An MMP-degradable and conductive hydrogel to stabilize HIF-1α for recovering cardiac functions

Xiaojuan Wei, Si Chen, Tian Xie, Hongchi Chen, Xin Jin, Jumin Yang, Shafaq Sahar, Huanlei Huang, Shuoji Zhu, Nanbo Liu, Changjiang Yu, Ping Zhu, Wei Wang, Wei Zhang, Xiaojuan Wei, Si Chen, Tian Xie, Hongchi Chen, Xin Jin, Jumin Yang, Shafaq Sahar, Huanlei Huang, Shuoji Zhu, Nanbo Liu, Changjiang Yu, Ping Zhu, Wei Wang, Wei Zhang

Abstract

Rationale: Although a few injectable hydrogels have shown a reliable biosafety and a moderate promise in treating myocardial infarction (MI), the updated hydrogel systems with an on-demand biodegradation and multi-biofunctions to deliver therapeutic drug would achieve more prominent efficacy in the future applications. In this report, a conductive and injectable hydrogel crosslinked by matrix metalloproteinase-sensitive peptides (MMP-SP) was rationally constructed to stabilize hypoxia-inducible factor-1α (HIF-1α) to recover heart functions after MI. Methods: Firstly, tetraaniline (TA) was incorporated into partially oxidized alginate (ALG-CHO) to endow the hydrogels with conductivity. The 1,4-dihydrophenonthrolin-4-one-3-carboxylic acid (DPCA) nanodrug was manufactured with high drug loading capacity and decorated with polymerized dopamine (PDA) to achieve a stable release of the drug. Both ALG-CHO and DPCA@PDA can be cross-linked by thiolated hyaluronic acid (HA-SH) and thiolated MMP-SP to construct a MMP-degradable and conductive hydrogel. After administration in the infarcted heart of rats, echocardiographic assessments, histological evaluation, and RT-PCR were used to evaluate therapeutic effects of hydrogels. Results: The cell viability and the results of subcutaneous implantation verify a good cytocompatibility and biocompatibility of the resulting hydrogels. The hydrogel shows remarkable strength in decreasing the expression of inflammatory factors, maintaining a high level of HIF-1α to promote the vascularization, and promoting the expression of junctional protein connexin 43. Meanwhile, the multifunctional hydrogels greatly reduce the infarcted area (by 33.8%) and improve cardiac functions dramatically with ejection fraction (EF) and fractional shortening (FS) being increased by 31.3% and 19.0%, respectively. Conclusion: The as-prepared hydrogels in this report achieve a favorable therapeutic effect, offering a promising therapeutic strategy for treating heart injury.

Trial registration: ClinicalTrials.gov NCT01226563.

Keywords: conductivity; hypoxia-inducible factor-1α; injectable hydrogel; matrix metalloproteinase; myocardial infarction.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interest exists.

© The author(s).

Figures

Figure 1
Figure 1
The schematic design of an MMP-degradable and conductive hydrogel to stabilize HIF-1α for recovering heart functions after MI. The hydrogel was fabricated based on functionalized HA and ALG. The conductive TA was incorporated into ALG-CHO to endow electric conductivity on the hydrogels. DPCA nanodrug was coated with PDA to achieve a high drug loading amount and maintain a stable releasing manner. Both ALG-CHO and DPCA@PDA are crosslinked by HA-SH and thiolated MMP-SP to construct the multifunctional hydrogels. The resulted hydrogels were administrated by intramyocardial injection into the infarcted rat hearts to evaluate therapy effects.
Figure 2
Figure 2
The synthesis and characterization of DPCA@PDA NPs. (A) The synthesis process of DPCA@PDA. (B) The size distribution of DPCA and DPCA@PDA NPs assessed by DLS. TEM images of DPCA NPs (C) and DPCA@PDA NPs (D).
Figure 3
Figure 3
Properties of the as-prepared ALG-CHO-TA/DPCA@PDA/MMP-SP/HA-SH hydrogels. (A) Rheological analysis based on a time-sweep model. (B) The macro picture to demonstrate the injectable capability of the hydrogels. (C) The shear-thinning behavior and (D) the fatigue resistance of ALG-CHO-TA/DPCA@PDA/MMP-SP/HA-SH hydrogel. (E) The macro picture indicates that the conductive hydrogel can act as a wire to light up LED. (F) The conductivity of the hydrogels. (G) The DPCA releasing behavior in vitro. (H) The macro pictures illustrate the subcutaneous injection of hydrogels within 7 d. (I) H&E staining of the tissue around the hydrogels (black zone in the images) after 7-day subcutaneous injection. (III: ALG-CHO/HA-SH; IV: ALG-CHO-TA/HA-SH; V: ALG-CHO-TA/DPCA@PDA/HA-SH; VI: ALG-CHO-TA/DPCA@PDA/MMP-SP/HA-SH.)
Figure 4
Figure 4
Evaluation of the cardiac functions after various treatments at different time points. (A) The representative echocardiographic images for various groups at 28 d. The statistical values of (B) EF, (C) FS, (D) LVID; d, and (E) LVID; s. (I: Sham; II: MI; III: ALG-CHO/HA-SH; IV: ALG-CHO-TA/HA-SH; V: ALG-CHO-TA/DPCA@PDA/HA-SH; VI: ALG-CHO-TA/DPCA@PDA/MMP-SP/HA-SH.)
Figure 5
Figure 5
Histological analysis of the cardiac tissue at 28 d. (A) TTC staining. (B) Masson staining. (C) Sirius red staining. Statistical data on LV wall thickness (D), the infarcted area (E), and the fibrosis area (F). (I: Sham; II: MI; III: ALG-CHO/HA-SH; IV: ALG-CHO-TA/HA-SH; V: ALG-CHO-TA/DPCA@PDA/HA-SH; VI: ALG-CHO-TA/DPCA@PDA/MMP-SP/HA-SH.)
Figure 6
Figure 6
Protein expression level of TNF-α (A) and Cas-3 (B) determined by immunofluorescent staining at 28 d. (I: Sham; II: MI; III: ALG-CHO/HA-SH; IV: ALG-CHO-TA/HA-SH; V: ALG-CHO-TA/DPCA@PDA/HA-SH; VI: ALG-CHO-TA/DPCA@PDA/MMP-SP/HA-SH.)
Figure 7
Figure 7
Immunofluorescent staining of cTnT (A, B) and Cx43 (C, D) at 28 d. B, D are the statistical data quantified and normalized to the number of nuclei. (I: Sham; II: MI; III: ALG-CHO/HA-SH; IV: ALG-CHO-TA/HA-SH; V: ALG-CHO-TA/DPCA@PDA/HA-SH; VI: ALG-CHO-TA/DPCA@PDA/MMP-SP/HA-SH.)
Figure 8
Figure 8
Protein expression level of HIF-1α (A, B), VEGFA (C, D), and α-SMA (E, F) at 28 d measured by immunofluorescent staining. B, D, and F are the statistical data quantified and normalized to the number of nuclei. (I: Sham; II: MI; III: ALG-CHO/HA-SH; IV: ALG-CHO-TA/HA-SH; V: ALG-CHO-TA/DPCA@PDA/HA-SH; VI: ALG-CHO-TA/DPCA@PDA/MMP-SP/HA-SH.)
Figure 9
Figure 9
Gene expression level at 28 d determined by RT-PCR. A-F indicate TNF-α, IL-1β, Ang-1, HIF-1α, α-Actinin, and cTnT. (I: Sham; II: MI; III: ALG-CHO/HA-SH; IV: ALG-CHO-TA/HA-SH; V: ALG-CHO-TA/DPCA@PDA/HA-SH; VI: ALG-CHO-TA/DPCA@PDA/MMP-SP/HA-SH.)

References

    1. Virani SS, Alonso A, Benjamin EJ, Bittencourt MS, Callaway CW, Carson AP. et al. Heart disease and stroke statistics-2020 update: a report from the American Heart Association. Circulation. 2020;141:E139–E596.
    1. Thavapalachandran S, Grieve SM, Hume RD, Le TYL, Raguram K, Hudson JE. et al. Platelet-derived growth factor-AB improves scar mechanics and vascularity after myocardial infarction. Sci Transl Med. 2020;12:eaay2140.
    1. Langin M, Mayr T, Reichart B, Michel S, Buchholz S, Guethoff S. et al. Consistent success in life-supporting porcine cardiac xenotransplantation. Nature. 2018;564:430–3.
    1. Hasan A, Khattab A, Islam MA, Hweij KA, Zeitouny J, Waters R. et al. Injectable hydrogels for cardiac tissue repair after myocardial Infarction. Adv Sci. 2015;2:1500122.
    1. Wang LL, Liu Y, Chung JJ, Wang T, Gaffey AC, Lu MM. et al. Sustained miRNA delivery from an injectable hydrogel promotes cardiomyocyte proliferation and functional regeneration after ischaemic injury. Nat Biomed Eng. 2017;1:983–92.
    1. Leor J, Tuvia S, Guetta V, Manczur F, Castel D, Willenz U. et al. Intracoronary injection of in situ forming alginate hydrogel reverses left ventricular remodeling after myocardial infarction in swine. J Am Coll Cardiol. 2009;54:1014–23.
    1. Sabbah HN, Wang MJ, Gupta RC, Rastogi S, Ilsar I, Sabbah MS. et al. Augmentation of left ventricular wall thickness with alginate hydrogel implants improves left ventricular function and prevents progressive remodeling in dogs with chronic heart failure. Jacc-Heart Fail. 2013;1:252–8.
    1. Rao SV, Zeymer U, Douglas PS, Al-Khalidi H, White JA, Liu JY. et al. Bioabsorbable intracoronary matrix for prevention of ventricular remodeling after myocardial infarction. J Am Coll of Cardiol. 2016;68:715–23.
    1. He HJ, Zhao YN, Zhang H, Wang B, Pan J, Li J. et al. Effect of intramyocardial grafting collagen scaffold with mesenchymal stromal cells in patients with chronic ischemic heart disease a randomized clinical trial. JAMA Netw Open. 2020;3:e2016236.
    1. Schirmer L, Chwalek K, Tsurkan MV, Freudenberg U, Werner C. Glycosaminoglycan-based hydrogels with programmable host reactions. Biomaterials. 2020;228:119557.
    1. Zhu Y, Jiang H, Ye SH, Yoshizumi T, Wagner WR. Tailoring the degradation rates of thermally responsive hydrogels designed for soft tissue injection by varying the autocatalytic potential. Biomaterials. 2015;53:484–93.
    1. Rane AA, Chuang JS, Shah A, Hu DP, Dalton ND, Gu YS. et al. Increased infarct wall thickness by a bio-inert material is insufficient to prevent negative left ventricular remodeling after myocardial infarction. Plos One. 2011;6:e21571.
    1. Lutolf MP, Lauer-Fields JL, Schmoekel HG, Metters AT, Weber FE, Fields GB. et al. Synthetic matrix metalloproteinase-sensitive hydrogels for the conduction of tissue regeneration: Engineering cell-invasion characteristics. P Natl Acad Sci USA. 2003;100:5413–8.
    1. Purcell BP, Lobb D, Charati MB, Dorsey SM, Wade RJ, Zellars KN. et al. Injectable and bioresponsive hydrogels for on-demand matrix metalloproteinase inhibition. Nat Mater. 2014;13:653–61.
    1. Deleon-Pennell KY, Altara R, Yabluchanskiy A, Modesti A, Lindsey ML. The circular relationship between matrix metalloproteinase-9 and inflammation following myocardial infarction. Iubmb Life. 2015;67:611–8.
    1. Fan CX, Shi JJ, Zhuang Y, Zhang LL, Huang L, Yang W. et al. Myocardial-infarction-responsive smart hydrogels targeting matrix metalloproteinase for on-demand growth factor delivery. Adv Mater. 2019;31:201902900.
    1. Ban K, Park HJ, Kim S, Andukuri A, Cho KW, Hwang JW. et al. Cell therapy with embryonic stem cell-derived cardiomyocytes encapsulated in injectable nanomatrix gel enhances cell engraftment and promotes cardiac repair. ACS Nano. 2014;8:10815–25.
    1. Lee SJ, Sohn YD, Andukuri A, Kim S, Byun J, Han JW. et al. Enhanced therapeutic and long-term dynamic vascularization effects of human pluripotent stem cell-derived endothelial cells encapsulated in a nanomatrix gel. Circulation. 2017;136:1939–54.
    1. Mihic A, Cui Z, Wu J, Vlacic G, Miyagi Y, Li SH. et al. A conductive polymer hydrogel supports cell electrical signaling and improves cardiac function after implantation into myocardial infarct. Circulation. 2015;132:772–84.
    1. Dong R, Ma PX, Guo BL. Conductive biomaterials for muscle tissue engineering. Biomaterials. 2020;229:119584.
    1. Zhang CY, Hsieh MH, Wu SY, Li SH, Wu J, Liu SM. et al. A self-doping conductive polymer hydrogel that can restore electrical impulse propagation at myocardial infarct to prevent cardiac arrhythmia and preserve ventricular function. Biomaterials. 2020;231:119672.
    1. Burnstine-Townley A, Eshel Y, Amdursky N. Conductive scaffolds for cardiac and neuronal tissue engineering: governing factors and mechanisms. Adv Funct Mater. 2020;30:201901369.
    1. Walker BW, Lara RP, Mogadam E, Yu CH, Kimball W, Annabi N. Rational design of microfabricated electroconductive hydrogels for biomedical applications. Prog Polym Sci. 2019;92:135–57.
    1. Wu YB, Wang L, Guo BL, Ma PX. Interwoven aligned conductive nanofiber yarn/hydrogel composite scaffolds for engineered 3D cardiac anisotropy. ACS Nano. 2017;11:5646–59.
    1. Liang W, Chen JR, Li LY, Li M, Wei XJ, Tan BY. et al. Conductive hydrogen sulfide-releasing hydrogel encapsulating ADSCs for myocardial infarction treatment. ACS Appl Mater Inter. 2019;11:14619–29.
    1. Wang W, Tan BY, Chen JR, Bao R, Zhang XR, Liang S. et al. An injectable conductive hydrogel encapsulating plasmid DNA-eNOs and ADSCs for treating myocardial infarction. Biomaterials. 2018;160:69–81.
    1. Thangarajah H, Yao D, Chang EI, Shi Y, Jazayeri L, Vial IN. et al. The molecular basis for impaired hypoxia-induced VEGF expression in diabetic tissues. P Natl Acad Sci USA. 2009;106:13505–10.
    1. Eltzschig HK, Bratton DL, Colgan SP. Targeting hypoxia signalling for the treatment of ischaemic and inflammatory diseases. Nat Rev Drug Discov. 2014;13:852–69.
    1. Zhang Y, Strehin I, Bedelbaeva K, Gourevitch D, Clark L, Leferovich J. et al. Drug-induced regeneration in adult mice. Sci Transl Med. 2015;7:290ra92.
    1. Stoehr A, Yang YQ, Patel S, Evangelista AM, Aponte A, Wang GH. et al. Prolyl hydroxylation regulates protein degradation, synthesis, and splicing in human induced pluripotent stem cell-derived cardiomyocytes. Cardiovasc Res. 2016;110:346–58.
    1. Heber-Katz E, Messersmith P. Drug delivery and epimorphic salamander-type mouse regeneration: A full parts and labor plan. Adv Drug Deliver Rev. 2018;129:254–61.
    1. Esser TU, Roshanbinfar K, Engel FB. Promoting vascularization for tissue engineering constructs: current strategies focusing on HIF-regulating scaffolds. Expert Opin Biol Ther. 2019;19:105–18.
    1. Cheng J, Amin D, Latona J, Heber-Katz E, Messersmith PB. Supramolecular polymer hydrogels for drug-induced tissue regeneration. ACS Nano. 2019;13:5493–501.
    1. Nagai K, Ideguchi H, Kajikawa T, Li XF, Chavakis T, Cheng J, An injectable hydrogel-formulated inhibitor of prolyl-4-hydroxylase promotes T regulatory cell recruitment and enhances alveolar bone regeneration during resolution of experimental periodontitis. Faseb J. 2020.
    1. Tambuwala MM, Manresa MC, Cummins EP, Aversa V, Coulter IS, Taylor CT. Targeted delivery of the hydroxylase inhibitor DMOG provides enhanced efficacy with reduced systemic exposure in a murine model of colitis. J Control Release. 2015;217:221–7.
    1. Piantanida E, Alonci G, Bertucci A, De Cola L. Design of Nanocomposite injectable hydrogels for minimally invasive surgery. Acc Chem Res. 2019;52:2101–12.
    1. Park J, Brust TF, Lee HJ, Lee SC, Watts VJ, Yeo Y. Polydopamine-based simple and versatile surface modification of polymeric nano drug carriers. ACS Nano. 2014;8:3347–56.
    1. Cheng W, Nie JP, Gao NS, Liu G, Tao W, Xiao XJ. et al. A multifunctional nanoplatform against multidrug resistant cancer: merging the best of targeted Chemo/Gene/Photothermal therapy. Adv Funct Mater. 2017;27:201704135.
    1. Wu YH, Wang HB, Gao F, Xu ZY, Dai FY, Liu WG. An injectable supramolecular polymer nanocomposite hydrogel for prevention of breast cancer recurrence with theranostic and mammoplastic functions. Adv Funct Mater. 2018;28:201801000.
    1. Wang W, Chen JR, Li M, Jia HZ, Han XX, Zhang JX. et al. Rebuilding postinfarcted cardiac functions by injecting TIIA@PDA nanoparticle-cross-linked ROS-sensitive hydrogels. ACS Appl Mater Inter. 2019;11:2880–90.
    1. Han X, Li L, Xie T, Chen S. et al. “Ferrero-like” nanoparticles knotted injectable hydrogels to initially scavenge ROS and lastingly promote vascularization in infarcted hearts. Sci China Tech Sci. 2020;63:2435–2448.
    1. Bao XF, Zhao JH, Sun J, Hu M, Yang XR. Polydopamine Nanoparticles as Efficient Scavengers for Reactive Oxygen Species in Periodontal Disease. ACS Nano. 2018;12:8882–92.
    1. Wang X, Wang CP, Wang XY, Wang YT, Zhang Q, Cheng YY. A Polydopamine nanoparticle-knotted poly(ethylene glycol) hydrogel for on-demand drug delivery and chemo-photothermal therapy. Chem Mater. 2017;29:1370–6.
    1. Zhu Y, Matsumura Y, Wagner WR. Ventricular wall biomaterial injection therapy after myocardial infarction: Advances in material design, mechanistic insight and early clinical experiences. Biomaterials. 2017;129:37–53.
    1. Ruvinov E, Cohen S. Alginate biomaterial for the treatment of myocardial infarction: Progress, translational strategies, and clinical outlook From ocean algae to patient bedside. Adv Drug Deliver Rev. 2016;96:54–76.
    1. Balakrishnan B, Joshi N, Jayakrishnan A, Banerjee R. Self-crosslinked oxidized alginate/gelatin hydrogel as injectable, adhesive biomimetic scaffolds for cartilage regeneration. Acta Biomater. 2014;10:3650–63.
    1. Hua YJ, Gan YB, Zhang YQ, Ouyang B, Tu B, Zhang CM. et al. Adaptable to mechanically stable hydrogels Based on the dynamic covalent cross-linking of thiol-aldehyde addition. ACS Macro Lett. 2019;8:310–4.
    1. Liang S, Zhang Y, Wang H, Xu Z, Chen J, Bao R. et al. Paintable and rapidly bondable bonductive hydrogels as therapeutic cardiac patches. Adv Mater. 2018;30:e1704235.
    1. Qazi TH, Rai R, Dippold D, Roether JE, Schubert DW, Rosellini E. et al. Development and characterization of novel electrically conductive PANI-PGS composites for cardiac tissue engineering applications. Acta Biomater. 2014;10:2434–45.
    1. Broughton KM, Wang BJ, Firouzi F, Khalafalla F, Dimmeler S, Fernandez-Aviles F. et al. Mechanisms of cardiac repair and regeneration. Circ Res. 2018;122:1151–63.
    1. Yao Y, Ding J, Wang Z, Zhang H, Xie J, Wang Y. et al. ROS-responsive polyurethane fibrous patches loaded with methylprednisolone (MP) for restoring structures and functions of infarcted myocardium in vivo. Biomaterials. 2020;232:119726.
    1. Yuan Z, Tsou YH, Zhang XQ, Huang S, Yang Y, Gao M. et al. Injectable citrate-based hydrogel as an angiogenic biomaterial improves cardiac repair after myocardial infarction. ACS Appl Mater Interfaces. 2019;11:38429–39.
    1. Yang H, Qin X, Wang H, Zhao X, Liu Y, Wo HT. et al. An in vivo miRNA delivery system for restoring infarcted myocardium. ACS Nano. 2019;13:9880–94.
    1. Dobaczewski M, Gonzalez-Quesada C, Frangogiannis NG. The extracellular matrix as a modulator of the inflammatory and reparative response following myocardial infarction. J Mol Cell Cardiol. 2010;48:504–11.
    1. Feiner R, Engel L, Fleischer S, Malki M, Gal I, Shapira A. et al. Engineered hybrid cardiac patches with multifunctional electronics for online monitoring and regulation of tissue function. Nat Mater. 2016;15:679–85.
    1. Guo BL, Ma PX. Conducting polymers for tissue engineering. Biomacromolecules. 2018;19:1764–82.
    1. Cui H, Liu Y, Cheng Y, Zhang Z, Zhang P, Chen X. et al. In vitro study of electroactive tetraaniline-containing thermosensitive hydrogels for cardiac tissue engineering. Biomacromolecules. 2014;15:1115–23.
    1. Zhou T, Yan LW, Xie CM, Li PF, Jiang LL, Fang J. et al. A Mussel-inspired persistent ROS-scavenging, electroactive, and osteoinductive scaffold based on electrochemical-driven in Situ nanoassembly. Small. 2019;15:e1805440.
    1. Nakada Y, Canseco DC, Thet S, Abdisalaam S, Asaithamby A, Santos CX. et al. Hypoxia induces heart regeneration in adult mice. Nature. 2017;541:222–7.
    1. Kimura W, Xiao F, Canseco DC, Muralidhar S, Thet S, Zhang HM. et al. Hypoxia fate mapping identifies cycling cardiomyocytes in the adult heart. Nature. 2015;523:226–43.

Source: PubMed

3
订阅