Next Article in Journal
Nucleic Acid Aptamers Emerging as Modulators of G-Protein-Coupled Receptors: Challenge to Difficult Cell Surface Proteins
Next Article in Special Issue
The Fatal Circle of NETs and NET-Associated DAMPs Contributing to Organ Dysfunction
Previous Article in Journal
Circulating miRNA Fingerprint and Endothelial Function in Myocardial Infarction: Comparison at Acute Event and One-Year Follow-Up
Previous Article in Special Issue
Analysis of Leukocyte Recruitment in Continuous Veno-Venous Hemofiltration with Regional Citrate vs. Systemic Heparin Anticoagulation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Immune Cell Plasticity in Inflammation: Insights into Description and Regulation of Immune Cell Phenotypes

Centre for Biochemical Pharmacology, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London EC1 6MQ, UK
*
Author to whom correspondence should be addressed.
Cells 2022, 11(11), 1824; https://doi.org/10.3390/cells11111824
Submission received: 12 May 2022 / Revised: 28 May 2022 / Accepted: 30 May 2022 / Published: 2 June 2022

Abstract

:
Inflammation is a life-saving immune reaction occurring in response to invading pathogens. Nonetheless, inflammation can also occur in an uncontrolled, unrestricted manner, leading to chronic disease and organ damage. Mechanisms triggering an inflammatory response, hindering such a response, or leading to its resolution are well-studied but so far insufficiently elucidated with regard to precise therapeutic interventions. Notably, as an immune reaction evolves, requirements and environments for immune cells change, and thus cellular phenotypes adapt and shift, leading to the appearance of distinct cellular subpopulations with new functional features. In this article, we aim to highlight properties of, and overarching regulatory factors involved in, the occurrence of immune cell phenotypes with a special focus on neutrophils, macrophages and platelets. Additionally, we point out implications for both diagnostics and therapeutics in inflammation research.

1. Introduction

Immune cells are key components of the natural defense of the human body against outside invaders, such as bacteria. The most prominent role for their involvement in pathogen defense is played by leukocytes. Even though leukocytes are crucially needed for killing and eliminating bacterial particles, excess activation of these cells can be associated with self-inflicted tissue damage and organ failure. Such overshooting immune activation is visible in sepsis patients and is associated with high mortality rates, even nowadays.
Pro-inflammatory properties of leukocytes are well-documented, whereas more recent work highlights a much more diverse immune repertoire of these cells, including: anti-inflammatory properties during models of chronic disease, as well as pro-resolution responses of cellular subsets, which need to be critically considered when assessing the impact of immune cells within the human body. Even though certain functions have been ascribed to cell populations over time (for example, efferocytosis in the case of macrophages), a cell can adapt to its requirements and its surroundings. Inflammation involves variations in plasma and organ concentrations of danger-associated molecular patterns (DAMPs) or pathogen-associated molecular patterns (PAMPs), summarized here as inflammation-associated molecular patterns (IAMPs), anti-inflammatory mediators and resolving-type mediators. All these mediators can differentially impact the cell directly and/or indirectly through the tissue-specific environment (organ). Additionally, specific peaks of plasma levels can favor production and/or mobilization of cellular subsets. However, what determines the pro- or anti-inflammatory phenotype of an immune cell? Which pathways or organ-specific environmental cues lead to cellular reprogramming or maturation? Additionally, how can this knowledge be used for diagnostic or therapeutic purposes? Before discussing underlying mechanisms, it is first important to examine how cell phenotypes can be defined and distinguished.

2. Techniques for Immune Cell Phenotype Assessment: A Matter of Perspective

The description of cellular properties ranges back to the very early days of microscopy, in which simple features such as color were used to discriminate a “white” or a “red” blood cell [1,2]. In recent years, research methods have become more advanced and technical, and with this, research questions have evolved to become more precise and targeted as various cellular subpopulations and phenotypic features have been exposed. Commonly used approaches for phenotypic assessment include flow cytometry and microscopy protocols, whereas lately transcriptomic techniques have also been used for identification of scarce, phenotypically distinct subclusters, even though clear international standardization measures are still missing in many areas of cellular phenotyping (Table 1).
Immune cells are frequently analyzed by flow cytometry and immunocytochemistry to define their surface characteristics [3]. This allows for the assessment of functionally relevant molecules capable of interacting with their surroundings. Furthermore, it can be used as a readout for regulative processes such as receptor internalization and shedding. Proteomics approaches are of equal importance when assessing the protein composition of a cell and showcase, for example, differences in signaling components in sterile inflammation [3], or adhesion proteins in leukocytes during atherosclerosis [4]. Aside from proteins, the lipid mediator content can also be assessed, and this mass-spectrometry-based technique was recently used to identify activation-dependent and -independent differences in lipid patterns of leukocyte subsets [5]. Many phenotypic descriptions are based on the notion of a large, overall circulating or compartment-based cellular population instead of focusing on each single cell. Recent advances in single-cell techniques yielded novel insights into cellular population and cluster-based research. Transcriptomics can be applied to study overall regulatory processes affecting gene expression on a single cell or cellular population level. Such techniques are useful in, for example, identification of spatially restricted, distinct immune cell responses, such as within the cerebrospinal fluid of multiple sclerosis patients [6]. The combination of such ‘omics’ approaches and large datasets were recently used to highlight the impact of transcription and translation within sepsis patients [7].
Assessment of physical properties can assist in identifying features which could not be revealed by the aforementioned techniques. As such, microfluidic observations together with atomic force microscopy showed that softening of leukocytes controls their dexamethasone- and catecholamine-driven demargination [8], while advanced high-throughput mechanophenotyping (termed “deformability cytometry”) was applied to differentiate active/non-active leukocytes in pleural effusions [9]. Functional assays, such as ligand-binding assays, ROS production, or migratory assays, are widely used to determine distinct responses of isolated cells to a pre-defined stimulus, which assists in identification of an activated or non-activatable cellular phenotype [10]. Most impressively, recent work by Andrés Hidalgo and colleagues elegantly showed that the integration of various data points from 4D in vivo experiments into a behavioral landscape profile allows for a much more holistic perspective on cellular phenotypes than a single targeted technique [11]. Therefore, cellular phenotype assessment depends on the context and required output, as well as the level of detail.

3. Mechanisms of Immune Cell Phenotype Switching: Outside In and Inside Out

Immune cell phenotype switches can be attributed to various processes, which operate in an interconnected manner (Figure 1). Generally, cellular characteristic changes will affect not only the surface composition, but equally the intracellular protein content and location, ultimately impacting the overall functionality of the immune cell.

3.1. Surface: Receptor Shedding, Internalization and Desensitization

Cell-surface receptor levels vary widely during inflammation based on various underlying mechanisms, including shedding and internalization. Receptor shedding requires sheddases, such as ADAMs, a class of ectodomain-shedding metalloproteinases [18]. A prominent example of receptor shedding is represented by L-selectin, which can be shed from the leukocyte surface in an iRhom2/ADAM17-dependent manner, impacting neutrophil effector functions and thus bacterial defense in vivo [19].
Receptor internalization equally occurs during inflammation/activation, impacting surface receptor levels. This process relies on endocytic mechanisms, generally involving caveolae- and/or clathrin-mediated endocytosis and redistribution mechanics [20,21]. As examples, CXCR1, 2 and 4 and GPIb have been described to undergo receptor internalization, modifying the interaction and activation potential of leukocytes and platelets, respectively, although significance for cellular functionality is context- and ligand-dependent [22,23,24]. Such events, making a receptor unavailable to its ligand or restricting signal-transduction, are also visible in the resolution-associated receptor FPR2. FPR2, like FPR1, is known to undergo desensitization, mediating excessive receptor-dependent responses, which then can be counteracted by reactivation by alternative signaling pathways, for example, by the platelet-activating-factor, allowing the cell to regain certain functionalities [25,26].

3.2. Regulation of Gene Expression

Inflammatory signals directly affect gene expression within immune cells. Assessment of neutrophils exposed to various stimuli identified 243 alterations in gene expression, occurring rapidly following exposure [27]. Such changes in gene expression are also detectable in leukocytes in various inflammatory diseases, such as amyotrophic lateral sclerosis, multiple sclerosis, infection, allergic reactions and rheumatoid arthritis [6,28,29,30,31]. Gene regulatory network analysis additionally highlights the effects of plasmatic sepsis-related changes on immune cell responses, affecting “hub” genes, such as STAT1, SOCS1 and ADORA3 [32]. Additionally, the (long) non-coding RNA of leukocytes is affected by endotoxemia, including GABPB-1AS1, THAP9-AS1 and SCARNA9 [33]. This showcases the sensitive regulatory interplay resulting in reprogramming and up-/downregulation of specific factors involved in immune cell functionality.

3.3. Activation-Dependent Vesicle Release and Membrane Fusion

Immune cells contain cargo, which can be mobilized, integrated into the membrane and/or released. An important example of exocytosis-mediated phenotype changes is the mobilization of P-selectin to the surface of platelets, which, amongst others, is affected by Ral GTPases and in a VAMP-7-dependent manner is linked to actin reorganization, subsequently resulting in platelet–leukocyte aggregate formation [34,35,36]. Additionally, in leukocytes, SNARE/NSF-dependent vesicle release will lead to reprogramming of a cell both at the surface and an intracellular level [37,38].

3.4. Re-Production and Re-Distribution

Sepsis evokes the extravasation of leukocytes and consumption of platelets [39,40]. Such events demand replenishing and mobilization of cellular pools to outweigh cellular deficiencies within the circulation [40]. HSCs express Toll-like receptors, which trigger myeloid differentiation and aid in replenishment of innate immune cells during infection [41]. Overall, inflammation is associated with changes in cytokine and chemokine profiles, differentially affecting myelopoiesis and cellular mobilization. As an example, IL-1 and IL-6, both of which are elevated during inflammation, are key triggers of hematopoietic stem cell proliferation and myelopoiesis [42]. Myelopoiesis and HSPC quiescence are additionally impacted by other factors, such as CXCL12 and leptin, which can be targeted by physical activity [43]. Such physical activity furthermore impacts emergency myelopoiesis, and absence or limitations of physical activity must therefore be considered in specific patient subsets, such as intensive care patients [43]. Notably, emergency myelopoiesis is regulated additionally by IL-3, affecting survival during sepsis [44], and has been linked to occurrence of neutrophil precursor cells in the circulation, visible, for example, in severe-COVID-19 patients [45]. Single-cell profiling furthermore identified shifting of progenitors to neutrophil lineage at the expense of monocytes during early sepsis [46].
In addition to novel production, organ resident cellular pools are mobilized during inflammation, such as the marginated pool of neutrophils, and also organ-specific (and possibly residing) leukocyte populations with distinct characteristics, including CD11bhiCD62LloCXCR4+ neutrophils in the lung, are observed [47,48,49]. Not only leukocytes but also platelet phenotypes are affected by new production; recent work has identified a spleen-based platelet production pool which allows for the appearance of a phenotypically distinct CD40Lhi platelet population in sepsis [50]. This is of note, since this CD40L phenotype is associated with outcomes in pulmonary infection [51].

3.5. Changed Physical Properties

Changes regarding immune cell phenotypes can also occur with regard to physical properties. Generally, immune cells are migratory cells, which can change their shape and physical characteristics. Notably, in response to different stimuli, including glucocorticoids and catecholamines, leukocytes exhibit actin reorganization and a reduction in overall stiffness. This softening itself is sufficient to impact cell demargination [8]. Therefore, it is plausible and likely that both “soft” and “stiff” leukocytes co-exist within different regions of the body, representing different physical phenotypes due to their activation status and actin reorganization processes. Importantly, various pathways have been implicated in the organization and re-organization of the actin cytoskeleton, impacting circulating leukocyte numbers as well as infiltrating leukocyte subsets [52,53]. Another physical–biochemical occurrence is the redistribution of lipid raft signaling platforms [54]. Lipid rafts aid in the directed partitioning of proteins into regions of signal intensity and control clathrin-independent recycling of integrins, such as LFA-1 during chemotaxis [55]. Notably, each leukocyte class can be distinguished by specific fatty acid profiles, likely influencing lipid rafts, whereas it is unclear how these processes vary amongst distinct subclass phenotypes within immune cell populations [56]. A lipidomic study indeed revealed that fatty acid processing and lipid composition differs amongst macrophage subtypes (M1 vs. M2) with a quantified increase in exogenous fatty acid channeling into sphingolipids in M2 macrophages [57]. Thus, taking into account the various activation states and motions an immune cell undergoes, an axis-polarized phenotypic distinction (stage of recruitment, uropodia/lamellipodia formation) can emerge. Another apparent physical phenotypic distinction can be made by size. Indeed, a population of large, immature platelets has been reported to be prominently detectable in inflammatory settings of emergency cellular replenishment [58].

3.6. Integrin-Mediated Signaling

Integrins are bi-directional transmembrane signaling receptors of immune cells, capable of changing their activation state upon stimulation (inside-out signaling) and transmitting signals following ligand-binding (outside-in signaling) [49]. Aside from the formerly described bent-closed, extended-closed and extended-open conformations, a bent-open configuration of β2-integrins has been observed to hinder leukocyte adhesion by cis-binding of ICAM-1 [59,60]. Judging by the integrin activation states, immune cells can therefore exist in up to four different phenotypically distinct states, although these activation states appear to be of a transitional nature and to exist in parallel on leukocyte surfaces [59]. Regarding the effects of outside-in signaling on cellular phenotypes, in the leukocyte, ligand binding to an integrin can evoke a response related to effector functions, leading to a Syk-kindlin-3-dependent switch towards a ROS-productive, pro-migratory, phagocytic phenotype [61,62,63], nicely summarized elsewhere [64]. Additionally, upon phagocytic priming, macrophages and neutrophils can acquire a phenotype of pro-phagocytic responses, aiding in facilitated bacterial clearance [65]. Speaking of cellular uptake events, another peculiarity of immune cells must be taken into consideration when interpreting appearance of phenotypically distinct cellular subclasses: the uptake of foreign cellular material. Such integration of foreign material can be detrimental; for example, membrane raft incorporation of mycobacterium tuberculosis virulence factor lipoarabinomannan leads to a macrophage phenotype of impaired phagocytic capacity [66]. However, it can also promote a pro-immunogenic phenotypic priming of cells. Thus, megakaryocytes, which at first sight in the past were not considered bona fide immune cells, though our perception has changed in recent years [67], are capable of integrating neutrophil membrane fragments into newly produced platelets, thereby altering their surface composition and interaction potential [68].

4. Principles and Regulatory Factors Affecting Immune Phenotypes: The Environment Shapes the Cell, and the Cell Impacts Its Environment

Phenotypic reprogramming of immune cells occurs due to various generalizable principles, which affect, in different ways, distribution, production and activation mechanisms within the body (Figure 2). All these regulatory processes occur simultaneously to a varying degree, leading to temporary dominance of distinct governing factors and phenotypic landscape composition.

4.1. Circadian Rhythms

The time of day leads to variations in leukocyte populations both in numbers and phenotypes with direct consequences for combatting invading pathogens [69]. Thus, the myeloid-specific brain and muscle Arntl-like protein 1 (Bmal1)-dependent control of inflammatory monocyte circadian rhythmicity is known to impact immune defense [70]. Tissue-specific circadian modulations of adhesion molecules and chemokines [71], together with cell autonomous oscillations, further promote organ selectivity and circadian rhythmicity of recruitment of distinct leukocyte subsets [72,73,74,75]. Additionally, plasmatic concentrations of specialized pro-resolving mediators, such as RvDn3 DPA, are diurnally regulated, thereby affecting both neutrophil and platelet activation [76].

4.2. Aging

The host, both as a whole, and each cell will mature and age throughout their lifespan, implicating differential cell-intrinsic and -extrinsic phenotypic modulators. Similar to extensive microenvironmental changes occurring in the developing organism, in which leukocytes with an impaired functional phenotype are observable [77], host ageing is equally associated with overall changes in the inflammatory milieu. Notably, increased basal levels of circulating pro-inflammatory mediators, reduced cellular regeneration potential due to double-stranded DNA breaks, as well as differentially modulated inflammatory responses of immune cells, including reduced neutrophil chemotaxis and peroxide production and macrophage cytokine production, can be observed during ageing [78,79]. Additionally, organ-specific phenotypic reprogramming occurs during ageing, implicating interaction of reverse-transmigrated neutrophils within the lung, which primes these cells for an activated phenotype responsible for the induction of remote organ damage [80]. Of note, ageing is also associated with disturbances in circadian rhythms, leading to imbalances in immune cell responses [81], and likewise, disturbed circadian rhythmicity due to genetic targeting of BMAL1 leads to premature ageing [82]. As for the cell-intrinsic maturation and ageing, aged neutrophils feature high CXCR4 expression, which mediates their clearance within the bone marrow [40,49]. In contrast to the notion of an aged cell as an exhausted “retired” cell, these are actually well-educated, “trained” and easily activatable cells, aiding in the immediate immune defense response [83,84]. The occurrence of these CXCR4hi-aged-neutrophils is regulated by microbiota-TLR-MyD88 signaling [83]. Nonetheless, as with most overtly active cellular phenotypes, a downside must also be considered, since persistence of these aged intravascular neutrophils is associated with thrombo-inflammation and reduced survival following myocardial infarction [85].

4.3. Stress

Acute and chronic stress have divergent effects on immune cell functionality; chronic stress is regarded to mainly evoke an immunosuppressive response, whereas acute stress can lead to immunostimulation [86,87,88]. Animal experiments using application of restrainer-mediated psychological stress thus led to increased leukocyte recruitment into a site of injury, showcasing modulated systemic factors and impacting phenotypic activation-dependent immune cell responses [89]. Similarly, changes in leukocyte subsets can be detected in humans when exposed to stress [90]. These responses are not surprising, in view of the hormonal changes occurring in response to stressors, including catecholamine release and glucocorticoids, and the variety of counter receptors expressed by immune cells. Nonetheless, immunologic responses following catecholamine stimulation are much more complex, indicating the dose and context dependency of leukocyte subsets, as discussed by Scheiermann et al. and others [91,92].

4.4. Environment/Compartment (Organ)

Each tissue/organ within the body features proprietary interaction and signaling modalities [49]. Therefore, the lung primes a noxious, overtly active reverse-transmigrated neutrophil phenotype, subsequently leading to secondary organ injury [80]. Similar observations of detectability and phenotypically distinct priming of reverse-transmigrated neutrophils with delayed apoptosis have been made, for example, in chronic inflammatory arthritis patients, highlighting either a potential reverse migration of neutrophils from the joint back into the blood stream or the return of neutrophils from other secondary injured organs to the circulation [93]. Overall, inflammatory-site-invading neutrophils take on modified transcriptional activities, involving neutrophil polarization and changing their phenotype in a site- and stimulus-specific manner [94]. As an example, in resolving inflammation settings, migrated neutrophils upregulate the pro-resolving protein annexin A1, and this could be part of resolution circuits in experimental peritonitis [95], sepsis [96] and colitis [97]. This is of special interest when taking into consideration that formation of membrane blebs, as has been described in neutrophils during apoptosis and compression-microenvironment-dependent membrane protrusion [98,99], is affected by Annexin A1 itself, regulating cell lysis and potentially membrane repair mechanisms [100]. Additionally, it is emerging that aside from traditional organ-resident cell populations, an inflammation-mediated imprinting of resident macrophages via modulated niche mechanisms leads to prolonged phenotypic re-composition of immune cell landscapes within organs [101].

4.5. Mechanical Forces

Immune cells exist not only within tissue compartments but also within the circulation. Circulating immune cells are constantly impacted by shear forces, leading to deformation and cell–cell collisions. Thus, taking into consideration margination and deformation capacities of immune cells, various force-adapted cellular phenotypes co-exist within the circulation in parallel. According to Simon et al.’s considerations of the modified Smoluchowski’s two-body collision theory, collisions and aggregate formation of neutrophils do occur in a shear-dependent manner, emphasizing that different immune cell phenotypic characteristics can be found within varying vascular beds [102]. Shear additionally impacts the biomechanical re-composition of leukocytes. Herein, pseudopod formation and leukocyte spreading occurs in a potassium- and calcium-dependent, shear-controlled manner with reduced shear favoring pseudopod projection [103]. Nonetheless, migration and podosome formation varies according to leukocyte and macrophage subsets [104]. Interestingly, leukocytes themselves also show a force-dependent migratory plasticity, in that they hibernate between adhesion-receptor-mediated responses of force transmission and receptor-independent nuclear-positioning-facilitated locomotion, as suggested by Renkawitz and Sixt [105,106].
Importantly, inflammation can lead to redistribution characteristics due to changed blood flow properties, involving vasodilation/vasoconstriction and endothelial damage, impacting the site- and subset-specific trafficking of immune cells in an activation- and shear-dependent manner [107,108]. Leukocytes are recruited in a simplified cascade-like manner, involving rolling, adhesion and extravasation. During rolling, sling formation and rupturing of slings can occur, which can conclusively lead to leukocyte subsets lacking certain membrane elements/particles [109]. Likewise, similar to the changed flow properties during sepsis, the introduction of artificial surfaces, for example, during renal replacement therapy or due to assist devices, also poses an environment for differential targeting and activation of leukocytes. Neutrophil activation and microparticle formation thus can be commonly observed in cardiopulmonary bypass and ventricular assist devices, as shear-dependent mechanical activation and phenotype switching appear to occur in a guanine-nucleotide-exchange-factor-dependent manner [110].

4.6. Inflammation and Injury—A Combination of Extremes

An inflammatory reaction in response to an injury or infection leads to disturbance and modification of several of these generalizable processes. Increased release of stress signals (catecholamines and glucocorticoids), together with disruption of circadian rhythms as well as cell consumption, are only part of the overarching modulatory environment during inflammation. Most prominently, increased levels of cytokines, chemokines, PAMPs and DAMPs lead to a complete re-composition of the immunologic landscape within the body [111].

5. Macrophage Phenotypes and Plasticity

Macrophages can be categorized by their localization (pulmonary, liver, interstitial, alveolar), function (pro-inflammatory, pro-resolution), characteristics (expression, cytokine release) or ontogeny (resident vs. monocyte-derived recruited). Each categorial variable is differentially impacted by inflammation, as, for example, pulmonary inflammation leads to changes in overall tissue resident monocyte/macrophage numbers and phenotypic predominance [112].

5.1. M1, M2 and Beyond

One of the most commonly used phenotypic distinctions involves the categorial description of M1 and M2 macrophages, implying a pro-inflammatory or anti-inflammatory/pro-resolution phenotype (Table 2) [113]. As described in the nomenclature perspective paper by Murray et al., these populations feature distinct activation patterns and can be obtained by differential activation in vitro, implicating STAT1 and STAT6, respectively [114]. Classically activated M1 macrophages are described as phagocytic, proinflammatory cytokine-releasing (IL-1, IL-6, TNF) macrophages, which can be obtained in vitro by LPS and interferon stimulation [114,115,116]. The phenotypic shifting towards M1 involves key pro-inflammatory triggers, such as TLR4, MyD88, IKK, NF-kB and IRF5, as well as JNK [117,118,119]. Notably, shifting of macrophages to an M1 phenotype favors a snowball response in that the release of proinflammatory mediators encourages the polarization of additional macrophages towards an M1 profile [120]. Alternatively activated M2 macrophages, on the other hand, can be obtained by IL-4, IL-10 and/or TGFβ stimulation and showcase subclass-specific production of IL-10, CCL22 and CCL24 [121]. Further subset categorization is used as M2a (wound-healing, CD206hi), M2b (regulatory, CD86hi), M2c (acquired deactivation, MerTKhi) and M2d (tumor-associated, VEGFhi) [114,122]. In septic baboons, a purely M1 phenotypic profile of circulating monocytes, and speculatively also macrophages, were associated with reduced survival compared to a mixed M1/M2 phenotypic profile [123]. It appears reasonable that during an acute infection an inflammatory response is required in order to clear invading pathogens; subsequently, such inflammation needs to be resolved and cleared in order to ensure organ homeostasis [124]. Aside from signaling mediators such as PI3K/Akt being involved in balancing M1/M2 profiles, NOTCH signaling (favoring M1 macrophage plasticity) and KLF4 signaling (increased in M2 macrophages) have also been reported to impact macrophage plasticity and their pro-/anti-inflammatory responses [125,126]. An M1 macrophage profile is also present during acute kidney injury and can be targeted by modulation of heparin-binding protein expression or the targeting of CLEC4e/Mincle-Syk selectively on macrophages [127,128]. Although locally, such an M1 profile is needed to combat invading pathogens, a systemic overactive M1-polarized macrophage profile is associated with deteriorated sepsis outcomes [129], whereas an M2 profile is required for resolution dynamics following an acute insult. Nonetheless, preponderance of M2-like macrophage phenotypes could hold risks of immunosuppression and thus of secondary infection. Such a post-sepsis M2-like phenotype, implicating immunosuppression, is regulated via p21, affecting p65-p50 and p50-p50-NFkB pathways within the macrophages [130]. Indeed, an incomplete M2-type conversion was hypothesized to be involved in post-sepsis immunosuppression, although this study leaves various questions still unanswered [131]. Macrophage phenotypes are not only modulated by underlying conditions or self-sustaining pro-inflammatory feedback loops; cellular interplay controls modulation of macrophage plasticity. Indeed, recent work highlights that during pulmonary infection, platelet–Treg interplay differentially primes the M2-like resolution responses in alveolar macrophages in post-pneumonic mice [51]. Nonetheless, these rather basal phenotypic attributions of M1/M2 profiles insufficiently describe the whole magnitude of cellular complexity present within the organism, since in vivo studies highlight presence of intermediate phenotypes and much more complex signaling environments [132]. Phenotypic switching of macrophages is not only impacted by pro-inflammatory mediators, but also by specialized pro-resolving mediators (SPMs). These mediators, such as Resolvin D2, are capable of controlling sepsis severity and protectin D1/neuroprotectin D1 offers protection in an influenza virus model, showing the diversity of these modulators [133,134,135]. Notably, the resolution-implicated GPCR GPR37, expressed on macrophages, controls neuroprotectin D1-mediated phagocytosis and protection against sepsis [136,137]. Thus, macrophage phenotypic changes are not only impacted by the pro-inflammatory environment, but equally by the presence of pro-resolution factors, aiding in the containment of infection and overshooting immune responses.

5.2. Tissue-Specific Macrophages

Aside from a purely functional connotation, localization and origin must also be taken into consideration when interpreting the magnitude of macrophage phenotypes during inflammation. Indeed, tissue origin and ontogenic programming of macrophages seem to partially outweigh the stimulatory responses during inflammation. More precisely, in a co-infection model of Heligmosomoides polygyrus bakeri and Salmonella enterica, F4/80hi-resident macrophages are primed for recruitment of neutrophils by release of neutrophil-attracting and -activating chemokines, such as CXCL1 and CXCL2, compared to F4/80lo-recruited macrophages [138].
Generally, organ-residing macrophages can be described based on their compartmentalization or cell surface markers. In the lung, the basic distinction involves alveolar- and tissue-resident macrophages, characterized by absence/lower expression of CD11b, presence of CD11c, absence of CD86 and presence of SiglecF on the surface of alveolar macrophages compared to interstitial macrophages [139]. scRNAseq techniques have aided in assessing more diverse subtypes of tissue-specific macrophages. Herein, a pulmonary SiglecF-positive (SiglecF+ CX3CR1+ MHCIIhi) interstitial macrophage was identified, causing pro-fibrotic changes within the lung in a bleomycin model and delineating an intermediate phenotype in-between classically referenced interstitial and alveolar macrophages [140]. In the spleen, marginal metallophilic (Siglec1+, CD169+), marginal zone (SIGNR1+ MARCO+) and red pulp (F4/80hiCD68+ CD11blo) macrophages can be distinguished [141]. Taking advantage of their spatial proximity, splenic CD169+ metallophilic macrophages via Usp18 aid in the activation of antiviral T- and B-cell responses during viral infection, thus bridging innate and adaptive immune responses [141,142,143]. Moreover, the liver is a major site of macrophage residency and houses Kupffer cells as well as monocyte-derived macrophages [144]. Once more, subset specific functional attributes can be found, as, for example, MerTK+ HLA-DRhi macrophages control the resolution of inflammation during acute liver failure via induction of neutrophil apoptosis and clearance [145]. Again, application of scRNAseq techniques aids in further characterization of macrophage subsets, as in human livers two subsets of CD68+ macrophages (set 1: Lyz+ CSTA+ CD74+: inflammatory macrophages; subset 2: CD5L+ MARCO+ VSIG4+ C5AR1+ KLF4+ and others: tolerogenic macrophages) were identified [146]. Similarly, in the heart, macrophages with varying functionalities have been described. Notably, IL-10-producing macrophages have been implicated in diastolic dysfunction, as connexin 43-expressing macrophages contribute to electrical conduction within the myocardium [147,148].

5.3. Overarching Macrophage Phenotypes

Aside from organ specific phenotypic attribution, distinct subsets can be detected across various organs. Using scRNAseq analyses, dichotomic LYVE1hi or MHCII+ macrophage subsets were previously defined [149]. Another study concluded that this characterization is insufficiently detailed, again using scRNAseq of CD45+ CD64+ macrophages within the heart, liver, lung, kidney and brain, and thereby revealing the presence of three distinct populations: (i) Timd4+ Lyve1+ Folr2+, (ii) Ccr2+ Cd52+ S100+ MHCIIhi and (iii) Trem2+ Apoe+ Cd14+ MHCIIhi macrophages. These macrophage subtypes, even though they show similar expression patterns across organs, could be linked to differential functions within each specific organ [150]. It is thus likely that not only expression by itself, but also imprinting via tissue microenvironments, contributes to immune cell phenotypes and function. Indeed, it has recently been suggested that imprinting of macrophages by host tissues might limit plasticity, especially under inflammatory conditions [101]. Impressively, macrophages also respond to remote damage, partially in a tissue-specific fashion. Herein, both macrophage numbers and macrophage responsiveness phenotypes in remote organs are affected differentially by, for example, myocardial infarction, CLP or stroke. This NK-cell-IFNγ-dependent response leads to priming of alveolar macrophages for combatting of bacterial pneumonia [151]. Although ontogeny of tissue-resident macrophages is known to be of yolk sac origin, and organ-detectable macrophage populations contain both pre-populated resident as well as recruited monocyte-derived macrophages, the macrophage landscape is even more complex [152,153,154]. For example, central nervous system macrophages consist of, on the one hand, prenatal progenitors (deriving into meningeal macrophages and microglia), whereas perivascular macrophages derive perinatally from meningeal macrophages dependent on arterial vascular smooth muscle cells and postnatally depend on Irf8 and Mafb, showcasing that tissue-specific niches are crucial for development and maintenance of macrophage subpopulations [155].

6. Neutrophil Phenotypes—The Many Faces of a Single Cell

Similar to macrophages, various neutrophil phenotypes have also been described (Table 2). Mechanisms for phenotype changes include activation/deactivation, (reverse) transmigration, reproduction and uptake of foreign (bacterial) material. Neutrophils can be distinguished by various properties, including surface receptor composition, gene expression and density gradient centrifugation characteristics [156]. A common categorization of neutrophil phenotypes similar to the categorization of macrophage subtypes into N0, N1 and N2 subclasses is performed, which has been extensively summarized already [157,158]. Similar to M2 macrophages, CD206 expression is a key feature of N2 neutrophils. Whereas N1 neutrophils have been primarily regarded as anti-tumorigenic and N2 as pro-tumorigenic [159], neutrophil populations appear to be a bit more complex when taking into consideration all phenotypic aspects. Interestingly, CD206+ N2 neutrophils also come into play, for example, in inflammatory ischemic damage, such as stroke. Herein, N2 polarization, in a PPARγ-dependent manner, increases neutrophil clearance and resolution of post-stroke inflammation [160]. Additionally, such temporal polarization of neutrophils could also be detected in the heart, following myocardial infarction [161]. Along this line, neutrophil-derived microvesicles indeed display AnxA1-dependent protective characteristics in, for example, the inflamed joint in experimental inflammatory arthritis [162].
Density gradient characterization of neutrophils includes normal-density neutrophils (NDNs), consisting of terminally differentiated and immature neutrophils, and low-density neutrophils (LDNs), which are quite heterogenous, including both pro-inflammatory and immunosuppressive populations and containing myelocytes and metamyelocytes as well as both CD16+ CD11b+ mature and CD11blo CD16lo immature neutrophil precursors, although functional distinctions of NDNs and LDNs are being debated [163,164]. Interestingly, LDNs showing CD16int expression are correlated with disease severity, for example, in COVID-19, showing either a potentially functional role or the regulatory mechanisms impacting neutrophil re-production and activation [165]. Characteristically, pathologic challenges impact the bone marrow and splenic neutrophil composition in that C/EBPe-dependent proliferative neutrophil precursors expand [166]. Indeed, in sepsis patients, CD16 immature granulocytes are associated with lymphopenia and immunosuppressive patient characteristics [167]. Not only CD16 int or immature granulocytes impact lymphocytes, but also CD16hi-identifiable subsets (CD16hi CD62Llo CD11chi CD11b hi) of mature neutrophils can equally impact T-cell functionality. Herein, these neutrophils can effectively hinder T-cell proliferation and responses via a Mac-1/ROS-dependent inflammation-mediated mechanism [168]. This population of mature NET-producing neutrophils can also target other disease categories, including NET targeting of tumor cells, and shows distinct characteristic features, such as reduced adhesion, potentially facilitating cellular translocation [169,170]. Functional relevance with regard to neutrophil subsets has been highlighted in a model of human-LPS-mediated systemic inflammation. In this model, overall neutrophil populations showed reduced levels of common surface receptors, such as CXCR1/2 and TLR4, but increased respiratory burst priming. Nonetheless, CD16(dim)-banded newly produced neutrophils exhibit reduced antimicrobial functionality, hinting at a rather complex time-dependent neutrophil population landscape present during systemic inflammation, possibly contributing to the various stages of hyperinflammation and secondary infection susceptibility [171].
Activation responses do not only include ROS formation, but also other changes in surface receptor compositions and intracellular signaling cascades. Notably, engagement of chemokines or selectins leads to activation of β2-integrins on the neutrophil surface, subsequently exposing an activated phenotype [3]. Additionally, receptor shedding occurs, and this leads to downregulation of L-selectin, which is by itself crucial for subsequent antibacterial and pro-migratory responses [19]. Other surface markers have also been associated with distinct neutrophil functions and phenotypes. One such marker relevant here is the glycoprotein CD177, which is expressed at variable levels in healthy humans [172]. Even though no apparent phenotype is visible in CD177lo or CD1177hi voluntary donors, and CD177 by itself is incapable of signal transmission, this receptor can signal via β2–integrin interaction and thereby mediate neutrophil migration [172,173]. This receptor, a marker for neutrophil activation, was additionally used for correlation with COVID-19 severity and is also linked to protective mechanisms in inflammatory bowel disease [174,175]. Aside from regulative mechanisms affecting CD177 and integrins, other receptors, such as CD66, are mobilized to the neutrophil surface upon activation and consecutively allow binding of ligands (such as galectins) for induction of secondary immune responses, thus showcasing activation-dependent neutrophil phenotypes [157]. Notably, neutrophil phenotype switching is not only traceable due to classical activation at sites of local inflammation, but distinct subpopulations can be identified, as formerly extravasated cells re-enter into the circulation via a process termed reverse migration [176]. In aged mice, these cells, which are characterized, amongst others, by CD54hi CXCR1lo expression, can traffic to the lung and cause endothelial barrier disruption [80,93,177]. Thus, the presence of neutrophil subpopulations is markedly affected by both local and systemic levels of inflammatory mediators and mechanisms of cellular interplay, impacting surface receptor composition as well as intracellular signaling elements, the latter of which includes the safeguard transcription factor PU.1 [178].
Immune cells are communicative cells. Neutrophils show swarm-like behavior in a LTB4-dependent manner, but such a swarm response is self-limited via GPCR desensitization, showcasing auto-responsive inflammatory imprinting mechanisms and presence of phenotypically distinct local-site neutrophil swarm subsets [179,180]. Importantly, overarching principles, such as CXCR2-Bmal1-dependent circadian proteome changes occurring in neutrophils, additionally impact the magnitude of neutrophil phenotypes detectable within the body [181]. Application of scRNAseq techniques for bone marrow, blood and spleen indeed corroborated the existence of neutrophil subsets during steady state and infection, highlighting eight distinct phenotypic populations [182].

7. Platelets as Immune Cells and Platelet Phenotypes during Inflammation

Platelets are prominently known for their hemostatic potential. Nonetheless, in recent years, their immunologic potential has been brought into focus [39,183]. Indeed, migration, bundling of bacteria and impacting of leukocyte recruitment are just a few inflammatory and immunomodulatory characteristics which make it valid to address these cellular particles as immune cells [39,51,184,185]. Similar to the distinction of neutrophils into low- and normal-density cells, platelets have also been determined by large-heavy (supposedly young) or small-light (supposedly old) characteristics with varying aggregation responses [186,187]. Platelets are commonly distinguished according to their activation state. Activated platelets can feature a high-affinity GPIIb/IIIa receptor conformation, reduced GPIb levels and P-selectin surface mobilization, amongst others, when confronted with various stimuli. Platelets additionally express the glucocorticoid receptor alpha, which, when activated by prednisolone, impacts P2Y12 ADP signaling to inhibit platelet aggregation [188,189]. Platelets are anucleate cellular particles, which partially restrict the capabilities for phenotypic changes. Nevertheless, they contain various mRNAs and—upon activation—synthesize pro-IL-1β protein and IL-1β in a β3-integrin-dependent manner [190]. Platelets overall contain not only cytokines but various chemokines, which can be rapidly released upon activation [191]. Taking into consideration the numerical magnitude of platelets, the effects of platelet activation and immediate chemokine/cytokine release can be detrimental for the host. Platelets can additionally release extracellular vesicles (EVs). This EV release differs according to cellular interplay, platelet stimulation and disease-specific microenvironmental cues [192,193,194,195]. Interestingly, when exposed to stressors such as inflammation, platelet production within the organism itself changes [196,197]. Occurrence of immature platelets in sepsis patients is well-documented and can serve as a predictor for mortality and identification of sepsis patients [198,199,200]. Equally, the immature platelet fraction can aid in identification of inflammatory complications in cardiac surgery patients [201]. Inflammatory changes, such as increased IL-1β levels, indeed favor megakaryocyte rupture, potentially leading to observable changes in platelet numbers and phenotypes [202]. The mode of production as well as the production site might impact the occurrence of distinct platelet populations. Aside from bone-marrow-derived platelets, pulmonary and splenic platelet production have also been reported. Whereas the lung continuously contributes to platelet production, inflammation or changed platelet demand can lead to shifting in platelet production sites with consequent changes in platelet phenotypes [203]. During sepsis, megakaryocyte progenitors are mobilized to the spleen, where they differentiate in an IL-3-dependent manner to fully mature megakaryocytes, which then produce a distinct population of CD40Lhi platelets [50]. Aside from such inflammation-induced or immature platelets, aged platelets can also be detected in the circulation. These platelets feature impaired P-selectin mobilization upon thrombin stimulation together with proteome changes linked to increased apoptosis and senescence and reduced spreading and secretion capacities [204,205]. The de-sialylation of these aged platelets results in a feedback loop, stimulating TPO synthesis and thus platelet reproduction, showcasing the interconnection between circulating platelet subpopulations [206,207]. Other approaches to identifying distinct platelet phenotypes or subpopulations involve discovery of eNOS-positive and -negative platelets within human blood, suggesting these two population phenotypes distinctly regulate thrombosis onset [208]. Other phenotypic occurrences include the detection of serotonin-enhanced procoagulant COAT platelets in stroke patients [209,210,211]. Extensive multiparameter phenotyping of platelets can also serve to identify prostacyclin-specific platelet responses [212], or specific human patient cohorts, even in a population of elsewise healthy-appearing humans, identifying six subgroups with distinct platelet phenotypes and reactivity patterns [213]. In addition, combinatorial proteomics and lipidomics studies have enabled deeper characterization of platelet responses and phenotypic subsets [214]. With regard to the COVID-19 pandemic, platelet phenotyping has aided in the identification of disease severity in patient populations [12]. Surprisingly, even though platelets express FPR2 and react to Annexin A1 [215], no examination has been performed to stratify platelets by their pro-inflammatory and pro-resolution potential, as it appears plausible to distinguish between Plat1 (pro-inflammatory) and Plat2 (pro-resolution) platelets, similar to the macrophage and neutrophil characterization, although more detail and comparative studies are still needed (Table 2) [215,216].
Table 2. Commonly ascribed immune cell phenotypes.
Table 2. Commonly ascribed immune cell phenotypes.
Cell TypePhenotypeMechanismCharacteristics
PlateletsImmature plateletsEmergency thrombopoiesis
-
High granularity
-
Increased size
Sepsis-induced splenic plateletsIL-3-mediated splenic megakaryocyte maturation
-
CD40Lhi
Lung-derived plateletsS1P-gradient-dependent platelet shedding in pulmonary microvasculature
-
Phenotypic distinction so far unclear [217]
Young vs. oldLife-cycle dependent aging
-
Desialylated old platelets
-
Stimulation of TPO production
COATed plateletsDual stimulation with collagen and thrombin
-
Phosphatidylserine exposure
-
High fibrinogen binding
Post-emperipolesis plateletsMegakaryocyte emperipolesis followed by thrombopoiesis
-
Containing neutrophil membrane fragments
Plat 1 vs. Plat 2Differing stimuli and microenvironments
-
Pro-inflammatory vs. pro-resolution
NeutrophilsNDNDensity gradient purification
-
Containing mostly mature neutrophils
LDNDensity gradient purification
-
Containing immature neutrophils
-
Containing neutrophils with elevated CD66b, CD11b
N1Increased following TGF-beta blockade
-
Immunostimulating
-
Cytotoxic, tumor-suppressive
-
Mature phenotype
-
Short lifecycle [218]
-
CD11blo
N2PPARy-dependent
-
Immunosuppressive,
-
Tumor-permitting, pro-metastatic
-
CD206+
-
Increasingly efferocytosed during stroke
Reverse-migrated neutrophilsLTB4–Neutrophil Elastase-dependent
-
Over-activated
-
CD54hi CXCR1lo
-
Capable of causing remote injury
Aged neutrophilsLife-cycle-dependent aging, microbiota-TLR-MyD88-dependent
-
CXCR4hi
MacrophagesM1LPS-, IFN-dependent
-
Pro-inflammatory
M2 (a, b, c, d)IL-10, IL-4-dependent
-
Anti-inflammatory
AlveolarLocalization
-
Patrolling of airways
-
Diverse population (including SiglecF+ and SiglecF cells)
InterstitialLocalization
-
Diverse population (including Lyve1+/−, MHCII+/−, CX3CR1+/− cells)
-
Localized in interstitium
ResidentOrgan-inherent resident immune cells
-
Not mobilized but detectable both under baseline and inflammatory conditions
PerivascularLocalization vs. organ-specific expression patterns
-
Diverse population [219]
-
Capable of modulating vascular development
ProfibroticIncreased in pulmonary bleomycin model
-
SiglecF+ CD11c+ MHCIIhi
Monocyte derivedRecruitment of monocytes and maturation to macrophages
-
CX3CR1+ (mouse)-derived
-
CD16/CD14 (human)-derived
Splenic macrophagesAnatomic localization
-
Marginal metallophilic macrophages
-
Marginal zone macrophages
-
Red pulp macrophages

8. Regulatory Aspects of DAMPS, PAMPs, Microenvironments and Cellular Interplay

Inflammation can be regarded as the body’s defense reaction to a stimulus, injury or infection which is either well-controlled and adequately triggered, resulting in the successful combating of an invading pathogen, or occurs in excess or under unwanted circumstances, as can be observed in atherosclerosis and rheumatoid arthritis. Pro-inflammatory characteristics are attributed to the notion that a distinct cellular phenotype or response can be perceived as a main driver of inflammation, leading to the putative identification of the main cell type. Distinct tissues contain various chemokine reservoirs, which can be released and differentially control immune cell influx and activation [111,220]. Depending on the site of injury, cause and duration of damage, the cellular as well as chemokine profiles will vary.
Overall, inflammation is associated with the release of a variety of cytokines and chemokines, including TNF, IL-1, IL-6, IL-8, IL-23, CXCL-1, CXCL-2 and procalcitonin, as well as autacoids, neuropeptides, prostanoids and complement factors. Cytokines and chemokines can differentially affect and activate immune cells and likewise contribute to regulation of vascular permeability and immune cell (re-)production. Upon stimulation with IFN and LPS, macrophages, for example, take on the classic M1 pro-inflammatory phenotype. As can be seen by this, engagement of TLR ligands, such as bacterial components including LPS, thus evokes a pro-inflammatory response which can be limited in a TRAF2-dependent, c-Rel, IRF5-dependent manner [221]. This pro-inflammatory polarization of macrophages by TLR4 is additionally controlled by Girdin, which dampens the extent of the pro-inflammatory responses by suppression of NFkB, CREB and p38 MAPK-pathway activation, as Girdin hinders TLR4 receptor dimerization, which is crucially needed for pro-inflammatory signal transduction [222]. Not only bacterial components do lead to pro-inflammatory phenotypes, but also engagement of, for example, calcium depositions within atherosclerotic lesions will trigger TNF secretion and proinflammatory macrophage responses in a PKC-, ERK- and JNK-dependent manner [223]. With regard to regulation of the dynamics of macrophage phenotypes during inflammation, a rapid mobilizable peritoneal population has been described, and this is capable of invading injured tissue in a CD44- and ATP-dependent manner while switching to an AAM phenotype [224], as peritoneal macrophage polarization depends on retinoic acid and GATA6 [225]. Importantly, various transcription factors have been implicated in the maturation and polarization of M1/M2 macrophages, including PU.1 (global enhancer repertoire, but no effect on polarization), STAT1 (M1), IRF5 (M1), STAT6 (M2), IRF4 (M2), C/EBPb (regulating Arg1, IL10 and Mrc1) and PPARγ (M2) [226,227]. Likewise, the repression of histone H3 proteolytic cleavage, which is mediated by CTSG, ELANE and PRTN3, is involved in the maturation of monocytes to fully functional macrophages [228]. Additionally, polarization responses of macrophages depend on the tissue environment and can be exemplary primed by β2-adrenergic responses and the local microenvironment itself [229,230,231]. Only recently, the olfactory receptor Olfr2/OR6A2 was identified in vascular macrophages and shown to be linked via circulating octanal levels to IL-1β release by macrophages, inflammation and stimulation of atherosclerosis [232]. This highlights the complex regulatory interaction of one signal (in this case octanal) leading to the boosting of an additional pro-inflammatory signal (IL-1). Aside from these proinflammatory signals, anti-inflammatory or signals of mixed phenotypic characteristics are also at play. Thus, as IL-4 directs macrophages towards an M2 phenotype, it also limits neutrophil expansion and interferes with CXCR2/4-dependent tissue translocation [233]. An interesting example is represented by IL-12, which plays a somewhat dual role as it contains a subunit as IL-35 (IL-12p35); this demonstrates that various influencing factors are at play for the priming of an inflammatory response. IL-12 can assist in activation of macrophages as well as NK cells, thus displaying properties of a pro-inflammatory cytokine. Nonetheless, in high-blood-pressure settings, the IL-35 subunit of IL-12 reduced M1 macrophages [234,235]. Equally contributing to the notion of diverse regulatory mechanisms, the developmental endothelial locus-1 protein, on the one hand, hinders leukocyte recruitment when secreted by endothelial cells, but, on the other hand, within macrophages, controls their efferocytotic and pro-resolving properties, indicating how cellular and microenvironmental factors do contribute to phenotypic cellular responses [236]. Although it might appear favorable to introduce M2-polarized macrophages into a setting in which a healing or resolution response is needed, this is not feasible in all conditions, as, for example, topical application of M2 macrophages into diabetic wounds does not aid in wound healing or neutrophil clearance [237].
Cytokines can be released by macrophages and surgery as well as other injuries that are associated with peak cytokine levels, depending on the site of the incision/damage [111]. Cytokines and chemokines bind to specific receptors to start cellular activation cascades, which differentially prime immune cell responses. In the lung, macrophages constantly clear the airways in order to guarantee homeostasis [238]. Comparable mechanisms are at play in other organs and environments, such as the peritoneum, where tissue-resident macrophages prevent neutrophil activation in order to maintain tissue homeostasis [239]. Nonetheless, under inflammatory conditions, neutrophils rapidly invade upon bacterial infection or toxic injury [111]. Endothelial cells and neutrophils are activated, allowing the phenomena of neutrophil rolling, adhesion and extravasation in a selectin- and chemokine-dependent manner [40]. DAMPs and PAMPs both prime neutrophils to take on an activated phenotype. Such an inflammatory microenvironment can also be elicited by the release of ATP from necrotic cells, which subsequently activates the Nlrp3 inflammasome via P2rx7 signaling and results in formyl peptide signals, attracting neutrophils that take on proinflammatory chemotactic functions [240]. Herein, interaction of fMLP with its receptor (FPR1) leads to a BTK-dependent pro-inflammatory phenotype switch, resulting in activation of β2-integrins, more specifically Mac-1 [241]. Nonetheless, in human neutrophils, absence of Btk can also be associated with plasma membrane association of Rac2, Mal, PI(3)K and PTKs, leading to a primed activation state, as BTK under baseline conditions interacts with Mal, controlling its cytoplasmic localization [242]. As an additional signaling response, engagement of the chemokine CXCL-1 and binding to its receptor CXCR2 results in activation of integrin LFA-1 in a talin-1, kindlin-3, Rap/Riam and ILK-dependent pathway [3,243]. Binding of a chemokine to its GPCR and subsequent downstream signaling is additionally controlled and self-limited by desensitization occurring in neutrophils in a GRK2-dependent manner, thereby limiting the swarming behavior of these cells when in a proinflammatory state [179]. Aside from activating signaling receptors, inhibitory receptors can also be found within neutrophils. For example, the ITIM receptor PILRα controls integrin activation and neutrophil responses, in which its absence leads to increased neutrophil recruitment, whereas absence of the ITIM receptor LY49Q is associated with overactivation of β2 integrins, resulting in inadequate leukocyte adhesion, preventing transmigration, by this showing partially opposing signaling responses even within similar receptor classes [244,245].
Neutrophils are migratory cells, relying on adequate actin cytoskeleton reorganization and integrin functionality. Crucial for the well-oriented directional response towards differential chemokine gradients is the PTEN pathway, which regulates PI(3)kinase activity, phospholipase A2 and p38-dependent migration, impacting bacterial defense and directionality phenotypes of pro-inflammatory neutrophils [246]. Another integrin regulatory element in neutrophils is the myosin light-chain kinase, which, through activation of Pyk2, mediates β2-integrin activation, additionally linking integrins to actin-cytoskeleton-related mechanistic features [247]. Neutrophil activation switching furthermore crucially relies on calcium and potassium channels, such as KV1.3, ORAI and STIM, which all allow for the coordination of calcium signaling and spikes within the cell, impacting all functions ranging from integrin activation to phagocytosis [248,249,250,251]. Involved in the regulation of β2-integrin activation is additionally the downstream regulatory element antagonist modulator (DREAM), which regulates neutrophil responses following TNF stimulation but not fMLP via interaction with A20, a NFkB regulator and IkB kinase [252]. The overall neutrophil responses are tightly controlled by various transcription factors, as shown in a zymosan air pouch model, including RUNX1 and KLF6, being implicated in both neutrophil maturation and recruitment, while RFX2 and RELB are implicated in neutrophil survival; in this experimental setting, RELB, IRF5 and JUNB regulate neutrophil activation and effector functions [253]. An interesting function has been attributed to neutrophils in injury settings; herein, they transport matrix components to a wounded site, leading to scar formation in a kindlin-3-integrin-activation-dependent manner in which heat shock proteins induce integrin heterodimerization, indicating that an activated, supposedly pro-inflammatory phenotype can be functionally rather diverse, and even take on a profile comparable to reparative M2 macrophage features [254]. Phenotypic modulation of neutrophil responses is due to various activation/deactivation mechanisms. Similarly, reverse migration leads to a phenotypic shift in neutrophils. Herein, cells exhibit ICAM1hiCXCR1lo expression levels and are primed for executing subsequent, often distant, damage at remote organs in settings of increased vascular permeability [177,255]. This is controlled by LTB4, neutrophil elastase, JAM-C and Mac-1 [256]. Notably, such reverse-migration-regulative phenotype responses might also impact secondary organ damage related to UV light damage of the skin, resulting in neutrophil-mediated kidney injury [257].
Finally, cellular interplay affects neutrophil responses and phenotypes. Therein, platelets prime neutrophils for their proinflammatory responses via PSGL-1 interaction with P-Selectin [258]. This priming goes even further as neutrophil GPR35 is activated by platelet-derived serotonin-derivative 5-HIAA, thereby promoting the extravasation of neutrophils [259]. This platelet–neutrophil serotonin interplay is also known to impact myocardial damage by controlling neutrophil degranulation [260]. Looking at platelet–cellular interplay in more detail, platelets via interaction with Tregs also contribute to macrophage polarization in airways during bacterial pneumonia.Platelet-mediated IL-10/TGFβ release leads to an M2-phenotypic priming of macrophages, impacting macrophage efferocytotic capacity and thereby neutrophil clearance from the airways [51]. Platelets by themselves are mainly regarded as taking on activated or inactive phenotypes. Mechanisms underlying these activation responses have been summarized in the past and include engagement of, for example, ADP receptors, thrombin receptors, vWF receptors or collagen receptors [261]. Importantly, focusing on the immunologic aspects of platelets, the migrating and bacterial-bundling capacity of platelets is linked to GPIIb/IIIa activity, and calcium and Myosin-IIa-dependent trailing edge formation [262]. This repositioning ability of platelets additionally aids in an Arp2/3-dependent manner in the maintenance of vascular integrity and surface repair during inflammatory microbleeding, hinting at a sealing phenotype of these migratory-active platelets [263].

9. Translational Findings and Immune Phenomics of COVID-19

Laboratory assessment of patients is vital for understanding current health status, disease progression and therapeutic implications. Mass cytometry and single-cell techniques have aided in the acquisition of large-scale clinical and animal study datasets. Cellular phenotyping and assessment of signaling pathways impacting phenotypic priming of immune cells are thus of tremendous interest as they can help us to understand the immunologic landscape of various patient populations, as summarized in Table 3 [264].
Aside from these purely diagnostic purposes, phenotypic assessment has led to advances in the understanding of mechanistic implications and signaling pathways regulating immune hypo- vs. hyper-activation. Targeting of specific populations as well as distinct pathway elements thus appears tempting for therapeutic purposes [272]. For example, local IL-1 administration might aid in the phenotypic priming of alternatively activated macrophages via activation of GATA-3, and timed local administration of platelets could also potentially aid in the shifting of inflammatory cell phenotypes [51,273]. Furthermore, the PAM3-based M2-phenotypic priming of macrophages was used in translational animal models to improve disease course in lupus and inflammatory colitis [274,275]. Likewise, administration of either high-dose dexamethasone or retinoic acid leads to a shifting in macrophage phenotypes towards M2 in a setting of immune thrombocytopenia, as combinatorial administration leads to a sustained response in these patients [276,277]. Active targeting of on–off switches of both inflammation and resolution might aid in the focused guiding of immune cellular subsets to sites of necessity. Exemplary, an agonistic ChemR23 antibody, activating this resolvin E1 receptor, promotes macrophage efferocytosis and aids in the reduction in neutrophil tissue accumulation, stimulating inflammation resolution [278]. While pursuing another pro-resolution pathway, the FPR2/ALX pathway additionally targets neutrophils themselves via Akt and ERK1/2 signaling in sickle cell disease, limiting the pro-NETosis phenotype of neutrophils and aiding in resolution of thrombo-inflammation [279]. Additionally, dietary peculiarities should be considered for switching of phenotypic properties of immune cells. Indeed, a high-salt diet via induction of hyperglucocorticoidism results in reduced bacterial defense capacity of neutrophils [280].
In addition to these translational disease settings and pathomechanistic peculiarities, recent focus was placed on the COVID-19 pandemic, highlighting consequences of an outraged immune system. As mentioned above, specific cellular subsets, such as neutrophil CD16int populations, correlate with hypercoagulation and disease severity in COVID-19 [165]. The Stark/Massberg laboratory used an intriguing approach in assessing phenotypic configurations of immune cells in COVID-19 patients, revealing disturbed neutrophil and platelet activation patterns in the blood of COVID-19 patients, correlating with disease severity [12]. Additionally, other groups have shown, through various techniques, the tremendous impact COVID-19 has on leukocytes and platelets in particular, including IL-6-mediated platelet activation and STAT3 phosphorylation within monocytes and T cells [281,282,283]. As leukocyte and platelet dysregulated functionality is observable, the neutrophil activation marker CD177 can also be used for assessment of COVID-19 severity [174]. Additionally, a CXCL-8/IL-8-dysregulated prothrombotic neutrophil phenotype, favoring degranulation and NETosis, was ascribed to thrombotic complications in COVID-19 [284,285]. Comparative analyses between rheumatoid arthritis and COVID-19 samples revealed similar pro-inflammatory disease patterns regulated via SPP1 and S100A12, leading to activation of proinflammatory CD14+ monocytes and PD-L1+ neutrophils [286]. Another study reported on the severity-related increase in immature CD16dim neutrophil subsets with increased levels of CD66b, LOX-1 and CD24 and overall changes in granulocyte populations, which are partly restored upon recovery from COVID-19, showing a near complete absence of immature CD16dim neutrophils and normalization of CXCR1, CXCR4 and CD147 levels [287]. McDonald et al. performed a prospective study in COVID-19 ARDS ICU patients, utilizing mass cytometry for single-cell analysis. This work highlights distinct neutrophil phenotypes, identifying a functional priming towards NET release without alteration of other cellular functions, such as ROS production. This phenotype existed independently of dexamethasone treatment, highlighting the necessity of other more accurate therapeutics targeting discrete neutrophil functional phenotypes [288]. Therapeutic consequences of these phenotypic pathway assessments are so far limited but might aid in the differentiation of patient subcategories and sooner or later the targeted intervention with regard to pro-/anti-inflammatory cell populations.

10. Outlook and Conclusions

Deeper analyses of distinct immune cell phenotypes are crucial for assessment and stratification of patient populations and disease patterns. Identification of overarching extracellular and intracellular factors contributing to phenotypic re-composition of the immune system will allow for direct targeting of distinct cellular responses for modes of action, such as combating invading pathogens, preventing microvascular leakage or restoring tissue integrity. Due to the large datasets implicated in phenotype assessment, artificial intelligence and machine learning ought to assist in pattern recognition in order to determine novel cellular subsets or behavioral cellular traits [11]. Nonetheless, we deem crucial the determination of functional and clinical relevance of such increasingly detailed cellular patterns and the link of phenotypic features to diseases; in other words, we need to ascertain what is crucial or important, and what is disposable from a pathomechanistic and pharmacological perspective. Additionally, it is important that we determine whether distinct phenotypes are transient or constant or even permanent, or rather simply fluid-phase patterns resulting from maturation steps of prematurely released populations. After all, the holy grail of immune cell phenotype research remains the discovery of specific cues which evoke a cellular response resembling the desired phenotype: for example, a pro-phagocytic macrophage, clearing all neutrophils, without exerting any damaging effects on the surrounding tissue or fibrotic effects. Then, we might be able to switch on/off as needed or trigger another functional type. Lastly, it is tempting to speculate about the pharmacological utility of distinct immune cell populations for therapy. Indeed, the Ley Lab recently demonstrated that disease-specific monocytes control inflammation and mortality in a cystic fibrosis model, hinting that healthy-phenotype monocytes could potentially aid in therapy for such a disease [289].

Author Contributions

A.M. conceived and wrote the manuscript. M.P. critically revised the manuscript and contributed to writing. Both authors edited the final version of the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported by the German Research Foundation (DFG MA9604/1-1 grant to A.M.).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Acknowledgments

We would like to thank Isobel A. Blacksell for critical review of the manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Leeuwenhoek, A.V. Arcana Naturae Detecta; Delphis Batavorum, Apud Henricum a Krooneveld: Delft, The Netherlands, 1695. [Google Scholar]
  2. Andral, G. Essai D’hématologie Pathologique; Fortin, Masson et Cie: Paris, France, 1843. [Google Scholar]
  3. Margraf, A.; Germena, G.; Drexler, H.C.A.; Rossaint, J.; Ludwig, N.; Prystaj, B.; Mersmann, S.; Thomas, K.; Block, H.; Gottschlich, W.; et al. The integrin-linked kinase is required for chemokine-triggered high-affinity conformation of the neutrophil beta2-integrin LFA-1. Blood 2020, 136, 2200–2205. [Google Scholar] [CrossRef] [PubMed]
  4. Tracz, J.; Handschuh, L.; Lalowski, M.; Marczak, L.; Kostka-Jeziorny, K.; Perek, B.; Wanic-Kossowska, M.; Podkowinska, A.; Tykarski, A.; Formanowicz, D.; et al. Proteomic Profiling of Leukocytes Reveals Dysregulation of Adhesion and Integrin Proteins in Chronic Kidney Disease-Related Atherosclerosis. J. Proteome Res. 2021, 20, 3053–3067. [Google Scholar] [CrossRef] [PubMed]
  5. Alarcon-Barrera, J.C.; von Hegedus, J.H.; Brouwers, H.; Steenvoorden, E.; Ioan-Facsinay, A.; Mayboroda, O.A.; Ondo-Mendez, A.; Giera, M. Lipid metabolism of leukocytes in the unstimulated and activated states. Anal. Bioanal. Chem. 2020, 412, 2353–2363. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Schafflick, D.; Xu, C.A.; Hartlehnert, M.; Cole, M.; Schulte-Mecklenbeck, A.; Lautwein, T.; Wolbert, J.; Heming, M.; Meuth, S.G.; Kuhlmann, T.; et al. Integrated single cell analysis of blood and cerebrospinal fluid leukocytes in multiple sclerosis. Nat. Commun. 2020, 11, 247. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Leite, G.G.F.; Ferreira, B.L.; Tashima, A.K.; Nishiduka, E.S.; Cunha-Neto, E.; Brunialti, M.K.C.; Assuncao, M.; Azevedo, L.C.P.; Freitas, F.; van der Poll, T.; et al. Combined Transcriptome and Proteome Leukocyte’s Profiling Reveals Up-Regulated Module of Genes/Proteins Related to Low Density Neutrophils and Impaired Transcription and Translation Processes in Clinical Sepsis. Front. Immunol. 2021, 12, 744799. [Google Scholar] [CrossRef] [PubMed]
  8. Fay, M.E.; Myers, D.R.; Kumar, A.; Turbyfield, C.T.; Byler, R.; Crawford, K.; Mannino, R.G.; Laohapant, A.; Tyburski, E.A.; Sakurai, Y.; et al. Cellular softening mediates leukocyte demargination and trafficking, thereby increasing clinical blood counts. Proc. Natl. Acad. Sci. USA 2016, 113, 1987–1992. [Google Scholar] [CrossRef] [Green Version]
  9. Tse, H.T.; Gossett, D.R.; Moon, Y.S.; Masaeli, M.; Sohsman, M.; Ying, Y.; Mislick, K.; Adams, R.P.; Rao, J.; Di Carlo, D. Quantitative diagnosis of malignant pleural effusions by single-cell mechanophenotyping. Sci. Transl. Med. 2013, 5, 212ra163. [Google Scholar] [CrossRef] [Green Version]
  10. Margraf, A.; Cappenberg, A.; Vadillo, E.; Ludwig, N.; Thomas, K.; Korner, K.; Zondler, L.; Rossaint, J.; Germena, G.; Hirsch, E.; et al. ArhGAP15, a RacGAP, Acts as a Temporal Signaling Regulator of Mac-1 Affinity in Sterile Inflammation. J. Immunol. 2020, 205, 1365–1375. [Google Scholar] [CrossRef]
  11. Crainiciuc, G.; Palomino-Segura, M.; Molina-Moreno, M.; Sicilia, J.; Aragones, D.G.; Li, J.L.Y.; Madurga, R.; Adrover, J.M.; Aroca-Crevillen, A.; Martin-Salamanca, S.; et al. Behavioural immune landscapes of inflammation. Nature 2022, 601, 415–421. [Google Scholar] [CrossRef]
  12. Nicolai, L.; Leunig, A.; Brambs, S.; Kaiser, R.; Weinberger, T.; Weigand, M.; Muenchhoff, M.; Hellmuth, J.C.; Ledderose, S.; Schulz, H.; et al. Immunothrombotic Dysregulation in COVID-19 Pneumonia Is Associated With Respiratory Failure and Coagulopathy. Circulation 2020, 142, 1176–1189. [Google Scholar] [CrossRef]
  13. Ballesteros, I.; Rubio-Ponce, A.; Genua, M.; Lusito, E.; Kwok, I.; Fernandez-Calvo, G.; Khoyratty, T.E.; van Grinsven, E.; Gonzalez-Hernandez, S.; Nicolas-Avila, J.A.; et al. Co-option of Neutrophil Fates by Tissue Environments. Cell 2020, 183, 1282–1297. [Google Scholar] [CrossRef] [PubMed]
  14. Peng, B.; Geue, S.; Coman, C.; Munzer, P.; Kopczynski, D.; Has, C.; Hoffmann, N.; Manke, M.C.; Lang, F.; Sickmann, A.; et al. Identification of key lipids critical for platelet activation by comprehensive analysis of the platelet lipidome. Blood 2018, 132, e1–e12. [Google Scholar] [CrossRef] [PubMed]
  15. Anders, C.B.; Lawton, T.M.W.; Smith, H.L.; Garret, J.; Doucette, M.M.; Ammons, M.C.B. Use of integrated metabolomics, transcriptomics, and signal protein profile to characterize the effector function and associated metabotype of polarized macrophage phenotypes. J. Leukoc. Biol. 2022, 111, 667–693. [Google Scholar] [CrossRef] [PubMed]
  16. Zeming, K.K.; Lu, R.; Woo, K.L.; Sun, G.; Quek, K.Y.; Cheow, L.F.; Chen, C.H.; Han, J.; Lim, S.L. Multiplexed Single-Cell Leukocyte Enzymatic Secretion Profiling from Whole Blood Reveals Patient-Specific Immune Signature. Anal. Chem. 2021, 93, 4374–4382. [Google Scholar] [CrossRef] [PubMed]
  17. Rossaint, J.; Berger, C.; Van Aken, H.; Scheld, H.H.; Zahn, P.K.; Rukosujew, A.; Zarbock, A. Cardiopulmonary bypass during cardiac surgery modulates systemic inflammation by affecting different steps of the leukocyte recruitment cascade. PLoS ONE 2012, 7, e45738. [Google Scholar] [CrossRef] [Green Version]
  18. Edwards, D.R.; Handsley, M.M.; Pennington, C.J. The ADAM metalloproteinases. Mol. Asp. Med. 2008, 29, 258–289. [Google Scholar] [CrossRef]
  19. Cappenberg, A.; Margraf, A.; Thomas, K.; Bardel, B.; McCreedy, D.A.; Van Marck, V.; Mellmann, A.; Lowell, C.A.; Zarbock, A. L-selectin shedding affects bacterial clearance in the lung: A new regulatory pathway for integrin outside-in signaling. Blood 2019, 134, 1445–1457. [Google Scholar] [CrossRef]
  20. Zhu, X.D.; Zhuang, Y.; Ben, J.J.; Qian, L.L.; Huang, H.P.; Bai, H.; Sha, J.H.; He, Z.G.; Chen, Q. Caveolae-dependent endocytosis is required for class A macrophage scavenger receptor-mediated apoptosis in macrophages. J. Biol. Chem. 2011, 286, 8231–8239. [Google Scholar] [CrossRef] [Green Version]
  21. Kumari, S.; Mg, S.; Mayor, S. Endocytosis unplugged: Multiple ways to enter the cell. Cell Res. 2010, 20, 256–275. [Google Scholar] [CrossRef] [Green Version]
  22. Rose, J.J.; Foley, J.F.; Murphy, P.M.; Venkatesan, S. On the mechanism and significance of ligand-induced internalization of human neutrophil chemokine receptors CXCR1 and CXCR2. J. Biol. Chem. 2004, 279, 24372–24386. [Google Scholar] [CrossRef] [Green Version]
  23. Forster, R.; Kremmer, E.; Schubel, A.; Breitfeld, D.; Kleinschmidt, A.; Nerl, C.; Bernhardt, G.; Lipp, M. Intracellular and surface expression of the HIV-1 coreceptor CXCR4/fusin on various leukocyte subsets: Rapid internalization and recycling upon activation. J. Immunol. 1998, 160, 1522–1531. [Google Scholar] [PubMed]
  24. Cramer, E.M.; Lu, H.; Caen, J.P.; Soria, C.; Berndt, M.C.; Tenza, D. Differential redistribution of platelet glycoproteins Ib and IIb-IIIa after plasmin stimulation. Blood 1991, 77, 694–699. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Gabl, M.; Holdfeldt, A.; Sundqvist, M.; Lomei, J.; Dahlgren, C.; Forsman, H. FPR2 signaling without beta-arrestin recruitment alters the functional repertoire of neutrophils. Biochem. Pharmacol. 2017, 145, 114–122. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Forsman, H.; Onnheim, K.; Andreasson, E.; Christenson, K.; Karlsson, A.; Bylund, J.; Dahlgren, C. Reactivation of desensitized formyl peptide receptors by platelet activating factor: A novel receptor cross talk mechanism regulating neutrophil superoxide anion production. PLoS ONE 2013, 8, e60169. [Google Scholar] [CrossRef] [Green Version]
  27. Zhang, X.; Kluger, Y.; Nakayama, Y.; Poddar, R.; Whitney, C.; DeTora, A.; Weissman, S.M.; Newburger, P.E. Gene expression in mature neutrophils: Early responses to inflammatory stimuli. J. Leukoc. Biol. 2004, 75, 358–372. [Google Scholar] [CrossRef] [Green Version]
  28. Andres-Benito, P.; Moreno, J.; Dominguez, R.; Aso, E.; Povedano, M.; Ferrer, I. Inflammatory Gene Expression in Whole Peripheral Blood at Early Stages of Sporadic Amyotrophic Lateral Sclerosis. Front. Neurol. 2017, 8, 546. [Google Scholar] [CrossRef] [Green Version]
  29. Ramilo, O.; Allman, W.; Chung, W.; Mejias, A.; Ardura, M.; Glaser, C.; Wittkowski, K.M.; Piqueras, B.; Banchereau, J.; Palucka, A.K.; et al. Gene expression patterns in blood leukocytes discriminate patients with acute infections. Blood 2007, 109, 2066–2077. [Google Scholar] [CrossRef] [Green Version]
  30. Liu, S.M.; Xavier, R.; Good, K.L.; Chtanova, T.; Newton, R.; Sisavanh, M.; Zimmer, S.; Deng, C.; Silva, D.G.; Frost, M.J.; et al. Immune cell transcriptome datasets reveal novel leukocyte subset-specific genes and genes associated with allergic processes. J. Allergy Clin. Immunol. 2006, 118, 496–503. [Google Scholar] [CrossRef]
  31. Lequerre, T.; Gauthier-Jauneau, A.C.; Bansard, C.; Derambure, C.; Hiron, M.; Vittecoq, O.; Daveau, M.; Mejjad, O.; Daragon, A.; Tron, F.; et al. Gene profiling in white blood cells predicts infliximab responsiveness in rheumatoid arthritis. Arthritis Res. Ther. 2006, 8, R105. [Google Scholar] [CrossRef] [Green Version]
  32. Godini, R.; Fallahi, H.; Ebrahimie, E. Network analysis of inflammatory responses to sepsis by neutrophils and peripheral blood mononuclear cells. PLoS ONE 2018, 13, e0201674. [Google Scholar] [CrossRef] [Green Version]
  33. Scicluna, B.P.; Uhel, F.; van Vught, L.A.; Wiewel, M.A.; Hoogendijk, A.J.; Baessman, I.; Franitza, M.; Nurnberg, P.; Horn, J.; Cremer, O.L.; et al. The leukocyte non-coding RNA landscape in critically ill patients with sepsis. Elife 2020, 9. [Google Scholar] [CrossRef] [PubMed]
  34. Margraf, A.; Nussbaum, C.; Rohwedder, I.; Klapproth, S.; Kurz, A.R.M.; Florian, A.; Wiebking, V.; Pircher, J.; Pruenster, M.; Immler, R.; et al. Maturation of Platelet Function During Murine Fetal Development In Vivo. Arter. Thromb. Vasc. Biol. 2017, 37, 1076–1086. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Koseoglu, S.; Peters, C.G.; Fitch-Tewfik, J.L.; Aisiku, O.; Danglot, L.; Galli, T.; Flaumenhaft, R. VAMP-7 links granule exocytosis to actin reorganization during platelet activation. Blood 2015, 126, 651–660. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Wersall, A.; Williams, C.M.; Brown, E.; Iannitti, T.; Williams, N.; Poole, A.W. Mouse Platelet Ral GTPases Control P-Selectin Surface Expression, Regulating Platelet-Leukocyte Interaction. Arter. Thromb. Vasc. Biol. 2018, 38, 787–800. [Google Scholar] [CrossRef] [Green Version]
  37. Rorvig, S.; Ostergaard, O.; Heegaard, N.H.; Borregaard, N. Proteome profiling of human neutrophil granule subsets, secretory vesicles, and cell membrane: Correlation with transcriptome profiling of neutrophil precursors. J. Leukoc. Biol. 2013, 94, 711–721. [Google Scholar] [CrossRef]
  38. Sheshachalam, A.; Srivastava, N.; Mitchell, T.; Lacy, P.; Eitzen, G. Granule protein processing and regulated secretion in neutrophils. Front. Immunol. 2014, 5, 448. [Google Scholar] [CrossRef] [Green Version]
  39. Margraf, A.; Zarbock, A. Platelets in Inflammation and Resolution. J. Immunol. 2019, 203, 2357–2367. [Google Scholar] [CrossRef]
  40. Margraf, A.; Lowell, C.A.; Zarbock, A. Neutrophils in acute inflammation: Current concepts and translational implications. Blood 2022, 139, 2130–2144. [Google Scholar] [CrossRef]
  41. Nagai, Y.; Garrett, K.P.; Ohta, S.; Bahrun, U.; Kouro, T.; Akira, S.; Takatsu, K.; Kincade, P.W. Toll-like receptors on hematopoietic progenitor cells stimulate innate immune system replenishment. Immunity 2006, 24, 801–812. [Google Scholar] [CrossRef] [Green Version]
  42. Mitroulis, I.; Kalafati, L.; Bornhauser, M.; Hajishengallis, G.; Chavakis, T. Regulation of the Bone Marrow Niche by Inflammation. Front. Immunol. 2020, 11, 1540. [Google Scholar] [CrossRef]
  43. Frodermann, V.; Rohde, D.; Courties, G.; Severe, N.; Schloss, M.J.; Amatullah, H.; McAlpine, C.S.; Cremer, S.; Hoyer, F.F.; Ji, F.; et al. Exercise reduces inflammatory cell production and cardiovascular inflammation via instruction of hematopoietic progenitor cells. Nat. Med. 2019, 25, 1761–1771. [Google Scholar] [CrossRef] [PubMed]
  44. Weber, G.F.; Chousterman, B.G.; He, S.; Fenn, A.M.; Nairz, M.; Anzai, A.; Brenner, T.; Uhle, F.; Iwamoto, Y.; Robbins, C.S.; et al. Interleukin-3 amplifies acute inflammation and is a potential therapeutic target in sepsis. Science 2015, 347, 1260–1265. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Schulte-Schrepping, J.; Reusch, N.; Paclik, D.; Bassler, K.; Schlickeiser, S.; Zhang, B.; Kramer, B.; Krammer, T.; Brumhard, S.; Bonaguro, L.; et al. Severe COVID-19 Is Marked by a Dysregulated Myeloid Cell Compartment. Cell 2020, 182, 1419–1440.e1423. [Google Scholar] [CrossRef]
  46. Kwok, I.; Becht, E.; Xia, Y.; Ng, M.; Teh, Y.C.; Tan, L.; Evrard, M.; Li, J.L.Y.; Tran, H.T.N.; Tan, Y.; et al. Combinatorial Single-Cell Analyses of Granulocyte-Monocyte Progenitor Heterogeneity Reveals an Early Uni-potent Neutrophil Progenitor. Immunity 2020, 53, 303–318.e305. [Google Scholar] [CrossRef] [PubMed]
  47. Kim, J.H.; Podstawka, J.; Lou, Y.; Li, L.; Lee, E.K.S.; Divangahi, M.; Petri, B.; Jirik, F.R.; Kelly, M.M.; Yipp, B.G. Aged polymorphonuclear leukocytes cause fibrotic interstitial lung disease in the absence of regulation by B cells. Nat. Immunol. 2018, 19, 192–201. [Google Scholar] [CrossRef] [PubMed]
  48. Yipp, B.G.; Kim, J.H.; Lima, R.; Zbytnuik, L.D.; Petri, B.; Swanlund, N.; Ho, M.; Szeto, V.G.; Tak, T.; Koenderman, L.; et al. The Lung is a Host Defense Niche for Immediate Neutrophil-Mediated Vascular Protection. Sci. Immunol. 2017, 2, eaam8929. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Margraf, A.; Ley, K.; Zarbock, A. Neutrophil Recruitment: From Model Systems to Tissue-Specific Patterns. Trends Immunol. 2019, 40, 613–634. [Google Scholar] [CrossRef] [PubMed]
  50. Valet, C.; Magnen, M.; Qiu, L.; Cleary, S.J.; Wang, K.M.; Ranucci, S.; Grockowiak, E.; Boudra, R.; Conrad, C.; Seo, Y.; et al. Sepsis promotes splenic production of a protective platelet pool with high CD40 ligand expression. J. Clin. Investig. 2022, 132. [Google Scholar] [CrossRef]
  51. Rossaint, J.; Thomas, K.; Mersmann, S.; Skupski, J.; Margraf, A.; Tekath, T.; Jouvene, C.C.; Dalli, J.; Hidalgo, A.; Meuth, S.G.; et al. Platelets orchestrate the resolution of pulmonary inflammation in mice by T reg cell repositioning and macrophage education. J. Exp. Med. 2021, 218, e20201353. [Google Scholar] [CrossRef]
  52. Kamnev, A.; Lacouture, C.; Fusaro, M.; Dupre, L. Molecular Tuning of Actin Dynamics in Leukocyte Migration as Revealed by Immune-Related Actinopathies. Front. Immunol. 2021, 12, 750537. [Google Scholar] [CrossRef]
  53. Sprenkeler, E.G.G.; Webbers, S.D.S.; Kuijpers, T.W. When Actin is Not Actin’ Like It Should: A New Category of Distinct Primary Immunodeficiency Disorders. J. Innate Immun. 2021, 13, 3–25. [Google Scholar] [CrossRef] [PubMed]
  54. Shao, B.; Yago, T.; Setiadi, H.; Wang, Y.; Mehta-D’souza, P.; Fu, J.; Crocker, P.R.; Rodgers, W.; Xia, L.; McEver, R.P. O-glycans direct selectin ligands to lipid rafts on leukocytes. Proc. Natl. Acad. Sci. USA 2015, 112, 8661–8666. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Fabbri, M.; Di Meglio, S.; Gagliani, M.C.; Consonni, E.; Molteni, R.; Bender, J.R.; Tacchetti, C.; Pardi, R. Dynamic partitioning into lipid rafts controls the endo-exocytic cycle of the alphaL/beta2 integrin, LFA-1, during leukocyte chemotaxis. Mol. Biol. Cell 2005, 16, 5793–5803. [Google Scholar] [CrossRef] [Green Version]
  56. Poggi, P.; Mirabella, R.; Neri, S.; Assirelli, E.; Dolzani, P.; Mariani, E.; Calder, P.C.; Chatgilialoglu, A. Membrane fatty acid heterogeneity of leukocyte classes is altered during in vitro cultivation but can be restored with ad-hoc lipid supplementation. Lipids Health Dis. 2015, 14, 165. [Google Scholar] [CrossRef] [Green Version]
  57. Morgan, P.K.; Huynh, K.; Pernes, G.; Miotto, P.M.; Mellett, N.A.; Giles, C.; Meikle, P.J.; Murphy, A.J.; Lancaster, G.I. Macrophage polarization state affects lipid composition and the channeling of exogenous fatty acids into endogenous lipid pools. J. Biol. Chem. 2021, 297, 101341. [Google Scholar] [CrossRef] [PubMed]
  58. Benlachgar, N.; Doghmi, K.; Masrar, A.; Mahtat, E.M.; Harmouche, H.; Tazi Mezalek, Z. Immature platelets: A review of the available evidence. Thromb. Res. 2020, 195, 43–50. [Google Scholar] [CrossRef] [PubMed]
  59. Fan, Z.; McArdle, S.; Marki, A.; Mikulski, Z.; Gutierrez, E.; Engelhardt, B.; Deutsch, U.; Ginsberg, M.; Groisman, A.; Ley, K. Neutrophil recruitment limited by high-affinity bent beta2 integrin binding ligand in cis. Nat. Commun. 2016, 7, 12658. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Fan, Z.; Kiosses, W.B.; Sun, H.; Orecchioni, M.; Ghosheh, Y.; Zajonc, D.M.; Arnaout, M.A.; Gutierrez, E.; Groisman, A.; Ginsberg, M.H.; et al. High-Affinity Bent beta2-Integrin Molecules in Arresting Neutrophils Face Each Other through Binding to ICAMs In cis. Cell Rep. 2019, 26, 119–130.e115. [Google Scholar] [CrossRef] [Green Version]
  61. Liu, H.; Zhu, L.; Dudiki, T.; Gabanic, B.; Good, L.; Podrez, E.A.; Cherepanova, O.A.; Qin, J.; Byzova, T.V. Macrophage Migration and Phagocytosis Are Controlled by Kindlin-3’s Link to the Cytoskeleton. J. Immunol. 2020, 204, 1954–1967. [Google Scholar] [CrossRef]
  62. Xue, Z.H.; Feng, C.; Liu, W.L.; Tan, S.M. A role of kindlin-3 in integrin alphaMbeta2 outside-in signaling and the Syk-Vav1-Rac1/Cdc42 signaling axis. PLoS ONE 2013, 8, e56911. [Google Scholar] [CrossRef] [Green Version]
  63. Feng, C.; Li, Y.F.; Yau, Y.H.; Lee, H.S.; Tang, X.Y.; Xue, Z.H.; Zhou, Y.C.; Lim, W.M.; Cornvik, T.C.; Ruedl, C.; et al. Kindlin-3 mediates integrin alphaLbeta2 outside-in signaling, and it interacts with scaffold protein receptor for activated-C kinase 1 (RACK1). J. Biol. Chem. 2012, 287, 10714–10726. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Mocsai, A.; Walzog, B.; Lowell, C.A. Intracellular signalling during neutrophil recruitment. Cardiovasc. Res. 2015, 107, 373–385. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Sharma, P.; Vijaykumar, A.; Raghavan, J.V.; Rananaware, S.R.; Alakesh, A.; Bodele, J.; Rehman, J.U.; Shukla, S.; Wagde, V.; Nadig, S.; et al. Particle uptake driven phagocytosis in macrophages and neutrophils enhances bacterial clearance. J. Control. Release 2022, 343, 131–141. [Google Scholar] [CrossRef] [PubMed]
  66. Welin, A.; Winberg, M.E.; Abdalla, H.; Sarndahl, E.; Rasmusson, B.; Stendahl, O.; Lerm, M. Incorporation of Mycobacterium tuberculosis lipoarabinomannan into macrophage membrane rafts is a prerequisite for the phagosomal maturation block. Infect. Immun. 2008, 76, 2882–2887. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Cunin, P.; Nigrovic, P.A. Megakaryocytes as immune cells. J. Leukoc. Biol. 2019, 105, 1111–1121. [Google Scholar] [CrossRef]
  68. Cunin, P.; Bouslama, R.; Machlus, K.R.; Martinez-Bonet, M.; Lee, P.Y.; Wactor, A.; Nelson-Maney, N.; Morris, A.; Guo, L.; Weyrich, A.; et al. Megakaryocyte emperipolesis mediates membrane transfer from intracytoplasmic neutrophils to platelets. Elife 2019, 8, e44031. [Google Scholar] [CrossRef]
  69. Aroca-Crevillen, A.; Adrover, J.M.; Hidalgo, A. Circadian Features of Neutrophil Biology. Front. Immunol. 2020, 11, 576. [Google Scholar] [CrossRef]
  70. Nguyen, K.D.; Fentress, S.J.; Qiu, Y.; Yun, K.; Cox, J.S.; Chawla, A. Circadian gene Bmal1 regulates diurnal oscillations of Ly6C(hi) inflammatory monocytes. Science 2013, 341, 1483–1488. [Google Scholar] [CrossRef] [Green Version]
  71. Scheiermann, C.; Kunisaki, Y.; Lucas, D.; Chow, A.; Jang, J.E.; Zhang, D.; Hashimoto, D.; Merad, M.; Frenette, P.S. Adrenergic nerves govern circadian leukocyte recruitment to tissues. Immunity 2012, 37, 290–301. [Google Scholar] [CrossRef] [Green Version]
  72. He, W.; Holtkamp, S.; Hergenhan, S.M.; Kraus, K.; de Juan, A.; Weber, J.; Bradfield, P.; Grenier, J.M.P.; Pelletier, J.; Druzd, D.; et al. Circadian Expression of Migratory Factors Establishes Lineage-Specific Signatures that Guide the Homing of Leukocyte Subsets to Tissues. Immunity 2018, 49, 1175–1190.e1177. [Google Scholar] [CrossRef] [Green Version]
  73. Holtkamp, S.J.; Ince, L.M.; Barnoud, C.; Schmitt, M.T.; Sinturel, F.; Pilorz, V.; Pick, R.; Jemelin, S.; Muhlstadt, M.; Boehncke, W.H.; et al. Circadian clocks guide dendritic cells into skin lymphatics. Nat. Immunol. 2021, 22, 1375–1381. [Google Scholar] [CrossRef]
  74. Druzd, D.; Matveeva, O.; Ince, L.; Harrison, U.; He, W.; Schmal, C.; Herzel, H.; Tsang, A.H.; Kawakami, N.; Leliavski, A.; et al. Lymphocyte Circadian Clocks Control Lymph Node Trafficking and Adaptive Immune Responses. Immunity 2017, 46, 120–132. [Google Scholar] [CrossRef] [Green Version]
  75. Pick, R.; He, W.; Chen, C.S.; Scheiermann, C. Time-of-Day-Dependent Trafficking and Function of Leukocyte Subsets. Trends Immunol. 2019, 40, 524–537. [Google Scholar] [CrossRef] [Green Version]
  76. Colas, R.A.; Souza, P.R.; Walker, M.E.; Burton, M.; Zaslona, Z.; Curtis, A.M.; Marques, R.M.; Dalli, J. Impaired Production and Diurnal Regulation of Vascular RvDn-3 DPA Increase Systemic Inflammation and Cardiovascular Disease. Circ. Res. 2018, 122, 855–863. [Google Scholar] [CrossRef] [PubMed]
  77. Sperandio, M.; Quackenbush, E.J.; Sushkova, N.; Altstatter, J.; Nussbaum, C.; Schmid, S.; Pruenster, M.; Kurz, A.; Margraf, A.; Steppner, A.; et al. Ontogenetic regulation of leukocyte recruitment in mouse yolk sac vessels. Blood 2013, 121, e118–e128. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Shaw, A.C.; Goldstein, D.R.; Montgomery, R.R. Age-dependent dysregulation of innate immunity. Nat. Rev. Immunol. 2013, 13, 875–887. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Nikolich-Zugich, J. Author Correction: The twilight of immunity: Emerging concepts in aging of the immune system. Nat. Immunol. 2018, 19, 1146. [Google Scholar] [CrossRef]
  80. Barkaway, A.; Rolas, L.; Joulia, R.; Bodkin, J.; Lenn, T.; Owen-Woods, C.; Reglero-Real, N.; Stein, M.; Vazquez-Martinez, L.; Girbl, T.; et al. Age-related changes in the local milieu of inflamed tissues cause aberrant neutrophil trafficking and subsequent remote organ damage. Immunity 2021, 54, 1494–1510.e1497. [Google Scholar] [CrossRef]
  81. Nathan, P.; Gibbs, J.E.; Rainger, G.E.; Chimen, M. Changes in Circadian Rhythms Dysregulate Inflammation in Ageing: Focus on Leukocyte Trafficking. Front. Immunol. 2021, 12, 673405. [Google Scholar] [CrossRef]
  82. Kondratov, R.V.; Kondratova, A.A.; Gorbacheva, V.Y.; Vykhovanets, O.V.; Antoch, M.P. Early aging and age-related pathologies in mice deficient in BMAL1, the core componentof the circadian clock. Genes Dev. 2006, 20, 1868–1873. [Google Scholar] [CrossRef] [Green Version]
  83. Zhang, D.; Chen, G.; Manwani, D.; Mortha, A.; Xu, C.; Faith, J.J.; Burk, R.D.; Kunisaki, Y.; Jang, J.E.; Scheiermann, C.; et al. Neutrophil ageing is regulated by the microbiome. Nature 2015, 525, 528–532. [Google Scholar] [CrossRef] [PubMed]
  84. Uhl, B.; Vadlau, Y.; Zuchtriegel, G.; Nekolla, K.; Sharaf, K.; Gaertner, F.; Massberg, S.; Krombach, F.; Reichel, C.A. Aged neutrophils contribute to the first line of defense in the acute inflammatory response. Blood 2016, 128, 2327–2337. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Adrover, J.M.; Del Fresno, C.; Crainiciuc, G.; Cuartero, M.I.; Casanova-Acebes, M.; Weiss, L.A.; Huerga-Encabo, H.; Silvestre-Roig, C.; Rossaint, J.; Cossio, I.; et al. A Neutrophil Timer Coordinates Immune Defense and Vascular Protection. Immunity 2019, 50, 390–402.e310. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Dhabhar, F.S. Enhancing versus suppressive effects of stress on immune function: Implications for immunoprotection and immunopathology. Neuroimmunomodulation 2009, 16, 300–317. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Dhabhar, F.S.; McEwen, B.S. Enhancing versus suppressive effects of stress hormones on skin immune function. Proc. Natl. Acad. Sci. USA 1999, 96, 1059–1064. [Google Scholar] [CrossRef] [Green Version]
  88. McEwen, B.S. Protective and damaging effects of stress mediators. N. Engl. J. Med. 1998, 338, 171–179. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. Viswanathan, K.; Dhabhar, F.S. Stress-induced enhancement of leukocyte trafficking into sites of surgery or immune activation. Proc. Natl. Acad. Sci. USA 2005, 102, 5808–5813. [Google Scholar] [CrossRef] [Green Version]
  90. Maes, M.; Van Bockstaele, D.R.; Gastel, A.; Song, C.; Schotte, C.; Neels, H.; DeMeester, I.; Scharpe, S.; Janca, A. The effects of psychological stress on leukocyte subset distribution in humans: Evidence of immune activation. Neuropsychobiology 1999, 39, 1–9. [Google Scholar] [CrossRef] [PubMed]
  91. Ince, L.M.; Weber, J.; Scheiermann, C. Control of Leukocyte Trafficking by Stress-Associated Hormones. Front. Immunol. 2018, 9, 3143. [Google Scholar] [CrossRef] [Green Version]
  92. Cosentino, M.; Marino, F.; Maestroni, G.J. Sympathoadrenergic modulation of hematopoiesis: A review of available evidence and of therapeutic perspectives. Front. Cell Neurosci. 2015, 9, 302. [Google Scholar] [CrossRef]
  93. Buckley, C.D.; Ross, E.A.; McGettrick, H.M.; Osborne, C.E.; Haworth, O.; Schmutz, C.; Stone, P.C.; Salmon, M.; Matharu, N.M.; Vohra, R.K.; et al. Identification of a phenotypically and functionally distinct population of long-lived neutrophils in a model of reverse endothelial migration. J. Leukoc. Biol. 2006, 79, 303–311. [Google Scholar] [CrossRef] [PubMed]
  94. Grieshaber-Bouyer, R.; Radtke, F.A.; Cunin, P.; Stifano, G.; Levescot, A.; Vijaykumar, B.; Nelson-Maney, N.; Blaustein, R.B.; Monach, P.A.; Nigrovic, P.A.; et al. The neutrotime transcriptional signature defines a single continuum of neutrophils across biological compartments. Nat. Commun. 2021, 12, 2856. [Google Scholar] [CrossRef] [PubMed]
  95. Damazo, A.S.; Yona, S.; Flower, R.J.; Perretti, M.; Oliani, S.M. Spatial and temporal profiles for anti-inflammatory gene expression in leukocytes during a resolving model of peritonitis. J. Immunol. 2006, 176, 4410–4418. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Damazo, A.S.; Yona, S.; D’Acquisto, F.; Flower, R.J.; Oliani, S.M.; Perretti, M. Critical protective role for annexin 1 gene expression in the endotoxemic murine microcirculation. Am. J. Pathol. 2005, 166, 1607–1617. [Google Scholar] [CrossRef] [Green Version]
  97. Babbin, B.A.; Laukoetter, M.G.; Nava, P.; Koch, S.; Lee, W.Y.; Capaldo, C.T.; Peatman, E.; Severson, E.A.; Flower, R.J.; Perretti, M.; et al. Annexin A1 regulates intestinal mucosal injury, inflammation, and repair. J. Immunol. 2008, 181, 5035–5044. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Nusbaum, P.; Laine, C.; Bouaouina, M.; Seveau, S.; Cramer, E.M.; Masse, J.M.; Lesavre, P.; Halbwachs-Mecarelli, L. Distinct signaling pathways are involved in leukosialin (CD43) down-regulation, membrane blebbing, and phospholipid scrambling during neutrophil apoptosis. J. Biol. Chem. 2005, 280, 5843–5853. [Google Scholar] [CrossRef] [Green Version]
  99. Graziano, B.R.; Town, J.P.; Sitarska, E.; Nagy, T.L.; Fosnaric, M.; Penic, S.; Iglic, A.; Kralj-Iglic, V.; Gov, N.S.; Diz-Munoz, A.; et al. Cell confinement reveals a branched-actin independent circuit for neutrophil polarity. PLoS Biol. 2019, 17, e3000457. [Google Scholar] [CrossRef] [Green Version]
  100. Babiychuk, E.B.; Monastyrskaya, K.; Potez, S.; Draeger, A. Blebbing confers resistance against cell lysis. Cell Death Differ. 2011, 18, 80–89. [Google Scholar] [CrossRef] [Green Version]
  101. Guilliams, M.; Svedberg, F.R. Does tissue imprinting restrict macrophage plasticity? Nat. Immunol. 2021, 22, 118–127. [Google Scholar] [CrossRef]
  102. Goldsmith, H.L.; Quinn, T.A.; Drury, G.; Spanos, C.; McIntosh, F.A.; Simon, S.I. Dynamics of neutrophil aggregation in couette flow revealed by videomicroscopy: Effect of shear rate on two-body collision efficiency and doublet lifetime. Biophys. J. 2001, 81, 2020–2034. [Google Scholar] [CrossRef] [Green Version]
  103. Moazzam, F.; DeLano, F.A.; Zweifach, B.W.; Schmid-Schonbein, G.W. The leukocyte response to fluid stress. Proc. Natl. Acad. Sci. USA 1997, 94, 5338–5343. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Cougoule, C.; Van Goethem, E.; Le Cabec, V.; Lafouresse, F.; Dupre, L.; Mehraj, V.; Mege, J.L.; Lastrucci, C.; Maridonneau-Parini, I. Blood leukocytes and macrophages of various phenotypes have distinct abilities to form podosomes and to migrate in 3D environments. Eur. J. Cell Biol. 2012, 91, 938–949. [Google Scholar] [CrossRef] [PubMed]
  105. Renkawitz, J.; Sixt, M. Mechanisms of force generation and force transmission during interstitial leukocyte migration. EMBO Rep. 2010, 11, 744–750. [Google Scholar] [CrossRef] [Green Version]
  106. Renkawitz, J.; Kopf, A.; Stopp, J.; de Vries, I.; Driscoll, M.K.; Merrin, J.; Hauschild, R.; Welf, E.S.; Danuser, G.; Fiolka, R.; et al. Nuclear positioning facilitates amoeboid migration along the path of least resistance. Nature 2019, 568, 546–550. [Google Scholar] [CrossRef] [PubMed]
  107. Lupu, F.; Kinasewitz, G.; Dormer, K. The role of endothelial shear stress on haemodynamics, inflammation, coagulation and glycocalyx during sepsis. J. Cell Mol. Med. 2020, 24, 12258–12271. [Google Scholar] [CrossRef]
  108. Ploppa, A.; Schmidt, V.; Hientz, A.; Reutershan, J.; Haeberle, H.A.; Nohe, B. Mechanisms of leukocyte distribution during sepsis: An experimental study on the interdependence of cell activation, shear stress and endothelial injury. Crit. Care 2010, 14, R201. [Google Scholar] [CrossRef] [Green Version]
  109. Marki, A.; Buscher, K.; Lorenzini, C.; Meyer, M.; Saigusa, R.; Fan, Z.; Yeh, Y.T.; Hartmann, N.; Dan, J.M.; Kiosses, W.B.; et al. Elongated neutrophil-derived structures are blood-borne microparticles formed by rolling neutrophils during sepsis. J. Exp. Med. 2021, 218, e20200551. [Google Scholar] [CrossRef]
  110. Radley, G.; Ali, S.; Pieper, I.L.; Thornton, C.A. Mechanical shear stress and leukocyte phenotype and function: Implications for ventricular assist device development and use. Int. J. Artif. Organs 2019, 42, 133–142. [Google Scholar] [CrossRef]
  111. Margraf, A.; Ludwig, N.; Zarbock, A.; Rossaint, J. Systemic Inflammatory Response Syndrome After Surgery: Mechanisms and Protection. Anesth. Analg. 2020, 131, 1693–1707. [Google Scholar] [CrossRef]
  112. Schyns, J.; Bai, Q.; Ruscitti, C.; Radermecker, C.; De Schepper, S.; Chakarov, S.; Farnir, F.; Pirottin, D.; Ginhoux, F.; Boeckxstaens, G.; et al. Non-classical tissue monocytes and two functionally distinct populations of interstitial macrophages populate the mouse lung. Nat. Commun. 2019, 10, 3964. [Google Scholar] [CrossRef] [Green Version]
  113. Ley, K. M1 Means Kill; M2 Means Heal. J. Immunol. 2017, 199, 2191–2193. [Google Scholar] [CrossRef] [PubMed]
  114. Murray, P.J.; Allen, J.E.; Biswas, S.K.; Fisher, E.A.; Gilroy, D.W.; Goerdt, S.; Gordon, S.; Hamilton, J.A.; Ivashkiv, L.B.; Lawrence, T.; et al. Macrophage activation and polarization: Nomenclature and experimental guidelines. Immunity 2014, 41, 14–20. [Google Scholar] [CrossRef] [Green Version]
  115. Chavez-Galan, L.; Olleros, M.L.; Vesin, D.; Garcia, I. Much More than M1 and M2 Macrophages, There are also CD169(+) and TCR(+) Macrophages. Front. Immunol. 2015, 6, 263. [Google Scholar] [CrossRef]
  116. Rostam, H.M.; Reynolds, P.M.; Alexander, M.R.; Gadegaard, N.; Ghaemmaghami, A.M. Image based Machine Learning for identification of macrophage subsets. Sci. Rep. 2017, 7, 3521. [Google Scholar] [CrossRef] [PubMed]
  117. Krausgruber, T.; Blazek, K.; Smallie, T.; Alzabin, S.; Lockstone, H.; Sahgal, N.; Hussell, T.; Feldmann, M.; Udalova, I.A. IRF5 promotes inflammatory macrophage polarization and TH1-TH17 responses. Nat. Immunol. 2011, 12, 231–238. [Google Scholar] [CrossRef] [PubMed]
  118. Wang, N.; Liang, H.; Zen, K. Molecular mechanisms that influence the macrophage m1-m2 polarization balance. Front. Immunol. 2014, 5, 614. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  119. Han, M.S.; Jung, D.Y.; Morel, C.; Lakhani, S.A.; Kim, J.K.; Flavell, R.A.; Davis, R.J. JNK expression by macrophages promotes obesity-induced insulin resistance and inflammation. Science 2013, 339, 218–222. [Google Scholar] [CrossRef] [Green Version]
  120. Yao, Y.; Xu, X.H.; Jin, L. Macrophage Polarization in Physiological and Pathological Pregnancy. Front. Immunol. 2019, 10, 792. [Google Scholar] [CrossRef]
  121. Orecchioni, M.; Ghosheh, Y.; Pramod, A.B.; Ley, K. Corrigendum: Macrophage Polarization: Different Gene Signatures in M1(LPS+) vs. Classically and M2(LPS-) vs. Alternatively Activated Macrophages. Front. Immunol. 2020, 11, 234. [Google Scholar] [CrossRef]
  122. Wang, L.X.; Zhang, S.X.; Wu, H.J.; Rong, X.L.; Guo, J. M2b macrophage polarization and its roles in diseases. J. Leukoc. Biol. 2019, 106, 345–358. [Google Scholar] [CrossRef] [Green Version]
  123. Mehta, A.; Brewington, R.; Chatterji, M.; Zoubine, M.; Kinasewitz, G.T.; Peer, G.T.; Chang, A.C.; Taylor, F.B., Jr.; Shnyra, A. Infection-induced modulation of m1 and m2 phenotypes in circulating monocytes: Role in immune monitoring and early prognosis of sepsis. Shock 2004, 22, 423–430. [Google Scholar] [CrossRef] [PubMed]
  124. Benoit, M.; Desnues, B.; Mege, J.L. Macrophage polarization in bacterial infections. J. Immunol. 2008, 181, 3733–3739. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Liao, X.; Sharma, N.; Kapadia, F.; Zhou, G.; Lu, Y.; Hong, H.; Paruchuri, K.; Mahabeleshwar, G.H.; Dalmas, E.; Venteclef, N.; et al. Kruppel-like factor 4 regulates macrophage polarization. J. Clin. Investig. 2011, 121, 2736–2749. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Chen, X.; Tang, J.; Shuai, W.; Meng, J.; Feng, J.; Han, Z. Macrophage polarization and its role in the pathogenesis of acute lung injury/acute respiratory distress syndrome. Inflamm. Res. 2020, 69, 883–895. [Google Scholar] [CrossRef] [PubMed]
  127. Xing, L.; Zhongqian, L.; Chunmei, S.; Pingfa, C.; Lei, H.; Qin, J.; Genhua, M.; Yijun, D. Activation of M1 macrophages in sepsis-induced acute kidney injury in response to heparin-binding protein. PLoS ONE 2018, 13, e0196423. [Google Scholar] [CrossRef] [Green Version]
  128. Lv, L.L.; Tang, P.M.; Li, C.J.; You, Y.K.; Li, J.; Huang, X.R.; Ni, J.; Feng, M.; Liu, B.C.; Lan, H.Y. The pattern recognition receptor, Mincle, is essential for maintaining the M1 macrophage phenotype in acute renal inflammation. Kidney Int. 2017, 91, 587–602. [Google Scholar] [CrossRef]
  129. Xu, F.; Ma, Y.; Huang, W.; Gao, J.; Guo, M.; Li, J.; Kong, L.; Liang, G.; Du, R.; Xu, Q.; et al. Typically inhibiting USP14 promotes autophagy in M1-like macrophages and alleviates CLP-induced sepsis. Cell Death Dis. 2020, 11, 666. [Google Scholar] [CrossRef]
  130. Rackov, G.; Hernandez-Jimenez, E.; Shokri, R.; Carmona-Rodriguez, L.; Manes, S.; Alvarez-Mon, M.; Lopez-Collazo, E.; Martinez, A.C.; Balomenos, D. p21 mediates macrophage reprogramming through regulation of p50-p50 NF-kappaB and IFN-beta. J. Clin. Investig. 2016, 126, 3089–3103. [Google Scholar] [CrossRef]
  131. Watanabe, N.; Suzuki, Y.; Inokuchi, S.; Inoue, S. Sepsis induces incomplete M2 phenotype polarization in peritoneal exudate cells in mice. J. Intensive Care 2016, 4, 6. [Google Scholar] [CrossRef] [Green Version]
  132. Sica, A.; Mantovani, A. Macrophage plasticity and polarization: In vivo veritas. J. Clin. Investig. 2012, 122, 787–795. [Google Scholar] [CrossRef]
  133. Spite, M.; Norling, L.V.; Summers, L.; Yang, R.; Cooper, D.; Petasis, N.A.; Flower, R.J.; Perretti, M.; Serhan, C.N. Resolvin D2 is a potent regulator of leukocytes and controls microbial sepsis. Nature 2009, 461, 1287–1291. [Google Scholar] [CrossRef] [Green Version]
  134. Chiang, N.; Fredman, G.; Backhed, F.; Oh, S.F.; Vickery, T.; Schmidt, B.A.; Serhan, C.N. Infection regulates pro-resolving mediators that lower antibiotic requirements. Nature 2012, 484, 524–528. [Google Scholar] [CrossRef] [Green Version]
  135. Morita, M.; Kuba, K.; Ichikawa, A.; Nakayama, M.; Katahira, J.; Iwamoto, R.; Watanebe, T.; Sakabe, S.; Daidoji, T.; Nakamura, S.; et al. The lipid mediator protectin D1 inhibits influenza virus replication and improves severe influenza. Cell 2013, 153, 112–125. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Bang, S.; Donnelly, C.R.; Luo, X.; Toro-Moreno, M.; Tao, X.; Wang, Z.; Chandra, S.; Bortsov, A.V.; Derbyshire, E.R.; Ji, R.R. Activation of GPR37 in macrophages confers protection against infection-induced sepsis and pain-like behaviour in mice. Nat. Commun. 2021, 12, 1704. [Google Scholar] [CrossRef] [PubMed]
  137. Bang, S.; Xie, Y.K.; Zhang, Z.J.; Wang, Z.; Xu, Z.Z.; Ji, R.R. GPR37 regulates macrophage phagocytosis and resolution of inflammatory pain. J. Clin. Investig. 2018, 128, 3568–3582. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. Ruckerl, D.; Campbell, S.M.; Duncan, S.; Sutherland, T.E.; Jenkins, S.J.; Hewitson, J.P.; Barr, T.A.; Jackson-Jones, L.H.; Maizels, R.M.; Allen, J.E. Macrophage origin limits functional plasticity in helminth-bacterial co-infection. PLoS Pathog. 2017, 13, e1006233. [Google Scholar] [CrossRef] [PubMed]
  139. Schyns, J.; Bureau, F.; Marichal, T. Lung Interstitial Macrophages: Past, Present, and Future. J. Immunol. Res. 2018, 2018, 5160794. [Google Scholar] [CrossRef] [PubMed]
  140. Aran, D.; Looney, A.P.; Liu, L.; Wu, E.; Fong, V.; Hsu, A.; Chak, S.; Naikawadi, R.P.; Wolters, P.J.; Abate, A.R.; et al. Reference-based analysis of lung single-cell sequencing reveals a transitional profibrotic macrophage. Nat. Immunol. 2019, 20, 163–172. [Google Scholar] [CrossRef]
  141. Borges da Silva, H.; Fonseca, R.; Pereira, R.M.; Cassado Ados, A.; Alvarez, J.M.; D’Imperio Lima, M.R. Splenic Macrophage Subsets and Their Function during Blood-Borne Infections. Front. Immunol. 2015, 6, 480. [Google Scholar] [CrossRef] [Green Version]
  142. Ludewig, B.; Cervantes-Barragan, L. CD169(+) macrophages take the bullet. Nat. Immunol. 2011, 13, 13–14. [Google Scholar] [CrossRef]
  143. Honke, N.; Shaabani, N.; Cadeddu, G.; Sorg, U.R.; Zhang, D.E.; Trilling, M.; Klingel, K.; Sauter, M.; Kandolf, R.; Gailus, N.; et al. Enforced viral replication activates adaptive immunity and is essential for the control of a cytopathic virus. Nat. Immunol. 2011, 13, 51–57. [Google Scholar] [CrossRef] [PubMed]
  144. Van der Heide, D.; Weiskirchen, R.; Bansal, R. Therapeutic Targeting of Hepatic Macrophages for the Treatment of Liver Diseases. Front. Immunol. 2019, 10, 2852. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Triantafyllou, E.; Pop, O.T.; Possamai, L.A.; Wilhelm, A.; Liaskou, E.; Singanayagam, A.; Bernsmeier, C.; Khamri, W.; Petts, G.; Dargue, R.; et al. MerTK expressing hepatic macrophages promote the resolution of inflammation in acute liver failure. Gut 2018, 67, 333–347. [Google Scholar] [CrossRef] [PubMed]
  146. MacParland, S.A.; Liu, J.C.; Ma, X.Z.; Innes, B.T.; Bartczak, A.M.; Gage, B.K.; Manuel, J.; Khuu, N.; Echeverri, J.; Linares, I.; et al. Single cell RNA sequencing of human liver reveals distinct intrahepatic macrophage populations. Nat. Commun. 2018, 9, 4383. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Hulsmans, M.; Sager, H.B.; Roh, J.D.; Valero-Munoz, M.; Houstis, N.E.; Iwamoto, Y.; Sun, Y.; Wilson, R.M.; Wojtkiewicz, G.; Tricot, B.; et al. Cardiac macrophages promote diastolic dysfunction. J. Exp. Med. 2018, 215, 423–440. [Google Scholar] [CrossRef]
  148. Hulsmans, M.; Clauss, S.; Xiao, L.; Aguirre, A.D.; King, K.R.; Hanley, A.; Hucker, W.J.; Wulfers, E.M.; Seemann, G.; Courties, G.; et al. Macrophages Facilitate Electrical Conduction in the Heart. Cell 2017, 169, 510–522.e520. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  149. Chakarov, S.; Lim, H.Y.; Tan, L.; Lim, S.Y.; See, P.; Lum, J.; Zhang, X.M.; Foo, S.; Nakamizo, S.; Duan, K.; et al. Two distinct interstitial macrophage populations coexist across tissues in specific subtissular niches. Science 2019, 363, eaau0964. [Google Scholar] [CrossRef]
  150. Dick, S.A.; Wong, A.; Hamidzada, H.; Nejat, S.; Nechanitzky, R.; Vohra, S.; Mueller, B.; Zaman, R.; Kantores, C.; Aronoff, L.; et al. Three tissue resident macrophage subsets coexist across organs with conserved origins and life cycles. Sci. Immunol. 2022, 7, eabf7777. [Google Scholar] [CrossRef]
  151. Hoyer, F.F.; Naxerova, K.; Schloss, M.J.; Hulsmans, M.; Nair, A.V.; Dutta, P.; Calcagno, D.M.; Herisson, F.; Anzai, A.; Sun, Y.; et al. Tissue-Specific Macrophage Responses to Remote Injury Impact the Outcome of Subsequent Local Immune Challenge. Immunity 2019, 51, 899–914.e897. [Google Scholar] [CrossRef]
  152. Gomez Perdiguero, E.; Klapproth, K.; Schulz, C.; Busch, K.; Azzoni, E.; Crozet, L.; Garner, H.; Trouillet, C.; de Bruijn, M.F.; Geissmann, F.; et al. Tissue-resident macrophages originate from yolk-sac-derived erythro-myeloid progenitors. Nature 2015, 518, 547–551. [Google Scholar] [CrossRef]
  153. Stremmel, C.; Schuchert, R.; Wagner, F.; Thaler, R.; Weinberger, T.; Pick, R.; Mass, E.; Ishikawa-Ankerhold, H.C.; Margraf, A.; Hutter, S.; et al. Author Correction: Yolk sac macrophage progenitors traffic to the embryo during defined stages of development. Nat. Commun. 2018, 9, 3699. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Honold, L.; Nahrendorf, M. Resident and Monocyte-Derived Macrophages in Cardiovascular Disease. Circ. Res. 2018, 122, 113–127. [Google Scholar] [CrossRef] [PubMed]
  155. Masuda, T.; Amann, L.; Monaco, G.; Sankowski, R.; Staszewski, O.; Krueger, M.; Del Gaudio, F.; He, L.; Paterson, N.; Nent, E.; et al. Specification of CNS macrophage subsets occurs postnatally in defined niches. Nature 2022, 604, 740–748. [Google Scholar] [CrossRef] [PubMed]
  156. Hong, C.W. Current Understanding in Neutrophil Differentiation and Heterogeneity. Immune Netw. 2017, 17, 298–306. [Google Scholar] [CrossRef] [Green Version]
  157. Silvestre-Roig, C.; Hidalgo, A.; Soehnlein, O. Neutrophil heterogeneity: Implications for homeostasis and pathogenesis. Blood 2016, 127, 2173–2181. [Google Scholar] [CrossRef] [Green Version]
  158. Fridlender, Z.G.; Sun, J.; Kim, S.; Kapoor, V.; Cheng, G.; Ling, L.; Worthen, G.S.; Albelda, S.M. Polarization of tumor-associated neutrophil phenotype by TGF-beta: “N1” versus “N2” TAN. Cancer Cell 2009, 16, 183–194. [Google Scholar] [CrossRef] [Green Version]
  159. Ohms, M.; Moller, S.; Laskay, T. An Attempt to Polarize Human Neutrophils Toward N1 and N2 Phenotypes in vitro. Front. Immunol. 2020, 11, 532. [Google Scholar] [CrossRef]
  160. Cuartero, M.I.; Ballesteros, I.; Moraga, A.; Nombela, F.; Vivancos, J.; Hamilton, J.A.; Corbi, A.L.; Lizasoain, I.; Moro, M.A. N2 neutrophils, novel players in brain inflammation after stroke: Modulation by the PPARgamma agonist rosiglitazone. Stroke 2013, 44, 3498–3508. [Google Scholar] [CrossRef] [Green Version]
  161. Ma, Y.; Yabluchanskiy, A.; Iyer, R.P.; Cannon, P.L.; Flynn, E.R.; Jung, M.; Henry, J.; Cates, C.A.; Deleon-Pennell, K.Y.; Lindsey, M.L. Temporal neutrophil polarization following myocardial infarction. Cardiovasc. Res. 2016, 110, 51–61. [Google Scholar] [CrossRef] [Green Version]
  162. Headland, S.E.; Jones, H.R.; Norling, L.V.; Kim, A.; Souza, P.R.; Corsiero, E.; Gil, C.D.; Nerviani, A.; Dell’Accio, F.; Pitzalis, C.; et al. Neutrophil-derived microvesicles enter cartilage and protect the joint in inflammatory arthritis. Sci. Transl. Med. 2015, 7, 315ra190. [Google Scholar] [CrossRef]
  163. Scapini, P.; Marini, O.; Tecchio, C.; Cassatella, M.A. Human neutrophils in the saga of cellular heterogeneity: Insights and open questions. Immunol. Rev. 2016, 273, 48–60. [Google Scholar] [CrossRef] [PubMed]
  164. Hardisty, G.R.; Llanwarne, F.; Minns, D.; Gillan, J.L.; Davidson, D.J.; Gwyer Findlay, E.; Gray, R.D. High Purity Isolation of Low Density Neutrophils Casts Doubt on Their Exceptionality in Health and Disease. Front. Immunol. 2021, 12, 625922. [Google Scholar] [CrossRef] [PubMed]
  165. Morrissey, S.M.; Geller, A.E.; Hu, X.; Tieri, D.; Ding, C.; Klaes, C.K.; Cooke, E.A.; Woeste, M.R.; Martin, Z.C.; Chen, O.; et al. A specific low-density neutrophil population correlates with hypercoagulation and disease severity in hospitalized COVID-19 patients. JCI Insight 2021, 6, e148435. [Google Scholar] [CrossRef] [PubMed]
  166. Evrard, M.; Kwok, I.W.H.; Chong, S.Z.; Teng, K.W.W.; Becht, E.; Chen, J.; Sieow, J.L.; Penny, H.L.; Ching, G.C.; Devi, S.; et al. Developmental Analysis of Bone Marrow Neutrophils Reveals Populations Specialized in Expansion, Trafficking, and Effector Functions. Immunity 2018, 48, 364–379.e368. [Google Scholar] [CrossRef] [Green Version]
  167. Guerin, E.; Orabona, M.; Raquil, M.A.; Giraudeau, B.; Bellier, R.; Gibot, S.; Bene, M.C.; Lacombe, F.; Droin, N.; Solary, E.; et al. Circulating immature granulocytes with T-cell killing functions predict sepsis deterioration*. Crit. Care Med. 2014, 42, 2007–2018. [Google Scholar] [CrossRef]
  168. Pillay, J.; Kamp, V.M.; van Hoffen, E.; Visser, T.; Tak, T.; Lammers, J.W.; Ulfman, L.H.; Leenen, L.P.; Pickkers, P.; Koenderman, L. A subset of neutrophils in human systemic inflammation inhibits T cell responses through Mac-1. J. Clin. Investig. 2012, 122, 327–336. [Google Scholar] [CrossRef]
  169. Kamp, V.M.; Pillay, J.; Lammers, J.W.; Pickkers, P.; Ulfman, L.H.; Koenderman, L. Human suppressive neutrophils CD16bright/CD62Ldim exhibit decreased adhesion. J. Leukoc. Biol. 2012, 92, 1011–1020. [Google Scholar] [CrossRef]
  170. Millrud, C.R.; Kagedal, A.; Kumlien Georen, S.; Winqvist, O.; Uddman, R.; Razavi, R.; Munck-Wikland, E.; Cardell, L.O. NET-producing CD16(high) CD62L(dim) neutrophils migrate to tumor sites and predict improved survival in patients with HNSCC. Int. J. Cancer 2017, 140, 2557–2567. [Google Scholar] [CrossRef] [Green Version]
  171. Pillay, J.; Ramakers, B.P.; Kamp, V.M.; Loi, A.L.; Lam, S.W.; Hietbrink, F.; Leenen, L.P.; Tool, A.T.; Pickkers, P.; Koenderman, L. Functional heterogeneity and differential priming of circulating neutrophils in human experimental endotoxemia. J. Leukoc. Biol. 2010, 88, 211–220. [Google Scholar] [CrossRef]
  172. Grieshaber-Bouyer, R.; Nigrovic, P.A. Neutrophil Heterogeneity as Therapeutic Opportunity in Immune-Mediated Disease. Front. Immunol. 2019, 10, 346. [Google Scholar] [CrossRef] [Green Version]
  173. Bai, M.; Grieshaber-Bouyer, R.; Wang, J.; Schmider, A.B.; Wilson, Z.S.; Zeng, L.; Halyabar, O.; Godin, M.D.; Nguyen, H.N.; Levescot, A.; et al. CD177 modulates human neutrophil migration through activation-mediated integrin and chemoreceptor regulation. Blood 2017, 130, 2092–2100. [Google Scholar] [CrossRef] [PubMed]
  174. Levy, Y.; Wiedemann, A.; Hejblum, B.P.; Durand, M.; Lefebvre, C.; Surenaud, M.; Lacabaratz, C.; Perreau, M.; Foucat, E.; Dechenaud, M.; et al. CD177, a specific marker of neutrophil activation, is associated with coronavirus disease 2019 severity and death. iScience 2021, 24, 102711. [Google Scholar] [CrossRef] [PubMed]
  175. Zhou, G.; Yu, L.; Fang, L.; Yang, W.; Yu, T.; Miao, Y.; Chen, M.; Wu, K.; Chen, F.; Cong, Y.; et al. CD177(+) neutrophils as functionally activated neutrophils negatively regulate IBD. Gut 2018, 67, 1052–1063. [Google Scholar] [CrossRef] [PubMed]
  176. Nourshargh, S.; Renshaw, S.A.; Imhof, B.A. Reverse Migration of Neutrophils: Where, When, How, and Why? Trends Immunol. 2016, 37, 273–286. [Google Scholar] [CrossRef]
  177. Ji, J.; Fan, J. Neutrophil in Reverse Migration: Role in Sepsis. Front. Immunol. 2021, 12, 656039. [Google Scholar] [CrossRef]
  178. Fischer, J.; Walter, C.; Tonges, A.; Aleth, H.; Jordao, M.J.C.; Leddin, M.; Groning, V.; Erdmann, T.; Lenz, G.; Roth, J.; et al. Safeguard function of PU.1 shapes the inflammatory epigenome of neutrophils. Nat. Immunol. 2019, 20, 546–558. [Google Scholar] [CrossRef]
  179. Kienle, K.; Glaser, K.M.; Eickhoff, S.; Mihlan, M.; Knopper, K.; Reategui, E.; Epple, M.W.; Gunzer, M.; Baumeister, R.; Tarrant, T.K.; et al. Neutrophils self-limit swarming to contain bacterial growth in vivo. Science 2021, 372, eabe7729. [Google Scholar] [CrossRef]
  180. Lammermann, T.; Afonso, P.V.; Angermann, B.R.; Wang, J.M.; Kastenmuller, W.; Parent, C.A.; Germain, R.N. Neutrophil swarms require LTB4 and integrins at sites of cell death in vivo. Nature 2013, 498, 371–375. [Google Scholar] [CrossRef]
  181. Adrover, J.M.; Aroca-Crevillen, A.; Crainiciuc, G.; Ostos, F.; Rojas-Vega, Y.; Rubio-Ponce, A.; Cilloniz, C.; Bonzon-Kulichenko, E.; Calvo, E.; Rico, D.; et al. Programmed ‘disarming’ of the neutrophil proteome reduces the magnitude of inflammation. Nat. Immunol. 2020, 21, 135–144. [Google Scholar] [CrossRef]
  182. Xie, X.; Shi, Q.; Wu, P.; Zhang, X.; Kambara, H.; Su, J.; Yu, H.; Park, S.Y.; Guo, R.; Ren, Q.; et al. Single-cell transcriptome profiling reveals neutrophil heterogeneity in homeostasis and infection. Nat. Immunol. 2020, 21, 1119–1133. [Google Scholar] [CrossRef]
  183. Leslie, M. Cell biology. Beyond clotting: The powers of platelets. Science 2010, 328, 562–564. [Google Scholar] [CrossRef] [PubMed]
  184. Rossaint, J.; Margraf, A.; Zarbock, A. Role of Platelets in Leukocyte Recruitment and Resolution of Inflammation. Front. Immunol. 2018, 9, 2712. [Google Scholar] [CrossRef] [PubMed]
  185. Nicolai, L.; Massberg, S. Platelets as key players in inflammation and infection. Curr. Opin. Hematol. 2020, 27, 34–40. [Google Scholar] [CrossRef] [PubMed]
  186. Karpatkin, S. Heterogeneity of human platelets. II. Functional evidence suggestive of young and old platelets. J. Clin. Investig. 1969, 48, 1083–1087. [Google Scholar] [CrossRef] [PubMed]
  187. Lesyk, G.; Jurasz, P. Advances in Platelet Subpopulation Research. Front. Cardiovasc. Med. 2019, 6, 138. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  188. Moraes, L.A.; Paul-Clark, M.J.; Rickman, A.; Flower, R.J.; Goulding, N.J.; Perretti, M. Ligand-specific glucocorticoid receptor activation in human platelets. Blood 2005, 106, 4167–4175. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  189. Liverani, E.; Banerjee, S.; Roberts, W.; Naseem, K.M.; Perretti, M. Prednisolone exerts exquisite inhibitory properties on platelet functions. Biochem. Pharmacol. 2012, 83, 1364–1373. [Google Scholar] [CrossRef] [Green Version]
  190. Lindemann, S.; Tolley, N.D.; Dixon, D.A.; McIntyre, T.M.; Prescott, S.M.; Zimmerman, G.A.; Weyrich, A.S. Activated platelets mediate inflammatory signaling by regulated interleukin 1beta synthesis. J. Cell Biol. 2001, 154, 485–490. [Google Scholar] [CrossRef] [Green Version]
  191. Manne, B.K.; Xiang, S.C.; Rondina, M.T. Platelet secretion in inflammatory and infectious diseases. Platelets 2017, 28, 155–164. [Google Scholar] [CrossRef]
  192. Oggero, S.; de Gaetano, M.; Marcone, S.; Fitzsimons, S.; Pinto, A.L.; Ikramova, D.; Barry, M.; Burke, D.; Montero-Melendez, T.; Cooper, D.; et al. Extracellular vesicles from monocyte/platelet aggregates modulate human atherosclerotic plaque reactivity. J. Extracell. Vesicles 2021, 10, 12084. [Google Scholar] [CrossRef]
  193. Boilard, E.; Nigrovic, P.A.; Larabee, K.; Watts, G.F.; Coblyn, J.S.; Weinblatt, M.E.; Massarotti, E.M.; Remold-O’Donnell, E.; Farndale, R.W.; Ware, J.; et al. Platelets amplify inflammation in arthritis via collagen-dependent microparticle production. Science 2010, 327, 580–583. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  194. Stone, A.P.; Nikols, E.; Freire, D.; Machlus, K.R. The pathobiology of platelet and megakaryocyte extracellular vesicles: A (c)lot has changed. J. Thromb. Haemost. 2022. [Google Scholar] [CrossRef] [PubMed]
  195. Puhm, F.; Boilard, E.; Machlus, K.R. Platelet Extracellular Vesicles: Beyond the Blood. Arter. Thromb. Vasc. Biol. 2021, 41, 87–96. [Google Scholar] [CrossRef] [PubMed]
  196. Pick, M.; Perry, C.; Lapidot, T.; Guimaraes-Sternberg, C.; Naparstek, E.; Deutsch, V.; Soreq, H. Stress-induced cholinergic signaling promotes inflammation-associated thrombopoiesis. Blood 2006, 107, 3397–3406. [Google Scholar] [CrossRef] [Green Version]
  197. Kaser, A.; Brandacher, G.; Steurer, W.; Kaser, S.; Offner, F.A.; Zoller, H.; Theurl, I.; Widder, W.; Molnar, C.; Ludwiczek, O.; et al. Interleukin-6 stimulates thrombopoiesis through thrombopoietin: Role in inflammatory thrombocytosis. Blood 2001, 98, 2720–2725. [Google Scholar] [CrossRef] [Green Version]
  198. Koyama, K.; Katayama, S.; Muronoi, T.; Tonai, K.; Goto, Y.; Koinuma, T.; Shima, J.; Nunomiya, S. Time course of immature platelet count and its relation to thrombocytopenia and mortality in patients with sepsis. PLoS ONE 2018, 13, e0192064. [Google Scholar] [CrossRef] [Green Version]
  199. Park, S.H.; Ha, S.O.; Cho, Y.U.; Park, C.J.; Jang, S.; Hong, S.B. Immature platelet fraction in septic patients: Clinical relevance of immature platelet fraction is limited to the sensitive and accurate discrimination of septic patients from non-septic patients, not to the discrimination of sepsis severity. Ann. Lab. Med. 2016, 36, 1–8. [Google Scholar] [CrossRef] [Green Version]
  200. Enz Hubert, R.M.; Rodrigues, M.V.; Andreguetto, B.D.; Santos, T.M.; de Fatima Pereira Gilberti, M.; de Castro, V.; Annichino-Bizzacchi, J.M.; Dragosavac, D.; Carvalho-Filho, M.A.; De Paula, E.V. Association of the immature platelet fraction with sepsis diagnosis and severity. Sci. Rep. 2015, 5, 8019. [Google Scholar] [CrossRef] [Green Version]
  201. Imperiali, C.E.; Lopez-Delgado, J.C.; Dastis-Arias, M.; Sanchez-Navarro, L. Evaluation of the delta of immature platelet fraction as a predictive biomarker of inflammatory response after cardiac surgery. J. Clin. Pathol. 2020, 73, 335–340. [Google Scholar] [CrossRef]
  202. Nishimura, S.; Nagasaki, M.; Kunishima, S.; Sawaguchi, A.; Sakata, A.; Sakaguchi, H.; Ohmori, T.; Manabe, I.; Italiano, J.E., Jr.; Ryu, T.; et al. IL-1alpha induces thrombopoiesis through megakaryocyte rupture in response to acute platelet needs. J. Cell Biol. 2015, 209, 453–466. [Google Scholar] [CrossRef] [Green Version]
  203. Lefrancais, E.; Ortiz-Munoz, G.; Caudrillier, A.; Mallavia, B.; Liu, F.C.; Sayah, D.M.; Thornton, E.E.; Headley, M.B.; David, T.; Coughlin, S.R.; et al. The lung is a site of platelet biogenesis and a reservoir for haematopoietic progenitors. Nature 2017, 544, 105–109. [Google Scholar] [CrossRef] [PubMed]
  204. Peng, J.; Friese, P.; Heilmann, E.; George, J.N.; Burstein, S.A.; Dale, G.L. Aged platelets have an impaired response to thrombin as quantitated by P-selectin expression. Blood 1994, 83, 161–166. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  205. Allan, H.E.; Hayman, M.A.; Marcone, S.; Chan, M.V.; Edin, M.L.; Maffucci, T.; Joshi, A.; Menke, L.; Crescente, M.; Mayr, M.; et al. Proteome and functional decline as platelets age in the circulation. J. Thromb. Haemost. 2021, 19, 3095–3112. [Google Scholar] [CrossRef] [PubMed]
  206. Grozovsky, R.; Begonja, A.J.; Liu, K.; Visner, G.; Hartwig, J.H.; Falet, H.; Hoffmeister, K.M. The Ashwell-Morell receptor regulates hepatic thrombopoietin production via JAK2-STAT3 signaling. Nat. Med. 2015, 21, 47–54. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  207. Kile, B.T. Aging platelets stimulate TPO production. Nat. Med. 2015, 21, 11–12. [Google Scholar] [CrossRef]
  208. Radziwon-Balicka, A.; Lesyk, G.; Back, V.; Fong, T.; Loredo-Calderon, E.L.; Dong, B.; El-Sikhry, H.; El-Sherbeni, A.A.; El-Kadi, A.; Ogg, S.; et al. Differential eNOS-signalling by platelet subpopulations regulates adhesion and aggregation. Cardiovasc. Res. 2017, 113, 1719–1731. [Google Scholar] [CrossRef]
  209. Prodan, C.I.; Joseph, P.M.; Vincent, A.S.; Dale, G.L. Coated-platelets in ischemic stroke: Differences between lacunar and cortical stroke. J. Thromb. Haemost. 2008, 6, 609–614. [Google Scholar] [CrossRef]
  210. Alberio, L.; Safa, O.; Clemetson, K.J.; Esmon, C.T.; Dale, G.L. Surface expression and functional characterization of alpha-granule factor V in human platelets: Effects of ionophore A23187, thrombin, collagen, and convulxin. Blood 2000, 95, 1694–1702. [Google Scholar] [CrossRef]
  211. Dale, G.L.; Friese, P.; Batar, P.; Hamilton, S.F.; Reed, G.L.; Jackson, K.W.; Clemetson, K.J.; Alberio, L. Stimulated platelets use serotonin to enhance their retention of procoagulant proteins on the cell surface. Nature 2002, 415, 175–179. [Google Scholar] [CrossRef]
  212. Hindle, M.S.; Spurgeon, B.E.J.; Cheah, L.T.; Webb, B.A.; Naseem, K.M. Multidimensional flow cytometry reveals novel platelet subpopulations in response to prostacyclin. J. Thromb. Haemost. 2021, 19, 1800–1812. [Google Scholar] [CrossRef]
  213. Dunster, J.L.; Bye, A.P.; Kriek, N.; Sage, T.; Mitchell, J.L.; Kempster, C.; Batista, J.; McKinney, H.; Thomas, P.; Jones, C.I.; et al. Multiparameter phenotyping of platelet reactivity for stratification of human cohorts. Blood Adv. 2021, 5, 4017–4030. [Google Scholar] [CrossRef] [PubMed]
  214. Aslan, J.E. Platelet Proteomes, Pathways, and Phenotypes as Informants of Vascular Wellness and Disease. Arter. Thromb. Vasc. Biol. 2021, 41, 999–1011. [Google Scholar] [CrossRef] [PubMed]
  215. Senchenkova, E.Y.; Ansari, J.; Becker, F.; Vital, S.A.; Al-Yafeai, Z.; Sparkenbaugh, E.M.; Pawlinski, R.; Stokes, K.Y.; Carroll, J.L.; Dragoi, A.M.; et al. Novel Role for the AnxA1-Fpr2/ALX Signaling Axis as a Key Regulator of Platelet Function to Promote Resolution of Inflammation. Circulation 2019, 140, 319–335. [Google Scholar] [CrossRef]
  216. Salamah, M.F.; Ravishankar, D.; Kodji, X.; Moraes, L.A.; Williams, H.F.; Vallance, T.M.; Albadawi, D.A.; Vaiyapuri, R.; Watson, K.; Gibbins, J.M.; et al. The endogenous antimicrobial cathelicidin LL37 induces platelet activation and augments thrombus formation. Blood Adv. 2018, 2, 2973–2985. [Google Scholar] [CrossRef] [PubMed]
  217. Lefrancais, E.; Looney, M.R. Platelet Biogenesis in the Lung Circulation. Physiology (Bethesda) 2019, 34, 392–401. [Google Scholar] [CrossRef] [PubMed]
  218. Masucci, M.T.; Minopoli, M.; Carriero, M.V. Tumor Associated Neutrophils. Their Role in Tumorigenesis, Metastasis, Prognosis and Therapy. Front. Oncol. 2019, 9, 1146. [Google Scholar] [CrossRef] [Green Version]
  219. Lapenna, A.; De Palma, M.; Lewis, C.E. Author Correction: Perivascular macrophages in health and disease. Nat. Rev. Immunol. 2021, 21, 752. [Google Scholar] [CrossRef]
  220. Hughes, C.E.; Nibbs, R.J.B. A guide to chemokines and their receptors. FEBS J. 2018, 285, 2944–2971. [Google Scholar] [CrossRef]
  221. Jin, J.; Xiao, Y.; Hu, H.; Zou, Q.; Li, Y.; Gao, Y.; Ge, W.; Cheng, X.; Sun, S.C. Proinflammatory TLR signalling is regulated by a TRAF2-dependent proteolysis mechanism in macrophages. Nat. Commun. 2015, 6, 5930. [Google Scholar] [CrossRef] [Green Version]
  222. Swanson, L.; Katkar, G.D.; Tam, J.; Pranadinata, R.F.; Chareddy, Y.; Coates, J.; Anandachar, M.S.; Castillo, V.; Olson, J.; Nizet, V.; et al. TLR4 signaling and macrophage inflammatory responses are dampened by GIV/Girdin. Proc. Natl. Acad. Sci. USA 2020, 117, 26895–26906. [Google Scholar] [CrossRef]
  223. Nadra, I.; Mason, J.C.; Philippidis, P.; Florey, O.; Smythe, C.D.; McCarthy, G.M.; Landis, R.C.; Haskard, D.O. Proinflammatory activation of macrophages by basic calcium phosphate crystals via protein kinase C and MAP kinase pathways: A vicious cycle of inflammation and arterial calcification? Circ. Res. 2005, 96, 1248–1256. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  224. Wang, J.; Kubes, P. A Reservoir of Mature Cavity Macrophages that Can Rapidly Invade Visceral Organs to Affect Tissue Repair. Cell 2016, 165, 668–678. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  225. Okabe, Y.; Medzhitov, R. Tissue-specific signals control reversible program of localization and functional polarization of macrophages. Cell 2014, 157, 832–844. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  226. Ruffell, D.; Mourkioti, F.; Gambardella, A.; Kirstetter, P.; Lopez, R.G.; Rosenthal, N.; Nerlov, C. A CREB-C/EBPbeta cascade induces M2 macrophage-specific gene expression and promotes muscle injury repair. Proc. Natl. Acad. Sci. USA 2009, 106, 17475–17480. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  227. Lawrence, T.; Natoli, G. Transcriptional regulation of macrophage polarization: Enabling diversity with identity. Nat. Rev. Immunol. 2011, 11, 750–761. [Google Scholar] [CrossRef] [PubMed]
  228. Cheung, P.; Schaffert, S.; Chang, S.E.; Dvorak, M.; Donato, M.; Macaubas, C.; Foecke, M.H.; Li, T.M.; Zhang, L.; Coan, J.P.; et al. Repression of CTSG, ELANE and PRTN3-mediated histone H3 proteolytic cleavage promotes monocyte-to-macrophage differentiation. Nat. Immunol. 2021, 22, 711–722. [Google Scholar] [CrossRef]
  229. Gabanyi, I.; Muller, P.A.; Feighery, L.; Oliveira, T.Y.; Costa-Pinto, F.A.; Mucida, D. Neuro-immune Interactions Drive Tissue Programming in Intestinal Macrophages. Cell 2016, 164, 378–391. [Google Scholar] [CrossRef] [Green Version]
  230. Lavin, Y.; Winter, D.; Blecher-Gonen, R.; David, E.; Keren-Shaul, H.; Merad, M.; Jung, S.; Amit, I. Tissue-resident macrophage enhancer landscapes are shaped by the local microenvironment. Cell 2014, 159, 1312–1326. [Google Scholar] [CrossRef] [Green Version]
  231. Gosselin, D.; Link, V.M.; Romanoski, C.E.; Fonseca, G.J.; Eichenfield, D.Z.; Spann, N.J.; Stender, J.D.; Chun, H.B.; Garner, H.; Geissmann, F.; et al. Environment drives selection and function of enhancers controlling tissue-specific macrophage identities. Cell 2014, 159, 1327–1340. [Google Scholar] [CrossRef]
  232. Orecchioni, M.; Kobiyama, K.; Winkels, H.; Ghosheh, Y.; McArdle, S.; Mikulski, Z.; Kiosses, W.B.; Fan, Z.; Wen, L.; Jung, Y.; et al. Olfactory receptor 2 in vascular macrophages drives atherosclerosis by NLRP3-dependent IL-1 production. Science 2022, 375, 214–221. [Google Scholar] [CrossRef]
  233. Woytschak, J.; Keller, N.; Krieg, C.; Impellizzieri, D.; Thompson, R.W.; Wynn, T.A.; Zinkernagel, A.S.; Boyman, O. Type 2 Interleukin-4 Receptor Signaling in Neutrophils Antagonizes Their Expansion and Migration during Infection and Inflammation. Immunity 2016, 45, 172–184. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  234. Balasubbramanian, D.; Goodlett, B.L.; Mitchell, B.M. Is IL-12 pro-inflammatory or anti-inflammatory? Depends on the blood pressure. Cardiovasc. Res. 2019, 115, 998–999. [Google Scholar] [CrossRef] [Green Version]
  235. Ye, J.; Que, B.; Huang, Y.; Lin, Y.; Chen, J.; Liu, L.; Shi, Y.; Wang, Y.; Wang, M.; Zeng, T.; et al. Interleukin-12p35 knockout promotes macrophage differentiation, aggravates vascular dysfunction, and elevates blood pressure in angiotensin II-infused mice. Cardiovasc. Res. 2019, 115, 1102–1113. [Google Scholar] [CrossRef] [PubMed]
  236. Kourtzelis, I.; Li, X.; Mitroulis, I.; Grosser, D.; Kajikawa, T.; Wang, B.; Grzybek, M.; von Renesse, J.; Czogalla, A.; Troullinaki, M.; et al. DEL-1 promotes macrophage efferocytosis and clearance of inflammation. Nat. Immunol. 2019, 20, 40–49. [Google Scholar] [CrossRef] [PubMed]
  237. Jetten, N.; Roumans, N.; Gijbels, M.J.; Romano, A.; Post, M.J.; de Winther, M.P.; van der Hulst, R.R.; Xanthoulea, S. Wound administration of M2-polarized macrophages does not improve murine cutaneous healing responses. PLoS ONE 2014, 9, e102994. [Google Scholar] [CrossRef] [Green Version]
  238. Neupane, A.S.; Willson, M.; Chojnacki, A.K.; Vargas, E.S.C.F.; Morehouse, C.; Carestia, A.; Keller, A.E.; Peiseler, M.; DiGiandomenico, A.; Kelly, M.M.; et al. Patrolling Alveolar Macrophages Conceal Bacteria from the Immune System to Maintain Homeostasis. Cell 2020, 183, 110–125.e111. [Google Scholar] [CrossRef] [PubMed]
  239. Uderhardt, S.; Martins, A.J.; Tsang, J.S.; Lammermann, T.; Germain, R.N. Resident Macrophages Cloak Tissue Microlesions to Prevent Neutrophil-Driven Inflammatory Damage. Cell 2019, 177, 541–555.e517. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  240. McDonald, B.; Pittman, K.; Menezes, G.B.; Hirota, S.A.; Slaba, I.; Waterhouse, C.C.; Beck, P.L.; Muruve, D.A.; Kubes, P. Intravascular danger signals guide neutrophils to sites of sterile inflammation. Science 2010, 330, 362–366. [Google Scholar] [CrossRef]
  241. Volmering, S.; Block, H.; Boras, M.; Lowell, C.A.; Zarbock, A. The Neutrophil Btk Signalosome Regulates Integrin Activation during Sterile Inflammation. Immunity 2016, 44, 73–87. [Google Scholar] [CrossRef] [Green Version]
  242. Honda, F.; Kano, H.; Kanegane, H.; Nonoyama, S.; Kim, E.S.; Lee, S.K.; Takagi, M.; Mizutani, S.; Morio, T. The kinase Btk negatively regulates the production of reactive oxygen species and stimulation-induced apoptosis in human neutrophils. Nat. Immunol. 2012, 13, 369–378. [Google Scholar] [CrossRef]
  243. Bromberger, T.; Klapproth, S.; Rohwedder, I.; Weber, J.; Pick, R.; Mittmann, L.; Min-Weissenhorn, S.J.; Reichel, C.A.; Scheiermann, C.; Sperandio, M.; et al. Binding of Rap1 and Riam to Talin1 Fine-Tune beta2 Integrin Activity During Leukocyte Trafficking. Front. Immunol. 2021, 12, 702345. [Google Scholar] [CrossRef] [PubMed]
  244. Wang, J.; Shiratori, I.; Uehori, J.; Ikawa, M.; Arase, H. Neutrophil infiltration during inflammation is regulated by PILRalpha via modulation of integrin activation. Nat. Immunol. 2013, 14, 34–40. [Google Scholar] [CrossRef] [PubMed]
  245. Margraf, A.; Volmering, S.; Skupski, J.; Van Marck, V.; Makrigiannis, A.P.; Block, H.; Zarbock, A. The ITIM Domain-Containing NK Receptor Ly49Q Impacts Pulmonary Infection by Mediating Neutrophil Functions. J. Immunol. 2018, 200, 4085–4093. [Google Scholar] [CrossRef] [PubMed]
  246. Heit, B.; Robbins, S.M.; Downey, C.M.; Guan, Z.; Colarusso, P.; Miller, B.J.; Jirik, F.R.; Kubes, P. PTEN functions to ‘prioritize’ chemotactic cues and prevent ‘distraction’ in migrating neutrophils. Nat. Immunol. 2008, 9, 743–752. [Google Scholar] [CrossRef]
  247. Xu, J.; Gao, X.P.; Ramchandran, R.; Zhao, Y.Y.; Vogel, S.M.; Malik, A.B. Nonmuscle myosin light-chain kinase mediates neutrophil transmigration in sepsis-induced lung inflammation by activating beta2 integrins. Nat. Immunol. 2008, 9, 880–886. [Google Scholar] [CrossRef] [Green Version]
  248. Immler, R.; Nadolni, W.; Bertsch, A.; Morikis, V.; Rohwedder, I.; Masgrau-Alsina, S.; Schroll, T.; Yevtushenko, A.; Soehnlein, O.; Moser, M.; et al. The voltage-gated potassium channel KV1.3 regulates neutrophil recruitment during inflammation. Cardiovasc. Res. 2022, 118, 1289–1302. [Google Scholar] [CrossRef]
  249. Clemens, R.A.; Chong, J.; Grimes, D.; Hu, Y.; Lowell, C.A. STIM1 and STIM2 cooperatively regulate mouse neutrophil store-operated calcium entry and cytokine production. Blood 2017, 130, 1565–1577. [Google Scholar] [CrossRef]
  250. Clemens, R.A.; Lowell, C.A. Store-operated calcium signaling in neutrophils. J. Leukoc. Biol. 2015, 98, 497–502. [Google Scholar] [CrossRef] [Green Version]
  251. Grimes, D.; Johnson, R.; Pashos, M.; Cummings, C.; Kang, C.; Sampedro, G.R.; Tycksen, E.; McBride, H.J.; Sah, R.; Lowell, C.A.; et al. ORAI1 and ORAI2 modulate murine neutrophil calcium signaling, cellular activation, and host defense. Proc. Natl. Acad. Sci. USA 2020, 117, 24403–24414. [Google Scholar] [CrossRef]
  252. Li, J.; Kumari, T.; Barazia, A.; Jha, V.; Jeong, S.Y.; Olson, A.; Kim, M.; Lee, B.K.; Manickam, V.; Song, Z.; et al. Neutrophil DREAM promotes neutrophil recruitment in vascular inflammation. J. Exp. Med. 2022, 219, e20211083. [Google Scholar] [CrossRef]
  253. Khoyratty, T.E.; Ai, Z.; Ballesteros, I.; Eames, H.L.; Mathie, S.; Martin-Salamanca, S.; Wang, L.; Hemmings, A.; Willemsen, N.; von Werz, V.; et al. Distinct transcription factor networks control neutrophil-driven inflammation. Nat. Immunol. 2021, 22, 1093–1106. [Google Scholar] [CrossRef] [PubMed]
  254. Fischer, A.; Wannemacher, J.; Christ, S.; Koopmans, T.; Kadri, S.; Zhao, J.; Gouda, M.; Ye, H.; Muck-Hausl, M.; Krenn, P.W.; et al. Neutrophils direct preexisting matrix to initiate repair in damaged tissues. Nat. Immunol. 2022, 23, 518–531. [Google Scholar] [CrossRef] [PubMed]
  255. Owen-Woods, C.; Joulia, R.; Barkaway, A.; Rolas, L.; Ma, B.; Nottebaum, A.F.; Arkill, K.P.; Stein, M.; Girbl, T.; Golding, M.; et al. Local microvascular leakage promotes trafficking of activated neutrophils to remote organs. J. Clin. Investig. 2020, 130, 2301–2318. [Google Scholar] [CrossRef] [PubMed]
  256. Colom, B.; Bodkin, J.V.; Beyrau, M.; Woodfin, A.; Ody, C.; Rourke, C.; Chavakis, T.; Brohi, K.; Imhof, B.A.; Nourshargh, S. Leukotriene B4-Neutrophil Elastase Axis Drives Neutrophil Reverse Transendothelial Cell Migration In Vivo. Immunity 2015, 42, 1075–1086. [Google Scholar] [CrossRef] [Green Version]
  257. Skopelja-Gardner, S.; Tai, J.; Sun, X.; Tanaka, L.; Kuchenbecker, J.A.; Snyder, J.M.; Kubes, P.; Mustelin, T.; Elkon, K.B. Acute skin exposure to ultraviolet light triggers neutrophil-mediated kidney inflammation. Proc. Natl. Acad. Sci. USA 2021, 118, e2019097118. [Google Scholar] [CrossRef]
  258. Sreeramkumar, V.; Adrover, J.M.; Ballesteros, I.; Cuartero, M.I.; Rossaint, J.; Bilbao, I.; Nacher, M.; Pitaval, C.; Radovanovic, I.; Fukui, Y.; et al. Neutrophils scan for activated platelets to initiate inflammation. Science 2014, 346, 1234–1238. [Google Scholar] [CrossRef] [Green Version]
  259. De Giovanni, M.; Tam, H.; Valet, C.; Xu, Y.; Looney, M.R.; Cyster, J.G. GPR35 promotes neutrophil recruitment in response to serotonin metabolite 5-HIAA. Cell 2022, 185, 815–830.e819. [Google Scholar] [CrossRef]
  260. Mauler, M.; Herr, N.; Schoenichen, C.; Witsch, T.; Marchini, T.; Hardtner, C.; Koentges, C.; Kienle, K.; Ollivier, V.; Schell, M.; et al. Platelet Serotonin Aggravates Myocardial Ischemia/Reperfusion Injury via Neutrophil Degranulation. Circulation 2019, 139, 918–931. [Google Scholar] [CrossRef]
  261. Estevez, B.; Du, X. New Concepts and Mechanisms of Platelet Activation Signaling. Physiology (Bethesda) 2017, 32, 162–177. [Google Scholar] [CrossRef] [Green Version]
  262. Gaertner, F.; Ahmad, Z.; Rosenberger, G.; Fan, S.; Nicolai, L.; Busch, B.; Yavuz, G.; Luckner, M.; Ishikawa-Ankerhold, H.; Hennel, R.; et al. Migrating Platelets Are Mechano-scavengers that Collect and Bundle Bacteria. Cell 2017, 171, 1368–1382. [Google Scholar] [CrossRef]
  263. Nicolai, L.; Schiefelbein, K.; Lipsky, S.; Leunig, A.; Hoffknecht, M.; Pekayvaz, K.; Raude, B.; Marx, C.; Ehrlich, A.; Pircher, J.; et al. Vascular surveillance by haptotactic blood platelets in inflammation and infection. Nat. Commun. 2020, 11, 5778. [Google Scholar] [CrossRef] [PubMed]
  264. Wu, X.; Liu, Y.; Jin, S.; Wang, M.; Jiao, Y.; Yang, B.; Lu, X.; Ji, X.; Fei, Y.; Yang, H.; et al. Single-cell sequencing of immune cells from anticitrullinated peptide antibody positive and negative rheumatoid arthritis. Nat. Commun. 2021, 12, 4977. [Google Scholar] [CrossRef] [PubMed]
  265. Koppejan, H.; Hameetman, M.; Beyrend, G.; van Unen, V.; Kwekkeboom, J.C.; van der Helm-van Mil, A.H.; Toes, R.E.M.; van Gaalen, F.A. Immunoprofiling of early, untreated rheumatoid arthritis using mass cytometry reveals an activated basophil subset inversely linked to ACPA status. Arthritis Res. Ther. 2021, 23, 272. [Google Scholar] [CrossRef]
  266. Reyes, M.; Filbin, M.R.; Bhattacharyya, R.P.; Billman, K.; Eisenhaure, T.; Hung, D.T.; Levy, B.D.; Baron, R.M.; Blainey, P.C.; Goldberg, M.B.; et al. An immune-cell signature of bacterial sepsis. Nat. Med. 2020, 26, 333–340. [Google Scholar] [CrossRef] [PubMed]
  267. Chen, T.; Conroy, J.; Wang, X.; Situ, M.; Namas, R.A.; Vodovotz, Y.; Chen, W.; Singh, H.; Billiar, T.R. The independent prognostic value of global epigenetic alterations: An analysis of single-cell ATAC-seq of circulating leukocytes from trauma patients followed by validation in whole blood leukocyte transcriptomes across three etiologies of critical illness. EBioMedicine 2022, 76, 103860. [Google Scholar] [CrossRef] [PubMed]
  268. Mitsialis, V.; Wall, S.; Liu, P.; Ordovas-Montanes, J.; Parmet, T.; Vukovic, M.; Spencer, D.; Field, M.; McCourt, C.; Toothaker, J.; et al. Single-Cell Analyses of Colon and Blood Reveal Distinct Immune Cell Signatures of Ulcerative Colitis and Crohn’s Disease. Gastroenterology 2020, 159, 591–608.e510. [Google Scholar] [CrossRef]
  269. Wauters, E.; Van Mol, P.; Garg, A.D.; Jansen, S.; Van Herck, Y.; Vanderbeke, L.; Bassez, A.; Boeckx, B.; Malengier-Devlies, B.; Timmerman, A.; et al. Discriminating mild from critical COVID-19 by innate and adaptive immune single-cell profiling of bronchoalveolar lavages. Cell Res. 2021, 31, 272–290. [Google Scholar] [CrossRef]
  270. Wilk, A.J.; Rustagi, A.; Zhao, N.Q.; Roque, J.; Martinez-Colon, G.J.; McKechnie, J.L.; Ivison, G.T.; Ranganath, T.; Vergara, R.; Hollis, T.; et al. A single-cell atlas of the peripheral immune response in patients with severe COVID-19. Nat. Med. 2020, 26, 1070–1076. [Google Scholar] [CrossRef]
  271. Heming, M.; Li, X.; Rauber, S.; Mausberg, A.K.; Borsch, A.L.; Hartlehnert, M.; Singhal, A.; Lu, I.N.; Fleischer, M.; Szepanowski, F.; et al. Neurological Manifestations of COVID-19 Feature T Cell Exhaustion and Dedifferentiated Monocytes in Cerebrospinal Fluid. Immunity 2021, 54, 164–175.e166. [Google Scholar] [CrossRef]
  272. Garash, R.; Bajpai, A.; Marcinkiewicz, B.M.; Spiller, K.L. Drug delivery strategies to control macrophages for tissue repair and regeneration. Exp. Biol. Med. (Maywood) 2016, 241, 1054–1063. [Google Scholar] [CrossRef] [Green Version]
  273. Faas, M.; Ipseiz, N.; Ackermann, J.; Culemann, S.; Gruneboom, A.; Schroder, F.; Rothe, T.; Scholtysek, C.; Eberhardt, M.; Bottcher, M.; et al. IL-33-induced metabolic reprogramming controls the differentiation of alternatively activated macrophages and the resolution of inflammation. Immunity 2021, 54, 2531–2546.e2535. [Google Scholar] [CrossRef] [PubMed]
  274. Horuluoglu, B.; Bayik, D.; Kayraklioglu, N.; Goguet, E.; Kaplan, M.J.; Klinman, D.M. PAM3 supports the generation of M2-like macrophages from lupus patient monocytes and improves disease outcome in murine lupus. J. Autoimmun. 2019, 99, 24–32. [Google Scholar] [CrossRef] [PubMed]
  275. Horuluoglu, B.H.; Kayraklioglu, N.; Tross, D.; Klinman, D. PAM3 protects against DSS-induced colitis by altering the M2:M1 ratio. Sci. Rep. 2020, 10, 6078. [Google Scholar] [CrossRef] [PubMed]
  276. Feng, Q.; Xu, M.; Yu, Y.Y.; Hou, Y.; Mi, X.; Sun, Y.X.; Ma, S.; Zuo, X.Y.; Shao, L.L.; Hou, M.; et al. High-dose dexamethasone or all-trans-retinoic acid restores the balance of macrophages towards M2 in immune thrombocytopenia. J. Thromb. Haemost. 2017, 15, 1845–1858. [Google Scholar] [CrossRef] [Green Version]
  277. Huang, Q.S.; Liu, Y.; Wang, J.B.; Peng, J.; Hou, M.; Liu, H.; Feng, R.; Wang, J.W.; Xu, L.P.; Wang, Y.; et al. All-trans retinoic acid plus high-dose dexamethasone as first-line treatment for patients with newly diagnosed immune thrombocytopenia: A multicentre, open-label, randomised, controlled, phase 2 trial. Lancet Haematol. 2021, 8, e688–e699. [Google Scholar] [CrossRef]
  278. Trilleaud, C.; Gauttier, V.; Biteau, K.; Girault, I.; Belarif, L.; Mary, C.; Pengam, S.; Teppaz, G.; Thepenier, V.; Danger, R.; et al. Agonist anti-ChemR23 mAb reduces tissue neutrophil accumulation and triggers chronic inflammation resolution. Sci. Adv. 2021, 7, eabd1453. [Google Scholar] [CrossRef] [PubMed]
  279. Ansari, J.; Senchenkova, E.Y.; Vital, S.A.; Al-Yafeai, Z.; Kaur, G.; Sparkenbaugh, E.M.; Orr, A.W.; Pawlinski, R.; Hebbel, R.P.; Granger, D.N.; et al. Targeting the AnxA1/Fpr2/ALX pathway regulates neutrophil function, promoting thromboinflammation resolution in sickle cell disease. Blood 2021, 137, 1538–1549. [Google Scholar] [CrossRef]
  280. Jobin, K.; Stumpf, N.E.; Schwab, S.; Eichler, M.; Neubert, P.; Rauh, M.; Adamowski, M.; Babyak, O.; Hinze, D.; Sivalingam, S.; et al. A high-salt diet compromises antibacterial neutrophil responses through hormonal perturbation. Sci. Transl. Med. 2020, 12, eaay3850. [Google Scholar] [CrossRef]
  281. Canzano, P.; Brambilla, M.; Porro, B.; Cosentino, N.; Tortorici, E.; Vicini, S.; Poggio, P.; Cascella, A.; Pengo, M.F.; Veglia, F.; et al. Platelet and Endothelial Activation as Potential Mechanisms Behind the Thrombotic Complications of COVID-19 Patients. JACC Basic Transl. Sci. 2021, 6, 202–218. [Google Scholar] [CrossRef]
  282. Turnbull, I.R.; Fuchs, A.; Remy, K.E.; Kelly, M.P.; Frazier, E.P.; Ghosh, S.; Chang, S.W.; Mazer, M.B.; Hess, A.; Leonard, J.M.; et al. Dysregulation of the leukocyte signaling landscape during acute COVID-19. PLoS ONE 2022, 17, e0264979. [Google Scholar] [CrossRef]
  283. Suvarna, K.; Salkar, A.; Palanivel, V.; Bankar, R.; Banerjee, N.; Gayathri, J.P.M.; Srivastava, A.; Singh, A.; Khatri, H.; Agrawal, S.; et al. A Multi-omics Longitudinal Study Reveals Alteration of the Leukocyte Activation Pathway in COVID-19 Patients. J. Proteome Res. 2021, 20, 4667–4680. [Google Scholar] [CrossRef] [PubMed]
  284. Kaiser, R.; Leunig, A.; Pekayvaz, K.; Popp, O.; Joppich, M.; Polewka, V.; Escaig, R.; Anjum, A.; Hoffknecht, M.L.; Gold, C.; et al. Self-sustaining IL-8 loops drive a prothrombotic neutrophil phenotype in severe COVID-19. JCI Insight 2021, 6, e150862. [Google Scholar] [CrossRef] [PubMed]
  285. Veras, F.P.; Pontelli, M.C.; Silva, C.M.; Toller-Kawahisa, J.E.; de Lima, M.; Nascimento, D.C.; Schneider, A.H.; Caetite, D.; Tavares, L.A.; Paiva, I.M.; et al. SARS-CoV-2-triggered neutrophil extracellular traps mediate COVID-19 pathology. J. Exp. Med. 2020, 217, e150862. [Google Scholar] [CrossRef]
  286. MacDonald, L.; Alivernini, S.; Tolusso, B.; Elmesmari, A.; Somma, D.; Perniola, S.; Paglionico, A.; Petricca, L.; Bosello, S.L.; Carfi, A.; et al. COVID-19 and RA share an SPP1 myeloid pathway that drives PD-L1+ neutrophils and CD14+ monocytes. JCI Insight 2021, 6, e150862. [Google Scholar] [CrossRef] [PubMed]
  287. Lourda, M.; Dzidic, M.; Hertwig, L.; Bergsten, H.; Palma Medina, L.M.; Sinha, I.; Kvedaraite, E.; Chen, P.; Muvva, J.R.; Gorin, J.B.; et al. High-dimensional profiling reveals phenotypic heterogeneity and disease-specific alterations of granulocytes in COVID-19. Proc. Natl. Acad. Sci. USA 2021, 118. [Google Scholar] [CrossRef] [PubMed]
  288. Panda, R.; Castanheira, F.V.; Schlechte, J.M.; Surewaard, B.G.; Shim, H.B.; Zucoloto, A.Z.; Slavikova, Z.; Yipp, B.G.; Kubes, P.; McDonald, B. A functionally distinct neutrophil landscape in severe COVID-19 reveals opportunities for adjunctive therapies. JCI Insight 2022, 7. [Google Scholar] [CrossRef] [PubMed]
  289. Fan, Z.; Pitmon, E.; Wen, L.; Miller, J.; Ehinger, E.; Herro, R.; Liu, W.; Chen, J.; Mikulski, Z.; Conrad, D.J.; et al. Bone Marrow Transplantation Rescues Monocyte Recruitment Defect and Improves Cystic Fibrosis in Mice. J. Immunol. 2022, 208, 745–752. [Google Scholar] [CrossRef]
Figure 1. Modes of cell phenotype switches. Immune cells are activated by ligand–receptor binding. This may evoke different responses, including shedding or internalization of surface molecules, exocytosis and vesicle–membrane fusion, integrin activation, lipid raft redistribution, actin reorganization and shape-changes and potentially the integration or ingestion of foreign material.
Figure 1. Modes of cell phenotype switches. Immune cells are activated by ligand–receptor binding. This may evoke different responses, including shedding or internalization of surface molecules, exocytosis and vesicle–membrane fusion, integrin activation, lipid raft redistribution, actin reorganization and shape-changes and potentially the integration or ingestion of foreign material.
Cells 11 01824 g001
Figure 2. Example of generalizable principles of immune cell phenotype changes. Age, lifespan and microenvironments are known to impact occurrence and regulation of immune cell phenotypes. Additionally, mechanical forces, leading to deformation and disruption of membrane components, can lead to changes in shape, membrane composition, activation patterns and overall phenotype appearance. Demand-dependent (re-)production leads to occurrence of various distinct phenotypes, including immature, pro-inflammatory and immunomodulatory cellular phenotypes.
Figure 2. Example of generalizable principles of immune cell phenotype changes. Age, lifespan and microenvironments are known to impact occurrence and regulation of immune cell phenotypes. Additionally, mechanical forces, leading to deformation and disruption of membrane components, can lead to changes in shape, membrane composition, activation patterns and overall phenotype appearance. Demand-dependent (re-)production leads to occurrence of various distinct phenotypes, including immature, pro-inflammatory and immunomodulatory cellular phenotypes.
Cells 11 01824 g002
Table 1. Techniques (selection) for cell phenotype assessment.
Table 1. Techniques (selection) for cell phenotype assessment.
TypeTechniqueReadoutProConExample
Surface receptor analysisFACS, MicroscopyDetection and expression levels of surface moleculesQuick, relatively cheapIncomplete, biased (depending on antibody selection), possibility of confounding effects (dilution, shedding, etc.)Nicolai et al. [12]
ProteomicsMassSpecProtein contentComplete representation of protein compositionRelatively expensive, machinery neededLeite et al. [7]
TranscriptomicsSequencingGene expression analysis“Wholistic” perspective on regulatory mechanisms and phenotypesExpensive, time-consumingBallesteros et al. [13]
LipidomicsMassSpecLipid mediator compositionDetailed lipid analysisExpensive, equipment intensivePeng et al. [14]
Metabolic profilingUHPLC/MS/MS “Metabotype”Leukocyte specific metabolite analysisMore detailed cellular characterizationConfounder, expensiveAnders et al. [15]
Single-cell enzyme secretionSingle-cell multiplex profilingLeukocyte enzyme secretion phenotypingPatient characteristic immune secretory signaturesNot routinely availableZeming et al. [16]
Functional assaysFlow chambers, ROS production, ligand binding, …Functionality in activation assaysDirect functional readoutNot routinely availableRossaint et al. [17]
Behavioral landscapeIntegrated phenotype assessmentIntegrated multiparametric overview of cellular motionGeneralistic perspective on cellular behavior and functionalityLabor-intensive workup, not routinely availableCrainiciuc et al. [11]
Table 3. Examples of applications of immune cell phenotyping.
Table 3. Examples of applications of immune cell phenotyping.
DiseaseTechniqueObservationPublication
Sero-positive/sero-negative rheumatoid arthritis (RA)Single-cell sequencingUpregulation of CCL13, 18 and MMP3 in circulating myeloid cells; lack of HLA-DRB5 of ACPA− vs. ACPA+ RA patients.Wu et al. [264]
Mass cytometryIdentification of increased CD62L+ basophil subset in ACPA+ vs. ACPA− patients.Koppejan et al. [265]
Multiple sclerosis (MS) phenotypingscRNAseqCompartmentalized immune cell mechanisms and altered expression profilesSchafflick et al. [6]
Sepsis phenotypingscRNAseqPBMC assessment reveals PLAC8 and CLU-dependent CD14+ IL1R2hiHLA-DRlo monocyte discrimination of bacterial sepsis vs. non-sepsis patients with visible expansion in septic patientsReyes et al. [266]
scATAC-seqPBMCs show prognostic value of overall epigenetic heterogeneity (EG-hi: worse survival)Chen et al. [267]
Inflammatory bowel disease (IBD)scRNAseqIdentification of mucosal IL1B+ macrophages and monocytes in IBD vs. control; PBMCs with increased IL-1β+ circulating monocytes in active Crohn’s disease vs. ulcerative colitis.Mitsialis et al. [268]
COVID-19 phenotypingscRNAseq of BALDiscrimination of disease severityWauters et al. [269]
scRNAseq of BloodCellular atlas of blood immune responses during COVID-19, including developing neutrophil population and HLAII downregulation within PBMCsWilk et al. [270]
scRNAseq of cerebrospinal fluidDedifferentiated monocytes in CSF in neuro-COVID-19 patientsHeming et al. [271]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Margraf, A.; Perretti, M. Immune Cell Plasticity in Inflammation: Insights into Description and Regulation of Immune Cell Phenotypes. Cells 2022, 11, 1824. https://doi.org/10.3390/cells11111824

AMA Style

Margraf A, Perretti M. Immune Cell Plasticity in Inflammation: Insights into Description and Regulation of Immune Cell Phenotypes. Cells. 2022; 11(11):1824. https://doi.org/10.3390/cells11111824

Chicago/Turabian Style

Margraf, Andreas, and Mauro Perretti. 2022. "Immune Cell Plasticity in Inflammation: Insights into Description and Regulation of Immune Cell Phenotypes" Cells 11, no. 11: 1824. https://doi.org/10.3390/cells11111824

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