Next Article in Journal
SAG-DTA: Prediction of Drug–Target Affinity Using Self-Attention Graph Network
Next Article in Special Issue
Novel Insight into the Serum Sphingolipid Fingerprint Characterizing Longevity
Previous Article in Journal
Mitochondrial Dysfunction in Vascular Wall Cells and Its Role in Atherosclerosis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Macrophage Motility in Wound Healing Is Regulated by HIF-1α via S1P Signaling

1
Department of Orthodontics and Dentofacial Orthopedics, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima 770-8504, Japan
2
Department of Orthodontics, Faculty of Dentistry, Sultan Agung Islamic University, Semarang 50112, Indonesia
3
Department of Orthodontics, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8525, Japan
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2021, 22(16), 8992; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22168992
Submission received: 22 July 2021 / Revised: 14 August 2021 / Accepted: 18 August 2021 / Published: 20 August 2021
(This article belongs to the Special Issue The Role of Sphingolipids in Oxidative Stress)

Abstract

:
Accumulating evidence indicates that the molecular pathways mediating wound healing induce cell migration and localization of cytokines to sites of injury. Macrophages are immune cells that sense and actively respond to disturbances in tissue homeostasis by initiating, and subsequently resolving, inflammation. Hypoxic conditions generated at a wound site also strongly recruit macrophages and affect their function. Hypoxia inducible factor (HIF)-1α is a transcription factor that contributes to both glycolysis and the induction of inflammatory genes, while also being critical for macrophage activation. For the latter, HIF-1α regulates sphingosine 1-phosphate (S1P) to affect the migration, activation, differentiation, and polarization of macrophages. Recently, S1P and HIF-1α have received much attention, and various studies have been performed to investigate their roles in initiating and resolving inflammation via macrophages. It is hypothesized that the HIF-1α/S1P/S1P receptor axis is an important determinant of macrophage function under inflammatory conditions and during disease pathogenesis. Therefore, in this review, biological regulation of monocytes/macrophages in response to circulating HIF-1α is summarized, including signaling by S1P/S1P receptors, which have essential roles in wound healing.

1. Introduction

Hypoxia-inducible factor (HIF) is a transcription factor that is composed of two basic helix-loop-helix proteins, α and β, which both belong to the PER-ARNT-SIM (PAS) family [1]. Under hypoxic conditions, HIF binds to a pentanucleotide sequence (RCGTG) present in hypoxic response elements (HREs) that are carried by certain target genes. All three subunits of HIF-α—1α, 2α, and 3α—are highly responsive to hypoxic conditions. There are only two types of β subunits, aryl hydrocarbon receptor nuclear translocator (ARNT, also known as HIF-1β) and ARNT2. The latter is expressed in the nucleus, is not responsive to oxygen, and has other functions in gene transcription [2,3,4,5,6].
The roles and functions of HIF-1α and HIF-2α have been more extensively studied than those of HIF-3α. Five variants of HIF-1α have been reported to be associated with its exon structure or amino acid sequence: HIF-1α557, HIF-1α736, HIF-1αFL (or HIF-1αZ), HIF-1α516, and HIF-1α785 [7,8,9,10]. For several different functions, HIF-1α and HIF-2α have overlapping roles. However, HIF-2α is characterized by more restricted expression, is also known as endothelial PAS domain protein 1 (EPAS1), and has a smaller structure than HIF-1α [11,12]. HIF-3α has a smaller transactivation domain than both HIF-1α and HIF-2α. In humans, six HIF-3α variants have been reported, HIF-3α1–6 [12,13].
In macrophages, HIF-α isoforms can be differentially activated, and they contribute to regulation of macrophage polarization depending on their microenvironment. Macrophages often localize to hypoxic tissues, and this environment can strongly affect essential macrophage functions [14,15]. During M1 and M2 macrophage polarization that is induced by Th1 and Th2 cytokines, respectively, mRNA levels of HIF-1α and HIF-2α are differentially expressed. HIF-1α and HIF-2α further contribute to M1 and M2 macrophage functions by binding inducible nitric oxidase synthase (iNOS) or arginase-1, respectively, to maintain nitric oxide (NO) homeostasis during inflammation [15,16,17].
Sphingosine-1-phosphate (S1P) plays a role in the activation of macrophages and is subsequently produced in and affects macrophage functions. The role of S1P under hypoxic conditions has been extensively investigated [18,19,20]. S1P production, S1P receptor (S1PR) expression, and signaling mechanisms mediated by both have been shown to be dynamic. During inflammation, higher levels of S1P are present in affected tissues, thereby leading to greater signal input to macrophages from their environment [21]. S1P also acts to guide macrophages in their migration to a site of inflammation and injury. Moreover, S1P-dependent migration has been found to be strictly dependent on the S1PR profiles [22].
Recent evidence showed that targeting the S1P/S1PR signaling pathway is crucial for the treatment of immune-mediated diseases (multiple sclerosis and rheumatoid arthritis) [23,24,25], inflammatory bowel diseases [26], lung diseases [27,28], liver diseases [29,30], vascular diseases [31,32,33], brain diseases [34,35], renal diseases [36], and allergy [37]. In the latest clinical trial research, S1P/S1PR also act as a potential new adjuvant therapy to alleviate viral infection of COVID-19 symptoms [38]. In addition, the long duration treatment has been observed to be effective by targeting S1P/S1PR signals in fingolimod (FTY720) [34], KRP-203 [39], and ozanimod (RPC1063) [26]. However, to establish the long-term effect by targeting S1P/S1PR in chronic diseases, larger samples and longer treatment duration for assessing clinical efficacy and safety remain to be studied.
The importance of the S1P/S1PR signaling axis is demonstrated in knockout (KO) mouse models. Amongst all S1PRs, S1P1 is the most critical S1P receptor for angiogenesis, while S1P2,3 is redundant or enhancing S1PR signaling function [40]. The defect also occurs in the nervous system development or hinders lymphocytes migration [40,41]. Identification of S1P4,5 in the KO mouse model suggested that S1P4 receptors play a role in the essential regulation of megakaryocytes morphology and platelet repopulation, while S1P5 acts as an oligodendroglial receptor that regulates oligodendrocytes development [42,43]. Disturbance during vascular development caused by a defect in the S1P/S1PR signal affects vascular integrity and coagulation processes [44]. An imbalance in the S1P gradient causes S1P to play the role of both a pro- and an anti-inflammatory agent in various immune cells, such as monocytes/macrophages, platelets, mast cells, lymphocytes, endothelial cells, and fibroblasts [45].
Macrophages all express S1PRs (S1P1–5), although their expression profile varies according to subtype and their distinct functional properties (Figure 1) [46]. For example, in peritoneal macrophages, S1P1 acts as a pro-migratory signal involving Rho kinase and PI3K-Akt1 [21,47]. Conversely, S1P2 signaling stimulates cAMP production to inhibit macrophage migration via NLRP3 inflammasome activation, thereby attenuating phosphorylation of Akt in peritoneal inflammation [30,48]. Meanwhile, S1P3 has been shown to promote activation of inflammatory macrophages in microglia, while a model of brain ischemia has demonstrated that S1P3 regulates expression levels of inflammatory genes (e.g., IL-6, IL-1β, TNF-α, iNOS, and COX-2) via induction of lipopolysaccharide (LPS) [49,50]. To date, S1P4 and S1P5 remain less studied compared with S1P1–3. S1P4 is expressed by leukocytes, T cells, monocytes, and macrophages [51,52]. Under pro-inflammatory conditions, macrophages downregulate S1P4, while S1P4 levels are higher on M2-polarized macrophages [53]. Furthermore, on T cells, S1P4 contributes to suppression of cellular proliferation by reducing expression of interleukin (IL)-2 and interferon (IFN)-γ and increasing levels of IL-10, while not affecting T cell migration [51,54]. Regarding S1P5, it contributes to the exit of monocytes from bone marrow and tissue macrophages during inflammation [55].
In order to survive hypoxic conditions during inflammation, macrophages need to migrate to specific tissue sites. Recent studies of HIF-1α have highlighted the important role of S1P/S1PR signaling for this migration phenotype [52,53,56,57,58]. However, the mechanistic details regarding the communication that takes place between these signaling pathways in macrophages remain to be elucidated. Therefore, in this review, HIF-1α and S1P/S1PR signaling in macrophages in response to inflammation in wound healing is presented, and insights into possible mechanisms are discussed.

2. Biological Regulation of HIF-1α in Macrophage

2.1. The Cellular Regulation of HIF-1α

Under normal oxygen conditions, HIF-1α proteins are continuously synthesized and degraded via the proteasome pathway [59,60,61]. As a result, HIF-1α has a very short half-life. However, in the presence of decreasing concentrations of oxygen, degradation of HIF-1α is reduced. This oxygen-dependent regulation is achieved due to an oxygen-dependent degradation domain (ODDD) in HIF-1α that contains Fe2+ and two key prolyl residues (Pro402 and Pro564). When these prolyl residues undergo oxygen-dependent hydroxylation, HIF-1α is recognized by a component of an E3 ubiquitin ligase complex, von Hippel–Lindau tumor suppressor protein (pVHL), and targeted for degradation via the ubiquitin-proteosome pathway [62,63,64,65,66,67,68,69,70]. Therefore, when oxygen levels are low in a hypoxic environment, HIF-1α exhibits a longer half-life.
While pVHL has been shown to have a vital role in controlling HIF-1α stability via ubiquitination [71], a pVHL-independent pathway also exists. The latter is mediated by tumor suppression protein p53 and forkhead box O4 (FOXO4) and promotes HIF-1α proteosomal-mediated proteolysis [72,73]. Other components of this proteolysis complex include elongin-B, elongin-C, Cul2, and Rbx1, which are components of other E3 ubiquitin ligase complexes as well. Degradation of HIF-1α effectively blocks transcriptional activation of its downstream genes under normoxic conditions. In contrast, under hypoxic conditions, prolyl hydroxylation is suppressed and HIF-1α accumulates and translocates to the nucleus. After HIF-1α forms a dimer with HIF-1β, this trans-activating complex is able to bind HREs or enhancer sequences present in target genes. Notable target genes include vascular endothelial growth factor (VEGF), erythropoietin, iNOS, heme oxygenase-1, enolase-1, aldolase-A, lactate dehydrogenase-A, and phosphoglycerate kinase-1 [2,74,75]. HIF-1α signaling pathways have also been shown to influence collagen-prolyl-4-hydroxylase, urokinase-type plasminogen activator receptor, matrix metalloproteinase-2 (MMP2), and tissue inhibitor of MMP (TIMP)-1 [76].
VEGF is directly regulated by HIF-1α [77], and the biological activity of VEGF is further enhanced when VEGF receptor-1 (VEGFR-1/Flt-1) is upregulated in response to hypoxia. VEGF mRNA also exhibits greater stability under hypoxic conditions [78]. In endothelial cells, there is an autocrine signaling loop that involves signaling through VEGF and VEGFR-1 to provide hypoxic induction of VEGFR-2. When HIF-1α is deleted, impaired vascularization of xenografts has been observed [79]. The phosphatidylinositol 3-kinase (PI3K)/Akt pathway has also been shown to affect HIF-1α/VEGF expression in the presence of vanadium, a carcinogenic agent, via reactive oxygen species (ROS) [80].
It is known that hypoxic conditions affect oxidative stress. There are several sources of ROS, including the respiratory electron transport chain in mitochondria, the family of NADPH-oxidase enzymes, xanthine oxidase, peroxisomes, cytochrome p450 enzymes, cyclooxygenase, and lipoxygenase [81]. Mitochondria are influenced by HIF-1α during the cellular respiration process. Under hypoxic conditions and in response to HIF-1α, the composition of the cytochrome oxidase complex is changed [76,82]. Moreover, mitochondria-derived ROS that are produced by electron transport chain complex III stabilize HIF-1α [82]. ROS also increase the transcriptional activity of HIF-1α [83]. Experimentally, this was demonstrated when cells incubated with H2O2 exhibited a longer half-life for HIF-1α and genes targeted under normoxic conditions were activated [84]. With greater stability, HIF-1 induces the mitochondrial protease LON, which is required for degradation of cytochrome c oxidase (COX)4-1 [76] and expression of pyruvate dehydrogenase kinase (PDK). The latter inhibits pyruvate dehydrogenase via phosphorylation. If pyruvate is not converted into acetyl CoA, pyruvate cannot enter the TCA cycle, and consumption of mitochondrial oxygen is reduced. ROS generation in response to hypoxia is also attenuated [76,85,86].
In keratinocytes, when SphK1 is deleted, ROS production is drastically reduced via NF-κB, as well as its anti-inflammatory effects via phorbol 12-myristate 13-acetate [87]. Deleted SphK1 also inhibits the accumulation of ceramides via S1P, which leads to ROS enhancement in non-alcohol liver injury [88]. N-acteylcysteine, a ROS scavenger, suppresses HIF-1α and VEGF via blockade of SphK1 activity under hypoxic conditions [89], while SphK1-dependent stabilization of HIF-1α is mediated via the Akt/GSK3β signaling pathway [90]. Cho et al. revealed a role for SphK1 in melantonin-induced inactivation of HIF-1α under hypoxic conditions [91]. Melantonin suppresses Akt/GSK3β and ROS-related signaling to decrease SphK1 activity and thereby decrease stabilization of HIF-1α expression [91]. Neutralization of ROS by antioxidants is known to abolish induction of HIF-1α during hypoxia [92,93]. HIF-α directly modulates ROS expression by inhibiting prolyl hydroxylases or their cofactors [94], and it indirectly modulates ROS expression by activating signaling upstream of HIF-α (e.g., mitogen-activated protein kinases (MAPK) [95,96], PI3K/Akt, and ERK-induced Rac1 [80,95,96,97,98]).
There are several molecules downstream of PI3K/Akt that regulate levels of HIF-1α protein. These include mammalian target of rapamycin (mTOR), glycogen synthase kinase-3-β (GSK3β), FOXO3, and Bcl-2 apoptosis-related family members [99,100,101,102]. GSK3β has been shown to contribute to the destabilization of HIF-1α in a VHL-independent manner [98,103,104]. Meanwhile, Akt inactivates phosphorylation of (p)-GSK3β in its ODDD and promotes HIF-1α accumulation [103]. Therefore, it is hypothesized that HIF-α and ROS activity are controlled via protein kinase phosphorylation, potentially through the universal phosphorylation signal transduction pathway of PI3K/Akt [105].
During cell cycle regulation, mTOR is a hypoxic sensor and a target of Akt. As an upstream mediator of HIF-1α activation, mTOR can also alter HIF-1α post-transcriptionally [106]. Previous studies have further revealed that signaling through the PI3K/Akt pathway via mTOR/FKBP-rapamycin-associated protein (FRAP) signaling pathways is regulated by HIF-1α [107]. Correspondingly, in the presence of individual inhibitors of PI3K and FRAP, or a dual inhibitor of PI3K/mTOR (LY294002, rapamycin, and NVP-BEZ235, respectively), activation of p-Akt is suppressed, and this is followed by reductions in expression levels of HIF-1α and VEGF under hypoxic conditions [106,107,108]. Wortmannin, a specific inhibitor of the PI3K/Akt pathway, has also been shown to inhibit expression of HIF-1α [109]. Thus, targeting of PI3K/Akt signaling pathways has led to repression and then sensitization to cellular death via HIF-1α gene expression [110].

2.2. Role of HIF-1α in Inflammation and Immune System Regulation Involving Macrophages

Macrophages represent one of the first barriers of the immune system that invading pathogens encounter [111,112,113]. Macrophages are able to engulf pathogens, including phagocytic dead cells and cellular debris, and perform degradation in phagolysosomes [114,115,116,117]. Macrophages also employ an array of direct antimicrobial mechanisms by generating ROS and reactive nitrogen species in phagosomes, delivering cathepsin and maturing phagosomes via hydrolase [118,119,120,121]. Therefore, the importance of ROS in macrophages acting as immune cells is well recognized.
Regarding the regulation of ROS in mitochondria, increased ROS expression catalyzes the PHD2 domain, thereby impairing its activity and inducing specific post-translational modifications. This inhibition of PHD2 subsequently promotes HIF activation to affect pro- and anti-inflammatory activities to regulate immune cells [16,122,123]. Macrophages, upon stimulation, can change their metabolic activity to acquire a phenotype that is usually associated with a pathological immunological niche. Consequently, decreases in local O2 can create a hypoxic microenvironment that leads to activation of HIF signaling and modulation of immune cell activity [124]. Thus, a correlation exists between hypoxic conditions, inflammation, and immune cell metabolism.
Hypoxic conditions stimulate the activity and survival of macrophages. Briefly, HIF activation induces ATP production, which increases cellular motility, invasiveness, and bactericidal activity [125,126]. Treatment of macrophage with LPS has been shown to upregulate pro-inflammatory genes, such as IL-10, IL-1β, HIF-1α, and NF-κB p65 [127]. Meanwhile, Aarup et al. have provided in vivo evidence that macrophages lacking HIF-1α in atherosclerosis mice exhibit decreased apoptosis, migration, and glucose uptake [128,129]. Conversely, overexpression of HIF-1α induces the polarization of pro-inflammatory macrophage (M1) through NF-κB to upregulate genes related to glycolysis metabolism (e.g., PDK1, PGK1, GLUT1, GCK, and PKM2) [130].
In vitro, M1 macrophage have been stimulated with intracellular proteins and nucleic acids from lysed cells, as well as with bacterial components (IFN-γ, LPS, and peptidoglycan, respectively) [131]. M1 macrophages are characterized by production of NO, iNOS, ROS, IFN-γ, IL-6, IL-1β, IL-12, CD86, CD80, MHC-II, and tumor necrosis factor (TNF)-α. They also secrete MMP-2 and MMP-9 for degradation of the extracellular matrix to facilitate cell migration and infiltration, and they produce growth factors such as platelet-derived growth factor, insulin-like growth factor-1, VEGF, and tumor growth factor (TGF)-β1 for cell proliferation, granulation, tissue formation, and angiogenesis [132,133,134,135,136].
HIF-1α expression is independent of NF-κB. On the other hand, the secondary target genes of NF-κB activation facilitate the chromatin remodeling of nucleosome of IkBζ (Nfkbiz) [137], and they may associate with the M1 phenotype. However, both the HIF-1α and HIF-2α subunits appear to be essential for maintaining levels of NF-κB p65 [100,138]. It appears that HIF-2α may promote anti-inflammatory and pro-resolving/regenerative M2 macrophages [17,139]. However, incongruent roles for HIF-2α have been reported. For example, HIF-2α promotes IL-1β expression [140] and competes with iNOS for arginine metabolism, thereby limiting synthesis of NO [17]. The latter is associated with an M1 phenotype rather than an M2 phenotype [140]. Thus, HIF-1α and HIF-2α appear to have overlapping functions, yet they also have distinct functions that cannot be compensated for by the other [139]. This spectrum of phenotype activation, which at times can be redundant, requires further study.
M2 macrophages reduce inflammation via upregulation of IL-10 expression, arginase-1, programmed death-ligand-2, resistin-like molecule-α, and TGF-β1. However, M2 macrophages also remodel and strengthen the extracellular matrix in the presence of MMP-12 and MMP-13 expression [134,141]. Additionally, M2 macrophages can be stimulated by IL-4 and IL-13, and they express mannose receptor CD206, dectin, IL-10, and TGF-β1 [131]. Upon IL-4 treatment, transcription factor STAT6 is activated, which then induces peroxisome proliferator-activated receptor (PPAR)γ-coactivator (PGC)-1β. PGC-1β subsequently induces biogenesis and mitochondrial respiration and further interacts with estrogen-related receptor-α and nuclear respiratory factor-1 to drive synthesis of cytochrome c and ATP [142,143]. When PGC-1β is knocked down, both the function and metabolic profile of M2 macrophages are impaired [144]. Jumonji domain-containing (Jmjd)-3 is a histone 3 Lys27 (H3K27) demethylase that has a role in macrophage activation that is NF-κB-dependent following stimulation of TLR [145]. The transcription factor IRF4 is crucial for the M2 macrophages response and is a direct target of Jmjd3-mediated demethylation [146]. Therefore, transcription factors may represent key metabolic switches in M2 macrophages (Figure 2).

3. The Role of S1P/S1PR in Macrophages during Inflammation

S1P and ceramide are potential bioactive lipid mediators that regulate cellular pleiotropic activities such as survival, proliferation, inflammation, and migration [147,148,149]. S1P is a pro-apoptotic backbone component of all sphingolipids [150] and is generated from ceramidase and sphingosine kinase (SphK). Correspondingly, SphK is a central component of sphingolipid metabolism, which both synthesizes and degrades sphingolipids [151]. Briefly, de novo ceramide synthesis initially combines palmitoyl-CoA and serine to form 3-keto-dihydrosphingosine, which is subsequently reduced to dihydroceramide [152]. A desaturate subsequently generates the corresponding ceramides that can undergo phosphorylation or glycosylation to form glucosylceramides. The latter are further processed to form glycosphingolipids that are presented at the plasma membrane. Alternatively, ceramides can be converted to sphingomyelin and incorporated into the outer cell membrane. If attacked by acidic or neutral sphingomyelinases, these sphingomyelins are converted back to ceramide. Following cleavage by ceramidases, the ceramides form sphingosine, which can be phosphorylated by two sphingosine kinase isoforms, SphK1 and SphK2, to generate S1P [45,153,154].
S1P is a ligand of five high-affinity (S1P1–5) G protein-coupled receptors that are linked to either Gi, Gq, and/or G12/13. These receptors exhibit distinct tissue expression profiles, and their specific effects are dependent on these profiles [155,156,157]. Myeloid cells express G12/13, Gq/13, and Gs [158]. G12/13 belongs to the α-subunit that shares 67% amino acid sequence identified, and it stimulates an effector pathway of GTPase RhoA [159,160]. S1P is produced intracellularly by Sphk1 that is activated in response to several stimuli, including pro-inflammatory cytokines [45]. It is hypothesized that S1P induces activation of inhibitor of κ kinase (IKK)-β and c-jun amino-terminal kinase (JNK) via upstream activation of TGF-β-activated kinase-1. SphK-1 can also block JNK activation and prevent inflammation, while inhibition of SphK1 leads to activation of JNK. Interestingly, S1P opposes the effects of ceramide by counteracting ceramide-induced activation of JNK [149,161]. Thus, the ratio of ceramide-to-S1P may function as an intracellular rheostat.
S1P can exert both paracrine and autocrine effects following secretion by the ATP-binding cassette transporter or the S1P transporter spinster homolog-2 [162,163,164]. In circulation, S1P is characterized by a high nanomolar concentration [165]. Recent studies have reported that S1P mediates intracellular functions, specifically histone acetylation in the nucleus [166]. If confirmed, a link between S1P and epigenetic regulation of gene expression would be established [166].
S1P has been shown to act as a cellular motility regulator in conjunction with TGF-β/Smad3 signaling to maintain cartilage homeostasis in osteoarthritis [167]; S1P also acts as a cofactor for TNF receptor-associated factor-2 to produce ubiquitin ligase activity for activation of NF-κB [168]. Activated NF-κB is then able to interact with prohibitin-2 and mediate mitochondrial respiration [169]. It may also modulate p21-activated kinase-1 activity [170]. Ishii et al. revealed that S1P/S1P1 interactions regulate the egress of osteoclast precursor cells from circulation to bone tissues [171]. Meanwhile, S1P2 modulates NFATc1 to affect osteoclastogenesis and pro-inflammatory cytokines [172]. The S1P2-mediated caspase-11 p26 subunit is also known to induce macrophage death under sepsis conditions [173]. Dying cells release S1P, which activates S1P1/3 in macrophages to upregulate COX-2 triggering of VEGF [174]. Moreover, Fas apoptotic signaling promotes osteoclast precursor cells or bone marrow macrophages (BMMs)-induced osteoclasts, in conjunction with S1P1 signaling through NF-κB p50 subunit activation in rheumatoid arthritis [23,175]. Thus, targeting components downstream of S1PR signaling may represent a promising therapeutic approach.
Signaling through S1P/S1PRs promotes M1/M2 polarization of macrophages by affecting cytokine production and migration phenotype [21,53]. Different subtypes of macrophages exhibit distinct SIPR profiles, yet all macrophages express all five S1PRs to some extent [53]. Moreover, receptor expression profiles appear to correspond to distinct functional properties (Figure 1) [46]. Under inflammatory conditions, levels of S1P increase, and this increase is sensed by various types of cells, including macrophages. Macrophages are exposed to multiple signals from their environment, and they adjust their response accordingly. It is hypothesized that inflammatory, homeostatic, or regenerative conditions, as well as S1P production and S1PR expression and/or signaling, further add to the complexity of the functional properties that characterize macrophage populations [21].
Inflammation is triggered by endogenous signals and microbial components such as LPS at sites of injury. Once established, sites of injury are characterized by paracrine recruitment, proliferation, and differentiation of circulating progenitor cells and diverse types of inflammatory cells [21]. Das et al. have demonstrated that activation of S1P/S1PRs during bone healing activates anti-inflammatory (M2) macrophages, promotes vascularization, and recruits bone marrow-derived mononuclear cells at the site of injury [176]. However, the specific types of S1PRs that regulate M1/M2 macrophages for various inflammatory or disease conditions remain unclear. It has been demonstrated that bacterial stimulation of macrophages leads to an increase in protein levels of S1P3 [177]. In the absence of S1P3, phagosome maturation of macrophages is affected via ROS activation [177]. When bone fractures have been treated with FTY720, a non-selective S1PR agonist, no improvement in healing was observed, and there was no difference in osteoclast numbers at the wound sites [178]. Yang et al. demonstrated that S1P2 and S1P3 have important roles in the polarization of M1 macrophages [179]. In both in vivo and in vitro studies, S1P-induced BMMs were observed to promote M1 macrophages, and this tightly correlated with expression of TNF-α and MCP-1 and the signaling pathway involving Gαi/o, PI3K, and JNK [179]. Various roles and actions of S1P/S1PRs signaling pathways in monocytes/macrophages are summarized in Table 1. Some of these studies explain the infiltration of M1/M2 macrophages under inflammatory conditions. For example, S1P3 knockout (KO) studies have shown that the number of M2 macrophages increases and fewer T cells infiltrate muscle wound sites [180]. In addition, it was observed that treatment with a S1P3 antagonist, VPC01091, improved tissue regeneration [180]. Decreased monocyte/macrophage motility detected in S1P4 [54] and SphK1 KO mice has also been associated with reduced S1P generation [181]. Under hypoxic conditions, SphK1-mediated accumulation of HIF-1α levels occurs and is dependent on Akt/GSK3β signaling [90]. In contrast, partial HIF-1α KO mice have been reported to aggravate the infiltration of M1/M2 macrophages via the S1P/S1P1 signaling axis [57]. Thus, overlap between the roles of S1P/S1PRs and HIF-1α signaling pathways may exist under inflammatory conditions and may also affect the function of M1/M2 macrophages.

4. Cellular Regulation of HIF-1α through S1P/S1P1 Signaling in Macrophages during Wound Healing

4.1. The Roles of Macrophage Profiles and Hypoxia on Wound Healing

In this section, the roles of HIF-1α and S1P in macrophages recruited to inflamed regions of wound healing are discussed. Specifically, activation, polarization, migration, and phagocytosis ability of these macrophages are discussed, as well as the pathways that potentially mediate these functions. Macrophages are present in all tissues. They are present as resident cells (Langerhans cells) or they are introduced as infiltrating monocyte-derived cells [182]. The tissue site is a predominant determinant of the phenotype of tissue-resident cells. The latter help to both maintain tissue homeostasis and act as sentinels of injury. Therefore, both recruited macrophages and tissue-resident macrophages substantially contribute to wound healing at a site of injury [183].
Wound healing is a tightly coordinated and highly dynamic process that is able to restore tissue integrity after hypoxia is induced with infection or physical trauma. As healing progresses, a decline in tissue hypoxia occurs. It has been demonstrated that hypoxia induces essential factors that stimulate the proliferation and migration of endothelial cells, macrophages, keratinocytes, and fibroblasts in wound areas [184,185,186]. There are three distinctive phases to the healing process: (1) coagulation and inflammation, (2) tissue formation, and (3) tissue remodeling [187]. In the first phase, a blood clot is established to provisionally close the wound. Concomitantly, recruitment of inflammatory cells is initiated [188]. In the subsequent tissue formation phase, cell proliferation is initiated by local growth factors, and pro-inflammatory signaling declines. Finally, the wound site is organized to restructure the tissue and complete the tissue remodeling phase [189]. In all three phases, macrophages are critical components [190].
It is possible that an imbalance in phenotype switching of M1/M2 macrophages induces tissue breakdown [191]. Furthermore, any cell depletion from monocyte or macrophage lineages will impair wound closure and the granulation of formation tissue [192]. Thus, macrophages fulfill distinct functional roles, and this highlights their diversity and plasticity in achieving these functions.

4.2. Differential Wound Healing Is Accelerated at Skin and Mucosal Sites of Injury

It is well-documented that oral mucosal wounds heal faster than skin wounds, despite having the same stages of wound healing at each site (Figure 3) [186,193]. Multiple pro-inflammatory cytokines, chemokines, growth factors, and ROS production contribute to successful wound healing [194]. It has been observed in oral mucosal wounds that fewer inflammatory cells infiltrate the wounds (e.g., neutrophils, macrophages, mannose receptor-positive M2 macrophages, and T cells) [195], and cytokine production (IL-6 and TGF-β1) is reduced [193]. However, a robust increase in re-epithelialization is detected 24 h post-injury compared with skin wound closure [193]. It has also been observed that oral keratinocytes express lower levels of VEGF than skin keratinocytes under hypoxic conditions [196], and skin wounds exhibit higher levels of hypoxia and elevated levels of HIF-1α compared with mucosal wounds under stressed conditions [186]. Higher levels of HIF-1α expression in skin may be due to a greater abundance of gene expression related to the HIF-1α pathway in skin than in oral mucosa. Relevant genes include MMP1, MMP8, MMP9, MMP10, MMP13, MMP23B, NOS3, SLC2A1, SLC2A3, PIK3R1, PIK3R2, PGF, RRAS2, and EGLN3. Meanwhile, in the tongue, fewer genes are expressed that relate to the HIF-1α pathway (e.g., MMP10, SLC2A1, PIK3R1, and PIK3R5) [186]. Fewer keratinocytes lead to less scar tissue formation in oral wounds than in skin wounds [197]. This is due to increased levels of SOX2 and PITX1 expression by primary human oral cells, which are unique to the oral cavity, and they accelerate wound closure [198]. In contrast, expression of α-SMA (myofibroblasts) is higher in oral sites of injury than in skin sites of injury during the late stages of wound healing. Expression of α-SMA is followed by elevated levels of TGF-β and pSmad3 [199]. The essential role of TGF-β/Smad3 signaling in wound healing [200] is consistent with the motility and infiltration of monocyte macrophages to wound areas. The former has been demonstrated in both Smad3-deficient mice and with use of Smad3-targeted siRNA, where acceleration in palatal wound repair was achieved [201,202]. Furthermore, the observed acceleration was accompanied by decreased expression of TGF-β, α-SMA, MCP-1, and MIP-1 α [201,202]. In a study of macrophage recruitment to oral wound sites vs. skin wound sites, it was confirmed that macrophages arrive earlier to oral wounds than to skin wounds, then were reduced in number to the level of unwounded tissue after 60 days [199]. Meanwhile, the number of macrophages at skin wounds remained high after 60 days [199]. Based on these results, it appears that a low number of macrophages (CD68, CD40, CD206, CD163) in oral wounds [195] may be beneficial for achieving scarless formation since they are known to abundantly produce more pro-fibrotic TGF-β1 in oral wounds than in skin wounds [199,203].
It is also possible that rapid oral wound healing is facilitated by the presence of saliva/mucous in the oral cavity, which can provide many necessary cytokines, growth factors, and protease inhibitors [193,204,205]. However, it is recognized that saliva alone is not responsible for the rapid healing of oral mucosal wounds. Rather, tissue characteristics, location, size, type of injury, salivary flow, microflora, and temperature are factors that also need to be considered [193]. Thus, macrophages provide critical, multi-faceted functions in wound repair by acting as pro- or anti-inflammatory agents. Given the importance of their role, the functions of macrophages in the oral mucosa remain to be further explored.
Several wound healing treatments related to oxygen therapy have been developed, which mediate anti-inflammatory effects. These include supplemental oxygen therapy and molecular hydrogen as an antioxidant, which are applied in preventive and therapeutic applications [186,206]. In both skin and mucosal wounds, it has been observed that hyperbaric oxygen therapy does not preserve HIF-1α and VEGF expression [186]. Consequently, no acceleration in wound closure is predicted. In palatal wound closures, hydrogen-rich water has been effective in accelerating healing via upregulation of the nuclear factor E2-related factor (Nrf)-2/antioxidant defense pathway. The latter is characterized by increases in heme oxygenase-1 and NAD(P)H quinone dehydrogenase-1 and reduced levels of iNOS [206]. Increased levels of healing-associated genes (e.g., FGF7, VEGF, TGF-1β, α-SMA) have also been observed [206].
An important mediator for skin and mucosal wound healing is IL-33, a potent Th2-type immune response [207,208,209]. Administration of IL-33 effectively accelerates wound healing by shrinking the wound area and increasing re-epithelialization and accumulation of collagen and fibronectin [207]. With an abundance of collagen and fibronectin, greater scar thickness is possible. IL-33 is also associated with an increased number of M2 macrophages (CD206), arginase-1 expression, and alternatively activated macrophages [207].

4.3. Cross Signaling among HIF-1α/S1P/S1PRs in Macrophages

Monocytes/macrophages are tightly dependent on HIF-1α expression in wound healing [210]. HIF-1α recruits monocytes/macrophages by promoting CCL2/MCP-1 secretion and also has a strong correlation with CD68 expression [211]. A hypoxic environment modulates macrophage functions both directly (oxidase, oxygenase, and hydroxylase) and indirectly (ATP/AMP, ROS, Krebs cycle, and Fe2+ levels) [212,213,214]. Meanwhile, S1P dependently affects chemotaxis of M1/M2 polarization without influencing phagocytic activity [53]. A hypoxic wound environment causes SphK1 to be rapidly activated via ROS, and this precedes accumulation of HIF-1α [90]. On the other hand, an increase in intracellular S1P occurs due to the conversion of ceramide to sphingosine, which is activated by HIF-1α via ERK, Akt, and the GSK3β pathway [215]. Thus, SIP activates and enhances the lifespan of macrophages [90,98]. In addition, S1P and adenosine [216] act to regulate HIF-1α production by controlling gene expression of VEGF and TGF-α through S1P3, Gi, and their downstream effectors (PKC-β1, MEK, PI3K, and mTOR) under normoxic conditions [56].
In macrophages, long-term hypoxic stress (chronic inflammation) upregulates Toll-like receptor (TLR)-4 mediated HIF-1α through the PI3K/Akt pathway, yet not via p38 [217,218]. Moreover, the PI3K/Akt pathway stabilizes HIF-1α via inhibition of GSK3β in the early stages of hypoxia [104]. When Semba et al. characterized the migration and activation of macrophage during the early stages of systemic inflammation, they found that HIF-1α-induced pyruvate dehydrogenase kinase (PDK)-1 promotes glycolysis, while cytochrome c oxidase remains unaffected [219]. In addition, it was observed that cross talk between HIF-1α and S1P/S1P1 signaling controls the migration of M1/M2 macrophages to a wound site via the downstream effectors ERK, Akt, and p38 [57]. Correspondingly, reduced macrophage migratory activity is associated with HIF-1α deficiency [57,219]. ATP is rapidly consumed in the cytosol due to the remodeling of cytoskeletal actin filaments that occurs with migration [220,221]. Thus, it is possible that cross talk exists between glycolytic reprogramming and actin filament remodeling during activation of macrophage migration under hypoxic conditions.
Interestingly, administration of dimethyloxalylglycine (DMOG), an inhibitor of PHD and α-ketoglutarate, helps accelerate wound closure and upregulates macrophage migration in a partial HIF-1α KO mouse model [57]. DMOG activates AMPK [222], HIF-1α, VEGF [223], and NF-κB [138,224], thereby abrogating pro-inflammatory cytokine expression. DMOG also activates PGC-1α to increase mitochondrial activity and glycolytic flux [225,226]. Endothelial-specific S1P1 KO mice exhibit a defect in vascular stabilization and systemic failure due to upregulation of VEGF by HIF-1α [227]. Lim et al. investigated a wound healing treatment involving the PHD inhibitor ciclopiroxolamine (CPX) and S1P [228]. In vitro, CPX and S1P extensively affected endothelia and the fibroblast formatting vascular network by expressing MCP-1 and VEGF [228]. Thus, stabilizing and inducing HIF-1α gene expression by inhibiting mitochondrial activity [224] can potentially enhance cellular sensitivity to S1P via Rho family GTPase Rac activation [228]. Thus, the HIF-1α/S1P1 signaling axis [57] [227] may represent a key regulator of inflammation not only in myeloid cells [185] but also in fibroblasts and endothelial cells [228].

5. Conclusions

In summary, the HIF-1α/S1P/S1PR signaling axis appears to play a critical role in the activation and polarization of macrophages, and this contributes to the inflammatory conditions at a wound site. However, additional studies are needed to better understand the exact role of S1P-induced functional consequences for macrophage biology in different wound healing entities, especially with respect to targeting the HIF-1α/S1P/S1PR signaling axis as a strategy for wound healing therapy.

Author Contributions

Conceptualization, T.I.; writing—original draft preparation, I.R.H. and T.I.; writing—review and editing, I.R.H., T.I., T.K.-O., T.S., A.I., and E.T.; funding acquisition, T.I. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported in part by JSPS KAKENHI grants from the Japan Society for the Promotion of Science (No. 18H03011 to T.I.).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Wang, G.L.; Jiang, B.-H.; Rue, E.A.; Semenza, G.L. Hypoxia-inducible factor 1 is a basic-helix-loop-helix-PAS heterodimer regulated by cellular O2 tension. Proc. Natl. Acad. Sci. USA 1995, 92, 5510–5514. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Weidemann, A.; Johnson, R.S. Biology of HIF-1α. Cell Death Differ. 2008, 15, 621–627. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Ema, M.; Hirota, K.; Mimura, J.; Abe, H.; Yodoi, J.; Sogawa, K.; Poellinger, L.; Fujii-Kuriyama, Y. Molecular mechanisms of transcription activation by HLF and HIF1α in response to hypoxia: Their stabilization and redox signal-induced interaction with CBP/p300. EMBO J. 1999, 18, 1905–1914. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Wang, G.L.; Jiang, B.H.; Semenza, G.L. Effect of protein kinase and phosphatase inhibitors on expression of hypoxia-inducible factor 1. Biochem. Biophys. Res. Commun. 1995, 216, 669–675. [Google Scholar] [CrossRef]
  5. Li, H.; Ko, H.P.; Whitlock, J.P. Induction of phosphoglycerate kinase 1 gene expression by hypoxia. Roles of Arnt and HIF1α. J. Biol. Chem. 1996, 271, 21262–21267. [Google Scholar] [CrossRef] [Green Version]
  6. Wiesener, M.S.; Jürgensen, J.S.; Rosenberger, C.; Scholze, C.K.; Hörstrup, J.H.; Warnecke, C.; Mandriota, S.J.; Bechmann, I.; Frei, U.A.; Pugh, C.W.; et al. Widespread, hypoxia-inducible expression of HIF-2α in distinct cell populations of different organs. FASEB J. 2002, 17, 271–273. [Google Scholar] [CrossRef] [Green Version]
  7. Gothie, E.; Richard, D.E.; Berra, E.; Pages, G.; Pouyssegur, J. Identification of alternative spliced variants of human hypoxia-inducible factor-1α. J. Biol. Chem. 2000, 275, 6922–6927. [Google Scholar] [CrossRef] [Green Version]
  8. Chun, Y.S.; Choi, E.; Yeo, E.J.; Lee, J.H.; Kim, M.S.; Park, J.W. A new HIF-1α variant induced by zinc ion suppresses HIF-1-mediated hypoxic responses. J. Cell. Sci. 2001, 114, 4051–4061. [Google Scholar] [CrossRef]
  9. Chun, Y.S.; Choi, E.; Kim, T.Y.; Kim, M.S.; Park, J.W. A dominant-negative isoform lacking exons 11 and 12 of the human hypoxia-inducible factor-1α gene. Biochem. J. 2002, 362, 71–79. [Google Scholar] [CrossRef]
  10. Chun, Y.S.; Lee, K.H.; Choi, E.; Bae, S.Y.; Yeo, E.J.; Huang, L.E.; Kim, M.S.; Park, J.W. Phorbol ester stimulates the nonhypoxic induction of a novel hypoxia-inducible factor 1α isoform: Implications for tumor promotion. Cancer Res. 2003, 63, 8700–8707. [Google Scholar]
  11. Tian, H.; McKnight, S.L.; Russell, D.W. Endothelial PAS domain protein 1 (EPAS1), a transcription factor selectively expressed in endothelial cells. Genes Dev. 1997, 11, 72–82. [Google Scholar] [CrossRef] [Green Version]
  12. Lee, J.W.; Bae, S.H.; Jeong, J.W.; Kim, S.H.; Kim, K.W. Hypoxia-inducible factor (HIF-1)α: Its protein stability and biological functions. Exp. Mol. Med. 2004, 36, 1–12. [Google Scholar] [CrossRef]
  13. Makino, Y.; Cao, R.; Svensson, K.; Bertilsson, G.; Asman, M.; Tanaka, H.; Cao, Y.; Berkenstam, A.; Poellinger, L. Inhibitory PAS domain protein is a negative regulator of hypoxia-inducible gene expression. Nature 2001, 414, 550–554. [Google Scholar] [CrossRef]
  14. Murdoch, C.; Muthana, M.; Lewis, C.E. Hypoxia regulates macrophage functions in inflammation. J. Immunol. 2005, 175, 6257–6263. [Google Scholar] [CrossRef] [Green Version]
  15. Nizet, V.; Johnson, R.S. Interdependence of hypoxic and innate immune responses. Nat. Rev. Immunol. 2009, 9, 609–617. [Google Scholar] [CrossRef] [Green Version]
  16. Corrado, C.; Fontana, S. Hypoxia and HIF signaling: One axis with divergent effects. Int. J. Mol. Sci. 2020, 21, 5611. [Google Scholar] [CrossRef] [PubMed]
  17. Takeda, N.; O’Dea, E.L.; Doedens, A.; Kim, J.W.; Weidemann, A.; Stockmann, C.; Asagiri, M.; Simon, M.C.; Hoffmann, A.; Johnson, R.S. Differential activation and antagonistic function of HIF-{α} isoforms in macrophages are essential for NO homeostasis. Genes Dev. 2010, 24, 491–501. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Wacker, B.K.; Park, T.S.; Gidday, J.M. Hypoxic preconditioning-induced cerebral ischemic tolerance: Role of microvascular sphingosine kinase 2. Stroke 2009, 40, 3342–3348. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Wacker, B.K.; Perfater, J.L.; Gidday, J.M. Hypoxic preconditioning induces stroke tolerance in mice via a cascading HIF, sphingosine kinase, and CCL2 signaling pathway. J. Neurochem. 2012, 123, 954–962. [Google Scholar] [CrossRef] [Green Version]
  20. Yung, L.M.; Wei, Y.; Qin, T.; Wang, Y.; Smith, C.D.; Waeber, C. Sphingosine kinase 2 mediates cerebral preconditioning and protects the mouse brain against ischemic injury. Stroke 2012, 43, 199–204. [Google Scholar] [CrossRef] [PubMed]
  21. Weigert, A.; Olesch, C.; Brüne, B. Sphingosine-1-phosphate and macrophage biology—How the sphinx tames the big eater. Front. Immunol. 2019, 10, 1706. [Google Scholar] [CrossRef]
  22. Gude, D.R.; Alvarez, S.E.; Paugh, S.W.; Mitra, P.; Yu, J.; Griffiths, R.; Barbour, S.E.; Milstien, S.; Spiegel, S. Apoptosis induces expression of sphingosine kinase 1 to release sphingosine-1-phosphate as a "come-and-get-me" signal. FASEB J. 2008, 22, 2629–2638. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Hutami, I.R.; Izawa, T.; Mino-Oka, A.; Shinohara, T.; Mori, H.; Iwasa, A.; Tanaka, E. Fas/S1P1 crosstalk via NF-κB activation in osteoclasts controls subchondral bone remodeling in murine TMJ arthritis. Biochem. Biophys. Res. Commun. 2017, 490, 1274–1281. [Google Scholar] [CrossRef] [PubMed]
  24. Roggeri, A.; Schepers, M.; Tiane, A.; Rombaut, B.; Van Veggel, L.; Hellings, N.; Prickaerts, J.; Pittaluga, A.; Vanmierlo, T. Sphingosine-1-phosphate receptor modulators and oligodendroglial cells: Beyond immunomodulation. Int. J. Mol. Sci. 2020, 21, 7537. [Google Scholar] [CrossRef] [PubMed]
  25. Baeyens, A.; Bracero, S.; Chaluvadi, V.S.; Khodadadi-Jamayran, A.; Cammer, M.; Schwab, S.R. Monocyte-derived S1P in the lymph node regulates immune responses. Nature 2021, 592, 290–295. [Google Scholar] [CrossRef]
  26. Sandborn, W.J.; Feagan, B.G.; Wolf, D.C.; D’Haens, G.; Vermeire, S.; Hanauer, S.B.; Ghosh, S.; Smith, H.; Cravets, M.; Frohna, P.A.; et al. Ozanimod induction and maintenance treatment for ulcerative colitis. N. Engl. J. Med. 2016, 374, 1754–1762. [Google Scholar] [CrossRef]
  27. Ebenezer, D.L.; Fu, P.; Natarajan, V. Targeting sphingosine-1-phosphate signaling in lung diseases. Pharmacol. Ther. 2016, 168, 143–157. [Google Scholar] [CrossRef] [Green Version]
  28. Barnawi, J.; Tran, H.; Jersmann, H.; Pitson, S.; Roscioli, E.; Hodge, G.; Meech, R.; Haberberger, R.V.; Hodge, S. Potential link between the sphingosine-1-phosphate (S1P) system and defective alveolar macrophage phagocytic function in chronic obstructive pulmonary disease (COPD). PLoS ONE 2015, 10, e0122771. [Google Scholar] [CrossRef]
  29. Hou, L.; Yang, L.; Chang, N.; Zhao, X.; Zhou, X.; Dong, C.; Liu, F.; Yang, L.; Li, L. Macrophage sphingosine 1-phosphate receptor 2 blockade attenuates liver inflammation and fibrogenesis triggered by NLRP3 inflammasome. Front. Immunol. 2020, 11, 1149. [Google Scholar] [CrossRef] [PubMed]
  30. Hou, L.; Zhang, Z.; Yang, L.; Chang, N.; Zhao, X.; Zhou, X.; Yang, L.; Li, L. NLRP3 inflammasome priming and activation in cholestatic liver injury via the sphingosine 1-phosphate/S1P receptor 2/Gα(12/13)/MAPK signaling pathway. J. Mol. Med. (Berl.) 2021, 99, 273–288. [Google Scholar] [CrossRef]
  31. Grimm, M.; Tischner, D.; Troidl, K.; Albarran Juarez, J.; Sivaraj, K.K.; Ferreiros Bouzas, N.; Geisslinger, G.; Binder, C.J.; Wettschureck, N. S1P2/G12/13 signaling negatively regulates macrophage activation and indirectly shapes the atheroprotective B1-cell population. Arterioscler. Thromb. Vasc. Biol. 2016, 36, 37–48. [Google Scholar] [CrossRef] [Green Version]
  32. Keul, P.; Lucke, S.; Lipinski, K.V.W.; Bode, C.; Gräler, M.; Heusch, G.; Levkau, B. Sphingosine-1-phosphate receptor 3 promotes recruitment of monocyte/macrophages in inflammation and atherosclerosis. Circ. Res. 2011, 108, 314–323. [Google Scholar] [CrossRef] [Green Version]
  33. Potì, F.; Gualtieri, F.; Sacchi, S.; Weißen-Plenz, G.; Varga, G.; Brodde, M.; Weber, C.; Simoni, M.; Nofer, J.-R. KRP-203, sphingosine 1-phosphate receptor type 1 agonist, ameliorates atherosclerosis in LDL-R−/−mice. Arter. Thromb. Vasc. Biol. 2013, 33, 1505–1512. [Google Scholar] [CrossRef] [Green Version]
  34. Wei, Y.; Yemisci, M.; Kim, H.-H.; Yung, L.M.; Shin, H.K.; Hwang, S.-K.; Guo, S.; Qin, T.; Alsharif, N.; Brinkmann, V.; et al. Fingolimod provides long-term protection in rodent models of cerebral ischemia. Ann. Neurol. 2010, 69, 119–129. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Grassi, S.; Mauri, L.; Prioni, S.; Cabitta, L.; Sonnino, S.; Prinetti, A.; Giussani, P. Sphingosine 1-phosphate receptors and metabolic enzymes as druggable targets for brain diseases. Front. Pharmacol. 2019, 10, 807. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Xu, M.; Liu, D.; Ding, L.-H.; Ma, K.-L.; Wu, M.; Lv, L.-L.; Wen, Y.; Liu, H.; Tang, R.-N.; Liu, B.-C. FTY720 inhibits tubulointerstitial inflammation in albumin overload-induced nephropathy of rats via the Sphk1 pathway. Acta Pharmacol. Sin. 2014, 35, 1537–1545. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Oskeritzian, C.A.; Hait, N.C.; Wedman, P.; Chumanevich, A.; Kolawole, E.M.; Price, M.M.; Falanga, Y.T.; Harikumar, K.B.; Ryan, J.J.; Milstien, S.; et al. The sphingosine-1-phosphate/sphingosine-1-phosphate receptor 2 axis regulates early airway T-cell infiltration in murine mast cell-dependent acute allergic responses. J. Allergy Clin. Immunol. 2015, 135, 1008–1018e1. [Google Scholar] [CrossRef] [Green Version]
  38. McGowan, E.M.; Haddadi, N.; Nassif, N.T.; Lin, Y. Targeting the SphK-S1P-SIPR pathway as a potential therapeutic approach for COVID-19. Int. J. Mol. Sci. 2020, 21, 7189. [Google Scholar] [CrossRef]
  39. Wenderfer, S.E.; Stepkowski, S.M.; Braun, M.C. Increased survival and reduced renal injury in MRL/lpr mice treated with a novel sphingosine-1-phosphate receptor agonist. Kidney Int. 2008, 74, 1319–1326. [Google Scholar] [CrossRef] [Green Version]
  40. Kono, M.; Allende, M.L.; Proia, R.L. Sphingosine-1-phosphate regulation of mammalian development. Biochim. Biophys. Acta 2008, 1781, 435–441. [Google Scholar] [CrossRef] [Green Version]
  41. Tsai, H.C.; Nguyen, K.; Hashemi, E.; Engleman, E.; Hla, T.; Han, M.H. Myeloid sphingosine-1-phosphate receptor 1 is important for CNS autoimmunity and neuroinflammation. J. Autoimmun. 2019, 105, 102290. [Google Scholar] [CrossRef] [PubMed]
  42. Golfier, S.; Kondo, S.; Schulze, T.; Takeuchi, T.; Vassileva, G.; Achtman, A.H.; Graler, M.H.; Abbondanzo, S.J.; Wiekowski, M.; Kremmer, E.; et al. Shaping of terminal megakaryocyte differentiation and proplatelet development by sphingosine-1-phosphate receptor S1P4. FASEB J. 2010, 24, 4701–4710. [Google Scholar] [CrossRef]
  43. Jaillard, C.; Harrison, S.; Stankoff, B.; Aigrot, M.S.; Calver, A.R.; Duddy, G.; Walsh, F.S.; Pangalos, M.N.; Arimura, N.; Kaibuchi, K.; et al. Edg8/S1P5: An oligodendroglial receptor with dual function on process retraction and cell survival. J. Neurosci. 2005, 25, 1459–1469. [Google Scholar] [CrossRef]
  44. Ruf, W.; Furlan-Freguia, C.; Niessen, F. Vascular and dendritic cell coagulation signaling in sepsis progression. J. Thromb. Haemost. 2009, 7 (Suppl. 1), S118–S121. [Google Scholar] [CrossRef] [PubMed]
  45. Spiegel, S.; Milstien, S. The outs and the ins of sphingosine-1-phosphate in immunity. Nat. Rev. Immunol. 2011, 11, 403–415. [Google Scholar] [CrossRef] [PubMed]
  46. Bryan, A.M.; Del Poeta, M. Sphingosine-1-phosphate receptors and innate immunity. Cell. Microbiol. 2018, 20, e12836. [Google Scholar] [CrossRef] [Green Version]
  47. Al-Jarallah, A.; Chen, X.; Gonzalez, L.; Trigatti, B.L. High density lipoprotein stimulated migration of macrophages depends on the scavenger receptor class B, type I, PDZK1 and Akt1 and is blocked by sphingosine 1 phosphate receptor antagonists. PLoS ONE 2014, 9, e106487. [Google Scholar] [CrossRef] [Green Version]
  48. Michaud, J.; Im, D.S.; Hla, T. Inhibitory role of sphingosine 1-phosphate receptor 2 in macrophage recruitment during inflammation. J. Immunol. 2010, 184, 1475–1483. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Gaire, B.P.; Song, M.R.; Choi, J.W. Sphingosine 1-phosphate receptor subtype 3 (S1P3) contributes to brain injury after transient focal cerebral ischemia via modulating microglial activation and their M1 polarization. J. Neuroinflamm. 2018, 15, 284. [Google Scholar] [CrossRef] [PubMed]
  50. Heo, J.Y.; Im, D.S. Pro-inflammatory role of S1P3 in macrophages. Biomol Ther. (Seoul) 2019, 27, 373–380. [Google Scholar] [CrossRef]
  51. Wang, W.; Graeler, M.H.; Goetzl, E.J. Type 4 sphingosine 1-phosphate G protein-coupled receptor (S1P 4) transduces S1P effects on T cell proliferation and cytokine secretion without signaling migration. FASEB J. 2005, 19, 1731–1733. [Google Scholar] [CrossRef]
  52. Duong, C.Q.; Bared, S.M.; Abu-Khader, A.; Buechler, C.; Schmitz, A.; Schmitz, G. Expression of the lysophospholipid receptor family and investigation of lysophospholipid-mediated responses in human macrophages. Biochim. Biophys. Acta 2004, 1682, 112–119. [Google Scholar] [CrossRef] [PubMed]
  53. Muller, J.; von Bernstorff, W.; Heidecke, C.D.; Schulze, T. Differential S1P receptor profiles on M1- and M2-polarized macrophages affect macrophage cytokine production and migration. Biomed. Res. Int. 2017, 2017, 7584621. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Schuster, C.; Huard, A.; Sirait-Fischer, E.; Dillmann, C.; Brüne, B.; Weigert, A. S1PR4-dependent CCL2 production promotes macrophage recruitment in a murine psoriasis model. Eur. J. Immunol. 2020, 50, 839–845. [Google Scholar] [CrossRef] [Green Version]
  55. Debien, E.; Mayol, K.; Biajoux, V.; Daussy, C.; De Agüero, M.G.; Taillardet, M.; Dagany, N.; Brinza, L.; Henry, T.; Dubois, B.; et al. S1PR5 is pivotal for the homeostasis of patrolling monocytes. Eur. J. Immunol. 2013, 43, 1667–1675. [Google Scholar] [CrossRef] [Green Version]
  56. Kalhori, V.; Kemppainen, K.; Asghar, M.Y.; Bergelin, N.; Jaakkola, P.; Törnquist, K. Sphingosine-1-phosphate as a regulator of hypoxia-induced factor-1α in thyroid follicular carcinoma cells. PLoS ONE 2013, 8, e66189. [Google Scholar] [CrossRef] [Green Version]
  57. Hutami, I.R.; Izawa, T.; Khurel-Ochir, T.; Sakamaki, T.; Iwasa, A.; Tomita, S.; Tanaka, E. HIF-1α controls palatal wound healing by regulating macrophage motility via S1P/S1P1 signaling axis. Oral. Dis. 2021. [Google Scholar] [CrossRef]
  58. Sanagawa, A.; Iwaki, S.; Asai, M.; Sakakibara, D.; Norimoto, H.; Sobel, B.E.; Fujii, S. Sphingosine 1phosphate induced by hypoxia increases the expression of PAI1 in HepG2 cells via HIF1α. Mol. Med. Rep. 2016, 14, 1841–1848. [Google Scholar] [CrossRef]
  59. Jewell, U.R.; Kvietikova, I.; Scheid, A.; Bauer, C.; Wenger, R.H.; Gassmann, M. Induction of HIF-1α in response to hypoxia is instantaneous. FASEB J. 2001, 15, 1312–1314. [Google Scholar] [CrossRef] [PubMed]
  60. Jiang, B.H.; Semenza, G.L.; Bauer, C.; Marti, H.H. Hypoxia-inducible factor 1 levels vary exponentially over a physiologically relevant range of O2 tension. Am. J. Physiol. 1996, 271, C1172–C1180. [Google Scholar] [CrossRef] [Green Version]
  61. Cockman, M.E.; Masson, N.; Mole, D.R.; Jaakkola, P.; Chang, G.W.; Clifford, S.C.; Maher, E.R.; Pugh, C.W.; Ratcliffe, P.J.; Maxwell, P.H. Hypoxia inducible factor-α binding and ubiquitylation by the von Hippel-Lindau tumor suppressor protein. J. Biol. Chem. 2000, 275, 25733–25741. [Google Scholar] [CrossRef] [Green Version]
  62. Ivan, M.; Kondo, K.; Yang, H.; Kim, W.; Valiando, J.; Ohh, M.; Salic, A.; Asara, J.M.; Lane, W.S.; Kaelin, W.G., Jr. HIFα targeted for VHL-mediated destruction by proline hydroxylation: Implications for O2 sensing. Science 2001, 292, 464–468. [Google Scholar] [CrossRef] [PubMed]
  63. Jaakkola, P.; Mole, D.R.; Tian, Y.-M.; Wilson, M.I.; Gielbert, J.; Gaskell, S.J.; von Kriegsheim, A.; Hebestreit, H.F.; Mukherji, M.; Schofield, C.; et al. Targeting of HIF-α to the von Hippel-Lindau ubiquitylation complex by O2-regulated prolyl hydroxylation. Science 2001, 292, 468–472. [Google Scholar] [CrossRef]
  64. Semenza, G.L. Hypoxia-inducible factor 1 and cancer pathogenesis. IUBMB Life 2008, 60, 591–597. [Google Scholar] [CrossRef]
  65. Maxwell, P.; Wiesener, M.S.; Chang, G.-W.; Clifford, S.C.; Vaux, E.C.; Cockman, M.; Wykoff, C.C.; Pugh, C.; Maher, E.; Ratcliffe, P. The tumour suppressor protein VHL targets hypoxia-inducible factors for oxygen-dependent proteolysis. Nature 1999, 399, 271–275. [Google Scholar] [CrossRef] [PubMed]
  66. Ohh, M.; Park, C.W.; Ivan, M.; Hoffman, M.A.; Kim, T.Y.; Huang, L.E.; Pavletich, N.; Chau, V.; Kaelin, W.G. Ubiquitination of hypoxia-inducible factor requires direct binding to the beta-domain of the von Hippel-Lindau protein. Nat. Cell Biol 2000, 2, 423–427. [Google Scholar] [CrossRef]
  67. Bai, S.; Datta, J.; Jacob, S.T.; Ghoshal, K. Treatment of PC12 cells with nerve growth factor induces proteasomal degradation of T-cadherin that requires tyrosine phosphorylation of its cadherin domain. J. Biol. Chem. 2018, 282, 27171–27180. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Kamura, T.; Sato, S.; Iwai, K.; Czyzyk-Krzeska, M.; Conaway, R.C.; Conaway, J.W. Activation of HIF1α ubiquitination by a reconstituted von Hippel-Lindau (VHL) tumor suppressor complex. Proc. Natl. Acad. Sci. USA 2000, 97, 10430–10435. [Google Scholar] [CrossRef] [Green Version]
  69. Tanimoto, K.; Makino, Y.; Pereira, T.; Poellinger, L. Mechanism of regulation of the hypoxia-inducible factor-1 α by the von Hippel-Lindau tumor suppressor protein. EMBO J. 2000, 19, 4298–4309. [Google Scholar] [CrossRef] [Green Version]
  70. Semenza, G.L. Regulation of cancer cell metabolism by hypoxia-inducible factor 1. Semin. Cancer Biol. 2009, 19, 12–16. [Google Scholar] [CrossRef]
  71. Metzen, E.; Zhou, J.; Jelkmann, W.; Fandrey, J.; Brune, B. Nitric oxide impairs normoxic degradation of HIF-1α by inhibition of prolyl hydroxylases. Mol. Biol. Cell 2003, 14, 3470–3481. [Google Scholar] [CrossRef] [Green Version]
  72. Ravi, R.; Mookerjee, B.; Bhujwalla, Z.M.; Sutter, C.H.; Artemov, D.; Zeng, Q.; Dillehay, L.E.; Madan, A.; Semenza, G.L.; Bedi, A. Regulation of tumor angiogenesis by p53-induced degradation of hypoxia-inducible factor 1α. Genes Dev. 2000, 14, 34–44. [Google Scholar]
  73. Tang, T.T.-L.; Lasky, L.A. The forkhead transcription factor FOXO4 induces the down-regulation of hypoxia-inducible factor 1 α by a von Hippel-Lindau protein-independent mechanism. J. Biol. Chem. 2003, 278, 30125–30135. [Google Scholar] [CrossRef] [Green Version]
  74. Lee, P.J.; Jiang, B.H.; Chin, B.Y.; Iyer, N.V.; Alam, J.; Semenza, G.L.; Choi, A.M. Hypoxia-inducible factor-1 mediates transcriptional activation of the heme oxygenase-1 gene in response to hypoxia. J. Biol. Chem. 1997, 272, P5375–P5381. [Google Scholar] [CrossRef] [Green Version]
  75. Semenza, G.L.; Agani, F.; Booth, G.; Forsythe, J.; Iyer, N.; Jiang, B.H.; Leung, S.; Roe, R.; Wiener, C.; Yu, A. Structural and functional analysis of hypoxia-inducible factor 1. Kidney Int. 1997, 51, 553–555. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Fukuda, R.; Zhang, H.; Kim, J.W.; Shimoda, L.; Dang, C.V.; Semenza, G.L. HIF-1 regulates cytochrome oxidase subunits to optimize efficiency of respiration in hypoxic cells. Cell 2007, 129, 111–122. [Google Scholar] [CrossRef] [Green Version]
  77. Forsythe, J.A.; Jiang, B.H.; Iyer, N.V.; Agani, F.; Leung, S.W.; Koos, R.D.; Semenza, G.L. Activation of vascular endothelial growth factor gene transcription by hypoxia-inducible factor 1. Mol. Cell. Biol. 1996, 16, 4604–4613. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Levy, N.S.; Chung, S.; Furneaux, H.; Levy, A.P. Hypoxic stabilization of vascular endothelial growth factor mRNA by the RNA-binding protein HuR. J. Biol. Chem. 1998, 273, 6417–6423. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Tang, N.; Wang, L.; Esko, J.; Giordano, F.J.; Huang, Y.; Gerber, H.P.; Ferrara, N.; Johnson, R.S. Loss of HIF-1α in endothelial cells disrupts a hypoxia-driven VEGF autocrine loop necessary for tumorigenesis. Cancer Cell 2004, 6, 485–495. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  80. Gao, N.; Ding, M.; Zheng, J.Z.; Zhang, Z.; Leonard, S.S.; Liu, K.J.; Shi, X.; Jiang, B.H. Vanadate-induced expression of hypoxia-inducible factor 1 α and vascular endothelial growth factor through phosphatidylinositol 3-kinase/Akt pathway and reactive oxygen species. J. Biol. Chem. 2002, 277, 31963–31971. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  81. Herb, M.; Schramm, M. Functions of ROS in macrophages and antimicrobial immunity. Antioxidants 2021, 10, 313. [Google Scholar] [CrossRef]
  82. Klimova, T.; Chandel, N.S. Mitochondrial complex III regulates hypoxic activation of HIF. Cell. Death Differ. 2008, 15, 660–666. [Google Scholar] [CrossRef] [Green Version]
  83. Jung, S.N.; Yang, W.K.; Kim, J.; Kim, H.S.; Kim, E.J.; Yun, H.; Park, H.; Kim, S.S.; Choe, W.; Kang, I.; et al. Reactive oxygen species stabilize hypoxia-inducible factor-1 α protein and stimulate transcriptional activity via AMP-activated protein kinase in DU145 human prostate cancer cells. Carcinogenesis 2008, 29, 713–721. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Chandel, N.S.; McClintock, D.S.; Feliciano, C.E.; Wood, T.M.; Melendez, J.A.; Rodriguez, A.M.; Schumacker, P.T. Reactive oxygen species generated at mitochondrial complex III stabilize hypoxia-inducible factor-1α during hypoxia: A mechanism of O2 sensing. J. Biol. Chem. 2000, 275, 25130–25138. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Kim, J.W.; Tchernyshyov, I.; Semenza, G.L.; Dang, C.V. HIF-1-mediated expression of pyruvate dehydrogenase kinase: A metabolic switch required for cellular adaptation to hypoxia. Cell Metab. 2006, 3, 177–185. [Google Scholar] [CrossRef] [Green Version]
  86. Papandreou, I.; Cairns, R.A.; Fontana, L.; Lim, A.L.; Denko, N.C. HIF-1 mediates adaptation to hypoxia by actively downregulating mitochondrial oxygen consumption. Cell Metab. 2006, 3, 187–197. [Google Scholar] [CrossRef] [Green Version]
  87. Sun, Q.; Kim, H.E.; Cho, H.; Shi, S.; Kim, B.; Kim, O. Red light-emitting diode irradiation regulates oxidative stress and inflammation through SPHK1/NF-κB activation in human keratinocytes. J. Photochem. Photobiol. B 2018, 186, 31–40. [Google Scholar] [CrossRef]
  88. Li, Q.; Qian, J.; Li, Y.; Huang, P.; Liang, H.; Sun, H.; Liu, C.; Peng, J.; Lin, X.; Chen, X.; et al. Generation of sphingosine-1-phosphate by sphingosine kinase 1 protects nonalcoholic fatty liver from ischemia/reperfusion injury through alleviating reactive oxygen species production in hepatocytes. Free Radic. Biol. Med. 2020, 159, 136–149. [Google Scholar] [CrossRef]
  89. Xia, C.; Meng, Q.; Liu, L.-Z.; Rojanasakul, Y.; Wang, X.-R.; Jiang, B.-H. Reactive oxygen species regulate angiogenesis and tumor growth through vascular endothelial growth factor. Cancer Res. 2007, 67, 10823–10830. [Google Scholar] [CrossRef] [Green Version]
  90. Ader, I.; Brizuela, L.; Bouquerel, P.; Malavaud, B.; Cuvillier, O. Sphingosine kinase 1: A new modulator of hypoxia inducible factor 1α during hypoxia in human cancer cells. Cancer Res. 2008, 68, 8635–8642. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  91. Cho, S.-Y.; Lee, H.-J.; Jeong, S.-J.; Kim, H.-S.; Chen, C.Y.; Lee, E.-O.; Kim, S.-H. Sphingosine kinase 1 pathway is involved in melatonin-induced HIF-1α inactivation in hypoxic PC-3 prostate cancer cells. J. Pineal Res. 2011, 51, 87–93. [Google Scholar] [CrossRef] [PubMed]
  92. Bell, E.L.; Klimova, T.A.; Eisenbart, J.; Moraes, C.T.; Murphy, M.P.; Budinger, G.R.; Chandel, N.S. The Qo site of the mitochondrial complex III is required for the transduction of hypoxic signaling via reactive oxygen species production. J. Cell Biol. 2007, 177, 1029–1036. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Chandel, N.S.; Maltepe, E.; Goldwasser, E.; Mathieu, C.E.; Simon, M.C.; Schumacker, P.T. Mitochondrial reactive oxygen species trigger hypoxia-induced transcription. Proc. Natl. Acad. Sci. USA 1998, 95, 11715–11720. [Google Scholar] [CrossRef] [Green Version]
  94. Kaelin, W.G., Jr.; Ratcliffe, P.J. Oxygen sensing by metazoans: The central role of the HIF hydroxylase pathway. Mol. Cell 2008, 30, 393–402. [Google Scholar] [CrossRef] [PubMed]
  95. Du, J.; Xu, R.; Hu, Z.; Tian, Y.; Zhu, Y.; Gu, L.; Zhou, L. PI3K and ERK-induced Rac1 activation mediates hypoxia-induced HIF-1α expression in MCF-7 breast cancer cells. PLoS ONE 2011, 6, e25213. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Mylonis, I.; Chachami, G.; Samiotaki, M.; Panayotou, G.; Paraskeva, E.; Kalousi, A.; Georgatsou, E.; Bonanou, S.; Simos, G. Identification of MAPK phosphorylation sites and their role in the localization and activity of hypoxia-inducible factor-1α. J. Biol. Chem. 2006, 281, 33095–33106. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Koshikawa, N.; Hayashi, J.; Nakagawara, A.; Takenaga, K. Reactive oxygen species-generating mitochondrial DNA mutation up-regulates hypoxia-inducible factor-1α gene transcription via phosphatidylinositol 3-kinase-Akt/protein kinase C/histone deacetylase pathway. J. Biol. Chem. 2009, 284, 33185–33194. [Google Scholar] [CrossRef] [Green Version]
  98. Flügel, D.; Görlach, A.; Michiels, C.; Kietzmann, T. Glycogen synthase kinase 3 phosphorylates hypoxia-inducible factor 1α and mediates its destabilization in a VHL-independent manner. Mol. Cell. Biol. 2007, 27, 3253–3265. [Google Scholar] [CrossRef] [Green Version]
  99. Zhang, Z.; Yao, L.; Yang, J.; Wang, Z.; Du, G. PI3K/Akt and HIF1 signaling pathway in hypoxiaischemia (Review). Mol. Med. Rep. 2018, 18, 3547–3554. [Google Scholar]
  100. Liu, M.-H.; Li, G.-H.; Peng, L.-J.; Qu, S.-L.; Zhang, Y.; Peng, J.; Luo, X.-Y.; Hu, H.-J.; Ren, Z.; Liu, Y.; et al. PI3K/Akt/FoxO3a signaling mediates cardioprotection of FGF-2 against hydrogen peroxide-induced apoptosis in H9c2 cells. Mol. Cell. Biochem. 2016, 414, 57–66. [Google Scholar] [CrossRef]
  101. Liu, J.; Wu, P.; Xu, Z.; Zhang, J.; Liu, J.; Yang, Z. Ginkgolide B inhibits hydrogen peroxide-induced apoptosis and attenuates cytotoxicity via activating the PI3K/Akt/mTOR signaling pathway in H9c2 cells. Mol. Med. Rep. 2020, 22, 310–316. [Google Scholar] [CrossRef]
  102. Galanis, A.; Pappa, A.; Giannakakis, A.; Lanitis, E.; Dangaj, D.; Sandaltzopoulos, R. Reactive oxygen species and HIF-1 signalling in cancer. Cancer Lett. 2008, 266, 12–20. [Google Scholar] [CrossRef] [PubMed]
  103. Sodhi, A.; Montaner, S.; Miyazaki, H.; Gutkind, J.S. MAPK and Akt act cooperatively but independently on hypoxia inducible factor-1α in rasV12 upregulation of VEGF. Biochem. Biophys. Res. Commun. 2001, 287, 292–300. [Google Scholar] [CrossRef]
  104. Mottet, D.; Dumont, V.; Deccache, Y.; Demazy, C.; Ninane, N.; Raes, M.; Michiels, C. Regulation of hypoxia-inducible factor-1α protein level during hypoxic conditions by the phosphatidylinositol 3-kinase/Akt/glycogen synthase kinase 3beta pathway in HepG2 cells. J. Biol. Chem. 2003, 278, 31277–31285. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Dimova, E.Y.; Michiels, C.; Kietzmann, T. Kinases as upstream regulators of the HIF system: Their emerging potential as anti-cancer drug targets. Curr. Pharm. Des. 2009, 15, 3867–3877. [Google Scholar] [CrossRef] [PubMed]
  106. Karar, J.; Cerniglia, G.J.; Lindsten, T.; Koumenis, C.; Maity, A. Dual PI3K/mTOR inhibitor NVP-BEZ235 suppresses hypoxia-inducible factor (HIF)-1α expression by blocking protein translation and increases cell death under hypoxia. Cancer Biol. Ther. 2012, 13, 1102–1111. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Zhong, H.; Chiles, K.; Feldser, D.; Laughner, E.; Hanrahan, C.; Georgescu, M.M.; Simons, J.W.; Semenza, G.L. Modulation of hypoxia-inducible factor 1α expression by the epidermal growth factor/phosphatidylinositol 3-kinase/PTEN/AKT/FRAP pathway in human prostate cancer cells: Implications for tumor angiogenesis and therapeutics. Cancer Res. 2000, 60, 1541–1545. [Google Scholar]
  108. Yang, X.-M.; Wang, Y.-S.; Zhang, J.; Li, Y.; Xu, J.-F.; Zhu, J.; Zhao, W.; Chu, D.-K.; Wiedemann, P. Role of PI3K/Akt and MEK/ERK in mediating hypoxia-induced expression of HIF-1 and VEGF in laser-induced rat choroidal neovascularization. Investig. Opthalmol. Vis. Sci. 2009, 50, 1873–1879. [Google Scholar] [CrossRef] [PubMed]
  109. Li, L.; Qu, Y.; Mao, M.; Xiong, Y.; Mu, D. The involvement of phosphoinositid 3-kinase/Akt pathway in the activation of hypoxia-inducible factor-1α in the developing rat brain after hypoxia–ischemia. Brain Res. 2008, 1197, 152–158. [Google Scholar] [CrossRef] [PubMed]
  110. Kilic-Eren, M.; Boylu, T.; Tabor, V. Targeting PI3K/Akt represses hypoxia inducible factor-1α activation and sensitizes Rhabdomyosarcoma and Ewing’s sarcoma cells for apoptosis. Cancer Cell Int. 2013, 13, 1–36. [Google Scholar] [CrossRef] [Green Version]
  111. Jung, S. Macrophages and monocytes in 2017: Macrophages and monocytes: Of tortoises and hares. Nat. Rev. Immunol. 2018, 18, 85–86. [Google Scholar] [CrossRef] [PubMed]
  112. Epelman, S.; Lavine, K.J.; Randolph, G.J. Origin and functions of tissue macrophages. Immunity 2014, 41, 21–35. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Davies, L.C.; Jenkins, S.J.; Allen, J.E.; Taylor, P.R. Tissue-resident macrophages. Nat. Immunol. 2013, 14, 986–995. [Google Scholar] [CrossRef] [PubMed]
  114. Mitchell, G.; Chen, C.; Portnoy, D.A. Strategies used by bacteria to grow in macrophages. Microbiol. Spectr. 2016, 4, 701–725. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Haas, A. The phagosome: Compartment with a license to kill. Traffic 2007, 8, 311–330. [Google Scholar] [CrossRef]
  116. Djaldetti, M.; Salman, H.; Bergman, M.; Djaldetti, R.; Bessler, H. Phagocytosis--the mighty weapon of the silent warriors. Microsc. Res. Tech. 2002, 57, 421–431. [Google Scholar] [CrossRef]
  117. Lemke, G. How macrophages deal with death. Nat. Rev. Immunol. 2019, 19, 539–549. [Google Scholar] [CrossRef]
  118. Weiss, G.; Schaible, U.E. Macrophage defense mechanisms against intracellular bacteria. Immunol. Rev. 2015, 264, 182–203. [Google Scholar] [CrossRef] [Green Version]
  119. Del Cerro-Vadillo, E.; Madrazo-Toca, F.; Marin, E.C.; Fernandez-Prieto, L.; Beck, C.; Leyva-Cobián, F.; Saftig, P.; Alvarez-Dominguez, C. Cutting edge: A novel nonoxidative phagosomal mechanism exerted by cathepsin-D controls listeria monocytogenes intracellular growth. J. Immunol. 2006, 176, 1321–1325. [Google Scholar] [CrossRef] [Green Version]
  120. Alvarez-Dominguez, C.; Carrasco-Marin, E.; Lopez-Mato, P.; Leyva-Cobian, F. The contribution of both oxygen and nitrogen intermediates to the intracellular killing mechanisms of C1q-opsonized Listeria monocytogenes by the macrophage-like IC-21 cell line. Immunology 2000, 101, 83–89. [Google Scholar] [CrossRef]
  121. Arango Duque, G.; Descoteaux, A. Macrophage cytokines: Involvement in immunity and infectious diseases. Front. Immunol. 2014, 5, 491. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Emerling, B.M.; Platanias, L.C.; Black, E.; Nebreda, A.R.; Davis, R.J.; Chandel, N.S. Mitochondrial reactive oxygen species activation of p38 mitogen-activated protein kinase is required for hypoxia signaling. Mol. Cell. Biol. 2005, 25, 4853–4862. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Salmeen, A.; Barford, D. Functions and mechanisms of redox regulation of cysteine-based phosphatases. Antioxid. Redox Signal 2005, 7, 560–577. [Google Scholar] [CrossRef] [PubMed]
  124. Taylor, C.T.; Colgan, S.P. Regulation of immunity and inflammation by hypoxia in immunological niches. Nat. Rev. Immunol. 2017, 17, 774–785. [Google Scholar] [CrossRef]
  125. Palazon, A.; Goldrath, A.W.; Nizet, V.; Johnson, R.S. HIF transcription factors, inflammation, and immunity. Immunity 2014, 41, 518–528. [Google Scholar] [CrossRef] [Green Version]
  126. Peyssonnaux, C.; Datta, V.; Cramer, T.; Doedens, A.; Theodorakis, E.A.; Gallo, R.L.; Hurtado-Ziola, N.; Nizet, V.; Johnson, R.S. HIF-1α expression regulates the bactericidal capacity of phagocytes. J. Clin. Investig. 2005, 115, 1806–1815. [Google Scholar] [CrossRef] [Green Version]
  127. Lopez-Pascual, A.; Lorente-Cebrian, S.; Moreno-Aliaga, M.J.; Martinez, J.A.; Gonzalez-Muniesa, P. Inflammation stimulates hypoxia-inducible factor-1α regulatory activity in 3T3-L1 adipocytes with conditioned medium from lipopolysaccharide-activated RAW 264.7 macrophages. J. Cell. Physiol. 2018, 234, 550–560. [Google Scholar] [CrossRef] [Green Version]
  128. Aarup, A.; Pedersen, T.X.; Junker, N.; Christoffersen, C.; Bartels, E.D.; Madsen, M.; Nielsen, C.H.; Nielsen, L.B. Hypoxia-inducible factor-1α expression in macrophages promotes development of atherosclerosis. Arter. Thromb. Vasc. Biol. 2016, 36, 1782–1790. [Google Scholar] [CrossRef] [Green Version]
  129. Watts, E.R.; Walmsley, S.R. Inflammation and hypoxia: HIF and PHD isoform selectivity. Trends Mol. Med. 2019, 25, 33–46. [Google Scholar] [CrossRef] [Green Version]
  130. Wang, T.; Liu, H.; Lian, G.; Zhang, S.-Y.; Wang, X.; Jiang, C. HIF1α-induced glycolysis metabolism is essential to the activation of inflammatory macrophages. Mediat. Inflamm. 2017, 2017, 9029327. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  131. Ferrante, C.J.; Leibovich, S.J. Regulation of macrophage polarization and wound healing. Adv. Wound Care (N. Rochelle) 2012, 1, 10–16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Murray, P.J.; Wynn, T.A. Protective and pathogenic functions of macrophage subsets. Nat. Rev. Immunol. 2011, 11, 723–737. [Google Scholar] [CrossRef] [PubMed]
  133. Murray, P.J.; Allen, J.; Biswas, S.K.; Fisher, E.; Gilroy, D.; 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] [Green Version]
  134. Vannella, K.M.; Wynn, T.A. Mechanisms of organ injury and repair by macrophages. Annu. Rev. Physiol. 2017, 79, 593–617. [Google Scholar] [CrossRef]
  135. Krzyszczyk, P.; Schloss, R.; Palmer, A.; Berthiaume, F. The role of macrophages in acute and chronic wound healing and interventions to promote pro-wound healing phenotypes. Front. Physiol. 2018, 9, 419. [Google Scholar] [CrossRef]
  136. Mantovani, A.; Sica, A.; Sozzani, S.; Allavena, P.; Vecchi, A.; Locati, M. The chemokine system in diverse forms of macrophage activation and polarization. Trends Immunol. 2004, 25, 677–686. [Google Scholar] [CrossRef]
  137. Hildebrand, D.G.; Alexander, E.; Horber, S.; Lehle, S.; Obermayer, K.; Munck, N.A.; Rothfuss, O.; Frick, J.S.; Morimatsu, M.; Schmitz, I.; et al. IκBzeta is a transcriptional key regulator of CCL2/MCP-1. J. Immunol. 2013, 190, 4812–4820. [Google Scholar] [CrossRef]
  138. Oliver, K.M.; Taylor, C.T.; Cummins, E.P. Hypoxia. Regulation of NFκB signalling during inflammation: The role of hydroxylases. Arthritis Res. Ther. 2009, 11, 215. [Google Scholar] [CrossRef] [Green Version]
  139. Fang, H.-Y.; Hughes, R.; Murdoch, C.; Coffelt, S.; Biswas, S.K.; Harris, A.; Johnson, R.; Imityaz, H.Z.; Simon, M.C.; Fredlund, E.; et al. Hypoxia-inducible factors 1 and 2 are important transcriptional effectors in primary macrophages experiencing hypoxia. Blood 2009, 114, 844–859. [Google Scholar] [CrossRef] [Green Version]
  140. Tannahill, G.M.; Curtis, A.M.; Adamik, J.; Palsson-McDermott, E.M.; McGettrick, A.F.; Goel, G.; Frezza, C.; Bernard, N.J.; Kelly, B.; Foley, N.H.; et al. Succinate is an inflammatory signal that induces IL-1beta through HIF-1α. Nature 2013, 496, 238–242. [Google Scholar] [CrossRef]
  141. Hesketh, M.; Sahin, K.B.; West, Z.E.; Murray, R.Z. Macrophage phenotypes regulate scar formation and chronic wound healing. Int. J. Mol. Sci. 2017, 18, 1545. [Google Scholar] [CrossRef] [Green Version]
  142. St-Pierre, J.; Lin, J.; Krauss, S.; Tarr, P.T.; Yang, R.; Newgard, C.B.; Spiegelman, B.M. Bioenergetic analysis of peroxisome proliferator-activated receptor gamma coactivators 1α and 1β (PGC-1α and PGC-1β) in muscle cells. J. Biol. Chem. 2003, 278, 26597–26603. [Google Scholar] [CrossRef] [Green Version]
  143. Shao, D.; Liu, Y.; Liu, X.; Zhu, L.; Cui, Y.; Cui, A.; Qiao, A.; Kong, X.; Liu, Y.; Chen, Q.; et al. PGC-1β-regulated mitochondrial biogenesis and function in myotubes is mediated by NRF-1 and ERR α. Mitochondrion 2010, 10, 516–527. [Google Scholar] [CrossRef] [PubMed]
  144. Vats, D.; Mukundan, L.; Odegaard, J.I.; Zhang, L.; Smith, K.L.; Morel, C.R.; Wagner, R.A.; Greaves, D.R.; Murray, P.J.; Chawla, A. Oxidative metabolism and PGC-1β attenuate macrophage-mediated inflammation. Cell Metab. 2006, 4, 13–24. [Google Scholar] [CrossRef] [Green Version]
  145. Desanta, F.; Totaro, M.G.; Prosperini, E.; Notarbartolo, S.; Testa, G.; Natoli, G. The histone H3 lysine-27 demethylase Jmjd3 links inflammation to inhibition of polycomb-mediated gene silencing. Cell 2007, 130, 1083–1094. [Google Scholar] [CrossRef] [Green Version]
  146. Satoh, T.; Takeuchi, O.; Vandenbon, A.; Yasuda, K.; Tanaka, Y.; Kumagai, Y.; Miyake, T.; Matsushita, K.; Okazaki, T.; Saitoh, T.; et al. The Jmjd3-Irf4 axis regulates M2 macrophage polarization and host responses against helminth infection. Nat. Immunol. 2010, 11, 936–944. [Google Scholar] [CrossRef] [PubMed]
  147. Cyster, J.G.; Schwab, S. Sphingosine-1-phosphate and lymphocyte egress from lymphoid organs. Annu. Rev. Immunol. 2012, 30, 69–94. [Google Scholar] [CrossRef] [PubMed]
  148. Spiegel, S.; Milstien, S. Sphingosine-1-phosphate: An enigmatic signalling lipid. Nat. Rev. Mol. Cell. Biol. 2003, 4, 397–407. [Google Scholar] [CrossRef] [PubMed]
  149. Bruce, C.R.; Risis, S.; Babb, J.R.; Yang, C.; Kowalski, G.M.; Selathurai, A.; Lee-Young, R.S.; Weir, J.M.; Yoshioka, K.; Takuwa, Y.; et al. Overexpression of sphingosine kinase 1 prevents ceramide accumulation and ameliorates muscle insulin resistance in high-fat diet-fed mice. Diabetes 2012, 61, 3148–3155. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  150. Spiegel, S.; Milstien, S. Functions of the multifaceted family of sphingosine kinases and some close relatives. J. Biol. Chem. 2007, 282, 2125–2129. [Google Scholar] [CrossRef] [Green Version]
  151. Hannun, Y.A.; Obeid, L. Principles of bioactive lipid signalling: Lessons from sphingolipids. Nat. Rev. Mol. Cell Biol. 2008, 9, 139–150. [Google Scholar] [CrossRef] [PubMed]
  152. Merrill, A.H., Jr.; Stokes, T.H.; Momin, A.; Park, H.; Portz, B.J.; Kelly, S.; Wang, E.; Sullards, M.C.; Wang, M.D. Sphingolipidomics: A valuable tool for understanding the roles of sphingolipids in biology and disease. J. Lipid Res. 2009, 50, S97–S102. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Pitman, M.; Costabile, M.; Pitson, S.M. Recent advances in the development of sphingosine kinase inhibitors. Cell. Signal. 2016, 28, 1349–1363. [Google Scholar] [CrossRef] [PubMed]
  154. Adams, D.R.; Pyne, S.; Pyne, N.J. Sphingosine kinases: Emerging structure-function insights. Trends Biochem. Sci. 2016, 41, 395–409. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Spiegel, S.; English, D.; Milstien, S. Sphingosine 1-phosphate signaling: Providing cells with a sense of direction. Trends Cell Biol. 2002, 12, 236–242. [Google Scholar] [CrossRef]
  156. Rosen, H.; Stevens, R.; Hanson, M.; Roberts, E.; Oldstone, M.B. Sphingosine-1-phosphate and its receptors: Structure, signaling, and influence. Annu. Rev. Biochem. 2013, 82, 637–662. [Google Scholar] [CrossRef]
  157. Blaho, V.; Hla, T. An update on the biology of sphingosine 1-phosphate receptors. J. Lipid Res. 2014, 55, 1596–1608. [Google Scholar] [CrossRef] [Green Version]
  158. Lattin, J.; Zidar, D.A.; Schroder, K.; Kellie, S.; Hume, D.A.; Sweet, M.J. G-protein-coupled receptor expression, function, and signaling in macrophages. J. Leukoc. Biol. 2007, 82, 16–32. [Google Scholar] [CrossRef] [Green Version]
  159. Worzfeld, T.; Wettschureck, N.; Offermanns, S. G12/G13-mediated signalling in mammalian physiology and disease. Trends Pharmacol. Sci. 2008, 29, 582–589. [Google Scholar] [CrossRef]
  160. Siehler, S. Regulation of RhoGEF proteins by G12/13-coupled receptors. Br. J. Pharmacol. 2009, 158, 41–49. [Google Scholar] [CrossRef] [Green Version]
  161. Di, A.; Kawamura, T.; Gao, X.P.; Tang, H.; Berdyshev, E.; Vogel, S.M.; Zhao, Y.Y.; Sharma, T.; Bachmaier, K.; Xu, J.; et al. A novel function of sphingosine kinase 1 suppression of JNK activity in preventing inflammation and injury. J. Biol. Chem. 2010, 285, 15848–15857. [Google Scholar] [CrossRef] [Green Version]
  162. Nishi, T.; Kobayashi, N.; Hisano, Y.; Kawahara, A.; Yamaguchi, A. Molecular and physiological functions of sphingosine 1-phosphate transporters. Biochim. Biophys. Acta 2014, 1841, 759–765. [Google Scholar] [CrossRef]
  163. Osborne, N.; Brand-Arzamendi, K.; Ober, E.A.; Jin, S.-W.; Verkade, H.; Holtzman, N.G.; Yelon, D.; Stainier, D.Y. The spinster homolog, two of hearts, is required for sphingosine 1-phosphate signaling in zebrafish. Curr. Biol. 2008, 18, 1882–1888. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Kawahara, A.; Nishi, T.; Hisano, Y.; Fukui, H.; Yamaguchi, A.; Mochizuki, N. The sphingolipid transporter spns2 functions in migration of zebrafish myocardial precursors. Science 2009, 323, 524–527. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Pappu, R.; Schwab, S.R.; Cornelissen, I.; Pereira, J.P.; Regard, J.B.; Xu, Y.; Camerer, E.; Zheng, Y.W.; Huang, Y.; Cyster, J.G.; et al. Promotion of lymphocyte egress into blood and lymph by distinct sources of sphingosine-1-phosphate. Science 2007, 316, 295–298. [Google Scholar] [CrossRef] [PubMed]
  166. Hait, N.C.; Allegood, J.; Maceyka, M.; Strub, G.M.; Harikumar, K.B.; Singh, S.K.; Luo, C.; Marmorstein, R.; Kordula, T.; Milstien, S.; et al. Regulation of histone acetylation in the nucleus by sphingosine-1-phosphate. Science 2009, 325, 1254–1257. [Google Scholar] [CrossRef] [Green Version]
  167. Mori, H.; Izawa, T.; Tanaka, E. Smad3 deficiency leads to mandibular condyle degradation via the sphingosine 1-phosphate (S1P)/S1P3 signaling axis. Am. J. Pathol. 2015, 185, 2742–2756. [Google Scholar] [CrossRef]
  168. Alvarez, S.E.; Harikumar, K.; Hait, N.C.; Allegood, J.; Strub, G.M.; Kim, E.Y.; Maceyka, M.; Jiang, H.; Luo, C.; Kordula, T.; et al. Sphingosine-1-phosphate is a missing cofactor for the E3 ubiquitin ligase TRAF2. Nat. Cell Biol. 2010, 465, 1084–1088. [Google Scholar] [CrossRef] [Green Version]
  169. Strub, G.M.; Paillard, M.; Liang, J.; Gomez, L.; Allegood, J.C.; Hait, N.C.; Maceyka, M.; Price, M.M.; Chen, Q.; Simpson, D.; et al. Sphingosine-1-phosphate produced by sphingosine kinase 2 in mitochondria interacts with prohibitin 2 to regulate complex IV assembly and respiration. FASEB J. 2010, 25, 600–612. [Google Scholar] [CrossRef] [Green Version]
  170. Maceyka, M.; Alvarez, S.E.; Milstien, S.; Spiegel, S. Filamin A links sphingosine kinase 1 and sphingosine-1-phosphate receptor 1 at lamellipodia to orchestrate cell migration. Mol. Cell Biol. 2008, 28, 5687–5697. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  171. Ishii, M.; Egen, J.G.; Klauschen, F.; Meier-Schellersheim, M.; Saeki, Y.; Vacher, J.; Proia, R.L.; Germain, R.N. Sphingosine-1-phosphate mobilizes osteoclast precursors and regulates bone homeostasis. Nature 2009, 458, 524–528. [Google Scholar] [CrossRef]
  172. Yu, H. Sphingosine-1-phosphate receptor 2 regulates proinflammatory cytokine production and osteoclastogenesis. PLoS ONE 2016, 11, e0156303. [Google Scholar] [CrossRef] [PubMed]
  173. Song, F.; Hou, J.; Chen, Z.; Cheng, B.; Lei, R.; Cui, P.; Sun, Y.; Wang, H.; Fang, X. Sphingosine-1-phosphate receptor 2 signaling promotes caspase-11-dependent macrophage pyroptosis and worsens escherichia coli sepsis outcome. Anesthesiology 2018, 129, 311–320. [Google Scholar] [CrossRef] [PubMed]
  174. Brecht, K.; Weigert, A.; Hu, J.; Popp, R.; Fisslthaler, B.; Korff, T.; Fleming, I.; Geisslinger, G.; Brune, B. Macrophages programmed by apoptotic cells promote angiogenesis via prostaglandin E2. FASEB J. 2011, 25, 2408–2417. [Google Scholar] [CrossRef]
  175. Hutami, I.R.; Tanaka, E.; Izawa, T. Crosstalk between Fas and S1P1 signaling via NF-kB in osteoclasts controls bone destruction in the TMJ due to rheumatoid arthritis. Jpn. Dent. Sci. Rev. 2019, 55, 12–19. [Google Scholar] [CrossRef] [PubMed]
  176. Das, A.; Segar, C.E.; Hughley, B.B.; Bowers, D.T.; Botchwey, E.A. The promotion of mandibular defect healing by the targeting of S1P receptors and the recruitment of alternatively activated macrophages. Biomaterials 2013, 34, 9853–9862. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  177. Hou, J.; Chen, Q.; Wu, X.; Zhao, D.; Reuveni, H.; Licht, T.; Xu, M.; Hu, H.; Hoeft, A.; Ben-Sasson, S.A.; et al. S1PR3 signaling drives bacterial killing and is required for survival in bacterial sepsis. Am. J. Respir. Crit. Care Med. 2017, 196, 1559–1570. [Google Scholar] [CrossRef] [PubMed]
  178. Heilmann, A.; Schinke, T.; Bindl, R.; Wehner, T.; Rapp, A.; Haffner-Luntzer, M.; Liedert, A.; Amling, M.; Ignatius, A. Systemic treatment with the sphingosine-1-phosphate analog FTY720 does not improve fracture healing in mice. J. Orthop. Res. 2013, 31, 1845–1850. [Google Scholar] [CrossRef]
  179. Yang, J.; Yang, L.; Tian, L.; Ji, X.; Yang, L.; Li, L. Sphingosine 1-phosphate (S1P)/S1P receptor2/3 axis promotes inflammatory M1 polarization of bone marrow-derived monocyte/macrophage via G(α)i/o/PI3K/JNK pathway. Cell Physiol. Biochem. 2018, 49, 1677–1693. [Google Scholar] [CrossRef] [PubMed]
  180. Hymel, L.A.; Ogle, M.E.; Anderson, S.E.; Emeterio, C.L.S.; Turner, T.C.; York, W.Y.; Liu, A.Y.; Olingy, C.E.; Sridhar, S.; Lim, H.S.; et al. Modulating local S1P receptor signaling as a regenerative immunotherapy after volumetric muscle loss injury. J. Biomed. Mater. Res. Part A 2020, 109, 695–712. [Google Scholar] [CrossRef] [PubMed]
  181. Yu, H.; Sun, C.; Argraves, K.M. Periodontal inflammation and alveolar bone loss induced by Aggregatibacter actinomycetemcomitans is attenuated in sphingosine kinase 1-deficient mice. J. Periodontal Res. 2016, 51, 38–49. [Google Scholar] [CrossRef] [Green Version]
  182. Hoeffel, G.; Wang, Y.; Greter, M.; See, P.; Teo, P.; Malleret, B.; Leboeuf, M.; Low, D.; Oller, G.; de Almeida, F.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] [PubMed] [Green Version]
  183. Minutti, C.; Knipper, J.; Allen, J.E.; Zaiss, D.M. Tissue-specific contribution of macrophages to wound healing. Semin. Cell Dev. Biol. 2017, 61, 3–11. [Google Scholar] [CrossRef] [Green Version]
  184. DiPietro, L. Wound healing: The role of the macrophage and other immune cells. Shock 1995, 4, 233–240. [Google Scholar] [CrossRef]
  185. Lin, N.; Simon, M.C. Hypoxia-inducible factors: Key regulators of myeloid cells during inflammation. J. Clin. Investig. 2016, 126, 3661–3671. [Google Scholar] [CrossRef] [PubMed]
  186. Chen, L.; Gajendrareddy, P.; DiPietro, L.A. Differential expression of HIF-1α in skin and mucosal wounds. J. Dent. Res. 2012, 91, 871–876. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  187. Le, A.D.; Basi, D.L.; Abubaker, A.O. Wound healing: Findings of the 2005 AAOMS research summit. J. Oral Maxillofac. Surg. 2005, 63, 1426–1435. [Google Scholar] [CrossRef]
  188. Martin, P.; Leibovich, S.J. Inflammatory cells during wound repair: The good, the bad and the ugly. Trends Cell Biol. 2005, 15, 599–607. [Google Scholar] [CrossRef]
  189. Reinke, J.M.; Sorg, H. Wound repair and regeneration. Eur. Surg. Res. 2012, 49, 35–43. [Google Scholar] [CrossRef]
  190. Brancato, S.K.; Albina, J.E. Wound macrophages as key regulators of repair: Origin, phenotype, and function. Am. J. Pathol. 2011, 178, 19–25. [Google Scholar] [CrossRef]
  191. Sindrilaru, A.; Peters, T.; Wieschalka, S.; Baican, C.; Baican, A.; Peter, H.; Hainzl, A.; Schatz, S.; Qi, Y.; Schlecht, A.; et al. An unrestrained proinflammatory M1 macrophage population induced by iron impairs wound healing in humans and mice. J. Clin. Investig. 2011, 121, 985–997. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  192. Lucas, T.; Waisman, A.; Ranjan, R.; Roes, J.; Krieg, T.; Müller, W.; Roers, A.; Eming, S.A. Differential roles of macrophages in diverse phases of skin repair. J. Immunol. 2010, 184, 3964–3977. [Google Scholar] [CrossRef] [Green Version]
  193. Szpaderska, A.M.; Zuckerman, J.D.; DiPietro, L.A. Differential injury responses in oral mucosal and cutaneous wounds. J. Dent. Res. 2003, 82, 621–626. [Google Scholar] [CrossRef] [PubMed]
  194. Yoshino, F.; Yoshida, A.; Nakajima, A.; Wada-Takahashi, -S.; Takahashi, S.S.-I.; Lee, M.C. Alteration of the redox state with reactive oxygen species for 5-fluorouracil-induced oral mucositis in hamsters. PLoS ONE 2013, 8, e82834. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  195. Glim, J.E.; Beelen, R.H.; Niessen, F.B.; Everts, V.; Ulrich, M.M. The number of immune cells is lower in healthy oral mucosa compared to skin and does not increase after scarring. Arch. Oral Biol. 2015, 60, 272–281. [Google Scholar] [CrossRef] [PubMed]
  196. Szpaderska, A.M.; Walsh, C.G.; Steinberg, M.J.; DiPietro, L.A. Distinct patterns of angiogenesis in oral and skin wounds. J. Dent. Res. 2005, 84, 309–314. [Google Scholar] [CrossRef]
  197. Wong, J.W.; Gallant-Behm, C.; Wiebe, C.; Mak, K.; Hart, D.A.; Larjava, H.; Häkkinen, L. Wound healing in oral mucosa results in reduced scar formation as compared with skin: Evidence from the red Duroc pig model and humans. Wound Repair Regen. 2009, 17, 717–729. [Google Scholar] [CrossRef]
  198. Iglesias-Bartolome, R.; Uchiyama, A.; Molinolo, A.A.; Abusleme, L.; Brooks, S.R.; Callejas-Valera, J.L.; Edwards, D.; Doci, C.; Asselin-Labat, M.-L.; Onaitis, M.W.; et al. Transcriptional signature primes human oral mucosa for rapid wound healing. Sci. Transl. Med. 2018, 10, eaap8798. [Google Scholar] [CrossRef] [Green Version]
  199. Mak, K.; Manji, A.; Gallant-Behm, C.; Wiebe, C.; Hart, D.A.; Larjava, H.; Häkkinen, L. Scarless healing of oral mucosa is characterized by faster resolution of inflammation and control of myofibroblast action compared to skin wounds in the red Duroc pig model. J. Dermatol. Sci. 2009, 56, 168–180. [Google Scholar] [CrossRef]
  200. O’Kane, S.; Ferguson, M.W. Transforming growth factor beta s and wound healing. Int. J. Biochem. Cell Biol. 1997, 29, 63–78. [Google Scholar] [CrossRef]
  201. Jinno, K.; Takahashi, T.; Tsuchida, K.; Tanaka, E.; Moriyama, K. Acceleration of palatal wound healing in Smad3-deficient mice. J. Dent. Res. 2009, 88, 757–761. [Google Scholar] [CrossRef] [PubMed]
  202. Yoneda, N.; Yasue, A.; Watanabe, T.; Tanaka, E. Down-regulation of Smad3 accelerates palatal wound repair. J. Dent. Res. 2013, 92, 716–720. [Google Scholar] [CrossRef]
  203. Wilgus, T.A. Immune cells in the healing skin wound: Influential players at each stage of repair. Pharmacol. Res. 2008, 58, 112–116. [Google Scholar] [CrossRef] [PubMed]
  204. Zelles, T.; Purushotham, K.R.; Macauley, S.P.; Oxford, G.E.; Humphreys-Beher, M.G. Saliva and growth factors: The fountain of youth resides in us all. J. Dent. Res. 1995, 74, 1826–1832. [Google Scholar] [CrossRef]
  205. Ashcroft, G.S.; Lei, K.; Jin, W.; Longenecker, G.; Kulkarni, A.B.; Greenwell-Wild, T.; Hale-Donze, H.; McGrady, G.; Song, X.-Y.; Wahl, S.M. Secretory leukocyte protease inhibitor mediates non-redundant functions necessary for normal wound healing. Nat. Med. 2000, 6, 1147–1153. [Google Scholar] [CrossRef]
  206. Tamaki, N.; Campos, R.C.O.; Fukui, M.; Ito, H.-O. Hydrogen-rich water intake accelerates oral palatal wound healing via activation of the Nrf2/antioxidant defense pathways in a rat model. Oxidative Med. Cell. Longev. 2015, 2016, 5679040. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  207. Yin, H.; Li, X.; Hu, S.; Liu, T.; Yuan, B.; Gu, H.; Ni, Q.; Zhang, X.; Zheng, F. IL-33 accelerates cutaneous wound healing involved in upregulation of alternatively activated macrophages. Mol. Immunol. 2013, 56, 347–353. [Google Scholar] [CrossRef]
  208. Yin, H.; Li, X.Y.; Yuan, B.H.; Zhang, B.B.; Hu, S.L.; Gu, H.B.; Jin, X.B.; Zhu, J.Y. Adenovirus-mediated overexpression of soluble ST2 provides a protective effect on lipopolysaccharide-induced acute lung injury in mice. Clin. Exp. Immunol. 2011, 164, 248–255. [Google Scholar] [CrossRef]
  209. Duan, L.; Chen, J.; Zhang, H.; Yang, H.; Zhu, P.; Xiong, A.; Xia, Q.; Zheng, F.; Tan, Z.; Gong, F.; et al. Interleukin-33 ameliorates experimental colitis through promoting Th2/Foxp3(+) regulatory T-cell responses in mice. Mol. Med. 2012, 18, 753–761. [Google Scholar] [CrossRef]
  210. Cramer, T.; Yamanishi, Y.; Clausen, B.E.; Forster, I.; Pawlinski, R.; Mackman, N.; Haase, V.H.; Jaenisch, R.; Corr, M.; Nizet, V.; et al. HIF-1α is essential for myeloid cell-mediated inflammation. Cell 2003, 112, 645–657. [Google Scholar] [CrossRef] [Green Version]
  211. Li, N.; Li, Y.; Li, Z.; Huang, C.; Yang, Y.; Lang, M.; Cao, J.; Jiang, W.; Xu, Y.; Dong, J.; et al. Hypoxia inducible factor 1 (HIF-1) recruits macrophage to activate pancreatic stellate cells in pancreatic ductal adenocarcinoma. Int. J. Mol. Sci. 2016, 17, 799. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  212. Prabhakar, N.R. Sensing hypoxia: Physiology, genetics and epigenetics. J. Physiol. 2013, 591, 2245–2257. [Google Scholar] [CrossRef]
  213. Ratcliffe, P.J. Oxygen sensing and hypoxia signalling pathways in animals: The implications of physiology for cancer. J. Physiol. 2013, 591, 2027–2042. [Google Scholar] [CrossRef]
  214. Loboda, A.; Jozkowicz, A.; Dulak, J. HIF-1 and HIF-2 transcription factors—Similar but not identical. Mol. Cells 2010, 29, 435–442. [Google Scholar] [CrossRef] [PubMed]
  215. Monick, M.M.; Mallampalli, R.K.; Bradford, M.; McCoy, D.; Gross, T.J.; Flaherty, D.M.; Powers, L.S.; Cameron, K.; Kelly, S.; Merrill, A.H.; et al. Cooperative prosurvival activity by ERK and Akt in human alveolar macrophages is dependent on high levels of acid ceramidase activity. J. Immunol. 2004, 173, 123–135. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  216. De Ponti, C.; Carini, R.; Alchera, E.; Nitti, M.P.; Locati, M.; Albano, E.; Cairo, G.; Tacchini, L. Adenosine A2a receptor-mediated, normoxic induction of HIF-1 through PKC and PI-3K-dependent pathways in macrophages. J. Leukoc. Biol. 2007, 82, 392–402. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  217. Blouin, C.C.; Page, E.L.; Soucy, G.M.; Richard, D.E. Hypoxic gene activation by lipopolysaccharide in macrophages: Implication of hypoxia-inducible factor 1α. Blood 2004, 103, 1124–1130. [Google Scholar] [CrossRef] [PubMed]
  218. Kim, S.Y.; Jeong, E.; Joung, S.M.; Lee, J.Y. PI3K/Akt contributes to increased expression of Toll-like receptor 4 in macrophages exposed to hypoxic stress. Biochem. Biophys. Res. Commun. 2012, 419, 466–471. [Google Scholar] [CrossRef] [PubMed]
  219. Semba, H.; Takeda, N.; Isagawa, T.; Sugiura, Y.; Honda, K.; Wake, M.; Miyazawa, H.; Yamaguchi, Y.; Miura, M.; Jenkins, D.M.R.; et al. HIF-1α-PDK1 axis-induced active glycolysis plays an essential role in macrophage migratory capacity. Nat. Commun. 2016, 7, 11635. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  220. Real-Hohn, A.; Zancan, P.; Da Silva, D.; Martins, E.R.; Salgado, L.T.; Mermelstein, C.S.; Gomes, A.M.; Sola-Penna, M. Filamentous actin and its associated binding proteins are the stimulatory site for 6-phosphofructo-1-kinase association within the membrane of human erythrocytes. Biochimie 2010, 92, 538–544. [Google Scholar] [CrossRef]
  221. Shiraishi, T.; Verdone, J.E.; Huang, J.; Kahlert, U.D.; Hernandez, J.R.; Torga, G.; Zarif, J.C.; Epstein, T.; Gatenby, R.; McCartney, A.; et al. Glycolysis is the primary bioenergetic pathway for cell motility and cytoskeletal remodeling in human prostate and breast cancer cells. Oncotarget 2015, 6, 130–143. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  222. Yan, H.; Zhang, D.-X.; Shi, X.; Zhang, Q.; Huang, Y.-S. Activation of the prolyl-hydroxylase oxygen-sensing signal cascade leads to AMPK activation in cardiomyocytes. J. Cell. Mol. Med. 2011, 16, 2049–2059. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  223. Takaku, M.; Tomita, S.; Kurobe, H.; Kihira, Y.; Morimoto, A.; Higashida, M.; Ikeda, Y.; Ushiyama, A.; Hashimoto, I.; Nakanishi, H.; et al. Systemic preconditioning by a prolyl hydroxylase inhibitor promotes prevention of skin flap necrosis via HIF-1-induced bone marrow-derived cells. PLoS ONE 2012, 7, e42964. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  224. Zhdanov, A.V.; Okkelman, I.A.; Collins, F.W.; Melgar, S.; Papkovsky, D.B. A novel effect of DMOG on cell metabolism: Direct inhibition of mitochondrial function precedes HIF target gene expression. Biochim. Biophys. Acta 2015, 1847, 1254–1266. [Google Scholar] [CrossRef] [Green Version]
  225. Reznick, R.M.; Shulman, G.I. The role of AMP-activated protein kinase in mitochondrial biogenesis. J. Physiol. 2006, 574, 33–39. [Google Scholar] [CrossRef] [PubMed]
  226. Jager, S.; Handschin, C.; St-Pierre, J.; Spiegelman, B.M. AMP-activated protein kinase (AMPK) action in skeletal muscle via direct phosphorylation of PGC-1α. Proc. Natl. Acad. Sci. USA 2007, 104, 12017–12022. [Google Scholar] [CrossRef] [Green Version]
  227. Chae, S.S.; Paik, J.H.; Allende, M.L.; Proia, R.L.; Hla, T. Regulation of limb development by the sphingosine 1-phosphate receptor S1p1/EDG-1 occurs via the hypoxia/VEGF axis. Dev. Biol. 2004, 268, 441–447. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  228. Lim, S.H.; Kim, C.; Aref, A.R.; Kamm, R.D.; Raghunath, M. Complementary effects of ciclopirox olamine, a prolyl hydroxylase inhibitor and sphingosine 1-phosphate on fibroblasts and endothelial cells in driving capillary sprouting. Integr. Biol. (Camb) 2013, 5, 1474–1484. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Role of S1P/S1PRs in macrophages. S1PRs on macrophages can be activated by extracellular molecules such as cytokines, chemokines, or growth factors. Receptor activation transmits a signal for sphingomyelinase (SMase) and ceramidase (CDase) to sequentially breakdown sphingomyelin and ceramide, respectively, to generate sphingosine. Phosphorylation (p) of the latter by Sphk1 produces S1P, which induces intracellular signaling to activate kinase-mediated phosphorylation of SphK1. Translocation of S1P from the cytosol to the cell membrane also leads to phosphorylation of sphingosine to S1P. SphK1 is essential for the TNF signaling pathway for downstream molecule NF-κB activation. Akt signaling molecules are activated by S1P1 and S1P4 and signal for macrophage activity and survival such as MCP-1 and MIP-1α. G protein through Rac1 and RhoA activation plays a role in S1P1 and S1P3 as the regulators of macrophage migration and infiltration. To the contrary, the migration signals are inhibited by S1P2, by which NLRP3 becomes a considerable signal to shade the S1P2 blockade mechanism to inhibit the macrophage death signal, caspase-11. Meanwhile, S1P5 signal activation regulates monocytes migration activity together with CCR2. TGF- β signaling pathway through molecules downstream of Smad3, Akt, and mTOR Smad3 mediates HIF-1α stabilization, and expression thus activates targeting genes containing HRE.
Figure 1. Role of S1P/S1PRs in macrophages. S1PRs on macrophages can be activated by extracellular molecules such as cytokines, chemokines, or growth factors. Receptor activation transmits a signal for sphingomyelinase (SMase) and ceramidase (CDase) to sequentially breakdown sphingomyelin and ceramide, respectively, to generate sphingosine. Phosphorylation (p) of the latter by Sphk1 produces S1P, which induces intracellular signaling to activate kinase-mediated phosphorylation of SphK1. Translocation of S1P from the cytosol to the cell membrane also leads to phosphorylation of sphingosine to S1P. SphK1 is essential for the TNF signaling pathway for downstream molecule NF-κB activation. Akt signaling molecules are activated by S1P1 and S1P4 and signal for macrophage activity and survival such as MCP-1 and MIP-1α. G protein through Rac1 and RhoA activation plays a role in S1P1 and S1P3 as the regulators of macrophage migration and infiltration. To the contrary, the migration signals are inhibited by S1P2, by which NLRP3 becomes a considerable signal to shade the S1P2 blockade mechanism to inhibit the macrophage death signal, caspase-11. Meanwhile, S1P5 signal activation regulates monocytes migration activity together with CCR2. TGF- β signaling pathway through molecules downstream of Smad3, Akt, and mTOR Smad3 mediates HIF-1α stabilization, and expression thus activates targeting genes containing HRE.
Ijms 22 08992 g001
Figure 2. Macrophage intracellular reprogramming during polarization. Balanced switching of M1/M2 macrophage action is crucial. Both the HIF-1α and HIF-2α subunits are essential to maintain the NF-κB level. However, due to the incongruent roles of HIF-2α, the part of the phenotypic switch between HIF-1α and HIF-2α in M1/M2 macrophages remains uncertain. M1 macrophage polarization is NF-κB-independent. NF-κB mediates the IkBζ recruitment to target promoter and acts as a transcriptional coactivator for chromatin remodeling that regulates the CCL2 (MCP-1) gene, thus producing cytokines such as TNF-α, IL-1β, IL-6, IL-12, and IFN-γ. M2 macrophages upon IL-4 stimulation activate STAT6, which then induces PGC-1β that is essential for the M2 macrophage profile and is characterized by IL-10 and TGF-β1 expression. Meanwhile, Jmjd-3, a histone 3 Lys27 (H3K27) demethylase that directly targets IRF-4, is crucial for M2 macrophages.
Figure 2. Macrophage intracellular reprogramming during polarization. Balanced switching of M1/M2 macrophage action is crucial. Both the HIF-1α and HIF-2α subunits are essential to maintain the NF-κB level. However, due to the incongruent roles of HIF-2α, the part of the phenotypic switch between HIF-1α and HIF-2α in M1/M2 macrophages remains uncertain. M1 macrophage polarization is NF-κB-independent. NF-κB mediates the IkBζ recruitment to target promoter and acts as a transcriptional coactivator for chromatin remodeling that regulates the CCL2 (MCP-1) gene, thus producing cytokines such as TNF-α, IL-1β, IL-6, IL-12, and IFN-γ. M2 macrophages upon IL-4 stimulation activate STAT6, which then induces PGC-1β that is essential for the M2 macrophage profile and is characterized by IL-10 and TGF-β1 expression. Meanwhile, Jmjd-3, a histone 3 Lys27 (H3K27) demethylase that directly targets IRF-4, is crucial for M2 macrophages.
Ijms 22 08992 g002
Figure 3. Role of metabolic reprogramming in oral wound healing under hypoxic conditions. Oral mucosal wounds heal faster than skin wounds, although having similar wound healing stages. The saliva is considered one factor that facilitates rapid mucosal wound closure by expressing secretory IgA. However, the essential cytokines, chemokines, growth factors, and protease inhibitors are interrelated and inseparable. Fewer M1/M2 macrophages and lower cytokine production (IL-6 and TGF-β1) during oral mucosal wound healing were observed. Lower VEGF expression found in the oral wound may be due to a decreased level of HIF-1α gene expression level, while higher expression levels of SOX2 and PITX1 followed by the increased level of α-SMA expression lead to acceleration and less scar tissue formation in oral wounds than in skin wounds. Wound S1P/HIF gradient cause S1P/HIF to act as pro- or anti-inflammatory agents for inflammatory cells such as M1/M2 macrophages.
Figure 3. Role of metabolic reprogramming in oral wound healing under hypoxic conditions. Oral mucosal wounds heal faster than skin wounds, although having similar wound healing stages. The saliva is considered one factor that facilitates rapid mucosal wound closure by expressing secretory IgA. However, the essential cytokines, chemokines, growth factors, and protease inhibitors are interrelated and inseparable. Fewer M1/M2 macrophages and lower cytokine production (IL-6 and TGF-β1) during oral mucosal wound healing were observed. Lower VEGF expression found in the oral wound may be due to a decreased level of HIF-1α gene expression level, while higher expression levels of SOX2 and PITX1 followed by the increased level of α-SMA expression lead to acceleration and less scar tissue formation in oral wounds than in skin wounds. Wound S1P/HIF gradient cause S1P/HIF to act as pro- or anti-inflammatory agents for inflammatory cells such as M1/M2 macrophages.
Ijms 22 08992 g003
Table 1. S1P/S1PRs signaling and monocyte/macrophage function under inflammatory and pathological conditions.
Table 1. S1P/S1PRs signaling and monocyte/macrophage function under inflammatory and pathological conditions.
Inflammatory ConditionsSignaling
Pathways
S1P ReceptorsRoles
Palatal wound healing [57]MCP-1, HIF-1α, MIP-1α, iNOS, TNF-α, Akt, p38S1P1Infiltration of M1/M2 macrophages
CNS autoimmunity and neuroinflammation [41]STAT-3, JAK, IL-6S1P1Pronounced activation of monocytes
Inflammation and atherosclerosis [21,24,25,31,32,33]TNF-α, MCP-1, IL-6, Akt, PI3K, PKC, ERK1/2, p38, Rho kinasesS1P1Activation and migration of macrophages
12/13, NF-κB, RhoAS1P2Inhibits macrophage migration (negative effect) and inhibits M1 activation
MCP-1, TNF-α, Sphk1S1P3Migration of monocytes/macrophages
Tubulointerstitial inflammation [36]MCP-1, SphK1, TNF-α, arginase-1, IL-6, IL-10S1P1
S1P3
Macrophage infiltration
Acute allergic [37]MCP-1/CCL2, MIP-1α, RANTES/CCL5S1P2Macrophage infiltration
Inflammation and liver injury [29,30,179]i/o, PI3K, JNK, Rac1S1P2
S1P3
Migration of BMMs,
M1 polarization
NLRP3, IL-1β, IL-18, p38, ERK, JNK, Gα12/13S1P2Blockade BMMs activation and M1 polarization
Rheumatoid arthritis [23,175]NF-κB, Fas, Akt, p38, ERK1/2, Rac, RhoS1P1Motility and number of BMMs induced osteoclast
Cerebral ischemia [49]NF-κB p65, ERK1/2, p38, AktS1P3M1 polarization
Bacterial sepsis [50,173,177]Caspase-11S1P2Macrophage pyroptosis
MCP-1, Sphk1, TNF-α, arginase-1, IL-6, IL-10S1P1
S1P3
Macrophage infiltration
Psoriasis [54]IL-6, CCL2, CXCL1S1P4M2 macrophage infiltration
Bone marrow sinusoidal inflammation [55]CCR2S1P5Monocyte migration
Pulmonary disease [27,28]CCR2S1P5Monocyte migration
Encephalomyelitis [25]CCR2S1P5Inflammatory monocytes supply the concentration of lymph node S1P
Several inflammatory conditions related to monocyte/macrophage disturbances (wound healing, allergies, liver injury, and arterial/pulmonary diseases) are a hallmark feature of hypoxia and are detected by S1P/S1PR signaling. These disturbances are related to various cellular signaling pathways that mediate cell survival, migration, and apoptosis. S1P1–3 contribute to the activation, motility, and infiltration of monocytes/macrophages. Meanwhile, S1P4–5 only contribute to the migration and infiltration of monocytes/macrophages. However, supporting research remains to be further analyzed. Thus, activation of S1P/S1PRs in monocytes/macrophages through various pathways can trigger functional responses as indicated. Targeting of the genes indicated may therefore be of interest in disease settings.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Hutami, I.R.; Izawa, T.; Khurel-Ochir, T.; Sakamaki, T.; Iwasa, A.; Tanaka, E. Macrophage Motility in Wound Healing Is Regulated by HIF-1α via S1P Signaling. Int. J. Mol. Sci. 2021, 22, 8992. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22168992

AMA Style

Hutami IR, Izawa T, Khurel-Ochir T, Sakamaki T, Iwasa A, Tanaka E. Macrophage Motility in Wound Healing Is Regulated by HIF-1α via S1P Signaling. International Journal of Molecular Sciences. 2021; 22(16):8992. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22168992

Chicago/Turabian Style

Hutami, Islamy Rahma, Takashi Izawa, Tsendsuren Khurel-Ochir, Takuma Sakamaki, Akihiko Iwasa, and Eiji Tanaka. 2021. "Macrophage Motility in Wound Healing Is Regulated by HIF-1α via S1P Signaling" International Journal of Molecular Sciences 22, no. 16: 8992. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22168992

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