Open access peer-reviewed chapter

Wnt Signaling Regulates Macrophage Mediated Immune Response to Pathogens

Written By

Suborno Jati and Malini Sen

Submitted: 04 February 2019 Reviewed: 19 April 2019 Published: 04 July 2019

DOI: 10.5772/intechopen.86433

From the Edited Volume

Macrophage Activation - Biology and Disease

Edited by Khalid Hussain Bhat

Chapter metrics overview

1,269 Chapter Downloads

View Full Metrics

Abstract

Infection with pathogenic microbes is a global threat. Macrophages play a fundamental role in promoting host resistance to deadly infections from pathogenic microbes by virtue of a well-orchestrated immune defense system. Phagocytosis and obliteration of invading pathogens by macrophages are an innate immune function that not only sustains immune homeostasis but also bolsters adaptive immune response through antigen processing and presentation. Wnt signaling, where Wnt, a secreted glycoprotein which interacts with Frizzled and ROR cell surface receptors to initiate cellular interactions, could be vital for the immune response executed and propagated by macrophages in both innate and adaptive immune responses. The goal of this chapter is to describe how Wnt signaling influences phagocytosis, autophagy, and transcriptional activation to enable the macrophage to exercise its immune response program to resist infection.

Keywords

  • macrophage
  • Wnt
  • phagocytosis
  • actin cytoskeleton
  • transcription
  • immunity

1. Introduction

1.1 Macrophages: innate and adaptive immunity

Macrophages are present as crucial members of a multitude of specialized cells that fortify our immune system by fighting against infection caused by pathogens [1]. Macrophages differentiate from tissue-infiltrated circulating monocytes, which originate from bone marrow resident myeloid precursors [2, 3]. All tissue macrophages, however, do not originate from monocytes. Although some macrophage origins have been studied carefully, the detailed molecular mechanisms toward the differentiation of different macrophage types remain mostly uncharacterized [4, 5, 6, 7]. Irrespective of their origin, most macrophages eliminate encountered pathogens through phagocytosis (element of innate immunity) and additionally present the foreign antigens derived from pathogens via major histocompatibility complex (MHC) molecules to lymphocytes leading to lymphocyte activation (element of adaptive immunity) [2, 8]. Cytoskeletal modulations and transcriptional activation programs intrinsically associated with macrophage-mediated immune functions (e.g. phagocytosis, autophagy/xenophagy) conform to the in-built maneuvering of macrophages as they confront with different kinds of pathogens. Several lines of evidence substantiate that Wnt signaling is important for the transcriptional programs and cytoskeletal modulations inherent to macrophages during immune surveillance and response to different kinds of infection [9, 10, 11, 12, 13].

1.2 Wnt signaling

Wnt signaling is an integral theme of tissue/organ morphogenesis, repair, and maintenance. Thus, it is not surprising that this central premise of life is also an important component of macrophage function [9, 10, 11, 12, 13, 14, 15, 16]. Wnts constitute a large family of secreted glycoprotein ligands, which bind to Frizzled and/or ROR cell surface receptors during various phases of tissue and organ development, morphogenesis, and homeostasis. Frizzleds are seven transmembrane-spanning receptors bearing homology to heterotrimeric G protein-coupled receptors, and RORs bear homology to tyrosine kinase receptors [17, 18, 19, 20]. Based on the gene database, there are about 19 Wnt ligands and about 12 and 2 Frizzled and ROR receptors, respectively [21, 22]. Whether all these gene products are expressed and functional in our system in different cellular contexts is unclear at this stage. Although there is evidence of co-receptor function by the ROR subtype receptors during Wnt-Frizzled signaling [22, 23], the degrees of coordination between the Frizzled and ROR receptors under different physiological conditions are yet to be characterized at the molecular level. Given the considerable homology among the respective members of the Wnt and Frizzled families, any one Wnt ligand may interact with multiple Frizzled receptors. Thus, the outcome of Wnt-Frizzled signaling in a particular cell type under a certain condition could be dependent precisely on the existing profile of Wnt-Frizzled stoichiometry [20].

Wnt signaling is broadly classified into two types—canonical or β-catenin-dependant and noncanonical or β-catenin-independent (Figure 1). The transcriptional coactivator β-catenin promotes gene expression by LEF/TCF family transcription factors in response to canonical Wnt signaling, and transcriptional activators such as NFκB, NFAT, and AP1 are associated with noncanonical Wnt signaling. Even though the ligands Wnt3A and Wnt5A are mostly considered as representatives of the canonical and noncanonical modes of Wnt signaling, respectively [21, 24], the mode of signaling is in reality governed by the receptor(s) receiving the Wnt signal as mentioned above and the associated adaptor molecule(s) transmitting it. Thus, some level of crosstalk between the two modes of signaling would not be uncommon. Interestingly, the intracellular adaptor molecule Disheveled acts as a mediator of both β-catenin-dependant and β-catenin-independent Wnt signaling. Heterotrimeric G proteins, which have been reported to couple with Frizzled receptors, add to the complexity of Wnt signaling [18, 25]. Whether heterotrimeric G proteins cooperate with Disheveled during canonical and noncanonical Wnt signaling is not known clearly. Although there is some evidence of the involvement of lipid molecules such as cholesterol in switching Disheveled between the canonical and noncanonical modes of Wnt signaling [25], the molecular details of such presumed conformational switches remain largely undefined. The reason behind the preference of cell surface coactivator receptors such as lipoprotein receptor-like protein (LRP) 5/6 for the canonical mode of Wnt signaling as opposed to the noncanonical mode also remains unclear (Figure 1).

Figure 1.

An overview of Wnt signaling cascade: in canonical mode of signaling, the association of Wnt-Fz and LRP activates a signaling cascade through Dvl and/or G-proteins that leads to inactivation of a GSK3 associated destruction complex which in the absence of Wnt would phosphorylate β-catenin for terminal destruction by proteasome. Via GSK3 inactivation, β-catenin gets stabilized and translocates to the nucleus where it acts as a co-activator of LEF/TCF (transcription factor). In the non-canonical mode of Wnt signaling (often β-catenin independent) the signaling cascade through Dvl and/or G-protiens leads to activation of Ca2+ mediated signaling where protein kinase C (PKC) and CaMKII gets activated and leads to translocation of NFκB, NFAT to the nucleus. Wnt also binds to ROR leading to activation of AP1. A crosstalk between the pathways is not uncommon.

1.3 Wnt signaling in immune system

Given that host cytoskeletal rearrangements encompassing phagocytosis and autophagy/xenophagy and transcriptional regulation of immune defense genes come into the direct line of control of pathogenic incursions and immune homeostasis [9, 10, 11, 12, 26], Wnt signaling aptly associates with host-pathogen interactions of macrophages at the crossroads of innate and adaptive immunity. The attributes of Wnt signaling and the microbe world being diverse, their mutual interactions in the various host defense programs are expected to be manifold. Although Wnt3A and Wnt5A are often represented as the prototypes for the two different modes of Wnt signaling (canonical and noncanonical) in the regulation of immune response, several molecular details of the balancing act of the Wnts in relation to the interactions of macrophages with different microbes remain unclear.

The primary objective of this chapter is to briefly summarize the conceptual advancement in the context of Wnt signaling and immune defense by macrophages, focusing mainly on transcriptional activation and the actin cytoskeleton-associated phagocytosis and autophagy machineries. Our aim is to also address unanswered questions, which may prove instrumental in bridging existing gaps in our evaluation of the Wnts in the context of macrophage host defense programs.

Advertisement

2. Sustenance of immune defense by macrophages through a steady state of transcriptional activation by Wnt signaling

2.1 Significance of constitutive transcriptional activation in macrophages by Wnt signaling

Macrophages have long been acknowledged for executing immune defense against microbial pathogens through diverse means of signaling that include several transcription factors including NFκB, AP1, and NFAT [27, 28, 29, 30]. The ability of macrophages to recognize and engulf pathogens, deliberate NADPH oxidase activity, and process antigens for presentation to MHC molecules and T cell activation place macrophages quite aptly at the crossroad of innate and adaptive immune defense programs [31, 32, 33]. Surely, macrophages have in-built mechanisms to execute innate immunity and translate it to adaptive immune response. However, not much is known about the molecular details of how macrophages are naturally geared to operate in such innate and adaptive modes of immune defense. We recently demonstrated that NF-κB (p65) [34], a transcription factor functioning at the core of our immune system, remains activated at a basal level in macrophages through a steady state of Wnt5A signaling. Administration of inhibitor of Wnt production2 (IWP2) to macrophages in culture or depletion of Wnt5A or Frizzled5 (putative Wnt5A receptor) gene expression in macrophages by silencing gene transcription through small interfering RNA blocks constitutive p65 activation and the steady-state immune activity of macrophages [10]. Sustained presence of the Wnt5A-p65 axis can potentially bridge innate and adaptive immune responses through regulation of the expression of immune response genes, such as CD14, interferons (IFN)s, and MHC, and elaboration of immune signaling networks that involve major immune response molecules such as the Toll-like receptors (TLR) and nucleotide-binding oligomerization domain-containing proteins (NOD) during challenge by pathogens [13, 35, 36]. The interrelation of this basal level Wnt5A-p65 signaling with other major transcription factors and coactivators of Wnt signaling that mediate immune response by macrophages remains to be deciphered at the molecular level.

2.2 NF-κB transcription factors

NF-κB transcription factors comprise a family of five members: p52, p50, p65 (RelA), c-Rel, and RelB, which regulate gene transcription as combinatorial dimers [34, 37, 38]. These dimers remain or become activated through different modes depending on the physiological context of cell signaling. In the classical mode of activation, the homo and heterodimers are translocated to the nucleus for gene expression after being released from the IκB-bound states in the cytoplasm in response to different stimuli that lead to proteasome-assisted IκB degradation through activation of the IκB kinase IKK2/β [34]. The p65 homo and heterodimers while being responsible for inflammatory gene expression are also significantly involved in the sustenance of innate immune response gene expression in a context-dependent manner [10]. Some of the NF-κB (p65) responsive immune response genes include CD14, MHC, and IFNs. A schematic of NF-κB activation is shown in Figure 2.

Figure 2.

An overview of NFKB activation pathway in the macrophage: During steady state a basal level of stimulus by Wnt signaling keeps IKK enough activated to result in inactivation of IκB and translocation of a certain pool of NFκB transcription factor (p65 homodimer) to the nucleus. A minimum pool of transcription factors contributes to survival and vigilance for immune response. In the activated state, during inflammation and chronic infection, stimuli (TNFα, LPS, IL1β) lead to an increase in NFκB combinatorial dimers in the nucleus.

2.3 Wnt5A signaling-mediated activation of transcription

As mentioned earlier in this chapter, Wnt5A is one of several members of the large family of Wnt glycoprotein ligands. Frizzled-5, Frizzled-4, and ROR1 are putative receptors for Wnt5A. It is to be noted that although modified versions of selective Wnt-Frizzled complex structures have been solved [39], none of the ligand-receptor complexes have been truly biochemically characterized in their physiological contexts. In the noncanonical mode of Wnt signaling of which Wnt5A is a representative, Wnt5A-Frizzled-ROR or Wnt5A-Frizzled-initiated signaling alters the activity of Rho/Rac family GTPases through differential activation of Disheveled [10, 40]. Within the Frizzled family of cell surface receptors, Frizzled2, Frizzled5, and Frizzled4 are some of the putative receptors for Wnt5A [17, 41, 42]. It is not known if Disheveled activation by Wnt5A signaling acts in concert with or is regulated by heterotrimeric G proteins, given that Frizzled receptors are homologous to heterotrimeric G protein-coupled receptors. The involvement of β-catenin by Wnt5A signaling is governed by the availability of receptors and cytoplasmic signaling intermediates [20, 43]. The subsequent activation of transcription factors such as AP1, NFAT, and NF-κB through complex signaling networks and crosstalk, either dependent or independent of nuclear translocation of β-catenin (explained in Figures 1 and 2), could lead to elaboration of context-dependent immune responses (Figure 3).

Figure 3.

A schematic of Wnt5A-p65 axis: Wnt5A binds with it’s putative receptor Frizzled5 (FZ) and transmits signal through intermediates like Disheveled (Dvl), trimeric G-proteins (Gα, β, ƴ ) activating Rac1. Activated Rac1 helps in translocation of NFκB from cytosol to nucleus via activation of IKK and proteasomal degradation of IKK-phosphorylated IκB. The translocated p65 in the nucleus helps to maintain expression of proteins such as CD14, IFNƴ, IFNb, MHC, needed for pathogen detection and clearance, and Bcl2, needed for cell survival. Amplification of signals by CD14-assisted molecules such as TLRs facilitate pathogen recognition and clearance.

The basal Wnt5A-Frizzled5 signaling-dependent NF-κB (p65) activity in macrophages that we observed is at least partly accountable for the steady-state expression of CD14/IFNβ, the promoter sequence of which at the genome level contains p65 binding elements [10, 13] (Figure 3). The constitutive p65 activity in the nucleus also contributes to sustaining Wnt5A expression [10]. Accordingly, the self-sustaining Wnt5A-p65 axis responsive CD14 and IFNβ expression helps to initiate and coordinate several aspects of macrophage function including interaction of pathogen recognition with TLR signaling, thus enabling adaptation to protective immune responses to bacteria, bacterial LPS (lipopolysaccharide), and virus as explained in Figure 3. The Wnt5A-NF-κB (p65) responsive gene expression declines upon exposing macrophages to an IKK2-specific inhibitor [10]. Wnt5A signaling is also responsible for a basal level of secretion of IFN-γ, another important regulator of innate immune signaling in macrophages. The steady-state Wnt5A signaling and NF-κB activity also promote macrophage survival through the expression of NF-κB-responsive survival genes such as Bcl2 [10]. These data are consistent with the dearth of survival of NF-κB-deficient mice due to different kinds of infection and apoptotic cell death [44]. The Wnt5A-Frizzled5 signaling-assisted constitutive p65 activity is dependent on Rac1 activation, which lies upstream of IKK2 activity [10]. The detailed mechanism of how the Rac1 GTPase activates IKK in a Wnt5A signaling-dependent mode is yet to be explored. It also remains to be tested how Wnt5A-responsive innate immune functions in macrophages relating to pathogen recognition and activation of several intracellular signaling pathways translate to adaptive immune responses encompassing antigen processing/presentation and lymphocyte activation.

2.4 Signaling and transcriptional activation by other Wnts

In light of the fact that Wnts comprise a large family of glycoprotein ligands sharing considerable amino acid sequence homology and bind to cell surface receptors that are equally homologous [21], the schemes of regulation and sustenance of immune responses in macrophages by Wnt signaling are likely to be manifold. Several reports have outlined the importance of canonical Wnt signaling and β-catenin in the development, sustenance, and elaboration of memory and effector T cells that comprise a crucially important component of immunity to infectious pathogens [45]. The role of the TCF family of transcription factors in this respect has generated considerable interest in our understanding of the importance of Wnt signaling in immune homeostasis. However, the precise role of canonical Wnt signaling by β-catenin and TCF transcription factors in macrophages in the generation and sustenance of T cell-mediated immunity remains unclear.

Advertisement

3. Role of Wnt signaling in macrophage phagocytosis: involvement of the actin cytoskeleton

3.1 Significance of phagocytosis

Phagocytosis of pathogens is one of the most important features of the host-pathogen communications and interactions mediated by macrophages. This element of host defense by macrophages not only operates toward host protection at the onset of infection but also makes room for the initiation and amplification of intracellular signals that can potentially mature to the generation of antigen-specific T cell responses and creation of immunological memory (explained in Figure 4).

Figure 4.

A schematic of maturation of pathogen containing vesicle and its outcome: After phagocytosis of pathogen there is fusion of early endosome and endoplasmic reticulum (ER) with the phagosome which helps in the maturation of the phagosome and fusion with lysosome. This is important for both innate and adaptive immunity.

As described earlier in this chapter, Wnt5A signaling aids in maintaining a steady-state expression of CD14 and IFNβ, two of the many molecules involved in innate immune defense. Although it is not exactly clear how CD14 and IFNβ fit into the program of phagocytosis in exact molecular terms, it is documented that while CD14 is instrumental in the recognition of structural motifs like lipopolysaccharide (LPS) intrinsic to certain pathogens, both CD14 and IFNβ facilitate pathogen clearance through the initiation and propagation of macrophage TLR signaling during phagocytosis and activation of immune responses [10, 13] (Figure 3). Following pathogen engulfment and phagosome formation during phagocytosis, macrophages rely mostly on endosomal and lysosomal proteases and NADPH oxidase-generated reactive oxygen species for both pathogen clearance as well as processing and presentation of antigenic peptides to MHC molecules for presentation to T lymphocytes [31, 46] and translation to memory.

3.2 Need for Wnt5A signaling-assisted actin rearrangement/assembly

At the core of all phagocytosis-related processes lies the involvement of the actin cytoskeleton through its influence on protein sorting/trafficking and intracellular organelle fusions that are crucial for the activation of phagosomal enzymes such as NADPH oxidase and phagosome maturation [31, 47]. Several cytoskeletal GTPases such as Rac1 and Disheveled, lipid rafts, and actin-nucleating proteins such as Arp2/3 and formins partake of the cytoskeletal actin modulations that accompany macrophage phagocytosis and phagosome maturation [47, 48, 49, 50]. There is evidence that Wnt5A signaling is important for such rearrangements of the actin cytoskeleton. Accordingly, Wnt5A signaling facilitates Rac1- Disheveled-lipid raft-dependent phagocytosis of bacteria and other foreign matter through modulations of the actin cytoskeleton [9]. Blockers of any of the cytoskeletal actin-associated signaling intermediates—Rac1, Disheveled, or lipid raft and cytochalasin-D, an inhibitor of actin assembly—are antagonistic to the effect of Wnt5A signaling on phagocytosis [9]. The influence of Wnt5A signaling on phagocytic uptake is usually dependent on the microbe under consideration, because while most bacterial species tested undergo facilitated phagocytic uptake by Wnt5A signaling in macrophages, phagocytic uptake of Leishmania donovani remains unaffected by it [11]. Perhaps Wnt5A-facilitated internalization encompasses distinct membranous domains depending on the availability of cognate receptors, which are not equally compatible with all microbes. That Wnt5A signaling also facilitates phagosome-lysosome fusion during phagosome maturation which is evident from the augmented appearance of lysosomal markers such as cathepsins in Wnt5A-induced phagosomes of bacteria-infected macrophages [12]. Wnt5A-facilitated alteration in cytoskeletal actin assembly that correlates with phagosome-lysosome fusion is concomitant with the killing of several microbes including bacterial pathogens (Pseudomonas aeruginosa, Streptococcus pneumoniae, etc.) and even Leishmania donovani, although it gets internalized independent of Wnt5A signaling [11, 12]. The mechanism of microbial killing is discussed at greater length in the following section of this chapter. Microbial killing is furthermore facilitated by Wnt5A-responsive NADPH oxidase activity, which is associated with cytoskeletal actin-dependent assembly of NADPH oxidase subunits [11]. Interestingly, nonpathogenic laboratory strains of bacteria that are engulfed by macrophages in increased numbers by Wnt5A signaling are not necessarily killed by it like the pathogenic bacterial strains [9, 12]. Such discrepancy in the fate of internalized microbes may be an outcome of notable differences in the interaction of different microbial components with Wnt5A-regulated cytoskeletal actin rearrangements. The interrelation between Wnt5A signaling and Ehrlichia infection is especially noteworthy in this context [51].

In light of the fact that the cytoskeletal actin-assisted phagosome is the originator and communicator of many signals generated by phagocytozed cargo-recognizing molecules such as TLR, NOD1, and NOD2 [35, 52, 53] (Figure 4), it is quite likely that the consequences of Wnt5A-assisted phagocytosis are numerous. Association of Wnt5A signaling with TLRs has already been reported [54]. Careful analysis of the consequences of such associations is important.

3.3 Role played by other Wnts and costimulatory molecules of Wnt signaling

Wnts other than Wnt5A are known to regulate macrophage phagocytosis as well. For example, the Drosophila Wnt has been reported to stimulate phagocytic uptake in the S2 cell, a macrophage-like line [55]. Moreover, Wnt1, Wnt7A, and Wnt3A have been reported as phagocytic modulators [56, 57]. The association or relation of these different modes of phagocytosis with Wnt5A signaling and cytoskeletal actin rearrangements is yet to be explored. At this point of our understanding of Wnt signaling with respect to phagocytosis, regulatory roles played by LRP5/6 and ROR, which act as co-receptors to Wnts [22, 58], remain unclear. It also remains to be seen if the influence of Wnt5A signaling on phagocytosis is in the canonical or noncanonical mode or is in fact an intermediary between the two depending on the context of infection, the available receptors, and coactivators.

Advertisement

4. Wnt signaling-induced actin-dependent autophagy-assisted xenophagy by macrophages and the potential link with antigen processing/presentation

4.1 Autophagy-assisted xenophagy

Several pathogenic microorganisms try to adapt to the intracellular milieu of macrophage creating a niche for their survival [59, 60, 61]. Nevertheless, as described earlier in this chapter, the host macrophage tries maneuvering elimination of infection by pathogens by several means. It has been reported that following phagocytosis of microbes by macrophages, the host autophagy machinery comes into play in the ultimate event of clearance of bacteria and other engulfed microbes (xenophagy) through coordinated alterations of the actin cytoskeleton. Autophagy involves the turnover and clearance of damaged organelles and proteins by the cell under both normal conditions as well as under stress in the maintenance of cellular homeostasis [62, 63]. During infection with pathogens, the autophagy program is often utilized for the incapacitation and eradication of engulfed pathogens [26, 64].

4.2 Role of Wnt signaling and cytoskeletal actin in autophagy-assisted xenophagy

Wnt signaling has been reported to play a significant role in the autophagy-assisted xenophagy of engulfed microbes by macrophages. Wnt5A signaling, for instance, has been documented to be an integral component of this theme in the killing of several bacterial pathogens through utilization of a Rac1-Disheveled-actin cytoskeleton circuit that involves interactions among several autophagy-associated proteins like microtubule-associated protein 1B-light chain 3B (LC3B), autophagy-related 5 (ATG5), ATG7, and Unc-51-like autophagy-activating kinase 1 (ULK1) [12]. The different nuances of Wnt5A signaling in connection with the actin cytoskeleton are depicted in Figure 5. Pathogen killing through autophagy machinery is blocked with the use of cytochalasin-D, an inhibitor of actin assembly as well as with inhibitors to Rac1 and Disheveled [12]. Although Wnt5A-assisted killing of L. donovani in macrophages has not been shown to directly involve autophagy, electron micrographs of L. donovani harboring parasitophorous vacuoles, which display distinct membranous aggregates, suggest that L. donovani containing parasitophorous vacuoles may be subjected to lysis by the host autophagy circuit activated by Wnt5A signaling [11]. The inactivation or lysis of microbe-carrying vacuoles, which happens in due course through fusion of autophagy-destined phagosome or autophagosome with the lysosome, may also be facilitated by Wnt5A signaling [12]. Although cholesterol and other lipids are known to partake of both Wnt5A signaling and actin dynamics [65, 66], at this stage much remains unknown about the specific roles of cholesterol and other lipids in the process of actin modulation during phagocytosis and autophagic clearance of bacteria and other microbes. It also remains to be seen if Wnt5A signaling during autophagy belongs strictly to the noncanonical mode or canonical mode based on the involvement of β-catenin.

Figure 5.

Schematic of Wnt5A signaling aided bacterial killing: Both autocrine and paracrine modes of Wnt5A signaling can lead to increase in phagocytosis of pathogenic bacteria. After phagocytosis, the Wnt5A mediated cytoskeletal modulation leads to fusion of early endosome and lysosome with the pathogen containing phagosome. Wnt5A signaling also activates Rac1 and Unc like kinase 1 (Ulk1) for initiation of autophagy. The subsequent steps of maturation lead to killing of pathogen in an autophagy dependent process (xenophagy).

4.3 Potential link with antigen presentation/adaptive immunity

In view of the fact that the autophagic or rather xenophagic removal of pathogens by macrophages involves reorganization and fusion of intracellular vesicles associated with at least partial lysis of pathogens, the processing and presentation of pathogen antigens to MHC molecules are a likely event during xenophagy in infected macrophages [67, 68]. Thus, autophagosome formation, autophagosome lysosome fusion, and T cell activation by the presentation of processed pathogenic antigens may prevail as a continuum during immune defense depending on the nature and degree of the infection. Given the intrinsic association of Wnt signaling with cytoskeletal dynamics and autophagy [11, 12], it is quite likely that Wnt signaling will influence the antigen processing and presentation linked with autophagy in infected macrophages. Detailed investigation in this respect, although important, remains to be documented.

Advertisement

5. Concluding remarks

Given the important role played by Wnt ligands in the transmission of signals associated with cytoskeletal modulation and transcriptional regulation which are part and parcel of host-pathogen communications [27, 28, 29, 69], a combination of Wnt signal transduction cascades is expected to hold a fundamental standing in the immune defense program operated by macrophages in both innate and adaptive immunity. Phagocytosis, autophagy/xenophagy (intracellular microbial killing), and a steady-state expression of immune defense molecules through transcriptional regulation appear as some of the major players of the immune defense program operated by Wnt signaling.

In respect of transcriptional regulation of immune defense molecules by steady-state Wnt5A-signaling as described in this chapter [10], it is not understood exactly what dictates the nuclear translocation of p65 and not the other NFκB isoforms for specific modes of gene expression. Additionally, how this regulation fits in with the activity of other major transcription factors like NFAT and AP1 in the macrophage is also not clearly understood. Moreover, details of the context dependence of Wnt5A signaling, wherein a certain level and mode of signal transmission will be beneficial for immune response, but excess will cause inflammation and disorder [70, 71, 72], remain largely unclear. Besides, a clear concept of how actin cytoskeleton-associated proteins such as Rac1 promote both NFκB activity as well as cytoskeletal rearrangements for phagocytosis and autophagy is yet to be achieved [10, 12]. Whether nuclear translocation of NFκB is a natural function of actin assembly or is executed by a separate pool of Rac1 associated cytoskeletal proteins is an important matter that deserves investigation.

With regard to phagocytosis and autophagy-assisted xenophagy, the molecular details of the actin rearrangements with actin binding proteins and the processing and presentation of antigens remain to be deciphered. This brings into question how different host-pathogen interactions within macrophages are guided by modulations of the actin cytoskeleton. Of special interest in this context is the interaction of the actin cytoskeleton with pathogenic mycobacteria, which thrive in self-generated niches within macrophages [60, 73]. The interrelation between different modes of Wnt signaling and mycobacterial infection, although much studied [74, 75], needs to be better understood with respect to actin dynamics. Now that Wnt5A signaling has been shown to play a major role in the regulation of actin cytoskeletal modulation and autophagy [11, 12, 76], future experiments addressing whether this can also facilitate the adaptive immune response through antigen processing and presentation may prove fruitful.

At this juncture of our understanding of Wnt signaling and immune response by macrophages, it is important to know how the different Wnt ligands operate in the regulation of immune response by the different types of macrophages that are distributed in different tissues under the varied conditions of intracellular milieu and infection. Macrophages (microglia) present in the brain and spinal cord maintain an active immune defense scheme against pathogens that affect the central nervous system. Alveolar and airway macrophages likewise protect the respiratory tract and lungs from the toxic effect of infectious agents. Peritoneal macrophages of the peritoneum and Kupffer cells of the liver also encounter and confront infectious agents for host protection. Quite naturally, the roles played by Wnt signaling in the combat mechanism of each macrophage type in its paradigm of immune defense is expected to be different at least to some extent on account of potential variations in cellular environmental cues and modes of host-pathogen interactions.

Advertisement

Acknowledgments

This work was supported by DBT, Government of India (BT/PR7106/MED/29/639/2012), Institutional funding (BSC0114, BSC0116). SJ was supported by the Research Scholar Fellowship from CSIR, Government of India and by The Company of Biologists, Journal of Cell Biology.

Advertisement

Conflict of interest

The authors declare that there is no conflict of interest.

References

  1. 1. Gordon S. The macrophage: Past, present and future. European Journal of Immunology. 2007;37(S1):S9-S17
  2. 2. Geissmann F, Auffray C, Palframan R, Wirrig C, Ciocca A, Campisi L, et al. Blood monocytes: Distinct subsets, how they relate to dendritic cells, and their possible roles in the regulation of T-cell responses. Immunology and Cell Biology. 2008;86(5):398-408
  3. 3. Geissmann F, Manz MG, Jung S, Sieweke MH, Merad M, Ley K. Development of monocytes, macrophages and dendritic cells. Science. 2010;327(5966):656-661
  4. 4. Anderson KL, Smith KA, Conners K, McKercher SR, Maki RA, Torbett BE. Myeloid development is selectively disrupted in PU. 1 null mice. Blood. 15 May 1998;91(10):3702-3710
  5. 5. Auffray C, Fogg D, Garfa M, Elain G, Join-Lambert O, Kayal S, et al. Monitoring of blood vessels and tissues by a population of monocytes with patrolling behavior. Science. 2007;317(5838):666-670
  6. 6. Davies LC, Taylor PR. Tissue-resident macrophages: Then and now. Immunology. 2015;144(4):541-548
  7. 7. Epelman S, Lavine KJ, Randolph GJ. Origin and functions of tissue macrophages. Immunity. 2014;41(1):21-35
  8. 8. Hume DA. Macrophages as APC and the dendritic cell myth. Journal of Immunology. 2008;181(9):5829-5835
  9. 9. Maiti G, Naskar D, Sen M. The wingless homolog Wnt5a stimulates phagocytosis but not bacterial killing. Proceedings of the National Academy of Sciences. 2012;109(41):16600-16605
  10. 10. Naskar D, Maiti G, Chakraborty A, Roy A, Chattopadhyay D, Sen M. Wnt5a-Rac1-NF-B Homeostatic circuitry sustains innate immune functions in macrophages. The Journal of Immunology. 2014;192(9):4386-4397
  11. 11. Chakraborty A, Kurati SP, Mahata SK, Sundar S, Roy S, Sen M. Wnt5a signaling promotes host defense against Leishmania donovani infection. Journal of Immunology. 2017;199(3):992-1002
  12. 12. Jati S, Kundu S, Chakraborty A, Mahata SK, Nizet V, Sen M. Wnt5A signaling promotes defense against bacterial pathogens by activating a host autophagy circuit. Frontiers in Immunology. 2018;9:679
  13. 13. Guha I, Naskar D, Sen M. Macrophage as a mediator of immune response: Sustenance of immune homeostasis
  14. 14. Clevers H. Wnt/β-catenin signaling in development and disease. Cell. 2006;127(3):469-480
  15. 15. Logan CY, Nusse R. The Wnt signaling pathway in development and disease. Annual Review of Cell and Developmental Biology. 2004;20:781-810
  16. 16. Staal FJT, Luis TC, Tiemessen MM. WNT signalling in the immune system: WNT is spreading its wings. Nature Reviews. Immunology. 2008;8(8):581-593
  17. 17. Liu X, Liu T, Slusarski DC, Yang-Snyder J, Malbon CC, Moon RT, et al. Activation of a frizzled-2/beta-adrenergic receptor chimera promotes Wnt signaling and differentiation of mouse F9 teratocarcinoma cells via Galphao and Galphat. Proceedings of the National Academy of Sciences of the United States of America. 1999;96(25):14383-14388
  18. 18. Schulte G, Bryja V. The frizzled family of unconventional G-protein-coupled receptors. Trends in Pharmacological Sciences. 2007;28(10):518-525
  19. 19. Wang H, Liu T, Malbon CC. Structure-function analysis of Frizzleds. Cellular Signalling. 2006;18(7):934-941
  20. 20. Mikels AJ, Nusse R. Purified Wnt5a protein activates or inhibits β-catenin–TCF signaling depending on receptor context (Arias AM, editor). PLoS Biology. 2006;4(4):e115
  21. 21. The Wnt Homepage [Internet]. Available from: https://web.stanford.edu/group/nusselab/cgi-bin/wnt/ [Accessed: 31 January 2019]
  22. 22. Green J, Nusse R, van Amerongen R. The role of Ryk and Ror receptor tyrosine kinases in Wnt signal transduction. Cold Spring Harbor Perspectives in Biology. 1 Feb 2014;6(2):a009175
  23. 23. Yu J, Chen L, Cui B, Widhopf GF, Shen Z, Wu R, et al. Wnt5a induces ROR1/ROR2 heterooligomerization to enhance leukemia chemotaxis and proliferation. Journal of Clinical Investigation. 2015;126(2):585-598
  24. 24. Grumolato L, Liu G, Mong P, Mudbhary R, Biswas R, Arroyave R, et al. Canonical and noncanonical Wnts use a common mechanism to activate completely unrelated coreceptors. Genes & Development. 2010;24(22):2517-2530
  25. 25. Aznar N, Ear J, Dunkel Y, Sun N, Satterfield K, He F, Kalogriopoulos NA, et al. Convergence of Wnt, growth factor, and heterotrimeric G protein signals on the guanine nucleotide exchange factor Daple. Science Signaling. 27 Feb 2018;11(519):eaao4220
  26. 26. Bah A, Vergne I. Macrophage autophagy and bacterial infections. Frontiers in immunology. 6 Nov 2017;8:1483
  27. 27. Fric J, Zelante T, Wong AYW, Mertes A, Yu H-B, Ricciardi-Castagnoli P. NFAT control of innate immunity. Blood. 2012;120(7):1380-1389
  28. 28. Newton K, Dixit VM. Signaling in innate immunity and inflammation. Cold Spring Harbor Perspectives in Biology. 1 Mar 2012;4(3):a006049
  29. 29. Foletta VC, Segal DH, Cohen DR. Transcriptional regulation in the immune system: All roads lead to AP-1. Journal of Leukocyte Biology. 1998;63(2):139-152
  30. 30. Zhong B, Tien P, Shu H-B. Innate immune responses: Crosstalk of signaling and regulation of gene transcription. Virology. 2006;352(1):14-21
  31. 31. Rybicka JM, Balce DR, Khan MF, Krohn RM, Yates RM. NADPH oxidase activity controls phagosomal proteolysis in macrophages through modulation of the lumenal redox environment of phagosomes. Proceedings of the National Academy of Sciences of the United States of America. 2010;107(23):10496-10501
  32. 32. Underhill DM, Bassetti M, Rudensky A, Aderem A. Dynamic interactions of macrophages with T cells during antigen presentation. The Journal of Experimental Medicine. 1999;190(12):1909-1914
  33. 33. Brode S, Macary PA. Cross-presentation: Dendritic cells and macrophages bite off more than they can chew! Immunology. 2004;112(3):345-351
  34. 34. Ghosh S, May MJ, Kopp EB. NF-kappa B and Rel proteins: Evolutionarily conserved mediators of immune responses. Annual Review of Immunology. 1998;16:225-260
  35. 35. Zabucchi G, Trevisan E, Vita F, Soranzo MR, Borelli V. NOD1 and NOD2 interact with the phagosome cargo in mast cells: A detailed morphological evidence. Inflammation. 2015;38(3):1113-1125
  36. 36. Garcia-Rodriguez KM, Goenka A, Alonso-Rasgado MT, Hernández-Pando R, Bulfone-Paus S. The role of mast cells in tuberculosis: Orchestrating innate immune crosstalk?. Frontiers in Immunology. 17 Oct 2017;8:1290
  37. 37. Sarnico I, Lanzillotta A, Benarese M, Alghisi M, Baiguera C, Battistin L, et al. NF-kappaB dimers in the regulation of neuronal survival. International Review of Neurobiology. 2009;85:351-362
  38. 38. Sen M, Ghosh G. Transcriptional outcome of Wnt-frizzled signal transduction in inflammation: Evolving concepts. Journal of Immunology. 2008;181(7):4441-4445
  39. 39. Janda CY, Waghray D, Levin AM, Thomas C, Garcia KC. Structural basis of Wnt recognition by frizzled. Science. 2012;337(6090):59-64
  40. 40. Schlessinger K, Hall A, Tolwinski N. Wnt signaling pathways meet Rho GTPases. Genes and Development. 2009;23(3):265-277
  41. 41. He X, Saint-Jeannet J-P, Wang Y, Nathans J, Dawid I, Varmus H. A member of the frizzled protein family mediating axis induction by Wnt-5A. Science. 1997;275(5306):1652-1654
  42. 42. Sato A, Yamamoto H, Sakane H, Koyama H, Kikuchi A. Wnt5a regulates distinct signalling pathways by binding to Frizzled2. The EMBO Journal. 2010;29(1):41-54
  43. 43. Bryja V, Schulte G, Rawal N, Grahn A, Arenas E. Wnt-5a induces dishevelled phosphorylation and dopaminergic differentiation via a CK1-dependent mechanism. Journal of Cell Science. 2007;120(4):586-595
  44. 44. Sha WC, Liou HC, Tuomanen EI, Baltimore D. Targeted disruption of the p50 subunit of NF-kappa B leads to multifocal defects in immune responses. Cell. 1995;80(2):321-330
  45. 45. Staal FJT, Arens R. Wnt Signaling as master regulator of T-lymphocyte responses: Implications for transplant therapy. Transplantation. 2016;100(12):2584-2592
  46. 46. Vyas JM, Van der Veen AG, Ploegh HL. The known unknowns of antigen processing and presentation. Nature Reviews. Immunology. 2008;8(8):607-618
  47. 47. Blocker A, Severin FF, Burkhardt JK, Bingham JB, Yu H, Olivo J-C, et al. Molecular requirements for bi-directional movement of phagosomes along microtubules. The Journal of Cell Biology. 1997;137(1):113-129
  48. 48. Rotty JD, Brighton HE, Craig SL, Asokan SB, Cheng N, Ting JP, et al. Arp2/3 complex is required for macrophage integrin functions but is dispensable for FcR phagocytosis and in vivo motility. Developmental Cell. 2017;42(5):498-513 e6
  49. 49. Clarke M, Engel U. Mechanically induced actin-mediated rocketing of phagosomes. Molecular Biology of the Cell. 2006;17:10
  50. 50. Nagao G, Ishii K, Hirota K, Makino K, Terada H. Role of lipid rafts in innate immunity and phagocytosis of polystyrene latex microspheres. Colloids and Surfaces. B, Biointerfaces. 2011;84(2):317-324
  51. 51. Luo T, Dunphy PS, Lina TT, McBride JW. Ehrlichia chaffeensis exploits canonical and noncanonical host Wnt signaling pathways to stimulate phagocytosis and promote intracellular survival. Infection and Immunity. 2016;84(3):686-700
  52. 52. Kong L, Ge B-X. MyD88-independent activation of a novel actin-Cdc42/Rac pathway is required for toll-like receptor-stimulated phagocytosis. Cell Research. 2008;18(7):745
  53. 53. Blander JM, Medzhitov R. Regulation of phagosome maturation by signals from toll-like receptors. Science. 2004;304(5673):1014-1018
  54. 54. Trinath J, Holla S, Mahadik K, Prakhar P, Singh V, Balaji KN. The WNT signaling pathway contributes to dectin-1-dependent inhibition of toll-like receptor-induced inflammatory signature. Molecular and Cellular Biology. 2014;34(23):4301-4314
  55. 55. Zhu F, Zhang X. The Wnt signaling pathway is involved in the regulation of phagocytosis of virus in Drosophila. Scientific Reports. 25 Jun 2013;3:2069
  56. 56. Wallace J, Lutgen V, Avasarala S, St Croix B, Winn RA, Al-Harthi L. Wnt7a induces a unique phenotype of monocyte-derived macrophages with lower phagocytic capacity and differential expression of pro- and anti-inflammatory cytokines. Immunology. 2018;153(2):203-213
  57. 57. Chen K, Fu Q , Li D, Wu Y, Sun S, Zhang X. Wnt3a suppresses Pseudomonas aeruginosa-induced inflammation and promotes bacterial killing in macrophages. Molecular Medicine Reports. 2016;13(3):2439-2446
  58. 58. Goel S, Chin EN, Fakhraldeen SA, Berry SM, Beebe DJ, Alexander CM. Both LRP5 and LRP6 receptors are required to respond to physiological Wnt ligands in mammary epithelial cells and fibroblasts. The Journal of Biological Chemistry. 2012;287(20):16454-16466
  59. 59. Moradin N, Descoteaux A. Leishmania promastigotes: Building a safe niche within macrophages. Frontiers in Cellular and Infection Microbiology. 19 Sep 2012;2:121
  60. 60. Stutz MD, Pellegrini M. Mycobacterium tuberculosis: Preparing and maintaining the replicative niche. Trends in Microbiology. 2018;26(10):813-814
  61. 61. Ribet D, Cossart P. How bacterial pathogens colonize their hosts and invade deeper tissues. Microbes and Infection. 2015;17(3):173-183
  62. 62. Monastyrska I, Klionsky DJ. Autophagy in organelle homeostasis: Peroxisome turnover. Molecular Aspects of Medicine. 2006;27(5-6):483-494
  63. 63. Farré JC, Krick R, Subramani S, Thumm M. Turnover of organelles by autophagy in yeast. Current Opinion in Cell Biology. 2009;21(4):522-530
  64. 64. Chargui A, El May MV. Autophagy mediates neutrophil responses to bacterial infection. APMIS. 2014;122(11):1047-1058
  65. 65. Iliev AI, Djannatian JR, Nau R, Mitchell TJ, Wouters FS. Cholesterol-dependent actin remodeling via RhoA and Rac1 activation by the Streptococcus pneumoniae toxin pneumolysin. Proceedings of the National Academy of Sciences. 2007;104(8):2897-2902
  66. 66. Woods A, James CG, Wang G, Dupuis H, Beier F. Control of chondrocyte gene expression by actin dynamics: A novel role of cholesterol/Ror-α signalling in endochondral bone growth. Journal of Cellular and Molecular Medicine. 2009;13(9b):3497-3516
  67. 67. Crotzer VL, Blum JS. Autophagy and its role in MHC-mediated antigen presentation. The Journal of Immunology. 2009;182(6):3335-3341
  68. 68. English L, Chemali M, Duron J, Rondeau C, Laplante A, Gingras D, et al. Autophagy enhances the presentation of endogenous viral antigens on MHC class I molecules during HSV-1 infection. Nature Immunology. May 2009;10(5):480
  69. 69. Mostowy S, Shenoy AR. The cytoskeleton in cell-autonomous immunity: Structural determinants of host defence. Nature Reviews. Immunology. 2015;15(9):559-573
  70. 70. Sen M, Chamorro M, Reifert J, Corr M, Carson DA. Blockade of Wnt-5A/frizzled 5 signaling inhibits rheumatoid synoviocyte activation. Arthritis and Rheumatism. 2001;44(4):772-781
  71. 71. Sen M, Lauterbach K, El-Gabalawy H, Firestein GS, Corr M, Carson DA. Expression and function of wingless and frizzled homologs in rheumatoid arthritis. Proceedings of the National Academy of Sciences. 2000;97(6):2791-2796
  72. 72. Sen M. Wnt signalling in rheumatoid arthritis. Rheumatology (Oxford, England). 2005;44(6):708-713
  73. 73. Nguyen L, Pieters J. The Trojan horse: Survival tactics of pathogenic mycobacteria in macrophages. Trends in Cell Biology. 2005;15(5):269-276
  74. 74. Brandenburg J, Reiling N. The Wnt blows: On the functional role of Wnt signaling in Mycobacterium tuberculosis infection and beyond. Frontiers in Immunology. 26 Dec 2016;7:635
  75. 75. Villaseñor T, Madrid-Paulino E, Maldonado-Bravo R, Urbán-Aragón A, Pérez-Martínez L, Pedraza-Alva G. Activation of the Wnt pathway by Mycobacterium tuberculosis: a Wnt–Wnt situation. Frontiers in Immunology. 1 Feb 2017;8:50
  76. 76. Witze ES, Litman ES, Argast GM, Moon RT, Ahn NG. Wnt5a control of cell polarity and directional movement by polarized redistribution of adhesion receptors. Science. 2008;320(5874):365-369

Written By

Suborno Jati and Malini Sen

Submitted: 04 February 2019 Reviewed: 19 April 2019 Published: 04 July 2019