Elsevier

Biomaterials

Volume 30, Issue 8, March 2009, Pages 1482-1491
Biomaterials

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

https://doi.org/10.1016/j.biomaterials.2008.11.040Get rights and content

Abstract

Recently, macrophages have been characterized as having an M1 or M2 phenotype based on receptor expression, cytokine and effector molecule production, and function. The effects of macrophage phenotype upon tissue remodeling following the implantation of a biomaterial are largely unknown. The objectives of this study were to determine the effects of a cellular component within an implanted extracellular matrix (ECM) scaffold upon macrophage phenotype, and to determine the relationship between macrophage phenotype and tissue remodeling. Partial-thickness defects in the abdominal wall musculature of Sprague–Dawley rats were repaired with autologous body wall tissue, acellular allogeneic rat body wall ECM, xenogeneic pig urinary bladder tissue, or acellular xenogeneic pig urinary bladder ECM. At 3, 7, 14, and 28 days the host tissue response was characterized using histologic, immunohistochemical, and RT-PCR methods. The acellular test articles were shown to elicit a predominantly M2 type response and resulted in constructive remodeling, while those containing a cellular component, even an autologous cellular component, elicited a predominantly M1 type response and resulted in deposition of dense connective tissue and/or scarring. We conclude that the presence of cellular material within an ECM scaffold modulates the phenotype of the macrophages participating in the host response following implantation, and that the phenotype of the macrophages participating in the host response appears to be related to tissue remodeling outcome.

Introduction

The extracellular matrix (ECM) represents the secreted products, both structural and functional, of the resident cells of each tissue and organ. The composition and ultrastructure of the ECM is determined by factors that influence the phenotype of its resident cells including mechanical forces, biochemical milieu, oxygen requirements, pH and inherent gene expression patterns, among others. In turn, the ECM influences cell attachment, migration, proliferation and three-dimensional organization, serving as an “information highway” between cells [1], [2], [3]. For these reasons, multiple forms of allogeneic and xenogeneic ECM from sources such as small intestine, urinary bladder, and skin have been investigated as biologic scaffolds for tissue reconstruction in both preclinical studies and human clinical applications [4], [5]. Some studies have shown improved tissue remodeling outcomes when site appropriate autologous cells are either seeded onto the ECM scaffold prior to implantation or placed in contact with the scaffold in-situ [6], [7], [8]. However, the survival and fate of such a cellular component during the remodeling process following in vivo implantation is largely unknown, and the effect of the presence of these cells upon the host macrophage response has not been investigated.

Macrophages are a heterogeneous subset of the mononuclear cell population [9], [10], [11] involved in the host response to implanted materials. Macrophages are activated in response to tissue damage or infection, causing an increase in the production of cytokines, chemokines, and other inflammatory molecules to which they are exposed [9], [12], [13], [14]. Recently, macrophage phenotype has been characterized based on distinct functional properties, surface markers, and the cytokine profile of the microenvironment [9], [14], [15]. Polarized macrophages are referred to as either M1 or M2 cells, mimicking the Th1/Th2 nomenclature [9]. However, M1 and M2 represent extremes along a continuum that includes multiple macrophage phenotypes (M1, M2a, M2b and M2c) [14]. M1, classically activated proinflammatory, macrophages are known to be induced by IFN-γ alone or in combination with LPS, TNF and GM-CSF. In general, M1 activated macrophages express IL-12high, IL-23high, IL-10low; metabolize arginine; produce high levels of inducible nitric oxide synthetase (iNOS); secrete toxic reactive oxygen and nitric oxygen intermediates and inflammatory cytokines such as IL-1β, IL-6, and TNF; and are inducer and effector cells in Th1 type inflammatory responses [15]. In contrast, M2, alternatively activated, macrophages are induced by exposure to a variety of signals including the cytokines IL-4, IL-13, and IL-10, immune complexes, and glucocorticoid or secosteroid (vitamin D3) hormones. M2 activated macrophages express IL-12low, IL-23low, and IL-10high; have high levels of scavenger, mannose, and galactose receptors; produce arginase in the place of arginine, subsequently producing ornithine and polyamines; are involved in polarized Th2 reactions; and possess the ability to facilitate tissue repair and regeneration [10], [12], [13], [15].

Macrophages are a plastic cell population capable of sequentially changing their polarization in response to local stimuli during the process of wound healing [16], [17], [18]. The macrophages participating in the host response to an implanted material are exposed to multiple stimuli including cytokines and effector molecules secreted by cells including other macrophages that are participating in the host response, microbial agents, epitopes associated with the implanted biomaterial, and the degradation products of the biomaterial, among others. Therefore, it is logical to assume that the host macrophage response after implantation of a biomaterial is modulated via “cross-talk” between macrophages and the other cells involved in the host response as well as factors within the local microenvironment. The effects of macrophage phenotype upon the tissue remodeling outcome following the implantation of a biomaterial are largely unknown, but recognition of the predominant phenotypic profile may provide a tool by which a constructive and functional tissue remodeling outcome can be predicted and/or promoted.

The objectives of the present study were twofold: (1) to determine the effects of the presence of cells, either autologous or xenogeneic, within an implanted ECM scaffold material upon the phenotype of the macrophages participating in the host response, and (2) to determine the relationship between the M1/M2 profile of the macrophages participating in the host response and the downstream tissue remodeling outcome.

Section snippets

Overview

Sixty-four Sprague–Dawley rats were randomly divided into four separate groups of sixteen each. 1 cm × 1 cm defects were created in the ventrolateral abdominal wall musculature, and repaired using one of the following materials: (1) cellular autograft (autologous body wall tissue), (2) acellular allograft (allogeneic rat body wall ECM), (3) cellular xenograft (xenogeneic pig urinary bladder tissue), or (4) acellular xenograft (xenogeneic pig urinary bladder ECM). See Table 1. The treatment

Results

All of the animals in this study survived the surgical procedure and post-operative period without complications.

Discussion

The present study examined the effects of the presence of a cellular component within a scaffold derived from extracellular matrix upon the polarization of the macrophages participating in the host response following implantation. The study also examined the relationship between macrophage polarization and host tissue remodeling events until 28 days post implantation. The results of the study indicate that the presence of a cellular component within an ECM derived scaffold shifts the macrophage

Conclusion

The present study showed that the presence of a cellular component within an extracellular matrix scaffold modulates the phenotype of the macrophages participating in the host response following implantation. It was observed that those test articles that contained a cellular component, even an autologous cellular component, elicited a predominantly M1 type macrophage response and resulted in the deposition of dense connective tissue and/or scarring. Those test articles that did not contain a

References (36)

  • D.O. Freytes et al.

    Biaxial strength of multilaminated extracellular matrix scaffolds

    Biomaterials

    (2004)
  • S. Badylak et al.

    Morphologic study of small intestinal submucosa as a body wall repair device

    J Surg Res

    (2002)
  • K.J. Livak et al.

    Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method

    Methods

    (2001)
  • T.W. Gilbert et al.

    Decellularization of tissues and organs

    Biomaterials

    (2006)
  • U. Galili

    The alpha-gal epitope (Gal alpha 1-3Gal beta 1-4GlcNAc-R) in xenotransplantation

    Biochimie

    (2001)
  • T.W. Gilbert et al.

    A quantitative method for evaluating the degradation of biologic scaffold materials

    Biomaterials

    (2007)
  • D. Ingber

    Extracellular matrix and cell shape: potential control points for inhibition of angiogenesis

    J Cell Biochem

    (1991)
  • C.D. Mills et al.

    M-1/M-2 macrophages and the Th1/Th2 paradigm

    J Immunol

    (2000)
  • Cited by (743)

    View all citing articles on Scopus
    1

    Suite 200, 100 Technology Drive, Pittsburgh, PA 15219, USA. Tel.: +1 412 235 5177; fax: +1 412 235 5224.

    2

    Mathematical Sciences Building, 150 N. University Street, West Lafayette, IN 47907-2067, USA. Tel.: +1 765 496 8378; fax: +1 765 494 1736.

    View full text