Elsevier

Biomaterials

Volume 255, October 2020, 120154
Biomaterials

Engineering 3D skeletal muscle primed for neuromuscular regeneration following volumetric muscle loss

https://doi.org/10.1016/j.biomaterials.2020.120154Get rights and content

Abstract

Volumetric muscle loss (VML) overwhelms the native regenerative capabilities of skeletal muscle and has few effective treatments to regain lost muscle mass and function. Tissue engineered muscle constructs designed to promote neuromuscular regeneration are a promising therapeutic avenue. To date, there has been no engineered muscle construct for VML treatment that has incorporated a pharmacologic agent to promote neuromuscular regeneration. Here, we have modified electrospun fibrin microfiber bundles, which have demonstrated muscle regenerative potential, with the heparan sulfate proteoglycan, agrin, to stimulate innervation post-VML. Myoblasts cultured on microfiber bundles with either soluble or chemically tethered agrin demonstrated statistically significant increased clustering of acetylcholine receptors (AChRs) with soluble agrin displaying AChR clusters throughout the myofiber bundles, and tethered agrin displaying AChR clusters only at 10 μm from the substrate surface. Following implantation into murine VML defects for 4 weeks, constructs pre-treated with soluble or tethered agrin resulted in statistically significant increased neuromuscular junctions, regenerating myofibers, vascular infiltration, neural infiltration, and nuclear yes-associated protein (YAP) expression within the defect site compared to the control without agrin. The agrin-tethered microfiber bundles provided sustained agrin signaling within the regenerating site during the 4-week post-implantation periods and further augmented the density of regenerating myofibers in regenerated tissue with statistical significance compared to constructs with soluble agrin. These data demonstrate the neuromuscular regenerative potential of engineered muscle constructs pre-treated to induce AChR clustering with locally delivered agrin at the site of VML regeneration.

Introduction

The repair mechanism for skeletal muscle is severely compromised following volumetric muscle loss (VML), thus leading to chronic functional deficits [1,2]. Current treatments are often unsuccessful in restoring muscle function and are limited by donor site morbidity, lack of donor tissue, and the need for highly skilled surgical teams [3]. It was recently demonstrated that VML is accompanied by significant motoneuron axotomy and lost interaction between neurons and the injured skeletal muscle, a probable cause for the heightened levels of lost muscle function seen post-VML [4]. Tissue engineered grafts have great potential to create clinical treatment options for regeneration of muscle with VML, but previous approaches to regenerate the injured muscle remain limited in their ability to encourage neuromuscular junction (NMJ) regeneration within the healing tissue. Engineered muscle constructs that incorporate methods to encourage neural infiltration and the formation of functional NMJs post-VML provide promising avenues to restore interaction between the muscle and nerve.

Prior strategies to promote NMJ regeneration post-VML have included neurotization [5,6], the use of rehabilitative exercise [[7], [8], [9], [10]], and implantation of a pre-innervated construct [11]. These approaches have moderately increased neural infiltration, force recovery, and NMJ formation within and around the defect site. Despite incremental neuromuscular regeneration, the nerve and NMJ densities within the defects remain low and the newly formed NMJs exhibit abnormal morphologies. In addition, there has been little effort to quantify neuromuscular regeneration across the entire defect area, instead relying on subsets of the defect region at high magnifications that likely do not accurately represent variability in expression across the defect region. In native tissues, the mature NMJ contains densely clustered postsynaptic acetylcholine receptors (AChR) optimized for efficient signal transfer across the neuromuscular synapse and effective muscle contraction [12]. Agrin, a large heparan sulfate proteoglycan secreted by the nerve terminal, is vital for AChR cluster stabilization during embryonic development [13] and has been utilized extensively to induce AChR clustering in cultured myotubes [[14], [15], [16], [17], [18], [19], [20], [21], [22]]. Muscle constructs containing agrin physically mixed into a fibrin hydrogel and seeded with mouse C2C12 myoblasts were implanted subcutaneously in a non-VML defect near the peroneal nerve for 8 weeks and resulted in increased nerve infiltration, NMJ formation, and vascular infiltration [14], demonstrating the potential neuromuscular therapeutic benefits of scaffold-mediated agrin delivery in vivo. In this study, we resolved to test whether the delivery of soluble or chemically-tethered agrin could promote improved NMJ regeneration in the treatment of VML defects.

Although release of bioactive molecules from engineered constructs has been utilized in VML treatment to promote vascular infiltration [[23], [24], [25], [26]] and myogenesis [[25], [26], [27]], there have been no previously published constructs used for VML treatment that have incorporated a bioactive factor to induce nerve infiltration and the formation of neuromuscular junctions. In addition, prior VML treatments with scaffold-mediated delivery of bioactive molecules have utilized physical entrapment or passive adsorption to the scaffold prior to implantation, which rely on diffusion of the bioactive agent to the surrounding tissues [28]. In contrast, scaffolds incorporating immobilized bioactive agents offer the ability to control spatiotemporal presentation and local signaling to the regenerating tissue, avoid poor targeting efficiency, and extend factor bioactivity over time following implantation [[29], [30], [31]]. Our group has previously utilized electrospun fibrin microfiber bundles that mimic the native properties of skeletal muscle combined with C2C12 myoblasts to promote functional and histological regeneration post-VML [32]. In the current study, we have enhanced our in vitro muscle constructs to promote neuromuscular regeneration in VML defects through the delivery of agrin. We tested the hypothesis that immobilized agrin tethered to the surface of C2C12 myoblast-seeded microfiber bundles would maintain its in vitro bioactivity and induce AChR clustering in myotubes cultured on the 3D constructs. In addition, we tested the ability of tethered agrin constructs implanted in a murine VML defect model to improve neuromuscular regeneration via recruitment of acetylcholine receptors to overlap with nerves and form neuromuscular junctions at 4 weeks compared to constructs pre-treated with soluble or zero agrin.

Section snippets

Electrospinning fibrin scaffolds

Fibrin scaffolds were electrospun in a sterile environment with sterile solutions as previously described [32,33]. Parallel syringes containing solutions of fibrinogen (Sigma) or sodium alginate (Sigma) were connected via a Y-connector and extruded by syringe pumps with an applied voltage of 3–5 kV applied to a 27-G needle tip utilizing 1% fibrinogen with an extrusion rate of 4 ml/h and 0.75% alginate with an extrusion rate of 1 ml/h. Poly(ethylene oxide) (PEO, average Mv ~ 4000 kDa, Sigma) was

AChR cluster 3D spatial distribution is determined by agrin delivery method

Electrospun fibrin hydrogels designed to mimic the hierarchical structure and alignment of skeletal muscle were fabricated as previously described. A protocol to chemically tether proteins to the electrospun fibrin scaffolds using the EDC zero-length crosslinker was initially developed and validated using a fluorescent Cy3 antibody which has a similar size to agrin (Fig. S1A). Agrin was chemically tethered to acellular scaffolds at 0.08–2 μg/scaffold prior to seeding with C2C12 myoblasts, after

Discussion

Successful regeneration of VML defects includes the formation of mature NMJs that allow for efficient neural signal propagation and subsequent muscle contraction. Significant advances in the field of skeletal muscle tissue engineering have demonstrated varied levels of muscle tissue regeneration and functional recovery post-VML [9,23,[38], [39], [40], [41], [42], [43], [44], [45], [46], [47], [48]], but remain limited in their ability to promote neuromuscular regeneration. To date, there have

Conclusions

In conclusion, engineered muscle constructs pre-treated with agrin form dense AChR clusters in vitro and promote neuromuscular regeneration upon implantation in VML defects for four weeks. While all treatment groups resulted in improved muscle function, constructs pre-treated with both soluble and tethered agrin caused increased neurofilament and blood vessel infiltration, NMJ formation, and the presence of eMHC + regenerating myofibers compared to no agrin controls. In addition, sustained in

Author contributions

Jordana Gilbert-Honick: Designed research, Performed research, Analyzed data, Wrote the paper, Shama R. Iyer: Performed research, Sarah M. Somers: Performed research, Analyzed data, Hannah Takasuka: Performed research, Richard M. Lovering: Designed research, Analyzed data, Kathryn R. Wagner: Designed research, Analyzed data, Hai-Quan Mao: Designed research, Analyzed data, Warren L. Grayson: Designed research, Analyzed data, Wrote the paper.

Data availability

The raw/processed data required to reproduce these findings cannot be shared at this time due to technical or time limitations.

Declaration of competing interest

The authors declare no conflict of interest.

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

Acknowledgements

Funding was provided by the Maryland Stem Cell Research Fund (2016-MSCRFI-2692), the Wilmer Core Grant for Vision Research, Microscopy and Imaging Core Module (EY001765), the NIH (NIAMS NRSA F31 AR071759 and K01 AR074048), and the MDA DG 577897.

References (63)

  • M.T.A. Li et al.

    Functional analysis of limb recovery following autograft treatment of volumetric muscle loss in the quadriceps femoris

    J. Biomech.

    (2014)
  • B.T. Corona et al.

    The promotion of a functional fibrosis in skeletal muscle with volumetric muscle loss injury following the transplantation of muscle-ECM

    Biomaterials

    (2013)
  • K. Matsumoto-Miyai et al.

    Coincident pre- and postsynaptic activation induces dendritic filopodia via neurotrypsin-dependent agrin cleavage

    Cell

    (2009)
  • K.I. Watt et al.

    Yap is a novel regulator of C2C12 myogenesis

    Biochem. Biophys. Res. Commun.

    (2010)
  • K. Garg et al.

    Volumetric muscle Loss : persistent functional deficits beyond frank loss of tissue

    J. Orthop. Res.

    (2015)
  • J. Gilbert-Honick et al.

    Vascularized and innervated skeletal muscle tissue engineering

    Adv Healthc Mater

    (2019)
  • B.F. Grogan et al.

    Volumetric muscle loss

    J. Am. Acad. Orthop. Surg.

    (2011)
  • B.T. Corona et al.

    Impact of volumetric muscle loss injury on persistent motoneuron axotomy

    Muscle Nerve

    (2018)
  • K.W. VanDusen et al.

    Engineered skeletal muscle units for repair of volumetric muscle loss in the tibialis anterior muscle of a rat

    Tissue Eng.

    (2014)
  • B.T. Corona et al.

    Autologous minced muscle grafts: a tissue engineering therapy for the volumetric loss of skeletal muscle

    Am J Physiol Physiol

    (2013)
  • M. Quarta et al.

    Bioengineered constructs combined with exercise enhance stem cell-mediated treatment of volumetric muscle loss

    Nat. Commun.

    (2017)
  • K.H. Nakayama et al.

    Rehabilitative exercise and spatially patterned nanofibrillar scaffolds enhance vascularization and innervation following volumetric muscle loss

    npj Regen Med

    (2018)
  • S. Das et al.

    Pre-innervated tissue engineered muscle promotes a pro-regenerative microenvironment following volumetric muscle loss

    bioRxiv 840124

    (2019)
  • N. Singhal et al.

    Role of extracellular matrix proteins and their receptors in the development of the vertebrate neuromuscular junction

    Dev Neurobiol

    (2011)
  • V. Witzemann

    Development of the neuromuscular junction

    Cell Tissue Res.

    (2006)
  • B.G.X. Zhang et al.

    Combination of agrin and laminin increase acetylcholine receptor clustering and enhance functional neuromuscular junction formation in vitro

    Dev Neurobiol

    (2016)
  • J.B. Scott et al.

    Achieving acetylcholine receptor clustering in tissue-engineered skeletal muscle constructs in vitro through a materials-directed agrin delivery approach

    Front. Pharmacol.

    (2017)
  • W. Bian et al.

    Soluble miniagrin enhances contractile function of engineered skeletal muscle

    Faseb. J.

    (2012)
  • L.K. Lee et al.

    Mechanistic distinctions between agrin and laminin-1 induced aggregation of acetylcholine receptors

    BMC Neurosci.

    (2002)
  • D.J. Burkin et al.

    Laminin and alpha7beta1 integrin regulate agrin-induced clustering of acetylcholine receptors

    J. Cell Sci.

    (2000)
  • L. Perry et al.

    Genetically engineered human muscle transplant enhances murine host neovascularization and myogenesis

    Commun Biol

    (2018)
  • Cited by (0)

    View full text