Next Article in Journal
Fabrication Scaffold with High Dimensional Control for Spheroids with Undifferentiated iPS Cell Properties
Next Article in Special Issue
Functionalizing Collagen Membranes with MSC-Conditioned Media Promotes Guided Bone Regeneration in Rat Calvarial Defects
Previous Article in Journal
The Distinct Assignments for Hsp90α and Hsp90β: More Than Skin Deep
Previous Article in Special Issue
Effect of Different Application Modalities on the Bonding Performance of Adhesive Systems to Dentin: A Systematic Review and Meta-Analysis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Macrophages: From Simple Phagocyte to an Integrative Regulatory Cell for Inflammation and Tissue Regeneration—A Review of the Literature

1
Institute of Pathology, University of Regensburg, 93053 Regensburg, Germany
2
Central Biobank Regensburg, University and University Hospital Regensburg, 93053 Regensburg, Germany
3
Department of Cardiology, University Medical Centre, Johannes Gutenberg University of Mainz, 55131 Mainz, Germany
4
German Center for Cardiovascular Research (DZHK), Partner Site Rhine Main, 55131 Mainz, Germany
5
Medical Center, Faculty of Medicine, Institute for Exercise and Occupational Medicine, University of Freiburg, 79106 Freiburg, Germany
6
Clinic and Polyclinic for Orthopedics and Trauma Surgery, University Hospital of Cologne, 50937 Cologne, Germany
7
Department of Diagnostic and Interventional Radiology, University Hospital Heidelberg, 69120 Heidelberg, Germany
8
Translational Lung Research Centre Heidelberg (TLRC), German Lung Research Centre (DZL), 69120 Heidelberg, Germany
9
Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
10
Department of Sports Medicine, Medical Clinic VII, University Hospital Heidelberg, 69120 Heidelberg, Germany
11
Department of Internal Medicine, St. Vincenz and Elisabeth Hospital of Mainz (KKM), 55131 Mainz, Germany
12
Department of Dermatology, University Medical Centre Regensburg, 93053 Regensburg, Germany
13
Department for Trauma Surgery, University Hospital Regensburg, 93053 Regensburg, Germany
14
Institute of Pathology, University Medical Centre Mannheim, Ruprecht-Karls-University Heidelberg, 68167 Mannheim, Germany
*
Author to whom correspondence should be addressed.
Cells 2023, 12(2), 276; https://doi.org/10.3390/cells12020276
Submission received: 29 November 2022 / Revised: 29 December 2022 / Accepted: 7 January 2023 / Published: 11 January 2023
(This article belongs to the Special Issue Mineralized Tissues Repair and Regeneration)

Abstract

:
The understanding of macrophages and their pathophysiological role has dramatically changed within the last decades. Macrophages represent a very interesting cell type with regard to biomaterial-based tissue engineering and regeneration. In this context, macrophages play a crucial role in the biocompatibility and degradation of implanted biomaterials. Furthermore, a better understanding of the functionality of macrophages opens perspectives for potential guidance and modulation to turn inflammation into regeneration. Such knowledge may help to improve not only the biocompatibility of scaffold materials but also the integration, maturation, and preservation of scaffold-cell constructs or induce regeneration. Nowadays, macrophages are classified into two subpopulations, the classically activated macrophages (M1 macrophages) with pro-inflammatory properties and the alternatively activated macrophages (M2 macrophages) with anti-inflammatory properties. The present narrative review gives an overview of the different functions of macrophages and summarizes the recent state of knowledge regarding different types of macrophages and their functions, with special emphasis on tissue engineering and tissue regeneration.

1. Introduction

In the 19th century, the Russian zoologist Ilja (Elie) Metchnikow [1] described a cell, which was able to engulf other cells, bacteria, or solid particles respectively [2,3,4,5]. He called this process phagocytosis. Based on the relatively large diameter of these “eating cells”, the early biologists called them macrophages. After cellular uptake, macrophages kill engulfed cells or organisms. Within the inflammatory process, macrophages, along with neutrophilic granulocytes, are the first line of cellular defense. Metchnikow hypothesized that the inflammatory reaction underwent evolutionary changes like any other biological process. Since macrophages can migrate on their own and integrate foreign bodies, Metchnikow set these cells as the first step in the inflammatory reaction [1]. According to this evolutionary hypothesis, the inflammatory reaction is developed step by step by the involvement of lymphocytes and granulocytes, the cascade of humoral factors of the complement system, and as the latest step, the production of antibodies [6].
Nowadays, our understanding of inflammation and the role of macrophages has dramatically changed. Macrophages are widely accepted to secrete pro-inflammatory or anti-inflammatory cytokines to achieve the orchestration between the different immune cells [7,8,9]. Cytokines are small protein signaling molecules that regulate cells’ growth, differentiation, and function [10]. Today, the term macrophage describes a heterogeneous group of cells with various functions in diverse cellular processes [7]. The first evidence for their heterogeneity was given by Aderem et al., who discovered that macrophages respond to bacterial lipopolysaccharide (LPS) without inducing an inflammatory response via T-cells [11]. Another milestone was the identification of a macrophage subpopulation called “alternatively activated macrophages” (AAM) [12]. Recent studies have shown that macrophages are also involved in synthesizing extracellular matrix (ECM) [13]. Mosser hypothesized that the potency of macrophages to synthesize ECM components gives evidence that these cells potentially have a primary role in tissue repair and not microbial killing [7]. These initial statements give insight into the concept that macrophages play a crucial role in the immune response to pathogens, tissue homeostasis and inflammation, as well as in regeneration and repair [13,14,15]. The present review gives an overview of the different functions of macrophages and summarizes the current state of the literature regarding different types of macrophages and their functions without claiming to be exhaustive.

2. The Origin and Formation of Macrophages

The origin of macrophages has been a matter of debate in recent years. Traditionally, macrophages were seen as descendants of monocytes [16,17]. Monocytes represent a group of white blood cells derived from the myelopoietic stem cells in the bone marrow as all other types of blood and immune cells [18,19,20]. Monocytes are primarily encountered in the blood as circulating cells, but also in the bone marrow and spleen and are incapable of steady state proliferation in these surroundings [21,22]. After their formation in the bone marrow, monocytes enter the blood, where they circulate [10,23] and migrate into various tissues reacting to different stimuli. Such stimuli for monocytic migration may either derive from inflammation or as a result of trauma. After migration into the tissue, monocytes form colonies under the action of chemotactic stimuli (Figure 1) [24].
The classical assumption that all macrophages originate from circulating monocytes was discarded years ago. Nowadays, it is well-accepted that hematopoiesis unfolds in three sequential waves, and macrophages undergo self-renewal within the tissue they reside [25]. The comprehensive debate about the origin of macrophages is not focused on in this review and is well demonstrated elsewhere [25,27,28].

3. Polarization of Macrophages

Once it became apparent that macrophages not only phagocytize but also express other functions, it was necessary to categorize them further. There is evidence that macrophages are a cell type that can assume various phenotypes based on the stimuli to which they are exposed. Because these cells respond differently to environmental signals [29], the classification centers on their activation mode [14,30,31,32,33]. In this context, macrophages have been categorized into the following subpopulations [31]:
  • Classically activated macrophages (CAM, M1-macrophages)
  • Alternatively activated macrophages (AAM, M2-macrophages)
In 2008, Mosser proposed an alternative classification of macrophages based on three homeostatic activities, which are host defense, wound healing, and immune regulation [22,34]. Furthermore, tumor-associated macrophages have also been identified as a separate group extensively studied in the last few years [35,36,37].

3.1. The Plasticity of Macrophages

The mechanisms resulting in the different macrophage phenotypes are crucial to understanding macrophage subpopulations. This process is called “plasticity” [5] and describes the ability of cells to respond to different microenvironmental influences by displaying diverse functional phenotypes [38,39]. Thus, plasticity results in a polarization of macrophages into different phenotypes assigned to the different subpopulations [14,30,40]. Taking these facts together, it is essential to realize that unlike other cells, which lose their heterogeneity during maturation, macrophages retain their plasticity and transform according to environmental signals [20,29]. Furthermore, there is evidence that the phenotype of polarized M1 and M2 macrophages could be reversed not only experimentally in vitro and in vivo but also in situ (Figure 2) [41,42,43,44]. For instance, in vitro analyses clearly demonstrated the capacity of macrophages to switch between M1 and M2 macrophages using different recombinant cytokines and biologically active substances measured by their CD163 and CD206 expression and their CCL18 and CCL3 production [41]. Furthermore, a switch from M1 to M2 polarized macrophages is described in experimental and human parasite infections [45,46].
In living organisms, the phenomenon of macrophage plasticity ensures that when M1-macrophages have completed the clearance of pathogens and any destroyed surrounding tissue, they transform into M2-macrophages to produce components of the extracellular matrix and simultaneously activate and induce other cells, such as fibroblasts, which also contribute to the formation of extracellular matrix, and thus initiate tissue regeneration [31,47,48]. Another important aspect regarding the plasticity of macrophages concerns their relationship to the modulation of chronic disease and autoimmunity [49]. For a long time, it was taken as a matter of fact that the incomplete or failed switching from one phenotype to another had an impact on chronic inflammation and autoimmune disorders [42]. In chronic venous ulcers, for example, it was shown that macrophages infiltrating the tissue fail to switch from an M1 to an M2 state due to iron overload. Therefore, ROS-mediated DNA damage, fibroblast cellular senescence, and defective tissue repair occur [50]. More recently, it has been demonstrated that even effective macrophage phagocytosis of apoptotic cells is crucially involved in the modulation of chronic inflammatory and autoimmune diseases, which underlines the active regulatory role of macrophages in these pathomechanisms [3,31].
Depending on their phenotype, macrophages differ regarding their metabolism being able to switch from an aerobic to an anaerobic state and vice versa. M1-macrophages use glycolysis and pentose phosphate pathways to meet their energy needs. The tricarboxylic acid cycle (TCA) is broken at two points, and itaconate and succinate accumulate. Besides, oxidative phosphorylation and fatty acid oxidation are downregulated. In contrast, M2 macrophages have an intact TCA and increased fatty acid oxidation and oxidative phosphorylation [51]. Another prime example demonstrating the importance of the plasticity of macrophages for their functionality is given by arginine metabolism in the differently polarized macrophages. M1 and M2 macrophages use different arginine-catabolizing enzymes. M1-macrophages metabolize arginine via inducible nitric oxide synthase (iNOS) into nitric oxide (NO) and citrulline, whereas arginase hydrolyzes arginine to ornithine and urea in M2-macrophages. In further downstream pathways, ornithine is broken down into polyamine and proline, which are essential for cellular proliferation and tissue repair [30,52]. M1 and M2 macrophages also differ in their iron metabolism, associated with the respective macrophage function. Iron is essential for bacterial growth as some bacteria obtain energy from the oxidation of divalent iron. Inflammatory M1 macrophages express low levels of hemoglobin receptors (CD163 and CD91), leading to a smaller heme pool within the macrophage. Also, they show high levels of ferritin, an iron storage protein, and low levels of ferroportin, an iron exporting channel affiliated with iron retention resulting in a bacteriostatic effect. In contrast, M2 macrophages express low ferritin levels and high ferroportin levels. The resulting iron release is linked to tissue repair, angiogenesis, and tumor promotion [8,53,54,55]. Therefore, metabolic adaption is a crucial feature of macrophage polarization.

3.2. Classically Activated Macrophages (CAM or M1-Macrophages)

CAMs or M1-macrophages are the best-characterized macrophage subpopulation. These cells represent the classical phagocyte [15]. The term “classically activated macrophages” describes those macrophages rising during cell-mediated immune responses [20]. These cells can elicit an effective innate immune response [31].

3.2.1. The Activation Process of Classically Activated Macrophages

One of the main activators of M1-macrophages is interferon-gamma (IFN-γ) [56,57]. This cytokine was originally called macrophage-activating factor (MAF). However, today the term MAF is not restricted to IFN-γ but also includes other cytokines and active molecules. IFN-γ has a variety of functions. Thus, it is involved in eliminating viral and intracellular bacterial infections and the mechanisms of tumor control. IFN-γ is an immunomodulator, an immunestimulus, and also can inhibit viral replication directly [58]. In innate immunity, IFN-γ is produced by natural killer T-lymphocytes (NKT) and natural killer (NK) cells. During an antigen-specific immunoreaction, IFN-γ is synthesized by CD8-positive cytotoxic T-lymphocytes (CTL) and CD4-positive TH1-T-helper cells. IFN-γ activates the transcription factors STAT-1 and STAT-2, which bind to gamma-activated sequences (GAS) at various immunological effector genes. As a result, activated macrophages secrete pro-inflammatory cytokines as well as oxygen and nitrogen radicals [7,59,60,61,62,63,64]. Another important activating molecule for macrophages is tumor necrosis factor-alpha (TNF-α), formerly known as cachectin. This molecule is produced by macrophages and is a member of the cytokine family of polypeptide mediators, which also contain interferons and interleukins. TNF-α is an important mediator during inflammation, immune responses, and infectious phenomena. One of the effects of TNF is the initiation of apoptosis, including in tumor cells [65,66,67,68,69].
The activation process in classically activated macrophages occurs either in the presence of IFN-γ alone or in combination with other co-stimulating factors [70]. However, a second stimulus is mandatory after the initial stimulation with IFN-γ. In these second stimulating processes, ligands to Toll-like receptors (TLRs) are intimately involved. TLRs bind several microbial components. After ligand-binding, the activated receptor initiates a signal transduction pathway which triggers the production of gene products, which control innate immune responses and further instruct the development of antigen-specific acquired immunity [71,72,73]. The ligands for TLRs are expressed on microorganisms and are known as so-called “pathogen-associated molecular patterns” (PAMPs) [60]. The PAMPs are defined as molecules associated with groups of pathogens that are recognized by cells of the innate immune system. These molecules can be described as low molecular weight signals in a class of microbes. TLRs and other pattern recognition receptors (PRRs) can recognize these PAMP molecules. PAMPs are concerned with the activation of innate immune responses. Essential components of the PAMP molecular family are endotoxins found on the cell membrane of gram-negative bacteria, also known as LPS [74,75,76].
The activation of TLRs induces the synthesis of TNF-α, which can act in an autocrine manner to amplify the stimulation of macrophages [77]. Some other TLR-Ligands are able to induce endogenous production of IFN-β [78], which can substitute for IFN-γ [20]. Therefore, after the first activation of CAMs with IFN-γ and LPS, the CAMs are further activated from the endogenously produced TNF-α and IFN-β (Figure 3) [20].
M1-macrophages are co-stimulated by additional pro-inflammatory cytokines such as IL-1, IL-12, or other stress signals [48,77]. Stress signals are endogenous factors released from damaged or stressed tissue [79], such as heat-shock proteins, fibronectin fragments, hyaluronan or high-mobility group box 1 proteins [80,81]. Heat-shock proteins, so-called chaperone molecules, are expressed as a reaction to several stressful conditions, such as infections or malignancies [82,83,84]. They are molecules, that detect proteins that had failed to fold or lost their native functional conformation in the cell preventing the aggregation of these proteins [85,86,87]. This phenomenon is described as the stress response [88,89,90]. Fibronectin fragments represent a cleavage product of the extracellular matrix due to the action of metalloproteinases, which are secreted by monocytes during inflammation [91,92]. Fibronectin is a macrophage activator [93]. Hyaluronan is a significant component of the ECM and modulates the inflammatory response [94,95,96]. In this context, larger polysaccharide chains promote anti-inflammatory activity, and smaller to medium size polysaccharide chains have pro-inflammatory properties [90,97,98,99]. High-mobility group box 1 (HMGB-1) is a structural co-factor critical for proper transcriptional regulation in somatic cells and is typically located in the nucleus [100,101]. This molecule, among others, induces inflammation, proliferation, and migration of cells [102,103,104]. HMGB-1 is also passively released by necrotic but not apoptotic cells. Furthermore, it is secreted by activated macrophages [105]. Besides the stimulating stress signals described above, classically activated macrophages can also be co-stimulated by a variety of other molecules [8,16] and hypoxia [106,107,108,109,110].

3.2.2. The Function of Classically Activated Macrophages (CAMs)

The best-described function of M1-macrophages or CAMs is the phagocytosis of pathogens [4,20,111,112,113]. Metchnikoff first described this function more than a hundred years ago [114,115]. Phagocytic activity and the synthesis of toxic agents, such as reactive oxygen and nitrogen species, are reasons why classically activated macrophages belong to the “microbicidal” repertoire of the organism [116,117]. It is important to underline that CAMs express an isoform of iNOS that cannot be detected until the CAMs are activated via IFN-γ and LPS [116,118]. iNOS is an enzyme that synthesizes NO by oxidation of the amino acid L-arginine. NO represents a critical mediator which reacts with superoxide anion (O2), resulting in the production of peroxynitrite (ONOO), and these radicals are responsible for the subsequent oxidative damage [119,120,121,122].
To effectively fulfill their function in hostdefense, CAMs secrete various pro-inflammatory cytokines, such as TNFα, IL-1, IL-6, IL-12, and IL-23 [22,123,124,125,126,127]. It is interesting to realize that especially IL-23 [128] but also IL-1 [129,130,131,132], and IL-6 [129,133] have been described as playing an important role in the development of the T-helper cell type 17 (Th17). These cells produce IL-17 [134], which triggers cascades involved in the induction of inflammation and autoimmunity [113,135,136,137,138,139]. In addition to the elimination of pathogens, classically activated macrophages also can present antigens via the MHC-II pathway [7].
As mentioned above, CAMs are also involved in destroying extracellular matrix and tissue reorganization during inflammation or trauma. To achieve this, CAMs produce and secrete various enzymes such as matrix-metalloproteinases (MMPs), macrophage metalloelastase (MMP12), collagenase and hyaluronidase [2,140,141,142,143,144]. MMPs form a group of zinc-dependent proteolytic endoproteinases, which degrade extracellular matrix proteins to support normal tissue remodeling and contribute to tissue destruction during various pathological conditions such as cell-material interactions and tumor cell invasion [145]. Some MMPs also play a role in macrophage polarization. For example, MMP8 has been shown to induce the M2 phenotype via the regulation of TGF-β expression [146,147]. The most important effect of extracellular matrix degradation is the support of macrophage migration through the inflamed tissue to facilitate their functions in clearing cell debris and pathogens (Figure 4).

3.3. M2-Macrophages

M2-macrophages are a subpopulation of macrophages that are not activated by the classical pathway via IFN-γ and TNF-α. Gordon et al. introduced the term “alternatively activated macrophages” to characterize a population of macrophages that have to be exposed to IL-4 for activation [60,148]. Also, extracellular nucleotides can influence the differentiation of macrophages into M2-macrophages [149]. M2-macrophages play an important role in various conditions, including immunoregulation, infections, wound healing, and modification of the extracellular matrix by the secretion of proteases and growth factors [150,151]. The group of M2-macrophages includes a minimum of three subpopulations [152,153], which are categorized based on their in vitro activation and polarization pathway:
  • M2a-Macrophages (alternatively activated macrophages, AAM)
  • M2b-macrophages (Type 2—macrophages)
  • M2c-macrophages (deactivated macrophages)
M2-macrophages are capable of releasing the anti-inflammatory cytokine IL-10, thus achieving a Th2-Response [31]. IL-10 is a cytokine with anti-inflammatory activity [154], which has been unequivocally established in various models of infection, inflammation, and even in cancer [155,156,157]. It is a potent inhibitor of antigen presentation and inhibits major histocompatibility complex class II expression and the upregulation of the co-stimulatory molecules CD80 and CD86 [158].

3.3.1. M2a-Macrophages

This subgroup of M2-macrophages is also termed “alternatively activated macrophages” (AAM) [12,159]. These macrophages are characterized by their low expression level of IL-12 [70,159]. In vitro monocytes transform into M2a-macrophages after treatment with IL-4 and IL-13 [14,159,160,161,162,163,164,165]. IL-13 shares a common receptor with IL-4 and exerts similar effects on macrophages [45,166]. IL-4 and IL-13 are cytokines released from various cell sources, including basophils, mast cells, Th2-T-Cells, and innate lymphoid cells. The two interleukins, IL-4 and IL-13, share several structural characteristics and both molecules antagonize the actions of IFN-γ [148,166,167] (Figure 5). After activation by IL-4 and IL-13, M2a-macrophages produce and release IL-1 receptor antagonists, which inhibit IL-1 function [168].
M2a-macrophages are further characterized by their abundant levels of non-opsonic receptors (such as mannose receptor, which is also known as CD206) and the failure (incompetence) to produce NO [45] via the induction of arginase [169], which leads to the generation of ornithine and polyamines [51,70]. They are also characterized by the production of low levels of pro-inflammatory cytokines (IL-1, TNF, and IL-6) and the low expression of the co-stimulatory molecule CD86 [170].
Functionally, macrophages are key regulators of fibrosis and resolution. This crucial mechanism is mediated by stabilin-1, a transmembrane glycoprotein expressed by endothelial cells and a subtype of macrophages. It was demonstrated that stabilin-1 expressed by macrophages regulates fibrosis in liver injury [171,172]. In this context, M2a-macrophages have also been described to have a pro-fibrotic potential [167]. In this context, in vitro studies demonstrated that after activation of macrophages with IL-4 or TGF-β, consecutively added myofibroblasts showed an increase in proliferation and the production of fibronectin and collagen I [173,174,175]. However, IL-4-activated macrophages can produce fibronectin and additional matrix proteins, including the TGF-ß-inducible gene H3 (bIG-H3), to a higher degree than classically activated macrophages. Furthermore, the “alternatively activated” macrophages that differentiate in response to IL-4 and IL-13 are involved in Th2-type responses (production of IL-10), including humoral immunity and wound healing [176]. An interesting finding was that activation through IL-4 could lead to an induction of a fusogenic status. This means these macrophages can build multinucleated giant cells (MNGCs) in the presence of other functional components [177,178] (Figure 6). Although some studies show that MNGCs express an M2 rather than M1 phenotype, the exact correlation between macrophage polarization and MNGC formation remains to be further investigated [179,180].

3.3.2. M2b-Macrophages

To achieve the M2b-macrophage polarization, the macrophages need to be exposed to lipopolysaccharides (agonists of TLR) [181] in the presence of IgG-immune complexes [182,183,184] (Figure 7). Macrophages which, by the time of activation, are exposed to IgG-immune complexes, synthesize large amounts of IL-10 but do not produce IL-12 [182,185].
Despite their high production of inflammatory cytokines and toxic molecules, it could be shown in animal studies that M2b macrophages protect mice against LPS toxicity. Moreover, they promote Th2 differentiation and humoral antibody production [7,70,186,187]. Thus, the M2b-macrophages are more similar to M1 macrophages than alternatively activated macrophages. The M2b are capable of synthesizing NO and have a low arginase activity compared to M2a- und M2c-macrophages. On the other hand, they express the CD86-receptor on their membrane and produce pro-inflammatory cytokines such as TNF, IL-1 and IL-6. One of the basic differences between the M1- and M2b- phenotype is that M2b are able to induce a Th2-response due to the production of IL-10 [20,183], whereas M1-subpopulations induce a Th1-response following their production of IL-12 [188].

3.3.3. M2c-Macrophages

IL-10, TGFβ, or glucocorticoids are required to polarize a macrophage population into the M2c-subgroup [160,162]. After polarization, the M2c-macrophages can produce IL-10 and TGFβ for self-stimulation (autocrine effect). The basic functions of M2c-macrophages are immunosuppression, remodeling of ECM, including matrix deposition, and tissue remodeling [34,189]. Furthermore, the induction of fibrosis triggered by M2c-macrophages has also been reported [31]. Fibrogenesis is a dynamic process in which the synthesis and deposition of ECM components occur as an answer to parenchymal tissue injury. This process plays a pivotal role in multiple physiological and pathological conditions, such as the granulation of wound healing, atherosclerosis, and chronic inflammation [174]. Fibrosis is characterized by the extensive proliferation and activation of tissue fibroblasts, the primary producers of extracellular components [190]. One of the important mediators to modulate proliferation and consecutive ECM components are transforming growth factor β 1 (TGFβ-1) (Figure 8) [191,192].

3.4. Tumor-Associated Macrophages

In addition to the functions mentioned above, macrophages are essential players in tumorigenesis, tumor promotion, and metastases as they orchestrate cancer-related inflammation and support angiogenesis [193]. During carcinogenesis, circulating monocytes and/ or tissue-resident macrophages are recruited to the tumor niche by mediators secreted by tumor cells and cells of the tumor microenvironment (TME) [193,194,195,196]. These so-called Tumor-Associated Macrophages (TAMs) are a highly plastic, heterogeneous subpopulation of macrophages that cannot be fully captured by the traditional M1/M2 dichotomy [197]. Often, TAMs are referred to as M2d or M2-like macrophages, which might lead to the assumption that only the M2 phenotype occurs in TME. However, M1-like and M2-like macrophages coexist within the TME, secreting opposing factors resulting in their distinct functions [198]. Remarkably, the TAM phenotype is not static and may switch from M1 to M2 as TAMs are sensitive to factors secreted by the TME. One possible explanation for the switch from M1 to M2-like is the expression of adenosine A2A receptors (A2AR) on the surface of M1 macrophages under hypoxic conditions. Adenosine binds to the A2AR suppressing the pro-inflammatory cytokine production (TNF-α, IL-12) and enhancing the secretion of anti-inflammatory and pro-angiogenic factors such as IL-10 and VEGF [187,199]. Furthermore, TAM subsets show a co-expression of M1 and M2 gene signatures, which underlines their broad phenotype spectrum [200].
As described earlier, phenotypical polarization to M1 occurs due to the effect of IFN-y, TNF-α, LPS, and others. The antitumor potential of M1-like TAMs is based on the lysis of tumor cells after phagocytosis or on the secretion of immunostimulatory cytokines and chemokines (e.g., IL-6, IL-12, TNF) which induce inflammation and thus tumor suppression [201]. In contrast, M2-like TAMs are more abundant in the TME and are accepted to be tumor-promoting. As described previously, M2-like TAMs are polarized by IL-4, IL-10, TGFβ-1, and PGE2. Across many cancer entities, the occurrence of M2-like TAMs is linked to numerous tumor-supportive properties such as enhanced tumor cell proliferation, angiogenesis, metastasis, immune suppression, drug resistance, and poor prognosis [202]. The role of TAMs is an extensively reviewed topic in the literature and remains an ongoing field of research. To complete the picture of macrophage heterogeneity, TAMs are mentioned very briefly in the present review but without the claim to be exhaustive.

4. Macrophages and Their Role in Tissue Regeneration

The changing view on macrophages and their different functions in inflammation, wound healing, and regeneration has begun to influence our understanding of their role in different cellular mechanisms in tissue engineering. This concerns not only the biocompatibility of scaffold materials but also the integration, maturation, and preservation of cell-scaffold constructs or induced regeneration. Macrophages are regulators in inflammatory and immunological processes within the tissue, and there are several aspects of tissue engineering in which macrophages play a pivotal role with respect to biomaterials. Relevant examples are the induction of inflammation and host responses as a reaction to biomaterial implants [8,31,48,203,204]. In this context, it is generally accepted that the macrophage is a central element of the inflammatory response, which is practically universally involved in the tissue reaction to implanted biomaterials [205].

4.1. Immunomodulatory Potential of IL-4

The role of macrophages is regulatory since the phenotypic differentiation to M1 or M2 macrophages is decisive for the secretion of pro- or anti-inflammatory cytokines. As the anti-inflammatory M2 phenotype is associated with improved tissue regeneration, biomaterials should be modified to avoid disadvantageous tissue reactions. Examples of disadvantageous tissue reactions are stenosis in grafts as a result of intima hyperplasia by excessive macrophage infiltration within tissue-engineered vascular grafts [206], but also the formation of peritoneal adhesions or fibrosis after surgical treatment and implantation of biomaterials within the peritoneal cavity [207]. Tan et al. could achieve advantageous tissue reactions in mice by using a bioactive vascular graft coated with IL-4 pushing macrophage polarization toward the M2 phenotype. Consequently, they observed a reduction of foreign body encapsulation and inhibition of neointimal hyperplasia compared to the control group [208]. Recently, resident peritoneal murine macrophages were found to represent an anti-adhesion cell barrier by forming a shield around surgery-induced fibrin clots. Nevertheless, this barrier is frequently inadequate, allowing adhesions to form. By injecting IL-4c, the macrophage barrier was strengthened, and post-operative adhesions were effectively prevented [209]. In the context of macrophage polarization, IL-4 seems to be a promising agent, and further research needs to be performed to establish it in clinical practice.

4.2. Importance of Nanomaterial Characteristics

With a view to the use of various biomaterials as cell carriers, scaffolds, or release systems for signaling molecules and growth factors to trigger tissue regeneration, there is a need to understand the role played by macrophages in the biocompatibility and biodegradation of such implanted materials. If the material is incompatible with the organism, either a severe inflammatory reaction or a foreign body reaction (FBR) is induced, in both of which macrophages are of pathogenetic importance [203]. For instance, Barsch et al. examined whether inflammation and FBR were significantly influenced by the 3D biomaterial design by comparing filamentous fleece and sponge-like biomaterial in a porcine model. Although no statistically significant difference could be found regarding FBR, the sponge-like synthetics showed a significantly lower inflammatory reaction which was quantified based on the density of polymorph-nucleated cells [210]. A further component of biocompatibility is the degradation of biomaterials and the tissue reaction to the degradation products. In this context, macrophages first trigger early acute inflammation, which is mandatory for the elimination of damaged molecules, and then they initiate and regulate the regenerative process [8,211,212,213,214]. However, macrophages are also involved in pathological processes resulting from disturbed wound healing, such as scar formation or delayed and failed regeneration [151]. Taking this pathophysiological function into account, recent strategies are aimed at controlling or modulating macrophages for tissue repair and regeneration. Garash et al. suggest strategies, which include controlled delivery of anti-inflammatory drugs, delivery of macrophages as a component of cellular therapy, controlled release of cytokines that modulate the macrophage phenotype and the design of nanoparticles that exploit the inherent phagocytic character of macrophages [215]. Nanomaterials are an emerging field of interest, but the potential of nanoparticles (NPs) as macrophage regulators has yet to be fully exploited. Ni et al. identified gold NPs as a potential periodontitis treatment option since injection of 45 nm NPs in induced periodontitis in rats resulted in significant anti-inflammatory effects such as M2 polarization [216].

4.3. Role of Hydrogels and Water-Soluble Substances

Furthermore, Kim and Tabata suggest an enhancement of wound healing by dual release patterns of stromal-derived cell factor-1 and a macrophage recruitment agent from gelatin Hydrogels. The authors demonstrated that culturing macrophages on fibrin gels stimulated the secretion of the anti-inflammatory cytokine interleucin-10 (IL-10) [217]. Hydrogels represent a three-dimensional network filled with water that mimics tissue microenvironment and is therefore considered to be biocompatible material. By conjugating signaling molecules, hydrogels can trigger cells to fulfill distinct functions. For tissue engineering, degradable hydrogels are favored as they can be replaced by growing tissue [218]. Regarding disease and cancer treatment, injectable hydrogels are auspicious biomaterials that can serve as scaffolds and carriers of therapeutic agents [219]. For instance, Xu et al. injected gelatin hydrogel into intracerebral hemorrhage lesions in mice. The researchers demonstrated that inflammation was suppressed in the intervention group. Macrophage polarization was observed to shift towards the M2 phenotype leading to a decline in the secretion of inflammatory cytokines, resulting in reduced neuronal loss and enhanced functional recovery [220].
Shiratori et al. showed that drugs could polarize macrophages into different subtypes. For example, Azithromycin, tofacitinib, hydroxychloroquine, and pioglitazone exhibit an anti-inflammatory profile by downregulation of M1 markers and upregulation of some M2 markers [221]. On the other hand, Huang et al. show that synthetic waterborne polyurethane nanoparticles (PU NPs) can inhibit the macrophage polarization toward the M1 phenotype but not toward the M2 phenotype [222]. In contrast, exposure of macrophages to soluble fibrinogen leads to the secretion of large amounts of inflammatory cytokine TNF-α. In conclusion, fibrin exerts a protective effect on macrophages, preventing inflammatory activation. From these findings, the authors concluded that fibrin and fibrinogen might represent key players in regulating macrophage phenotype behavior [217]. A differential regulation of macrophage inflammatory activation by fibrin and fibrinogen was also shown by Hsieh, Smith et al. [223].

4.4. Role of Iron in Macrophage Polarization

As described above, M1 and M2 macrophages differ in their iron metabolism, so iron oxide nanoparticles (IONPs) are a potent inducer of a switch of polarization. On the one hand, IONPs have been shown to activate macrophages and inhibit tumor growth on their own; on the other hand, IONPs have been used to deliver tumor-suppressing or macrophage-activating biomolecules [224]. Another option to shift TAMs from an M2-like to an M1-like phenotype was reported by Sang et al. They used Sulfur Quantum Dots as a nano trap for free iron ions, which then led to the production of reactive oxygen species and consequently to the reprogramming of macrophages to an M1-like phenotype. The so-activated macrophages could then suppress tumor growth via the activation of immune responses [225].

4.5. Influence of the Injury Microenvironment

In each tissue, the injury microenvironment is different. Although the injury triggers, in general, a cascade of more or less the same reactions, the microenvironment and the tissue responses to damage are derived from the tissue composition and the nature of the injury. These unique microenvironments were shown in sterile inflammation against pathogen-mediated inflammation due to the damage-associated molecular patterns recognized by inflammatory cells and not by pathogen-associated molecular patterns. For example, bone regeneration is based on different activities than skeletal muscle regeneration, as reflected by the differences in cytokine, chemokine, and growth factors present during homeostasis and wound healing in these two tissues [226]. If we understand the microenvironment of the inflamed tissue, then it might be easier to develop strategies regarding the reactions between cells and biomaterials. Also, for biomaterial-mediated tissue repair strategies that use endogenous monocyte/macrophage populations, the microenvironment of inflammatory damage can decisively contribute to the criteria for material design. Immunoregenerative materials can be designed to release molecules to enhance or disrupt specific features of the lesion to facilitate repair [227,228], but they should also prioritize the general healing goals of a particular tissue. Firstly, we must understand the properties of a tissue and its responses to injury into consideration of its caused microenvironment and then combine all this knowledge with the polarization of macrophages to achieve proper healing. The cells have to be “guided” in a particular wound environment, according to the damaged tissue [226]. The modulation of macrophages and their phenotype polarization was also shown by Lee et al. They suggest, for example, positive modulation of macrophage phenotype polarization (i.e., towards the regenerative M2 rather than the inflammatory M1 phenotype) with a modified surface, which is essential for the osteogenesis function of Titanium (Ti) bone implants. They showed that nanoscale topographical modification and surface bioactive ion chemistry could positively modulate the macrophage phenotype in a Ti implant surface. They induced the regenerative M2 macrophage phenotype of cells in nanostructured Ti surfaces [229]. Zhu et al. analyzed the modifying role of surface topography on macrophage polarization. The minimal scale of TiO2 honeycomb-like structures of 90 nanometers was most effective in stimulating the M2 phenotype. Thus, a favorable anti-inflammatory microenvironment was created, being beneficial for bone formation and osteointegration [230]. Thus, macrophages are also of particular importance for bone and cartilage formation as well as their remodeling [231,232]. From these examples, we can conclude that by controlling the tissue environment and microenvironment, we can control the macrophage behavior and modulate the macrophage phenotype [215].

4.6. Role of Macrophages in Angiogenesis

Angiogenesis is a further crucial mechanism that combines macrophages with various processes in tissue engineering. Macrophages take part not only in wound healing as such but also in angiogenesis to support the development and remodeling of vascular networks [110,233,234,235]. Angiogenesis is a multistep process in which macrophages are involved in each step. M2 macrophages secrete proteases (e.g., MMP9) and thus cleave the ECM to create space for the newly forming vessels. In parallel, resting endothelial cells are activated by paracrine stimulation [235]. For this purpose, mainly M1 macrophages secrete pro-angiogenic factors such as VEGF-A, TNF, or FGF2 [236,237,238,239]. After loosening the basement membrane, endothelial sprouting gives rise to new capillaries that migrate toward angiogenic stimuli and then fuse with other sprouts or capillaries to form anastomoses. In “tip cell guidance”, M2 macrophages wrap around the sprouts to facilitate anastomosis formation. Subsequently, the new capillaries maturate, and M2 macrophages remove redundant vessels via phagocytosis [235]. By imitating the physiological process of Angiogenesis, researchers aim to endorse vascularization in tissue engineering scaffolds [240]. In this context, Spiller et al. analyzed the role of macrophage phenotype in the vascularization of scaffolds. They found that M1 and M2c macrophages cause endothelial sprouting and M2a macrophages supported anastomoses. The researchers could control macrophage response by modifying scaffold properties [241]. Another recent study investigated whether reprogramming macrophages with KGM-modified SiO2 nanoparticles influences diabetic wound healing. The researchers demonstrated that an M2-like phenotype was linked to angiogenesis, enhanced ECM production, and accelerated wound healing by repressing extensive or persistent inflammation and fibrosis [242]. Targeting angiogenesis in a macrophage-centered treatment approach, therefore, represents a promising target in tissue engineering.
In conclusion, the differentiation of cells of the monocyte-macrophage lineage into M1 and M2 subpopulations is of major relevance for biomaterial applications in tissue engineering. M1 macrophages are mainly active in the immune system during inflammation. Excessive or prolonged M1 macrophage activation could result in “tissue injury” and thus negatively affect the clinical course of a tissue-engineered implant. On the other hand, M2 macrophages are important for the resolution of inflammation due to their ability to produce anti-inflammatory cytokines. They are also important cells for homeostasis and tissue regeneration. Being able to find the optimal balance between these subpopulations remains a prime challenge in regenerative medicine but holds great promise for the future.
Taking all these findings together, which demonstrate the broad spectrum of macrophage functions, it becomes clear that these cells should be essentially involved in biomaterial- and tissue-engineered-based strategies, and their specific role should be taken into account. Such considerations could open new pathways to modulate the plasticity of macrophages in various tissue engineering approaches.
Metchnikow described macrophages for the first time. They were supposed to phagocytize foreign bodies and bacteria. Now, a hundred years after his death, macrophages are still in trend with a variety of functions and subpopulations. It is our duty to continue to examine them so that we can understand and explain different pathological processes so that we can apply their functions in vitro and in vivo research fields regarding tissue regeneration and engineering, but also in honor and memory of Metchnikow.

Author Contributions

Conceptualization, A.M., V.H.S., F.B., W.W. and C.B.; Literature request, A.M., L.W., C.S., M.B., S.S., M.R., V.H.S., W.W.; investigation, C.B., V.A., K.K.; writing—original draft preparation, A.M., L.W., M.B., F.B., D.G.; writing—review and editing, C.B., A.M., M.B., L.W., V.H.S., T.N., F.G., F.B., W.W., D.G., K.K., C.S., S.S., M.R., V.A.; visualization, A.M., L.W., F.B.; supervision, C.B.; project administration, C.B., T.N., F.G.; funding acquisition, C.B., T.N., F.G., M.R. All authors have read and agreed to the published version of the manuscript.

Funding

This work was funded by a grant of the European Union, Ziel ETZ-352 2014–2020, (Ref.: Interreg 289).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

List of Abbreviations

A2AR: adenosine A2A receptors; AAM: alternatively activated macrophages; CAM: Classically activated macrophages; CTL: cytotoxic T-lymphocytes; ECM: extracellular matrix; EMP: Erythro-Myeloid Progenitors; FBR: foreign body reaction; GAS: gamma-activated sequences; GC: glucocorticoids; HMGB-1: High-mobility group box 1; HSC: Hematopoietic Stem Cells; HSPs: heat-shock proteins; iNOS: inducible nitric oxide synthase; LIF: leukemia inhibitory factor; LPS: bacterial lipopolysaccharide; MAF: macrophage-activating factor; MMPs: matrix-metalloproteinases; MNGC: multinucleated giant cell; NK: natural killer cells; NKT: natural killer T-lymphocytes; NO: nitric oxide; NP. Nanoparticles; PAMPs: pathogen-associated molecular patterns; PPR: pattern recognition receptors; PU NPs: polyurethane nanoparticles; TAM: tumor-associated macrophages; TCA: tricarboxylic acid cycle; TGFβ-1: transforming growth factor β 1; Th17: T-helper cell type 17; Ti: Titanium; TME: tumor microenvironment; TLRs: Toll-like receptors; TNF-α: tumor necrosis factor-alpha.

References

  1. Gordon, S. Elie Metchnikoff, the Man and the Myth. J. Innate Immun. 2016, 8, 223–227. [Google Scholar] [CrossRef] [PubMed]
  2. Xia, Z.; Triffitt, J.T. A review on macrophage responses to biomaterials. Biomed. Mater. 2006, 1, R1–R9. [Google Scholar] [CrossRef] [PubMed]
  3. Erwig, L.P.; Henson, P.M. Clearance of apoptotic cells by phagocytes. Cell Death Differ. 2008, 15, 243–250. [Google Scholar] [CrossRef] [PubMed]
  4. Flannagan, R.S.; Jaumouillé, V.; Grinstein, S. The cell biology of phagocytosis. Annu. Rev. Pathol. 2012, 7, 61–98. [Google Scholar] [CrossRef] [PubMed]
  5. Braga, T.T.; Agudelo, J.S.; Camara, N.O. Macrophages During the Fibrotic Process: M2 as Friend and Foe. Front. Immunol. 2015, 6, 602. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Yona, S.; Gordon, S. From the Reticuloendothelial to Mononuclear Phagocyte System—The Unaccounted Years. Front. Immunol. 2015, 6, 328. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Mosser, D.M. The many faces of macrophage activation. J. Leukoc. Biol. 2003, 73, 209–212. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Shapouri-Moghaddam, A.; Mohammadian, S.; Vazini, H.; Taghadosi, M.; Esmaeili, S.A.; Mardani, F.; Seifi, B.; Mohammadi, A.; Afshari, J.T.; Sahebkar, A. Macrophage plasticity, polarization, and function in health and disease. J. Cell. Physiol. 2018, 233, 6425–6440. [Google Scholar] [CrossRef]
  9. Schreml, S.; Szeimies, R.M.; Prantl, L.; Landthaler, M.; Babilas, P. Wound healing in the 21st century. J. Am. Acad. Dermatol. 2010, 63, 866–881. [Google Scholar] [CrossRef]
  10. Geissmann, F.; Manz, M.G.; Jung, S.; Sieweke, M.H.; Merad, M.; Ley, K. Development of monocytes, macrophages, and dendritic cells. Science 2010, 327, 656–661. [Google Scholar] [CrossRef]
  11. Aderem, A.A.; Cohen, D.S.; Wright, S.D.; Cohn, Z.A. Bacterial lipopolysaccharides prime macrophages for enhanced release of arachidonic acid metabolites. J. Exp. Med. 1986, 164, 165–179. [Google Scholar] [CrossRef]
  12. Varin, A.; Gordon, S. Alternative activation of macrophages: Immune function and cellular biology. Immunobiology 2009, 214, 630–641. [Google Scholar] [CrossRef]
  13. Oishi, Y.; Manabe, I. Macrophages in inflammation, repair and regeneration. Int. Immunol. 2018, 30, 511–528. [Google Scholar] [CrossRef] [Green Version]
  14. Martinez, F.O.; Helming, L.; Gordon, S. Alternative activation of macrophages: An immunologic functional perspective. Annu. Rev. Immunol. 2009, 27, 451–483. [Google Scholar] [CrossRef] [Green Version]
  15. Gordon, S. The macrophage: Past, present and future. Eur. J. Immunol. 2007, 37 (Suppl. 1), S9–S17. [Google Scholar] [CrossRef]
  16. Zhang, X.; Mosser, D.M. Macrophage activation by endogenous danger signals. J. Pathol. 2008, 214, 161–178. [Google Scholar] [CrossRef]
  17. Tacke, F. Monocyte subpopulations in inflammation processes: Principles and perspectives. Dtsch. Med. Wochenschr. 2009, 134, 1645–1648. [Google Scholar] [CrossRef]
  18. Shizuru, J.A.; Negrin, R.S.; Weissman, I.L. Hematopoietic stem and progenitor cells: Clinical and preclinical regeneration of the hematolymphoid system. Annu. Rev. Med. 2005, 56, 509–538. [Google Scholar] [CrossRef] [Green Version]
  19. Kondo, M.; Wagers, A.J.; Manz, M.G.; Prohaska, S.S.; Scherer, D.C.; Beilhack, G.F.; Shizuru, J.A.; Weissman, I.L. Biology of hematopoietic stem cells and progenitors: Implications for clinical application. Annu. Rev. Immunol. 2003, 21, 759–806. [Google Scholar] [CrossRef]
  20. Mosser, D.M.; Edwards, J.P. Exploring the full spectrum of macrophage activation. Nat. Rev. Immunol. 2008, 8, 958–969. [Google Scholar] [CrossRef]
  21. Auffray, C.; Sieweke, M.H.; Geissmann, F. Blood monocytes: Development, heterogeneity, and relationship with dendritic cells. Annu. Rev. Immunol. 2009, 27, 669–692. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Swirski, F.K.; Nahrendorf, M.; Etzrodt, M.; Wildgruber, M.; Cortez-Retamozo, V.; Panizzi, P.; Figueiredo, J.L.; Kohler, R.H.; Chudnovskiy, A.; Waterman, P.; et al. Identification of splenic reservoir monocytes and their deployment to inflammatory sites. Science 2009, 325, 612–616. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Randolph, G.J. No need to coax monocytes. Although inflammation has been thought to rely on recruitment of macrophages from the blood, tissue macrophages can proliferate for an inflammatory response. Science 2011, 332, 1268–1269. [Google Scholar] [CrossRef] [PubMed]
  24. Shi, C.; Pamer, E.G. Monocyte recruitment during infection and inflammation. Nat. Rev. Immunol. 2011, 11, 762–774. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Hoeffel, G.; Ginhoux, F. Fetal monocytes and the origins of tissue-resident macrophages. Cell. Immunol. 2018, 330, 5–15. [Google Scholar] [CrossRef]
  26. Lamb, B.W.; Green, J.S.; Benn, J.; Brown, K.F.; Vincent, C.A.; Sevdalis, N. Improving decision making in multidisciplinary tumor boards: Prospective longitudinal evaluation of a multicomponent intervention for 1421 patients. J. Am. Coll. Surg. 2013, 217, 412–420. [Google Scholar] [CrossRef]
  27. Yoder, M.C. Inducing definitive hematopoiesis in a dish. Nat. Biotechnol. 2014, 32, 539–541. [Google Scholar] [CrossRef]
  28. Wu, Y.; Hirschi, K.K. Tissue-Resident Macrophage Development and Function. Front. Cell Dev. Biol. 2020, 8, 617879. [Google Scholar] [CrossRef]
  29. Stout, R.D.; Jiang, C.; Matta, B.; Tietzel, I.; Watkins, S.K.; Suttles, J. Macrophages sequentially change their functional phenotype in response to changes in microenvironmental influences. J. Immunol. 2005, 175, 342–349. [Google Scholar] [CrossRef] [Green Version]
  30. Mantovani, A.; Sozzani, S.; Locati, M.; Allavena, P.; Sica, A. Macrophage polarization: Tumor-associated macrophages as a paradigm for polarized M2 mononuclear phagocytes. Trends Immunol. 2002, 23, 549–555. [Google Scholar] [CrossRef]
  31. Kou, P.M.; Babensee, J.E. Macrophage and dendritic cell phenotypic diversity in the context of biomaterials. J. Biomed. Mater. Res. A 2011, 96, 239–260. [Google Scholar] [CrossRef]
  32. Cassetta, L.; Cassol, E.; Poli, G. Macrophage polarization in health and disease. Sci. World J. 2011, 11, 213962. [Google Scholar] [CrossRef] [Green Version]
  33. Biswas, S.K.; Mantovani, A. Macrophage plasticity and interaction with lymphocyte subsets: Cancer as a paradigm. Nat. Immunol. 2010, 11, 889–896. [Google Scholar] [CrossRef]
  34. Luo, C.; Chen, M.; Madden, A.; Xu, H. Expression of complement components and regulators by different subtypes of bone marrow-derived macrophages. Inflammation 2012, 35, 1448–1461. [Google Scholar] [CrossRef]
  35. Zhang, Q.W.; Liu, L.; Gong, C.Y.; Shi, H.S.; Zeng, Y.H.; Wang, X.Z.; Zhao, Y.W.; Wei, Y.Q. Prognostic significance of tumor-associated macrophages in solid tumor: A meta-analysis of the literature. PLoS ONE 2012, 7, e50946. [Google Scholar] [CrossRef] [Green Version]
  36. Mantovani, A.; Bottazzi, B.; Colotta, F.; Sozzani, S.; Ruco, L. The origin and function of tumor-associated macrophages. Immunol. Today 1992, 13, 265–270. [Google Scholar] [CrossRef]
  37. Yang, L.; Zhang, Y. Tumor-associated macrophages: From basic research to clinical application. J. Hematol. Oncol. 2017, 10, 58. [Google Scholar] [CrossRef] [Green Version]
  38. Biswas, S.K.; Chittezhath, M.; Shalova, I.N.; Lim, J.Y. Macrophage polarization and plasticity in health and disease. Immunol. Res. 2012, 53, 11–24. [Google Scholar] [CrossRef]
  39. Murray, P.J. Macrophage Polarization. Annu. Rev. Physiol. 2017, 79, 541–566. [Google Scholar] [CrossRef]
  40. Laria, A.; Lurati, A.; Marrazza, M.; Mazzocchi, D.; Re, K.A.; Scarpellini, M. The macrophages in rheumatic diseases. J. Inflamm. Res. 2016, 9, 1–11. [Google Scholar] [CrossRef]
  41. Porcheray, F.; Viaud, S.; Rimaniol, A.C.; Leone, C.; Samah, B.; Dereuddre-Bosquet, N.; Dormont, D.; Gras, G. Macrophage activation switching: An asset for the resolution of inflammation. Clin. Exp. Immunol. 2005, 142, 481–489. [Google Scholar] [CrossRef] [PubMed]
  42. Sica, A.; Mantovani, A. Macrophage plasticity and polarization: In vivo veritas. J. Clin. Investig. 2012, 122, 787–795. [Google Scholar] [CrossRef] [PubMed]
  43. Saccani, A.; Schioppa, T.; Porta, C.; Biswas, S.K.; Nebuloni, M.; Vago, L.; Bottazzi, B.; Colombo, M.P.; Mantovani, A.; Sica, A. p50 nuclear factor-κB overexpression in tumor-associated macrophages inhibits M1 inflammatory responses and antitumor resistance. Cancer Res. 2006, 66, 11432–11440. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Guiducci, C.; Vicari, A.P.; Sangaletti, S.; Trinchieri, G.; Colombo, M.P. Redirecting in vivo elicited tumor infiltrating macrophages and dendritic cells towards tumor rejection. Cancer Res. 2005, 65, 3437–3446. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Noël, W.; Raes, G.; Hassanzadeh Ghassabeh, G.; De Baetselier, P.; Beschin, A. Alternatively activated macrophages during parasite infections. Trends Parasitol. 2004, 20, 126–133. [Google Scholar] [CrossRef]
  46. Babu, S.; Blauvelt, C.P.; Kumaraswami, V.; Nutman, T.B. Chemokine receptors of T cells and of B cells in lymphatic filarial infection: A role for CCR9 in pathogenesis. J. Infect. Dis. 2005, 191, 1018–1026. [Google Scholar] [CrossRef]
  47. Gratchev, A.; Guillot, P.; Hakiy, N.; Politz, O.; Orfanos, C.E.; Schledzewski, K.; Goerdt, S. Alternatively activated macrophages differentially express fibronectin and its splice variants and the extracellular matrix protein βIG-H3. Scand. J. Immunol. 2001, 53, 386–392. [Google Scholar] [CrossRef]
  48. Yunna, C.; Mengru, H.; Lei, W.; Weidong, C. Macrophage M1/M2 polarization. Eur. J. Pharmacol. 2020, 877, 173090. [Google Scholar] [CrossRef]
  49. Funes, S.C.; Rios, M.; Escobar-Vera, J.; Kalergis, A.M. Implications of macrophage polarization in autoimmunity. Immunology 2018, 154, 186–195. [Google Scholar] [CrossRef] [Green Version]
  50. Sindrilaru, A.; Peters, T.; Wieschalka, S.; Baican, C.; Baican, A.; Peter, H.; Hainzl, A.; Schatz, S.; Qi, Y.; Schlecht, A.; et al. An unrestrained proinflammatory M1 macrophage population induced by iron impairs wound healing in humans and mice. J. Clin. Investig. 2011, 121, 985–997. [Google Scholar] [CrossRef]
  51. Viola, A.; Munari, F.; Sánchez-Rodríguez, R.; Scolaro, T.; Castegna, A. The Metabolic Signature of Macrophage Responses. Front. Immunol. 2019, 10, 1462. [Google Scholar] [CrossRef] [Green Version]
  52. Rath, M.; Müller, I.; Kropf, P.; Closs, E.I.; Munder, M. Metabolism via Arginase or Nitric Oxide Synthase: Two Competing Arginine Pathways in Macrophages. Front. Immunol. 2014, 5, 532. [Google Scholar] [CrossRef] [Green Version]
  53. Biswas, S.K.; Mantovani, A. Orchestration of metabolism by macrophages. Cell Metab. 2012, 15, 432–437. [Google Scholar] [CrossRef] [Green Version]
  54. Zhu, L.; Zhao, Q.; Yang, T.; Ding, W.; Zhao, Y. Cellular metabolism and macrophage functional polarization. Int. Rev. Immunol. 2015, 34, 82–100. [Google Scholar] [CrossRef]
  55. Xia, Y.; Li, Y.; Wu, X.; Zhang, Q.; Chen, S.; Ma, X.; Yu, M. Ironing Out the Details: How Iron Orchestrates Macrophage Polarization. Front. Immunol. 2021, 12, 669566. [Google Scholar] [CrossRef]
  56. Martinez, F.O.; Sica, A.; Mantovani, A.; Locati, M. Macrophage activation and polarization. Front. Biosci. 2008, 13, 453–461. [Google Scholar] [CrossRef] [Green Version]
  57. Martinez, F.O. Regulators of macrophage activation. Eur. J. Immunol. 2011, 41, 1531–1534. [Google Scholar] [CrossRef]
  58. Kang, S.; Brown, H.M.; Hwang, S. Direct Antiviral Mechanisms of Interferon-γ. Immune Netw. 2018, 18, e33. [Google Scholar] [CrossRef]
  59. Dale, D.C.; Boxer, L.; Liles, W.C. The phagocytes: Neutrophils and monocytes. Blood 2008, 112, 935–945. [Google Scholar] [CrossRef]
  60. Mosser, D.M.; Zhang, X. Activation of murine macrophages. Curr. Protoc. Immunol. 2008, 83, 14.2.1–14.2.8. [Google Scholar] [CrossRef]
  61. Schoenborn, J.R.; Wilson, C.B. Regulation of interferon-γ during innate and adaptive immune responses. Adv. Immunol. 2007, 96, 41–101. [Google Scholar] [CrossRef] [PubMed]
  62. Gray, P.W.; Goeddel, D.V. Structure of the human immune interferon gene. Nature 1982, 298, 859–863. [Google Scholar] [CrossRef] [PubMed]
  63. Xie, C.; Liu, C.; Wu, B.; Lin, Y.; Ma, T.; Xiong, H.; Wang, Q.; Li, Z.; Ma, C.; Tu, Z. Effects of IRF1 and IFN-β interaction on the M1 polarization of macrophages and its antitumor function. Int. J. Mol. Med. 2016, 38, 148–160. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Klose, C.S.N.; Artis, D. Innate lymphoid cells control signaling circuits to regulate tissue-specific immunity. Cell Res. 2020, 30, 475–491. [Google Scholar] [CrossRef] [PubMed]
  65. Baeyens, K.J.; De Bondt, H.L.; Raeymaekers, A.; Fiers, W.; De Ranter, C.J. The structure of mouse tumour-necrosis factor at 1.4 A resolution: Towards modulation of its selectivity and trimerization. Acta Crystallogr. Sect. D Biol. Crystallogr. 1999, 55, 772–778. [Google Scholar] [CrossRef] [Green Version]
  66. Sherry, B.; Jue, D.M.; Zentella, A.; Cerami, A. Characterization of high molecular weight glycosylated forms of murine tumor necrosis factor. Biochem. Biophys. Res. Commun. 1990, 173, 1072–1078. [Google Scholar] [CrossRef]
  67. Fransen, L.; Muller, R.; Marmenout, A.; Tavernier, J.; Van der Heyden, J.; Kawashima, E.; Chollet, A.; Tizard, R.; Van Heuverswyn, H.; Van Vliet, A.; et al. Molecular cloning of mouse tumour necrosis factor cDNA and its eukaryotic expression. Nucleic Acids Res. 1985, 13, 4417–4429. [Google Scholar] [CrossRef] [Green Version]
  68. Cseh, K.; Beutler, B. Alternative cleavage of the cachectin/tumor necrosis factor propeptide results in a larger, inactive form of secreted protein. J. Biol. Chem. 1989, 264, 16256–16260. [Google Scholar] [CrossRef]
  69. Kalliolias, G.D.; Ivashkiv, L.B. TNF biology, pathogenic mechanisms and emerging therapeutic strategies. Nat. Rev. Rheumatol. 2016, 12, 49–62. [Google Scholar] [CrossRef]
  70. Mantovani, A.; Sica, A.; Sozzani, S.; Allavena, P.; Vecchi, A.; Locati, M. The chemokine system in diverse forms of macrophage activation and polarization. Trends Immunol. 2004, 25, 677–686. [Google Scholar] [CrossRef]
  71. Takeda, K.; Akira, S. Toll-like receptors in innate immunity. Int. Immunol. 2005, 17, 1–14. [Google Scholar] [CrossRef] [Green Version]
  72. Rock, F.L.; Hardiman, G.; Timans, J.C.; Kastelein, R.A.; Bazan, J.F. A family of human receptors structurally related to Drosophila Toll. Proc. Natl. Acad. Sci. USA 1998, 95, 588–593. [Google Scholar] [CrossRef] [Green Version]
  73. Turvey, S.E.; Broide, D.H. Innate immunity. J. Allergy Clin. Immunol. 2010, 125, S24–S32. [Google Scholar] [CrossRef]
  74. Ausubel, F.M. Are innate immune signaling pathways in plants and animals conserved? Nat. Immunol. 2005, 6, 973–979. [Google Scholar] [CrossRef]
  75. Didierlaurent, A.; Simonet, M.; Sirard, J.C. Innate and acquired plasticity of the intestinal immune system. Cell. Mol. Life Sci. 2005, 62, 1285–1287. [Google Scholar] [CrossRef] [Green Version]
  76. Rumbo, M.; Nempont, C.; Kraehenbuhl, J.-P.; Sirard, J.-C. Mucosal interplay among commensal and pathogenic bacteria: Lessons from flagellin and Toll-like receptor 5. FEBS Lett. 2006, 580, 2976–2984. [Google Scholar] [CrossRef]
  77. Hamidzadeh, K.; Mosser, D.M. Purinergic Signaling to Terminate TLR Responses in Macrophages. Front. Immunol. 2016, 7, 74. [Google Scholar] [CrossRef] [Green Version]
  78. Yamamoto, M.; Sato, S.; Hemmi, H.; Hoshino, K.; Kaisho, T.; Sanjo, H.; Takeuchi, O.; Sugiyama, M.; Okabe, M.; Takeda, K.; et al. Role of adaptor TRIF in the MyD88-independent toll-like receptor signaling pathway. Science 2003, 301, 640–643. [Google Scholar] [CrossRef]
  79. Gallucci, S.; Matzinger, P. Danger signals: SOS to the immune system. Curr. Opin. Immunol. 2001, 13, 114–119. [Google Scholar] [CrossRef]
  80. Yu, L.; Wang, L.; Chen, S. Endogenous toll-like receptor ligands and their biological significance. J. Cell. Mol. Med. 2010, 14, 2592–2603. [Google Scholar] [CrossRef]
  81. El-Zayat, S.R.; Sibaii, H.; Mannaa, F.A. Toll-like receptors activation, signaling, and targeting: An overview. Bull. Natl. Res. Cent. 2019, 43, 187. [Google Scholar] [CrossRef] [Green Version]
  82. Costa, T.E.M.M.; Raghavendra, N.M.; Penido, C. Natural heat shock protein 90 inhibitors in cancer and inflammation. Eur. J. Med. Chem. 2020, 189, 112063. [Google Scholar] [CrossRef]
  83. Vostakolaei, M.A.; Abdolalizadeh, J.; Hejazi, M.S.; Kordi, S.; Molavi, O. Hsp70 in Cancer: Partner or Traitor to Immune System. Iran. J. Allergy Asthma Immunol. 2019, 18, 589–604. [Google Scholar] [CrossRef]
  84. Bolhassani, A.; Agi, E. Heat shock proteins in infection. Clin. Chim. Acta 2019, 498, 90–100. [Google Scholar] [CrossRef]
  85. Kaushik, S.; Cuervo, A.M. Chaperones in autophagy. Pharmacol. Res. 2012, 66, 484–493. [Google Scholar] [CrossRef] [Green Version]
  86. Young, J.C.; Agashe, V.R.; Siegers, K.; Hartl, F.U. Pathways of chaperone-mediated protein folding in the cytosol. Nat. Rev. Mol. Cell Biol. 2004, 5, 781–791. [Google Scholar] [CrossRef]
  87. Buchner, J. Molecular chaperones and protein quality control: An introduction to the JBC Reviews thematic series. J. Biol. Chem. 2019, 294, 2074–2075. [Google Scholar] [CrossRef] [Green Version]
  88. Basu, S.; Binder, R.J.; Suto, R.; Anderson, K.M.; Srivastava, P.K. Necrotic but not apoptotic cell death releases heat shock proteins, which deliver a partial maturation signal to dendritic cells and activate the NF-κB pathway. Int. Immunol. 2000, 12, 1539–1546. [Google Scholar] [CrossRef] [Green Version]
  89. Lindquist, S.; Craig, E.A. The heat-shock proteins. Annu. Rev. Genet. 1988, 22, 631–677. [Google Scholar] [CrossRef]
  90. Calapre, L.; Gray, E.S.; Ziman, M. Heat stress: A risk factor for skin carcinogenesis. Cancer Lett. 2013, 337, 35–40. [Google Scholar] [CrossRef]
  91. Trial, J.; Rubio, J.A.; Birdsall, H.H.; Rodriguez-Barradas, M.; Rossen, R.D. Monocyte activation by circulating fibronectin fragments in HIV-1-infected patients. J. Immunol. 2004, 173, 2190–2198. [Google Scholar] [CrossRef] [Green Version]
  92. Wright, S.D.; Meyer, B.C. Fibronectin receptor of human macrophages recognizes the sequence Arg-Gly-Asp-Ser. J. Exp. Med. 1985, 162, 762–767. [Google Scholar] [CrossRef] [Green Version]
  93. Digiacomo, G.; Tusa, I.; Bacci, M.; Cipolleschi, M.G.; Dello Sbarba, P.; Rovida, E. Fibronectin induces macrophage migration through a SFK-FAK/CSF-1R pathway. Cell Adhes. Migr. 2017, 11, 327–337. [Google Scholar] [CrossRef] [Green Version]
  94. Jiang, D.; Liang, J.; Noble, P.W. Hyaluronan in tissue injury and repair. Annu. Rev. Cell Dev. Biol. 2007, 23, 435–461. [Google Scholar] [CrossRef] [Green Version]
  95. Jiang, D.; Liang, J.; Fan, J.; Yu, S.; Chen, S.; Luo, Y.; Prestwich, G.D.; Mascarenhas, M.M.; Garg, H.G.; Quinn, D.A.; et al. Regulation of lung injury and repair by Toll-like receptors and hyaluronan. Nat. Med. 2005, 11, 1173–1179. [Google Scholar] [CrossRef]
  96. Mummert, M.E. Immunologic roles of hyaluronan. Immunol. Res. 2005, 31, 189–206. [Google Scholar] [CrossRef]
  97. Cantor, J.O.; Nadkarni, P.P. Hyaluronan: The Jekyll and Hyde molecule. Inflamm. Allergy Drug Targets 2006, 5, 257–260. [Google Scholar] [CrossRef]
  98. Stern, R.; Asari, A.A.; Sugahara, K.N. Hyaluronan fragments: An information-rich system. Eur. J. Cell Biol. 2006, 85, 699–715. [Google Scholar] [CrossRef]
  99. Avenoso, A.; Bruschetta, G.; D’Ascola, A.; Scuruchi, M.; Mandraffino, G.; Saitta, A.; Campo, S.; Campo, G.M. Hyaluronan Fragmentation During Inflammatory Pathologies: A Signal that Empowers Tissue Damage. Mini-Rev. Med. Chem. 2020, 20, 54–65. [Google Scholar] [CrossRef]
  100. Ulloa, L.; Messmer, D. High-mobility group box 1 (HMGB1) protein: Friend and foe. Cytokine Growth Factor Rev. 2006, 17, 189–201. [Google Scholar] [CrossRef]
  101. Lotze, M.T.; Tracey, K.J. High-mobility group box 1 protein (HMGB1): Nuclear weapon in the immune arsenal. Nat. Rev. Immunol. 2005, 5, 331–342. [Google Scholar] [CrossRef] [PubMed]
  102. Erlandsson Harris, H.; Andersson, U. Mini-review: The nuclear protein HMGB1 as a proinflammatory mediator. Eur. J. Immunol. 2004, 34, 1503–1512. [Google Scholar] [CrossRef] [PubMed]
  103. Harris, H.E.; Raucci, A. Alarmin(g) news about danger: Workshop on innate danger signals and HMGB1. EMBO Rep. 2006, 7, 774–778. [Google Scholar] [CrossRef] [Green Version]
  104. Klune, J.R.; Dhupar, R.; Cardinal, J.; Billiar, T.R.; Tsung, A. HMGB1: Endogenous danger signaling. Mol. Med. 2008, 14, 476–484. [Google Scholar] [CrossRef]
  105. Liu, T.; Xiang, A.; Peng, T.; Doran, A.C.; Tracey, K.J.; Barnes, B.J.; Tabas, I.; Son, M.; Diamond, B. HMGB1-C1q complexes regulate macrophage function by switching between leukotriene and specialized proresolving mediator biosynthesis. Proc. Natl. Acad. Sci. USA 2019, 116, 23254–23263. [Google Scholar] [CrossRef]
  106. Albina, J.E.; Henry, W.L., Jr.; Mastrofrancesco, B.; Martin, B.A.; Reichner, J.S. Macrophage activation by culture in an anoxic environment. J. Immunol. 1995, 155, 4391–4396. [Google Scholar] [CrossRef]
  107. Nickel, D.; Busch, M.; Mayer, D.; Hagemann, B.; Knoll, V.; Stenger, S. Hypoxia Triggers the Expression of Human β Defensin 2 and Antimicrobial Activity against Mycobacterium tuberculosis in Human Macrophages. J. Immunol. 2012, 188, 4001–4007. [Google Scholar] [CrossRef] [Green Version]
  108. Murdoch, C.; Muthana, M.; Lewis, C.E. Hypoxia regulates macrophage functions in inflammation. J. Immunol. 2005, 175, 6257–6263. [Google Scholar] [CrossRef] [Green Version]
  109. Grimshaw, M.J.; Balkwill, F.R. Inhibition of monocyte and macrophage chemotaxis by hypoxia and inflammation—A potential mechanism. Eur. J. Immunol. 2001, 31, 480–489. [Google Scholar] [CrossRef]
  110. Mosser, D.M.; Hamidzadeh, K.; Goncalves, R. Macrophages and the maintenance of homeostasis. Cell. Mol. Immunol. 2021, 18, 579–587. [Google Scholar] [CrossRef]
  111. Aderem, A. Phagocytosis and the inflammatory response. J. Infect. Dis. 2003, 187 (Suppl. S2), S340–S345. [Google Scholar] [CrossRef]
  112. Aderem, A.; Underhill, D.M. Mechanisms of phagocytosis in macrophages. Annu. Rev. Immunol. 1999, 17, 593–623. [Google Scholar] [CrossRef]
  113. Galli, S.J.; Borregaard, N.; Wynn, T.A. Phenotypic and functional plasticity of cells of innate immunity: Macrophages, mast cells and neutrophils. Nat. Immunol. 2011, 12, 1035–1044. [Google Scholar] [CrossRef] [Green Version]
  114. Metschnikoff, E. Ueber die Beziehung der Phagocyten zu Milzbrandbacillen. Virchows Arch. 1884, 97, 502–526. [Google Scholar] [CrossRef]
  115. Metschnikoff, E. Lecture on Phagocytosis and Immunity. Br. Med. J. 1891, 1, 213–217. [Google Scholar] [CrossRef] [Green Version]
  116. MacMicking, J.; Xie, Q.W.; Nathan, C. Nitric oxide and macrophage function. Annu. Rev. Immunol. 1997, 15, 323–350. [Google Scholar] [CrossRef]
  117. Raupach, B.; Kaufmann, S.H. Immune responses to intracellular bacteria. Curr. Opin. Immunol. 2001, 13, 417–428. [Google Scholar] [CrossRef]
  118. Kamijo, R.; Harada, H.; Matsuyama, T.; Bosland, M.; Gerecitano, J.; Shapiro, D.; Le, J.; Koh, S.I.; Kimura, T.; Green, S.J.; et al. Requirement for transcription factor IRF-1 in NO synthase induction in macrophages. Science 1994, 263, 1612–1615. [Google Scholar] [CrossRef]
  119. Abu-Amara, M.; Yang, S.Y.; Seifalian, A.; Davidson, B.; Fuller, B. The nitric oxide pathway—Evidence and mechanisms for protection against liver ischaemia reperfusion injury. Liver Int. 2012, 32, 531–543. [Google Scholar] [CrossRef]
  120. Ricciardolo, F.L.; Nijkamp, F.P.; Folkerts, G. Nitric oxide synthase (NOS) as therapeutic target for asthma and chronic obstructive pulmonary disease. Curr. Drug Targets 2006, 7, 721–735. [Google Scholar] [CrossRef]
  121. Bartosz, G. Peroxynitrite: Mediator of the toxic action of nitric oxide. Acta Biochim. Pol. 1996, 43, 645–659. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Pacher, P.; Beckman, J.S.; Liaudet, L. Nitric oxide and peroxynitrite in health and disease. Physiol. Rev. 2007, 87, 315–424. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Fieren, M.W. The local inflammatory responses to infection of the peritoneal cavity in humans: Their regulation by cytokines, macrophages, and other leukocytes. Mediat. Inflamm. 2012, 2012, 976241. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Wang, J.; Nikrad, M.P.; Travanty, E.A.; Zhou, B.; Phang, T.; Gao, B.; Alford, T.; Ito, Y.; Nahreini, P.; Hartshorn, K.; et al. Innate immune response of human alveolar macrophages during influenza A infection. PLoS ONE 2012, 7, e29879. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Verreck, F.A.; de Boer, T.; Langenberg, D.M.; Hoeve, M.A.; Kramer, M.; Vaisberg, E.; Kastelein, R.; Kolk, A.; de Waal-Malefyt, R.; Ottenhoff, T.H. Human IL-23-producing type 1 macrophages promote but IL-10-producing type 2 macrophages subvert immunity to (myco)bacteria. Proc. Natl. Acad. Sci. USA 2004, 101, 4560–4565. [Google Scholar] [CrossRef] [Green Version]
  126. Baay, M.; Brouwer, A.; Pauwels, P.; Peeters, M.; Lardon, F. Tumor cells and tumor-associated macrophages: Secreted proteins as potential targets for therapy. Clin. Dev. Immunol. 2011, 2011, 565187. [Google Scholar] [CrossRef] [Green Version]
  127. Xue, Q.; Lu, Y.; Eisele, M.R.; Sulistijo, E.S.; Khan, N.; Fan, R.; Miller-Jensen, K. Analysis of single-cell cytokine secretion reveals a role for paracrine signaling in coordinating macrophage responses to TLR4 stimulation. Sci. Signal. 2015, 8, ra59. [Google Scholar] [CrossRef] [Green Version]
  128. Hou, Y.; Zhu, L.; Tian, H.; Sun, H.X.; Wang, R.; Zhang, L.; Zhao, Y. IL-23-induced macrophage polarization and its pathological roles in mice with imiquimod-induced psoriasis. Protein Cell 2018, 9, 1027–1038. [Google Scholar] [CrossRef]
  129. Sutton, C.E.; Lalor, S.J.; Sweeney, C.M.; Brereton, C.F.; Lavelle, E.C.; Mills, K.H. Interleukin-1 and IL-23 induce innate IL-17 production from γδ T cells, amplifying Th17 responses and autoimmunity. Immunity 2009, 31, 331–341. [Google Scholar] [CrossRef] [Green Version]
  130. Sutton, C.; Brereton, C.; Keogh, B.; Mills, K.H.; Lavelle, E.C. A crucial role for interleukin (IL)-1 in the induction of IL-17-producing T cells that mediate autoimmune encephalomyelitis. J. Exp. Med. 2006, 203, 1685–1691. [Google Scholar] [CrossRef]
  131. Zielinski, C.E.; Mele, F.; Aschenbrenner, D.; Jarrossay, D.; Ronchi, F.; Gattorno, M.; Monticelli, S.; Lanzavecchia, A.; Sallusto, F. Pathogen-induced human T(H)17 cells produce IFN-γ or IL-10 and are regulated by IL-1β. Nature 2012, 484, 514–518. [Google Scholar] [CrossRef]
  132. Mufazalov, I.A.; Schelmbauer, C.; Regen, T.; Kuschmann, J.; Wanke, F.; Gabriel, L.A.; Hauptmann, J.; Müller, W.; Pinteaux, E.; Kurschus, F.C.; et al. IL-1 signaling is critical for expansion but not generation of autoreactive GM-CSF+ Th17 cells. EMBO J. 2017, 36, 102–115. [Google Scholar] [CrossRef] [Green Version]
  133. Duvallet, E.; Semerano, L.; Assier, E.; Falgarone, G.; Boissier, M.C. Interleukin-23: A key cytokine in inflammatory diseases. Ann. Med. 2011, 43, 503–511. [Google Scholar] [CrossRef]
  134. Dong, W.; Zhu, P. Functional niche of inflamed synovium for Th17-cell expansion and activation in rheumatoid arthritis: Implication to clinical therapeutics. Autoimmun. Rev. 2012, 11, 844–851. [Google Scholar] [CrossRef]
  135. Kolls, J.K.; Linden, A. Interleukin-17 family members and inflammation. Immunity 2004, 21, 467–476. [Google Scholar] [CrossRef] [Green Version]
  136. Waite, J.C.; Skokos, D. Th17 response and inflammatory autoimmune diseases. Int. J. Inflamm. 2012, 2012, 819467. [Google Scholar] [CrossRef] [Green Version]
  137. Hemdan, N.Y.; Birkenmeier, G.; Wichmann, G.; Abu El-Saad, A.M.; Krieger, T.; Conrad, K.; Sack, U. Interleukin-17-producing T helper cells in autoimmunity. Autoimmun. Rev. 2010, 9, 785–792. [Google Scholar] [CrossRef]
  138. Tesmer, L.A.; Lundy, S.K.; Sarkar, S.; Fox, D.A. Th17 cells in human disease. Immunol. Rev. 2008, 223, 87–113. [Google Scholar] [CrossRef]
  139. McGinley, A.M.; Sutton, C.E.; Edwards, S.C.; Leane, C.M.; DeCourcey, J.; Teijeiro, A.; Hamilton, J.A.; Boon, L.; Djouder, N.; Mills, K.H.G. Interleukin-17A Serves a Priming Role in Autoimmunity by Recruiting IL-1β-Producing Myeloid Cells that Promote Pathogenic T Cells. Immunity 2020, 52, 342–356.e6. [Google Scholar] [CrossRef]
  140. Fingleton, B. Matrix metalloproteinases as regulators of inflammatory processes. Biochim. Biophys. Acta Mol. Cell Res. 2017, 1864, 2036–2042. [Google Scholar] [CrossRef]
  141. Van Lint, P.; Libert, C. Chemokine and cytokine processing by matrix metalloproteinases and its effect on leukocyte migration and inflammation. J. Leukoc. Biol. 2007, 82, 1375–1381. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Nénan, S.; Boichot, E.; Lagente, V.; Bertrand, C.P. Macrophage elastase (MMP-12): A pro-inflammatory mediator? Mem. Inst. Oswaldo Cruz 2005, 100 (Suppl. S1), 167–172. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Lagente, V.; Le Quement, C.; Boichot, E. Macrophage metalloelastase (MMP-12) as a target for inflammatory respiratory diseases. Expert Opin. Ther. Targets 2009, 13, 287–295. [Google Scholar] [CrossRef] [PubMed]
  144. Pierce, R.A.; Sandefur, S.; Doyle, G.A.; Welgus, H.G. Monocytic cell type-specific transcriptional induction of collagenase. J. Clin. Investig. 1996, 97, 1890–1899. [Google Scholar] [CrossRef] [PubMed]
  145. McCawley, L.J.; Matrisian, L.M. Matrix metalloproteinases: They’re not just for matrix anymore! Curr. Opin. Cell Biol. 2001, 13, 534–540. [Google Scholar] [CrossRef]
  146. Wen, G.; Zhang, C.; Chen, Q.; Luong, L.A.; Mustafa, A.; Ye, S.; Xiao, Q. A Novel Role of Matrix Metalloproteinase-8 in Macrophage Differentiation and Polarization. J. Biol. Chem. 2015, 290, 19158–19172. [Google Scholar] [CrossRef] [Green Version]
  147. Chen, J.; Huang, Z.B.; Liao, C.J.; Hu, X.W.; Li, S.L.; Qi, M.; Fan, X.G.; Huang, Y. LncRNA TP73-AS1/miR-539/MMP-8 axis modulates M2 macrophage polarization in hepatocellular carcinoma via TGF-β1 signaling. Cell Signal. 2020, 75, 109738. [Google Scholar] [CrossRef]
  148. Gordon, S. Alternative activation of macrophages. Nat. Rev. Immunol. 2003, 3, 23–35. [Google Scholar] [CrossRef]
  149. Barberà-Cremades, M.; Baroja-Mazo, A.; Pelegrín, P. Purinergic signaling during macrophage differentiation results in M2 alternative activated macrophages. J. Leukoc. Biol. 2016, 99, 289–299. [Google Scholar] [CrossRef] [Green Version]
  150. Duan, M.; Li, W.C.; Vlahos, R.; Maxwell, M.J.; Anderson, G.P.; Hibbs, M.L. Distinct macrophage subpopulations characterize acute infection and chronic inflammatory lung disease. J. Immunol. 2012, 189, 946–955. [Google Scholar] [CrossRef]
  151. Snyder, R.J.; Lantis, J.; Kirsner, R.S.; Shah, V.; Molyneaux, M.; Carter, M.J. Macrophages: A review of their role in wound healing and their therapeutic use. Wound Repair Regen. 2016, 24, 613–629. [Google Scholar] [CrossRef]
  152. Das, A.; Sinha, M.; Datta, S.; Abas, M.; Chaffee, S.; Sen, C.K.; Roy, S. Monocyte and macrophage plasticity in tissue repair and regeneration. Am. J. Pathol. 2015, 185, 2596–2606. [Google Scholar] [CrossRef] [Green Version]
  153. Wang, L.X.; Zhang, S.X.; Wu, H.J.; Rong, X.L.; Guo, J. M2b macrophage polarization and its roles in diseases. J. Leukoc. Biol. 2019, 106, 345–358. [Google Scholar] [CrossRef] [Green Version]
  154. Schwarz, M.; Rottgen, P.; Takada, Y.; Le Gall, F.; Knackmuss, S.; Bassler, N.; Buttner, C.; Little, M.; Bode, C.; Peter, K. Single-chain antibodies for the conformation-specific blockade of activated platelet integrin αIIbβ3 designed by subtractive selection from naive human phage libraries. FASEB J. 2004, 18, 1704–1706. [Google Scholar] [CrossRef] [Green Version]
  155. Tang, X. Tumor-associated macrophages as potential diagnostic and prognostic biomarkers in breast cancer. Cancer Lett. 2013, 332, 3–10. [Google Scholar] [CrossRef]
  156. Mantovani, A.; Biswas, S.K.; Galdiero, M.R.; Sica, A.; Locati, M. Macrophage plasticity and polarization in tissue repair and remodelling. J. Pathol. 2013, 229, 176–185. [Google Scholar] [CrossRef]
  157. Tang, X.; Mo, C.; Wang, Y.; Wei, D.; Xiao, H. Anti-tumour strategies aiming to target tumour-associated macrophages. Immunology 2013, 138, 93–104. [Google Scholar] [CrossRef] [Green Version]
  158. Mosser, D.M.; Zhang, X. Interleukin-10: New perspectives on an old cytokine. Immunol. Rev. 2008, 226, 205–218. [Google Scholar] [CrossRef]
  159. Kreider, T.; Anthony, R.M.; Urban, J.F., Jr.; Gause, W.C. Alternatively activated macrophages in helminth infections. Curr. Opin. Immunol. 2007, 19, 448–453. [Google Scholar] [CrossRef] [Green Version]
  160. Laskin, D.L.; Sunil, V.R.; Gardner, C.R.; Laskin, J.D. Macrophages and tissue injury: Agents of defense or destruction? Annu. Rev. Pharmacol. Toxicol. 2011, 51, 267–288. [Google Scholar] [CrossRef]
  161. Graff, J.W.; Dickson, A.M.; Clay, G.; McCaffrey, A.P.; Wilson, M.E. Identifying functional microRNAs in macrophages with polarized phenotypes. J. Biol. Chem. 2012, 287, 21816–21825. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. Chinetti-Gbaguidi, G.; Staels, B. Macrophage polarization in metabolic disorders: Functions and regulation. Curr. Opin. Lipidol. 2011, 22, 365–372. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Lolmede, K.; Campana, L.; Vezzoli, M.; Bosurgi, L.; Tonlorenzi, R.; Clementi, E.; Bianchi, M.E.; Cossu, G.; Manfredi, A.A.; Brunelli, S.; et al. Inflammatory and alternatively activated human macrophages attract vessel-associated stem cells, relying on separate HMGB1- and MMP-9-dependent pathways. J. Leukoc. Biol. 2009, 85, 779–787. [Google Scholar] [CrossRef] [PubMed]
  164. Munitz, A.; Brandt, E.B.; Mingler, M.; Finkelman, F.D.; Rothenberg, M.E. Distinct roles for IL-13 and IL-4 via IL-13 receptor α1 and the type II IL-4 receptor in asthma pathogenesis. Proc. Natl. Acad. Sci. USA 2008, 105, 7240–7245. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Witherel, C.E.; Graney, P.L.; Freytes, D.O.; Weingarten, M.S.; Spiller, K.L. Response of human macrophages to wound matrices in vitro. Wound Repair Regen. 2016, 24, 514–524. [Google Scholar] [CrossRef]
  166. Junttila, I.S. Tuning the Cytokine Responses: An Update on Interleukin (IL)-4 and IL-13 Receptor Complexes. Front. Immunol. 2018, 9, 888. [Google Scholar] [CrossRef] [Green Version]
  167. Anders, H.J.; Ryu, M. Renal microenvironments and macrophage phenotypes determine progression or resolution of renal inflammation and fibrosis. Kidney Int. 2011, 80, 915–925. [Google Scholar] [CrossRef] [Green Version]
  168. Dinarello, C.A. Interleukin-1 and interleukin-1 antagonism. Blood 1991, 77, 1627–1652. [Google Scholar] [CrossRef] [Green Version]
  169. Modolell, M.; Corraliza, I.M.; Link, F.; Soler, G.; Eichmann, K. Reciprocal regulation of the nitric oxide synthase/arginase balance in mouse bone marrow-derived macrophages by TH1 and TH2 cytokines. Eur. J. Immunol. 1995, 25, 1101–1104. [Google Scholar] [CrossRef]
  170. Edwards, J.P.; Zhang, X.; Frauwirth, K.A.; Mosser, D.M. Biochemical and functional characterization of three activated macrophage populations. J. Leukoc. Biol. 2006, 80, 1298–1307. [Google Scholar] [CrossRef]
  171. Rantakari, P.; Patten, D.A.; Valtonen, J.; Karikoski, M.; Gerke, H.; Dawes, H.; Laurila, J.; Ohlmeier, S.; Elima, K.; Hübscher, S.G.; et al. Stabilin-1 expression defines a subset of macrophages that mediate tissue homeostasis and prevent fibrosis in chronic liver injury. Proc. Natl. Acad. Sci. USA 2016, 113, 9298–9303. [Google Scholar] [CrossRef] [Green Version]
  172. Fabre, T.; Barron, A.M.S.; Christensen, S.M.; Asano, S.; Wadsworth, M.H.; Chen, X.; Wang, J.; McMahon, J.; Schlerman, F.; White, A.; et al. Identification of a Broadly Fibrogenic Macrophage Subset Induced by Type 3 Inflammation in Human and Murine Liver and Lung Fibrosis. bioRxiv 2022. [Google Scholar] [CrossRef]
  173. Duffield, J.S. The inflammatory macrophage: A story of Jekyll and Hyde. Clin. Sci. (Lond.) 2003, 104, 27–38. [Google Scholar] [CrossRef]
  174. Song, E.; Ouyang, N.; Horbelt, M.; Antus, B.; Wang, M.; Exton, M.S. Influence of alternatively and classically activated macrophages on fibrogenic activities of human fibroblasts. Cell. Immunol. 2000, 204, 19–28. [Google Scholar] [CrossRef]
  175. Pierce, G.F.; Mustoe, T.A.; Lingelbach, J.; Masakowski, V.R.; Griffin, G.L.; Senior, R.M.; Deuel, T.F. Platelet-derived growth factor and transforming growth factor-beta enhance tissue repair activities by unique mechanisms. J. Cell Biol. 1989, 109, 429–440. [Google Scholar] [CrossRef]
  176. Qian, B.Z.; Pollard, J.W. Macrophage diversity enhances tumor progression and metastasis. Cell 2010, 141, 39–51. [Google Scholar] [CrossRef] [Green Version]
  177. Helming, L.; Gordon, S. Macrophage fusion induced by IL-4 alternative activation is a multistage process involving multiple target molecules. Eur. J. Immunol. 2007, 37, 33–42. [Google Scholar] [CrossRef]
  178. Milde, R.; Ritter, J.; Tennent, G.A.; Loesch, A.; Martinez, F.O.; Gordon, S.; Pepys, M.B.; Verschoor, A.; Helming, L. Multinucleated Giant Cells Are Specialized for Complement-Mediated Phagocytosis and Large Target Destruction. Cell Rep. 2015, 13, 1937–1948. [Google Scholar] [CrossRef] [Green Version]
  179. Prokop, S.; Heppner, F.L.; Goebel, H.H.; Stenzel, W. M2 polarized macrophages and giant cells contribute to myofibrosis in neuromuscular sarcoidosis. Am. J. Pathol. 2011, 178, 1279–1286. [Google Scholar] [CrossRef]
  180. Miron, R.J.; Bosshardt, D.D. Multinucleated Giant Cells: Good Guys or Bad Guys? Tissue Eng. Part B Rev. 2018, 24, 53–65. [Google Scholar] [CrossRef]
  181. Fleming, B.D.; Mosser, D.M. Regulatory macrophages: Setting the threshold for therapy. Eur. J. Immunol. 2011, 41, 2498–2502. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  182. Anderson, C.F.; Mosser, D.M. Cutting edge: Biasing immune responses by directing antigen to macrophage Fcγ receptors. J. Immunol. 2002, 168, 3697–3701. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  183. Anderson, C.F.; Mosser, D.M. A novel phenotype for an activated macrophage: The type 2 activated macrophage. J. Leukoc. Biol. 2002, 72, 101–106. [Google Scholar] [CrossRef] [PubMed]
  184. Wilson, H.M.; Walbaum, D.; Rees, A.J. Macrophages and the kidney. Curr. Opin. Nephrol. Hypertens. 2004, 13, 285–290. [Google Scholar] [CrossRef] [PubMed]
  185. Anderson, C.F.; Gerber, J.S.; Mosser, D.M. Modulating macrophage function with IgG immune complexes. J. Endotoxin Res. 2002, 8, 477–481. [Google Scholar] [CrossRef] [Green Version]
  186. Mosser, D.M.; Karp, C.L. Receptor mediated subversion of macrophage cytokine production by intracellular pathogens. Curr. Opin. Immunol. 1999, 11, 406–411. [Google Scholar] [CrossRef]
  187. Abdelaziz, M.H.; Abdelwahab, S.F.; Wan, J.; Cai, W.; Wang, H.; Chen, J.; Kumar, K.D.; Vasudevan, A.; Sadek, A.; Su, Z.; et al. Alternatively activated macrophages; a double-edged sword in allergic asthma. J. Transl. Med. 2020, 18, 58. [Google Scholar] [CrossRef] [Green Version]
  188. Hesse, M.; Modolell, M.; La Flamme, A.C.; Schito, M.; Fuentes, J.M.; Cheever, A.W.; Pearce, E.J.; Wynn, T.A. Differential regulation of nitric oxide synthase-2 and arginase-1 by type 1/type 2 cytokines in vivo: Granulomatous pathology is shaped by the pattern of L-arginine metabolism. J. Immunol. 2001, 167, 6533–6544. [Google Scholar] [CrossRef] [Green Version]
  189. Sapudom, J.; Karaman, S.; Mohamed, W.K.E.; Garcia-Sabaté, A.; Quartey, B.C.; Teo, J.C.M. 3D in vitro M2 macrophage model to mimic modulation of tissue repair. NPJ Regen. Med. 2021, 6, 83. [Google Scholar] [CrossRef]
  190. Powell, D.W.; Mifflin, R.C.; Valentich, J.D.; Crowe, S.E.; Saada, J.I.; West, A.B. Myofibroblasts. I. Paracrine cells important in health and disease. Am. J. Physiol. 1999, 277, C1–C9. [Google Scholar] [CrossRef]
  191. Clouthier, D.E.; Comerford, S.A.; Hammer, R.E. Hepatic fibrosis, glomerulosclerosis, and a lipodystrophy-like syndrome in PEPCK-TGF-β1 transgenic mice. J. Clin. Investig. 1997, 100, 2697–2713. [Google Scholar] [CrossRef] [Green Version]
  192. White, J.C.; Jiang, Z.L.; Diamond, M.P.; Saed, G.M. Macrophages induce the adhesion phenotype in normal peritoneal fibroblasts. Fertil. Steril. 2011, 96, 758–763.e3. [Google Scholar] [CrossRef]
  193. Mantovani, A.; Marchesi, F.; Malesci, A.; Laghi, L.; Allavena, P. Tumour-associated macrophages as treatment targets in oncology. Nat. Rev. Clin. Oncol. 2017, 14, 399–416. [Google Scholar] [CrossRef]
  194. Reed, J.R.; Stone, M.D.; Beadnell, T.C.; Ryu, Y.; Griffin, T.J.; Schwertfeger, K.L. Fibroblast growth factor receptor 1 activation in mammary tumor cells promotes macrophage recruitment in a CX3CL1-dependent manner. PLoS ONE 2012, 7, e45877. [Google Scholar] [CrossRef] [Green Version]
  195. Ge, Z.; Ding, S. The Crosstalk Between Tumor-Associated Macrophages (TAMs) and Tumor Cells and the Corresponding Targeted Therapy. Front. Oncol. 2020, 10, 590941. [Google Scholar] [CrossRef]
  196. Zhou, S.L.; Zhou, Z.J.; Hu, Z.Q.; Huang, X.W.; Wang, Z.; Chen, E.B.; Fan, J.; Cao, Y.; Dai, Z.; Zhou, J. Tumor-Associated Neutrophils Recruit Macrophages and T-Regulatory Cells to Promote Progression of Hepatocellular Carcinoma and Resistance to Sorafenib. Gastroenterology 2016, 150, 1646–1658.e17. [Google Scholar] [CrossRef] [Green Version]
  197. Pittet, M.J.; Michielin, O.; Migliorini, D. Clinical relevance of tumour-associated macrophages. Nat. Rev. Clin. Oncol. 2022, 19, 402–421. [Google Scholar] [CrossRef]
  198. Wu, K.; Lin, K.; Li, X.; Yuan, X.; Xu, P.; Ni, P.; Xu, D. Redefining Tumor-Associated Macrophage Subpopulations and Functions in the Tumor Microenvironment. Front. Immunol. 2020, 11, 1731. [Google Scholar] [CrossRef]
  199. Ferrante, C.J.; Pinhal-Enfield, G.; Elson, G.; Cronstein, B.N.; Hasko, G.; Outram, S.; Leibovich, S.J. The adenosine-dependent angiogenic switch of macrophages to an M2-like phenotype is independent of interleukin-4 receptor alpha (IL-4Rα) signaling. Inflammation 2013, 36, 921–931. [Google Scholar] [CrossRef]
  200. Cheng, S.; Li, Z.; Gao, R.; Xing, B.; Gao, Y.; Yang, Y.; Qin, S.; Zhang, L.; Ouyang, H.; Du, P.; et al. A pan-cancer single-cell transcriptional atlas of tumor infiltrating myeloid cells. Cell 2021, 184, 792–809.e23. [Google Scholar] [CrossRef]
  201. Liu, J.; Geng, X.; Hou, J.; Wu, G. New insights into M1/M2 macrophages: Key modulators in cancer progression. Cancer Cell Int. 2021, 21, 389. [Google Scholar] [CrossRef]
  202. Kumari, N.; Choi, S.H. Tumor-associated macrophages in cancer: Recent advancements in cancer nanoimmunotherapies. J. Exp. Clin. Cancer Res. 2022, 41, 68. [Google Scholar] [CrossRef] [PubMed]
  203. Anderson, J.M.; Rodriguez, A.; Chang, D.T. Foreign body reaction to biomaterials. Semin. Immunol. 2008, 20, 86–100. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  204. Brochhausen, C.; Schmitt, V.H.; Mamilos, A.; Schmitt, C.; Planck, C.N.; Rajab, T.K.; Hierlemann, H.; Kirkpatrick, C.J. Expression of CD68 positive macrophages in the use of different barrier materials to prevent peritoneal adhesions—An animal study. J. Mater. Sci. Mater. Med. 2017, 28, 15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  205. Kirkpatrick, C.J.; Bittinger, F.; Wagner, M.; Kohler, H.; van Kooten, T.G.; Klein, C.L.; Otto, M. Current trends in biocompatibility testing. Proc. Inst. Mech. Eng. Part H J. Eng. Med. 1998, 212, 75–84. [Google Scholar] [CrossRef]
  206. Kurobe, H.; Maxfield, M.W.; Breuer, C.K.; Shinoka, T. Concise review: Tissue-engineered vascular grafts for cardiac surgery: Past, present, and future. Stem Cells Transl. Med. 2012, 1, 566–571. [Google Scholar] [CrossRef]
  207. Brochhausen, C.; Schmitt, V.H.; Rajab, T.K.; Planck, C.N.; Kramer, B.; Wallwiener, M.; Hierlemann, H.; Kirkpatrick, C.J. Intraperitoneal adhesions—An ongoing challenge between biomedical engineering and the life sciences. J. Biomed. Mater. Res. A 2011, 98, 143–156. [Google Scholar] [CrossRef]
  208. Tan, R.P.; Chan, A.H.P.; Wei, S.; Santos, M.; Lee, B.S.L.; Filipe, E.C.; Akhavan, B.; Bilek, M.M.; Ng, M.K.C.; Xiao, Y.; et al. Bioactive Materials Facilitating Targeted Local Modulation of Inflammation. JACC Basic Transl. Sci. 2019, 4, 56–71. [Google Scholar] [CrossRef]
  209. Ito, T.; Shintani, Y.; Fields, L.; Shiraishi, M.; Podaru, M.N.; Kainuma, S.; Yamashita, K.; Kobayashi, K.; Perretti, M.; Lewis-McDougall, F.; et al. Cell barrier function of resident peritoneal macrophages in post-operative adhesions. Nat. Commun. 2021, 12, 2232. [Google Scholar] [CrossRef]
  210. Barsch, F.; Mamilos, A.; Schmitt, V.H.; Babel, M.; Winter, L.; Wagner, W.; Winther, H.; Ottomann, C.; Niedermair, T.; Schreml, S.; et al. In Vivo Comparison of Synthetic Macroporous Filamentous and Sponge-like Skin Substitute Matrices Reveals Morphometric Features of the Foreign Body Reaction According to 3D Biomaterial Designs. Cells 2022, 11, 2834. [Google Scholar] [CrossRef]
  211. Murray, P.J.; Wynn, T.A. Protective and pathogenic functions of macrophage subsets. Nat. Rev. Immunol. 2011, 11, 723–737. [Google Scholar] [CrossRef] [Green Version]
  212. Wynn, T.A.; Chawla, A.; Pollard, J.W. Macrophage biology in development, homeostasis and disease. Nature 2013, 496, 445–455. [Google Scholar] [CrossRef] [Green Version]
  213. Schuppan, D.; Kim, Y.O. Evolving therapies for liver fibrosis. J. Clin. Investig. 2013, 123, 1887–1901. [Google Scholar] [CrossRef] [Green Version]
  214. Krzyszczyk, P.; Schloss, R.; Palmer, A.; Berthiaume, F. The Role of Macrophages in Acute and Chronic Wound Healing and Interventions to Promote Pro-wound Healing Phenotypes. Front. Physiol. 2018, 9, 419. [Google Scholar] [CrossRef]
  215. Garash, R.; Bajpai, A.; Marcinkiewicz, B.M.; Spiller, K.L. Drug delivery strategies to control macrophages for tissue repair and regeneration. Exp. Biol. Med. 2016, 241, 1054–1063. [Google Scholar] [CrossRef] [Green Version]
  216. Ni, C.; Zhou, J.; Kong, N.; Bian, T.; Zhang, Y.; Huang, X.; Xiao, Y.; Yang, W.; Yan, F. Gold nanoparticles modulate the crosstalk between macrophages and periodontal ligament cells for periodontitis treatment. Biomaterials 2019, 206, 115–132. [Google Scholar] [CrossRef]
  217. Kim, Y.H.; Tabata, Y. Enhancement of wound closure by modifying dual release patterns of stromal-derived cell factor-1 and a macrophage recruitment agent from gelatin hydrogels. J. Tissue Eng. Regen. Med. 2017, 11, 2999–3013. [Google Scholar] [CrossRef]
  218. Lee, J.-H.; Kim, H.-W. Emerging properties of hydrogels in tissue engineering. J. Tissue Eng. 2018, 9, 2041731418768285. [Google Scholar] [CrossRef] [Green Version]
  219. Lee, J.H. Injectable hydrogels delivering therapeutic agents for disease treatment and tissue engineering. Biomater. Res. 2018, 22, 27. [Google Scholar] [CrossRef] [Green Version]
  220. Xu, J.; Duan, Z.; Qi, X.; Ou, Y.; Guo, X.; Zi, L.; Wei, Y.; Liu, H.; Ma, L.; Li, H.; et al. Injectable Gelatin Hydrogel Suppresses Inflammation and Enhances Functional Recovery in a Mouse Model of Intracerebral Hemorrhage. Front. Bioeng. Biotechnol. 2020, 8, 785. [Google Scholar] [CrossRef]
  221. Shiratori, H.; Feinweber, C.; Luckhardt, S.; Wallner, N.; Geisslinger, G.; Weigert, A.; Parnham, M.J. An in vitro test system for compounds that modulate human inflammatory macrophage polarization. Eur. J. Pharmacol. 2018, 833, 328–338. [Google Scholar] [CrossRef]
  222. Huang, Y.J.; Hung, K.C.; Hung, H.S.; Hsu, S.H. Modulation of Macrophage Phenotype by Biodegradable Polyurethane Nanoparticles: Possible Relation between Macrophage Polarization and Immune Response of Nanoparticles. ACS Appl. Mater. Interfaces 2018, 10, 19436–19448. [Google Scholar] [CrossRef]
  223. Hsieh, J.Y.; Smith, T.D.; Meli, V.S.; Tran, T.N.; Botvinick, E.L.; Liu, W.F. Differential regulation of macrophage inflammatory activation by fibrin and fibrinogen. Acta Biomater. 2017, 47, 14–24. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  224. Chung, S.; Revia, R.A.; Zhang, M. Iron oxide nanoparticles for immune cell labeling and cancer immunotherapy. Nanoscale Horiz. 2021, 6, 696–717. [Google Scholar] [CrossRef]
  225. Sang, Y.; Deng, Q.; Cao, F.; Liu, Z.; You, Y.; Liu, H.; Ren, J.; Qu, X. Remodeling Macrophages by an Iron Nanotrap for Tumor Growth Suppression. ACS Nano 2021, 15, 19298–19309. [Google Scholar] [CrossRef]
  226. Ogle, M.E.; Segar, C.E.; Sridhar, S.; Botchwey, E.A. Monocytes and macrophages in tissue repair: Implications for immunoregenerative biomaterial design. Exp. Biol. Med. 2016, 241, 1084–1097. [Google Scholar] [CrossRef] [PubMed]
  227. Krieger, J.R.; Ogle, M.E.; McFaline-Figueroa, J.; Segar, C.E.; Temenoff, J.S.; Botchwey, E.A. Spatially localized recruitment of anti-inflammatory monocytes by SDF-1α-releasing hydrogels enhances microvascular network remodeling. Biomaterials 2016, 77, 280–290. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  228. Ogle, M.E.; Sefcik, L.S.; Awojoodu, A.O.; Chiappa, N.F.; Lynch, K.; Peirce-Cottler, S.; Botchwey, E.A. Engineering in vivo gradients of sphingosine-1-phosphate receptor ligands for localized microvascular remodeling and inflammatory cell positioning. Acta Biomater. 2014, 10, 4704–4714. [Google Scholar] [CrossRef] [Green Version]
  229. Lee, C.H.; Kim, Y.J.; Jang, J.H.; Park, J.W. Modulating macrophage polarization with divalent cations in nanostructured titanium implant surfaces. Nanotechnology 2016, 27, 085101. [Google Scholar] [CrossRef]
  230. Zhu, Y.; Liang, H.; Liu, X.; Wu, J.; Yang, C.; Wong, T.M.; Kwan, K.Y.H.; Cheung, K.M.C.; Wu, S.; Yeung, K.W.K. Regulation of macrophage polarization through surface topography design to facilitate implant-to-bone osteointegration. Sci. Adv. 2021, 7, eabf6654. [Google Scholar] [CrossRef]
  231. Boyce, B.F.; Rosenberg, E.; de Papp, A.E.; Duong, L.T. The osteoclast, bone remodelling and treatment of metabolic bone disease. Eur. J. Clin. Investig. 2012, 42, 1332–1341. [Google Scholar] [CrossRef]
  232. Schulze-Tanzil, G.; Zreiqat, H.; Sabat, R.; Kohl, B.; Halder, A.; Muller, R.D.; John, T. Interleukin-10 and articular cartilage: Experimental therapeutical approaches in cartilage disorders. Curr. Gene Ther. 2009, 9, 306–315. [Google Scholar] [CrossRef]
  233. Fantin, A.; Vieira, J.M.; Gestri, G.; Denti, L.; Schwarz, Q.; Prykhozhij, S.; Peri, F.; Wilson, S.W.; Ruhrberg, C. Tissue macrophages act as cellular chaperones for vascular anastomosis downstream of VEGF-mediated endothelial tip cell induction. Blood 2010, 116, 829–840. [Google Scholar] [CrossRef] [Green Version]
  234. Grunewald, M.; Avraham, I.; Dor, Y.; Bachar-Lustig, E.; Itin, A.; Jung, S.; Chimenti, S.; Landsman, L.; Abramovitch, R.; Keshet, E. VEGF-induced adult neovascularization: Recruitment, retention, and role of accessory cells. Cell 2006, 124, 175–189. [Google Scholar] [CrossRef] [Green Version]
  235. Du Cheyne, C.; Tay, H.; De Spiegelaere, W. The complex TIE between macrophages and angiogenesis. Anat. Histol. Embryol. 2020, 49, 585–596. [Google Scholar] [CrossRef]
  236. Debels, H.; Galea, L.; Han, X.; Palmer, J.; Van Rooijen, N.; Morrison, W.A.; Abberton, K.M. Macrophages play a key role in angiogenesis and adipogenesis in a mouse tissue engineering model. Tissue Eng. Part A 2013, 19, 2615–2625. [Google Scholar] [CrossRef] [Green Version]
  237. Kirkpatrick, C.J.; Krump-Konvalinkova, V.; Unger, R.E.; Bittinger, F.; Otto, M.; Peters, K. Tissue response and biomaterial integration: The efficacy of in vitro methods. Biomol. Eng. 2002, 19, 211–217. [Google Scholar] [CrossRef]
  238. Ono, M.; Torisu, H.; Fukushi, J.; Nishie, A.; Kuwano, M. Biological implications of macrophage infiltration in human tumor angiogenesis. Cancer Chemother. Pharmacol. 1999, 43, S69–S71. [Google Scholar] [CrossRef]
  239. Guida, E.; Stewart, A. Influence of hypoxia and glucose deprivation on tumour necrosis factor-α and granulocyte-macrophage colony-stimulating factor expression in human cultured monocytes. Cell. Physiol. Biochem. 1998, 8, 75–88. [Google Scholar] [CrossRef]
  240. Mastrullo, V.; Cathery, W.; Velliou, E.; Madeddu, P.; Campagnolo, P. Angiogenesis in Tissue Engineering: As Nature Intended? Front. Bioeng. Biotechnol. 2020, 8, 188. [Google Scholar] [CrossRef]
  241. Spiller, K.L.; Anfang, R.R.; Spiller, K.J.; Ng, J.; Nakazawa, K.R.; Daulton, J.W.; Vunjak-Novakovic, G. The role of macrophage phenotype in vascularization of tissue engineering scaffolds. Biomaterials 2014, 35, 4477–4488. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  242. Gan, J.; Liu, C.; Li, H.; Wang, S.; Wang, Z.; Kang, Z.; Huang, Z.; Zhang, J.; Wang, C.; Lv, D.; et al. Accelerated wound healing in diabetes by reprogramming the macrophages with particle-induced clustering of the mannose receptors. Biomaterials 2019, 219, 119340. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Formation and differentiation of macrophages. Schematic depiction of the differentiation of monocytes during embryonic development unfolding in three sequential waves and adulthood as well as their further differentiation to different subpopulations of macrophages in the tissue [25] (AGM region: aorta-gonad-mesonephros region; EMP: Erythro-Myeloid Progenitors, YS: yolk sac), modified after Corliss et al. [26].
Figure 1. Formation and differentiation of macrophages. Schematic depiction of the differentiation of monocytes during embryonic development unfolding in three sequential waves and adulthood as well as their further differentiation to different subpopulations of macrophages in the tissue [25] (AGM region: aorta-gonad-mesonephros region; EMP: Erythro-Myeloid Progenitors, YS: yolk sac), modified after Corliss et al. [26].
Cells 12 00276 g001
Figure 2. Plasticity of macrophages: a brief schematic depiction of the plasticity of macrophages from an inactive macrophage (MΦ) into either an M1-macrophage or M2-macrophage according to different stimuli. The scheme represents macrophages as an activations state of cells that can be changed along a continuum into the different sub-populations according to various stimuli in the environment. In addition, M1 and M2 macrophages can be turned into inactivated macrophages if there is a lack of stimuli.
Figure 2. Plasticity of macrophages: a brief schematic depiction of the plasticity of macrophages from an inactive macrophage (MΦ) into either an M1-macrophage or M2-macrophage according to different stimuli. The scheme represents macrophages as an activations state of cells that can be changed along a continuum into the different sub-populations according to various stimuli in the environment. In addition, M1 and M2 macrophages can be turned into inactivated macrophages if there is a lack of stimuli.
Cells 12 00276 g002
Figure 3. Activation of classically activated macrophages. In the presence of INF-γ-receptor ligands or PAMP, the macrophages adopt the M1 phenotype, which is characterized by the expression of INF-β and TNF-α –receptors. Furthermore, these cells are now capable of synthesizing TNF-α and INF-β and thus achieving self-activation.
Figure 3. Activation of classically activated macrophages. In the presence of INF-γ-receptor ligands or PAMP, the macrophages adopt the M1 phenotype, which is characterized by the expression of INF-β and TNF-α –receptors. Furthermore, these cells are now capable of synthesizing TNF-α and INF-β and thus achieving self-activation.
Cells 12 00276 g003
Figure 4. Summary of the basic functions of M1-Macrophages.
Figure 4. Summary of the basic functions of M1-Macrophages.
Cells 12 00276 g004
Figure 5. The activation of M2a-Macrophages. In the presence of IL-4- and IL-13- receptor ligands, the macrophages adopt the M2a phenotype, which is characterized by the expression of INF-β and TNF-α –receptors. Furthermore, these cells are now able to synthesize TNF-α and INF-β and thus undergo self-activation.
Figure 5. The activation of M2a-Macrophages. In the presence of IL-4- and IL-13- receptor ligands, the macrophages adopt the M2a phenotype, which is characterized by the expression of INF-β and TNF-α –receptors. Furthermore, these cells are now able to synthesize TNF-α and INF-β and thus undergo self-activation.
Cells 12 00276 g005
Figure 6. Summary of the function of M2a-macrophages at the cellular level.
Figure 6. Summary of the function of M2a-macrophages at the cellular level.
Cells 12 00276 g006
Figure 7. The polarization of M2b-Macrophages. In the presence of immune complexes, TLR-ligands, and IL-1 receptor ligands, the macrophages adopt the M2b phenotype.
Figure 7. The polarization of M2b-Macrophages. In the presence of immune complexes, TLR-ligands, and IL-1 receptor ligands, the macrophages adopt the M2b phenotype.
Cells 12 00276 g007
Figure 8. The polarization to M2c-Macrophages. In the presence of glucocorticoids (GC), IL-10 or TGF-β receptor ligands, the macrophages adopt the M2c phenotype, which is capable of synthesizing IL-10 and TGF-β and thus reaching a state of self-activation.
Figure 8. The polarization to M2c-Macrophages. In the presence of glucocorticoids (GC), IL-10 or TGF-β receptor ligands, the macrophages adopt the M2c phenotype, which is capable of synthesizing IL-10 and TGF-β and thus reaching a state of self-activation.
Cells 12 00276 g008
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Mamilos, A.; Winter, L.; Schmitt, V.H.; Barsch, F.; Grevenstein, D.; Wagner, W.; Babel, M.; Keller, K.; Schmitt, C.; Gürtler, F.; et al. Macrophages: From Simple Phagocyte to an Integrative Regulatory Cell for Inflammation and Tissue Regeneration—A Review of the Literature. Cells 2023, 12, 276. https://doi.org/10.3390/cells12020276

AMA Style

Mamilos A, Winter L, Schmitt VH, Barsch F, Grevenstein D, Wagner W, Babel M, Keller K, Schmitt C, Gürtler F, et al. Macrophages: From Simple Phagocyte to an Integrative Regulatory Cell for Inflammation and Tissue Regeneration—A Review of the Literature. Cells. 2023; 12(2):276. https://doi.org/10.3390/cells12020276

Chicago/Turabian Style

Mamilos, Andreas, Lina Winter, Volker H. Schmitt, Friedrich Barsch, David Grevenstein, Willi Wagner, Maximilian Babel, Karsten Keller, Christine Schmitt, Florian Gürtler, and et al. 2023. "Macrophages: From Simple Phagocyte to an Integrative Regulatory Cell for Inflammation and Tissue Regeneration—A Review of the Literature" Cells 12, no. 2: 276. https://doi.org/10.3390/cells12020276

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop