Elsevier

Biomaterials

Volume 33, Issue 34, December 2012, Pages 8793-8801
Biomaterials

Effect of modulating macrophage phenotype on peripheral nerve repair

https://doi.org/10.1016/j.biomaterials.2012.08.050Get rights and content

Abstract

Peripheral nerve repair across long gaps remains clinically challenging despite progress made with autograft transplantation. While scaffolds that present trophic factors and extracellular matrix molecules have been designed, matching the performance of autograft-induced repair has been challenging. In this study, we explored the effect of cytokine mediated ‘biasing’ of macrophage phenotypes on Schwann cell (SC) migration and axonal regeneration in vitro and in vivo. Macrophage phenotype was successfully modulated by local delivery of either Interferon-gamma (IFN-γ) or Interleukin-4 (IL-4) within polymeric nerve guidance channels, polarizing them toward pro-inflammatory (M1) or pro-healing (M2a and M2c) phenotypes, respectively. The initial polarization of macrophages to M2a and M2c phenotype results in enhanced SC infiltration and substantially faster axonal growth in a critically-sized rat sciatic nerve gap model (15 mm). The ratio of pro-healing to pro-inflammatory population of macrophages (CD206+/CCR7+), defined as regenerative bias, demonstrates a linear relationship with the number of axons at the distal end of the nerve scaffolds. The present results clearly suggest that rather than the extent of macrophage presence, their specific phenotype at the site of injury regulates the regenerative outcomes.

Introduction

Peripheral nerve defects are caused due to trauma or in the course of surgery [1]; however, bridging long gaps remains a clinical challenge [2], [3]. The current clinical approach for repairing long nerve deficits (15 mm or longer) is to use nerve autografts; however, the paucity of donor nerves, the disparity of donor nerve sizes with the recipient sites and the modality mismatch has compelled a search for alternatives that can match or exceed autograft performance [1], [2], [3], [4].

Several approaches to enhance regeneration have been explored including the design of novel nerve guidance channels [2], fillers within nerve guidance channels [5], local delivery of neurotrophic factors [6], [7], transplantation of cells [8], [9], and/or application of topographical cues [4], [10], [11]. While these approaches are promising, the overall success rate in matching autograft performance has been limited. Current approaches focus on enhancing axon growth by direct action on nerves, or glial cells, and here we investigate an alternative approach to influencing regenerative outcomes by modulating the initial inflammatory sequence via macrophages [12]. Since the sequence of cellular and molecular events associated with nerve regeneration is influenced by immune cells [13], [14], we hypothesize that modulating immune cells upstream of action on nerves or Schwann cells (SC) triggers endogenous repair mechanisms that can stimulate nerve repair across long gaps [12].

It is evident that the immune response plays an important role during regeneration in many tissues [15], [16], [17]. Macrophages are quite abundant and phenotypically diverse immune cell populations presented during nerve degeneration and regeneration [12], [13], [18]. Macrophages arrive at the site of injury within 24 h and their numbers at the site peak within 14–21 days [19], whereas it takes at least one week for lymphocyte influx to occur [13]. Macrophages, which are mainly recruited from circulation, account for the bulk of phagocytosis within days of peripheral nerve injury and play a critical role in debris removal, growth factor production, and remodeling of the extracellular matrix (ECM) of the distal nerve [20], [21]. Thus macrophages might represent an upstream ‘lever’ to influence downstream axon and SC regeneration (Fig. 1a).

Recently, macrophages have been demonstrated to have a spectrum of activation states/phenotypes which has led to categorizing them broadly as classically activated (M1) and alternatively activated (M2) macrophages [22], [23], [24]. M1 macrophages, which are activated by injury-triggered endogenous inflammatory signals, such as T-helper 1 (Th1) cytokine IFN-γ, considered to be inflammatory, microbicidal, and tumor destructive [23], [24]. M1 macrophages produce high levels of oxidative metabolites (e.g. nitric oxide and superoxide) as well as pro-inflammatory cytokines. On the other hand, M2 macrophages, which can be activated by Th2 cytokines such as IL-4, IL-10 and IL-13, support tissue repair by producing anti-inflammatory cytokines which mediate angiogenesis, cell replacement and matrix remodeling while suppressing destructive immunity [24], [25]. The M2 phenotype has also been shown to have its own subtypes (M2a, M2b and M2c) [26], [27]. While the exact role of each M2 subtype is not well defined, it has been shown that M2a and M2c enhance tissue repair and pro-healing functionality, but the M2b subtype possesses regulatory function with some characteristics of both anti-inflammatory and pro-inflammatory macrophages [26], [27].

Despite the fact that macrophages constitute a substantial fraction of the post-injury cellular milieu, very little attention has been devoted to understanding the contribution from different phenotypes of macrophages and the effect of their modulation on regeneration after injury, especially in the context of the peripheral nervous system (PNS) [13], [28]. Here, we modulate the phenotype of macrophages by employing two well-known cytokines IFN-γ or IL-4, polarizing them toward pro-inflammatory (M1) or pro-healing (M2a and M2c) phenotypes, respectively. Then, we investigate the influence of macrophage phenotype on the peripheral nerve regeneration in a critically-sized, non-healing 15 mm rat sciatic nerve gap.

Section snippets

SC proliferation study

Alveolar rat macrophage cells (NR8383)(2 × 105) were placed in serum-free medium (F12K with 1% P/S; 1% l-glutamine) and incubated, allowing attachment for 4 h (37 °C, 5% CO2). After 4 h, IL-4 or IFN-γ (50 ng/mL in 1 mL) was added and cells were incubated for 24 h (n = 4 per group). Then LPS (100 ng/mL) was added to IFN-γ containing medium and all cells were incubated for an additional 7 h. Then medium for all cells was changed to fresh serum-free medium and cells were incubated for another 24 h

Effect of polarized macrophages on SC proliferation and migration in vitro

Using a Boyden Chamber (BC), the effect of M1 or M2 macrophage phenotype on SC migration was explored in vitro. Because some of the secretory products of macrophages are labile, with short half-lives (e.g., oxygen and nitrogen free radicals), media transfer assays may underestimate the migratory potential of conditioned media (CM) from macrophages. Therefore, SCs grown on the trans-well of the BC were placed in the wells of pro-inflammatory and anti-inflammatory plated macrophages. The

Discussion

Although there is strong evidence to support the notion that macrophages modulate regeneration in the nervous system [12], [13], [18], it is unclear if they support [17], [33], [34] or hinder [35], [36] regenerative biochemical cascades after injury [37]. This apparent lack of clarity might stem from the ‘double-edged’ characteristics of macrophages [12], [38], [39]. On one hand macrophages, as the primary phagocytes of the innate immune system, release inflammatory neurotoxin and cytokines

Conclusions

Our data demonstrates that modulation of macrophage phenotype at the site of peripheral nerve injury can favorably bias the endogenous regenerative potential after injury that obviates the need for the downstream modulation of multiple factors and has significant implications for the treatment of long peripheral nerve gaps. Moreover, the present study strongly suggests that more than the extent of macrophage presence, their specific phenotype at the site of injury influence the regenerative

Acknowledgments

We would like to thank Dr. Balakrishna Pai, Dr. Tarun Saxena and Johnathan Lyon in the Bellamkonda Laboratory for helpful scientific and editorial discussions. We would also like to thank Dr. Balakrishna Pai for generating the GFP-expressing RSC96 Schwann cells used in the study. This work was supported by grants from the National Institutes of Health (NS44409, NS65109, 1R41NS06777-1).

References (60)

  • B.N. Brown et al.

    Macrophage polarization: an opportunity for improved outcomes in biomaterials and regenerative medicine

    Biomaterials

    (2012)
  • B.N. Brown et al.

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

    Biomaterials

    (2009)
  • I. Clements et al.

    Thin-film enhanced nerve guidance channels for peripheral nerve repair

    Biomaterials

    (2009)
  • P.G. Popovich et al.

    Depletion of hematogenous macrophages promotes partial hindlimb recovery and neuroanatomical repair after experimental spinal cord injury

    Exp Neurol

    (1999)
  • M. Schwartz et al.

    Microglial phenotype: is the commitment reversible?

    Trends Neurosci

    (2006)
  • M. Dodla et al.

    Differences between the effect of anisotropic and isotropic laminin and nerve growth factor presenting scaffolds on nerve regeneration across long peripheral nerve gaps

    Biomaterials

    (2008)
  • A. Balgude et al.

    Agarose gel stiffness determines rate of DRG neurite extension in 3D cultures

    Biomaterials

    (2001)
  • O. Butovsky et al.

    Microglia activated by IL-4 or IFN-[gamma] differentially induce neurogenesis and oligodendrogenesis from adult stem/progenitor cells

    Mol Cell Neurosci

    (2006)
  • K. Krick et al.

    Signaling cue presentation and cell delivery to promote nerve regeneration

    Curr Opin Biotechnol

    (2011)
  • B. Pfister et al.

    Biomedical engineering strategies for peripheral nerve repair: surgical applications, state of the art, and future challenges

    Crit Rev Biomed Eng

    (2011)
  • V. Mukhatyar et al.

    Tissue engineering strategies designed to realize the endogenous regenerative potential of peripheral nerves

    Adv Mater

    (2009)
  • M. Madaghiele et al.

    Collagen-based matrices with axially oriented pores

    J Biomed Mater Res A

    (2008)
  • E.G. Fine et al.

    GDNF and NGF released by synthetic guidance channels support sciatic nerve regeneration across a long gap

    Eur J Neurosci

    (2002)
  • E.C. Spivey et al.

    The fundamental role of subcellular topography in peripheral nerve repair therapies

    Biomaterials

    (2012)
  • D. Hoffman-Kim et al.

    Topography, cell response, and nerve regeneration

    Annu Rev Biomed Eng

    (2010)
  • N. Mokarram et al.

    Overcoming endogenous constraints on neuronal regeneration

    IEEE Trans Biomed Eng

    (2011)
  • A.D. Gaudet et al.

    Wallerian degeneration: gaining perspective on inflammatory events after peripheral nerve injury

    J Neuroinflammation

    (2011)
  • M. Brown et al.

    Consequences of slow Wallerian degeneration for regenerating motor and sensory axons

    J Neurobiol

    (1992)
  • L.A. DiPietro

    Wound healing: the role of the macrophage and other immune cells

    Shock

    (1995)
  • B. Chazaud et al.

    Dual and beneficial roles of macrophages during skeletal muscle regeneration

    Exerc Sport Sci Rev

    (2009)
  • Cited by (268)

    View all citing articles on Scopus
    View full text