Elsevier

Experimental Neurology

Volume 224, Issue 2, August 2010, Pages 403-414
Experimental Neurology

Anti-IL-6-receptor antibody promotes repair of spinal cord injury by inducing microglia-dominant inflammation

https://doi.org/10.1016/j.expneurol.2010.04.020Get rights and content

Abstract

We previously reported the beneficial effect of administering an anti-mouse IL-6 receptor antibody (MR16-1) immediately after spinal cord injury (SCI). The purpose of our present study was to clarify the mechanism underlying how MR16-1 improves motor function after SCI. Quantitative analyses of inflammatory cells using flow cytometry, and immunohistochemistry with bone marrow-chimeric mice generated by transplanting genetically marked purified hematopoietic stem cells, revealed that MR16-1 dramatically switched the central player in the post-traumatic inflammation, from hematogenous macrophages to resident microglia. This change was accompanied by alterations in the expression of relevant cytokines within the injured spinal cord; the expression of recruiting chemokines including CCL2, CCL5, and CXCL10 was decreased, while that of Granulocyte/Macrophage-Colony Stimulating Factor (GM-CSF), a known mitogen for microglia, was increased. We also showed that the resident microglia expressed higher levels of phagocytic markers than the hematogenous macrophages. Consistent with these findings, we observed significantly decreased tissue damage and reduced levels of myelin debris and Nogo-A, the axonal growth inhibitor, by MR16-1 treatment. Moreover, we observed increased axonal regeneration and/or sprouting in the MR16-1-treated mice. Our findings indicate that the functional improvement elicited by MR16-1 involves microglial functions, and provide new insights into the role of IL-6 signaling in the pathology of SCI.

Introduction

In the pathology of spinal cord injury (SCI), the primary mechanical injury is followed by post-traumatic inflammation, in which inflammatory cells such as neutrophils, hematogenous macrophages (blood-borne macrophages), and resident microglia accumulate at the lesion site. These inflammatory cells release reactive oxygen, nitrogen radicals, and proteases, which exacerbate tissue damage (Hausmann, 2003). Because the inflammation is regulated by pro-inflammatory cytokines, such as TNFα, IL-1β, and IL-6, these cytokines have been targets for potential pharmaceutical interventions for SCI (Nesic et al., 2001, Okada et al., 2004, Sharma et al., 2003, Tuna et al., 2001). Among these cytokines, IL-6 is known to promote the activation and infiltration of macrophages/microglia (Hurst et al., 2001, Van Wagoner and Benveniste, 1999), which are the major inflammatory cells in the injured spinal cord, and the overexpression of IL-6 extends the inflammation to worsen the tissue injury (Klusman and Schwab, 1997, Lacroix et al., 2002). Hypothesizing that a blockade of IL-6 signaling might reduce the extension of injury by post-SCI inflammation, we previously administered the anti-mouse IL6-receptor antibody (MR16-1) after SCI and demonstrated reduced inflammation, decreased astrogliosis, and enhanced tissue sparing, leading to improved functional recovery (Okada et al., 2004). As a humanized antibody for the human IL-6 receptor (MRA; tocilizumab) is already in clinical use for the treatment of Castleman's disease and rheumatoid arthritis (Choy et al., 2002, Nishimoto et al., 2000, Sato et al., 1993), this drug might represent a new option for the treatment of SCI.

However, recent studies using gene-knockout animals revealed that the continuous inhibition of IL-6 signaling is detrimental to functional recovery, by inhibiting axonal regeneration or causing failed gliosis, implying that IL-6 may also have a beneficial function in spinal cord repair (Cafferty et al., 2004, Okada et al., 2006). Furthermore, numerous studies suggest that inflammation is beneficial or even essential for spinal cord repair, because it clears tissue debris and involves the secretion of various neurotrophic factors (Donnelly and Popovich, 2008, Hashimoto et al., 2005, McTigue et al., 2000). This discrepancy prompted us to investigate how the administration of an anti-IL-6 receptor antibody immediately after SCI promotes the repair process.

One of the important determinants of the extent of secondary damage by inflammation is the nature of the recruited inflammatory cells. For example, the transplantation of macrophages that have been co-incubated with peripheral nerves or skin improves spinal cord repair (Bomstein et al., 2003, Schwartz et al., 1999). On the other hand, zymosan-activated macrophages have neurotoxic properties, although they can also have proregenerative effects (Gensel et al., 2009, Popovich et al., 2002, Steinmetz et al., 2005). Previous reports have shown that these differences in the characteristics of inflammatory cells depend not only on their state of activation, but also on their origin. A subpopulation of hematogenous macrophages is more cytotoxic than microglia, and their excessive infiltration into a lesion is detrimental to spinal cord repair (Gris et al., 2004, Popovich et al., 1999). Since IL-6 is known to promote macrophage infiltration after central nervous system (CNS) trauma (Klusman and Schwab, 1997, Lacroix et al., 2002), here we focused on the effect of the temporary inhibition of IL-6 signaling by MR16-1 on macrophages and microglia after SCI. The administration of MR16-1 reduced the infiltration of macrophages into the injured spinal cord, but increased the number of microglia residing there, thus switching the major inflammatory cell type at the lesion from hematogenous macrophages to resident microglia. A comparison of the expression of phagocytic markers by hematogenous macrophages and microglia revealed that the microglia had greater phagocytic ability against myelin debris after SCI. Consequently, this switch in major inflammatory cell type resulted in improved tissue sparing and debris clearance, which promoted neural repair after SCI.

Section snippets

Animals

74 adult female C57BL/6 J mice (8–10 weeks old) were used. C57BL/6 background CAG-EGFP transgenic mice that ubiquitously express EGFP under the control of the CAG promoter (Kawamoto et al., 2000) were kindly provided by Professor Jun-ichi Miyazaki (Osaka University, Osaka, Japan) and were bred in our animal facility. The ethics committee of our institution approved all the surgical and animal care procedures, in accordance with the Laboratory Animal Welfare Act, the Guide for the Care and Use of

Anti-IL-6 receptor antibody treatment reduced inflammatory cell accumulation

To examine the effect of MR16-1 on the infiltration of inflammatory cells after SCI, immunostaining for CD11b and Iba-1 was performed. Although CD11b is known to be expressed by granulocytes and some T cells, 93.6 ± 3.3% of the infiltrated cells were CD11b and Iba-1 double-positive at 4, 7, and 14 days post-injury (dpi), indicating that the immunocompetent cells present at the injured site at those times after SCI were mostly hematogenous macrophages and resident microglia (Figs. 1A–E). While the

Discussion

IL-6 is a pro-inflammatory cytokine that triggers secondary injury in the pathophysiology of SCI. IL-6 binds to soluble and membrane-bound IL-6-receptor to form a complexed ligand for gp130, the common signal transducer of IL-6 and its related cytokines. MR16-1 is a neutralizing antibody for IL-6-receptor that competitively inhibits its binding to IL-6, thereby blocking IL-6-receptor-mediated cell signaling. We previously reported that the systemic administration of MR16-1 decreases the

Author contributions

M.M., M.N., Y.T., M.L. and H.O. designed the research; M.M., O.Y., A.I., T.I., F.RM. and O.T. performed the in vivo experiments; S.M. and Y.M. generated the chimeric mice; Y.O. supplied the anti-IL-6 receptor antibody and analyzed the antibody results; M.M., M.N., S.O., F.RM. and H.K. analyzed the data; M.M., M.N. and H.O. wrote the paper; M.N. and H.O. supervised all the experiments.

Acknowledgments

This work was supported by grants from the Project for the Realization of Regenerative Medicine from the Ministry of Education, Culture, Sports, Science and Technology (MEXT), Japan to H.O.; the General Insurance Association of Japan to M.N., A.I. and Y.T.; and a Grant-in-Aid for the Global COE Program from MEXT to Keio University. This work was also supported by grants from the Grant of Orthopaedics and Traumatology Foundation, Inc. No. 0178 to A.I. We are grateful to Professor Claude Bernard

References (57)

  • D.M. McTigue et al.

    Localization of transforming growth factor-beta1 and receptor mRNA after experimental spinal cord injury

    Exp. Neurol.

    (2000)
  • M. Mildenberger et al.

    An animal model of prophylactic cranial irradiation: histologic effects at acute, early and delayed stages

    Int. J. Radiat. Oncol. Biol. Phys.

    (1990)
  • N. Nishimoto et al.

    Improvement in Castleman's disease by humanized anti-interleukin-6 receptor antibody therapy

    Blood

    (2000)
  • M. Okazaki et al.

    Characterization of anti-mouse interleukin-6 receptor antibody

    Immunol. Lett.

    (2002)
  • M. Penkowa et al.

    Astrocyte-targeted expression of IL-6 protects the CNS against a focal brain injury

    Exp. Neurol.

    (2003)
  • 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. Romano et al.

    Role of IL-6 and its soluble receptor in induction of chemokines and leukocyte recruitment

    Immunity

    (1997)
  • L.R. Saville et al.

    A monoclonal antibody to CD11d reduces the inflammatory infiltrate into the injured spinal cord: a potential neuroprotective treatment

    J. Neuroimmunol.

    (2004)
  • M. Schilling et al.

    Predominant phagocytic activity of resident microglia over hematogenous macrophages following transient focal cerebral ischemia: an investigation using green fluorescent protein transgenic bone marrow chimeric mice

    Exp. Neurol.

    (2005)
  • K.R. Swartz et al.

    Interleukin-6 promotes post-traumatic healing in the central nervous system

    Brain Res.

    (2001)
  • N.J. Van Wagoner et al.

    Interleukin-6 expression and regulation in astrocytes

    J. Neuroimmunol.

    (1999)
  • A.A. Babcock et al.

    Chemokine expression by glial cells directs leukocytes to sites of axonal injury in the CNS

    J. Neurosci.

    (2003)
  • B.S. Bregman et al.

    Recovery from spinal cord injury mediated by antibodies to neurite growth inhibitors

    Nature

    (1995)
  • W.B. Cafferty et al.

    Conditioning injury-induced spinal axon regeneration fails in interleukin-6 knock-out mice

    J. Neurosci.

    (2004)
  • E.H. Choy et al.

    Therapeutic benefit of blocking interleukin-6 activity with an anti-interleukin-6 receptor monoclonal antibody in rheumatoid arthritis: a randomized, double-blind, placebo-controlled, dose-escalation trial

    Arthritis Rheum.

    (2002)
  • J.C. Gensel et al.

    Macrophages promote axon regeneration with concurrent neurotoxicity

    J. Neurosci.

    (2009)
  • R.S. Ghirnikar et al.

    Chemokine antagonist infusion promotes axonal sparing after spinal cord contusion injury in rat

    J. Neurosci. Res.

    (2001)
  • D. Giulian et al.

    Colony-stimulating factors as promoters of ameboid microglia

    J. Neurosci.

    (1988)
  • Cited by (98)

    • Distribution and polarization of microglia and macrophages at injured sites and the lumbar enlargement after spinal cord injury

      2020, Neuroscience Letters
      Citation Excerpt :

      After SCI, secondary tissue damage occurs through an inflammatory response due to infiltration of the injured site by two monocytic lineage cell types: macrophages and activated resident microglia [6]. The macrophages have positive [7] and negative [8] effects on spinal repair, with an unclear overall effect. Activated resident microglia and macrophages differ in origin, and their relationship to the chronic inflammatory response, secondary damage and central NeP after SCI is unclear, particularly because differentiation of the cell types is difficult due to their morphological and phenotypic similarities [9] and a lack of cell-specific markers.

    View all citing articles on Scopus
    View full text