Elsevier

Biomaterials

Volume 177, September 2018, Pages 176-185
Biomaterials

Decellularized peripheral nerve supports Schwann cell transplants and axon growth following spinal cord injury

https://doi.org/10.1016/j.biomaterials.2018.05.049Get rights and content

Abstract

Schwann cell (SC) transplantation has been comprehensively studied as a strategy for spinal cord injury (SCI) repair. SCs are neuroprotective and promote axon regeneration and myelination. Nonetheless, substantial SC death occurs post-implantation, which limits therapeutic efficacy. The use of extracellular matrix (ECM)-derived matrices, such as Matrigel, supports transplanted SC survival and axon growth, resulting in improved motor function. Because appropriate matrices are needed for clinical translation, we test here the use of an acellular injectable peripheral nerve (iPN) matrix. Implantation of SCs in iPN into a contusion lesion did not alter immune cell infiltration compared to injury only controls. iPN implants were larger and contained twice as many SC-myelinated axons as Matrigel grafts. SC/iPN animals performed as well as the SC/Matrigel group in the BBB locomotor test, and made fewer errors on the grid walk at 4 weeks, equalizing at 8 weeks. The fact that this clinically relevant iPN matrix is immunologically tolerated and supports SC survival and axon growth within the graft offers a highly translational possibility for improving efficacy of SC treatment after SCI. To our knowledge, it is the first time that an injectable PN matrix is being evaluated to improve the efficacy of SC transplantation in SCI repair.

Introduction

Following an insult to the mammalian spinal cord, a complex cascade of reactions contributes to an aggravated expansion of the initial injury, including the formation of fluid-filled cavities that partly contribute to the regenerative failure and cause extreme functional deficits [1]. Cell transplantation strategies following spinal cord injury (SCI) rely on reducing cavitation following damage, re-bridging the injured tissue and creating more favorable conditions for axonal regeneration [2]. Among the prime candidates for repair are Schwann cells (SCs), the myelinating glia of the peripheral nervous system. SCs can be obtained from SCI patient nerve, purified and expanded in culture, and autologously transplanted into the lesioned spinal cord [3]. SCs have been extensively studied and found to be neuroprotective, reduce cavitation, promote axon regeneration and myelination, and modestly improve hindlimb movements in rat SCI models [[4], [5], [6]]. These findings contributed to FDA-approved clinical trials evaluating the safety and efficacy of autologous SC therapy in sub-acute and chronic SCI subjects (www.clinicaltrials.gov NCT01739023; NCT02354625) [[7], [8], [9]].

After transplantation, however, SC survival rates are considerably low which limits the extent of their therapeutic action. Previous studies have shown that after a contusion injury in rats, only approximately 20% of the SCs survived when transplanted in culture medium [[10], [11], [12]]. The harsh injury environment, where oxidative stress, inflammation and immune responses occur, undoubtedly contributes to these low survival levels. In addition, the SC detachment from extracellular matrix (ECM) in the culture dish before transplantation, contributes to significant cell death through an apoptotic process designated anoikis [13]. The vehicle in which SCs are transplanted is thus very important and affects the treatment efficacy. Patel et al. [14] compared different ECM-derived in situ gelling formulations with culture medium as vehicles for SC implantation into a SCI site. Transplanting SCs in these matrices, particularly Matrigel, enhanced SC survival, axon growth, re-vascularization and functional recovery compared to a SC suspension. Matrigel is not clinically relevant, however, as it is derived from a mouse tumor and its composition varies from batch to batch [15]. A supportive and clinically acceptable matrix is needed for human SC implantation.

We hypothesize that peripheral nerve (PN) tissue, the natural environment of SCs, could provide an excellent support scaffold for SC transplantation after SCI. Decellularization technologies allow the removal of cellular components from source tissues that can elicit an immune response, while preserving the ECM components, thus providing attractive biomaterials for tissue engineering [16]. Implantation of decellularized biomaterials does not require immunosuppression, and the chemical intrinsic cues for cell function and guidance are maintained [17]. Nagao et al. [18] have developed an optimized acellular PN graft that has been shown to elicit similar functional recovery to an autograft in a rat sciatic nerve injury model. A similar human-derived scaffold is now FDA-approved for use in patients with PN discontinuities. An injectable formulation of acellular PN, recently established, mimics the composition of native nerve ECM, gels at physiological temperature and possesses mechanical properties similar to rat neural tissue [19]. We here investigate the potential of injectable PN (iPN) matrix to support transplanted SC survival and axon growth following SCI. Accordingly, we transplanted SCs in iPN and compared this to Matrigel, in a rat model of thoracic contusion. We evaluated the subacute immune response, graft volume, axon ingrowth and SC myelination, and locomotor function 8 weeks post-transplantation. SCs transplanted in iPN promoted twice as many axons and equivalent locomotor recovery, compared to Matrigel, raising exciting possibilities that can directly translate to future SCI clinical trial strategies.

Section snippets

Injectable acellular peripheral nerve matrix preparation

Decellularization of rat sciatic nerve was conducted according to methods previously developed in the Schmidt laboratory [20]. Briefly, sciatic nerves were aseptically harvested from Sprague Dawley rats and the epineurium was removed. The nerves were then stored in 1× phosphate buffered saline (PBS) at −20 °C until processing. Nerves were washed with double distilled water and detergent, with buffer washes (100 mM sodium/50 mM phosphate buffer) performed between detergent washes. The detergent

Quantification of SC viability in iPN and Matrigel in vitro

In vitro data demonstrated that the iPN scaffold was not cytotoxic to the seeded SCs. SCs were able to grow abundantly in both iPN and Matrigel, and in both conditions they were evenly distributed. SCs seeded in Matrigel exhibited a bipolar morphology (Fig. 2A), whereas in iPN displayed a rounded phenotype (Fig. 2B). Despite the unusual morphological appearance, SCs cultured in iPN maintained robust viability that was significantly higher (90.0 ± 1.7%) than the cells cultured in Matrigel

Discussion

The current study demonstrates that iPN is an appropriate injectable biomaterial to support transplanted SC survival and axon growth into a SCI site. Moreover, injection of iPN into a lesion is immunologically tolerated and supports as much locomotor recovery as Matrigel, making this matrix an attractive possibility for future translational studies. SC transplantation in Matrigel has been compared to other matrices as well as with SC suspension and shown superior SC survival, axon growth and

Conclusions

Matrices to support SC grafts have been recently explored and shown to improve SC transplantation efficacy after SCI. Nonetheless, no clinically relevant ECM matrix that could be used in human subjects has been reported to date. Our results with iPN revealed this material to be highly supportive for SC survival and myelination, axon growth, and locomotor recovery. Additionally, implantation of this matrix does not aggravate immune responses in the lesion site. As both SC transplantation in SCI

Disclosures

The authors declare no conflict of interest.

Author contributions

SRC,YL, RAW, CES and MBB designed the experiments; SRC, YL, MWM and RCC performed the experimental studies and acquired data; SRC, MWM and RAW analyzed data and performed statistical analysis; SRC prepared the manuscript; CES and MBB edited and reviewed the manuscript.

Acknowledgements

This work was supported by the National Institutes of Health [NIH NS 09923 (MBB)], The Miami Project to Cure Paralysis (MPCP) at the University of Miami, The Buoniconti Fund, the Craig Neilsen Foundation [222456 (CES)] and Conquer Paralysis Now (SRC). The authors would like to acknowledge: Yelena Pressman for preparing SCs for transplantation; the MPCP Virus Core for the virus production; the Animal Facility Core at the MPCP for assisting in animal care; Margaret Bates and Vania Almeida for TEM

References (59)

  • J.E. Reing

    The effects of processing methods upon mechanical and biologic properties of porcine dermal extracellular matrix scaffolds

    Biomaterials

    (2010)
  • R.J. Playford

    Epidermal growth factor is digested to smaller, less active forms in acidic gastric juice

    Gastroenterology

    (1995)
  • D.J. Donnelly et al.

    Inflammation and its role in neuroprotection, axonal regeneration and functional recovery after spinal cord injury

    Exp. Neurol.

    (2008)
  • B.N. Brown

    Macrophage phenotype and remodeling outcomes in response to biologic scaffolds with and without a cellular component

    Biomaterials

    (2009)
  • A. Trivedi et al.

    Inflammation and spinal cord injury: infiltrating leukocytes as determinants of injury and repair processes

    Clin. Neurosci. Res.

    (2006)
  • J.C. Fleming

    Alpha4beta1 integrin blockade after spinal cord injury decreases damage and improves neurological function

    Exp. Neurol.

    (2008)
  • J.W. Rowland

    Current status of acute spinal cord injury pathophysiology and emerging therapies: promise on the horizon

    Neurosurg. Focus

    (2008)
  • W. Tetzlaff

    A systematic review of cellular transplantation therapies for spinal cord injury

    J. Neurotrauma

    (2011)
  • M.B. Bunge

    Efficacy of Schwann cell transplantation for spinal cord repair is improved with combinatorial strategies

    J. Physiol.

    (2016)
  • H. Kanno

    Schwann cell transplantation for spinal cord injury repair: its significant therapeutic potential and prospectus

  • J. Guest et al.

    Clinical translation of autologous Schwann cell transplantation for the treatment of spinal cord injury

    Curr. Opin. Organ Transplant.

    (2013)
  • K.D. Anderson

    Safety of autologous human Schwann cell transplantation in subacute thoracic spinal cord injury

    J. Neurotrauma

    (2017)
  • C.E. Hill

    Early necrosis and apoptosis of Schwann cells transplanted into the injured rat spinal cord

    Eur. J. Neurosci.

    (2007)
  • D.D. Pearse

    Transplantation of Schwann cells and/or olfactory ensheathing glia into the contused spinal cord: survival, migration, axon association, and functional recovery

    Glia

    (2007)
  • C.E. Hill

    Labeled Schwann cell transplantation: cell loss, host Schwann cell replacement, and strategies to enhance survival

    Glia

    (2006)
  • V. Patel

    Suspension matrices for improved Schwann-cell survival after implantation into the injured rat spinal cord

    J. Neurotrauma

    (2010)
  • T. Hoshiba

    Decellularized matrices for tissue engineering

    Expet Opin. Biol. Ther.

    (2010)
  • R.J. Nagao

    Functional characterization of optimized acellular peripheral nerve graft in a rat sciatic nerve injury model

    Neurol. Res.

    (2011)
  • R.C. Cornelison

    Injectable hydrogels of optimized acellular nerve for injection in the injured spinal cord

    Biomed. Mater.

    (2018)
  • Cited by (76)

    View all citing articles on Scopus
    View full text