It is now widely recognized that neutrophils are highly versatile and sophisticated cells that display de novo synthetic capacity and may greatly extend their lifespan. In addition, concepts such as “neutrophil heterogeneity” and “neutrophil plasticity” have started to emerge, implying that, under pathological conditions, neutrophils may differentiate into discrete subsets defined by distinct phenotypic and functional profiles. A number of studies have shown that neutrophils act as effectors in both innate and adaptive immunoregulatory networks. In fact, once recruited into inflamed tissues, neutrophils engage into complex bidirectional interactions with macrophages, natural killer, dendritic and mesenchymal stem cells, B and T lymphocytes, or platelets. As a result of this cross-talk, mediated either by contact-dependent mechanisms or cell-derived soluble factors, neutrophils and target cells reciprocally modulate their survival and activation status. Altogether, these novel aspects of neutrophil biology have shed new light not only on the potential complex roles that neutrophils play during inflammation and immune responses, but also in the pathogenesis of several inflammatory disorders including infection, autoimmunity, and cancer.

Over the last 2 decades, hundreds of reports have clearly documented that neutrophils are highly versatile and sophisticated cells, whose functions go far beyond the elimination of microorganisms. New fascinating aspects of typical activities, as well as novel and unanticipated functions, have been recently attributed to neutrophils. In this context, the notion that the average circulatory lifespan of neutrophils is much longer than previously thought1  and that their longevity increases several-fold during inflammation2  has changed the view under which these cells have long been considered. Accordingly, during their persistence in tissues, neutrophils can exert complex activities, including orchestration of the immune response.3,,,-7  In addition, during the late final phases of acute inflammatory responses, neutrophils become involved in the active induction of inflammation resolution through the production of proresolving lipid mediators.8  Moreover, the findings that neutrophils are also capable of reverse transmigration add another dimension to our understanding of the possible fates of these cells once they have migrated into inflamed tissues.7 

Among unanticipated findings, concepts such as neutrophil heterogeneity and neutrophil plasticity have also started to emerge.9,,-12  The notion that neutrophils are able to shape the inflammatory/immune responses through de novo production of cytokines and a release of preformed proinflammatory mediators, such as proteases and alarmins, is also now well established.13  Finally, the observation that neutrophils can infiltrate lymphoid organs, including spleen and lymph nodes, as well as the demonstration that neutrophils exhibit complex cross-talk with components of the innate and adaptive immune system, which may contribute to the pathogenesis of numerous chronic inflammatory disorders, has renewed interest in these cells within the immunology community.3,,,-7 

In this article, we focus on human neutrophils and summarize the recent findings on their heterogeneity and plasticity, as well as on the role that these cells play in linking the innate and adaptive arms of the immune response and in driving immune-mediated pathologies. Observations recently generated in mouse models will also be described when offering novel information on neutrophil functions.

Evidence for the existence of neutrophil subsets with functional and phenotypic heterogeneity has recently emerged in both humans and mice.7,12  For instance, small fractions of human neutrophils have been reported to express selected molecules under both physiological and pathological conditions, including T-cell receptor-associated variable immunoreceptor,14  olfactomedin 4,12,15  neutrophil antigen B1 (NB1/CD177),12,16,17  or CD49d.18  Similarly, while senescent neutrophils express elevated levels of CXCR4, neutrophils undergoing reverse transendothelial cell migration express elevated levels of intercellular adhesion molecule-1 (ICAM-1) and low levels of CXCR1.7,12  However, the biological and functional implications of these observations remain mostly unclear.

Conversely, detection of distinct circulating subsets displaying neutrophil-like morphology and showing immunosuppressive or proinflammatory functions has been well documented in systemic inflammation, autoimmune diseases, and cancer.9,-11  Some of these neutrophil populations (arbitrarily named here as low-density neutrophils [LDNs]; Figure 1A) were found to settle within the peripheral blood mononuclear cell fraction after density gradient centrifugation of the blood. Immunosuppressive subsets were also identified either within the normal density neutrophil fraction (arbitrarily named here as NDNs; Figure 1A) or within total leukocytes simply obtained by red cell lysis of whole blood (arbitrarily named here as unfractionated neutrophils [UNs]; Figure 1B). Notably, although all these neutrophil populations display a neutrophil-like morphology and the specific granulocyte marker CD66b, their phenotype, maturation/activation status, and function result differently depending on the disease type.9,-11  Such heterogeneity, particularly observed in immunosuppressive subsets, could derive, at least in part, from the fact that both immature and in vivo-activated neutrophils display altered buoyancy and sediment within the peripheral blood mononuclear cell fraction (Figure 1A). In addition, it has been recently shown that, in HIV patients, a subset of immunosuppressive neutrophils, characterized by markedly elevated levels of surface programmed death receptor 1 ligand (PDL-1), are recovered as both PDL-1high LDNs and PDL-1high NDNs after density gradient centrifugation.19  These observations would suggest that certain phenotypic/functional changes, instead of being intrinsic features of specialized neutrophil subsets, might occur in total circulating neutrophils as a result of systemic inflammation. Clearly, this issue might be solved when a careful comparison of UNs, LDNs, and NDNs from the same diseased individuals is performed.

Figure 1

Main neutrophil subsets identified in the peripheral blood of patients with diseases. Mature neutrophils from healthy donors, after blood centrifugation over density gradients, typically sediment on top of red cells (arbitrarily indicated as NDNs in A). By contrast, immature neutrophils, as well as mature neutrophils activated in vivo under inflammatory settings, display altered cell buoyancy properties and thus sediment within the mononuclear cell fraction (arbitrarily indicated as LDNs in A). According to the literature, LDNs may include (1) immature neutrophils found in patients with sepsis and with function mostly undefined; (2) immunosuppressive neutrophil subsets, also known as G-MDSCs, found in cancer and HIV patients or in G-CSF–treated donors and displaying either immature or activated phenotypes; and (3) proinflammatory neutrophil subsets found in patients with autoimmune diseases, recently named LDGs and consisting of a mixed population of immature and mature cells. Circulating mature neutrophil subsets, displaying immunosuppressive properties, have also been identified within either the NDN fraction in cancer and HIV patients (A) or the total leukocytes (obtained after red cell lysis of whole blood) from healthy volunteers administered with endotoxin or from patients with severe injury, cancer, or HIV infection (arbitrarily indicated as UNs in B).

Figure 1

Main neutrophil subsets identified in the peripheral blood of patients with diseases. Mature neutrophils from healthy donors, after blood centrifugation over density gradients, typically sediment on top of red cells (arbitrarily indicated as NDNs in A). By contrast, immature neutrophils, as well as mature neutrophils activated in vivo under inflammatory settings, display altered cell buoyancy properties and thus sediment within the mononuclear cell fraction (arbitrarily indicated as LDNs in A). According to the literature, LDNs may include (1) immature neutrophils found in patients with sepsis and with function mostly undefined; (2) immunosuppressive neutrophil subsets, also known as G-MDSCs, found in cancer and HIV patients or in G-CSF–treated donors and displaying either immature or activated phenotypes; and (3) proinflammatory neutrophil subsets found in patients with autoimmune diseases, recently named LDGs and consisting of a mixed population of immature and mature cells. Circulating mature neutrophil subsets, displaying immunosuppressive properties, have also been identified within either the NDN fraction in cancer and HIV patients (A) or the total leukocytes (obtained after red cell lysis of whole blood) from healthy volunteers administered with endotoxin or from patients with severe injury, cancer, or HIV infection (arbitrarily indicated as UNs in B).

Close modal

LDN subsets

LDNs have been detected not only in patients with autoimmune disorders,11  sepsis,20  HIV infections,19,21  and cancer,10  but also in patients with graft-versus-host disease undergoing extracorporeal photopheresis treatments,22  pregnant women,23  and in healthy donors receiving granulocyte-colony-stimulating factor (G-CSF) for stem cell mobilization.24,25  LDNs described in sepsis seem to be mostly immature and functionally poorly characterized (Figure 1A).20  LDNs isolated from patients suffering from autoimmune diseases, such as systemic lupus erythematosus (SLE) and psoriasis, have been instead more recently defined as low-density granulocytes (LDGs) (Figure 1A) and mostly consist of a mixed population of immature and mature cells with neutrophil-like morphology.11  LDGs display proinflammatory functions and induce vascular damage via their enhanced ability to release inflammatory molecules and autoantigens, as well as neutrophil extracellular traps (NETs, which result from the extrusion of nuclear DNA together with antimicrobial proteins26 ).11  By contrast, LDNs identified in solid tumors, G-CSF-treated donors, and HIV patients are generally defined as granulocytic myeloid-derived suppressor cells (G-MDSCs) because of their immunosuppressive functions (Figure 1A).10,19,21,24,25  Confusion still exists regarding the phenotype of G-MDSCs, with some reports describing these cells as activated, whereas others describe them as immature cells with neutrophil-like morphology.10  The immunosuppressive function exerted by G-MDSCs is mainly defined according to their ability to suppress T-cell activation/proliferation, and it has been shown to be primarily mediated by overproduction of arginase 1 and/or reactive oxygen species (ROS).10  It should be pointed out that the relationship existing among G-MDSCs and the other identified immunosuppressive neutrophil subsets (see below) is a matter of extensive investigation in the field.9,10  Indeed, whether G-MDSCs are specialized subsets of neutrophils or originate through an altered process of granulopoiesis is still unclear.10  The recent observations that monocytic MDSCs (Mo-MDSCs) can differentiate into G-MDSCs in tumor-bearing mice and in patients with multiple myeloma27  have further complicated the scenario. It has been also suggested that splenic granulocyte and macrophage progenitors are significantly increased in patients with invasive cancer.28  The fact that freshly isolated splenic neutrophils from healthy donors were shown to inhibit the proliferation of CD4+ T cells29  raises the possibility that, similar to mice, human G-MDSCs might originate through a process of inflammation-induced extramedullary granulopoiesis.

Other neutrophil immunosuppressive subsets

To date, several NDNs inhibiting T-cell proliferation through diverse mechanisms have been identified (Figure 1A), including the CD15+CD16low and the CCL2-producing subsets detected in cancer patients30,31  and the PDL-1high subset detected in HIV patients.19  The presence of immunosuppressive UNs has been instead reported in healthy volunteers administered with endotoxin or in patients with severe injury,32,33  cancer,34  giant-cell arteritis under glucocorticoid treatment,35  and HIV-1 infection19  (Figure 1B). However, at least to our knowledge, only Pillay et al32  have formally sorted a subset of activated (CD16brightCD62Ldim) mature neutrophils from total leukocytes and shown it to effectively inhibit T-cell responses through macrophage-1 antigen (Mac-1)- and ROS-dependent mechanisms.32 

The concept that neutrophils may display a previously unanticipated plasticity derives from observations on their ability, under inflammatory settings, to differentiate into other myeloid cell types. Accordingly, the fact that neutrophils can acquire antigen presenting (APC)-like properties and dendritic cell (DC) characteristics on long-term incubation with discrete cytokine combinations, or be reprogrammed into macrophages, has been known for a long time.3,,-6,36  More recently, both immature and mature mouse neutrophils have been shown to differentiate into hybrid populations showing dual phenotypes and properties of both neutrophils and DCs, either when cultured in vitro with granulocyte macrophage-colony-stimulating factor (GM-CSF) or when transferred to inflammatory settings in vivo.37  This study has been corroborated in humans, in whom purified bone marrow-derived neutrophils were demonstrated to differentiate into a hybrid population characterized by the expression of both neutrophil and DC markers on in vitro treatment with GM-CSF, tumor necrosis factor-α (TNFα), and interleukin-4 (IL-4) for 7 days.37  Further depicting their extraordinary plasticity, a very small fraction of neutrophils has been shown to survive in vitro without the addition of exogenous cytokines or growth factors and, after 7 days of culture, ultimately transform into giant phagocytic cells.38  However, the potential significance of these giant neutrophils to inflammatory/anti-inflammatory processes in vivo remains to be elucidated. In this context, it is plausible that with the development of very efficient cell isolation techniques and the increased availability of neutrophils purified from various compartments, such as spleen, peritoneal exudates, lungs, oral cavity, skin, bone marrow, cord blood, and placenta, other neutrophil subpopulations with specialized functions will be discovered.

It has been demonstrated that human neutrophils, other than interacting with nonimmune cell types such as platelets39  and mesenchimal stem cells,3  can establish, in vitro and in vivo, cross-talk with innate immune cells, such as DCs, monocytes, macrophages, and natural killer (NK) cells, as well as with adaptive immune cells, such as T and B cells, or related subpopulations3,,-6  (summarized in Tables 1 and 2). As highlighted below, by establishing such bidirectional interactions, neutrophils receive signals modulating their survival and effector functions on the one hand, whereas they initiate, amplify, and/or suppress innate or adaptive immune effector responses on the other hand. Studies made in a variety of experimental animal models (listed in supplemental Tables 1 and 2 on the Blood Web site) not only have corroborated the existence of neutrophil-centered cross-talk, but have also highlighted their pathophysiological significance.

Table 1

Cross-talk between human neutrophils and innate immune cells

Neutrophil cross-talk withCross-talk outcomeReferences
DCs: moDCs Reduced CD40, CD80, and CD86 expression, and decreased ability to stimulate T cell proliferation in modcs engulfing apoptotic and/or necrotic neutrophils 
 Enhancement, by fMLF-, TNFα- or LPS-activated neutrophils, of moDC maturation and ability to promote T cell proliferation and Th1 polarization, via Mac-1/DC-SIGN and/or Mac-1/CEACAM1 interactions and TNFα release 2-3 
 Enhancement, by apoptotic and/or live neutrophils, of moDC maturation and ability to promote T cell proliferation, via CD18-mediated contact dependent mechanisms and release of soluble factors 
 Inhibition, by non-infected apoptotic neutrophils, of M tuberculosis-induced moDC maturation and ability to induce lymphocyte proliferation. Enhancement of moDC ability to drive lymphocyte proliferation by M tuberculosis-induced apoptotic neutrophils 
 Promotion of CD4+FOXP3+ Treg differentiation by moDCs treated with neutrophil-derived elastase 6-7 
 Inhibition, by neutrophil-derived EVs, of moDC maturation and capacity to induce T cell proliferation 
 Enhancement, by BCG-infected neutrophils, of moDC maturation and ability to recall reactivity of T cells isolated from vaccinated donors, via cell-contact dependent mechanisms 
 Modulation of DC functions by neutrophil-derived alarmins (defensins, cathelicidin, lactoferrin and high-mobility group box-1 protein) Reviewed in Dumitru et al10  
 moDC internalization and cross-presentation of antigens previously processed by neutrophils 11 
 Inhibition of moDC maturation and cytokine production by neutrophil-derived myeloperoxidase 12 
pDCs Enhancement of pDC-derived IFNα by NETs released by SLE neutrophils containing self-DNA in complex with antimicrobial peptides 13-14 
 Enhancement of pDC-derived IFNα by NETs containing DNA complexed with secretory leukocyte protease inhibitor and neutrophil elastase or cathepsin G 15-16 
slanDCs Enhancement, by neutrophils, of slanDC-derived IL-12p70, via CD18/ICAM-1 interactions 17 
 Enhancement of neutrophil and slanDC survival by reciprocal interactions occurring through contact-dependent mechanisms 18 
Macrophages Inhibition of proinflammatory cytokine production in macrophages engulfing apoptotic neutrophils Reviewed in Bowers et al19  
 Inhibition of macrophage activation, cytokine production and phagocytosis by neutrophil-derived EVs 20-21 
 Enhancement of macrophage antimicrobial activity by the uptake of antimicrobial peptides from neutrophils Reviewed in Rieber et al22  
 Enhancement of macrophage phagocytosis and reactive oxygen species (ROS) production by neutrophil-derived primary granule proteins 23-24 
 Enhancement of macrophage-derived cytokines by M tuberculosis-induced NETs 25 
 Activation of NLRP3 inflammasome and induction of IL-1β and IL-18 release in macrophages by NETs from LPS-activated neutrophils or resting LDGs from SLE patients 26 
 Enhancement, by PMA-induced NETs, of cytokine production in LPS-stimulated macrophages. Clearance of PMA-induced NETs by resting macrophages 27 
NK cells Modulation, by NK cells, of neutrophil survival, activation and HB-EGF release, via GM-CSF, IFNγ and TNFα release as well as contact-dependent mechanisms 28-29 
 Enhancement, by neutrophils, of NK-derived IFNγ, via ICAM-3 and CD11d/CD18 interactions 17,30 
 Modulation of NK cell functions by neutrophil-derived molecules, such as arginase-1, serine proteases, defensins and ROS Reviewed in Tsuda et al31  
 Impairment of NK cell maturation and functions in neutropenic patients, additionally supported by in vivo experimental models 32 
 Induction, by NK cells, of caspase-dependent neutrophil apoptosis, via NKp46- and Fas-dependent mechanisms 33 
 Enhancement, by NK cells, of neutrophil antifungal activity. Inhibition of NK cell activation by neutrophils in the presence of Candida albicans 34 
 Inhibition of pro-inflammatory, and enhancement of anti-inflammatory, cytokine production in NK cells, by neutrophil–derived EVs 35 
 Induction, by NK cells, of apoptosis in galactosaminogalactan (GG)-treated neutrophils, via NKG2D mediated interactions 36 
ILCs Enhancement of neutrophil B-cell helper functions by splenic ILCs, via GM-CSF 37 
Neutrophil cross-talk withCross-talk outcomeReferences
DCs: moDCs Reduced CD40, CD80, and CD86 expression, and decreased ability to stimulate T cell proliferation in modcs engulfing apoptotic and/or necrotic neutrophils 
 Enhancement, by fMLF-, TNFα- or LPS-activated neutrophils, of moDC maturation and ability to promote T cell proliferation and Th1 polarization, via Mac-1/DC-SIGN and/or Mac-1/CEACAM1 interactions and TNFα release 2-3 
 Enhancement, by apoptotic and/or live neutrophils, of moDC maturation and ability to promote T cell proliferation, via CD18-mediated contact dependent mechanisms and release of soluble factors 
 Inhibition, by non-infected apoptotic neutrophils, of M tuberculosis-induced moDC maturation and ability to induce lymphocyte proliferation. Enhancement of moDC ability to drive lymphocyte proliferation by M tuberculosis-induced apoptotic neutrophils 
 Promotion of CD4+FOXP3+ Treg differentiation by moDCs treated with neutrophil-derived elastase 6-7 
 Inhibition, by neutrophil-derived EVs, of moDC maturation and capacity to induce T cell proliferation 
 Enhancement, by BCG-infected neutrophils, of moDC maturation and ability to recall reactivity of T cells isolated from vaccinated donors, via cell-contact dependent mechanisms 
 Modulation of DC functions by neutrophil-derived alarmins (defensins, cathelicidin, lactoferrin and high-mobility group box-1 protein) Reviewed in Dumitru et al10  
 moDC internalization and cross-presentation of antigens previously processed by neutrophils 11 
 Inhibition of moDC maturation and cytokine production by neutrophil-derived myeloperoxidase 12 
pDCs Enhancement of pDC-derived IFNα by NETs released by SLE neutrophils containing self-DNA in complex with antimicrobial peptides 13-14 
 Enhancement of pDC-derived IFNα by NETs containing DNA complexed with secretory leukocyte protease inhibitor and neutrophil elastase or cathepsin G 15-16 
slanDCs Enhancement, by neutrophils, of slanDC-derived IL-12p70, via CD18/ICAM-1 interactions 17 
 Enhancement of neutrophil and slanDC survival by reciprocal interactions occurring through contact-dependent mechanisms 18 
Macrophages Inhibition of proinflammatory cytokine production in macrophages engulfing apoptotic neutrophils Reviewed in Bowers et al19  
 Inhibition of macrophage activation, cytokine production and phagocytosis by neutrophil-derived EVs 20-21 
 Enhancement of macrophage antimicrobial activity by the uptake of antimicrobial peptides from neutrophils Reviewed in Rieber et al22  
 Enhancement of macrophage phagocytosis and reactive oxygen species (ROS) production by neutrophil-derived primary granule proteins 23-24 
 Enhancement of macrophage-derived cytokines by M tuberculosis-induced NETs 25 
 Activation of NLRP3 inflammasome and induction of IL-1β and IL-18 release in macrophages by NETs from LPS-activated neutrophils or resting LDGs from SLE patients 26 
 Enhancement, by PMA-induced NETs, of cytokine production in LPS-stimulated macrophages. Clearance of PMA-induced NETs by resting macrophages 27 
NK cells Modulation, by NK cells, of neutrophil survival, activation and HB-EGF release, via GM-CSF, IFNγ and TNFα release as well as contact-dependent mechanisms 28-29 
 Enhancement, by neutrophils, of NK-derived IFNγ, via ICAM-3 and CD11d/CD18 interactions 17,30 
 Modulation of NK cell functions by neutrophil-derived molecules, such as arginase-1, serine proteases, defensins and ROS Reviewed in Tsuda et al31  
 Impairment of NK cell maturation and functions in neutropenic patients, additionally supported by in vivo experimental models 32 
 Induction, by NK cells, of caspase-dependent neutrophil apoptosis, via NKp46- and Fas-dependent mechanisms 33 
 Enhancement, by NK cells, of neutrophil antifungal activity. Inhibition of NK cell activation by neutrophils in the presence of Candida albicans 34 
 Inhibition of pro-inflammatory, and enhancement of anti-inflammatory, cytokine production in NK cells, by neutrophil–derived EVs 35 
 Induction, by NK cells, of apoptosis in galactosaminogalactan (GG)-treated neutrophils, via NKG2D mediated interactions 36 
ILCs Enhancement of neutrophil B-cell helper functions by splenic ILCs, via GM-CSF 37 

References to Table 1 are listed in the supplemental Materials.

Table 2

Cross-talk between human neutrophils and adaptive immune cells

Neutrophil cross-talk withCross-talk outcomeReferences
T cells: CD4+ and/ or CD8+ T cells Induction, by antigen-pulsed neutrophils, of lymphocyte proliferation in a non-MHC-restricted fashion 
 Induction of antigen specific T cell activation by neutrophil precursors that have acquired DC-like properties after treatment with GM-CSF, IL-4 and TNFα 
 MHC class II-restricted antigen presentation to T cells by GM-CSF plus IFNγ-treated neutrophils 3-4 
 Enhancement of T cell proliferation by neutrophils from patients with Staphylococcus aureus infections 
 Cross-presentation, by neutrophils, of soluble antigens to CD8+T cells 
 Neutrophil and T cell reciprocal modulation of lifespan and function, via cytokine release and/or cell-contact dependent mechanisms Reviewed in Kolaczkowska and Kubes7  
 Enhancement of neutrophil survival and activation by anti-CD3-activated CD4+T and, more potently, CD8+T cells, via TNFα, IFNγ and GM-CSF release 
 Recruitment of Th1 and Th17, but not Th2, cells by neutrophils. Enhancement of neutrophil recruitment, survival and activation by Th17 cells, mostly via CXCL8/IL-8 and GM-CSF release, respectively 
 Reduction of viability, activation and proliferation of CD4+T cells by unstimulated neutrophils 10 
 Inhibition of T cell activation, proliferation and function by activated neutrophils, via release of arginase-1, production of ROS and/or cell-contact dependent mechanisms 11-13 
 Inhibition of CD4+T cell activation and proliferation by splenic neutrophils 14 
 Inhibition of T cell activation and proliferation by CD11cbright/CD62Ldim/CD11bbright/CD16bright neutrophils isolated from individuals injected with endotoxin, via Mac-1-, ROS- and/or PDL-1 (B7-H1)- dependent mechanisms 15-16 
 Inhibition of T cell activation and proliferation by activated mature neutrophils or granulocytic myeloid-derived suppressor cells (G-MDSCs) from cancer patients, mainly via arginase-1 and ROS overproduction Reviewed in Wang et al17  
 Priming of T cell responses to specific antigens by NETs 18 
Tregs Recruitment of neutrophils by Tregs, via CXCL8/IL-8 release 19 
 Promotion of CD4+FOXP3+ Treg differentiation by moDCs treated with neutrophil-derived elastase 20-21 
γδ T cells Killing of LPS-treated neutrophils by γδ T cells, via contact dependent mechanisms mediated by surface heat shock protein-72 22 
 Enhancement of neutrophil migration, phagocytosis and α-defensin release by zoledronic acid-activated γδ T cells, via release of soluble factors 23 
 Enhancement of neutrophil survival and activation by phosphoantigen activated-γδ T cells. Activation of γδ T cells by neutrophils harboring phagocytosed bacteria 24 
 Suppression of spontaneous and phosphoantigen-induced activation in γδ T cells by neutrophils, via ROS production 25 
 Inhibition of zoledronate-mediated Vγ9Vδ2 T cell activation by neutrophils, via hydrogen peroxide, serine proteases and arginase-1 release 26 
 Enhancement of the migration, survival and proliferation of tumor infiltrated G-MDSCs by activated γδ T isolated from tumor tissues 27 
iNKT cells Inhibition of iNKT-derived IFNγ and iNKT cytotoxicity by neutrophils, via contact dependent mechanisms 28 
B cells Enhancement of B cell survival and proliferation by G-CSF-or antineutrophil cytoplasmic antibody (ANCA)-stimulated neutrophils, via BAFF production 29-30 
 Enhancement of plasma cell survival by neutrophils, via APRIL secretion 31 
 Induction of immunoglobulin (Ig) class switching, somatic hypermutation and antibody production in MZ B cells by splenic neutrophils, via BAFF, APRIL and IL-21 production 14 
Neutrophil cross-talk withCross-talk outcomeReferences
T cells: CD4+ and/ or CD8+ T cells Induction, by antigen-pulsed neutrophils, of lymphocyte proliferation in a non-MHC-restricted fashion 
 Induction of antigen specific T cell activation by neutrophil precursors that have acquired DC-like properties after treatment with GM-CSF, IL-4 and TNFα 
 MHC class II-restricted antigen presentation to T cells by GM-CSF plus IFNγ-treated neutrophils 3-4 
 Enhancement of T cell proliferation by neutrophils from patients with Staphylococcus aureus infections 
 Cross-presentation, by neutrophils, of soluble antigens to CD8+T cells 
 Neutrophil and T cell reciprocal modulation of lifespan and function, via cytokine release and/or cell-contact dependent mechanisms Reviewed in Kolaczkowska and Kubes7  
 Enhancement of neutrophil survival and activation by anti-CD3-activated CD4+T and, more potently, CD8+T cells, via TNFα, IFNγ and GM-CSF release 
 Recruitment of Th1 and Th17, but not Th2, cells by neutrophils. Enhancement of neutrophil recruitment, survival and activation by Th17 cells, mostly via CXCL8/IL-8 and GM-CSF release, respectively 
 Reduction of viability, activation and proliferation of CD4+T cells by unstimulated neutrophils 10 
 Inhibition of T cell activation, proliferation and function by activated neutrophils, via release of arginase-1, production of ROS and/or cell-contact dependent mechanisms 11-13 
 Inhibition of CD4+T cell activation and proliferation by splenic neutrophils 14 
 Inhibition of T cell activation and proliferation by CD11cbright/CD62Ldim/CD11bbright/CD16bright neutrophils isolated from individuals injected with endotoxin, via Mac-1-, ROS- and/or PDL-1 (B7-H1)- dependent mechanisms 15-16 
 Inhibition of T cell activation and proliferation by activated mature neutrophils or granulocytic myeloid-derived suppressor cells (G-MDSCs) from cancer patients, mainly via arginase-1 and ROS overproduction Reviewed in Wang et al17  
 Priming of T cell responses to specific antigens by NETs 18 
Tregs Recruitment of neutrophils by Tregs, via CXCL8/IL-8 release 19 
 Promotion of CD4+FOXP3+ Treg differentiation by moDCs treated with neutrophil-derived elastase 20-21 
γδ T cells Killing of LPS-treated neutrophils by γδ T cells, via contact dependent mechanisms mediated by surface heat shock protein-72 22 
 Enhancement of neutrophil migration, phagocytosis and α-defensin release by zoledronic acid-activated γδ T cells, via release of soluble factors 23 
 Enhancement of neutrophil survival and activation by phosphoantigen activated-γδ T cells. Activation of γδ T cells by neutrophils harboring phagocytosed bacteria 24 
 Suppression of spontaneous and phosphoantigen-induced activation in γδ T cells by neutrophils, via ROS production 25 
 Inhibition of zoledronate-mediated Vγ9Vδ2 T cell activation by neutrophils, via hydrogen peroxide, serine proteases and arginase-1 release 26 
 Enhancement of the migration, survival and proliferation of tumor infiltrated G-MDSCs by activated γδ T isolated from tumor tissues 27 
iNKT cells Inhibition of iNKT-derived IFNγ and iNKT cytotoxicity by neutrophils, via contact dependent mechanisms 28 
B cells Enhancement of B cell survival and proliferation by G-CSF-or antineutrophil cytoplasmic antibody (ANCA)-stimulated neutrophils, via BAFF production 29-30 
 Enhancement of plasma cell survival by neutrophils, via APRIL secretion 31 
 Induction of immunoglobulin (Ig) class switching, somatic hypermutation and antibody production in MZ B cells by splenic neutrophils, via BAFF, APRIL and IL-21 production 14 

iNKT, invariant NKT cells.

References to Table 2 are listed in the supplemental Materials.

Cross-talk between neutrophils and innate immune cells

Neutrophils and DCs.

Neutrophils can modulate, either positively or negatively, the survival, maturation status, and functions of monocyte-derived DCs (moDCs), 6-sulfo LacNAc+ DCs (slanDCs), and plasmacytoid DCs (pDCs) (Table 1; supplemental Table 1).3,5,6,40  Survival and other effector activities of neutrophils can be modulated by DCs in a reciprocal manner, as for instance demonstrated to occur under neutrophil/slanDC coculture conditions (Table 1). Neutrophil-DC cross-talk is regulated either by contact-dependent mechanisms or via the release of cell-derived products, such as cytokines, inflammatory mediators, and, in the case of neutrophils, extracellular vesicles (EVs, also known as microparticles or ectosomes).3  EVs are cell-derived membrane vesicles of heterogeneous size, containing hundreds of distinct proteins, lipids, and microRNAs,41  that have been shown to mainly down-modulate the inflammatory responses in activated DCs (Table 1). In this context, recent publications have reported that NETs can also mediate cross-talk between neutrophils and several DC subsets and suggested that they represent a novel mechanism involved in the pathogenesis of SLE42,43  and psoriasis in humans,44,45  or type 1 diabetes (T1D)46  and autoimmune vasculitis47  in mice. Interestingly, inhibitory effects of NETs on lipopolysaccharide (LPS)-induced DC maturation and cytokine production have also been observed, given that NET-treated DCs inhibit T-lymphocyte proliferation and skew T-cell differentiation toward a T-helper type 2 (Th2) phenotype (L. S. Barrientos, V. Marin-Esteban, and S. Chollet-Martin, unpublished data, 2014). Likewise, myeloperoxidase release by neutrophils has recently been identified as another manner through which DC activation and function can be negatively modulated.48 

Neutrophils and macrophages.

Neutrophils may positively modulate cytokine production and microbicidal activity in macrophages49  (Table 1; supplemental Table 1), for instance, via NET release, as observed in models using human neutrophils isolated from SLE patients or activated in vitro with LPS or Mycobacterium tuberculosis (Table 1). Human neutrophils may also inhibit macrophage activation and/or macrophage-derived TNFα and CXCL8 by either efferocytosis-dependent mechanisms or EV release (Table 1).

Neutrophils and NK cells.

It has been widely documented that activated NK cells secrete multiple cytokines (including GM-CSF, interferon-γ [IFNγ], and TNFα) able to influence the activation status of neutrophils under coculture conditions, by extending their lifespan, by up-regulating their expression of activation markers, and by potentiating their capacity to phagocytose, to produce ROS, and to synthesize heparin binding-epidermal growth factor (HB-EGF) (Table 1; supplemental Table 1).3,50  Evidence that NK cells may instead induce neutrophil apoptosis under specific coculture conditions, either via NKp46- and Fas-dependent mechanisms or via interactions between NKG2D and MHC class I chain-related molecule A, also exist in the literature (Table 1). Reciprocally, neutrophils modulate NK cell survival, proliferation, cytotoxic activity and production of IFNγ, via the generation of prostaglandins or ROS, through release of granule components or via contact-dependent mechanisms (Table 1).3,50  In addition, recent observations suggest that, by releasing EVs, neutrophils negatively modulate NK cell functions, in particular skewing NK-derived cytokines from a proinflammatory to an anti-inflammatory profile.51  The relevance of the in vitro findings on cross-talk between human neutrophils and NK cells has been highlighted by studies proving that, under steady state, neutrophils are crucial for NK-cell development in both humans and mice.52  Additional evidence that cross-talk between these 2 cell types can also occur in vivo has recently been shown in a mouse model of systemic fungal infection, in which an IL-17-NK cell-GM-CSF axis was found to be critical for the modulation of neutrophil fungicidal activity.53  In line with these findings, in vitro coculture of human NK cells and neutrophils in the presence of Candida albicans resulted in a NK-mediated enhancement of neutrophil antifungal activity, concomitantly with a neutrophil-mediated inhibition of NK-cell activation,54  suggesting that interactions between these 2 cell types might serve to reciprocally modulate their responses.

Neutrophils and innate lymphoid cell types.

Mucosal tissues contain a large number of innate lymphocytes now collectively referred to as innate lymphoid cells (ILCs).55  Evidence has been emerging that the various ILC subsets play a key role in the orchestration of immunity to infection and in the pathogenesis of allergic and autoimmune diseases.55  In this context, a recently described human splenic ILC subset, with mucosa-like properties, has been found to activate marginal zone (MZ) B cells, either directly via B cell-activating factor (BAFF), CD40 ligand, and Delta-like 1 production, or indirectly by producing GM-CSF.56  The latter cytokine, in turn, has been shown to potentiate the ability of splenic neutrophils to directly activate B-cell functions.56  It is likely that additional cross-talk between neutrophils and other ILC subsets will be soon uncovered.

Cross-talk between neutrophils and adaptive immune cells

Neutrophils and B cells.

Neutrophils can modulate B-cell functions (Table 2; supplemental Table 2) in part through the production of cytokines crucial for B-cell survival, maturation, and differentiation, such as BAFF and a proliferation-inducing ligand (APRIL).57  The concept that human neutrophils can function as B-cell helpers has emerged by the demonstration that freshly isolated splenic neutrophils induce a T cell-independent antibody response by MZ B cells, via BAFF, APRIL, and IL-21 release.29  Intriguingly, B-cell helper functions by splenic neutrophils have not been reproduced by other groups,58  likely due to differences in the protocol used to collect, store, and process the splenic tissue and/or in the purity/manipulation of the neutrophil or B-cell populations, as pointed out by the same authors.58  Conversely, recent in vivo data confirm that splenic mouse neutrophils support preimmune immunoglobulin (Ig)G3 production, as well as postimmune IgM, IgG2b, IgG2c, and IgG3 responses to repetitive immunization with polysaccharides such as 2,4,6-trinitrophenyl-ficoll (A. Chorny and A. Cerutti, unpublished data, 2014). Neutrophils have also been proposed to play a role in the pathogenesis of B-cell lymphomas, either through the production of APRIL59  or, based on studies in mice, through NET release60  (supplemental Table 2).

Neutrophils and T cells.

The ability of both naïve/polarized T cells and neutrophils to reciprocally influence their effector functions under coculture conditions, either via chemokine and cytokine production or contact-dependent mechanisms, have been extensively discussed in previous reviews3,5,6,61  (Table 2; supplemental Table 2). Earlier studies, however, had already uncovered that T cell-derived cytokines, particularly IFNγ, GM-CSF, and TNFα, function as priming agents for human neutrophils, namely as factors able to greatly enhance neutrophil responsiveness on secondary stimulation.62  A variety of fast neutrophil responses involved in inflammatory and immune processes may be primed, ultimately leading to increased oxidative metabolism (eg, ROS production), degranulation, surface receptor expression, phagocytosis, and cytotoxicity.62,63  At the molecular level, neutrophil priming by T cell-derived cytokines may occur through multiple, but not yet completely identified, mechanisms, including de novo gene induction, post-translational modifications of specific effector proteins, and/or modification of signaling pathways.62,63  Neutrophil-T cell bidirectional interactions are, in turn, supported by original reports suggesting that humans and mouse neutrophils can positively modulate T-cell functions, either indirectly, via DC activation, or directly, through chemokine/cytokine production, APC-like properties, or NET-mediated mechanisms.3,-5  However, because several studies also suggest that, as described above, activated neutrophils or neutrophil subsets have a predominantly suppressive function on T cells,9,10  it is clear that the outcome of neutrophil-T cell cross-talk is a function of the experimental settings during which these interactions occur (Table 2; supplemental Table 2).

Controversial observations are emerging on the cross-talk occurring between human neutrophils and γδ T cells. Although initial evidence suggested that γδ T cells are involved in neutrophil killing to limit host tissue damage during sepsis, more recent studies report that γδ T cells positively modulate neutrophil recruitment, activation, and survival (Table 2; supplemental Table 2). Similarly, although human neutrophils were initially shown to stimulate γδ T cells,64  more recent evidence suggests that neutrophils may, mostly via ROS production, negatively modulate spontaneous and phosphoantigen-induced γδ T-cell activation65  or contribute to the loss of peripheral blood Vγ9Vδ2 T cells observed after long-term or high-dose administration of zoledronate.66  Considering that the stimulatory functions of neutrophils on γδ T cells were reported to occur only in the presence of autologous monocytes,64  it is plausible that technical differences, intrinsic to the experimental conditions used, might influence the resulting neutrophil-γδ T-cell interactions. Very recently, human γδ T cells have also shown to contribute to the recruitment, survival, and proliferation of tumor-infiltrated G-MDCs in colorectal cancer, through the release of IL-17A, CXCL8, GM-CSF, and TNFα.67  Strikingly, it has been recently observed that unconventional T cells, including γδ T cells and mucosal-associated invariant T cells, induce neutrophils to acquire a unique activated phenotype with APC properties not only for CD4+ but also for CD8+ T cells (M. S. Davey, B. Moser, and M. Eberl, unpublished data, 2014).

Finally, neutrophils have been shown to impair iNKT-cell function, in both mice and humans, through cell-cell contact-dependent mechanisms (Table 2; supplemental Table 2).

Most of our knowledge on neutrophil functions in human diseases derives from correlative studies on cells isolated from patients. However, because the isolation procedures, cell purity, or drug treatment these patients are receiving often limit the reliability of the observations made, the role of neutrophils in disease pathogenesis is mostly extrapolated from studies in experimental animal models. In this context, remarkable improvements have been made in developing tools suitable for studying the immunoregulatory role of neutrophils in vivo, including the availability of neutrophil-specific depleting antibodies (such as the anti-Ly6G/1A8 monoclonal antibody), the generation of mice carrying the conditional deletion of the loxP-flanked allele of interest under the control of neutrophil-specific promoters, as well as of humanized mice carrying functional human neutrophils. In any case, crucial neutrophil functions in humans, such as their indispensable role in providing protection against bacterial and fungal infections have been substantiated by studying patients affected by genetically determined neutrophil disorders.68  In this context, novel mutations causing neutrophil defects, specifically in genes enconding caspase recruitment domain-containing protein 9,69  IFN-stimulated gene 15,70  vacuolar protein sorting 45,71  and syntaxin–binding protein 272  have been recently identified. For instance, the lack of Mycobacterium-induced secretion of INF-stimulated gene 15, an IFNα/β-inducible ubiquitin-like intracellular protein, mostly by neutrophils, determined a reduced ability of lymphocytes and, especially, NK cells, to produce IFNγ, pointing for a novel player involved in the neutrophil-NK cell cross-talk for optimal antimycobacterial immunity.70 

A growing amount of evidence suggests that neutrophils play critical roles in chronic inflammatory conditions, such as atherosclerosis, type 2 diabetes, vascular, liver, and intestinal inflammation, and local and systemic allergic reactions.5,-7,73,74  Herein, we focus on recent discoveries concerning novel neutrophil functions in infection, autoimmunity, and cancer.

Neutrophils in infection

The importance of neutrophils in host defense and infectious diseases has been underscored by recent discoveries demonstrating that their function is essential in combating intracellular bacteria (such as mycobacteria or Brucella abortus),5,75  parasites,5,76,77  and pathogenic viruses (such as human immunodeficiency virus-1 or influenza virus).78,79  The latter data are further supported by the documentation that, in addition to Toll-like receptors (TLRs), C-type lectin receptors,80,81  and inflammasome components,82  neutrophils also express cytoplasmic sensors. The latter include receptors recognizing pathogen-derived intracellular RNA, such as retinoic acid inducible-I and melanoma differentiation-associated antigen 5, and receptors recognizing pathogen-derived intracellular DNA, such as IFN-inducible protein 16, leucine-rich repeat in Flightless I interacting protein-1, DEAD (Asp-Glu-Ala-Asp) box polypeptide 41, or stimulator of interferon genes.83  In such regard, the description of surface TLR9 in neutrophils points for additional mechanisms whereby neutrophils could sense microbial-derived hypomethylated CpG DNA.84  Similarly, TLR4, the receptor for LPS, was shown to function, in neutrophils, also as receptor for shiga toxin,85  which is responsible for the development of hemolytic uremic syndrome. The recent identification of CD11b as a cellular receptor for leukocidin A/B, a toxin that contributes to the S aureus-mediated killing of neutrophils,86  exemplifies how much our understanding of the neutrophil-pathogen interaction is increasing. Furthermore, recent findings have demonstrated that also neutrophil-derived EVs may contribute to restrict bacterial growth and dissemination, in addition to NETs.87,-89  Accordingly, depending on the nature of the stimulus activating neutrophils, various EVs types have been identified, all with different protein composition and biological properties, as well as exerting antibacterial effects distinct from those of NETs.87,90,91  However, whether neutrophils kill bacteria through NET and EVs release, or instead entrap them only, remains to be definitively solved.5,89,91 

Intriguingly, it is now appreciated that neutrophils may actually favor disease progression, depending on the infectious agent.3  For example, it has been shown that neutrophils contribute to the development of Lyme arthritis, a chronic inflammatory conditions occurring several months after Borrelia Burgdorferi infection, by recruiting pathogenic Th1/Th17 clones to the inflamed joint.92  Furthermore, pathogens can evade neutrophil clearance, as recently reported for Neisseria gonorrheae93  or Mycobacterium abscessus,94  which were shown to promote their survival by delaying primary granule-phagosome fusion93  or by inducing a limited pattern of neutrophil activation,94  respectively. Similarly, community-associated methicillin-resistant Staphylococcus aureus has been shown to survive within neutrophil phagosome and inhibit macrophage-mediated efferocytosis of community-associated methicillin-resistant S aureus-infected neutrophils.95  On the same line, the ability of Francisella tularensis to parasitize neutrophils and evade elimination has also been recently reviewed.96 

Neutrophils in autoimmunity

It has long been known that poorly controlled neutrophil activation is responsible for much of the tissue/organ damage in a variety of autoimmune diseases. More recently, a number of studies have highlighted the observation that neutrophils are a major source of autoantigens in these diseases.97,98  In this context, “NETting” neutrophils were recently found to release potential autoantigens, including deaminated histones in Felty’s syndrome, proteinase 3, or myeloperoxidase in autoimmune vasculitis, self-DNA, and antimicrobial peptides in SLE or citrullinated histones in rheumatoid arthritis (RA).98,99  There is therefore a strong interest in trying to understand how modified proteins externalized during neutrophil death by apoptosis or “NETosis” can function as autoantigens, leading to loss of tolerance and promoting autoimmune disease development.98  NETs have been shown to act as vehicles for proinflammatory molecules11,100  or tissue factor.101  The latter observation is of particular interest, considering that thrombotic risk is elevated not only in autoimmune vasculitis but also in other chronic diseases in which NETs form, including SLE, RA, colitis, and cancer.89,102  Impairment in NET clearance has also been associated to the pathogenesis of autoimmune diseases such as SLE98  or antiphospholipid syndrome.103  Interestingly, signal inhibitory receptor on leukocytes-1, a surface molecule previously shown to act as a negative regulator of phagocytes,104  can inhibit spontaneous and anti-neutrophil antibody-induced NET formation in SLE, likely by suppressing nicotinamide adenine dinucleotide phosphate oxidase and mitogen activated protein kinase kinase-extracellular signal-regulated kinase activity.105  Thus, recombinant signal inhibitory receptor on leukocytes-1 or other NET inhibitors might be considered for autoimmune disease treatment.98  Finally, human neutrophils have been recently suggested to contribute to the pathogenesis of autoimmune T1D, further confirming what recently observed in mice with spontaneous autoimmune T1D.106  If verified by more comprehensive studies, these data might lead to the definition of new therapeutic strategies for a disease that to date has proved controllable but incurable.106 

Neutrophils in cancer

Recent reviews elegantly discuss the multiple (pro- and antitumor) roles of neutrophils in cancer.59,107,108  Current evidence mostly support the idea that neutrophils facilitate, rather than inhibit, cancer progression primarily through their ability to promote tumor angiogenesis, invasion, and metastasis.107,,-110  This concept is also supported by dozens of reports correlating elevated numbers of tumor-infiltrating and/or blood neutrophils, as well as elevated blood neutrophil/lymphocyte ratios, with poor clinical outcome in several cancers.111  The mechanisms whereby neutrophils contribute to cancer metastasis, in particular, have recently gained more attention. For instance, even NETs have been involved in metastasis, by both trapping cancer cells and favoring their dissemination through the microvasculature.112  Histopathological evaluation of biopsies from a small cohort of pediatric patients with Ewing sarcoma has indeed revealed the presence of tumor-associated neutrophils undergoing NETosis in samples from patients with metastasis.113  In this context, we have shown that metastatic tumor-draining lymph nodes from carcinoma patients infiltrated by slanDCs also contain, in some cases, CD66b+neutrophils within an immunosuppressed like microenvironment.114  A very recent observation in a head and neck cancer model suggests that neutrophils, previously exposed to tumor-conditioned medium, induce an invasive, NK-resistant, and highly metastatic tumor cell phenotype (S. Brandau, unpublished data, 2014). Another mechanism proposed to explain protumor activities by human and mouse tumor-associated neutrophils consists in their ability to recruit regulatory T cells (Tregs) to the tumor sites, via CCL17 release.115  Based on all these findings, inducing the conversion of neutrophils from protumor to antitumor cells may represent a new form of immunotherapy for cancer.116  In this context, because neutrophils are the most abundant population of circulating white blood cells expressing FcγR and FcαR able to execute potent cytotoxic functions, the possibility of exploiting neutrophils for antibody-based cancer immunotherapy holds significant promise and deserves further investigation.117 

It is now beyond doubt that the traditional view of neutrophils as short-lived effector cells with limited functional capacity is incomplete. Neutrophils display many more functions than previously suspected, including the capacity to influence regulatory circuits in the innate and adaptive immune systems. The increasing availability of experimental animal models, including the zebrafish system, which allows prompt genetic and chemical manipulation,118  will provide new approaches to expand our knowledge of neutrophil biology.

As mentioned, an additional way whereby neutrophils may orchestrate, under physiological and pathological conditions, the evolution of inflammation, immune response, hematopoiesis, wound healing, angiogenesis, tissue remodeling, and bone reabsorption, occurs via the production and release of cytokines.13  In this context, as our technologies become more powerful and new molecules are cloned, the number of cytokines and chemokines that neutrophils can potentially produce continues to expand (Table 3), in some cases being regulated in a neutrophil-specific manner.13,57,59  Although the molecular mechanisms underlying the peculiar regulation of cytokine expression in neutrophils remain to be defined, recent studies suggest that they may include neutrophil-specific chromatin organization programs.119  Future challenges for scientists in the field will be to translate all these new insights into efficacious neutrophil-targeted therapies for the treatment of inflammatory conditions without compromising immunity.

Table 3

Cytokines that human neutrophils can potentially express and/or produce

Cytokine familySpecific cytokines
C-X-C chemokines CXCL1, CXCL2, CXCL3, CXCL4*, CXCL5, CXCL6, CXCL8, CXCL9, CXCL10, CXCL11, CXCL12*, CXCL13* 
C-C chemokines CCL2, CCL3, CCL4, CCL17, CCL18, CCL19, CCL20, CCL22 
Proinflammatory cytokines IL-1α, IL-1β, IL-6, IL-7, IL-9(?), IL-16(?), IL-17A, IL-17B, IL-18, MIF 
Anti-inflammatory cytokines IL-1ra, IL-4(?), TGFβ1, TGFβ2 
Immunoregulatory cytokines IFNα(?), IFNβ*, IFNγ(?), IL-12, IL-21, IL-23, IL-27, IL-18BP 
TNF superfamily members TNFα, TNFSF6/FasL, TNFSF8/CD30L, TNFSF10/TRAIL, TNFSF14/LIGHT*, TNFSF3/Lymphotoxin-β*, TNFSF13/APRIL, TNFSF13B/BAFF/BLyS, TNFSF5/CD40L, TNFSF11/RANKL 
Colony-stimulating factors G-CSF, M-CSF(?), GM-CSF(?), IL-3(?), SCF* 
Angiogenic and fibrogenic factors VEGFs, BV8 (prokineticin 2), HB-EGF, FGF-2, TGFα, HGF, angiopoietin-1 
Other cytokines NGF*, BDNF*, NT4*, PBEF (visfatin/NAMPT), amphiregulin, midkine, oncostatin M, activin A, endothelin 
Cytokine familySpecific cytokines
C-X-C chemokines CXCL1, CXCL2, CXCL3, CXCL4*, CXCL5, CXCL6, CXCL8, CXCL9, CXCL10, CXCL11, CXCL12*, CXCL13* 
C-C chemokines CCL2, CCL3, CCL4, CCL17, CCL18, CCL19, CCL20, CCL22 
Proinflammatory cytokines IL-1α, IL-1β, IL-6, IL-7, IL-9(?), IL-16(?), IL-17A, IL-17B, IL-18, MIF 
Anti-inflammatory cytokines IL-1ra, IL-4(?), TGFβ1, TGFβ2 
Immunoregulatory cytokines IFNα(?), IFNβ*, IFNγ(?), IL-12, IL-21, IL-23, IL-27, IL-18BP 
TNF superfamily members TNFα, TNFSF6/FasL, TNFSF8/CD30L, TNFSF10/TRAIL, TNFSF14/LIGHT*, TNFSF3/Lymphotoxin-β*, TNFSF13/APRIL, TNFSF13B/BAFF/BLyS, TNFSF5/CD40L, TNFSF11/RANKL 
Colony-stimulating factors G-CSF, M-CSF(?), GM-CSF(?), IL-3(?), SCF* 
Angiogenic and fibrogenic factors VEGFs, BV8 (prokineticin 2), HB-EGF, FGF-2, TGFα, HGF, angiopoietin-1 
Other cytokines NGF*, BDNF*, NT4*, PBEF (visfatin/NAMPT), amphiregulin, midkine, oncostatin M, activin A, endothelin 

The expression and/or production of most of these cytokines (updated from Mantovani et al3 ) has been validated in human neutrophils by gene expression techniques, immunohistochemistry, enzyme-linked immunosorbent assay, or biological assays. *, studies performed at the mRNA level only; (?), controversial data; APRIL, proliferation-inducing ligand; BAFF/BLyS, B-cell activating factor/B lymphocyte stimulator; BDNF, brain-derived neurotrophic factor; CCL, CC chemokine ligand; CXCL, CXC chemokine ligand; FasL, Fas ligand; FGF, fibroblast growth factor; HB-EGF, heparin binding-like epidermal growth factor; HGF, hepatocyte growth factor; IFN, interferon; IL-1ra, IL-1 receptor antagonist; IL18BP, IL-18 binding protein; LIGHT, homologous to lymphotoxins; M-CSF, macrophage-colony stimulating factor; MIF, macrophage inhibitory factor; NAMPT, nicotinamide phosphoribosyl transferase; NGF, nerve growth factor; NT4, neurotrophin-4; PBEF, pre-B-cell colony-enhancing factor; RANKL, receptor activator of nuclear factor k-B ligand; SCF, stem cell factor; TGF, transforming growth factor; TNFSF, TNF super family; TRAIL, TNF-related apoptosis-inducing ligand; VEGF, vascular endothelial growth factor.

The online version of this article contains a data supplement.

We thank C. A. Lowell for critical suggestions and helpful comments.

This work was supported by Associazione Italiana per la Ricerca sul Cancro, grant IG-11782 (M.A.C.) and Ministero dell’Istruzione dell’Universita’ e della Ricerca grant RBFR12I3UB_003 (P.S.).

Contribution: P.S. and M.A.C. wrote the review.

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

Correspondence: Patrizia Scapini, Department of Pathology and Diagnostics, Section of General Pathology, Strada Le Grazie 8, 37134 Verona, Italy; e-mail: patrizia.scapini@univr.it; or Marco A. Cassatella, Department of Pathology and Diagnostics, Section of General Pathology, Strada Le Grazie 8, 37134 Verona, Italy; e-mail: marco.cassatella@univr.it.

1
Tak
T
Tesselaar
K
Pillay
J
Borghans
JA
Koenderman
L
What’s your age again? Determination of human neutrophil half-lives revisited.
J Leukoc Biol
2013
94
4
595
601
2
Witko-Sarsat
V
Pederzoli-Ribeil
M
Hirsch
E
Sozzani
S
Cassatella
MA
Regulating neutrophil apoptosis: new players enter the game.
Trends Immunol
2011
32
3
117
124
3
Mantovani
A
Cassatella
MA
Costantini
C
Jaillon
S
Neutrophils in the activation and regulation of innate and adaptive immunity.
Nat Rev Immunol
2011
11
8
519
531
4
Amulic
B
Cazalet
C
Hayes
GL
Metzler
KD
Zychlinsky
A
Neutrophil function: from mechanisms to disease.
Annu Rev Immunol
2012
30
459
489
5
Mócsai
A
Diverse novel functions of neutrophils in immunity, inflammation, and beyond.
J Exp Med
2013
210
7
1283
1299
6
Mayadas
TN
Cullere
X
Lowell
CA
The multifaceted functions of neutrophils.
Annu Rev Pathol
2014
9
181
218
7
Kolaczkowska
E
Kubes
P
Neutrophil recruitment and function in health and inflammation.
Nat Rev Immunol
2013
13
3
159
175
8
Serhan
CN
Pro-resolving lipid mediators are leads for resolution physiology.
Nature
2014
510
7503
92
101
9
Pillay
J
Tak
T
Kamp
VM
Koenderman
L
Immune suppression by neutrophils and granulocytic myeloid-derived suppressor cells: similarities and differences.
Cell Mol Life Sci
2013
70
20
3813
3827
10
Dumitru
CA
Moses
K
Trellakis
S
Lang
S
Brandau
S
Neutrophils and granulocytic myeloid-derived suppressor cells: immunophenotyping, cell biology and clinical relevance in human oncology.
Cancer Immunol Immunother
2012
61
8
1155
1167
11
Carmona-Rivera
C
Kaplan
MJ
Low-density granulocytes: a distinct class of neutrophils in systemic autoimmunity.
Semin Immunopathol
2013
35
4
455
463
12
Beyrau
M
Bodkin
JV
Nourshargh
S
Neutrophil heterogeneity in health and disease: a revitalized avenue in inflammation and immunity.
Open Biol
2012
2
11
120134
13
Cassatella
MA
Neutrophil-derived proteins: selling cytokines by the pound.
Adv Immunol
1999
73
369
509
14
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
419
2
309
315
15
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
8
7
e69575
16
Ramirez-Velazquez
C
Castillo
EC
Guido-Bayardo
L
Ortiz-Navarrete
V
IL-17-producing peripheral blood CD177+ neutrophils increase in allergic asthmatic subjects.
Allergy Asthma Clin Immunol
2013
9
1
23
17
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
90
5
436
437
18
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, e5
19
Bowers
NL
Helton
ES
Huijbregts
RP
Goepfert
PA
Heath
SL
Hel
Z
Immune suppression by neutrophils in HIV-1 infection: role of PD-L1/PD-1 pathway.
PLoS Pathog
2014
10
3
e1003993
20
Drifte
G
Dunn-Siegrist
I
Tissières
P
Pugin
J
Innate immune functions of immature neutrophils in patients with sepsis and severe systemic inflammatory response syndrome.
Crit Care Med
2013
41
3
820
832
21
Cloke
T
Munder
M
Bergin
P
et al
Phenotypic alteration of neutrophils in the blood of HIV seropositive patients.
PLoS ONE
2013
8
9
e72034
22
Rieber
N
Wecker
I
Neri
D
et al
Extracorporeal photopheresis increases neutrophilic myeloid-derived suppressor cells in patients with GvHD.
Bone Marrow Transplant
2014
49
4
545
552
23
Ssemaganda
A
Kindinger
L
Bergin
P
et al
Characterization of neutrophil subsets in healthy human pregnancies.
PLoS ONE
2014
9
2
e85696
24
Vasconcelos
ZF
Santos
BM
Costa
ES
et al
T-lymphocyte function from peripheral blood stem-cell donors is inhibited by activated granulocytes.
Cytotherapy
2003
5
4
336
345
25
Luyckx
A
Schouppe
E
Rutgeerts
O
et al
G-CSF stem cell mobilization in human donors induces polymorphonuclear and mononuclear myeloid-derived suppressor cells.
Clin Immunol
2012
143
1
83
87
26
Brinkmann
V
Zychlinsky
A
Neutrophil extracellular traps: is immunity the second function of chromatin?
J Cell Biol
2012
198
5
773
783
27
Youn
JI
Kumar
V
Collazo
M
et al
Epigenetic silencing of retinoblastoma gene regulates pathologic differentiation of myeloid cells in cancer.
Nat Immunol
2013
14
3
211
220
28
Cortez-Retamozo
V
Etzrodt
M
Newton
A
et al
Origins of tumor-associated macrophages and neutrophils.
Proc Natl Acad Sci USA
2012
109
7
2491
2496
29
Puga
I
Cols
M
Barra
CM
et al
B cell-helper neutrophils stimulate the diversification and production of immunoglobulin in the marginal zone of the spleen.
Nat Immunol
2012
13
2
170
180
30
Choi
J
Suh
B
Ahn
YO
et al
CD15+/CD16low human granulocytes from terminal cancer patients: granulocytic myeloid-derived suppressor cells that have suppressive function.
Tumour Biol
2012
33
1
121
129
31
Tsuda
Y
Fukui
H
Asai
A
et al
An immunosuppressive subtype of neutrophils identified in patients with hepatocellular carcinoma.
J Clin Biochem Nutr
2012
51
3
204
212
32
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
122
1
327
336
33
de Kleijn
S
Langereis
JD
Leentjens
J
et al
IFN-γ-stimulated neutrophils suppress lymphocyte proliferation through expression of PD-L1.
PLoS ONE
2013
8
8
e72249
34
Hao
S
Andersen
M
Yu
H
Detection of immune suppressive neutrophils in peripheral blood samples of cancer patients.
Am J Blood Res
2013
3
3
239
245
35
Nadkarni
S
Dalli
J
Hollywood
J
Mason
JC
Dasgupta
B
Perretti
M
Investigational analysis reveals a potential role for neutrophils in giant-cell arteritis disease progression.
Circ Res
2014
114
2
242
248
36
Araki
H
Katayama
N
Yamashita
Y
et al
Reprogramming of human postmitotic neutrophils into macrophages by growth factors.
Blood
2004
103
8
2973
2980
37
Matsushima
H
Geng
S
Lu
R
et al
Neutrophil differentiation into a unique hybrid population exhibiting dual phenotype and functionality of neutrophils and dendritic cells.
Blood
2013
121
10
1677
1689
38
Dyugovskaya
L
Berger
S
Polyakov
A
Lavie
L
The development of giant phagocytes in long-term neutrophil cultures [published online ahead of print February 27, 2014].
J Leukoc Biol
39
Ghasemzadeh
M
Hosseini
E
Platelet-leukocyte crosstalk: Linking proinflammatory responses to procoagulant state.
Thromb Res
2013
131
3
191
197
40
Schuster
S
Hurrell
B
Tacchini-Cottier
F
Crosstalk between neutrophils and dendritic cells: a context-dependent process.
J Leukoc Biol
2013
94
4
671
675
41
Candelario
KM
Steindler
DA
The role of extracellular vesicles in the progression of neurodegenerative disease and cancer [published online ahead of print May 14, 2014].
Trends Mol Med
42
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
3
73
73ra19
43
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
3
73
73ra20
44
Skrzeczynska-Moncznik
J
Wlodarczyk
A
Banas
M
et al
DNA structures decorated with cathepsin G/secretory leukocyte proteinase inhibitor stimulate IFNI production by plasmacytoid dendritic cells.
Am J Clin Exp Immunol
2013
2
2
186
194
45
Skrzeczynska-Moncznik
J
Wlodarczyk
A
Zabieglo
K
et al
Secretory leukocyte proteinase inhibitor-competent DNA deposits are potent stimulators of plasmacytoid dendritic cells: implication for psoriasis.
J Immunol
2012
189
4
1611
1617
46
Diana
J
Simoni
Y
Furio
L
et al
Crosstalk between neutrophils, B-1a cells and plasmacytoid dendritic cells initiates autoimmune diabetes.
Nat Med
2013
19
1
65
73
47
Sangaletti
S
Tripodo
C
Chiodoni
C
et al
Neutrophil extracellular traps mediate transfer of cytoplasmic neutrophil antigens to myeloid dendritic cells toward ANCA induction and associated autoimmunity.
Blood
2012
120
15
3007
3018
48
Odobasic
D
Kitching
AR
Yang
Y
et al
Neutrophil myeloperoxidase regulates T-cell-driven tissue inflammation in mice by inhibiting dendritic cell function.
Blood
2013
121
20
4195
4204
49
Silva
MT
Macrophage phagocytosis of neutrophils at inflammatory/infectious foci: a cooperative mechanism in the control of infection and infectious inflammation.
J Leukoc Biol
2011
89
5
675
683
50
Costantini
C
Cassatella
MA
The defensive alliance between neutrophils and NK cells as a novel arm of innate immunity.
J Leukoc Biol
2011
89
2
221
233
51
Pliyev
BK
Kalintseva
MV
Abdulaeva
SV
Yarygin
KN
Savchenko
VG
Neutrophil microparticles modulate cytokine production by natural killer cells.
Cytokine
2014
65
2
126
129
52
Jaeger
BN
Donadieu
J
Cognet
C
et al
Neutrophil depletion impairs natural killer cell maturation, function, and homeostasis.
J Exp Med
2012
209
3
565
580
53
Bär
E
Whitney
PG
Moor
K
Reis e Sousa
C
LeibundGut-Landmann
S
IL-17 regulates systemic fungal immunity by controlling the functional competence of NK cells.
Immunity
2014
40
1
117
127
54
Voigt
J
Hünniger
K
Bouzani
M
et al
Human natural killer cells acting as phagocytes against Candida albicans and mounting an inflammatory response that modulates neutrophil antifungal activity.
J Infect Dis
2014
209
4
616
626
55
Walker
JA
Barlow
JL
McKenzie
AN
Innate lymphoid cells—how did we miss them?
Nat Rev Immunol
2013
13
2
75
87
56
Magri
G
Miyajima
M
Bascones
S
et al
Innate lymphoid cells integrate stromal and immunological signals to enhance antibody production by splenic marginal zone B cells.
Nat Immunol
2014
15
4
354
364
57
Scapini
P
Bazzoni
F
Cassatella
MA
Regulation of B-cell-activating factor (BAFF)/B lymphocyte stimulator (BLyS) expression in human neutrophils.
Immunol Lett
2008
116
1
1
6
58
Nagelkerke
SQ
aan de Kerk
DJ
Jansen
MH
van den Berg
TK
Kuijpers
TW
Failure to detect functional neutrophil B helper cells in the human spleen.
PLoS ONE
2014
9
2
e88377
59
Tecchio
C
Scapini
P
Pizzolo
G
Cassatella
MA
On the cytokines produced by human neutrophils in tumors.
Semin Cancer Biol
2013
23
3
159
170
60
Sangaletti
S
Tripodo
C
Vitali
C
et al
Defective stromal remodeling and neutrophil extracellular traps in lymphoid tissues favor the transition from autoimmunity to lymphoma.
Cancer Discov
2014
4
1
110
129
61
Müller
I
Munder
M
Kropf
P
Hänsch
GM
Polymorphonuclear neutrophils and T lymphocytes: strange bedfellows or brothers in arms?
Trends Immunol
2009
30
11
522
530
62
Berton
G
Cassatella
MA
Modulation of neutrophil functions by g-interferon. Granulocyte Responses to Cytokines, Coffey NG ed. New York: Marcel Dekker; 1992:437-456
63
Wright
HL
Moots
RJ
Bucknall
RC
Edwards
SW
Neutrophil function in inflammation and inflammatory diseases.
Rheumatology (Oxford)
2010
49
9
1618
1631
64
Davey
MS
Lin
CY
Roberts
GW
et al
Human neutrophil clearance of bacterial pathogens triggers anti-microbial γδ T cell responses in early infection.
PLoS Pathog
2011
7
5
e1002040
65
Sabbione
F
Gabelloni
ML
Ernst
G
et al
Neutrophils suppress γδ T-cell function.
Eur J Immunol
2014
44
3
819
830
66
Kalyan
S
Chandrasekaran
V
Quabius
ES
Lindhorst
TK
Kabelitz
D
Neutrophil uptake of nitrogen-bisphosphonates leads to the suppression of human peripheral blood γδ T cells.
Cell Mol Life Sci
2014
71
12
2335
2346
67
Wu
P
Wu
D
Ni
C
et al
GammadeltaT17 cells promote the accumulation and expansion of myeloid-derived suppressor cells in human colorectal cancer. Immunity. 2014;15;40(5):785-800
68
Dinauer
MC
Disorders of neutrophil function: an overview.
Methods Mol Biol
2014
1124
501
515
69
Drewniak
A
Gazendam
RP
Tool
AT
et al
Invasive fungal infection and impaired neutrophil killing in human CARD9 deficiency.
Blood
2013
121
13
2385
2392
70
Bogunovic
D
Byun
M
Durfee
LA
et al
Mycobacterial disease and impaired IFN-γ immunity in humans with inherited ISG15 deficiency.
Science
2012
337
6102
1684
1688
71
Vilboux
T
Lev
A
Malicdan
MC
et al
A congenital neutrophil defect syndrome associated with mutations in VPS45.
N Engl J Med
2013
369
1
54
65
72
Zhao
XW
Gazendam
RP
Drewniak
A
et al
Defects in neutrophil granule mobilization and bactericidal activity in familial hemophagocytic lymphohistiocytosis type 5 (FHL-5) syndrome caused by STXBP2/Munc18-2 mutations.
Blood
2013
122
1
109
111
73
Fournier
BM
Parkos
CA
The role of neutrophils during intestinal inflammation.
Mucosal Immunol
2012
5
4
354
366
74
Xu
R
Huang
H
Zhang
Z
Wang
FS
The role of neutrophils in the development of liver diseases.
Cell Mol Immunol
2014
11
3
224
231
75
Lowe
DM
Redford
PS
Wilkinson
RJ
O’Garra
A
Martineau
AR
Neutrophils in tuberculosis: friend or foe?
Trends Immunol
2012
33
1
14
25
76
Abi Abdallah
DS
Denkers
EY
Neutrophils cast extracellular traps in response to protozoan parasites.
Front Immunol
2012
3
382
77
Makepeace
BL
Martin
C
Turner
JD
Specht
S
Granulocytes in helminth infection — who is calling the shots?
Curr Med Chem
2012
19
10
1567
1586
78
Drescher
B
Bai
F
Neutrophil in viral infections, friend or foe?
Virus Res
2013
171
1
1
7
79
Gabriel
C
Her
Z
Ng
LF
Neutrophils: neglected players in viral diseases.
DNA Cell Biol
2013
32
12
665
675
80
Thomas
CJ
Schroder
K
Pattern recognition receptor function in neutrophils.
Trends Immunol
2013
34
7
317
328
81
Prüfer
S
Weber
M
Sasca
D
et al
Distinct signaling cascades of TREM-1, TLR and NLR in neutrophils and monocytic cells.
J Innate Immun
2014
6
3
339
352
82
Bakele
M
Joos
M
Burdi
S
et al
Localization and functionality of the inflammasome in neutrophils.
J Biol Chem
2014
289
8
5320
5329
83
Tamassia
N
Cassatella
MA
Cytoplasmic receptors recognizing nucleic acids and mediating immune functions in neutrophils.
Curr Opin Pharmacol
2013
13
4
547
554
84
Lindau
D
Mussard
J
Wagner
BJ
et al
Primary blood neutrophils express a functional cell surface Toll-like receptor 9.
Eur J Immunol
2013
43
8
2101
2113
85
Brigotti
M
Carnicelli
D
Arfilli
V
et al
Identification of TLR4 as the receptor that recognizes Shiga toxins in human neutrophils.
J Immunol
2013
191
9
4748
4758
86
DuMont
AL
Yoong
P
Day
CJ
et al
Staphylococcus aureus LukAB cytotoxin kills human neutrophils by targeting the CD11b subunit of the integrin Mac-1.
Proc Natl Acad Sci USA
2013
110
26
10794
10799
87
Timár
CI
Lorincz
AM
Csépányi-Kömi
R
et al
Antibacterial effect of microvesicles released from human neutrophilic granulocytes.
Blood
2013
121
3
510
518
88
Borregaard
N
Neutrophils, from marrow to microbes.
Immunity
2010
33
5
657
670
89
Yipp
BG
Kubes
P
NETosis: how vital is it?
Blood
2013
122
16
2784
2794
90
Dalli
J
Montero-Melendez
T
Norling
LV
et al
Heterogeneity in neutrophil microparticles reveals distinct proteome and functional properties.
Mol Cell Proteomics
2013
12
8
2205
2219
91
Lacy
P
A new way of trapping bugs: neutrophil microvesicles.
Blood
2013
121
3
420
421
92
Codolo
G
Bossi
F
Durigutto
P
et al
Orchestration of inflammation and adaptive immunity in Borrelia burgdorferi-induced arthritis by neutrophil-activating protein A.
Arthritis Rheum
2013
65
5
1232
1242
93
Johnson
MB
Criss
AK
Neisseria gonorrhoeae phagosomes delay fusion with primary granules to enhance bacterial survival inside human neutrophils.
Cell Microbiol
2013
15
8
1323
1340
94
Malcolm
KC
Nichols
EM
Caceres
SM
et al
Mycobacterium abscessus induces a limited pattern of neutrophil activation that promotes pathogen survival.
PLoS ONE
2013
8
2
e57402
95
Greenlee-Wacker
MC
Rigby
KM
Kobayashi
SD
Porter
AR
DeLeo
FR
Nauseef
WM
Phagocytosis of Staphylococcus aureus by human neutrophils prevents macrophage efferocytosis and induces programmed necrosis.
J Immunol
2014
192
10
4709
4717
96
Schwartz
JT
Bandyopadhyay
S
Kobayashi
SD
et al
Francisella tularensis alters human neutrophil gene expression: insights into the molecular basis of delayed neutrophil apoptosis.
J Innate Immun
2013
5
2
124
136
97
Kaplan
MJ
Role of neutrophils in systemic autoimmune diseases.
Arthritis Res Ther
2013
15
5
219
98
Radic
M
Marion
TN
Neutrophil extracellular chromatin traps connect innate immune response to autoimmunity.
Semin Immunopathol
2013
35
4
465
480
99
Khandpur
R
Carmona-Rivera
C
Vivekanandan-Giri
A
et al
NETs are a source of citrullinated autoantigens and stimulate inflammatory responses in rheumatoid arthritis. Sci Transl Med. 2013;5(178):178ra140
100
Lindau
D
Mussard
J
Rabsteyn
A
et al
TLR9 independent interferon α production by neutrophils on NETosis in response to circulating chromatin, a key lupus autoantigen [published online ahead of print September 7, 2013].
Ann Rheum Dis
101
Kambas
K
Chrysanthopoulou
A
Vassilopoulos
D
et al
Tissue factor expression in neutrophil extracellular traps and neutrophil derived microparticles in antineutrophil cytoplasmic antibody associated vasculitis may promote thromboinflammation and the thrombophilic state associated with the disease [published online ahead of print July 19, 2013]. Ann Rheum Dis
102
Martinod
K
Wagner
DD
Thrombosis: tangled up in NETs.
Blood
2014
123
18
2768
2776
103
Leffler
J
Stojanovich
L
Shoenfeld
Y
Bogdanovic
G
Hesselstrand
R
Blom
AM
Degradation of neutrophil extracellular traps is decreased in patients with antiphospholipid syndrome.
Clin Exp Rheumatol
2014
32
1
66
70
104
Steevels
TA
Lebbink
RJ
Westerlaken
GH
Coffer
PJ
Meyaard
L
Signal inhibitory receptor on leukocytes-1 is a novel functional inhibitory immune receptor expressed on human phagocytes.
J Immunol
2010
184
9
4741
4748
105
Van Avondt
K
Fritsch-Stork
R
Derksen
RH
Meyaard
L
Ligation of signal inhibitory receptor on leukocytes-1 suppresses the release of neutrophil extracellular traps in systemic lupus erythematosus.
PLoS ONE
2013
8
10
e78459
106
Battaglia
M
Neutrophils and type 1 autoimmune diabetes.
Curr Opin Hematol
2014
21
1
8
15
107
Fridlender
ZG
Albelda
SM
Tumor-associated neutrophils: friend or foe?
Carcinogenesis
2012
33
5
949
955
108
Brandau
S
Dumitru
CA
Lang
S
Protumor and antitumor functions of neutrophil granulocytes.
Semin Immunopathol
2013
35
2
163
176
109
Tazzyman
S
Niaz
H
Murdoch
C
Neutrophil-mediated tumour angiogenesis: subversion of immune responses to promote tumour growth.
Semin Cancer Biol
2013
23
3
149
158
110
Tecchio
C
Cassatella
MA
Neutrophil-derived cytokines involved in physiological and pathological angiogenesis.
Chem Immunol Allergy
2014
99
123
137
111
Donskov
F
Immunomonitoring and prognostic relevance of neutrophils in clinical trials.
Semin Cancer Biol
2013
23
3
200
207
112
Cools-Lartigue
J
Spicer
J
McDonald
B
et al
Neutrophil extracellular traps sequester circulating tumor cells and promote metastasis [published online ahead of print July 1, 2013].
J Clin Invest
113
Berger-Achituv
S
Brinkmann
V
Abed
UA
et al
A proposed role for neutrophil extracellular traps in cancer immunoediting.
Front Immunol
2013
4
48
114
Vermi
W
Micheletti
A
Lonardi
S
et al
slanDCs selectively accumulate in carcinoma-draining lymph nodes and marginate metastatic cells.
Nat Commun
2014
5
3029
115
Mishalian
I
Bayuh
R
Eruslanov
E
et al
Neutrophils recruit regulatory T-cells into tumors via secretion of CCL17-A new mechanism of impaired antitumor immunity [published online ahead of print February 6, 2014].
Int J Cancer
116
Dumitru
CA
Lang
S
Brandau
S
Modulation of neutrophil granulocytes in the tumor microenvironment: mechanisms and consequences for tumor progression.
Semin Cancer Biol
2013
23
3
141
148
117
van Egmond
M
Bakema
JE
Neutrophils as effector cells for antibody-based immunotherapy of cancer.
Semin Cancer Biol
2013
23
3
190
199
118
Henry
KM
Loynes
CA
Whyte
MK
Renshaw
SA
Zebrafish as a model for the study of neutrophil biology.
J Leukoc Biol
2013
94
4
633
642
119
Tamassia
N
Zimmermann
M
Castellucci
M
et al
Cutting edge: An inactive chromatin configuration at the IL-10 locus in human neutrophils.
J Immunol
2013
190
5
1921
1925
Sign in via your Institution