Neutrophils are polymorphonuclear leukocytes of the phagocytic system that act as first line of host defense against invading pathogens but are also important mediators of inflammation-induced injury. In contrast to other members of the innate immune system, neutrophils are classically considered a homogenous population of terminally differentiated cells with a well-defined and highly conserved function. Indeed, their short lifespan, the absent proliferative capacity, their limited ability to produce large amounts of cytokines, and the failure to recirculate from the tissue to the bloodstream have sustained this idea. However, increasing evidence over the last decade has demonstrated an unexpected phenotypic heterogeneity and functional versatility of the neutrophil population. Far beyond their antimicrobial functions, neutrophils are emerging as decision-shapers during innate and adaptive immune responses. These emerging discoveries open a new door to understand the role of neutrophils during homeostatic but also pathogenic immune processes. Thus, this review details novel insights of neutrophil phenotypic and functional heterogeneity during homeostasis and disease.

Myeloid and lymphoid members of the hematopoietic system are composed of subsets with distinct phenotypic and functional characteristics. As an example, a wide variety of monocyte1  and macrophage2  subpopulations has been identified that display a unique transcriptional profile defining their function during homeostasis and immune responses.1  The diversity of tissue macrophages can partly be explained by their differential embryonic origin.3  Beyond ontogeny, macrophage heterogeneity is dictated by environmental signals inducing differentiation or activation.4  The recognition of external cues by macrophages triggers different patterns of epigenetic modifications, gene expression, and protein synthesis and instructs them to exert a specific function in homeostasis4  or inflammation.5  Such activation has been modeled as 2 opposing phenotypic states with different functional properties: M1 macrophage polarization stimulates Th1 effector and microbicidal responses, and M2 macrophage polarization fosters a Th2 and reparative response. Multiple studies have identified these phenotypes in different pathological situations and demonstrated their importance in disease outcome.2  The dichotomous view of macrophage activation, however, is shifting toward a more plastic understanding of macrophage polarization where macrophages can exhibit multiple phenotypic and functional signatures combining the responses to different environmental stimuli that may vary in a spatiotemporal fashion. This new concept has recently been exemplified in tissue-resident4  or tumor-associated macrophages.6  Thus, heterogeneity and plasticity of leukocyte subsets integrate ontogenic and environmental imprints.

In contrast to other leukocyte populations, the idea of neutrophil heterogeneity has received less attention, likely based on the traditional view of unique properties of neutrophils: Limited lifespan, reduced transcriptional activity, and inability to return to the circulation after migrating to extravascular tissue. Hence, we here review emerging evidence challenging these classical concepts of neutrophil biology and further discuss how this may set the foundation for neutrophil heterogeneity.

Although the lifespan of circulating neutrophils is classically thought to be short,7  this view was recently challenged by a study estimating the lifespan of human neutrophils to be 5.4 days.8  Hence, this longer lifespan of neutrophils may set the basis for neutrophils to undergo phenotypic and functional changes. To further understand neutrophil kinetics, assessment of neutrophil lifespan within tissues is of importance (Figure 1). At sites of inflammation, neutrophil lifespan is increased through inhibition of cell apoptosis, an effect triggered by cytokines, pathogen-associated molecular patterns (PAMPs), damage-associated molecular pattern molecules (DAMPs), or environmental factors.9,10  Indeed, in chronic inflammation, neutrophil lifespan is abnormally prolonged, thereby worsening disease prognosis. Extended neutrophil lifespan through decreased apoptosis is observed in patients with asthma11  with acute coronary syndrome12  and results in increased disease severity. Lifespan extension through prosurvival signals produced under inflammatory conditions may also increase the capacity of neutrophils to undergo phenotypic and functional changes and account for neutrophil heterogeneity. For example, oxygen deprivation in tissue during inflammation drives hypoxia-inducible factor–dependent activation of neutrophil prosurvival pathways13,14  and directly impacts on the bactericidal activity of neutrophils.13  Similarly, in the low oxygen and nutrient environment of cystic fibrosis airways, neutrophils undergo metabolic reprogramming,15  trigger prosurvival mechanisms,16  and acquire distinct phenotypic and functional properties (supplemental Discussion 1, available on the Blood Web site).

Figure 1

Integration of factors determining neutrophil lifespan. The bone marrow neutrophil lineage is composed of 3 compartments: (1) the hematopoietic stem cell (HSC) pool, (2) the mitotic pool that is comprised of myeloblasts, promyelocytes, and myelocytes, and (3) the postmitotic pool that is comprised of metamyelocytes, band cells, and mature neutrophils. The size of the mitotic and postmitotic pool is estimated at 4.36 × 109 and 8.8 × 109 cells/kg, respectively. After 4 to 6 days in the bone marrow, neutrophils are released to the bloodstream (4.3-4.5 × 109 cells/L). The estimated period of time where neutrophils circulate is controversial. Although some studies suggest that the circulating lifespan is around 5 hours, alternative experimental calculation raises neutrophil lifespan up to 5.4 days, as reported in different studies. Neutrophils are also located in the “marginated pools,” vascular pools located in the lungs, spleen, and liver where the turnover time is still unclear. After neutrophils infiltrate tissues during inflammation, interaction with tissue-derived signals such as cytokines, PAMPs, DAMPs, or environmental factors reduces neutrophil apoptosis and increases their lifespan to an extent that remains unknown. fMLF, formyl-methionyl-leucyl phenylalanine; HMGB1, high-mobility group box-1.

Figure 1

Integration of factors determining neutrophil lifespan. The bone marrow neutrophil lineage is composed of 3 compartments: (1) the hematopoietic stem cell (HSC) pool, (2) the mitotic pool that is comprised of myeloblasts, promyelocytes, and myelocytes, and (3) the postmitotic pool that is comprised of metamyelocytes, band cells, and mature neutrophils. The size of the mitotic and postmitotic pool is estimated at 4.36 × 109 and 8.8 × 109 cells/kg, respectively. After 4 to 6 days in the bone marrow, neutrophils are released to the bloodstream (4.3-4.5 × 109 cells/L). The estimated period of time where neutrophils circulate is controversial. Although some studies suggest that the circulating lifespan is around 5 hours, alternative experimental calculation raises neutrophil lifespan up to 5.4 days, as reported in different studies. Neutrophils are also located in the “marginated pools,” vascular pools located in the lungs, spleen, and liver where the turnover time is still unclear. After neutrophils infiltrate tissues during inflammation, interaction with tissue-derived signals such as cytokines, PAMPs, DAMPs, or environmental factors reduces neutrophil apoptosis and increases their lifespan to an extent that remains unknown. fMLF, formyl-methionyl-leucyl phenylalanine; HMGB1, high-mobility group box-1.

Close modal

Leukocyte differentiation toward functionally distinct subpopulations requires the ability of these cells to synthesize and release a new set of proteins in response to environmental signals. Although mature neutrophils are classically considered to have limited capacity for de novo protein synthesis,17  recent work has explored the magnitude and mechanisms of neutrophil transcriptional18  and translational regulation.19  It is important to recognize that human neutrophil mRNA content is 10 to 20 times lower than that of other leukocytes,20  which translates into a lower protein production on a per cell basis. However, because neutrophils recruited to inflamed tissues greatly outnumber other leukocytes, the overall impact of neutrophil-derived cytokines in the inflammatory response can be significant. Beyond cytokines, de novo protein synthesis in neutrophils extends to membrane receptors (eg, Fcγ receptor-121 ), which may additionally alter neutrophil heterogeneity and function. In line with the ability of neutrophils to synthesize de novo proteins, recent studies report an active chromatin remodeling and a fine regulation of mRNA expression. On one side, neutrophil gene expression is controlled on an epigenetic level by chromatin histone modification such as methylation22  or acetylation,23  processes that tightly influence gene transcription. On the other side, neutrophils display a cell-specific pattern of noncoding regulatory regions,24  elements that are involved in the regulation of gene expression. Interestingly, interleukin (IL)-6 expression by human neutrophils after exposure to Toll-like receptor (TLR)-8 ligands involved chromatin remodeling and increased activity of noncoding regulatory elements (eg, enhancers).23  Thus, this evidence supports an inducible epigenetic and genetic regulation of neutrophil gene expression. Moreover, the association between genetic variants24  and altered methylation profiles25  in neutrophils associated with disease susceptibility emphasizes the importance of understanding how gene expression is controlled in neutrophils.

Collectively, these studies indicate that neutrophils are endowed with an active transcriptomic function, the disturbance of which may provoke abnormal gene expression and impact on disease outcome. Moreover, neutrophil transcriptional modulation during communication with the immune network or interaction with environmental signals may dictate the function required in a specific situation. For instance, IL-13+ neutrophils prime macrophages toward an anti-inflammatory phenotype that enhances parasite clearance during nematode infection (supplemental Discussion 2). Similarly, neutrophil-derived IL-12 or IL-10 secreted by CD49d+ neutrophil subsets shape macrophage function during bacterial infection (supplemental Discussion 3).

Abluminal to luminal transmigration

During the last decade, a number of studies have reported the ability of neutrophils to return to the bloodstream after migrating to the extravascular space,26-28  thus challenging the classical concept of unidirectional neutrophil migration. In vitro, reversely migrated neutrophils were shown to display a different phenotype (intercellular adhesion molecule-1 [ICAM-1]highCXCR1low) compared with circulating (ICAM-1lowCXCR1high) or tissue-resident (ICAM-1lowCXCR1low) neutrophils, a phenotype also found in low abundance in the circulation.26  Interestingly, reverse-transmigrated neutrophils were not able to transmigrate again into the tissue and had a prolonged lifespan and an increased capacity to produce superoxide species.26  The in vivo proof of reverse transmigration was described using intravital microscopy.28  Although luminal to abluminal transmigration is regulated by various junctional proteins (eg, platelet endothelial cell adhesion molecule-1, VE-cadherin, and CD99), reverse transmigration predominantly depends on junctional adhesion molecule (JAM)-C.28  Under ischemia-reperfusion, JAM-C is redistributed to a nonjunctional membrane location triggering neutrophil adhesion during luminal to abluminal migration,29  an event that was also observed after leukotriene B4 (LTB4) treatment. On the contrary, excessive LTB4 induced presentation of neutrophil elastase on CD11b, JAM-C degradation, and subsequent reverse transmigration (Figure 2).27  Neutrophils undergoing reverse transmigration exhibited a proinflammatory phenotype characterized by a high ICAM-1 expression.28  Interestingly, ICAM-1high neutrophils were found in distant organs after ischemia/reperfusion or LTB4 infusion, suggesting a potential implication in the systemic propagation of inflammation.27,28  Overall, reverse transmigration prolongs neutrophil lifespan and modulates their phenotype and function, thereby contributing to neutrophil heterogeneity.

Figure 2

Mutidirectional neutrophil migration. Neutrophil reverse transmigration from the subendothelial space to the circulation occurs predominantly after ischemia/reperfusion injury. After LTB4-driven neutrophil recruitment (1), infiltrated neutrophils interact with endothelial cells via CD11b and release NE, which degrades the JAM-C (2), allowing their circulation back to the bloodstream (3). Reverse-transmigrated neutrophils exhibit a distinct phenotype characterize by ICAM-1highCXCR1low. During pathogen-driven inflammation, infiltrated neutrophils may also return to lymphoid organs (bone marrow or lymph node) through the circulation or the lymphatic system. Inside the lymph nodes, neutrophils display an activated phenotype (CD11Bhigh, CD62Llow, CXCR2low) and express MHC (CH)-II and the costimulatory molecules CD80 and CD86, suggesting a newly acquired ability to present antigens (4). Furthermore, this subpopulation of neutrophils is able to modulate the adaptive immune response by promoting or repressing the T- and B-cell function in the lymph node (6). Neutrophils migrating toward the bone marrow induce CD8+ T cell–dependent antiviral responses (5).

Figure 2

Mutidirectional neutrophil migration. Neutrophil reverse transmigration from the subendothelial space to the circulation occurs predominantly after ischemia/reperfusion injury. After LTB4-driven neutrophil recruitment (1), infiltrated neutrophils interact with endothelial cells via CD11b and release NE, which degrades the JAM-C (2), allowing their circulation back to the bloodstream (3). Reverse-transmigrated neutrophils exhibit a distinct phenotype characterize by ICAM-1highCXCR1low. During pathogen-driven inflammation, infiltrated neutrophils may also return to lymphoid organs (bone marrow or lymph node) through the circulation or the lymphatic system. Inside the lymph nodes, neutrophils display an activated phenotype (CD11Bhigh, CD62Llow, CXCR2low) and express MHC (CH)-II and the costimulatory molecules CD80 and CD86, suggesting a newly acquired ability to present antigens (4). Furthermore, this subpopulation of neutrophils is able to modulate the adaptive immune response by promoting or repressing the T- and B-cell function in the lymph node (6). Neutrophils migrating toward the bone marrow induce CD8+ T cell–dependent antiviral responses (5).

Close modal

Lymphatic transit of neutrophils

An alternative way of neutrophil emigration from the tissue involves the exit through the lymphatic vessels, a route that gives neutrophils access to secondary lymphoid organs (Figure 2). During infection, mouse neutrophils are able to migrate to the lymph node through lymph and blood vessels.30-36  The fact that neutrophils were detected carrying living bacteria from infected tissue to the lymph node underlines their migratory capacity and their potential to interact with the adaptive immune system.30  In agreement with these observations, Hampton and colleagues recently confirmed the ability of neutrophils to migrate into the lymph nodes after skin infection.34  Interestingly, migrated neutrophils exhibited a distinct activated phenotype (CD11Bhigh, CD62Llow, CXCR2low), which was required for migration to the lymph nodes. Functionally, specific blocking of neutrophil migration to the lymph nodes reduced T-cell proliferation, thereby demonstrating the importance of neutrophils in regulating the adaptive response.34  Accordingly, neutrophils were able to enter lymph nodes in a mouse model of antigen immunization and to modulate CD4+ T-cell and B-cell responses.36  Another example of neutrophil recircularization comes from the observation that neutrophils egress from virus-infected skin and infiltrate the bone marrow to interact with resident CD8+ T cells resulting in enhanced antiviral responses.32  Hence, recirculation of neutrophils to draining lymph nodes may act as an early step of antigen presentation and initiation of adaptive immune responses. In accordance with this, neutrophils recruited into lymph nodes upregulated major histocompatibility complex (MHC)-II and costimulatory molecules (CD80, CD86), which may be involved in neutrophil-dependent antigen presentation.34  Alternatively, it has been proposed that neutrophils carrying bacteria or viruses may serve as “Trojan horses” that permit the dissemination of the infection on macrophage engulfment.37,38  Collectively, the ability of neutrophils to egress from tissue, to re-enter into the circulation, and to migrate into secondary lymphoid organs represents 1 source of neutrophil plasticity and heterogeneity.

Recent reviews have listed various forms of neutrophil heterogeneity.39,40  Below we discuss the most recent studies describing neutrophil heterogeneity under homeostatic and disease conditions (Table 1).41-72 

Table 1

Neutrophil heterogeneity in homeostatic and pathological conditions

Neutrophil subsetPrevalencePhenotypic propertiesFunctional propertiesHomeostatic/pathological relevanceReference
CD177+ Human: ∼50% of circulating neutrophils CD177+ Proteinase 3 = Apoptosis = transmigration in human peritonitis↓ granule protein mRNA content CD177 autoantibodies in ANCA-derived vasculitis patients 41,,,-45  
“Aged” MouseCXCR4highCD62LlowCD11Bhigh CD49high; hypersegmented nucleus; Reduced size and granularity  Regulation of stem cell niche and circadian release of HSPCs to the circulation / Enhanced intravascular inflammation 46,47  
55 ± 14% in ZT5 (clearance from the circulation period) = Apoptosis 
8 ± 3% in Z17 (release from the circulation period) ↑ Phagocytosis and NETosis 
OLFM4+ Human: 20-25% of circulating neutrophils OLFM4 expression in neutrophil-specific granules = Apoptosis OLFM4 autoantibodies in ANCA-derived vasculitis patients 48,,,-52  
= Phagocytosis Enhanced immune response against S aureus and E coli infection in absence of OLFM4 
= Transmigration 
↓ Cathepsin C activity 
OLFM4 + NET formation 
TCR+ Human: 3-5% of circulating neutrophils Human: TCRα,β variants Delayed apoptosis and IL-8 production on CD3/CD28 stimulation Reduced TCR variants in aged subjects compared to young individuals 53,54  
Mouse: Vα2, Vα5, Vβ1, Vβ16 
Angiogenic Mouse: 2.8% CD49d+VEGFR1highCXCR4high ↑MMP9 Promotes angiogenesis in hypoxic tissue 55,56  
Human: 3.2% 
CD62Ldim/CD16brightCD62Lbright/CD16dim Human: 3 hours after LPS administration: CD62Lbright; CD16bright; multi-lobular nucleus CD62Ldim/CD16bright: -Inhibition of T-cell proliferation through ROS release and dependent on CD11B expression Potential implication in sepsis-related immunosuppresion 57,58  
CD62Lbright/CD16bright: 60-70% CD62Ldim; CD16bright; CD11Chigh; CD11Bhigh; CD54high ; hypersegmented nucleus 
CD62Ldim/CD16bright:20-25% CD62Lbright; CD16dim; CD11Clow; CD11Blow; CD54low ; band-form nucleus 
CD62Lbright/CD16dim:10-15% 
CD63+ Human: neutrophils isolated from lung sputum CD11Bhigh CD66high CD63+CD80+MHC-II+ ↓ Glutathione activity Neutrophil elastase-dependent perpetuation of inflammation, infection and progression of cystic fibrosis airway disease 59,60  
↑ Neutrophil elastase activity 
↑ Arg1 
IL-13+ Not defined Ring-form nucleus; IL-5; IL-13; IL-33; insulin-like growth factor-1; Resistin-like molecule alpha/FIZZ1; chitinase-like 3. Priming of macrophages towards alternative or M2 phenotype Accelerated clearance of nematodes by primed macrophages 61  
CD49+ MousePMN-I: CD11B, CD49+, TLR2+, TLR4+, TLR5+, TLR8+; multilobular nucleus, MPOhigh PMN-I: IL-12, CCL3, classic activation of macrophages CD49d+ neutrophils promote virus-challenged experimental asthma 62,-64  
Sendai virus infection: 50% of BAL neutrophils 
MRSA infection: not defined PMN-II: IL-10, CCL2, alternative activation of macrophages CD49+ but not CD49 neutrophils induce resistance to MRSA infection 
HumanPMN-II: CD11B+, CD49-, TLR2+, TLR4+, TLR7+, TLR9+, ring-form nucleus; MPOlow Association between CD49d+ neutrophils and allergic disease in humans 
Non atopic patients: 1.625%; atopic patients: 6.57% 
IL-17+ Human: ∼70% of circulating neutrophils upon IL-6 and IL-23 stimulation IL-17A+; IL-17ra+, RORγt+; dectin-2 ↑ ROS production Reduction of Aspergillus fumigatus–mediated keratitis 65  
Mouse: ∼17% of bone marrow neutrophils on IL-6 and IL-23 stimulation ↑ Increase fungal killing capacity 
LDGs HumanMixed population with cells with band, lobular, or myelocyte-like nuclei ↓ Phagocytosis, = MPO, =respiratory burst Promotion of SLE-associated inflammation: induction of IFN-α production by pDCs through NET release 66,-68  
Healthy donors: ∼17% ↑ IFN-γ; TNF-α 
SLE patients: 1.2-54% ↑ Endothelial cell killing capacity 
 ↑ NET production 
TAN Not defined N1 TAN: Met+; hypersegmented nuclei N1 TAN: ↑ tumor cell killing capacity; ↑ NO; ↑ H2O2; ↑ TNF-α; ↑ ICAM-1; ↓ arg1; ↓ CCL2, ↓CCL5, ↓VEGF; ↓ MMP9 N1 and N2 TANs inhibit and promotes tumor development, respectively 69,,-72  
N2 TAN: Rounded nuclei N2 TAN: Rounded nuclei ↓ tumor cell killing capacity; ↓ NO; ↓ H2O2; ↓ TNF-α; ↓ ICAM-1; ↑ arg1; ↑ CCL2, ↑ CCL5, ↑ VEGF; ↑ MMP9 ↑ S100a8; ↑ S100a9; ↑ Prok2; 
Neutrophil subsetPrevalencePhenotypic propertiesFunctional propertiesHomeostatic/pathological relevanceReference
CD177+ Human: ∼50% of circulating neutrophils CD177+ Proteinase 3 = Apoptosis = transmigration in human peritonitis↓ granule protein mRNA content CD177 autoantibodies in ANCA-derived vasculitis patients 41,,,-45  
“Aged” MouseCXCR4highCD62LlowCD11Bhigh CD49high; hypersegmented nucleus; Reduced size and granularity  Regulation of stem cell niche and circadian release of HSPCs to the circulation / Enhanced intravascular inflammation 46,47  
55 ± 14% in ZT5 (clearance from the circulation period) = Apoptosis 
8 ± 3% in Z17 (release from the circulation period) ↑ Phagocytosis and NETosis 
OLFM4+ Human: 20-25% of circulating neutrophils OLFM4 expression in neutrophil-specific granules = Apoptosis OLFM4 autoantibodies in ANCA-derived vasculitis patients 48,,,-52  
= Phagocytosis Enhanced immune response against S aureus and E coli infection in absence of OLFM4 
= Transmigration 
↓ Cathepsin C activity 
OLFM4 + NET formation 
TCR+ Human: 3-5% of circulating neutrophils Human: TCRα,β variants Delayed apoptosis and IL-8 production on CD3/CD28 stimulation Reduced TCR variants in aged subjects compared to young individuals 53,54  
Mouse: Vα2, Vα5, Vβ1, Vβ16 
Angiogenic Mouse: 2.8% CD49d+VEGFR1highCXCR4high ↑MMP9 Promotes angiogenesis in hypoxic tissue 55,56  
Human: 3.2% 
CD62Ldim/CD16brightCD62Lbright/CD16dim Human: 3 hours after LPS administration: CD62Lbright; CD16bright; multi-lobular nucleus CD62Ldim/CD16bright: -Inhibition of T-cell proliferation through ROS release and dependent on CD11B expression Potential implication in sepsis-related immunosuppresion 57,58  
CD62Lbright/CD16bright: 60-70% CD62Ldim; CD16bright; CD11Chigh; CD11Bhigh; CD54high ; hypersegmented nucleus 
CD62Ldim/CD16bright:20-25% CD62Lbright; CD16dim; CD11Clow; CD11Blow; CD54low ; band-form nucleus 
CD62Lbright/CD16dim:10-15% 
CD63+ Human: neutrophils isolated from lung sputum CD11Bhigh CD66high CD63+CD80+MHC-II+ ↓ Glutathione activity Neutrophil elastase-dependent perpetuation of inflammation, infection and progression of cystic fibrosis airway disease 59,60  
↑ Neutrophil elastase activity 
↑ Arg1 
IL-13+ Not defined Ring-form nucleus; IL-5; IL-13; IL-33; insulin-like growth factor-1; Resistin-like molecule alpha/FIZZ1; chitinase-like 3. Priming of macrophages towards alternative or M2 phenotype Accelerated clearance of nematodes by primed macrophages 61  
CD49+ MousePMN-I: CD11B, CD49+, TLR2+, TLR4+, TLR5+, TLR8+; multilobular nucleus, MPOhigh PMN-I: IL-12, CCL3, classic activation of macrophages CD49d+ neutrophils promote virus-challenged experimental asthma 62,-64  
Sendai virus infection: 50% of BAL neutrophils 
MRSA infection: not defined PMN-II: IL-10, CCL2, alternative activation of macrophages CD49+ but not CD49 neutrophils induce resistance to MRSA infection 
HumanPMN-II: CD11B+, CD49-, TLR2+, TLR4+, TLR7+, TLR9+, ring-form nucleus; MPOlow Association between CD49d+ neutrophils and allergic disease in humans 
Non atopic patients: 1.625%; atopic patients: 6.57% 
IL-17+ Human: ∼70% of circulating neutrophils upon IL-6 and IL-23 stimulation IL-17A+; IL-17ra+, RORγt+; dectin-2 ↑ ROS production Reduction of Aspergillus fumigatus–mediated keratitis 65  
Mouse: ∼17% of bone marrow neutrophils on IL-6 and IL-23 stimulation ↑ Increase fungal killing capacity 
LDGs HumanMixed population with cells with band, lobular, or myelocyte-like nuclei ↓ Phagocytosis, = MPO, =respiratory burst Promotion of SLE-associated inflammation: induction of IFN-α production by pDCs through NET release 66,-68  
Healthy donors: ∼17% ↑ IFN-γ; TNF-α 
SLE patients: 1.2-54% ↑ Endothelial cell killing capacity 
 ↑ NET production 
TAN Not defined N1 TAN: Met+; hypersegmented nuclei N1 TAN: ↑ tumor cell killing capacity; ↑ NO; ↑ H2O2; ↑ TNF-α; ↑ ICAM-1; ↓ arg1; ↓ CCL2, ↓CCL5, ↓VEGF; ↓ MMP9 N1 and N2 TANs inhibit and promotes tumor development, respectively 69,,-72  
N2 TAN: Rounded nuclei N2 TAN: Rounded nuclei ↓ tumor cell killing capacity; ↓ NO; ↓ H2O2; ↓ TNF-α; ↓ ICAM-1; ↑ arg1; ↑ CCL2, ↑ CCL5, ↑ VEGF; ↑ MMP9 ↑ S100a8; ↑ S100a9; ↑ Prok2; 

arg1, arginase 1; BAL, bronchoalveolar lavage; MMP9, metalloproteinase-9; MPO, myeloperoxidase; MRSA, methicillin-resistant Staphylococcus aureus; NO, nitric oxide; pDCs, plasmacytoid dendritic cells; PMN, polymorphonuclear leukocyte; VEGF, vascular endothelial growth factor; ZT, zeitgeber time; ↑, increase; ↓, decrease; =, no change.

Phenotypic and functional adaption by activation

Neutrophil activation by cues derived from sites of inflammation results in exposure of distinct surface markers or de novo synthesis of cytokines, characteristics that are both commonly used to define neutrophil subsets. Neutrophil heterogeneity may thus partly be explained by a differential activation stage of neutrophil subpopulations. After interaction with endothelial selectins and chemotactic molecules neutrophil secretory vesicles are fused with the surface membrane exposing the proteins contained in these vesicles.73-75  In this way, neutrophils activated on the endothelium quickly upregulate CD11b and CD18, thus contributing to firm adhesion.76  In addition, tumor necrosis factor (TNF) receptors located in specific and gelatinase granules are redistributed to the cell membrane after activation.77  Galectin-3 released by macrophages or mast cells is a potent activator of neutrophils. Its receptor, CD66, is stored in specific granules, and stimulation with lipopolysaccharide (LPS)78  or granulocyte colony-stimulating factor (G-CSF)79  induces the secretion of specific granules exposing CD66 and hence primes the neutrophil for a second response to galectin-3. Laminin receptor-1 is located in the specific granules and its binding to the extracellular matrix protein laminin induces the secretion of lysozyme and the production of superoxide, thereby enhancing “second-hit” responses.80  Interestingly, circulating platelets activated by danger signals can also deliver signals through protruding domains presented by a fraction of intravascular neutrophils, a process that promotes neutrophil crawling and recruitment into inflamed tissues.81 

Neutrophil subsets in homeostasis

CD177.

In humans, CD177 glycoprotein (NB1) is exclusively expressed on the surface of neutrophils and regulates transmigration across the endothelium through the interaction with platelet endothelial cell adhesion molecule-1.82  In humans, a fraction of CD177+ neutrophils has been identified83  coexpressing membrane-bound proteinase-3,41  a serine protease normally located in primary granules. CD177 expression is required for surface presentation of proteinase-3,84  which in turn facilitates the transmigration of CD177+ neutrophils.85  The variable inter- and intraindividual expression of the CD177–proteinase-3 partnership in circulating neutrophils gained importance when membrane proteinase-3 was identified as an antigen antineutrophil cytoplasmic antibody (ANCA)-dependent vasculitis.86  Accordingly, the levels of neutrophils coexpressing proteinase-3 and CD177 are augmented in patients of ANCA-dependent vasculitis87  and associate with increased risk of relapse.88  However, the direct implication of CD177+ or CD177 subsets in ANCA-derived vasculitis and other inflammatory diseases remains unknown. To understand the potential role of CD177, a mouse genetically deficient in this protein was generated.89  As in humans,82  the absence of CD177 did not affect the migratory capacity of neutrophils; however, it caused cell death in this mouse model.89  In conclusion, although the frequency of CD177+ neutrophils during inflammatory disease is consistently augmented, the functional properties of this neutrophil subset are controversial and will require further investigation.

Aged neutrophils.

Neutrophil numbers are controlled by a fine balance between production, retention, mobilization, margination, and clearance. Neutrophil retention in the bone marrow is regulated by the coordinated action of CXCL12 and its receptor CXCR4, whereas neutrophil egress depends on CXCR2 and its ligands.90,91  CXCR4 is expressed at low levels in bone marrow–resident neutrophils while almost undetectable on the surface of circulating neutrophils.92  Interestingly, culture of human neutrophils for 20 hours triggers the upregulation of CXCR4 expression and increases their migratory capacity toward CXCL12 and the predilection to home to the bone marrow where they are cleared.93  In mice, this subpopulation of neutrophils, named “senescent” or “aged” neutrophils, are characterized by the upregulation of CXCR4 and CD11b and the loss of CD62L, size reduction, and hypersegmentation of the nucleus.46  Interestingly, clearance of aged neutrophils by bone marrow–resident macrophages downregulated CXCL12 production by stromal cells and induced the mobilization of hematopoietic stem progenitor cells (HSPCs) to the circulation.46  Moreover, numbers of circulating aged neutrophils and their clearance in the bone marrow fluctuated during the day, a process that controlled the circadian-oscillations of HSPC numbers in the blood.46  Besides this noninflammatory function, aging predisposes overactivation of neutrophils, which increases CD11b expression and susceptibility of neutrophil extracellular trap (NET) formation.47  Furthermore, this recent study provides evidence that neutrophil ageing is regulated by the microbiota in a TLR-dependent fashion. Interestingly, this aged subpopulation predominates in mice and humans under pathological conditions and dramatically worsens the disease outcome in a mouse model of sickle cell disease.47  Overall, these findings illustrate that aging may be a source of neutrophil heterogeneity that predisposes to functional changes with implications in homeostasis and inflammation.

Olfactomedin-4.

Olfactomedin-4 (OLFM4) is present in neutrophil-specific granules of 20% to 25% of human circulating neutrophils.48  Although the specific function of this protein in neutrophils is still unknown, neutrophil-derived OLFM4 has been suggested to induce autoimmune responses (supplemental Discussion 4).

T-cell receptor.

A repertoire of T-cell receptor (TCR) variants has unexpectedly been detected in a subset of circulating human neutrophils.53  A detailed discussion on this subset can be found in the supplemental Discussion 5.

Angiogenic neutrophils.

Under ischemia conditions, a distinct subset of neutrophils is recruited to hypoxic areas with unique phenotypic and angiogenic properties that facilitate restoration of the oxygen supply in the affected tissue55  (supplemental Discussion 6).

Neutrophil subsets in pathologic conditions

Pathogen-mediated systemic inflammation.

Systemic inflammation elicited by injury or sepsis causes overproduction and hyperreaction of neutrophils. After the initial immune response, nonresolved inflammation results in a prolonged action of neutrophils, which may cause collateral damage.94  However, it is during this latter phase when an anti-inflammatory response is produced, resulting in immunosuppression and increasing the risk for mortality.95  Because neutrophilia is still occurring during this phase of the disease, Pillay and colleagues aimed to examine specific alterations in the phenotype and function of neutrophils that may explain the observed immunosuppression.57  Analysis of circulating neutrophils in LPS-challenged healthy volunteers identified fully competent mature CD16bright neutrophils that were accompanied by the release of banded CD16dim neutrophils with increased reactive oxygen species (ROS) production but impaired antimicrobial function.57  Consequent analysis of LPS-driven neutrophil heterogeneity by the same group revealed the existence of a novel subset of mature, hypersegmented human neutrophils with immunosuppressive activity.58  This subset was defined by the surface markers CD11cbrightCD62LdimCD11bbrightCD16bright and was able to suppress T-cell proliferation through the release of ROS and required the expression of CD11b. The ability to suppress T-cell proliferation and responses has also been described for myeloid-derived suppressor cells (MDSCs), a leukocytic population identified in different pathologies such as infection or cancer.96  This MDSC population is composed of a monocytic and granulocytic subpopulation with the ability to suppress T-cell proliferation and activation in a ROS- and Arginase I–dependent fashion. Although the granulocytic counterparts of this population (G-MDSC) exhibit phenotypic and functional differences compared with those observed by Pillay and colleagues,96  both share this immunosuppressive activity. The lack of a consensus when phenotyping G-MDSCs96  makes it difficult to discern whether these 2 populations belong to a unique heterogeneous population with immunosuppressive abilities.

In pathogen-mediated inflammation, a number of other neutrophil subsets have been described. Refer to the supplemental Discussion for information on CD63+, IL-13+, CD49+, and IL-17+ neutrophil subsets (supplemental Discussions 1, 2, 3, and 7).

Systemic lupus erythematosus.

Initially an abnormal population of neutrophils isolated from patients of systemic lupus erythematosus (SLE) and other autoimmune disorders was characterized by displaying different density properties.97  The functional characterization of this subpopulation of neutrophils, referred to as low-density neutrophils (LDGs), revealed a proinflammatory, activated phenotype with exacerbated production of type I interferons (IFNs) and reduced phagocytic capacity.66  Human LDGs were also shown to be highly susceptible to form NETs in the absence of stimulation, which was responsible for the enhanced endothelial cell (EC) toxicity displayed by this subset.67  Indeed, anti-ribonucleoprotein antibodies developed in SLE patients induced NET release by neutrophils causing a potent activation of dendritic cells and production of IFN-α.68,98  Self-DNA and antimicrobial peptides present in the NET structures led to the generation of autoantibodies triggering a positive feedback loop that amplified the inflammatory response. Of note, LDGs have also been identified in other physiological or pathogenic immune alterations such as cancer,99  sepsis,100  pregnancy,101  diabetes,102  or HIV infection.103  Interestingly, these LDG subpopulations share the ability to suppress the immune response, a characteristic that contrasts the proinflammatory phenotype of SLE-associated neutrophils.

Cancer.

During tumor-driven inflammation, ≥2 different subpopulations of neutrophils have been described that exert antagonist functions. Fridlender et al demonstrated in mice that tumors were infiltrated by tumor-associated neutrophils (TANs) characterized by a hypersegmented nucleus, increased production of proinflammatory cytokines, and enhanced tumor cell killing capacity.69  In contrast to this antitumoral population, referred to as N1, an alternative population of TANs (N2) was identified that displayed an immature phenotype and increased arginase activity and promoted tumor growth. TANs exhibit clear phenotypic and functional parallels with M1 and M2 macrophage polarization patterns. For instance, N2 neutrophils and M2 macrophages, both of which favor tumor growth, share elevated arginase activity, which is responsible for inhibition of T-cell proliferation by depletion of l-arginine.104  The “polarization” of TANs from a protumoral toward an antitumoral phenotype was attributed to the action of the anti-inflammatory cytokine transforming growth factor (TGF)-β.69  Interesting in this context is that the primary tumor hijacked the IL-17/G-CSF axis to promote the mobilization of protumoral neutrophils.70  It is presently unclear whether these reported populations of protumoral neutrophils share a common origin and function. On the other hand, IFN-β has been involved in the activation of mouse and human neutrophils toward an antitumor phenotype, as demonstrated by genetically deficient mouse models71,105  and IFN therapies.105  Similarly, Finisguerra and colleagues recently demonstrated the importance of Met, a proto-oncogene whose expression in mouse and human neutrophils is essential in the transmigration, cytotoxic, and protumoral capacity of these cells.72 

As mentioned before, MDSCs are a separate population of cells with monocytic or granulocytic phenotypes that appear in tumor-bearing mice and cancer patients and are characterized by their ability to suppress T-cell proliferation and activity.96  Transcriptomic analysis of TANs compared with G-MDSCs and naïve neutrophils suggested that these 3 populations are distinct, with a surprisingly close proximity between G-MDSCs and naïve neutrophils compared with the TAN subpopulation.106  Hence, it is conceivable that the N1 antitumoral and N2 protumoral phenotypes described by Fridlender and others originated from naïve neutrophils or G-MDSCs, respectively. However, because naïve neutrophils were isolated from tumor-free mice, a more detailed study would be required to define the functional differences between suppressive (G-MDSCs) or nonsuppressive (naïve) neutrophils isolated from tumor-bearing mice. Unfortunately, the only method to reliably distinguish G-MDSCs from normal neutrophils depends on the analysis of their suppressive capacity, a technical handicap that impedes the proper isolation of both populations without compromising their function during manipulation. In this regard, advantage should be taken of the density properties attributed to G-MDSCs that resemble those exhibited by LDGs. Sagiv et al demonstrated that tumor-associated mouse and human LDGs are a heterogeneous population composed of mature and immature neutrophils with suppressive capacity.99  Interestingly, the mature members of this population originated from high-density neutrophils, which acquire immunosuppressive functions after exposure to TGF-β. Thus, this study provides a plausible explanation as to the origin of suppressive, mature neutrophils (N2 TAN). On the other hand, the authors suggested that the immature LDGs originate from immature neutrophils that are released from the bone marrow before completing their maturation. Because LDGs isolated from SLE patients do not exhibit immunosuppressive capacity, it is reasonable to considerer that the acquisition of low-density properties and the immunosuppressive capacity are independent processes that occur after neutrophil activation to thus far unknown environmental signals. Interestingly, in vitro activation of circulating neutrophils isolated from healthy donors induced the acquisition of the suppressive capacity and low-density properties probably after degranulation.107  In line with these results, LDGs101,103  and G-MDSCs107  express high levels of CD66b, a receptor mobilized from the granules to the surface on cell activation. However, additional studies are needed to further clarify the exact origin of these cells and the external signals that confer their immunosuppressive capacities.

As technical handicaps are being solved by the generation of novel techniques and animal models, a wide range of new and fascinating functions attributed to neutrophils is being discovered. The classical view of neutrophils does not accommodate all aspects of neutrophil biology, and thus the previously ascribed functions of neutrophils seem incomplete. The recently described prolonged lifespan of neutrophils, their ability to de novo synthesize cytokines, and the capacity to recirculate through different tissues and organs extend their repertoire of immunomodulatory functions by interacting and modulating the immune response exerted by both the innate and adaptive system. Moreover, these recently identified properties of neutrophils align with the increasing evidence for the existence of neutrophil subpopulations with distinct phenotypic and functional characteristics. It is indeed this renovated view of neutrophils as a plastic cell that may partly explain neutrophil heterogeneity. Understanding neutrophil heterogeneity may be instrumental to develop novel therapies that specifically target pathogenic neutrophil subsets without compromising immunity.

The online version of this article contains a data supplement.

O.S. is supported by the Nederlandse Organisatie voor Wetenschappelijk Onderzoek (VIDI project 91712303), the Academic Medical Center Research Council, the Deutsche Forschungsgemeinschaft (SO876/6-1, SFB914-B08, SFB1123-A06/B05), and the Ludwig Maximilian University excellent program. C.S.-R. is supported by the FöFoLe program of the LMU Medical Faculty. A.H. is supported by SAF2012-31142 from the Ministry of Economy and Competitiveness. The Centro Nacional de Investigaciones Cardiovasculares Carlos III is supported by the Ministry of Economy and Competitiveness and the Pro-CNIC Foundation and is a Severo Ochoa Center of Excellence (Ministry of Economy and Competitiveness award SEV-2015-0505).

Contribution: C.S.-R. wrote the manuscript; and A.H. and O.S. contributed to manuscript preparation and revision.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Carlos Silvestre-Roig, Institute for Cardiovascular Prevention, LMU Munich, Pettenkoferstrasse 9, 80336 Munich, Germany; e-mail: carlos.silvestre@med.uni-muenchen.de; or Oliver Soehnlein, Institute for Cardiovascular Prevention, LMU Munich, Pettenkoferstrasse 9, 80336 Munich, Germany; e-mail: oliver.soehnlein@gmail.com.

1
Ginhoux
 
F
Jung
 
S
Monocytes and macrophages: developmental pathways and tissue homeostasis.
Nat Rev Immunol
2014
, vol. 
14
 
6
(pg. 
392
-
404
)
2
Murray
 
PJ
Wynn
 
TA
Protective and pathogenic functions of macrophage subsets.
Nat Rev Immunol
2011
, vol. 
11
 
11
(pg. 
723
-
737
)
3
Gomez Perdiguero
 
E
Klapproth
 
K
Schulz
 
C
, et al. 
Tissue-resident macrophages originate from yolk-sac-derived erythro-myeloid progenitors.
Nature
2015
, vol. 
518
 
7540
(pg. 
547
-
551
)
4
Lavin
 
Y
Winter
 
D
Blecher-Gonen
 
R
, et al. 
Tissue-resident macrophage enhancer landscapes are shaped by the local microenvironment.
Cell
2014
, vol. 
159
 
6
(pg. 
1312
-
1326
)
5
Murray
 
PJ
Allen
 
JE
Biswas
 
SK
, et al. 
Macrophage activation and polarization: nomenclature and experimental guidelines.
Immunity
2014
, vol. 
41
 
1
(pg. 
14
-
20
)
6
Kratochvill
 
F
Neale
 
G
Haverkamp
 
JM
, et al. 
TNF counterbalances the emergence of M2 tumor macrophages.
Cell Reports
2015
, vol. 
12
 
11
(pg. 
1902
-
1914
)
7
Cartwright
 
GE
Athens
 
JW
Wintrobe
 
MM
The kinetics of granulopoiesis in normal man.
Blood
1964
, vol. 
24
 (pg. 
780
-
803
)
8
Pillay
 
J
den Braber
 
I
Vrisekoop
 
N
, et al. 
In vivo labeling with 2H2O reveals a human neutrophil lifespan of 5.4 days.
Blood
2010
, vol. 
116
 
4
(pg. 
625
-
627
)
9
Geering
 
B
Stoeckle
 
C
Conus
 
S
Simon
 
HU
Living and dying for inflammation: neutrophils, eosinophils, basophils.
Trends Immunol
2013
, vol. 
34
 
8
(pg. 
398
-
409
)
10
Sundqvist
 
M
Wekell
 
P
Osla
 
V
, et al. 
Increased intracellular oxygen radical production in neutrophils during febrile episodes of periodic fever, aphthous stomatitis, pharyngitis, and cervical adenitis syndrome.
Arthritis Rheum
2013
, vol. 
65
 
11
(pg. 
2971
-
2983
)
11
Uddin
 
M
Nong
 
G
Ward
 
J
, et al. 
Prosurvival activity for airway neutrophils in severe asthma.
Thorax
2010
, vol. 
65
 
8
(pg. 
684
-
689
)
12
Garlichs
 
CD
Eskafi
 
S
Cicha
 
I
, et al. 
Delay of neutrophil apoptosis in acute coronary syndromes.
J Leukoc Biol
2004
, vol. 
75
 
5
(pg. 
828
-
835
)
13
Peyssonnaux
 
C
Datta
 
V
Cramer
 
T
, et al. 
HIF-1alpha expression regulates the bactericidal capacity of phagocytes.
J Clin Invest
2005
, vol. 
115
 
7
(pg. 
1806
-
1815
)
14
Thompson
 
AA
Elks
 
PM
Marriott
 
HM
, et al. 
Hypoxia-inducible factor 2α regulates key neutrophil functions in humans, mice, and zebrafish.
Blood
2014
, vol. 
123
 
3
(pg. 
366
-
376
)
15
Laval
 
J
Touhami
 
J
Herzenberg
 
LA
, et al. 
Metabolic adaptation of neutrophils in cystic fibrosis airways involves distinct shifts in nutrient transporter expression.
J Immunol
2013
, vol. 
190
 
12
(pg. 
6043
-
6050
)
16
Makam
 
M
Diaz
 
D
Laval
 
J
, et al. 
Activation of critical, host-induced, metabolic and stress pathways marks neutrophil entry into cystic fibrosis lungs.
Proc Natl Acad Sci USA
2009
, vol. 
106
 
14
(pg. 
5779
-
5783
)
17
Cassatella
 
MA
The production of cytokines by polymorphonuclear neutrophils.
Immunol Today
1995
, vol. 
16
 
1
(pg. 
21
-
26
)
18
Tecchio
 
C
Micheletti
 
A
Cassatella
 
MA
Neutrophil-derived cytokines: facts beyond expression.
Front Immunol
2014
, vol. 
5
 pg. 
508
 
19
Yost
 
CC
Denis
 
MM
Lindemann
 
S
, et al. 
Activated polymorphonuclear leukocytes rapidly synthesize retinoic acid receptor-alpha: a mechanism for translational control of transcriptional events.
J Exp Med
2004
, vol. 
200
 
5
(pg. 
671
-
680
)
20
Scapini
 
P
Calzetti
 
F
Cassatella
 
MA
On the detection of neutrophil-derived vascular endothelial growth factor (VEGF).
J Immunol Methods
1999
, vol. 
232
 
1-2
(pg. 
121
-
129
)
21
Cassatella
 
MA
Flynn
 
RM
Amezaga
 
MA
Bazzoni
 
F
Vicentini
 
F
Trinchieri
 
G
Interferon gamma induces in human neutrophils and macrophages expression of the mRNA for the high affinity receptor for monomeric IgG (Fc gamma R-I or CD64).
Biochem Biophys Res Commun
1990
, vol. 
170
 
2
(pg. 
582
-
588
)
22
Zilbauer
 
M
Rayner
 
TF
Clark
 
C
, et al. 
Genome-wide methylation analyses of primary human leukocyte subsets identifies functionally important cell-type-specific hypomethylated regions.
Blood
2013
, vol. 
122
 
25
(pg. 
e52
-
e60
)
23
Zimmermann
 
M
Aguilera
 
FB
Castellucci
 
M
, et al. 
Chromatin remodelling and autocrine TNFα are required for optimal interleukin-6 expression in activated human neutrophils.
Nat Commun
2015
, vol. 
6
 pg. 
6061
 
24
Naranbhai
 
V
Fairfax
 
BP
Makino
 
S
, et al. 
Genomic modulators of gene expression in human neutrophils.
Nat Commun
2015
, vol. 
6
 pg. 
7545
 
25
Coit
 
P
Yalavarthi
 
S
Ognenovski
 
M
, et al. 
Epigenome profiling reveals significant DNA demethylation of interferon signature genes in lupus neutrophils.
J Autoimmun
2015
, vol. 
58
 (pg. 
59
-
66
)
26
Buckley
 
CD
Ross
 
EA
McGettrick
 
HM
, 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
, vol. 
79
 
2
(pg. 
303
-
311
)
27
Colom
 
B
Bodkin
 
JV
Beyrau
 
M
, et al. 
Leukotriene B4-Neutrophil Elastase Axis Drives Neutrophil Reverse Transendothelial Cell Migration In Vivo.
Immunity
2015
, vol. 
42
 
6
(pg. 
1075
-
1086
)
28
Woodfin
 
A
Voisin
 
MB
Beyrau
 
M
, et al. 
The junctional adhesion molecule JAM-C regulates polarized transendothelial migration of neutrophils in vivo.
Nat Immunol
2011
, vol. 
12
 
8
(pg. 
761
-
769
)
29
Scheiermann
 
C
Colom
 
B
Meda
 
P
, et al. 
Junctional adhesion molecule-C mediates leukocyte infiltration in response to ischemia reperfusion injury.
Arterioscler Thromb Vasc Biol
2009
, vol. 
29
 
10
(pg. 
1509
-
1515
)
30
Abadie
 
V
Badell
 
E
Douillard
 
P
, et al. 
Neutrophils rapidly migrate via lymphatics after Mycobacterium bovis BCG intradermal vaccination and shuttle live bacilli to the draining lymph nodes.
Blood
2005
, vol. 
106
 
5
(pg. 
1843
-
1850
)
31
Chtanova
 
T
Schaeffer
 
M
Han
 
SJ
, et al. 
Dynamics of neutrophil migration in lymph nodes during infection.
Immunity
2008
, vol. 
29
 
3
(pg. 
487
-
496
)
32
Duffy
 
D
Perrin
 
H
Abadie
 
V
, et al. 
Neutrophils transport antigen from the dermis to the bone marrow, initiating a source of memory CD8+ T cells.
Immunity
2012
, vol. 
37
 
5
(pg. 
917
-
929
)
33
Gorlino
 
CV
Ranocchia
 
RP
Harman
 
MF
, et al. 
Neutrophils exhibit differential requirements for homing molecules in their lymphatic and blood trafficking into draining lymph nodes.
J Immunol
2014
, vol. 
193
 
4
(pg. 
1966
-
1974
)
34
Hampton
 
HR
Bailey
 
J
Tomura
 
M
Brink
 
R
Chtanova
 
T
Microbe-dependent lymphatic migration of neutrophils modulates lymphocyte proliferation in lymph nodes.
Nat Commun
2015
, vol. 
6
 pg. 
7139
 
35
Yang
 
CW
Strong
 
BS
Miller
 
MJ
Unanue
 
ER
Neutrophils influence the level of antigen presentation during the immune response to protein antigens in adjuvants.
J Immunol
2010
, vol. 
185
 
5
(pg. 
2927
-
2934
)
36
Yang
 
CW
Unanue
 
ER
Neutrophils control the magnitude and spread of the immune response in a thromboxane A2-mediated process.
J Exp Med
2013
, vol. 
210
 
2
(pg. 
375
-
387
)
37
Coombes
 
JL
Charsar
 
BA
Han
 
SJ
, et al. 
Motile invaded neutrophils in the small intestine of Toxoplasma gondii-infected mice reveal a potential mechanism for parasite spread.
Proc Natl Acad Sci USA
2013
, vol. 
110
 
21
(pg. 
E1913
-
E1922
)
38
Peters
 
NC
Egen
 
JG
Secundino
 
N
, et al. 
 
In vivo imaging reveals an essential role for neutrophils in leishmaniasis transmitted by sand flies. Science (New York). 2008;321(5891):970-974
39
Beyrau
 
M
Bodkin
 
JV
Nourshargh
 
S
Neutrophil heterogeneity in health and disease: a revitalized avenue in inflammation and immunity.
Open Biol
2012
, vol. 
2
 
11
pg. 
120134
 
40
Kruger
 
P
Saffarzadeh
 
M
Weber
 
AN
, et al. 
Neutrophils: between host defence, immune modulation, and tissue injury.
PLoS Pathog
2015
, vol. 
11
 
3
pg. 
e1004651
 
41
Bauer
 
S
Abdgawad
 
M
Gunnarsson
 
L
Segelmark
 
M
Tapper
 
H
Hellmark
 
T
Proteinase 3 and CD177 are expressed on the plasma membrane of the same subset of neutrophils.
J Leukoc Biol
2007
, vol. 
81
 
2
(pg. 
458
-
464
)
42
Abdgawad
 
M
Pettersson
 
Å
Gunnarsson
 
L
, et al. 
Decreased neutrophil apoptosis in quiescent ANCA-associated systemic vasculitis.
PLoS One
2012
, vol. 
7
 
3
pg. 
e32439
 
43
Hu
 
N
Mora-Jensen
 
H
Theilgaard-Mönch
 
K
, et al. 
Differential expression of granulopoiesis related genes in neutrophil subsets distinguished by membrane expression of CD177.
PLoS One
2014
, vol. 
9
 
6
pg. 
e99671
 
44
Stroncek
 
DF
Neutrophil-specific antigen HNA-2a, NB1 glycoprotein, and CD177.
Curr Opin Hematol
2007
, vol. 
14
 
6
(pg. 
688
-
693
)
45
Wang
 
L
Ge
 
S
Agustian
 
A
Hiss
 
M
Haller
 
H
von Vietinghoff
 
S
Surface receptor CD177/NB1 does not confer a recruitment advantage to neutrophilic granulocytes during human peritonitis.
Eur J Haematol
2013
, vol. 
90
 
5
(pg. 
436
-
437
)
46
Casanova-Acebes
 
M
Pitaval
 
C
Weiss
 
LA
, et al. 
Rhythmic modulation of the hematopoietic niche through neutrophil clearance.
Cell
2013
, vol. 
153
 
5
(pg. 
1025
-
1035
)
47
Zhang
 
D
Chen
 
G
Manwani
 
D
, et al. 
Neutrophil ageing is regulated by the microbiome.
Nature
2015
, vol. 
525
 
7570
(pg. 
528
-
532
)
48
Clemmensen
 
SN
Bohr
 
CT
Rørvig
 
S
, et al. 
Olfactomedin 4 defines a subset of human neutrophils.
J Leukoc Biol
2012
, vol. 
91
 
3
(pg. 
495
-
500
)
49
Amirbeagi
 
F
Thulin
 
P
Pullerits
 
R
, et al. 
Olfactomedin-4 autoantibodies give unusual c-ANCA staining patterns with reactivity to a subpopulation of neutrophils.
J Leukoc Biol
2015
, vol. 
97
 
1
(pg. 
181
-
189
)
50
Liu
 
W
Yan
 
M
Liu
 
Y
McLeish
 
KR
Coleman
 
WG
Rodgers
 
GP
Olfactomedin 4 inhibits cathepsin C-mediated protease activities, thereby modulating neutrophil killing of Staphylococcus aureus and Escherichia coli in mice.
J Immunol
2012
, vol. 
189
 
5
(pg. 
2460
-
2467
)
51
Liu
 
W
Yan
 
M
Sugui
 
JA
, et al. 
Olfm4 deletion enhances defense against Staphylococcus aureus in chronic granulomatous disease.
J Clin Invest
2013
, vol. 
123
 
9
(pg. 
3751
-
3755
)
52
Welin
 
A
Amirbeagi
 
F
Christenson
 
K
, et al. 
The human neutrophil subsets defined by the presence or absence of OLFM4 both transmigrate into tissue in vivo and give rise to distinct NETs in vitro.
PLoS One
2013
, vol. 
8
 
7
pg. 
e69575
 
53
Puellmann
 
K
Kaminski
 
WE
Vogel
 
M
, et al. 
A variable immunoreceptor in a subpopulation of human neutrophils.
Proc Natl Acad Sci USA
2006
, vol. 
103
 
39
(pg. 
14441
-
14446
)
54
Fuchs
 
T
Püellmann
 
K
Scharfenstein
 
O
, et al. 
The neutrophil recombinatorial TCR-like immune receptor is expressed across the entire human life span but repertoire diversity declines in old age.
Biochem Biophys Res Commun
2012
, vol. 
419
 
2
(pg. 
309
-
315
)
55
Christoffersson
 
G
Vågesjö
 
E
Vandooren
 
J
, et al. 
VEGF-A recruits a proangiogenic MMP-9-delivering neutrophil subset that induces angiogenesis in transplanted hypoxic tissue.
Blood
2012
, vol. 
120
 
23
(pg. 
4653
-
4662
)
56
Massena
 
S
Christoffersson
 
G
Vågesjö
 
E
, et al. 
Identification and characterization of VEGF-A-responsive neutrophils expressing CD49d, VEGFR1, and CXCR4 in mice and humans.
Blood
2015
, vol. 
126
 
17
(pg. 
2016
-
2026
)
57
Pillay
 
J
Ramakers
 
BP
Kamp
 
VM
, et al. 
Functional heterogeneity and differential priming of circulating neutrophils in human experimental endotoxemia.
J Leukoc Biol
2010
, vol. 
88
 
1
(pg. 
211
-
220
)
58
Pillay
 
J
Kamp
 
VM
van Hoffen
 
E
, et al. 
A subset of neutrophils in human systemic inflammation inhibits T cell responses through Mac-1.
J Clin Invest
2012
, vol. 
122
 
1
(pg. 
327
-
336
)
59
Ingersoll
 
SA
Laval
 
J
Forrest
 
OA
, et al. 
Mature cystic fibrosis airway neutrophils suppress T cell function: evidence for a role of arginase 1 but not programmed death-ligand 1.
J Immunol
2015
, vol. 
194
 
11
(pg. 
5520
-
5528
)
60
Tirouvanziam
 
R
Gernez
 
Y
Conrad
 
CK
, et al. 
Profound functional and signaling changes in viable inflammatory neutrophils homing to cystic fibrosis airways.
Proc Natl Acad Sci USA
2008
, vol. 
105
 
11
(pg. 
4335
-
4339
)
61
Chen
 
F
Wu
 
W
Millman
 
A
, et al. 
Neutrophils prime a long-lived effector macrophage phenotype that mediates accelerated helminth expulsion.
Nat Immunol
2014
, vol. 
15
 
10
(pg. 
938
-
946
)
62
Cheung
 
DS
Ehlenbach
 
SJ
Kitchens
 
RT
, et al. 
Cutting edge: CD49d+ neutrophils induce FcepsilonRI expression on lung dendritic cells in a mouse model of postviral asthma.
J Immunol
2010
, vol. 
185
 
9
(pg. 
4983
-
4987
)
63
Sigua
 
JA
Buelow
 
B
Cheung
 
DS
, et al. 
 
CD49d-expressing neutrophils differentiate atopic from nonatopic individuals. J Allergy Clin Immunol. 2014;133(3):901-904
64
Tsuda
 
Y
Takahashi
 
H
Kobayashi
 
M
Hanafusa
 
T
Herndon
 
DN
Suzuki
 
F
Three different neutrophil subsets exhibited in mice with different susceptibilities to infection by methicillin-resistant Staphylococcus aureus.
Immunity
2004
, vol. 
21
 
2
(pg. 
215
-
226
)
65
Taylor
 
PR
Roy
 
S
Leal
 
SM
, et al. 
Activation of neutrophils by autocrine IL-17A-IL-17RC interactions during fungal infection is regulated by IL-6, IL-23, RORγt and dectin-2.
Nat Immunol
2014
, vol. 
15
 
2
(pg. 
143
-
151
)
66
Denny
 
MF
Yalavarthi
 
S
Zhao
 
W
, et al. 
A distinct subset of proinflammatory neutrophils isolated from patients with systemic lupus erythematosus induces vascular damage and synthesizes type I IFNs.
J Immunol
2010
, vol. 
184
 
6
(pg. 
3284
-
3297
)
67
Villanueva
 
E
Yalavarthi
 
S
Berthier
 
CC
, et al. 
Netting neutrophils induce endothelial damage, infiltrate tissues, and expose immunostimulatory molecules in systemic lupus erythematosus.
J Immunol
2011
, vol. 
187
 
1
(pg. 
538
-
552
)
68
Garcia-Romo
 
GS
Caielli
 
S
Vega
 
B
, et al. 
Netting neutrophils are major inducers of type I IFN production in pediatric systemic lupus erythematosus.
Sci Transl Med
2011
, vol. 
3
 
73
pg. 
73ra20
 
69
Fridlender
 
ZG
Sun
 
J
Kim
 
S
, et al. 
Polarization of tumor-associated neutrophil phenotype by TGF-beta: “N1” versus “N2” TAN.
Cancer Cell
2009
, vol. 
16
 
3
(pg. 
183
-
194
)
70
Coffelt
 
SB
Kersten
 
K
Doornebal
 
CW
, et al. 
IL-17-producing γδ T cells and neutrophils conspire to promote breast cancer metastasis.
Nature
2015
, vol. 
522
 
7556
(pg. 
345
-
348
)
71
Jablonska
 
J
Leschner
 
S
Westphal
 
K
Lienenklaus
 
S
Weiss
 
S
Neutrophils responsive to endogenous IFN-beta regulate tumor angiogenesis and growth in a mouse tumor model.
J Clin Invest
2010
, vol. 
120
 
4
(pg. 
1151
-
1164
)
72
Finisguerra
 
V
Di Conza
 
G
Di Matteo
 
M
, et al. 
MET is required for the recruitment of anti-tumoural neutrophils.
Nature
2015
, vol. 
522
 
7556
(pg. 
349
-
353
)
73
Borregaard
 
N
Kjeldsen
 
L
Rygaard
 
K
, et al. 
Stimulus-dependent secretion of plasma proteins from human neutrophils.
J Clin Invest
1992
, vol. 
90
 
1
(pg. 
86
-
96
)
74
Hidalgo
 
A
Peired
 
AJ
Wild
 
MK
Vestweber
 
D
Frenette
 
PS
Complete identification of E-selectin ligands on neutrophils reveals distinct functions of PSGL-1, ESL-1, and CD44.
Immunity
2007
, vol. 
26
 
4
(pg. 
477
-
489
)
75
Zarbock
 
A
Lowell
 
CA
Ley
 
K
Spleen tyrosine kinase Syk is necessary for E-selectin-induced alpha(L)beta(2) integrin-mediated rolling on intercellular adhesion molecule-1.
Immunity
2007
, vol. 
26
 
6
(pg. 
773
-
783
)
76
Kolaczkowska
 
E
Kubes
 
P
Neutrophil recruitment and function in health and inflammation.
Nat Rev Immunol
2013
, vol. 
13
 
3
(pg. 
159
-
175
)
77
Porteu
 
F
Nathan
 
CF
Mobilizable intracellular pool of p55 (type I) tumor necrosis factor receptors in human neutrophils.
J Leukoc Biol
1992
, vol. 
52
 
1
(pg. 
122
-
124
)
78
Almkvist
 
J
Fäldt
 
J
Dahlgren
 
C
Leffler
 
H
Karlsson
 
A
Lipopolysaccharide-induced gelatinase granule mobilization primes neutrophils for activation by galectin-3 and formylmethionyl-Leu-Phe.
Infect Immun
2001
, vol. 
69
 
2
(pg. 
832
-
837
)
79
de Haas
 
M
Kerst
 
JM
van der Schoot
 
CE
, et al. 
Granulocyte colony-stimulating factor administration to healthy volunteers: analysis of the immediate activating effects on circulating neutrophils.
Blood
1994
, vol. 
84
 
11
(pg. 
3885
-
3894
)
80
Yoon
 
PS
Boxer
 
LA
Mayo
 
LA
Yang
 
AY
Wicha
 
MS
Human neutrophil laminin receptors: activation-dependent receptor expression.
J Immunol
1987
, vol. 
138
 
1
(pg. 
259
-
265
)
81
Sreeramkumar
 
V
Adrover
 
JM
Ballesteros
 
I
, et al. 
 
Neutrophils scan for activated platelets to initiate inflammation. Science (New York). 2014;346(6214):1234-1238
82
Sachs
 
UJ
Andrei-Selmer
 
CL
Maniar
 
A
, et al. 
The neutrophil-specific antigen CD177 is a counter-receptor for platelet endothelial cell adhesion molecule-1 (CD31).
J Biol Chem
2007
, vol. 
282
 
32
(pg. 
23603
-
23612
)
83
Verheugt
 
FW
von dem Borne
 
AE
van Noord-Bokhorst
 
JC
van Elven
 
EH
Engelfriet
 
CP
Serological, immunochemical and immuoncytological properties of granulocyte antibodies.
Vox Sang
1978
, vol. 
35
 
5
(pg. 
294
-
303
)
84
von Vietinghoff
 
S
Tunnemann
 
G
Eulenberg
 
C
, et al. 
NB1 mediates surface expression of the ANCA antigen proteinase 3 on human neutrophils.
Blood
2007
, vol. 
109
 
10
(pg. 
4487
-
4493
)
85
Kuckleburg
 
CJ
Tilkens
 
SB
Santoso
 
S
Newman
 
PJ
Proteinase 3 contributes to transendothelial migration of NB1-positive neutrophils.
J Immunol
2012
, vol. 
188
 
5
(pg. 
2419
-
2426
)
86
Jennette
 
JC
Hoidal
 
JR
Falk
 
RJ
Specificity of anti-neutrophil cytoplasmic autoantibodies for proteinase 3.
Blood
1990
, vol. 
75
 
11
(pg. 
2263
-
2264
)
87
Hu
 
N
Westra
 
J
Huitema
 
MG
, et al. 
Coexpression of CD177 and membrane proteinase 3 on neutrophils in antineutrophil cytoplasmic autoantibody-associated systemic vasculitis: anti-proteinase 3-mediated neutrophil activation is independent of the role of CD177-expressing neutrophils.
Arthritis Rheum
2009
, vol. 
60
 
5
(pg. 
1548
-
1557
)
88
Rarok
 
AA
Stegeman
 
CA
Limburg
 
PC
Kallenberg
 
CG
Neutrophil membrane expression of proteinase 3 (PR3) is related to relapse in PR3-ANCA-associated vasculitis.
J Am Soc Nephrol
2002
, vol. 
13
 
9
(pg. 
2232
-
2238
)
89
Xie
 
Q
Klesney-Tait
 
J
Keck
 
K
, et al. 
Characterization of a novel mouse model with genetic deletion of CD177.
Protein Cell
2015
, vol. 
6
 
2
(pg. 
117
-
126
)
90
Eash
 
KJ
Greenbaum
 
AM
Gopalan
 
PK
Link
 
DC
CXCR2 and CXCR4 antagonistically regulate neutrophil trafficking from murine bone marrow.
J Clin Invest
2010
, vol. 
120
 
7
(pg. 
2423
-
2431
)
91
Köhler
 
A
De Filippo
 
K
Hasenberg
 
M
, et al. 
G-CSF-mediated thrombopoietin release triggers neutrophil motility and mobilization from bone marrow via induction of Cxcr2 ligands.
Blood
2011
, vol. 
117
 
16
(pg. 
4349
-
4357
)
92
Martin
 
C
Burdon
 
PC
Bridger
 
G
Gutierrez-Ramos
 
JC
Williams
 
TJ
Rankin
 
SM
Chemokines acting via CXCR2 and CXCR4 control the release of neutrophils from the bone marrow and their return following senescence.
Immunity
2003
, vol. 
19
 
4
(pg. 
583
-
593
)
93
Nagase
 
H
Miyamasu
 
M
Yamaguchi
 
M
, et al. 
Cytokine-mediated regulation of CXCR4 expression in human neutrophils.
J Leukoc Biol
2002
, vol. 
71
 
4
(pg. 
711
-
717
)
94
Ortega-Gómez
 
A
Perretti
 
M
Soehnlein
 
O
Resolution of inflammation: an integrated view.
EMBO Mol Med
2013
, vol. 
5
 
5
(pg. 
661
-
674
)
95
Hotchkiss
 
RS
Monneret
 
G
Payen
 
D
Sepsis-induced immunosuppression: from cellular dysfunctions to immunotherapy.
Nat Rev Immunol
2013
, vol. 
13
 
12
(pg. 
862
-
874
)
96
Gabrilovich
 
DI
Ostrand-Rosenberg
 
S
Bronte
 
V
Coordinated regulation of myeloid cells by tumours.
Nat Rev Immunol
2012
, vol. 
12
 
4
(pg. 
253
-
268
)
97
Hacbarth
 
E
Kajdacsy-Balla
 
A
Low density neutrophils in patients with systemic lupus erythematosus, rheumatoid arthritis, and acute rheumatic fever.
Arthritis Rheum
1986
, vol. 
29
 
11
(pg. 
1334
-
1342
)
98
Lande
 
R
Ganguly
 
D
Facchinetti
 
V
, et al. 
Neutrophils activate plasmacytoid dendritic cells by releasing self-DNA-peptide complexes in systemic lupus erythematosus.
Sci Transl Med
2011
, vol. 
3
 
73
pg. 
73ra19
 
99
Sagiv
 
JY
Michaeli
 
J
Assi
 
S
, et al. 
Phenotypic diversity and plasticity in circulating neutrophil subpopulations in cancer.
Cell Reports
2015
, vol. 
10
 
4
(pg. 
562
-
573
)
100
Janols
 
H
Bergenfelz
 
C
Allaoui
 
R
, et al. 
A high frequency of MDSCs in sepsis patients, with the granulocytic subtype dominating in gram-positive cases.
J Leukoc Biol
2014
, vol. 
96
 
5
(pg. 
685
-
693
)
101
Ssemaganda
 
A
Kindinger
 
L
Bergin
 
P
, et al. 
Characterization of neutrophil subsets in healthy human pregnancies.
PLoS One
2014
, vol. 
9
 
2
pg. 
e85696
 
102
Wong
 
SL
Demers
 
M
Martinod
 
K
, et al. 
Diabetes primes neutrophils to undergo NETosis, which impairs wound healing.
Nat Med
2015
, vol. 
21
 
7
(pg. 
815
-
819
)
103
Cloke
 
T
Munder
 
M
Taylor
 
G
Müller
 
I
Kropf
 
P
Characterization of a novel population of low-density granulocytes associated with disease severity in HIV-1 infection.
PLoS One
2012
, vol. 
7
 
11
pg. 
e48939
 
104
Fletcher
 
M
Ramirez
 
ME
Sierra
 
RA
, et al. 
l-Arginine depletion blunts antitumor T-cell responses by inducing myeloid-derived suppressor cells.
Cancer Res
2015
, vol. 
75
 
2
(pg. 
275
-
283
)
105
Andzinski
 
L
Kasnitz
 
N
Stahnke
 
S
, et al. 
Type I IFNs induce anti-tumor polarization of tumor associated neutrophils in mice and human.
Int J Cancer
2016
, vol. 
138
 
8
(pg. 
1982
-
1993
)
106
Fridlender
 
ZG
Sun
 
J
Mishalian
 
I
, et al. 
Transcriptomic analysis comparing tumor-associated neutrophils with granulocytic myeloid-derived suppressor cells and normal neutrophils.
PLoS One
2012
, vol. 
7
 
2
pg. 
e31524
 
107
Rodriguez
 
PC
Ernstoff
 
MS
Hernandez
 
C
, et al. 
Arginase I-producing myeloid-derived suppressor cells in renal cell carcinoma are a subpopulation of activated granulocytes.
Cancer Res
2009
, vol. 
69
 
4
(pg. 
1553
-
1560
)
Sign in via your Institution