Next Article in Journal
Naturally-Occurring Tyrosinase Inhibitors Classified by Enzyme Kinetics and Copper Chelation
Next Article in Special Issue
Extracellular Vesicles Released from Macrophages Infected with Mycoplasma pneumoniae Stimulate Proinflammatory Response via the TLR2-NF-κB/JNK Signaling Pathway
Previous Article in Journal
Molecular Target and Action Mechanism of Anti-Cancer Agents
Previous Article in Special Issue
Macrophage Migration Inhibitory Factor in Psoroptes ovis: Molecular Characterization and Potential Role in Eosinophil Accumulation of Skin in Rabbit and Its Implication in the Host–Parasite Interaction
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Multiple Shades of Gray—Macrophages in Acute Allograft Rejection

1
Daniel Swarovski Research Laboratory, Department of Visceral, Transplant and Thoracic Surgery, Medical University of Innsbruck, 6020 Innsbruck, Austria
2
Institute of Biological Chemistry, Biocenter, Medical University of Innsbruck, 6020 Innsbruck, Austria
3
Department of Visceral, Transplant, and Thoracic Surgery, Medical University of Innsbruck, 6020 Innsbruck, Austria
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(9), 8257; https://doi.org/10.3390/ijms24098257
Submission received: 25 March 2023 / Revised: 27 April 2023 / Accepted: 1 May 2023 / Published: 4 May 2023
(This article belongs to the Special Issue Macrophage Polarization: Learning to Manage It 2.0)

Abstract

:
Long-term results following solid organ transplantation do not mirror the excellent short-term results achieved in recent decades. It is therefore clear that current immunosuppressive maintenance protocols primarily addressing the adaptive immune system no longer meet the required clinical need. Identification of novel targets addressing this shortcoming is urgently needed. There is a growing interest in better understanding the role of the innate immune system in this context. In this review, we focus on macrophages, which are known to prominently infiltrate allografts and, during allograft rejection, to be involved in the surge of the adaptive immune response by expression of pro-inflammatory cytokines and direct cytotoxicity. However, this active participation is janus-faced and unspecific targeting of macrophages may not consider the different subtypes involved. Under this premise, we give an overview on macrophages, including their origins, plasticity, and important markers. We then briefly describe their role in acute allograft rejection, which ranges from sustaining injury to promoting tolerance, as well as the impact of maintenance immunosuppressants on macrophages. Finally, we discuss the observed immunosuppressive role of the vitamin-like compound tetrahydrobiopterin and the recent findings that suggest the innate immune system, particularly macrophages, as its target.

1. Introduction

Over the last five decades, organ transplantation has developed from an experimental treatment strategy to standard of care for end-stage failure of vital organs. Continuous advances in preservation strategies, surgical techniques, perioperative antibiotic prophylaxis and anesthesia were and are still pivotal for this development [1,2,3,4]. However, the major cornerstone for its success was the introduction of modern immunosuppression starting with cyclosporine A (CsA) in the 1980s [5,6]. Despite constant improvements, drug toxicities resulting from lifelong maintenance immunosuppressive medication still compromise graft and recipient survival, resulting in unvaried long-term attrition of transplanted organs that does not reflect the excellent short-term improvements [7]. Additionally, important adverse effects of lifelong immunosuppression such as hepatic and renal toxicity, hyperlipidemia and infections hamper graft and patient survival, limiting their quality of life [8,9,10,11]. As a result, currently available maintenance immunosuppressive protocols do not meet the required clinical needs and identifying novel molecular targets and pathways to improve long-term graft survival remains of utmost importance [12,13,14].
Historically, the research addressing allograft rejection mainly focused on the adaptive immune system, increasing the knowledge regarding different cell subsets involved in this process [15,16]. This goes back to early publications showing that T-cells played a crucial role in allograft rejection [17,18]. As a result, inhibiting differentiation and proliferation of the activated adaptive immune cells and their products became the focus of various immunosuppressive agents that entered the market [19].
Even though the presence of mononuclear cells infiltrating the rejected graft was already described in the 1980s [20] and macrophages were found to make up 60% of these infiltrating cells in organs undergoing acute rejection [21], cells of the innate immune system are only nowadays increasingly recognized as crucial contributors in transplant immunology [22]. Macrophages are among the first cells populating injured tissue, such as a transplanted organ [23]. Thanks to a myriad of diverse surface receptors such as pattern recognition receptors (PRR) that recognize damage-associated molecular patterns (DAMPs), macrophages locate to the injured region, fuel transcription of pro-inflammatory mediators [24] and act as a bridge for the initiation of the adaptive immune response [25]. However, pro-inflammatory mediators do not tell the whole story about macrophages.
In this review, we give an overview about general aspects of macrophages including their origin, classification, polarization markers and plasticity. We then focus on the evolving role of macrophages in acute allograft rejection, which ranges from pro-inflammatory to tolerogenic, and on the impact of clinically used immunosuppressive drugs on macrophage function. Finally, we review emerging results on tetrahydrobiopterin (BH4), a vitamin-like compound readily synthesized in the human body and cofactor to eight vital reactions, which has been shown to impact ischemia reperfusion injury (IRI), acute rejection as well as chronic rejection following solid organ rejection, rendering it a potentially novel immunosuppressive agent addressing the innate immune system.

2. Types of Macrophages, Polarization and Markers

2.1. Tissue-Resident Macrophages (TRM) and Monocyte-Derived Macrophages

Macrophages (large eaters, from Greek: μακρός and φαγεῖν) are white blood cells that are part of the innate immune system and their specialty is to detect and destroy pathogens [26,27]. In the body, two distinct pools of these cells exist, i.e., monocyte-derived macrophages that originate from bone marrow and tissue-resident macrophages (TRM) that arise during fetal development. The latter derive from the yolk sac and the fetal liver [28] and can generate specific phenotypes depending on the organ where they are located [29]. In the liver, they can be found as Kupffer cells, whereas bone-located macrophages are called osteoclasts and microglia are resident macrophages in the brain [22]. Additionally, the gut presents with resident macrophage populations that have distinct phenotypes and functions and are crucial for maintaining tolerance to the gut flora and orally administered antigens [30,31]. In the spleen, a special subtype of resident macrophages suppresses immunogenic reactions to apoptotic cells by efficiently clearing their material [32] and macrophages lining the subcapsular sinus of lymph nodes are involved in capturing lymph-borne pathogens [33]. For a comprehensive review, describing these different types of TRM, including others such as Langerhans cells and alveolar macrophages, the reader is referred to [28]. In contrast to the TRM, monocyte-derived macrophages originating from the bone marrow are patrolling in the blood circulation as part of the peripheral blood mononuclear cells (PBMCs). They can adhere to the vascular endothelium and pass through the endothelial cell gap, migrate to the tissues and then develop locally into differently polarized macrophages [34].

2.2. Macrophage Phenotypes and Respective Markers

Back in the 1980s, interferon (IFN) γ produced by Th1 helper cells was found to activate macrophages [35], while the Th2 cytokine interleukin (IL)-4 was identified as a mediator of macrophage activation, which however led to a different phenotype as that observed with IFNγ, in the 1990s [36]. Finally, in 2000, nitric oxide (NO) and arginine production profiles were compared in two mouse strains, a prototypical Th1 (C57Bl/6) and Th2 strain (BALB/c), and the authors proposed the Th1-elicited macrophages to be called M1, while the Th2-elicited macrophages were since then terminated M2 [37]. Metabolic studies have recently shown that certain metabolic pathways are closely related to the phenotype and function of macrophages, as M1 macrophages display increased glycolysis, fatty acid synthesis (FAS) and pentose phosphate pathway (PPP) metabolism. In M2 macrophages, high mitochondrial oxidative phosphorylation (OXPHOS) can be found and enhanced fatty acid oxidation (FAO) and glutamine metabolism are linked to M2 activation (for detailed information the reader is referred to [38]).
Phenotypically, macrophages are identified as CD45+ cells that express myeloid cell markers (general phenotypic marker for mice are, e.g., CD11b, F4/80, CD68, MERTK, and Ly6C; markers for humans are, e.g., CD11b, CD14, CD16, CD33, and CD68) but lack surface markers that are associated with adaptive immune cells (e.g., CD3, CD19, CD56, NKp46, TCR, and BCR). Regulatory macrophages (Mregs) in mice additionally express CD11c, CD169 and Dectin-1 [39], while CD11c [40] and DHRS9 [41,42] are found in humans. It should be emphasized that macrophages also express receptors for chemokines, cytokines, DAMPs, Fc fragments and complement products. These receptors empower macrophages to respond to a broad bundle of immune and inflammatory mediators as a result of complement activation, tissue inflammation, T-cell activation, antibody production, as well as dysregulated cell death [41]. Hence, the expression of such additional receptors further divides macrophages into complex subsets, especially in relation to their activation status, anatomical location or division of functional activities. Table 1 gives an overview of the so far described main monocyte-derived macrophages and TRM concerning their corresponding functional characteristics and markers.
M1 macrophages (also named classically activated or pro-inflammatory macrophages) can be induced by Th1 cytokines such as IFNγ and tumor necrosis factor (TNF) α as well as microbial products or DAMPs. From a molecular point of view, the induction by these factors leads to the activation of signal transducer and activator of transcription (STAT) 1, interferon-regulatory factor (IRF) 5 and, nuclear factor kappa B (NF-κB), which drive the expression of M1 markers such as inducible nitric oxide synthase (iNOS), MHC class II and inflammatory mediators such as IL-1β, IL-6, IL-12, IL-23, TNFα and cyclooxy-genase (COX) 2 [57]. Functionally, M1 macrophages play a role in the removal of pathogens during infection, harbor anti-microbial activity, mediate reactive oxygen species (ROS)-induced tissue damage and impair tissue regeneration and wound healing [27].
M2 macrophages (also named alternatively activated, anti-inflammatory macrophages) are activated by IL-4 or IL-13 which by binding to the IL-4 receptor activate STAT6 and IRF4 that drive transcription of M2 markers such as arginase (ARG) 1, peroxisome proliferator-activated receptor (PPAR) γ, suppressor of cytokine signaling (SOCS) 1, CD163, CD206, anti-inflammatory mediators such as IL-10 and transforming growth factor (TGF) β as well as other growth factors such as vascular endothelial growth factor (VEGF), platelet-derived growth factor (PDGF) and insulin-like growth factor (IGF). M2 macrophages are involved in, e.g., fibrosis, angiogenesis, wound healing, and phagocytosis of dying cells, and usually, the phenotype of TRM reflects such an anti-inflammatory M2 state [57]. The M2 phenotype can be further subdivided into type 2a (induced by IL-4 and IL-13, promoting fibrosis), type 2b (induced by immune complexes (ICs), toll-like receptor (TLR) agonists or IL-1 receptor ligands, producing pro- and anti-inflammatory cytokines), type 2c (induced by IL-10 and glucocorticoids (GC), inhibiting inflammation and eliminating apoptotic cells via phagocytosis), and type 2d (activated by TLR ligands through A2 adeno-sine receptor (A2R) agonists, inducing anti-inflammatory cytokines, favoring angiogenesis, features similar to tumor-associated macrophages (TAMs) (for a review, see [27,58]).

2.3. Macrophage Plasticity

In recent decades, the original classification of merely M1, M2 and M2 subtypes, which arose mostly from in vitro experiments and does not represent the actual in vivo situation, was challenged by the emergence of more and more phenotypes lying in between the two polarization extremes, M1 and M2 (see Table 1). As a clear and comprehensive nomenclature for the variety of known macrophage states was missing, a fact that greatly impacts on the possibility to compare the data of different published studies, there have been efforts to implement a more universal nomenclature. A suggestion to better describe the remarkable and interchangeable heterogeneity of polarization phenotypes was published in 2014 which postulated to include source (e.g., macrophages generated in vitro from human peripheral blood or murine bone marrow and macrophages isolated from human or murine tissues) and conditions used for macrophage polarization/activation as well as the activation agent (e.g., M(IL-4), M(IL-10), and M(IFNγ)) [53].
Even once polarized to a certain subtype, macrophages remain highly plastic in vivo and can quickly readapt their phenotype depending on the surroundings and the demand of the tissue they are located in [59]. Due to their highly plastic properties, macrophages can assist in all steps of inflammation starting from the initial inflammatory mounting phase and ranging to the resolution phase where tissue regeneration and repair is finalized [60]. In a zebrafish fin amputation model, it could be shown that macrophages were attracted to the wound in a single wave and changed their shape and behavior depending on the requirements directly at the wounded site [61]. The same study also provided a link to lipid metabolism, by showing the implication of polyunsaturated fatty acid (PUFA) metabolism and 15-lipoxygenase on the morphology and polarization of macrophages. In macrophages harvested from either aged or tumor-bearing mice, it was shown that they could be reprogrammed by exposing them to a different cytokine environment [62]. Still, the exact mechanisms leading to reprogramming and adaptation are not yet fully understood. As macrophage plasticity is important also for other immunological challenges such as organ transplantation [58] and disturbances in macrophage plasticity can lead to pathogenic conditions such as cancer, asthma and atherosclerosis, there is an urgent need to better understand the underlying processes at a molecular level.

3. Macrophages in Acute Rejection and Organ Tolerance

In contrast to the adaptive immune system entailing T-cell- and B-cell-mediated immunity, the role of the innate immune system is still less appreciated in allograft recognition. This, despite the already mentioned observation, that macrophages represent up to 60% of cellular infiltrates of the allograft during the rejection process [20] and that the amount of graft infiltrating macrophages has been correlated with worse outcomes [63].
The peak time of infiltrating monocyte-derived cells occurs within the first 48 h after reperfusion of the transplanted organ, with M1 macrophages initially dominating the scene. Three to five days later another phenotype predominates the graft, the M2 macrophages [64]. M1 macrophages are involved in tissue injury as a response to damage and pro-inflammatory signals while M2 macrophages mediate tissue remodeling and injury resolution, suggesting them as promoters of graft damage repair [21]. M2 macrophages might however also prevent and not only resolve the damage. In murine corneal allografts, an increase in M1 macrophages was observed during acute rejection while M2 macrophages were present in those mice that did not reject the transplant [65]. However, the simplified discrimination into classically activated M1 macrophages induced by Th1 cytokines presenting pro-inflammatory properties and alternatively activated M2 macrophages polarized by Th2 cytokines harboring anti-inflammatory and immunoregulatory properties is often challenging as switches between both macrophage phenotypes occur [27]. This complexity based on interactions of macrophages with their environment is also reflected in the different roles they assume during the entire process of allograft engraftment. This starts with IRI of the transplanted organ, passes over to acute allograft rejection and eventually ends in chronic allograft rejection characterized by allograft vasculopathy and graft failure (for a comprehensive review, we refer readers to [58]).

3.1. M1 Macrophages

It is nowadays accepted that the innate and the adaptive immune system interact with each other, with the former acting as a bridge to the activation and initiation of the latter resulting finally in allograft rejection. The basis for the injury promoting deleterious processes in the transplanted grafts starts already before the implantation of the graft. The brain death of the donor, the retrieval of the organ with its associated ischemia (which is even worse in case of donations after cardiac death) and the subsequent reperfusion of the organ result in the so-called IRI [66].
This initial injury triggers several processes promoting infiltration of mononuclear phagocytic cells with a predominant pro-inflammatory M1 phenotype. Crucial is the occurrence of tissue injury-associated DAMPs, which are recognized and processed by these innate immune cells thanks to PRRs such as TLR, NOD-like receptors or C-type lectin receptors, resulting in increased transcription of pro-inflammatory mediators. In addition, the increased expression of the transcription factor hypoxia-inducible factor 1α (HIF1α) induced by tissue hypoxia also triggers increased expression and production of pro-inflammatory cytokines. As a result, the classically activated macrophages that predominate in the initial phase generate high amounts of ROS and reactive nitrogen species (RNS) [67] aggravating allograft injury and secrete pro-inflammatory cytokines such as IL-1β, IL-12, IL-18, TNFα and IFNγ [68,69,70,71,72]. This results in activation of endothelial cells and promotion of cytotoxic T-cell generation [73,74]. The pivotal role of macrophages during acute allograft rejection is not only suggested by the presence of macrophages in specimens showing acute cellular rejection [75] but also by transcriptome analyses of such biopsies. In the latter, prominent transcript levels of the macrophage colony-stimulating factor (M-CSF) are noteworthy since this chemokine produced by the injured renal tubular epithelial cells (TEC) induces increased surface expression of MHC class II (in humans HLA-DR) and co-stimulatory molecules CD40 and CD80 on monocytes transforming them into antigen presenting cells (APC) [76]. Still, the initiation seems to be based on promoting cell differentiation of activated T-cells rather than antigen presentation since macrophages are weak antigen presenters compared to dendritic cells [77]. In fact, alteration of surface molecules such as phosphatidylserine receptor TIM4 or DC-SIGN (also known as CD209) can switch their ability from promoting differentiation of effector or helper T-cell to enhance the expansion of CD4+CD25+FoxP3+ regulatory T-cells (Tregs) [78,79]. Similarly, depending on the secreted cytokines macrophages might promote an allograft rejecting Th1/Th17 [80] or an allograft accepting CD4+CD25+FoxP3+ environment [40].
Recent findings challenge the traditional view of macrophages as innate immune cells mounting a non-allogeneic-specific inflammatory reaction. In mice lacking adaptive immune cells, rejection of the allograft resulted in an innate immune response based on the activation of the CD47/SIRPα pathway. SIRPα is a receptor of the immunoglobulin superfamily that mediates a “do not eat me” signal to docking macrophages. In this non-MHC-dependent allorecognition, SIRPα polymorphism of the donor was recognized by infiltrating macrophages allowing them to discriminate between self and non-self and to further start the rejection response [40,81].
Another finding challenging the traditional view of innate immune cells is the “trained immunity”. Macrophages can not only sense non-self antigens, but they might also develop immune memory. In a mouse model, macrophages exposed to allogeneic stimuli on day zero were able to develop an elevated immune response up to four weeks after the first exposure. The allogeneic memory was mediated by the paired immunoglobulin-like receptors-A (PIR-A) receptor binding the non-self MHC class I antigen and blocking this receptor resulted in prolonged allograft survival [82,83].
In line with these findings, in a rat kidney transplantation model of T-cell-mediated acute rejection, depletion of macrophages with liposomal-clodronate or c-fms kinase inhibitor improved allograft function attenuating its injury [84,85].
However, rather than just unspecifically deleting the presence of macrophages, shaping polarization of macrophages emerges as another strategy to prevent organ rejection.

3.2. M2 Macrophages

The role of M2 macrophages in acute rejection is heavily discussed as well as whether targeting macrophages in transplant immunology would present a promising treatment approach [78,86,87]. As they are able to amplify (anti-)inflammatory processes by cross-talking with the adaptive immune system, some therapeutic approaches of macrophage manipulation have recently moved into the focus of transplant immunology research [88,89]. As already mentioned above, M2 macrophages play a janus-faced role in solid organ transplantation. While playing an anti-inflammatory role, limiting excessive tissue injury caused by the initial infiltration of the predominantly M1 phenotype, M2 macrophages are crucial in the development of chronic allograft rejection promoting allograft fibrosis and chronic allograft vasculopathy.
Regarding acute rejection, there are different modulatory effects attributed to alternatively activated M2 macrophages. In mice, one of these effects could be related to the high expression of the M2-associated enzyme ARG1. Consumption of L-arginine leads to reduced T-cell proliferation and effector response [90]. Next, production of anti-inflammatory cytokines such as IL-10 promotes induction and proliferation of FoxP3+ Tregs [78]. Additionally, also increased phagocytosis has been recently observed to modulate acute rejection. Treatment with mesenchymal stem cells (MSC) overexpressing soluble fibrinogen-like protein 2 (sFgl2) which is mainly produced by Treg cells significantly delayed acute rejection and promoted the polarization of M2 macrophages in the graft. These M2 macrophages showed a higher migration and phagocytosis capacity in vitro when treated with IFNγ and lipopolysaccharides (LPS), suggesting a high activity in resolving the existing damage [87]. With regard to MSC, in a mouse transplantation model, local co-application of MSC and CsA resulted in a phenotype switch of infiltrating macrophages from M1 to M2. This was characterized by a decrease in intragraft NO and IFNγ production, while alternatively activated F4/80+ CD206+ M2 macrophages and IL-10 became predominant [91].
In a mouse cardiac transplantation model, tolerance acquired by co-stimulatory blockade with anti-CD40L mAb resulted in a conversion of pro-inflammatory macrophage precursors to M2-like regulatory macrophages. Interestingly, depletion of CD11b expressing monocytes of the recipients prevented occurrence of tolerance [78,92]. Using the same tolerance model, the presence of M2 macrophages was associated with graft infiltrating neutrophils secreting higher levels of M-CSF, a growth factor known to regulate macrophage proliferation and differentiation [93].

3.3. Regulatory Macrophages (Mregs)

Allograft-specific tolerance of the recipient immune system is the holy grail of transplant medicine. Since lifelong maintenance of immunosuppression is associated with serious long-term complications, allograft-specific tolerance would allow to significantly reduce the need for pharmacological immunosuppression. Historically, these studies include depletion protocols aimed to generate hematopoietic chimerism and focused on the adaptive immune system [94].
However, recent data provide evidence that a subtype of macrophages, the so-called regulatory macrophages (Mregs), can also induce a tolerogenic environment. Mregs can be seen as a different state of polarization since they differ from both the M1 and the M2 subtypes (Table 1). They possess a strong antigen presenting capacity due to highly expressed CD80 and MHC class II receptors and harbor an anti-inflammatory property producing large amounts of IL-10 and TGF-β and low levels of IL-12. For an in-depth review, we would refer the readers Zhang and colleagues [95]. The immunosuppressive capacity of Mregs is described to rely on different mechanisms. One is the secretion of anti-inflammatory cytokines such as IL-4, IL-10 and TGFβ [96]. Using a tolerogenic mouse cardiac transplantation model based on anti-CD40L mAb administration, the presence of Mregs characterized by DC-SIGN expression was flanked by increased numbers of CD4+ CD25+ FoxP3+ Tregs, decreased CD8+ T-cell activity and prominent intragraft levels of IL-10 [78]. From a mechanistic point of view, direct delivery of inhibitors affecting the M1 polarization pathways mTOR and NF-κB via nanoparticles also resulted in shifting macrophages towards a suppressive phenotype reducing CD8+ T-cell-mediated alloreactivity and Treg expansion [97]. In vitro, Mregs have also been shown to phagocyte and inactivate allogeneic T-cells by direct contact in an iNOS-dependent fashion. A single administration of ex vivo-generated donor-derived CD11b+ Ly6C-/low Ly6G- CD169+ Mregs before transplantation (either eight or 35 days before transplantation) resulted in prolonged allograft survival of fully allogeneic heart transplantation in mice. Of note, third party or recipient-derived Mregs did not confer the observed protective effect [39]. In this regard, the missed benefit of postoperative administration of in vitro cultured CD14+, CD16+, and CD18+ Mregs in a porcine lung transplantation model points to the importance of a preoperative administration as an optimal time frame [98].
Mregs have already found their way into clinical trials. Early clinical trials using these cells showed technical feasibility and safety of this immunosuppressive strategy [99]; however, as for experimental studies, only the switch from a postoperative [100] to a preoperative application [101] of these cells led to successful minimization of the immunosuppression in some patients. The EU-funded consortium called The ONE study developed a range of cell-based medicinal products (CBMPs) thought to be administered to allograft recipients with the final goal to reduce standard-of-care maintenance immunosuppression. The six developed CBMPs included two polyclonal Tregs (pTreg-1 or pTreg-2), two donor-antigen-specific Tregs, one tolerogenic dendritic cell and one Mreg cell product. Of note, all regulatory cell products except Mregs were derived from the recipient. The Mregs were the only donor-derived cell product. Phase 1/2A trials in living-donor kidney recipients using these CBMPs showed similar incidences of biopsy-proven acute rejections compared to standard-of-care maintenance immunosuppression, successful reduction in maintenance immunosuppression in nearly half of the patients and fewer infectious complications [102]. From a mechanistic point of view, co-culture and humanized mouse experiments showed that donor-derived Mregs induced a high proportion of FoxP3- CD4+ T-cells to become IL-10 producing FoxP3+ Tregs. These macrophage-induced Tregs did not derive from natural (n)Treg developed in the thymus and expressed cell-surface T-cell immunoreceptor with Ig and ITIM domains (TIGIT). Of note, these TIGIT+ iTregs were found in a recipient of The ONE study receiving Mregs (ONEmreg12 trial) at the seven year follow-up post transplantation and might be crucial in contributing to long-term acceptance of the graft. Among the multiple non-redundant molecules involved in TIGIT+ iTregs development also indoleamine-2,3-dioxygenase (IDO) and progestagen-associated endometrial protein (PEAP) were identified, two crucial players in fetal tolerance [40,103,104]. Even though the outcomes following Mregs infusion in human transplant recipients are still unclear, they clearly point at the importance of donor-derived macrophages in orchestrating allograft rejection and at the necessity to intervene in this deleterious process before antigen presentation in the recipient occurs.
Another promising approach is the use of mediators secreted by macrophages during efferocytosis. In a model of collagen-induced arthritis, injection of the supernatant from macrophages that are eliminating apoptotic cells (SuperMApo) resulted in resolution of ongoing inflammation. This was supported by in vitro data showing that co-culturing APC with SuperMApo triggered their return to homeostasis downregulating the molecular expression of co-stimulatory molecules. Of note, this resolution of inflammation was antigen-specific, and animals treated with SuperMApo were still competent to reject skin allografts [105]. Pro-resolving mediators in chronic diseases are already in clinical trials to test their safety and efficacy for the signs and symptoms of dry eyes as well as treatment of gingivitis (NCT00799552 and NCT02342691, available on the clinicaltrial.gov website).

3.4. Tissue-Resident Macrophages (TRM)

Residing in different organs, TRM further highlight the complex heterogeneity of macrophages. Osteoclasts in the bone, Kupffer cells in the liver or microglia in the brain are tissue-specific macrophages with the specific function to maintain tissue homeostasis and normal organ function. In mice, TRM are generally considered to have immunosuppressive function for maintaining tissue homeostasis, inhibiting T-cell activation and resolving ongoing inflammatory processes [106,107]. As a result, macrophages identified in injured organs derive from two different sources, TRM and circulating monocytes [108]. Unique to allograft transplantation is the fact that donor TRM residing in the transplanted graft are transferred to the recipient.
In this regard, the role of TRM is not clearly defined. Still, there is important data pointing at a steering role of TRM. In a mouse cardiac transplantation model, a subset of TRM expressing high levels of CD169 and TIM4 was shown to migrate to the draining lymph nodes inducing antigen-stimulated Tregs and finally promoting cardiac allograft engraftment [109]. On the contrary, in the lung, severe IRI has been observed to trigger increased TLR4 expression on alveolar macrophages, resulting in increased endotoxin levels and neutrophil recruitment in the donor organ aggravating allograft injury [110,111].
These different outcomes could be related to different TRM populations present in distinct organs. In the human heart, TRM can be distinguished by the expression of CCR2. While CCR2+ macrophages derive from adult hematopoietic progenitor cells, depend on circulating monocytes for maintenance and are involved in pro-inflammatory processes recruiting regularly monocytes and neutrophils, CCR2- macrophages derive from embryogenic progenitors, are independent of circulating monocyte replenishment and promote tissue repair [112]. In a recently published study using a murine heart transplantation model, donor CCR2+ macrophages and MYD88 signaling were identified as crucial drivers of allograft rejection. Depletion of either donor CCR2+ macrophages or MYD88 signaling resulted in prolonged allograft survival, in contrast to the deletion of donor CCR2- macrophages which resulted in a more rapid allograft rejection and cellular infiltration. Of note, targeting recipient CCR2+ macrophages did not show any survival benefit and the application of CTL4-Ig immunosuppression preserved donor CCR2- but not CCR2+ macrophages [113]. Hence, similarly to the polarization of infiltrating macrophages, there also seems to exist a fragile equilibrium between TRM. The exponential development and use of ex vivo normothermic perfusion techniques could represent the ideal platform to further pursue TRM as treatment target [114,115].
Another aspect underlining the complexity of TRM is the role of phagocytosis-mediated clearance of either injured graft cells or infiltrating lymphocytes. During renal injury, renal TEC transform themselves into semi-professional phagocytes expressing kidney injury molecule 1 (KIM-1) and are capable of clearing apoptotic cells [116]. In a murine model of kidney transplantation, the improvement of the phagocytic activity from TEC by the administration of recombinant apoptosis inhibitor of macrophages (rAIM) marked-ly reduced the delay of graft function. A single dose of rAIM resulted in almost normal kidney function within 48 h post-transplantation with reduced tissue inflammation and necrotic debris [117]. In the same line, in a mouse liver transplantation model, the promotion of Kupffer cell clearance capacity from apoptotic T-cells resulted in a reduced rejection activity index. The improved liver graft function was associated with an M2 polarization of Kupffer cells triggered by PUFAs from degraded T-cells which in turn activates PPARγ [118].

4. Effect of Maintenance Immunosuppressive Agents on Macrophages

Lifelong immunosuppression constitutes the pillar of long-term success in solid organ transplantation; however, the agents used for maintenance immunosuppression are hampered by drug toxicity and adverse effects are harming patient as well as graft survival [7]. The effect of immunosuppressants on T-cells has been extensively studied in recent years as T-cells were assumed to be the main immunological contributors of transplant rejection. However, their effect on innate immune cells, especially macrophages, which increasingly move into the focus of transplant rejection, remains limited and poorly investigated. In this chapter, we summarize recent findings to gain a better insight into the effects of maintenance immunosuppressants that are currently used in clinics including the mammalian target of rapamycin (mTOR) inhibitor rapamycin, the two calcineurin inhibitors CsA and tacrolimus, mycophenolate mofetil (MMF), GC as well as belatacept on macrophages [119]. An overview about the main effects of these immunosuppressives can be found in Table 2.

4.1. mTOR Inhibitor (Rapamycin/Sirolimus)

Rapamycin, also known as sirolimus, is an immunosuppressive drug that inhibits mTOR, a serine-threonine kinase involved in cell growth, protein synthesis, proliferation and apoptosis [149]. mTOR inhibitors are known to suppress T-cell proliferation thereby influencing graft rejection [150].
Studies investigating the role of mTOR inhibition on macrophages in a severe combined immunodeficiency (SCID) model showed that treatment of T-/B-cell-deficient mice and immunocompetent C57Bl/6 mice with rapamycin as well as mTOR knockout decreased the levels of CD11b+ F4/80+ macrophages. This impaired macrophage/monocyte development during mTOR deficiency goes in hand with an overexpression of STAT5 and further downregulation of IRF8 that downregulates M-CSF/CD115 signaling which is important for the differentiation and proliferation of monocytes/macrophages [120].
Additionally, in a murine heart transplantation model with antibody-mediated rejection, mTOR inhibitors were found to attenuate macrophage infiltration as well as transplant allograft vasculopathy [121]. Furthermore, rapamycin treatment in human macrophages polarized to M1- and M2-like phenotypes was found to induce apoptosis exclusively of M2 macrophages and the cytokine profile of TLR4-stimulated PBMCs from rapamycin-treated patients prior islet transplantation showed a shift towards a M1-like macrophage phenotype [122]. Such influence on M2-like macrophages was also found in bone marrow-derived macrophages with mTOR deletion in which M2 polarization was inhibited. Macrophage-specific knockout of mTOR in a murine heart transplantation model had no influence on acute allograft rejection while additional treatment of mTOR-deficient mice with the immunosuppressive agent CTLA4-Ig inhibited chronic rejection and expanded Foxp3+ Tregs in allografts [123]. In another murine heart transplantation model, the treatment of an mTOR-specific high-density lipoprotein (HDL) was shown to prevent aerobic glycolysis in macrophages and epigenetic modifications for the production of inflammatory cytokines. Simultaneously, the development of Mregs was induced which inhibited alloreactive CD8+ T-cell proliferation and expanded immunosuppressive Foxp3-expressing Tregs [97]. A reduction in inflammatory cytokines by rapamycin was also found in vitro in a human monocyte cell line as well as in primary monocytes from humans, in which LPS-induced chemokine expression of monocyte chemoattractant protein 1 (MCP-1, CCL2), IL-8, RANTES (CCL5), macrophage inflammatory protein (MIP) 1α and MIP-1β was decreased after rapamycin treatment [124].
All these findings point to a strong effect of rapamycin on macrophages by impairing their development, inhibiting the polarization to a M2-like phenotype in the setting of chronic rejection, decreasing their release of inflammatory cytokines and boosting the formation of Mregs.

4.2. Calcineurin Inhibitors (Cyclosporine A and Tacrolimus)

Calcineurin inhibitors, such as CsA and tacrolimus, are immunosuppressive drugs that act through inhibition of the calcineurin pathway, which decreases expression of IL-2 and other cytokines in T-cells, inhibits NF-κB activation and impairs T-cell activation, proliferation and differentiation [151,152].
A consequence of tacrolimus treatment on macrophages was identified in a rat model of spinal cord injury where the treatment led to lower TNFα pro-inflammatory cytokine production and M1 marker expression than in controls [125]. Additionally, polarization of PBMCs indicated a shift to M2-like surface marker after tacrolimus treatment of blood samples while IL-1β production and phagocytosis function of macrophages were not altered [126]. Tacrolimus-treated IL-10-deficient mice presented with lower pro-inflammatory cytokine production, suppressed LPS-induced activation of both NF-κB and MAPK in peritoneal macrophages and induced apoptosis of macrophages via the activation of caspases 3 and 9 [127]. Furthermore, in a urinary tract infection model, tacrolimus pretreated mice displayed higher bacterial loads than control mice. This impaired host innate immune response after tacrolimus treatment was characterized with reduced TLR5 expression in bladder macrophages [153]. In accordance with these findings, macrophages treated with tacrolimus in vitro showed a decreased responsiveness to LPS induction and macrophages from tacrolimus-treated mice or mice with myeloid-specific calcineurin B1 (CnB1) knockout had diminished LPS-induced inflammatory responses [128]. Additionally, in human monocytes/macrophages, CsA reduced NF-κB activation and dose-dependently inhibited the LPS-induced procoagulant activity, which was identified as tissue factor activity associated with coronary lesions and fibrin deposition [129]. In a rat kidney transplantation model, CsA treatment led to increased macrophage infiltration and lower graft survival when compared to a CCR5/CXCR3 antagonist treatment [154]. Moreover, such an accumulated macrophage influx together with increased interstitial fibrosis was seen in kidneys of rats suffering from CsA-caused nephrotoxicity [155].
Comparison of CsA and tacrolimus in rat renal allografts indicated that tacrolimus treatment improved changes in acute and chronic rejection and additionally inhibited the fibrogenic PDGF, while moderate to intense histological changes as well as an induction of PDGF were identified upon CsA treatment [130]. Consistent with this finding, renal biopsies from CsA-treated patients during chronic rejection showed increased tubulointerstitial CD68-positive macrophages together with a lower graft survival rate compared to tacrolimus-treated biopsies [131]. A clinical trial also showed that CsA treatment led to increased interstitial fibrosis as well as total cholesterol and low-density lipoprotein (LDL) levels while tacrolimus treatment showed a trend for insulin resistance after renal transplantation. However, no difference in the incidence of acute rejection in both treatments were identified [132].
In general, both calcineurin inhibitors affect macrophages by decreasing their NF-κB pathway activation and response to LPS with tacrolimus leading to a decrease in M1 surface markers with a shift to a M2-like phenotype and reduced production of pro-inflammatory cytokines. Of note, in the setting of chronic rejection, CsA treatment is characterized by increased macrophage infiltration and fibrosis compared to tacrolimus.

4.3. Mycophenolate Mofetil (MMF)

The immunosuppressive compound mycophenolate mofetil (MMF) is the ester prodrug of mycophenolic acid (MPA), an inhibitor of the enzyme inosine-5′-monophosphate dehydrogenase which is involved in nucleotide synthesis. MPA was shown to deplete guanosin nucleotides in T- and B-cells thereby inhibiting their proliferation and suppressing cell-mediated immune responses [156].
A possible effect of MPA on macrophages has already been assumed in inhibited synthesis of BH4, which is built from guanosine triphosphate (GTP). BH4 is the cofactor of several enzymes including iNOS, an enzyme involved in the inflammatory response of macrophages [157].
In MRL/lpr mice, a genetic model of generalized autoimmune disease, the treatment of dextran mycophenolate-based nanoparticles lowered the levels of pro-inflammatory mediators and reduced kidney injury. Additionally, these MPA nanoparticles were found to be phagocytosed in the spleen and kidney by macrophages which were characterized as CD206+ M2-like phenotypes with downregulated CD80 and CD40 surface markers and reduced TNFα production [133]. Likewise, treatment of PBMCs with MPA was also shown to have no influence on the phagocytosis function of macrophages but IL-1β production was reduced to 50% and an increase in M2 surface markers such as CD163 and CD200R on M1-polarized macrophages could be identified [126]. In addition, LPS-stimulated monocytes derived from renal transplant recipients that were treated with MMF showed decreased secretion of IL-1β together with lower IL-10 and TNFα secretion, reduced TNF-R1 expression and suppressed antigen presenting capacity [134]. Moreover, MMF-treated human monocytes presented with reduced binding to endothelial cells, inhibited upregulation of intracellular adhesion molecule 1 (ICAM-1) and decreased MHC-II expression on monocytes that were stimulated with either LPS or IFNγ [135].
Overall, MPA influences macrophages by promoting an M2-like macrophage phenotype, decreasing pro-inflammatory mediators and lowering the interactions of monocytes/macrophages and endothelial cells.

4.4. Glucocorticoids (GC)

GC are steroid hormones that are widely used due to their anti-inflammatory and immunomodulatory properties. They are involved, e.g., in the stabilization of lysosomal membranes, the suppression of prostaglandin synthesis and reduction in histamine release [150,158]. GC bind to their glucocorticoid receptor (GR), resulting in inhibition of transcription and production of several pro-inflammatory cytokines such as IL-1, IL-2, IL-6, IFNγ and TNFα by several complex steps [150].
A possible influence of GC on macrophages was speculated decades ago due to a side effect of GC treatment leading to reduced bone density where GC were found to directly act on bone marrow-resident macrophages, namely osteoclasts [60,159]. In a mouse model of contact allergy, the ablation of the GR in macrophages abolished downregulation of the inflammatory cytokines and chemokines such as IL-1b, MCP1, MIP2 and IFNγ-inducible protein 10 (IP10) by GC while its inactivation in keratinocytes or T-cells did not attenuate the effects of GC [136]. In the same line, macrophage-specific GR-deletion in a mouse model of induced colitis presented with increased macrophage infiltration at inflammatory sites in the colon as well as pro-inflammatory cytokine expression while expression of scavenger receptors and IL-10 were diminished [160]. Treatment of the macrophage cell line J774 with dexamethasone or hydrocortisone was found to cause a concentration-dependent inhibition of NO formation, suggesting an inhibition of nitric oxide synthases [137]. In contrast, the administration of corticoids during transient cerebral ischemia were found to increase the enzymatic activity of endothelial nitric oxide synthase [161]. However, in macrophages a dose-dependent effect of GC on NO production characterized by enhanced NO production after administration of low corticosterone levels concomitant with increased expression of pro-inflammatory cytokines and enzymes for mediator synthesis were identified while high doses of corticosterone strongly repressed macrophage functions [138]. In human kidney allografts with antibody-mediated or T-cell-mediated rejection, steroid treatment increased macrophage infiltration into the allograft, which was additionally accompanied with increased cell proliferation and antigen presentation [139]. Furthermore, prednisolone treatment of mice with mesangial proliferative glomerulonephritis was found to induce a M2-like macrophage phenotype and intensified glomerulosclerosis [140]. Similarly, GC treatment in human monocyte-derived macrophages induced differentiation of a specifically activated, anti-inflammatory subtype which enhances phagocytosis and motility as well as repression of adhesion, apoptosis, and oxidative burst [141]. In addition to the effect of GC to promote phagocytosis, they were also identified to enhance bacterial killing in vitro [142] and enhance CD163 expression and production of TGFβ1, fibroblast growth factor 2 (FGF-2), connective tissue growth factor (CTGF) indicating that steroids mimic a pro-fibrotic phenotype [143].
In summary, GC act on macrophages by enhancing pro-fibrotic, M2-like macrophage phenotypes and promoting phagocytosis while downregulating pro-inflammatory cytokines.

4.5. Belatacept

Belatacept is a second-generation fusion protein combining the extracellular domain of the human cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) with the Fc domain of human immunoglobulin G1 (CTLA4-Ig). It binds to the co-stimulatory molecules CD80 and CD86 on APCs thereby preventing T-cell binding to these molecules and inhibiting T-cell activation [162]. There is an obvious link between belatacept and macrophages as this compound blocks CD80/86 co-stimulatory molecules on APCs, which also include macrophages [163]. A study of renal transplant biopsies from patients treated with either calcineurin inhibitor or belatacept indicated that co-stimulatory blockade with belatacept was positively associated with genes related to innate immune responses including receptors of phagocytes (e.g., CD16, CD14, CD68, CD209, and TLR4) [144]. However, the effect of belatacept on macrophages is not well investigated. Nevertheless, there is evidence that CTLA4-Ig are affecting macrophages, which was already shown for abatacept, a first-generation CTLA4-Ig that is frequently used in the treatment of patients with rheumatoid arthritis. Bonelli and colleagues could show that CD14+ monocytes isolated from human PBMCs showed reduced migration capacity after treatment with abatacept [145]. Additionally, abatacept was found to reduce the production of pro-inflammatory cytokines such as TNFα, IL-12 and IFNγ in human macrophages generated from PBMCs [146]. Similarly, Cutolo and colleagues detected decreased IL-6, TNFα, IL-1β and TGFβ cytokine production in synovial macrophages from patients with rheumatoid arthritis compared to controls [147]. A recent study by Cutolo and colleagues could also show in monocyte-derived macrophages from peripheral blood that abatacept treatment in vitro induced the shift from M1 macrophages to M2 macrophages in LPS-induced macrophages from healthy subjects as well as macrophages obtained from patients with rheumatoid arthritis [148]. In general, an effect of belatacept on macrophages can be assumed as belatacept differs from its first-generation variant abatacept in two amino acid substitutions thereby leading to a higher affinity in binding to the co-stimulatory molecules [164].

5. BH4 as Immunosuppressive Player in Transplantation

In the last decade, we and others described BH4 in several animal studies to modulate the immune response in the setting of allograft transplantation. BH4 is a vitamin-like compound produced by probably almost every cell in higher organisms. It is an essential cofactor for a group of eight enzymes including (i) the three nitric oxide synthases (NOS), i.e., neuronal NOS (nNOS), inducible NOS (iNOS) and endothelial NOS (eNOS); (ii) four aromatic amino acid hydroxylases, i.e., tryptophan hydroxylases (TPH) 1 and 2, phenylalanine hydroxylase (PAH) and tyrosine hydroxylase (TH); and (iii) alkylglycerol monooxygenase (AGMO). These enzymes play a key role in several processes linked to cardiovascular function, neurotransmitter function and immune responses. Biosynthesis of BH4 occurs either via a de novo pathway requiring three enzymes, i.e., GTP-cyclohydrolase 1 (GCH1), 6-pyruvoyl tetrahydropterin synthase (PTPS) and sepiapterin reductase (SR), with GCH1 representing the rate-limiting enzyme or via a salvage pathway. In the latter, SR converts sepiapterin to 7,8-dihydrobiopterin which is finally transformed to BH4 by dihydrofolate reductase (DHFR) [165]. Several studies on IRI in transplantation and non-transplantation models observed a critical role of BH4 in preventing severe injury and loss of organ function. In a Langendorff perfused heart model, the endothelial-specific knockout of GCH1 led to a loss of BH4 in endothelial cells, resulting in endothelial dysfunction and increased cardiac infarct size following ischemic injury [166]. In the same line, supplementation with BH4 or its precursor sepiapterin reduced ischemic injury in rat models of cardiac infarction [167,168]. Implementation to a transplantation model revealed a similar protective effect in a murine pancreas transplantation model. Comparison with other pteridin-analoga showed that the protective effect was not due to the antioxidative capacity of BH4 but rather BH4-specific [169] and using knockout mice of the three different NOS isoforms revealed nNOS as BH4 target [170]. Similarly, treating the donor animal with BH4 resulted in a significant reduction in neointima formation in an aortic transplantation model. In this setting, BH4 supplementation was suggested to target eNOS preventing its uncoupling and the subsequent production of superoxide [171]. The importance of having saturated BH4 already before the major injury occurs was also observed in two ischemia preconditioning models where the increased expression of BH4 was clearly related to the protective effects of ischemic preconditioning [172,173].
With different groups describing a crucial role of iNOS in allograft transplantation, we tested in a murine allograft transplantation model the selective iNOS inhibitor 4-amino-BH4 (4-ABH4), the NOS-cofactor BH4 as well as the non-pterin inhibitor of iNOS N-(iminoethyl)-L-lysine (L-NIL). Only the two pteridines 4-ABH4 and BH4 resulted in prevention of allograft rejection while L-NIL did not affect the immune response, suggesting a pteridin-specific, iNOS-independent immunosuppressive effect of 4-ABH4 and BH4 [174]. Confirming this observation, overexpression of GCH1 was shown by another group to prevent allograft rejection pointing at an immunosuppressive role of pteridin compounds [175]. Of note, in this first publication demonstrating an immunosuppressive effect of BH4 in an acute rejection model, the microarray data showed that the repressed intragraft gene expression of CD163 during acute rejection was reversed by BH4 treatment [174]. This different expression in the M2 macrophage marker suggested that the effect might be related to the modulation of the innate immune system. In a follow-up study aimed at further dissecting the role of BH4 on the immune response during acute allograft rejection, we observed on day six following transplantation in BH4-treated animals increased levels of M2-related cytokines IL-10 and IL-4 in the graft as well as in the serum of the recipients. Of note, this was accompanied by a prominent mobilization of CD4+ CD25+ FoxP3+ Tregs in the spleen, a T-lymphocyte subpopulation known to be induced by alternatively activated macrophages (unpublished data). In the context of macrophages, BH4 has been mostly associated with iNOS since induction of this BH4-dependent enzyme is closely related to proinflammatory, classically activated macrophages [176]. However, a recent study using BH4-deficient Gch1fl/flTie2cre macrophages described BH4-dependent immune alterations that were not iNOS dependent. In particular, gene expression associated with the redox-sensitive transcription factor nuclear factor erythroid 2-related factor 2 (NRF2) were significantly reduced in BH4-deficient macrophages compared to wild-type ones [177]. Similarly, BH4-deficient Gch1fl/flTie2cre mice showed lower lung mycobacterial load following mycobacterial infection compared to wild-type animals, suggesting an anti-inflammatory role of BH4. Additionally, in this model, the effect could not be reproduced in mice lacking iNOS protein. In vitro quantification of mycobacterial growth confirmed the enhanced control of Gch1fl/flTie2cre macrophages compared to wild-type and iNOS-deficient cells [178].
Since iNOS, the hallmark molecule of M1 macrophages, has already been excluded as BH4 target in the context of acute rejection, the BH4-associated immunosuppressive effects might be related to another BH4-dependent enzyme, the recently described AGMO. AGMO is a lipid-cleaving enzyme that degrades ether lipids (especially alkylglycerols and lyso-alkylglycerophospholipids) into a free glycerol and a toxic fatty aldehyde, which is immediately converted to the corresponding fatty acid [179]. Ether lipids are a class of less well-studied lipids that harbor an ether bond at the sn-1 position of the glycerol backbone compared to the commonly known ester lipids. Similar to their ester counterparts, ether lipids can also be found in biological membranes and can act as immune-related signaling molecules [180,181]. For AGMO, the (patho)physiological role has not yet been fully elucidated; however, there is increasing evidence that AGMO is involved in immunological processes [182]. In general, AGMO gene expression was found to be upregulated in M2 macrophages while it is strongly downregulated in M1 macrophages [183]. Of note, the rate-limiting enzyme in BH4 biosynthesis, GCH1, is inversely regulated as it is upregulated in M1 (most probably to boost iNOS) but not in M2 macrophages [183] potentially causing suboptimal AGMO turnover rates. This assumption is also supported by observations in rat livers where iNOS had an about 200-fold higher affinity to BH4 (Km between 0.02 and 0.3 µM) compared to AGMO (Km = 2.6 µM), which had a similar affinity to BH4 such as PAH (Km = 2 µM) [184,185]. In this context, BH4 treatment in the acute rejection model might fuel AGMO in M2 macrophages leading to increased lipid degradation. Such an upregulation of a lipid-cleaving enzyme would also fit to the already described metabolism of M2 macrophages in which enhanced lipolysis leads to increased OXPHOS and FAO activating M2 macrophages [38]. An overview about the immunosuppressive effect of BH4 and its targets in transplantation can be seen in Figure 1.
Last but not least, treatment with the orally active synthetic BH4-analogon sapro-pterin dihydrochloride has been already approved by the FDA (US Food and Drug Administration) and the EMA (European Medicines Agency) for patients with BH4-deficient phenylketonuria [186]. Unraveling novel mechanisms of a compound which is already in clinical use makes BH4-related immunosuppression an appealing strategy. Repurposing of a clinically used drug would eliminate time-consuming pharmacological development and safety testing required for novel drugs.

6. Conclusions

In recent years, a substantial amount of knowledge has been gathered regarding the role of macrophages in organ transplantation. The observation that these cells occur in different phenotypes displaying either pro- or anti-inflammatory properties makes them an appealing target to prevent acute allograft rejection or even induce graft tolerance. So far, one might only speculate on the factors affecting polarization of infiltrating or resident macrophages during acute allograft rejection. Considering, however, the different injuries an organ encounters already before the implantation procedure and then during the allograft recognition processes the ultimate amount of damage itself as well as the time point of counteracting these damaging factors might be crucial for the balance between pro- and anti-inflammatory macrophages.
In this regard, a better understanding of the effects of currently used maintenance immunosuppressive agents on macrophage polarization might result in the development of newer treatment protocols. Dosages as well as different application time points could be based not only on their effect of infiltrating lymphocytes but also on their effect on plasticity of TRM and infiltrating macrophages.
In addition, with lifelong immunosuppression still jeopardizing long-term graft and recipient survival due to drug toxicities and adverse effects, the development of new immunosuppressive agents addressing innate immune cells are urgently needed. We propose herein BH4 as a promising candidate. Administration of this vitamin-like compound has already been shown to prevent acute allograft rejection, and there is increasing evidence that its immunomodulatory role is not associated with iNOS activity but presumably with another BH4-dependent enzyme expressed in macrophages, AGMO. Both these BH4-dependent enzymes are abundantly found in activated macrophages. However, they are differently upregulated, iNOS in classically and AGMO in alternatively activated macrophage phenotypes. Successfully addressing the balance between different phenotypes by BH4 supplementation might be the starting point for a new generation of immunomodulatory drugs.

Author Contributions

Writing—original draft preparation, K.L., S.E., K.W. and M.M.; writing—review and editing, K.L., S.E., K.W. and M.M. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the “In memoriam Dr. Gabriel Salzner Stiftung” to M.M. The funding institute was not involved in study design, manuscript preparation or publication decisions.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The authors would like to thank Jakob Troppmair for constructive criticism of the manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

4-ABH4, 4-amino-BH4; A2R, A2 adenosine receptor; AGMO, alkylglycerol monooxygenase; APC, antigen presenting cell; ARG, arginase; BH4, tetrahydrobiopterin; CBMPs, cell-based medicinal products; CnB1, calcineurin B1; COX, cyclooxygenase; CsA, CTLA-4, cytotoxic T-lymphocyte-associated protein 4; cyclosporine A; CTGF, connective tissue growth factor; DAMPs, damage-associated molecular patterns; DHFR, dihydrofolate reductase; eNOS, endothelial nitric oxide synthase; FAO, fatty acid oxidation; FAS, fatty acid synthesis; FGF-2, fibroblast growth factor 2; GC, glucocorticoids; GCH1, GTP-cyclohydrolase 1; GR, glucocorticoid receptor; GTP, guanosine triphosphate; HDL, high-density lipoprotein; HIF1α, hypoxia-inducible factor 1α; ICAM-1, intracellular adhesion molecule 1; ICs, immune complexes; IDO, indoleamine-2,3-dioxygenase; IFN, interferon; IGF, insulin-like growth factor; IL, interleukin; iNOS, inducible nitric oxide synthase; IP10, IFNγ-inducible protein 10; IRF, interferon-regulatory factor; IRI, ischemia reperfusion injury; KIM-1, kidney injury molecule; LDL, low-density lipoprotein; L-NIL, N-(iminoethyl)-L-lysine; LPS, lipopolysaccharides; MCP-1 (CCL2), monocyte chemoattractant protein 1; M-CSF, macrophage colony-stimulating factor; MIP, macrophage inflammatory protein; MMF, mycophenolate mofetil; MPA, mycophenolic acid; Mregs, regulatory macrophages; MSC, mesenchymal stem cells; mTOR, mammalian target of rapamycin; NF-κB, nuclear factor kappa B; NO, nitric oxide; nNOS, neuronal nitric oxide synthase; NRF2, nuclear factor erythroid 2-related factor 2; OXPHOS, oxidative phosphorylation; PAH, phenylalanine hydroxylase; PBMC, peripheral blood mononuclear cell; PDGF, platelet-derived growth factor; PEAP, progestagen-associated endometrial protein; PIR-A, paired immunoglobulin-like receptors-A; PPAR, peroxisome proliferator-activated receptor; PPP, pentose phosphate pathway; PRR, pattern recognition receptors; PTPS, 6-pyruvoyl tetrahydropterin synthase; PUFA, polyunsaturated fatty acid; rAIM, recombinant apoptosis inhibitor of macrophages; RNS, reactive nitrogen species; ROS, reactive oxygen species; SCID, severe combined immunodeficiency; sFgl2, soluble fibrinogen-like protein 2; SOCS, suppressor of cytokine signaling; SR, sepiapterin reductase; STAT, signal transducer and activator of transcription; TAMs, tumor-associated macrophages; TEC, tubular epithelial cell; TGF, transforming growth factor; TH, tyrosine hydroxylase; TIGIT, T-cell immunoreceptor with Ig and ITIM domains; TLR, toll-like receptors; TNF, tumor necrosis factor; TPH, tryptophan hydroxylase; TRM, tissue-resident macrophages; Treg, regulatory T-cells; VEGF, vascular endothelial growth factor.

References

  1. Guo, Z.; Zhao, Q.; Jia, Z.; Huang, C.; Wang, D.; Ju, W.; Zhang, J.; Yang, L.; Huang, S.; Chen, M.; et al. A Randomized-Controlled Trial of Ischemia-Free Liver Transplantation for End-Stage Liver Disease. J. Hepatol. 2023. [Google Scholar] [CrossRef] [PubMed]
  2. Nasralla, D.; Coussios, C.C.; Mergental, H.; Akhtar, M.Z.; Butler, A.J.; Ceresa, C.D.L.; Chiocchia, V.; Dutton, S.J.; García-Valdecasas, J.C.; Heaton, N.; et al. A Randomized Trial of Normothermic Preservation in Liver Transplantation. Nature 2018, 557, 50–56. [Google Scholar] [CrossRef] [PubMed]
  3. Kumar, L.; Sahu, S.; Deo, A.S.; Selvakumar, R.; Panchwag, A.A.; Pavithran, P. Recent Advances in Anaesthesia for Abdominal Solid Organ Transplantation. Indian J. Anaesth. 2023, 67, 32–38. [Google Scholar] [CrossRef] [PubMed]
  4. Chan, S.; Ng, S.; Chan, H.P.; Pascoe, E.M.; Playford, E.G.; Wong, G.; Chapman, J.R.; Lim, W.H.; Francis, R.S.; Isbel, N.M.; et al. Perioperative Antibiotics for Preventing Post-Surgical Site Infections in Solid Organ Transplant Recipients. Cochrane Database Syst. Rev. 2020, 8, CD013209. [Google Scholar] [CrossRef]
  5. Calne, R.Y.; Rolles, K.; Thiru, S.; Mcmaster, P.; Craddock, G.N.; Aziz, S.; White, D.J.G.; Evans, D.B.; Dunn, D.C.; Henderson, R.G.; et al. Cyclosporin a Initially as the Only Immunosuppressant in 34 Recipients of Cadaveric Organs: 32 Kidneys, 2 Pancreases, and 2 Livers. Lancet 1979, 314, 1033–1036. [Google Scholar] [CrossRef]
  6. Gordon, R.D.; Shaw, B.W., Jr.; Iwatsuki, S.; Esquivel, C.O.; Starzl, T.E. Indications for Liver Transplantation in the Cyclosporine Era. Surg. Clin. N. Am. 1986, 66, 541–556. [Google Scholar] [CrossRef]
  7. Lodhi, S.A.; Lamb, K.E.; Meier-Kriesche, H.U. Solid Organ Allograft Survival Improvement in the United States: The Long-Term Does Not Mirror the Dramatic Short-Term Success. Am. J. Transplant 2011, 11, 1226–1235. [Google Scholar] [CrossRef]
  8. Tustumi, F.; de Miranda Neto, A.A.; Silveira Júnior, S.; Fernandes, F.A.; de Brito e Silva, M.B.; Ernani, L.; Nacif, L.S.; Coelho, F.F.; Andraus, W.; Bernardo, W.M.; et al. Safety and Effectiveness of Mycophenolate Mofetil Associated with Tacrolimus for Liver Transplantation Immunosuppression: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Clinics 2021, 76, e2597. [Google Scholar] [CrossRef]
  9. Nguyen, L.S.; Vautier, M.; Allenbach, Y.; Zahr, N.; Benveniste, O.; Funck-Brentano, C.; Salem, J.-E. Sirolimus and mTOR Inhibitors: A Review of Side Effects and Specific Management in Solid Organ Transplantation. Drug Saf. 2019, 42, 813–825. [Google Scholar] [CrossRef]
  10. Peters, D.H.; Fitton, A.; Plosker, G.L.; Faulds, D. Tacrolimus. A Review of Its Pharmacology, and Therapeutic Potential in Hepatic and Renal Transplantation. Drugs 1993, 46, 746–794. [Google Scholar] [CrossRef]
  11. Scott, L.J.; McKeage, K.; Keam, S.J.; Plosker, G.L. Tacrolimus: A Further Update of Its Use in the Management of Organ Transplantation. Drugs 2003, 63, 1247–1297. [Google Scholar] [CrossRef] [PubMed]
  12. Dharnidharka, V.R.; Lamb, K.E.; Zheng, J.; Schechtman, K.B.; Meier-Kriesche, H.-U. Lack of Significant Improvements in Long-Term Allograft Survival in Pediatric Solid Organ Transplantation: A US National Registry Analysis. Pediatr. Transplant. 2015, 19, 477–483. [Google Scholar] [CrossRef] [PubMed]
  13. Wekerle, T.; Segev, D.; Lechler, R.; Oberbauer, R. Strategies for Long-Term Preservation of Kidney Graft Function. Lancet 2017, 389, 2152–2162. [Google Scholar] [CrossRef] [PubMed]
  14. Bamoulid, J.; Staeck, O.; Halleck, F.; Khadzhynov, D.; Brakemeier, S.; Dürr, M.; Budde, K. The Need for Minimization Strategies: Current Problems of Immunosuppression. Transpl. Int. 2015, 28, 891–900. [Google Scholar] [CrossRef] [PubMed]
  15. Hennessy, C.; Lewik, G.; Cross, A.; Hester, J.; Issa, F. Recent Advances in Our Understanding of the Allograft Response. Fac. Rev. 2021, 10, 21. [Google Scholar] [CrossRef] [PubMed]
  16. Short, S.; Lewik, G.; Issa, F. An Immune Atlas of T Cells in Transplant Rejection: Pathways and Therapeutic Opportunities. Transplantation 2023. [Google Scholar] [CrossRef] [PubMed]
  17. Hall, B.M.; Dorsch, S.; Roser, B. The Cellular Basis of Allograft Rejection in Vivo. I. The Cellular Requirements for First-Set Rejection of Heart Grafts. J. Exp. Med. 1978, 148, 878–889. [Google Scholar] [CrossRef] [PubMed]
  18. Bradley, J.A.; Sarawar, S.R.; Porteous, C.; Wood, P.J.; Cård, S.; Ager, A.; Bolton, E.M.; Bell, E.B. Allograft Rejection in CD4+ T Cell-Reconstituted Athymic Nude Rats—The Nonessential Role of Host-Derived CD8+ Cells. Transplantation 1992, 53, 477–482. [Google Scholar] [CrossRef] [PubMed]
  19. Halloran, P.F. Immunosuppressive Drugs for Kidney Transplantation. N. Engl. J. Med. 2004, 351, 2715–2729. [Google Scholar] [CrossRef]
  20. Hancock, W.W.; Thomson, N.M.; Atkins, R.C. Composition of Interstitial Cellular Infiltrate Identified by Monoclonal Antibodies in Renal Biopsies of Rejecting Human Renal Allografts. Transplantation 1983, 35, 458–463. [Google Scholar] [CrossRef]
  21. Salehi, S.; Reed, E.F. The Divergent Roles of Macrophages in Solid Organ Transplantation. Curr. Opin. Organ Transplant. 2015, 20, 446–453. [Google Scholar] [CrossRef] [PubMed]
  22. Yu, S.; Lu, J. Macrophages in Transplant Rejection. Transpl. Immunol. 2022, 71, 101536. [Google Scholar] [CrossRef] [PubMed]
  23. McLean, A.G.; Hughes, D.; Welsh, K.I.; Gray, D.W.; Roake, J.; Fuggle, S.V.; Morris, P.J.; Dallman, M.J. Patterns of Graft Infiltration and Cytokine Gene Expression during the First 10 Days of Kidney Transplantation. Transplantation 1997, 63, 374–380. [Google Scholar] [CrossRef] [PubMed]
  24. Lucas-Ruiz, F.; Mateo, S.V.; Jover-Aguilar, M.; Alconchel, F.; Martínez-Alarcón, L.; de Torre-Minguela, C.; Vidal-Correoso, D.; Villalba-López, F.; López-López, V.; Ríos-Zambudio, A.; et al. Danger Signals Released during Cold Ischemia Storage Activate NLRP3 Inflammasome in Myeloid Cells and Influence Early Allograft Function in Liver Transplantation. eBioMedicine 2023, 87, 104419. [Google Scholar] [CrossRef]
  25. Xu, H.; Dhanireddy, K.K.; Kirk, A.D. Human Monocytes as Intermediaries between Allogeneic Endothelial Cells and Allospecific T Cells: A Role for Direct Scavenger Receptor-Mediated Endothelial Membrane Uptake in the Initiation of Alloimmunity. J. Immunol. 2006, 176, 750–761. [Google Scholar] [CrossRef]
  26. Gordan, S.; Biburger, M.; Nimmerjahn, F. bIgG Time for Large Eaters: Monocytes and Macrophages as Effector and Target Cells of Antibody-Mediated Immune Activation and Repression. Immunol. Rev. 2015, 268, 52–65. [Google Scholar] [CrossRef]
  27. 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]
  28. Blériot, C.; Chakarov, S.; Ginhoux, F. Determinants of Resident Tissue Macrophage Identity and Function. Immunity 2020, 52, 957–970. [Google Scholar] [CrossRef]
  29. Hoeffel, G.; Wang, Y.; Greter, M.; See, P.; Teo, P.; Malleret, B.; Leboeuf, M.; Low, D.; Oller, G.; Almeida, F.; et al. Adult Langerhans Cells Derive Predominantly from Embryonic Fetal Liver Monocytes with a Minor Contribution of Yolk Sac-Derived Macrophages. J. Exp. Med. 2012, 209, 1167–1181. [Google Scholar] [CrossRef]
  30. Davies, L.C.; Jenkins, S.J.; Allen, J.E.; Taylor, P.R. Tissue-Resident Macrophages. Nat. Immunol. 2013, 14, 986–995. [Google Scholar] [CrossRef]
  31. Varol, C.; Mildner, A.; Jung, S. Macrophages: Development and Tissue Specialization. Annu. Rev. Immunol. 2015, 33, 643–675. [Google Scholar] [CrossRef] [PubMed]
  32. McGaha, T.L.; Chen, Y.; Ravishankar, B.; van Rooijen, N.; Karlsson, M.C.I. Marginal Zone Macrophages Suppress Innate and Adaptive Immunity to Apoptotic Cells in the Spleen. Blood 2011, 117, 5403–5412. [Google Scholar] [CrossRef] [PubMed]
  33. Louie, D.A.P.; Liao, S. Lymph Node Subcapsular Sinus Macrophages as the Frontline of Lymphatic Immune Defense. Front. Immunol. 2019, 10, 347. [Google Scholar] [CrossRef] [PubMed]
  34. Zhao, Y.; Zou, W.; Du, J.; Zhao, Y. The Origins and Homeostasis of Monocytes and Tissue-Resident Macrophages in Physiological Situation. J. Cell. Physiol. 2018, 233, 6425–6439. [Google Scholar] [CrossRef]
  35. Nathan, C.F.; Murray, H.W.; Wiebe, M.E.; Rubin, B.Y. Identification of Interferon-Gamma as the Lymphokine That Activates Human Macrophage Oxidative Metabolism and Antimicrobial Activity. J. Exp. Med. 1983, 158, 670–689. [Google Scholar] [CrossRef]
  36. Stein, M.; Keshav, S.; Harris, N.; Gordon, S. Interleukin 4 Potently Enhances Murine Macrophage Mannose Receptor Activity: A Marker of Alternative Immunologic Macrophage Activation. J. Exp. Med. 1992, 176, 287–292. [Google Scholar] [CrossRef]
  37. Mills, C.D.; Kincaid, K.; Alt, J.M.; Heilman, M.J.; Hill, A.M. M-1/M-2 Macrophages and the Th1/Th2 Paradigm. J. Immunol. 2000, 164, 6166–6173. [Google Scholar] [CrossRef]
  38. Liu, Y.; Xu, R.; Gu, H.; Zhang, E.; Qu, J.; Cao, W.; Huang, X.; Yan, H.; He, J.; Cai, Z. Metabolic Reprogramming in Macrophage Responses. Biomark Res. 2021, 9, 1. [Google Scholar] [CrossRef]
  39. Riquelme, P.; Tomiuk, S.; Kammler, A.; Fändrich, F.; Schlitt, H.J.; Geissler, E.K.; Hutchinson, J.A. IFN-γ-Induced iNOS Expression in Mouse Regulatory Macrophages Prolongs Allograft Survival in Fully Immunocompetent Recipients. Mol. Ther. 2013, 21, 409–422. [Google Scholar] [CrossRef]
  40. Riquelme, P.; Haarer, J.; Kammler, A.; Walter, L.; Tomiuk, S.; Ahrens, N.; Wege, A.K.; Goecze, I.; Zecher, D.; Banas, B.; et al. TIGIT iTregs Elicited by Human Regulatory Macrophages Control T Cell Immunity. Nat. Commun. 2018, 9, 2858. [Google Scholar] [CrossRef]
  41. Martinez, F.O.; Sica, A.; Mantovani, A.; Locati, M. Macrophage Activation and Polarization. Front. Biosci. 2008, 13, 453–461. [Google Scholar] [CrossRef] [PubMed]
  42. Riquelme, P.; Amodio, G.; Macedo, C.; Moreau, A.; Obermajer, N.; Brochhausen, C.; Ahrens, N.; Kekarainen, T.; Fändrich, F.; Cuturi, C.; et al. DHRS9 Is a Stable Marker of Human Regulatory Macrophages. Transplantation 2017, 101, 2731–2738. [Google Scholar] [CrossRef] [PubMed]
  43. Wuest, S.J.A.; Crucet, M.; Gemperle, C.; Loretz, C.; Hersberger, M. Expression and Regulation of 12/15-Lipoxygenases in Human Primary Macrophages. Atherosclerosis 2012, 225, 121–127. [Google Scholar] [CrossRef] [PubMed]
  44. Snodgrass, R.G.; Zezina, E.; Namgaladze, D.; Gupta, S.; Angioni, C.; Geisslinger, G.; Lütjohann, D.; Brüne, B. A Novel Function for 15-Lipoxygenases in Cholesterol Homeostasis and CCL17 Production in Human Macrophages. Front. Immunol. 2018, 9, 1906. [Google Scholar] [CrossRef] [PubMed]
  45. Golden, T.N.; Venosa, A.; Gow, A.J. Cell Origin and iNOS Function Are Critical to Macrophage Activation Following Acute Lung Injury. Front. Pharmacol. 2021, 12, 761496. [Google Scholar] [CrossRef]
  46. Rückerl, D.; Campbell, S.M.; Duncan, S.; Sutherland, T.E.; Jenkins, S.J.; Hewitson, J.P.; Barr, T.A.; Jackson-Jones, L.H.; Maizels, R.M.; Allen, J.E. Macrophage Origin Limits Functional Plasticity in Helminth-Bacterial Co-Infection. PLoS Pathog. 2017, 13, e1006233. [Google Scholar] [CrossRef]
  47. Cambier, C.J.; O’Leary, S.M.; O’Sullivan, M.P.; Keane, J.; Ramakrishnan, L. Phenolic Glycolipid Facilitates Mycobacterial Escape from Microbicidal Tissue-Resident Macrophages. Immunity 2017, 47, 552–565.e4. [Google Scholar] [CrossRef]
  48. 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]
  49. Sironi, M.; Martinez, F.O.; D’Ambrosio, D.; Gattorno, M.; Polentarutti, N.; Locati, M.; Gregorio, A.; Iellem, A.; Cassatella, M.A.; Van Damme, J.; et al. Differential Regulation of Chemokine Production by Fcgamma Receptor Engagement in Human Monocytes: Association of CCL1 with a Distinct Form of M2 Monocyte Activation (M2b, Type 2). J. Leukoc. Biol. 2006, 80, 342–349. [Google Scholar] [CrossRef]
  50. Rőszer, T. Understanding the Mysterious M2 Macrophage through Activation Markers and Effector Mechanisms. Mediators Inflamm. 2015, 2015, 816460. [Google Scholar] [CrossRef]
  51. Duluc, D.; Delneste, Y.; Tan, F.; Moles, M.-P.; Grimaud, L.; Lenoir, J.; Preisser, L.; Anegon, I.; Catala, L.; Ifrah, N.; et al. Tumor-Associated Leukemia Inhibitory Factor and IL-6 Skew Monocyte Differentiation into Tumor-Associated Macrophage-like Cells. Blood 2007, 110, 4319–4330. [Google Scholar] [CrossRef] [PubMed]
  52. Chen, S.; Lakkis, F.G.; Li, X.C. The Many Shades of Macrophages in Regulating Transplant Outcome. Cell. Immunol. 2020, 349, 104064. [Google Scholar] [CrossRef] [PubMed]
  53. Murray, P.J.; Allen, J.E.; Biswas, S.K.; Fisher, E.A.; Gilroy, D.W.; Goerdt, S.; Gordon, S.; Hamilton, J.A.; Ivashkiv, L.B.; Lawrence, T.; et al. Macrophage Activation and Polarization: Nomenclature and Experimental Guidelines. Immunity 2014, 41, 14–20. [Google Scholar] [CrossRef] [PubMed]
  54. Sica, A.; Mantovani, A. Macrophage Plasticity and Polarization: In Vivo Veritas. J. Clin. Investig. 2012, 122, 787–795. [Google Scholar] [CrossRef]
  55. Chávez-Galán, L.; Olleros, M.L.; Vesin, D.; Garcia, I. Much More than M1 and M2 Macrophages, There Are Also CD169(+) and TCR(+) Macrophages. Front. Immunol. 2015, 6, 263. [Google Scholar]
  56. Di Benedetto, P.; Ruscitti, P.; Vadasz, Z.; Toubi, E.; Giacomelli, R. Macrophages with Regulatory Functions, a Possible New Therapeutic Perspective in Autoimmune Diseases. Autoimmun. Rev. 2019, 18, 102369. [Google Scholar] [CrossRef]
  57. Panzer, S.E. Macrophages in Transplantation: A Matter of Plasticity, Polarization, and Diversity. Transplantation 2022, 106, 257–267. [Google Scholar] [CrossRef]
  58. Ordikhani, F.; Pothula, V.; Sanchez-Tarjuelo, R.; Jordan, S.; Ochando, J. Macrophages in Organ Transplantation. Front. Immunol. 2020, 11, 582939. [Google Scholar] [CrossRef]
  59. 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]
  60. Desgeorges, T.; Caratti, G.; Mounier, R.; Tuckermann, J.; Chazaud, B. Glucocorticoids Shape Macrophage Phenotype for Tissue Repair. Front. Immunol. 2019, 10, 1591. [Google Scholar] [CrossRef]
  61. Sipka, T.; Park, S.A.; Ozbilgic, R.; Balas, L.; Durand, T.; Mikula, K.; Lutfalla, G.; Nguyen-Chi, M. Macrophages Undergo a Behavioural Switch during Wound Healing in Zebrafish. Free Radic. Biol. Med. 2022, 192, 200–212. [Google Scholar] [CrossRef] [PubMed]
  62. 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] [PubMed]
  63. Tinckam, K.J.; Djurdjev, O.; Magil, A.B. Glomerular Monocytes Predict Worse Outcomes after Acute Renal Allograft Rejection Independent of C4d Status. Kidney Int. 2005, 68, 1866–1874. [Google Scholar] [CrossRef] [PubMed]
  64. Lee, S.; Huen, S.; Nishio, H.; Nishio, S.; Lee, H.K.; Choi, B.-S.; Ruhrberg, C.; Cantley, L.G. Distinct Macrophage Phenotypes Contribute to Kidney Injury and Repair. J. Am. Soc. Nephrol. 2011, 22, 317–326. [Google Scholar] [CrossRef]
  65. Oh, J.Y.; Lee, H.J.; Ko, A.Y.; Ko, J.H.; Kim, M.K.; Wee, W.R. Analysis of Macrophage Phenotype in Rejected Corneal Allografts. Investig. Ophthalmol. Vis. Sci. 2013, 54, 7779–7784. [Google Scholar] [CrossRef]
  66. Mori, D.N.; Kreisel, D.; Fullerton, J.N.; Gilroy, D.W.; Goldstein, D.R. Inflammatory Triggers of Acute Rejection of Organ Allografts. Immunol. Rev. 2014, 258, 132–144. [Google Scholar] [CrossRef]
  67. Paller, M.S.; Hoidal, J.R.; Ferris, T.F. Oxygen Free Radicals in Ischemic Acute Renal Failure in the Rat. J. Clin. Investig. 1984, 74, 1156–1164. [Google Scholar] [CrossRef]
  68. Niederkorn, J.Y. Immune Mechanisms of Corneal Allograft Rejection. Curr. Eye Res. 2007, 32, 1005–1016. [Google Scholar] [CrossRef]
  69. Medzhitov, R.; Janeway, C.A., Jr. Decoding the Patterns of Self and Nonself by the Innate Immune System. Science 2002, 296, 298–300. [Google Scholar] [CrossRef]
  70. Min, X.; Liu, C.; Wei, Y.; Wang, N.; Yuan, G.; Liu, D.; Li, Z.; Zhou, W.; Li, K. Expression and Regulation of Complement Receptors by Human Natural Killer Cells. Immunobiology 2014, 219, 671–679. [Google Scholar] [CrossRef]
  71. Guilliams, M.; Bruhns, P.; Saeys, Y.; Hammad, H.; Lambrecht, B.N. The Function of Fcγ Receptors in Dendritic Cells and Macrophages. Nat. Rev. Immunol. 2014, 14, 94–108. [Google Scholar] [CrossRef] [PubMed]
  72. de Paiva, V.N.; Monteiro, R.M.M.; de Paiva Marques, V.; Cenedeze, M.A.; de, P.A. Teixeira, V.; dos Reis, M.A.; Pacheco-Silva, A.; Câmara, N.O.S. Critical Involvement of Th1-Related Cytokines in Renal Injuries Induced by Ischemia and Reperfusion. Int. Immunopharmacol. 2009, 9, 668–672. [Google Scholar] [CrossRef] [PubMed]
  73. Wyburn, K.R.; Jose, M.D.; Wu, H.; Atkins, R.C.; Chadban, S.J. The Role of Macrophages in Allograft Rejection. Transplantation 2005, 80, 1641–1647. [Google Scholar] [CrossRef] [PubMed]
  74. Wilson, N.A.; Dylewski, J.; Degner, K.R.; O’Neill, M.A.; Reese, S.R.; Hidalgo, L.G.; Blaine, J.; Panzer, S.E. An in Vitro Model of Antibody-Mediated Injury to Glomerular Endothelial Cells: Upregulation of MHC Class II and Adhesion Molecules. Transpl. Immunol. 2020, 58, 101261. [Google Scholar] [CrossRef]
  75. Stewart, S.; Winters, G.L.; Fishbein, M.C.; Tazelaar, H.D.; Kobashigawa, J.; Abrams, J.; Andersen, C.B.; Angelini, A.; Berry, G.J.; Burke, M.M.; et al. Revision of the 1990 Working Formulation for the Standardization of Nomenclature in the Diagnosis of Heart Rejection. J. Heart Lung Transplant. 2005, 24, 1710–1720. [Google Scholar] [CrossRef]
  76. Singh, K.A.; Kampen, R.L.; Hoffmann, S.C.; Eldaif, S.M.; Kirk, A.D. Renal Epithelial Cell-Derived Monocyte Colony Stimulating Factor as a Local Informant of Renal Injury and Means of Monocyte Activation. Transpl. Int. 2009, 22, 730–737. [Google Scholar] [CrossRef]
  77. Xu, Y.; Liu, Y.; Yang, C.; Kang, L.; Wang, M.; Hu, J.; He, H.; Song, W.; Tang, H. Macrophages Transfer Antigens to Dendritic Cells by Releasing Exosomes Containing Dead-Cell-Associated Antigens Partially through a Ceramide-Dependent Pathway to Enhance CD4(+) T-Cell Responses. Immunology 2016, 149, 157–171. [Google Scholar] [CrossRef]
  78. Conde, P.; Rodriguez, M.; van der Touw, W.; Jimenez, A.; Burns, M.; Miller, J.; Brahmachary, M.; Chen, H.-M.; Boros, P.; Rausell-Palamos, F.; et al. DC-SIGN+ Macrophages Control the Induction of Transplantation Tolerance. Immunity 2015, 42, 1143–1158. [Google Scholar] [CrossRef]
  79. Wu, H.; Xu, X.; Li, J.; Gong, J.; Li, M. TIM-4 Blockade of KCs Combined with Exogenous TGF-β Injection Helps to Reverse Acute Rejection and Prolong the Survival Rate of Mice Receiving Liver Allografts. Int. J. Mol. Med. 2018, 42, 346–358. [Google Scholar] [CrossRef]
  80. Krausgruber, T.; Blazek, K.; Smallie, T.; Alzabin, S.; Lockstone, H.; Sahgal, N.; Hussell, T.; Feldmann, M.; Udalova, I.A. IRF5 Promotes Inflammatory Macrophage Polarization and TH1-TH17 Responses. Nat. Immunol. 2011, 12, 231–238. [Google Scholar] [CrossRef]
  81. Dai, H.; Friday, A.J.; Abou-Daya, K.I.; Williams, A.L.; Mortin-Toth, S.; Nicotra, M.L.; Rothstein, D.M.; Shlomchik, W.D.; Matozaki, T.; Isenberg, J.S.; et al. Donor SIRPα Polymorphism Modulates the Innate Immune Response to Allogeneic Grafts. Sci. Immunol. 2017, 2, eaam6202. [Google Scholar] [CrossRef] [PubMed]
  82. Dai, H.; Lan, P.; Zhao, D.; Abou-Daya, K.; Liu, W.; Chen, W.; Friday, A.J.; Williams, A.L.; Sun, T.; Chen, J.; et al. PIRs Mediate Innate Myeloid Cell Memory to Nonself MHC Molecules. Science 2020, 368, 1122–1127. [Google Scholar] [CrossRef] [PubMed]
  83. Ochando, J.; Mulder, W.J.M.; Madsen, J.C.; Netea, M.G.; Duivenvoorden, R. Trained Immunity-Basic Concepts and Contributions to Immunopathology. Nat. Rev. Nephrol. 2023, 19, 23–37. [Google Scholar] [CrossRef] [PubMed]
  84. Jose, M.D.; Ikezumi, Y.; van Rooijen, N.; Atkins, R.C.; Chadban, S.J. Macrophages Act as Effectors of Tissue Damage in Acute Renal Allograft Rejection. Transplantation 2003, 76, 1015–1022. [Google Scholar] [CrossRef]
  85. Ma, F.Y.; Woodman, N.; Mulley, W.R.; Kanellis, J.; Nikolic-Paterson, D.J. Macrophages Contribute to Cellular but Not Humoral Mechanisms of Acute Rejection in Rat Renal Allografts. Transplantation 2013, 96, 949–957. [Google Scholar] [CrossRef]
  86. Zhao, Z.; Pan, G.; Tang, C.; Li, Z.; Zheng, D.; Wei, X.; Wu, Z. IL-34 Inhibits Acute Rejection of Rat Liver Transplantation by Inducing Kupffer Cell M2 Polarization. Transplantation 2018, 102, e265–e274. [Google Scholar] [CrossRef]
  87. Gao, C.; Wang, X.; Lu, J.; Li, Z.; Jia, H.; Chen, M.; Chang, Y.; Liu, Y.; Li, P.; Zhang, B.; et al. Mesenchymal Stem Cells Transfected with sFgl2 Inhibit the Acute Rejection of Heart Transplantation in Mice by Regulating Macrophage Activation. Stem Cell Res. Ther. 2020, 11, 241. [Google Scholar] [CrossRef]
  88. Van den Bosch, T.P.P.; Kannegieter, N.M.; Hesselink, D.A.; Baan, C.C.; Rowshani, A.T. Targeting the Monocyte–Macrophage Lineage in Solid Organ Transplantation. Front. Immunol. 2017, 8, 153. [Google Scholar] [CrossRef]
  89. Ochando, J.; Ordikhani, F.; Boros, P.; Jordan, S. The Innate Immune Response to Allotransplants: Mechanisms and Therapeutic Potentials. Cell. Mol. Immunol. 2019, 16, 350–356. [Google Scholar] [CrossRef]
  90. Bronte, V.; Serafini, P.; De Santo, C.; Marigo, I.; Tosello, V.; Mazzoni, A.; Segal, D.M.; Staib, C.; Lowel, M.; Sutter, G.; et al. IL-4-Induced Arginase 1 Suppresses Alloreactive T Cells in Tumor-Bearing Mice. J. Immunol. 2003, 170, 270–278. [Google Scholar] [CrossRef]
  91. Hajkova, M.; Javorkova, E.; Zajicova, A.; Trosan, P.; Holan, V.; Krulova, M. A Local Application of Mesenchymal Stem Cells and Cyclosporine a Attenuates Immune Response by a Switch in Macrophage Phenotype. J. Tissue Eng. Regen. Med. 2017, 11, 1456–1465. [Google Scholar] [CrossRef] [PubMed]
  92. Garcia, M.R.; Ledgerwood, L.; Yang, Y.; Xu, J.; Lal, G.; Burrell, B.; Ma, G.; Hashimoto, D.; Li, Y.; Boros, P.; et al. Monocytic Suppressive Cells Mediate Cardiovascular Transplantation Tolerance in Mice. J. Clin. Investig. 2010, 120, 2486–2496. [Google Scholar] [CrossRef] [PubMed]
  93. Braza, M.S.; Conde, P.; Garcia, M.; Cortegano, I.; Brahmachary, M.; Pothula, V.; Fay, F.; Boros, P.; Werner, S.A.; Ginhoux, F.; et al. Neutrophil Derived CSF1 Induces Macrophage Polarization and Promotes Transplantation Tolerance. Am. J. Transplant 2018, 18, 1247–1255. [Google Scholar] [CrossRef] [PubMed]
  94. Rickert, C.G.; Markmann, J.F. Current State of Organ Transplant Tolerance. Curr. Opin. Organ Transplant. 2019, 24, 441–450. [Google Scholar] [CrossRef]
  95. Zhang, F.; Zhang, J.; Cao, P.; Sun, Z.; Wang, W. The Characteristics of Regulatory Macrophages and Their Roles in Transplantation. Int. Immunopharmacol. 2021, 91, 107322. [Google Scholar] [CrossRef]
  96. Schmidt, A.; Zhang, X.-M.; Joshi, R.N.; Iqbal, S.; Wahlund, C.; Gabrielsson, S.; Harris, R.A.; Tegnér, J. Human Macrophages Induce CD4(+)Foxp3(+) Regulatory T Cells via Binding and Re-Release of TGF-β. Immunol. Cell Biol. 2016, 94, 747–762. [Google Scholar]
  97. Braza, M.S.; van Leent, M.M.T.; Lameijer, M.; Sanchez-Gaytan, B.L.; Arts, R.J.W.; Pérez-Medina, C.; Conde, P.; Garcia, M.R.; Gonzalez-Perez, M.; Brahmachary, M.; et al. Inhibiting Inflammation with Myeloid Cell-Specific Nanobiologics Promotes Organ Transplant Acceptance. Immunity 2018, 49, 819–828.e6. [Google Scholar] [CrossRef]
  98. Warnecke, G.; Hutchinson, J.A.; Riquelme, P.; Kruse, B.; Thissen, S.; Avsar, M.; Zehle, G.; Steinkamp, T.; Peters, C.; Baumann, R.; et al. Postoperative Intravenous Infusion of Donor-Derived Transplant Acceptance-Inducing Cells as an Adjunct Immunosuppressive Therapy in a Porcine Pulmonary Allograft Model. Transpl. Int. 2009, 22, 332–341. [Google Scholar] [CrossRef]
  99. Hutchinson, J.A.; Gövert, F.; Riquelme, P.; Bräsen, J.H.; Brem-Exner, B.G.; Matthäi, M.; Schulze, M.; Renders, L.; Kunzendorf, U.; Geissler, E.K.; et al. Administration of Donor-Derived Transplant Acceptance-Inducing Cells to the Recipients of Renal Transplants from Deceased Donors Is Technically Feasible. Clin. Transplant. 2009, 23, 140–145. [Google Scholar] [CrossRef]
  100. Hutchinson, J.A.; Riquelme, P.; Brem-Exner, B.G.; Schulze, M.; Matthäi, M.; Renders, L.; Kunzendorf, U.; Geissler, E.K.; Fändrich, F. Transplant Acceptance-Inducing Cells as an Immune-Conditioning Therapy in Renal Transplantation. Transpl. Int. 2008, 21, 728–741. [Google Scholar] [CrossRef]
  101. Hutchinson, J.A.; Brem-Exner, B.G.; Riquelme, P.; Roelen, D.; Schulze, M.; Ivens, K.; Grabensee, B.; Witzke, O.; Philipp, T.; Renders, L.; et al. A Cell-Based Approach to the Minimization of Immunosuppression in Renal Transplantation. Transpl. Int. 2008, 21, 742–754. [Google Scholar] [CrossRef] [PubMed]
  102. Sawitzki, B.; Harden, P.N.; Reinke, P.; Moreau, A.; Hutchinson, J.A.; Game, D.S.; Tang, Q.; Guinan, E.C.; Battaglia, M.; Burlingham, W.J.; et al. Regulatory Cell Therapy in Kidney Transplantation (The ONE Study): A Harmonised Design and Analysis of Seven Non-Randomised, Single-Arm, Phase 1/2A Trials. Lancet 2020, 395, 1627–1639. [Google Scholar] [CrossRef] [PubMed]
  103. Mellor, A.L.; Munn, D.H. Creating Immune Privilege: Active Local Suppression That Benefits Friends, but Protects Foes. Nat. Rev. Immunol. 2008, 8, 74–80. [Google Scholar] [CrossRef] [PubMed]
  104. Lee, C.-L.; Lam, K.K.W.; Vijayan, M.; Koistinen, H.; Seppala, M.; Ng, E.H.Y.; Yeung, W.S.B.; Chiu, P.C.N. The Pleiotropic Effect of Glycodelin-A in Early Pregnancy. Am. J. Reprod. Immunol. 2016, 75, 290–297. [Google Scholar] [CrossRef] [PubMed]
  105. Bonnefoy, F.; Gauthier, T.; Vallion, R.; Martin-Rodriguez, O.; Missey, A.; Daoui, A.; Valmary-Degano, S.; Saas, P.; Couturier, M.; Perruche, S. Factors Produced by Macrophages Eliminating Apoptotic Cells Demonstrate Pro-Resolutive Properties and Terminate Ongoing Inflammation. Front. Immunol. 2018, 9, 2586. [Google Scholar] [CrossRef]
  106. 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]
  107. Ginhoux, F.; Schultze, J.L.; Murray, P.J.; Ochando, J.; Biswas, S.K. New Insights into the Multidimensional Concept of Macrophage Ontogeny, Activation and Function. Nat. Immunol. 2016, 17, 34–40. [Google Scholar] [CrossRef]
  108. Barclay, A.N.; Van den Berg, T.K. The Interaction between Signal Regulatory Protein Alpha (SIRPα) and CD47: Structure, Function, and Therapeutic Target. Annu. Rev. Immunol. 2014, 32, 25–50. [Google Scholar] [CrossRef]
  109. Thornley, T.B.; Fang, Z.; Balasubramanian, S.; Larocca, R.A.; Gong, W.; Gupta, S.; Csizmadia, E.; Degauque, N.; Kim, B.S.; Koulmanda, M.; et al. Fragile TIM-4-Expressing Tissue Resident Macrophages Are Migratory and Immunoregulatory. J. Clin. Investig. 2014, 124, 3443–3454. [Google Scholar] [CrossRef]
  110. Akbarpour, M.; Lecuona, E.; Chiu, S.F.; Wu, Q.; Querrey, M.; Fernandez, R.; Núñez-Santana, F.L.; Sun, H.; Ravi, S.; Kurihara, C.; et al. Residual Endotoxin Induces Primary Graft Dysfunction through Ischemia/reperfusion-Primed Alveolar Macrophages. J. Clin. Investig. 2020, 130, 4456–4469. [Google Scholar] [CrossRef]
  111. Bilyk, N.; Holt, P.G. Inhibition of the Immunosuppressive Activity of Resident Pulmonary Alveolar Macrophages by Granulocyte/macrophage Colony-Stimulating Factor. J. Exp. Med. 1993, 177, 1773–1777. [Google Scholar] [CrossRef] [PubMed]
  112. Bajpai, G.; Schneider, C.; Wong, N.; Bredemeyer, A.; Hulsmans, M.; Nahrendorf, M.; Epelman, S.; Kreisel, D.; Liu, Y.; Itoh, A.; et al. The Human Heart Contains Distinct Macrophage Subsets with Divergent Origins and Functions. Nat. Med. 2018, 24, 1234–1245. [Google Scholar] [CrossRef] [PubMed]
  113. Kopecky, B.J.; Dun, H.; Amrute, J.M.; Lin, C.-Y.; Bredemeyer, A.L.; Terada, Y.; Bayguinov, P.O.; Koenig, A.L.; Frye, C.C.; Fitzpatrick, J.A.J.; et al. Donor Macrophages Modulate Rejection After Heart Transplantation. Circulation 2022, 146, 623–638. [Google Scholar] [CrossRef] [PubMed]
  114. Hofmann, J.; Hackl, V.; Esser, H.; Meszaros, A.T.; Fodor, M.; Öfner, D.; Troppmair, J.; Schneeberger, S.; Hautz, T. Cell-Based Regeneration and Treatment of Liver Diseases. Int. J. Mol. Sci. 2021, 22, 276. [Google Scholar] [CrossRef]
  115. Gao, J.; He, K.; Xia, Q.; Zhang, J. Research Progress on Hepatic Machine Perfusion. Int. J. Med. Sci. 2021, 18, 1953–1959. [Google Scholar] [CrossRef] [PubMed]
  116. Ichimura, T.; Asseldonk, E.J.P.V.; Humphreys, B.D.; Gunaratnam, L.; Duffield, J.S.; Bonventre, J.V. Kidney Injury Molecule-1 Is a Phosphatidylserine Receptor That Confers a Phagocytic Phenotype on Epithelial Cells. J. Clin. Investig. 2008, 118, 1657–1668. [Google Scholar] [CrossRef]
  117. Lee, J.Y.; Arumugarajah, S.; Lian, D.; Maehara, N.; Haig, A.R.; Suri, R.S.; Miyazaki, T.; Gunaratnam, L. Recombinant Apoptosis Inhibitor of Macrophage Protein Reduces Delayed Graft Function in a Murine Model of Kidney Transplantation. PLoS ONE 2021, 16, e0249838. [Google Scholar] [CrossRef]
  118. Chen, Y.; He, Y.; Wu, X.; Xu, X.; Gong, J.; Chen, Y.; Gong, J. Rubicon Promotes the M2 Polarization of Kupffer Cells via LC3-Associated Phagocytosis-Mediated Clearance to Improve Liver Transplantation. Cell. Immunol. 2022, 378, 104556. [Google Scholar] [CrossRef]
  119. Nelson, J.; Alvey, N.; Bowman, L.; Schulte, J.; Segovia, M.C.; McDermott, J.; Te, H.S.; Kapila, N.; Levine, D.J.; Gottlieb, R.L.; et al. Consensus Recommendations for Use of Maintenance Immunosuppression in Solid Organ Transplantation: Endorsed by the American College of Clinical Pharmacy, American Society of Transplantation, and the International Society for Heart and Lung Transplantation. Pharmacotherapy 2022, 42, 599–633. [Google Scholar] [CrossRef]
  120. Zhao, Y.; Shen, X.; Na, N.; Chu, Z.; Su, H.; Chao, S.; Shi, L.; Xu, Y.; Zhang, L.; Shi, B.; et al. mTOR Masters Monocyte Development in Bone Marrow by Decreasing the Inhibition of STAT5 on IRF8. Blood 2018, 131, 1587–1599. [Google Scholar] [CrossRef]
  121. Salehi, S.; Sosa, R.A.; Jin, Y.-P.; Kageyama, S.; Fishbein, M.C.; Rozengurt, E.; Kupiec-Weglinski, J.W.; Reed, E.F. Outside-in HLA Class I Signaling Regulates ICAM-1 Clustering and Endothelial Cell-Monocyte Interactions via mTOR in Transplant Antibody-Mediated Rejection. Am. J. Transplant 2018, 18, 1096–1109. [Google Scholar] [CrossRef] [PubMed]
  122. Mercalli, A.; Calavita, I.; Dugnani, E.; Citro, A.; Cantarelli, E.; Nano, R.; Melzi, R.; Maffi, P.; Secchi, A.; Sordi, V.; et al. Rapamycin Unbalances the Polarization of Human Macrophages to M1. Immunology 2013, 140, 179–190. [Google Scholar] [CrossRef] [PubMed]
  123. Zhao, Y.; Chen, S.; Lan, P.; Wu, C.; Dou, Y.; Xiao, X.; Zhang, Z.; Minze, L.; He, X.; Chen, W.; et al. Macrophage Subpopulations and Their Impact on Chronic Allograft Rejection versus Graft Acceptance in a Mouse Heart Transplant Model. Am. J. Transplant 2018, 18, 604–616. [Google Scholar] [CrossRef] [PubMed]
  124. Lin, H.Y.-H.; Chang, K.-T.; Hung, C.-C.; Kuo, C.-H.; Hwang, S.-J.; Chen, H.-C.; Hung, C.-H.; Lin, S.-F. Effects of the mTOR Inhibitor Rapamycin on Monocyte-Secreted Chemokines. BMC Immunol. 2014, 15, 37. [Google Scholar] [CrossRef]
  125. Wang, J.; Xie, T.; Long, X.; Gao, R.; Kang, L.; Wang, Q.; Jiang, J.; Ye, L.; Lyu, J. The Effect of Tacrolimus-Containing Polyethylene Glycol-Modified Maghemite Nanospheres on Reducing Oxidative Stress and Accelerating the Healing Spinal Cord Injury of Rats Based on Increasing M2 Macrophages. Arab. J. Chem. 2022, 15, 103534. [Google Scholar] [CrossRef]
  126. Kannegieter, N.M.; Hesselink, D.A.; Dieterich, M.; Kraaijeveld, R.; Rowshani, A.T.; Leenen, P.J.M.; Baan, C.C. The Effect of Tacrolimus and Mycophenolic Acid on CD14+ Monocyte Activation and Function. PLoS ONE 2017, 12, e0170806. [Google Scholar] [CrossRef]
  127. Yoshino, T.; Nakase, H.; Honzawa, Y.; Matsumura, K.; Yamamoto, S.; Takeda, Y.; Ueno, S.; Uza, N.; Masuda, S.; Inui, K.; et al. Immunosuppressive Effects of Tacrolimus on Macrophages Ameliorate Experimental Colitis. Inflamm. Bowel Dis. 2010, 16, 2022–2033. [Google Scholar] [CrossRef]
  128. Jennings, C.; Kusler, B.; Jones, P.P. Calcineurin Inactivation Leads to Decreased Responsiveness to LPS in Macrophages and Dendritic Cells and Protects against LPS-Induced Toxicity in Vivo. Innate Immun. 2009, 15, 109–120. [Google Scholar] [CrossRef]
  129. Hölschermann, H.; Dürfeld, F.; Maus, U.; Bierhaus, A.; Heidinger, K.; Lohmeyer, J.; Nawroth, P.P.; Tillmanns, H.; Haberbosch, W. Cyclosporine a Inhibits Tissue Factor Expression in Monocytes/macrophages. Blood 1996, 88, 3837–3845. [Google Scholar] [CrossRef]
  130. Savikko, J.; Teppo, A.-M.; Taskinen, E.; von Willebrand, E. Different Effects of Tacrolimus and Cyclosporine on PDGF Induction and Chronic Allograft Injury: Evidence for Improved Kidney Graft Outcome. Transpl. Immunol. 2014, 31, 145–151. [Google Scholar] [CrossRef]
  131. Mizuiri, S.; Iwamoto, M.; Miyagi, M.; Kawamura, T.; Sakai, K.; Arai, K.; Aikawa, A.; Ohara, T.; Hemmi, H.; Hasegawa, A. Activation of Nuclear Factor-Kappa B and Macrophage Invasion in Cyclosporin A-and Tacrolimus-Treated Renal Transplants. Clin. Transplant. 2004, 18, 14–20. [Google Scholar] [CrossRef] [PubMed]
  132. Murphy, G.J.; Waller, J.R.; Sandford, R.S.; Furness, P.N.; Nicholson, M.L. Randomized Clinical Trial of the Effect of Microemulsion Cyclosporin and Tacrolimus on Renal Allograft Fibrosis. Br. J. Surg. 2003, 90, 680–686. [Google Scholar] [CrossRef] [PubMed]
  133. Jiang, B.; Zhang, Y.; Li, Y.; Chen, Y.; Sha, S.; Zhao, L.; Li, D.; Wen, J.; Lan, J.; Lou, Y.; et al. A Tissue-Tended Mycophenolate-Modified Nanoparticle Alleviates Systemic Lupus Erythematosus in MRL/ Mouse Model Mainly by Promoting Local M2-Like Macrophagocytes Polarization. Int. J. Nanomed. 2022, 17, 3251–3267. [Google Scholar] [CrossRef] [PubMed]
  134. Weimer, R.; Mytilineos, J.; Feustel, A.; Preiss, A.; Daniel, V.; Grimm, H.; Wiesel, M.; Opelz, G. Mycophenolate Mofetil-Based Immunosuppression and Cytokine Genotypes: Effects on Monokine Secretion and Antigen Presentation in Long-Term Renal Transplant Recipients. Transplantation 2003, 75, 2090–2099. [Google Scholar]
  135. Glomsda, B.A.; Blaheta, R.A.; Hailer, N.P. Inhibition of Monocyte/endothelial Cell Interactions and Monocyte Adhesion Molecule Expression by the Immunosuppressant Mycophenolate Mofetil. Spinal Cord 2003, 41, 610–619. [Google Scholar] [CrossRef]
  136. Tuckermann, J.P.; Kleiman, A.; Moriggl, R.; Spanbroek, R.; Neumann, A.; Illing, A.; Clausen, B.E.; Stride, B.; Förster, I.; Habenicht, A.J.R.; et al. Macrophages and Neutrophils Are the Targets for Immune Suppression by Glucocorticoids in Contact Allergy. J. Clin. Investig. 2007, 117, 1381–1390. [Google Scholar] [CrossRef]
  137. Di Rosa, M.; Radomski, M.; Carnuccio, R.; Moncada, S. Glucocorticoids Inhibit the Induction of Nitric Oxide Synthase in Macrophages. Biochem. Biophys. Res. Commun. 1990, 172, 1246–1252. [Google Scholar] [CrossRef]
  138. Lim, H.-Y.; Müller, N.; Herold, M.J.; van den Brandt, J.; Reichardt, H.M. Glucocorticoids Exert Opposing Effects on Macrophage Function Dependent on Their Concentration. Immunology 2007, 122, 47–53. [Google Scholar] [CrossRef]
  139. Bergler, T.; Jung, B.; Bourier, F.; Kühne, L.; Banas, M.C.; Rümmele, P.; Wurm, S.; Banas, B. Infiltration of Macrophages Correlates with Severity of Allograft Rejection and Outcome in Human Kidney Transplantation. PLoS ONE 2016, 11, e0156900. [Google Scholar] [CrossRef]
  140. Ikezumi, Y.; Suzuki, T.; Karasawa, T.; Hasegawa, H.; Kawachi, H.; Nikolic-Paterson, D.J.; Uchiyama, M. Contrasting Effects of Steroids and Mizoribine on Macrophage Activation and Glomerular Lesions in Rat Thy-1 Mesangial Proliferative Glomerulonephritis. Am. J. Nephrol. 2010, 31, 273–282. [Google Scholar] [CrossRef]
  141. Ehrchen, J.; Steinmüller, L.; Barczyk, K.; Tenbrock, K.; Nacken, W.; Eisenacher, M.; Nordhues, U.; Sorg, C.; Sunderkötter, C.; Roth, J. Glucocorticoids Induce Differentiation of a Specifically Activated, Anti-Inflammatory Subtype of Human Monocytes. Blood 2007, 109, 1265–1274. [Google Scholar] [CrossRef]
  142. Van der Goes, A.; Hoekstra, K.; van den Berg, T.K.; Dijkstra, C.D. Dexamethasone Promotes Phagocytosis and Bacterial Killing by Human Monocytes/macrophages in Vitro. J. Leukoc. Biol. 2000, 67, 801–807. [Google Scholar] [CrossRef]
  143. Ikezumi, Y.; Suzuki, T.; Yamada, T.; Hasegawa, H.; Kaneko, U.; Hara, M.; Yanagihara, T.; Nikolic-Paterson, D.J.; Saitoh, A. Alternatively Activated Macrophages in the Pathogenesis of Chronic Kidney Allograft Injury. Pediatr. Nephrol. 2015, 30, 1007–1017. [Google Scholar] [CrossRef]
  144. Dobi, D.; Vincenti, F.; Chandran, S.; Greenland, J.R.; Bowman, C.; Chen, A.; Junger, H.; Laszik, Z.G. The Impact of Belatacept on the Phenotypic Heterogeneity of Renal T Cell-Mediated Alloimmune Response: The Critical Role of Maintenance Treatment and Inflammatory Load. Clin. Transplant. 2020, 34, e14084. [Google Scholar] [CrossRef]
  145. Bonelli, M.; Ferner, E.; Göschl, L.; Blüml, S.; Hladik, A.; Karonitsch, T.; Kiener, H.P.; Byrne, R.; Niederreiter, B.; Steiner, C.W.; et al. Abatacept (CTLA-4IG) Treatment Reduces the Migratory Capacity of Monocytes in Patients with Rheumatoid Arthritis. Arthritis Rheum. 2013, 65, 599–607. [Google Scholar] [CrossRef]
  146. Wenink, M.H.; Santegoets, K.C.M.; Platt, A.M.; van den Berg, W.B.; van Riel, P.L.C.M.; Garside, P.; Radstake, T.R.D.J.; McInnes, I.B. Abatacept Modulates Proinflammatory Macrophage Responses upon Cytokine-Activated T Cell and Toll-like Receptor Ligand Stimulation. Ann. Rheum. Dis. 2012, 71, 80–83. [Google Scholar] [CrossRef]
  147. Cutolo, M.; Soldano, S.; Montagna, P.; Sulli, A.; Seriolo, B.; Villaggio, B.; Triolo, P.; Clerico, P.; Felli, L.; Brizzolara, R. CTLA4-Ig Interacts with Cultured Synovial Macrophages from Rheumatoid Arthritis Patients and Downregulates Cytokine Production. Arthritis Res. Ther. 2009, 11, R176. [Google Scholar] [CrossRef]
  148. Cutolo, M.; Soldano, S.; Gotelli, E.; Montagna, P.; Campitiello, R.; Paolino, S.; Pizzorni, C.; Sulli, A.; Smith, V.; Tardito, S. CTLA4-Ig Treatment Induces M1-M2 Shift in Cultured Monocyte-Derived Macrophages from Healthy Subjects and Rheumatoid Arthritis Patients. Arthritis Res. Ther. 2021, 23, 306. [Google Scholar] [CrossRef]
  149. Granata, S.; Dalla Gassa, A.; Carraro, A.; Brunelli, M.; Stallone, G.; Lupo, A.; Zaza, G. Sirolimus and Everolimus Pathway: Reviewing Candidate Genes Influencing Their Intracellular Effects. Int. J. Mol. Sci. 2016, 17, 735. [Google Scholar] [CrossRef]
  150. Taylor, A.L.; Watson, C.J.E.; Bradley, J.A. Immunosuppressive Agents in Solid Organ Transplantation: Mechanisms of Action and Therapeutic Efficacy. Crit. Rev. Oncol. Hematol. 2005, 56, 23–46. [Google Scholar] [CrossRef]
  151. Cangemi, M.; Montico, B.; Faè, D.A.; Steffan, A.; Dolcetti, R. Dissecting the Multiplicity of Immune Effects of Immunosuppressive Drugs to Better Predict the Risk of Malignancies in Solid Organ Transplant Patients. Front. Oncol. 2019, 9, 160. [Google Scholar] [CrossRef] [PubMed]
  152. Vafadari, R.; Kraaijeveld, R.; Weimar, W.; Baan, C.C. Tacrolimus Inhibits NF-κB Activation in Peripheral Human T Cells. PLoS ONE 2013, 8, e60784. [Google Scholar] [CrossRef] [PubMed]
  153. Emal, D.; Rampanelli, E.; Claessen, N.; Bemelman, F.J.; Leemans, J.C.; Florquin, S.; Dessing, M.C. Calcineurin Inhibitor Tacrolimus Impairs Host Immune Response against Urinary Tract Infection. Sci. Rep. 2019, 9, 106. [Google Scholar] [CrossRef] [PubMed]
  154. Kakuta, Y.; Okumi, M.; Miyagawa, S.; Tsutahara, K.; Abe, T.; Yazawa, K.; Matsunami, K.; Otsuka, H.; Takahara, S.; Nonomura, N. Blocking of CCR5 and CXCR3 Suppresses the Infiltration of Macrophages in Acute Renal Allograft Rejection. Transplantation 2012, 93, 24–31. [Google Scholar] [CrossRef] [PubMed]
  155. Young, B.A.; Burdmann, E.A.; Johnson, R.J.; Alpers, C.E.; Giachelli, C.M.; Eng, E.; Andoh, T.; Bennett, W.M.; Couser, W.G. Cellular Proliferation and Macrophage Influx Precede Interstitial Fibrosis in Cyclosporine Nephrotoxicity. Kidney Int. 1995, 48, 439–448. [Google Scholar] [CrossRef]
  156. Allison, A.C. Mechanisms of Action of Mycophenolate Mofetil. Lupus 2005, 14, S2–S8. [Google Scholar] [CrossRef]
  157. Allison, A.C.; Eugui, E.M. Mycophenolate Mofetil and Its Mechanisms of Action. Immunopharmacology 2000, 47, 85–118. [Google Scholar] [CrossRef]
  158. Adcock, I.M.; Ito, K. Molecular Mechanisms of Corticosteroid Actions. Monaldi Arch. Chest Dis. 2000, 55, 256–266. [Google Scholar]
  159. Jia, D.; O’Brien, C.A.; Stewart, S.A.; Manolagas, S.C.; Weinstein, R.S. Glucocorticoids Act Directly on Osteoclasts to Increase Their Life Span and Reduce Bone Density. Endocrinology 2006, 147, 5592–5599. [Google Scholar] [CrossRef]
  160. Meers, G.K.; Bohnenberger, H.; Reichardt, H.M.; Lühder, F.; Reichardt, S.D. Impaired Resolution of DSS-Induced Colitis in Mice Lacking the Glucocorticoid Receptor in Myeloid Cells. PLoS ONE 2018, 13, e0190846. [Google Scholar] [CrossRef]
  161. Limbourg, F.P.; Huang, Z.; Plumier, J.-C.; Simoncini, T.; Fujioka, M.; Tuckermann, J.; Schütz, G.; Moskowitz, M.A.; Liao, J.K. Rapid Nontranscriptional Activation of Endothelial Nitric Oxide Synthase Mediates Increased Cerebral Blood Flow and Stroke Protection by Corticosteroids. J. Clin. Investig. 2002, 110, 1729–1738. [Google Scholar] [CrossRef]
  162. Ippoliti, G.; D’Armini, A.M.; Lucioni, M.; Marjieh, M.; Viganò, M. Introduction to the Use of Belatacept: A Fusion Protein for the Prevention of Posttransplant Kidney Rejection. Biologics 2012, 6, 355–362. [Google Scholar] [CrossRef]
  163. Muntjewerff, E.M.; Meesters, L.D.; van den Bogaart, G. Antigen Cross-Presentation by Macrophages. Front. Immunol. 2020, 11, 1276. [Google Scholar] [CrossRef]
  164. Larsen, C.P.; Pearson, T.C.; Adams, A.B.; Tso, P.; Shirasugi, N.; Strobert, E.; Anderson, D.; Cowan, S.; Price, K.; Naemura, J.; et al. Rational Development of LEA29Y (belatacept), a High-Affinity Variant of CTLA4-Ig with Potent Immunosuppressive Properties. Am. J. Transplant 2005, 5, 443–453. [Google Scholar] [CrossRef]
  165. Werner, E.R.; Blau, N.; Thöny, B. Tetrahydrobiopterin: Biochemistry and Pathophysiology. Biochem. J. 2011, 438, 397–414. [Google Scholar] [CrossRef]
  166. Chuaiphichai, S.; Chu, S.M.; Carnicer, R.; Kelly, M.; Bendall, J.K.; Simon, J.N.; Douglas, G.; Crabtree, M.J.; Casadei, B.; Channon, K.M. Endothelial Cell-Specific Roles for Tetrahydrobiopterin in Myocardial Function, Cardiac Hypertrophy, and Response to Myocardial Ischemia-Reperfusion Injury. Am. J. Physiol. Heart Circ. Physiol. 2023, 324, H430–H442. [Google Scholar] [CrossRef]
  167. Dumitrescu, C.; Biondi, R.; Xia, Y.; Cardounel, A.J.; Druhan, L.J.; Ambrosio, G.; Zweier, J.L. Myocardial Ischemia Results in Tetrahydrobiopterin (BH4) Oxidation with Impaired Endothelial Function Ameliorated by BH4. Proc. Natl. Acad. Sci. USA 2007, 104, 15081–15086. [Google Scholar] [CrossRef]
  168. Tiefenbacher, C.P.; Lee, C.-H.; Kapitza, J.; Dietz, V.; Niroomand, F. Sepiapterin Reduces Postischemic Injury in the Rat Heart. Pflugers Arch. 2003, 447, 1–7. [Google Scholar] [CrossRef]
  169. Maglione, M.; Cardini, B.; Oberhuber, R.; Watschinger, K.; Jenny, M.; Gostner, J.; Hermann, M.; Obrist, P.; Margreiter, R.; Pratschke, J.; et al. Prevention of Lethal Murine Pancreas Ischemia Reperfusion Injury Is Specific for Tetrahydrobiopterin. Transpl. Int. 2012, 25, 1084–1095. [Google Scholar] [CrossRef]
  170. Cardini, B.; Watschinger, K.; Hermann, M.; Obrist, P.; Oberhuber, R.; Brandacher, G.; Chuaiphichai, S.; Channon, K.M.; Pratschke, J.; Maglione, M.; et al. Crucial Role for Neuronal Nitric Oxide Synthase in Early Microcirculatory Derangement and Recipient Survival Following Murine Pancreas Transplantation. PLoS ONE 2014, 9, e112570. [Google Scholar] [CrossRef]
  171. Oberhuber, R.; Riede, G.; Cardini, B.; Bernhard, D.; Messner, B.; Watschinger, K.; Steger, C.; Brandacher, G.; Pratschke, J.; Golderer, G.; et al. Impaired Endothelial Nitric Oxide Synthase Homodimer Formation Triggers Development of Transplant Vasculopathy-Insights from a Murine Aortic Transplantation Model. Sci. Rep. 2016, 6, 37917. [Google Scholar] [CrossRef] [PubMed]
  172. Ge, Z.-D.; Ionova, I.A.; Vladic, N.; Pravdic, D.; Hirata, N.; Vásquez-Vivar, J.; Pratt, P.F., Jr.; Warltier, D.C.; Pieper, G.M.; Kersten, J.R. Cardiac-Specific Overexpression of GTP Cyclohydrolase 1 Restores Ischaemic Preconditioning during Hyperglycaemia. Cardiovasc. Res. 2011, 91, 340–349. [Google Scholar] [CrossRef] [PubMed]
  173. Hiroi, T.; Wajima, T.; Kaneko, Y.; Kiuchi, Y.; Shimizu, S. An Important Role of Increase in Tetrahydrobiopterin via H2O2-JAK2 Signalling Pathway in Late Phase of Ischaemic Preconditioning. Exp. Physiol. 2010, 95, 609–621. [Google Scholar] [CrossRef] [PubMed]
  174. Brandacher, G.; Maglione, M.; Schneeberger, S.; Obrist, P.; Thoeni, G.; Wrulich, O.A.; Werner-Felmayer, G.; Margreiter, R.; Werner, E.R. Tetrahydrobiopterin Compounds Prolong Allograft Survival Independently of Their Effect on Nitric Oxide Synthase Activity. Transplantation 2006, 81, 583–589. [Google Scholar] [CrossRef] [PubMed]
  175. Ionova, I.A.; Vásquez-Vivar, J.; Cooley, B.C.; Khanna, A.K.; Whitsett, J.; Herrnreiter, A.; Migrino, R.Q.; Ge, Z.-D.; Regner, K.R.; Channon, K.M.; et al. Cardiac Myocyte-Specific Overexpression of Human GTP Cyclohydrolase I Protects against Acute Cardiac Allograft Rejection. Am. J. Physiol. Heart Circ. Physiol. 2010, 299, H88–H96. [Google Scholar] [CrossRef] [PubMed]
  176. MacMicking, J.D.; Nathan, C.; Hom, G.; Chartrain, N.; Fletcher, D.S.; Trumbauer, M.; Stevens, K.; Xie, Q.W.; Sokol, K.; Hutchinson, N. Altered Responses to Bacterial Infection and Endotoxic Shock in Mice Lacking Inducible Nitric Oxide Synthase. Cell 1995, 81, 641–650. [Google Scholar] [CrossRef] [PubMed]
  177. McNeill, E.; Crabtree, M.J.; Sahgal, N.; Patel, J.; Chuaiphichai, S.; Iqbal, A.J.; Hale, A.B.; Greaves, D.R.; Channon, K.M. Regulation of iNOS Function and Cellular Redox State by Macrophage Gch1 Reveals Specific Requirements for Tetrahydrobiopterin in NRF2 Activation. Free Radic. Biol. Med. 2015, 79, 206–216. [Google Scholar] [CrossRef]
  178. McNeill, E.; Stylianou, E.; Crabtree, M.J.; Harrington-Kandt, R.; Kolb, A.-L.; Diotallevi, M.; Hale, A.B.; Bettencourt, P.; Tanner, R.; O’Shea, M.K.; et al. Regulation of Mycobacterial Infection by Macrophage Gch1 and Tetrahydrobiopterin. Nat. Commun. 2018, 9, 5409. [Google Scholar] [CrossRef]
  179. Tietz, A.; Lindberg, M.; Kennedy, E.P. A New Pteridine-Requiring Enzyme System for the Oxidation of Glyceryl Ethers. J. Biol. Chem. 1964, 239, 4081–4090. [Google Scholar] [CrossRef]
  180. Magnusson, C.D.; Haraldsson, G.G. Ether Lipids. Chem. Phys. Lipids 2011, 164, 315–340. [Google Scholar] [CrossRef]
  181. Dorninger, F.; Forss-Petter, S.; Wimmer, I.; Berger, J. Plasmalogens, Platelet-Activating Factor and beyond-Ether Lipids in Signaling and Neurodegeneration. Neurobiol. Dis. 2020, 145, 105061. [Google Scholar] [CrossRef] [PubMed]
  182. Sailer, S.; Keller, M.A.; Werner, E.R.; Watschinger, K. The Emerging Physiological Role of AGMO 10 Years after Its Gene Identification. Life 2021, 11, 88. [Google Scholar] [CrossRef] [PubMed]
  183. Watschinger, K.; Keller, M.A.; McNeill, E.; Alam, M.T.; Lai, S.; Sailer, S.; Rauch, V.; Patel, J.; Hermetter, A.; Golderer, G.; et al. Tetrahydrobiopterin and Alkylglycerol Monooxygenase Substantially Alter the Murine Macrophage Lipidome. Proc. Natl. Acad. Sci. USA 2015, 112, 2431–2436. [Google Scholar] [CrossRef] [PubMed]
  184. Werner, E.R.; Hermetter, A.; Prast, H.; Golderer, G.; Werner-Felmayer, G. Widespread Occurrence of Glyceryl Ether Monooxygenase Activity in Rat Tissues Detected by a Novel Assay. J. Lipid Res. 2007, 48, 1422–1427. [Google Scholar] [CrossRef]
  185. Pastor, C.M.; Williams, D.; Yoneyama, T.; Hatakeyama, K.; Singleton, S.; Naylor, E.; Billiar, T.R. Competition for Tetrahydrobiopterin between Phenylalanine Hydroxylase and Nitric Oxide Synthase in Rat Liver. J. Biol. Chem. 1996, 271, 24534–24538. [Google Scholar] [CrossRef]
  186. Opladen, T.; López-Laso, E.; Cortès-Saladelafont, E.; Pearson, T.S.; Sivri, H.S.; Yildiz, Y.; Assmann, B.; Kurian, M.A.; Leuzzi, V.; Heales, S.; et al. Consensus Guideline for the Diagnosis and Treatment of Tetrahydrobiopterin (BH) Deficiencies. Orphanet J. Rare Dis. 2020, 15, 126. [Google Scholar] [CrossRef]
Figure 1. Immunosuppressive effect of BH4 in transplantation. (a) NOSs are identified as treatment targets for BH4-related immunosuppression in ischemia reperfusion injury and chronic rejection while NOS could be excluded as target for acute rejection. (b) Due to its upregulation in M2 macrophages that are linked with enhanced lipolysis, FAO and OXPHOS, AGMO might be the target for BH4-related immunosuppression in acute rejection.
Figure 1. Immunosuppressive effect of BH4 in transplantation. (a) NOSs are identified as treatment targets for BH4-related immunosuppression in ischemia reperfusion injury and chronic rejection while NOS could be excluded as target for acute rejection. (b) Due to its upregulation in M2 macrophages that are linked with enhanced lipolysis, FAO and OXPHOS, AGMO might be the target for BH4-related immunosuppression in acute rejection.
Ijms 24 08257 g001
Table 1. Functional characteristics and markers of different macrophage types *.
Table 1. Functional characteristics and markers of different macrophage types *.
MΦ TypeM1AtypicalM2aM2bM2cM2dMregTRM
stimulation/activationIFNγ
LPS
GM-CSF
DAMPs
PAMPs
mycobacterial infection
IL-33
IL-4
IL-13
fungal and helminth infection
ICs
IL-1R
LPS
IL-10
TGFβ
GCs
IL-6
LIF
adenosine
M-CSF
IFNγ
tissue environment
IL-4
IL-13
IL-10
C1q, C3b
normal
flora (?)
functional characteristicsCD16
CD32
CD64
CD68
CD80
CD86
CD115
CCR7
MHCII
IL-1R
TLR2
TLR4
COX2
SOCS3
MARCO
iNOS
IDO
CD163
CD206
MGL1
YM1
iNOS
CD163
CD206
CD209
CD200R
IL-1R
MHCII
Dectin-1
P2x7R
SR
TGM2
DecoyR
ARG1
YM1/2
FIZZ1
ALOX15
[43,44]
CD86
MHCII
CD163
TLR1
TLR8
IL-21R
MERTK

VEGF
CD301
CD209
CD169
CD11a
Podoplanin
CD127
CD204
CD80
iNOS
DHRS9
XCR1
CD123
CD163
CD169
CD172a
CD204
CD206
CD304
MER
IDO
Axl
CD192 (CCR2)
CD68
CD115
CD206
CD273
(PD-L2)
CD369 (Dectin-1)
MHCII
CD163
MGL-1
CD209
ARG1
iNOS
[45,46,47]
cytokine secretionIFNγ
TNFα
IL-1β
Il-6
IL-12
IL-23
TNFα
IL-6
IL-10
IL-12
IL-10
TGFβ
IL1-RA
IL-10 high
IL-1
IL-6
TNFα
IL-10
TGFβ
IL-10
TGFβ
VEGF
IL-12 low
TNFα low
IL-10
TGFβ
IL-10
chemokine secretionCCL10
CCL11
CCL5
CCL8
CCL9
CCL2
CCL3
CCL4
CCL18CCL17
CCL18
CCL22
CCL24
CCL1CCR2
CXCL13
CCL16
CCL18 [48]
CCL5
CXCL10
CXCL16
CCL1 [49]
CCR1
CCR2
* Red letters highlight markers for mice only, blue letters indicate specific human markers and black letters are used for markers present in both organisms. ALOX15, arachidonate 15-lipoxygenase; FIZZ1/RETNLA/RELMα, resistin-like molecule-alpha; IL-1RA, IL-1 receptor antagonist; LIF, leukocyte inhibitory factor; MARCO, macrophage receptor with collagenous structure; MERTK, proto-oncogene tyrosine-protein kinase MER; MGL1, macrophage galactose-type lectin-1; TGM2, transglutaminase 2; MMR (CD206), macrophage mannose receptor; SR, scavenger receptor; YM1/CHI3L3, chitinase-3-like protein-3; information adapted from [39,40,42,50,51,52,53,54,55,56].
Table 2. Main effects of maintenance immunosuppressive agents on macrophages.
Table 2. Main effects of maintenance immunosuppressive agents on macrophages.
ImmunosuppressiveMode of ActionEffect on MacrophagesRef.
rapamycin inhibition of mTOR (kinase)impairs macrophage development/infiltration
reduces inflammatory cytokine production
inhibits polarization to M2-like macrophages
promotes development of regulatory macrophages
[97,120,121,122,123,124]
tacrolimusinhibition of calcineurin (phosphatase)lowers pro-inflammatory cytokine production
reduces expression of M1 surface marker
shifts surface markers to M2 phenotype
reduces response to LPS
reduces NF-κB activation
[125,126,127,128]
cyclosporine Ainhibition of calcineurin (phosphatase)reduces response to LPS
reduces NF-κB activation
increases macrophage filtration and fibrosis (compared to tacrolismus)
[129,130,131,132]
mycophenolate mofetilinhibition of nucleotide synthesislowers pro-inflammatory mediators
favors M2-like macrophage phenotype
increases M2-like surface markers on M1-stimulated macrophages
reduces monocyte/endothelial cell interactions
[126,133,134,135]
glucocorticoidsregulation of gene expressiondownregulates pro-inflammatory cytokines
influences NO production
increases macrophage infiltration
induces a M2-like phenotype
promotes phagocytosis
mimics pro-fibrotic phenotype
[136,137,138,139,140,141,142,143]
belataceptblocking of co-stimulatory molecules CD80/86enhances phagocyte cell-surface markers
reduces migration capacity
reduces pro-inflammatory cytokine production
induces M1 to M2 macrophage shift
[144,145,146,147,148]
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

Lackner, K.; Ebner, S.; Watschinger, K.; Maglione, M. Multiple Shades of Gray—Macrophages in Acute Allograft Rejection. Int. J. Mol. Sci. 2023, 24, 8257. https://doi.org/10.3390/ijms24098257

AMA Style

Lackner K, Ebner S, Watschinger K, Maglione M. Multiple Shades of Gray—Macrophages in Acute Allograft Rejection. International Journal of Molecular Sciences. 2023; 24(9):8257. https://doi.org/10.3390/ijms24098257

Chicago/Turabian Style

Lackner, Katharina, Susanne Ebner, Katrin Watschinger, and Manuel Maglione. 2023. "Multiple Shades of Gray—Macrophages in Acute Allograft Rejection" International Journal of Molecular Sciences 24, no. 9: 8257. https://doi.org/10.3390/ijms24098257

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