeutic potential of proteins, and using protein as the corona could provide new potential applications of core/satellite nanoparticles in biomedical research. ASSOCIATED CONTENT Supporting information The Supporting Information is available free of charge on the ACS Publications website at DOI: Synthetic routes about the compounds used for preparation of core/satellite nanoparticles. AUTHOR INFORMATION Corresponding authors ∗Email: [email protected]; [email protected] NOTES The authors declare no competing financial interests. ACKNOWLEDGEMENT. This work was financially supported by the Startup Funding for “Hundred Young-Talent Scheme” Professorship provided by the Chongqing University in China (0236011104419) and National Natural Science Foundation of China (21605011). REFERENCES (1) Gu, Z.; Biswas, A.; Zhao, M.; Tang, Y. Tailoring Nanocarriers for Intracellular Protein Delivery. Chem Soc Rev. 2011, 40, 3638-3655. (2) Tang, R.; Jiang, Z.; Ray, M.; Hou, S.; Rotello, V. M. Cytosolic Delivery of Large Proteins Using Nanoparticle-Stabilized Nanocapsules. Nanoscale. 2016, 8, 18038-18041. (3) Wang, M.; Alberti, K.; Sun, S.; Arellano, C. L.; Xu, Q. Combinatorially Designed Lipid-like Nanoparticles for Intracellular Delivery of Cytotoxic Protein for Cancer Therapy. Angew Chem Int Ed. 2014, 53, 2893-2898.

ACS Paragon Plus Environment

14

Page 15 of 20

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Applied Materials & Interfaces

(4) Zuris, J. A.; Thompson, D. B.; Shu, Y.; Guilinger, J. P.; Bessen, J. L.; Hu, J. H.; Maeder, M. L.; Joung, J. K.; Chen, Z. Y.; Liu, D. R. Cationic Lipid-mediated Delivery of Proteins Enables Efficient Protein-based Genome Editing in Vitro and in Vivo. Nat Biotechnol. 2015, 33, 73-80. (5) Akash, M. S.; Rehman, K.; Chen, S. IL-1Ra and Its Delivery Strategies: Inserting the Association in Perspective. Pharm Res. 2013, 30, 2951-2966. (6) Kraus, V. B.; Birmingham, J.; Stabler, T. V.; Feng, S.; Taylor, D. C.; Moorman, C. T. 3rd.; Garrett, W. E.; Toth, A. P. Effects of Intraarticular IL1-Ra for Acute Anterior Cruciate Ligament Knee Injury: A Randomized Controlled Pilot Trial (NCT00332254). Osteoarthritis Cartilage 2012, 20, 271-278. (7) Hannum, C. H.; Wilcox, C. J.; Arend, W. P.; Joslin, F. G.; Dripps, D. J.; Heimdal, P. L.; Armes, L. G.; Sommer, A.; Eisenberg, S. P.; Thompson, R. C. Interleukin-1 Receptor Antagonist Activity of a Human Interleukin-1 Inhibitor. Nature. 1990, 343, 336-340. (8) Gabay, C.; Lamacchia, C.; Palmer, G. IL-1 Pathways in Inflammation and Human Diseases. Nat Rev Rheumatol. 2010, 6, 232-241. (9) Freeman, B. D.; Buchman, T. G. Interleukin-1 Receptor Antagonist as Therapy for Inflammatory Disorders. Expert Opin Biol Ther. 2001, 1, 301-308. (10) Akash, M. S. H.; Shen, Q.; Rehman, K.; Chen, S. Interleukin-1 Receptor Antagonist: a New Therapy for Type 2 Diabetes Mellitus. J Pharm Sci. 2012, 101, 1647-1658. (11) Cawthorne, C.; Prenant, C.; Smigova, A.; Julyan, P.; Maroy, R.; Herholz, K.; Rothwell, N.; Boutin, H. Biodistribution, Pharmacokinetics and Metabolism of Interleukin-1 Receptor Antagonist (IL-1RA) Using [(1)(8)F]-IL1RA and PET Imaging in Rats. Br J Pharmacol. 2011, 162, 659-672.

ACS Paragon Plus Environment

15

ACS Applied Materials & Interfaces

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 16 of 20

(12) Singh, A.; Agarwal, R.; Diaz-Ruiz, C. A.; Willett, N. J.; Wang, P.; Lee, L. A.; Wang, Q.; Guldberg, R. E.; Garcia, A. J. Nanoengineered Particles for Enhanced Intra-articular Retention and Delivery of Proteins. Adv. Healthc. Mater. 2014, 3, 1562-1567. (13) Lavi, G.; Voronov, E.; Dinarello, C. A.; Apte, R. N.; Cohen, S. Sustained Delivery of IL-1Ra From Biodegradable Microspheres Reduces the Number of Murine B16 Melanoma Lung Metastases. J Control Release. 2007, 123, 123-130. (14) Gorth, D. J.; Mauck, R. L.; Chiaro, J. A.; Mohanraj, B.; Hebela, N. M.; Dodge, G. R.; Elliott, D. M.; Smith, L. J. IL-1ra Delivered From Poly(lactic-co-glycolic acid) Microspheres Attenuates IL-1β-mediated Degradation of Nucleus Pulposus in Vitro. Arthritis Res Ther. 2012, 14, R179. (15) Yu, P.; Zheng, C.; Chen, J.; Zhang, G.; Liu, Y.; Suo, X.; Zhang, G.; Su, Z. Investigation on PEGylation Strategy of Recombinant Human Interleukin-1 Receptor Antagonist. BioorgMed Chem 2007, 15, 5396-5405. (16) Pasi, S.; Kant, R.; Gupta, S.; Surolia, A. Novel Multimeric IL-1 Receptor Antagonist for the Treatment of Rheumatoid Arthritis. Biomaterials. 2015, 42, 121-133. (17) Whitmire, R. E.; Wilson, D. S.; Singh, A.; Levenston, M. E.; Murthy, N.; Garcia, A. J. Selfassembling Nanoparticles for Intra-articular Delivery of Anti-inflammatory Proteins. Biomaterials 2012, 33, 7665-7675. (18) Liu, N.; Hentschel, M.; Weiss, T.; Alivisatos, A. P.; Giessen, H. Three-Dimensional Plasmon Rulers. Science 2011, 332, 1407-1410. (19) Chen, P. Y.; Soric, J.; Alù, A. Invisibility and Cloaking Based on Scattering Cancellation. Adv. Mater. 2012, 24, OP281-OP304.

ACS Paragon Plus Environment

16

Page 17 of 20

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Applied Materials & Interfaces

(20) Prasad, J.; Zins, I.; Branscheid, R.; Becker, J.; Koch, A. H. R.; Fytas,G.; Kolb, U.; Sönnichsen, C. Plasmonic Core-Satellite Assemblies as Highly Sensitive Refractive Index Sensors. J. Phys. Chem. C 2015, 119, 5577-5582. (21) Xiao, Q.; Zheng, X.; Bu, W.; Ge, W.; Zhang, S.; Chen, F.; Xing, H.; Ren, Q.; Fan, W.; Zhao, K.; Hua, Y.; Shi, J. A Core/satellite Multifunctional Nanotheranostic for in Vivo Imaging and Tumor Eradication by Radiation/photothermal Synergistic Therapy. J Am Chem Soc. 2013, 135, 13041-13048. (22) Qian, Z.; Hastings, S. P.; Li, C.; Edward, B.; McGinn, C. K.; Engheta, N.; Fakhraai, Z.; Park, S.-J. Raspberry-like Metamolecules Exhibiting Strong Magnetic Resonances. ACS Nano 2015, 9, 1263-1270. (23) Yoon, J. H.; Lim, J.; Yoon, S. Controlled Assembly and Plasmonic Properties of Asymmetric Core−Satellite Nanoassemblies. ACS Nano 2012, 6, 7199-7208. (24) Lebedev, N.; Griva, I.; Dressick, W. J.; Phelps, J.; Johnson, J. E.; Meshcheriakova, Y.; Lomonossoff, G. P.; Soto, C. M. A Virus-Based Nanoplasmonic Structure as a SurfaceEnhanced Raman Biosensor. Biosens. Bioelectron. 2016, 77, 306-314. (25) Schütz, M.; Schlücker, S. Molecularly linked 3D plasmonic nanoparticle core/satellite assemblies: SERS nanotags with single-particle Raman sensitivity. Phys Chem Chem Phys. 2015, 17, 24356-24360. (26) Choi, I.; Song, H. D.; Lee, S.; Yang, Y. I.; Kang, T.; Yi, J. Core-Satellites Assembly of Silver Nanoparticles on a Single Gold Nanoparticle via Metal Ion-Mediated Complex. J. Am. Chem. Soc. 2012, 134, 12083-12090. (27) Wang, X. D.; Rabe, K. S.; Ahmed, I.; Niemeyer, C. M. Multifunctional Silica Nanoparticles for Covalent Immobilization of Highly Sensitive Proteins. Adv Mater. 2015, 27, 7945-7950.

ACS Paragon Plus Environment

17

ACS Applied Materials & Interfaces

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 18 of 20

(28) Zheng, Y.; Thai, T.; Reineck, P.; Qiu, L.; Guo, Y.; Bach, U. DNA Directed Self-Assembly of Core-Satellite Plasmonic Nanostructures: A Highly Sensitive and Reproducible Near-IR SERS Sensor. Adv. Funct. Mater. 2013, 23, 1519-1526. (29) Wang, C.; Du, Y.; Wu, Q.; Xuan, S.; Zhou, J.; Song, J.; Shao, F.; Duan, H. StimuliResponsive Plasmonic Core-Satellite Assemblies: imotif DNA Linker Enabled Intracellular pH Sensing. Chem. Commun. 2013, 49, 5739-5741. (30) Li, F.; Chen, H.; Zhang, Y.; Chen, Z.; Zhang, Z.-P.; Zhang, X.-E.; Wang, Q. ThreeDimensional Gold Nanoparticle Clusters with Tunable Cores Templated by a Viral Protein Scaffold. Small 2012, 8, 3832-3838. (31) Höller, R. P.; Dulle, M.; Thomä, S.; Mayer, M.; Steiner, A. M, Förster, S.; Fery, A.; Kuttner, C.; Chanana, M. Protein-Assisted Assembly of Modular 3D Plasmonic Raspberry-like Core/Satellite Nanoclusters: Correlation of Structure and Optical Properties. ACS Nano. 2016, 10, 5740-5750. (32) Guo, D. S.; Liu, Y. Calixarene-based Supramolecular Polymerization in Solution. Chem Soc Rev. 2012, 41, 5907-5911. (33) Ma, X.; Tian, H. Stimuli-responsive Supramolecular Polymers in Aqueous Solution. Acc Chem Res. 2014, 47, 1971-1981. (34) Chen, L.; Tian, Y.; Ding, Y.; Tian, Y.; Wang, F. Multistimuli Responsive Supramolecular Cross-Linked etworks On the Basis of the Benzo-21-Crown-7/Secondary Ammonium Salt Recognition Motif. Macromolecules 2012, 45, 8412-8419. (35) Chi, X.; Ji, X.; Xia, D.; Huang, F. A Dual-responsive Supra-amphiphilic Polypseudorotaxane Constructed From A Water-soluble Pillar[7]arene and An Azobenzene-containing Random Copolymer. J Am Chem Soc. 2015, 137, 1440-1443.

ACS Paragon Plus Environment

18

Page 19 of 20

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Applied Materials & Interfaces

(36) Stoffelen, C.; Voskuhl, J.; Jonkheijm, P.; Huskens, J. Dual Stimuli-responsive Selfassembled Supramolecular Nanoparticles. Angew Chem Int Ed. 2014, 53, 3400-3404. (37) Lan, Y.; Wu, Y.; Karas, A.; Scherman, O. A. Photoresponsive Hybrid Raspberry-Like Colloids Based On Cucurbit[8]uril Host-guest Interactions. Angew Chem Int Ed. 2014, 53, 2166-2169. (38) Liu, Y.; Yang, H.; Wang, Z.; Zhang, X. Cucurbit[8]uril-based Supramolecular Polymers. Chem Asian J. 2013, 8, 1626-1632. (39) Ni, X. L.; Chen, S.; Yang, Y.; Tao, Z. Facile Cucurbit[8]uril-Based Supramolecular Approach To Fabricate Tunable Luminescent Materials in Aqueous Solution. J Am Chem Soc. 2016, 138, 6177-6183. (40) Wen, F.; Zhang, W. Q.; Zheng, P. W.; Zhang, X.; Yang, X. L.; Wang, Y.; Jiang, X. W.; Wei, G.W.; Shi, L. Q. One-Stage Synthesis of Narrowly Dispersed Polymeric Core-shell Microspheres. J Polym Sci Part A: Polym Chem. 2008, 46, 1192-1202.

ACS Paragon Plus Environment

19

ACS Applied Materials & Interfaces

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 20 of 20

Table of Contents

ACS Paragon Plus Environment

20