Europe PMC
Nothing Special   »   [go: up one dir, main page]

Europe PMC requires Javascript to function effectively.

Either your web browser doesn't support Javascript or it is currently turned off. In the latter case, please turn on Javascript support in your web browser and reload this page.

This website requires cookies, and the limited processing of your personal data in order to function. By using the site you are agreeing to this as outlined in our privacy notice and cookie policy.

Abstract 


Neutrophils, the most abundant white blood cells in the human circulation, play crucial roles in various diseases, including kidney disease. Traditionally viewed as short-lived pro-inflammatory phagocytes that release reactive oxygen species, cytokines and neutrophil extracellular traps, recent studies have revealed their complexity and heterogeneity, thereby challenging this perception. Neutrophils are now recognized as transcriptionally active cells capable of proliferation and reverse migration, displaying phenotypic and functional heterogeneity. They respond to a wide range of signals and deploy various cargo to influence the activity of other cells in the circulation and in tissues. They can regulate the behavior of multiple immune cell types, exhibit innate immune memory, and contribute to both acute and chronic inflammatory responses while also promoting inflammation resolution in a context-dependent manner. Here, we explore the origin and heterogeneity of neutrophils, their functional diversity, and the cues that regulate their effector functions. We also examine their emerging role in infectious and non-infectious diseases with a particular emphasis on kidney disease. Understanding the complex behavior of neutrophils during tissue injury and inflammation may provide novel insights, thereby paving the way for potential therapeutic strategies to manage acute and chronic conditions. By deciphering their multifaceted role, targeted interventions can be developed to address the intricacies of neutrophil-mediated immune responses and improve disease outcomes.

Free full text 


Logo of ndtLink to Publisher's site
Nephrol Dial Transplant. 2024 Oct; 39(10): 1551–1564.
Published online 2023 Dec 19. https://doi.org/10.1093/ndt/gfad266
PMCID: PMC11427074
PMID: 38115607

Neutrophils—biology and diversity

Associated Data

Data Availability Statement

ABSTRACT

Neutrophils, the most abundant white blood cells in the human circulation, play crucial roles in various diseases, including kidney disease. Traditionally viewed as short-lived pro-inflammatory phagocytes that release reactive oxygen species, cytokines and neutrophil extracellular traps, recent studies have revealed their complexity and heterogeneity, thereby challenging this perception. Neutrophils are now recognized as transcriptionally active cells capable of proliferation and reverse migration, displaying phenotypic and functional heterogeneity. They respond to a wide range of signals and deploy various cargo to influence the activity of other cells in the circulation and in tissues. They can regulate the behavior of multiple immune cell types, exhibit innate immune memory, and contribute to both acute and chronic inflammatory responses while also promoting inflammation resolution in a context-dependent manner. Here, we explore the origin and heterogeneity of neutrophils, their functional diversity, and the cues that regulate their effector functions. We also examine their emerging role in infectious and non-infectious diseases with a particular emphasis on kidney disease. Understanding the complex behavior of neutrophils during tissue injury and inflammation may provide novel insights, thereby paving the way for potential therapeutic strategies to manage acute and chronic conditions. By deciphering their multifaceted role, targeted interventions can be developed to address the intricacies of neutrophil-mediated immune responses and improve disease outcomes.

Keywords: autoimmunity, cardiovascular disease, infection, kidney disease, neutrophils

INTRODUCTION

Neutrophils play a crucial role in host defense in urinary tract infection and bacterial pyelonephritis by degrading virulence factors and identifying and eradicating pathogens, but on the other hand contribute to tissue damage in several non-infectious forms of kidney disease, e.g. ischemic injury and glomerulonephritis. Their migration from the blood into the injured tissue occurs in a tightly regulated cascade via adhesion molecules and circulating factors such as cytokines and chemokines. Once recruited to the site of tissue injury, neutrophils contribute to inflammation by releasing pro-inflammatory mediators such as reactive oxygen species (ROS) and neutrophil extracellular traps (NETs), and subsequently can promote further organ damage. While there is no doubt that the antimicrobial effector functions of neutrophils are lifesaving, e.g. in the context of immediate host defense, it has become evident that neutrophils can act as a key player in numerous inflammatory disease contexts including kidney disease.

Neutrophils are classically viewed as short-lived, terminally differentiated phagocytic cells with a rather unspecific mode of action. It was thought that neutrophils originate from hematopoietic stem cells through common bone marrow–derived progenitors, are pro-inflammatory immune cells and have an inability to proliferate, with unidirectional trafficking. This classical concept has been challenged by emerging evidence over recent years describing neutrophils as heterogenous population [1] that originate not only from the bone marrow but also the spleen, with the ability to rapidly adapt to the changing environment [2], to reversely migrate into other tissues [3], to communicate with other cell types and to promote even tissue repair [4]. Recent work revealed that whereas neutrophils indeed exhibit a short half-life in the circulation and some tissues, the life span differs between tissues, suggesting an adaptation of neutrophils to the microenvironment [5].

This review highlights the thus far underestimated phenotypic heterogeneity and functional diversity of neutrophils. We provide mechanistic evidence for their emerging role as effector cells during homeostasis but also during infectious and non-infectious diseases with a particular emphasis on kidney disease. Finally, we discuss how these new insights can be used to target neutrophils therapeutically.

NEUTROPHIL ORIGIN, DIFFERENTIATION AND AGING

Neutrophils are thought to originate from hematopoietic stem cells through common myeloid progenitors, granulo-monocytic progenitors and recently identified unipotent neutrophil progenitors (pro-neutrophils 1 and pro-neutrophils 2) [6, 7]. In recent decades, the classical concept that neutrophils are a homogenous terminally differentiated population has been challenged. Recent evidence points toward a phenotypic and functional heterogeneity of neutrophils under homeostatic and pathogenic conditions [8]. Neutrophils are continuously generated in the bone marrow. In humans, neutrophils mature in the bone marrow from committed myeloid precursors that, through subsequent differentiation stages (here defined as “immature neutrophils”), differentiate into segmented mature neutrophils (Fig. 1) [9]. In mice, three neutrophil subsets have been identified within the bone marrow: a committed proliferative neutrophil precursor (pre-neutrophils), which differentiates into non-proliferating immature neutrophils and mature neutrophils under the control of transcription factors [mainly PU.1 and CCAAT/enhancer-binding protein (C/EBP)α–ζ] [7, 10].

An external file that holds a picture, illustration, etc.
Object name is gfad266fig1.jpg

Neutrophil populations in homeostatic conditions. In mice, neutrophils develop from myeloid precursors within the bone marrow and are comprised of three distinct subpopulations, namely pre-neutrophil (proliferative neutrophils precursor), immature and segmented mature neutrophils. Under resting conditions, only terminally differentiated neutrophils are released into the blood and recovered as normal-density neutrophils (NDN). Evidence suggests that different neutrophil subpopulations, defined by the indicated markers, are present in the blood under homeostatic conditions. Neutrophils reside in pools within the lung vasculature, spleen and liver, but also transit and reside in other organs such as muscle, skin, lymph nodes and intestine. In the mouse spleen, three neutrophil subpopulations localized in the red pulp or marginal zone (MZ) participate in emergency granulopoiesis (Ly6Gint) and pneumococcal clearance (Ly6Ghigh), and can regulate antibody production and activation in MZ B cells (termed as B cell helper neutrophils, NBH). In humans, neutrophils mature in the bone marrow and subsequently differentiate from immature into mature neutrophils. As observed in mice, based on differential surface marker expression, circulating human neutrophils exhibit heterogeneity that associates with distinct effector activities. Under homeostatic conditions, these mature neutrophils can marginate in tissues including spleen (as NBH), lung and liver, but the phenotypes and functions of these cells remain poorly defined. This figure was generated with BioRender software. OLFM4, glycoprotein olfactomedin 4; TCRαβ, T cell receptor αβ; VEGFR1, vascular endothelial growth factor receptor 1.

The pool of neutrophils is maintained by a fine balance between granulopoiesis, neutrophil release from the bone marrow into the circulation and ultimately their return/homing to the bone marrow for final clearance by resident macrophages. Humans and mice differ in their numbers of circulating neutrophils. In humans, 50%–70% of circulating leukocytes are neutrophils, whereas only 10%–25% are in mice. Evidence suggests that different neutrophil subpopulations exist in the human and mouse blood depending on the course of their lifespan (approximately 5.4 days in humans and 12.5 h in mice). For example, 45%–65% of circulating human neutrophils are CD177+ and 20%–25% of neutrophils express the glycoprotein olfactomedin 4 (OLFM4) [8]. Moreover, as shown by flow cytometry analysis performed at different times of the day, the proportion of CXCR4CD62L+ young and CXCR4+CD62L aged neutrophil populations among circulating neutrophils from healthy individuals has been shown to be regulated by circadian oscillations [11].

Healthy tissues were traditionally considered to be devoid of neutrophils in the absence of inflammatory insults. However, under physiological conditions, neutrophils can be found in the lung, liver and spleen [8], but the phenotypes and functions of these cells remain poorly defined. For example, a human splenic neutrophil population that resides within the splenic marginal zone has been identified as B-cell helper neutrophils (NBH cells) with an ability to promote B-cell proliferation and antibody production [12]. Further research is needed to gain a better understanding of the phenotypic and functional features of neutrophil subpopulations in the circulation and tissues of healthy individuals.

The phenotypic and functional diversity of neutrophils change and progressively deteriorate with increased life span and aging (reviewed in [13, 14]). While the number of neutropihls remain unchanged with increasing age, neutrophils in the elderly tend to exhibit dysfunctional phagocytic and migratory abilities [15]. Inaccurate migration of aged neutrophils via increased activity of phosphorylated phosphatidylinositol 3-kinase can even cause damage to normal tissues [16]. The reduced phagocytic and bactericidal activity of neutrophils in the elderly is associated with increased intracellular calcium concentrations and reduced hexose uptake [15]. Moreover, aged neutrophils are less able to form NETs and to produce ROS. Thus, age-related changes in neutrophils are associated with immunosenescence, inflammaging (chronic inflammation), gut dysbiosis, increased susceptibility to infection and impaired vaccine response in older adults [17], and represent risk factors for several pathologies including cancer, cardiovascular disease (CVD) and kidney disease. Aging and chronic kidney disease (CKD) are interconnected as kidney function declines with age and kidney disease is more common in the elderly. Both share similar features such as cellular senescence, oxidative stress, DNA damage, mitochondrial dysfunction, telomere shortening and Wnt/β-catenin signaling (reviewed in [18]) that contribute to age-related neutrophil dysfunction.

NEUTROPHIL MIGRATION FROM THE CIRCULATION INTO TISSUES AND VICE VERSA

Neutrophil recruitment from the circulation into the infected or injured tissue during an inflammatory response follows a well-defined cascade of adhesion and activation events including rolling, slow rolling, adhesion, adhesion strengthening, crawling and transmigration (Fig. 2). This cascade is tightly regulated by a broad range of receptors and their respective ligands, including selectins, β2 integrins and chemokine receptors (reviewed in [19]). Efficient neutrophil recruitment is of the utmost importance, since inadequate recruitment results in either impaired host response that might lead to recurrent and life-threatening infections or to hyperinflammation and subsequent tissue damage [20]. Although leukocyte rolling and adhesion has been known since the 19th century, the involved signaling pathways are still not fully understood.

An external file that holds a picture, illustration, etc.
Object name is gfad266fig2.jpg

Neutrophil recruitment cascade from the vasculature to the tissue. Tethering and slow rolling are selectin-dependent, while β2 integrin interactions mediate slow rolling, adhesion strengthening, crawling and transmigration. Following rolling, neutrophils are activated by chemokines lining the luminal part of the endothelium. Neutrophil activation induces conformational changes in β2 surface integrins. Following a chemokine gradient, neutrophils crawl along the endothelium, guiding them to preferential sites of transmigration such as inflamed tissues either through a paracellular (between endothelial cells) or transcellular (through endothelial cells) mechanism. Activated neutrophils have several different fates mediated by factors such as chemokines, cytokines, ROS, MVs, and lipid mediators. These neutrophil fates include: degranulation, where neutrophil granules release antibacterial proteins such as defensins, lactoferrin, cathepsins and lysozyme, into either phagosomes or the extracellular space to kill pathogens; phagocytosis, where neutrophils engulf microorganisms into phagosomes to kill the pathogens using granules or various other mechanisms such as NADPH oxygenase-dependent mechanisms including ROS; and NET formation, which can eliminate extracellular microorganisms via PAD4 and the release of DNA, NE, MPO, histones and S1000A8/A9 proteins to immobilize pathogens and thereby to prevent spreading and facilitating phagocytosis of trapped microorganisms or direct killing of pathogens via antimicrobial histones and proteases. Neutrophils also die via apoptosis and are recognized by other phagocytes including macrophages by “eat-me signals” such as phosphatidylserine on their plasma membrane. Extravasated neutrophils can also migrate from the inflamed tissue back into circulation, a mechanisms called reverse transmigration [156]. This figure was generated with BioRender software. NADPH, nicotinamide adenine dinucleotide phosphate; MMP, matrix metalloproteinase; MVs, microvesicles; NE, neutrophil elastase.

Neutrophil recruitment highly depends on the activation of β2 integrins [lymphocyte function-associated antigen 1 (LFA-1, CD18/CD11a), macrophage-1 antigen (MAC-1, CD18/CD11b)] during rolling and adhesion induced by selectin-, chemokine- and toll-like receptor (TLR)-mediated signaling [21–25]. Regulation of β2 integrins and subsequent linkage to the actin cytoskeleton occurs via their intracellular domains supported by various adapter molecules including talin 1 and kindlin3 [26], but also new regulators have been identified recently, among them coronin-1A, hematopoietic progenitor kinase 1, integrin-linked kinase and the ion channel KV1.3 [27–30]. Furthermore, new molecular players such as myosin-9, mammalian STE20-like protein kinase 1 and unconventional myosin-IXb have been identified to be important during neutrophil migration, transmigration and interstitial migration, respectively [31–33]. The central role of src family kinases (SFKs) in neutrophil recruitment and migration has been known for years [34, 35]. In a recent multidimensional study, neutrophils were screened for their migratory behavior and linked to their biochemical properties in order to identify non-pathogenic and pathogenic neutrophil states. Here, the SFK member tyrosine-protein kinase Fgr was identified as a central regulator of the pathogenic state [36], further highlighting a role of SFKs in neutrophils. Neutrophil recruitment also depends on tissue-specific recruitment cues [2]. In the kidney, neutrophil recruitment within the glomerulus occurs without neutrophil rolling, but is dependent on selectins and β2 integrins [37], whereas “classical” selectin-dependent rolling and downstream integrin activation occur in the kidney cortex [38, 39].

It is thought that most neutrophils die in the injured and inflamed tissue, and are cleared by macrophages. Experimental evidence now suggests that migrated neutrophils can re-enter the vasculature through downregulation of the junctional adhesion protein JAM-C during inflammation [40], a process referred to as reverse neutrophil migration (Fig. 2) [3, 41, 42]. So far, reverse-migrating neutrophils (1%–2% of peripheral blood neutrophils) have been reported in patients with rheumatoid arthritis (RA) [43]. Notably, these reverse-migrated neutrophils were more resistant to apoptosis [43]. Explanations for this phenomenon of reverse neutrophil migration could be that it preserves neutrophils when they are not needed for host defense, or neutrophils re-entering the circulation could disseminate systemic inflammation or even chronic inflammation in other organs and contribute to tissue damage. Support for the contribution of reverse neutrophil transmigration to organ damage comes from recent experimental evidence [44, 45].

NEUTROPHIL CROSSTALK WITH PLATELETS AND OTHER IMMUNE AND NON-IMMUNE CELL SUBSETS

Although neutrophil interactions are crucial during the inflammatory response, additionally, neutrophils contribute to their own homeostasis through their communication with niche cells in the bone marrow. The effector functions of neutrophils in several inflammatory diseases occur through their mutual interactions with other cell types, either by physical contact or through the release of cytokines, chemokines or granule proteins. Neutrophil communication with endothelial cells has been widely studied in the context of CVDs, where they increase endothelial permeability, contributing to the recruitment of other immune cells such as monocytes. Furthermore, the communication of neutrophils with endothelial cells in inflammatory conditions is usually concomitant with the interactions of further cell types. For example, in CVD, the communication between platelets and neutrophils is crucial to the combination of inflammation and thrombosis. Similarly, in acute kidney injury (AKI) and CKD, the interactions between neutrophils and platelets facilitate thrombosis through the formation of NETs. These NETs create a platform that contributes to platelet activation, which in turn accumulates and favors thrombosis, e.g. cholesterol crystal embolism [46]. However, this interaction is not always detrimental and its outcome depends on the tissue microenvironment, the nature of the disease and the inflammatory stimuli. In the case of bacterial pneumonia, the depletion of platelets promoted the accumulation of neutrophils in the lung, aggravating lung injury [47]. Hence, the fine tuning of these interactions is essential to avoid the perpetuation of inflammation or cardiovascular damage. Neutrophils remarkably impact the pathogenicity of sickle cell disease through their promiscuous interactions with red blood cells, platelets and endothelial cells, which initiate vaso-occlusion, through regulating adhesion molecules [48].

Neutrophils interact closely with monocytes and macrophages, starting in the bone marrow where macrophages influence granulopoiesis via granulocyte colony-stimulating factor release [49] and modulate neutrophil retention by CXCL12/CXCR4 [50] or by direct interaction [51] (Fig. 3). Macrophages induce neutrophil recruitment to the site of inflammation through the secretion of chemoattractants such as CXCL1/CXCL2 and MCP-1 [52, 53]. Apoptotic neutrophils are cleared by macrophages promoting the production of interleukin-4 (IL-4), which in turn reduces neutrophil trafficking, activation and effector functions [54]. Conversely, neutrophils promote monocyte and macrophage recruitment by secreting LL37 and azurocidin [55]. The release of azurocidin and lactoferrin supports a proinflammatory (M1-like) macrophage phenotype, while neutrophil-generated ROS and gelatinase-associated lipocalin triggers a reparative (M2-like) macrophage phenotype [56, 57]. Enhanced NETosis and granule proteins further enhance the phagocytosis of pathogens by macrophages [58, 59].

An external file that holds a picture, illustration, etc.
Object name is gfad266fig3.jpg

Crosstalk of neutrophils with other cells. Platelets: neutrophil interactions with platelets occurs by direct adhesion through PSGL-1, integrin αLβ2, CD40 on neutrophils and P-selectin, CD40L, ICAM-2 and CD40L on platelets. The release of C5a, C3a, HMGB1 and S100A8/A9 proteins by activated neutrophils promotes megakaryocyte proliferation and platelet production. Platelet secretion of extracellular MVs containing chemokines and the release of chemokines from platelet granules activate neutrophils and promote NET formation. Vice versa, NET proteins can activate the coagulation cascade in platelets. Endothelial cell: L-selectin mediates neutrophil activation of endothelial cells with E-selectin, P-selectin, Tissue Factor and ICAM-1 via MMP, C5a, S100A8/A9 and ROS. Endothelial cells recruit activated neutrophils via Integrin αMβ2 after undergoing NET release–related damage. Monocyte/macrophage: monocytes can extend the lifespan of neutrophils and promote neutrophil recruitment through the secretion of GM-CSF, TNF-α and G-CSF. Recruitment of monocytes is mediated by granule proteins (PR3, LL-37, CTSG, azurocidin and HNP1–3) released from neutrophils. Lactoferrin, azurocidin, S100A8/A9, HNP1–3, LL-37 and NETs induce M1-like macrophage polarization, while for example Anx1 promotes M2-like macrophage polarization. T and B cell: neutrophils can act as APCs to promote T-cell differentiation into effector T cells through MHC molecules but also inhibit T cell proliferation. Activated neutrophils express intracellular and extracellular BAFF, which binds to the BCMA receptor on B cells and promotes B-cell maturation, survival and differentiation into plasma cells [61]. Tubular epithelial cells: kidney injury causes tubular epithelial cell necrosis and subsequent release of pro-inflammatory mediators that indirectly trigger neutrophil activation and induce NET formation and NETosis. The induced inflammatory response (release of NETs) contributes to further tubular cell necrosis in a vicious cycle known as necroinflammation. This figure was generated with BioRender software. Anx1, annexin 1; APCs, antigen presenting cells; BAFF, B cell activating factor; BCMA, B cell maturation antigen; CD40L, CD40 ligand; CTSG, cathepsin G; C3a and C5a, complement component 3a and 5a; HNP1–3, human neutrophil peptides 1–3; HMGB1, high mobility group box 1; ICAM, intercellular adhesion molecule; LL-37, cathelicidin; MHC, major histocompatibility complex; MMP, matrix metalloproteinase; MVs, microvesicles; NE, neutrophil elastase; PSGL-1, P-selectin granulocyte ligand 1; PR3, proteinase 3.

Neutrophils interact not only with innate immune cells but also with the adaptive immune system (reviewed in [51]). For example, in the marginal zone of human spleens neutrophils acquire B cell–stimulating characteristics through the production of chemokines and cytokines including B-cell activating factor as well as the pattern recognition receptor pentraxin 3 (see Fig. 3), thus stimulating immunoglobulin production by nearby B cells [60–62]. The functions of T cells can be either positively or negatively modulated by neutrophils [63–65]. However, it remains unclear whether the capacity to influence T-cell functions is exerted by pre‐existing neutrophils, that under specific circumstances acquire immunomodulatory properties, or by discrete neutrophil populations that emerge under physiopathological conditions and become specialized in either promoting or inhibiting T‐cell responses [51].

Neutrophils indirectly interact with non-immune cells such as tubular epithelial cells in the kidney. Upon AKI, necrotic tubular epithelial cells release danger-associated molecular patterns (DAMPs) including cytokines and histones, which in turn promote neutrophil infiltration in the kidney and subsequent peptidyl argine deiminase 4 (PAD4)-mediated NETs formation and NETosis [66]. Such NETs contribute to further tubular epithelial cell death and DAMPs release in a vicious cycle known as necroinflammation (see Fig. 3) [67].

NEUTROPHILS IN INFECTIOUS DISEASES

Infections (e.g. COVID-19, sepsis, pneumonia, hepatitis, herpes zoster and tuberculosis) affecting millions of people each year are associated with high mortality rates. In patients with kidney disease, infectious complications are the second most common cause of death [17]. Complications of severe infections can range from thrombosis, acute respiratory distress syndrome and arterial hypotension to organ damage including the kidney [68]. Certain infections can also trigger vasculitis through immune complex deposition and dysregulated complement activation, such as anti-neutrophil cytoplasmic antibodies (ANCA)-associated vasculitis (AAV), hepatitis B virus–associated polyarteritis nodosa and hepatitis C virus–related cryoglobulinaemia, but conversely, infections are also very common in patients with vasculitis (reviewed in [69, 70]). Myeloid cells are known to contribute to the immunopathology of infection, with neutrophils being implicated in both hyperinflammation and immunosuppression through well-defined mechanisms including oxidative burst, degranulation, phagocytosis and NET formation. Interestingly, lymph-borne hematopoietic stem cells are shown to originate from the bone marrow and can proliferate to myeloid cells within the kidney when implanted under the kidney capsule in response to TLR pathogens, suggesting a role for neutrophil granulopoiesis during infection [71, 72].

Recent evidence points toward a heterogeneity and functional diversity of neutrophils. Of note, four clusters of immature neutrophils—termed IL1R2+, PADI4+, MPO+ and MKI67+CYP1B1+ neutrophils—were identified in the blood of people with sepsis [73]. Such neutrophils display transcriptional features of so-called granulocytic myeloid-derived suppressor cells, and they are able to inhibit the proliferation and activation of CD4+ T cells via prostaglandin E2 [73]. Additionally, low-density neutrophils (LDN) are also high in numbers in patients with severe COVID-19 during the acute phase, where they display enhanced phagocytic capacity, spontaneous NET formation and elevated cytokine production [68]. A subset of immunosuppressive CD16brightCD62Ldim neutrophils was identified in patients with severe COVID-19 that secondarily developed pulmonary embolism. In convalescent individuals, LDNs decrease in numbers, while CD16low neutrophils remain expanded over the disease course [74].

Studies have illustrated a phenomenon called neutrophil swarming, a rapid accumulation of neutrophils mediated by intercellular neutrophil communication and sensing of pathogen and damage signals at sites of infection or injury (reviewed in [75, 76]). These swarming neutrophils produce chemoattractants such as CXCL2 and lipid mediator leukotriene B4 that are positively regulated by calcium, complement, Bruton's tyrosine kinase [35] and integrins, while NADPH oxidase 2 rather negatively regulates neutrophil swarming [77]. Whether neutrophil swarming also occurs in kidney disease is currently unknown. It is possible that neutrophil swarming might contribute to granulomatous interstitial nephritis similar to that observed in chronic granulomatous lung disease [78].

Of note, some bacterial pathogens have even developed mechanisms to escape neutrophil control, leading to chronic infections in the host associated with complications such as tissue damage or eventually death. One such example is Mycobacterium tuberculosis. Recent insights suggest a beneficial role of neutrophils for the initial M. tuberculosis clearance but if this fails, neutrophils are detrimental due to their excessive recruitment, neutrophil necrosis and tissue damage [79].

In high-risk patients where neutrophil immunity to host defense is impaired, clinical outcomes are worse. Diseases that interfere with the immune system and contribute to neutrophil dysfunction include cancer, cardiovascular disease, diabetes, inflammatory disorders and kidney disease. Thus, patients with kidney disease are more susceptible to infections due to the secondary immunodeficiency related to kidney disease (SIDKD) (reviewed in [17]). Neutrophil dysfunctions include decreased phagocytic capability to clear pathogens, reduced respiratory burst, abolished selectin-induced slow leukocyte rolling and transmigration [80, 81], and increased neutrophil apoptosis but diminished NET release and NETosis [17, 82] (see Table 1). Further studies are needed to unravel the diverse functional phenotypes of neutrophils during infections but also during SIDKD.

Table 1:

Mechanistic features of neutrophils in health and disease.

Physiological statesMechanisms of actionRef
Health
 HomeostasisCXCR4 expression in circulating neutrophils from healthy subjects correlates with neutrophil migration and aging. CXCR4CD62L+ young and CXCR4+CD62L aged neutrophils are regulated by circadian oscillation[11, 145]
CD177 is a glycoprotein on the plasma membrane and can serve as receptor of membrane-bound proteinase 3). CD177+ neutrophils in the blood of healthy individuals can promote the interaction of neutrophils with endothelial cells and their transmigration into the tissue via PECAM-1 or β2 integrins[1, 146]
Human OLFM4+ and OLFM4 neutrophils do not display major functional differences other than a differential ability to produce NETs in vitro. OLFM4+ neutrophils can migrate and increase in numbers during inflammation relative to healthy conditions[147]
Other neutrophil subpopulations are present in healthy individuals such as neutrophils expressing the T-cell receptor αβ or proangiogenic CD49+CXCR4+ vascular endothelial growth factor (VEGFR1)+[148, 149]
Disease
 InfectionIncreased numbers of LDNs (CD66b+CD16intCD44lowCD11bint) in patients with COVID-19 during the acute phase[68]
Contribution of neutrophils to hyperinflammation via oxidative burst (ROS production), degranulation, phagocytosis and NET formation[73]
Identification of four immature neutrophil subsets (IL1R2+, PADI4+, MPO+ and MKI67+CYP1B1+) in the blood of patients with sepsis[73]
Neutrophils contribute to immunosuppression and act as granulocytic myeloid-derived suppressor cells (PMN-MDSCs, CD16brightCD62Ldim neutrophils)[74]
 Non-infectious diseases
  Kidney cancerTGF-β, G-CSF and GM-CSF drive neutrophils toward a pro-tumor phenotype, while type I IFNs facilitate anti-tumor activity of these cells[89, 91, 92]
  Autoimmune diseasesIn AAV: ANCAs bind to self-antigens such as MPO and proteinase 3, and trigger neutrophil activation and the release of ANCAs, inflammatory cytokines (e.g. IL-6), ROS and lytic enzymes (e.g. histones, neutrophil elastase, MMPs), and NET formation[150]
In SLE: three subpopulations of neutrophils exist in the blood of SLE patients, LDNs (CD11bbright, CD15+, CD16+CD33+CD14neg-loCD66bhighCD10+), mature high-density neutrophils (CD11b+CD15+CD16+CD66b+CD10+) and PMN-MDSCs (CD11b+CD14CD15+CD66b+). LDNs release pro-inflammatory cytokines (TNF, IFN-α, IL-18), have impaired phagocytosis, enhanced NET formation (NETosis) via release of mitochondrial ROS, GSDMD oligomerization-dependent mitochondrial DNA and IL-17 release[151, 152]
Neutrophils are involved in disrupting endothelial permeability through the secretion of granule proteins like azurocidin, proteinase-3 and α-defensin
  Cardiovascular diseaseNeutrophils mediate monocyte recruitment and endothelial adhesion through secretion of cathepsin G, cathelicidin and α-defensin-platelet-derived CCL5 complexes[111, 153–155]
Neutrophil recruitment occurs via β2 integrin activation, and arrest and crawling via E-selectin, ITAM and SLP-76/ADAP
  Acute and chronic kidney diseaseContribution of neutrophils to necroinflammation through release of pro-inflammatory mediators (ROS, MPO, neutrophil elastase and histones), NET formation and RIPK1–RIPK3-MLKL-dependent necroptosis[38]
Granulopoiesis: two distinct LDG subsets, CD14lowCD16+CD15+ (mature neutrophil-like phenotype with increased size and granularity, higher expression of CD10, CD35) and CD14CD16CD15+ (immature neutrophil-like phenotype, derived from the bone marrow, higher expression of CD68 and DEF3) were identified in CKD patients on dialysis[136]

ADAP, adhesion and degranulation-promoting adaptor protein; DEF3, defensing 3; GSDMD, gasdermin D; ITAM, immunoreceptor tyrosine-based activation motif; MMP, matrix metalloproteinase; SLP-76, SH2-domain-containing leukocyte protein of 76 kDa.

NEUTROPHILS IN NON-INFECTIOUS DISEASES

The role of neutrophils in kidney cancer

The role of neutrophils in cancer is heterogeneous, as they can play both pro- but also anti-tumoral roles [83, 84]. Emergency granulopoiesis that is induced by the growing tumor leads to neutrophil expansion and activation, resulting in a high neutrophil-to-lymphocyte ratio that is often observed in the blood of cancer patients, which is mainly associated with a poor prognosis in the majority of cancer types including kidney cancer, or head and neck cancer [85, 86]. As a consequence, increased numbers of neutrophils are also recruited into other tissues, such as tumor or tumor-draining lymph nodes (TDLNs). Elevated numbers of tumor-associated neutrophils (TANs) or TDLN neutrophils are mainly a negative prognostic marker for the majority of cancers. However, recent findings indicate that at the early stage of cancerogenesis, neutrophils can play also an immunostimulatory, anti-tumor role, and their presence in tissues correlates with a better patient prognosis [87, 88].

Tumorigenic activity of neutrophils is mediated via different cytokines and growth factors that are present in the cancer microenvironment. For example, transforming growth factor (TGF)-β, granulocyte colony-stimulating factor (G-CSF), tumor necrosis factor (TNF)-α, ceruloplasmin and granulocyte–macrophage colony-stimulating factor (GM-CSF) [89, 90], drive infiltrating neutrophils toward a pro-tumor inflammatory phenotype within the tumor milieu, while type I interferons (IFN) facilitate anti-tumor activity of these cells [91, 92]. Due to this plasticity, therapeutic approaches aiming at boosting their anti-tumor phenotype or reversing their pro-tumor phenotype may provide novel therapeutic targets. As an example, induction of trained innate immunity using β-glucan was shown to increase type I IFN responses and to promote anti-tumor activity of neutrophils [93].

Similar to other cancer types, also in kidney cancer a high neutrophil-to-lymphocyte ratio [86] and high frequencies of tumor-infiltrating neutrophils [94] are associated with poor prognosis. Consequently, recent evidence from transcriptomic studies [95] suggested that NETs may also promote human kidney cancer. For example, renal cell carcinoma was one of the first cancer types in which neutrophils with T-cell suppressive activity, also defined as polymorphonuclear myeloid-derived suppressor cells (PMN-MDSC) [84], were characterized and associated with poor outcome [96].

Neutrophils and their role in autoimmunity

In several autoimmune diseases such as RA, systemic lupus erythematosus (SLE), primary Sjögren's Syndrome, psoriasis, AAV or multiple sclerosis, neutrophils contribute to pathogenicity sustaining inflammation through their constant recruitment, the release of cytokines, chemokines and complement factors, as well as their own clearance [97]. In addition, in RA, psoriasis and SLE patients a subset of neutrophils, low-density granulocytes (LDGs), show elevated levels in the serum. These LDGs are considered pro-inflammatory and show an increased tendency to form NETs, in contrast to the normal-density granulocytes [83]. Besides creating a pro-inflammatory niche and tissue damage, the continuous release of NETs by neutrophils or their defective clearance by phagocytes can further contribute to autoimmunity by releasing self-nuclear antigens into the tissue [98]. Similarly, the defective clearance of apoptotic neutrophils is a well-described mechanism of generating autoimmunity via the generation of auto-antibodies against exposed self-antigens from the uncleared cell (reviewed in [99]). Intriguingly, in some tissues affected by autoimmune diseases, such as in the joint during RA or the spleen during SLE, both apoptotic and NETotic neutrophils are found in specific sub-tissular compartments. This zonation of the neutrophil death modality emerges as an active mechanism of self-tolerance breakdown and targeting their clearance by macrophages is proposed as a therapeutic approach. Yet, the mechanisms by which neutrophils selectively undergo different cell death modalities in specific locations within the same tissue need further investigation.

Furthermore, neutrophils can also be targeted by auto-antibodies against neutrophil-specific self-antigens such as myeloperoxidase (MPO) and proteinase 3 in AAV. Targeting of self MPO by auto-antibodies triggers excessive activation of neutrophils that subsequently release inflammatory cytokines, ROS and lytic enzymes, and form NETs [100]. Such excessive NET formation and reduced NET degradation/DNase I activity are harmful to small vessels. Moreover, NETs are involved not only in ANCA-mediated vascular injury but also in the production of ANCAs themselves through the release of citrullinated histones and double-stranded DNA. Therefore, a vicious cycle of NET formation and ANCA production is considered to be involved in the pathogenesis of AAV [100].

Neutrophils in cardiovascular disease

Cardiovascular disease is a leading cause of death affecting patients with CKD and end-stage kidney disease (ESKD). The increased risk for cardiovascular events in these patients emerges during early stages of kidney disease in the form of atherosclerosis, heart failure, valvular heart disease and sudden cardiac death [101]. Recent evidence has highlighted the critical role of neutrophils and their mediators in several stages of atherosclerosis. Neutrophil counts correlate with plaque size extension and higher neutrophil-to-lymphocyte ratio in CKD patients [102], and subsequently, studies using neutropenic mice demonstrated reduced atheroma plaques [103]. A causal relation of blood neutrophil counts and cardiovascular endpoints has recently been shown in a Mendelian randomization study [104]. Thus, the increase in neutrophil counts in patients with CKD may indeed stand out as important regulator of cardiovascular inflammation. In fact, patients with ESKD who undergo hemodialysis present with more cardiovascular events [105] and these correlated with higher MPO and DNAse I serum levels, and circulating DNA–histone complex [106–108].

In atherosclerosis, neutrophils and NETs are highly involved. For example, neutrophils can increase endothelial adhesion molecule expression and disrupt endothelial permeability through the secretion of granule proteins [109]. They mediate monocyte recruitment by secretion of chemotactic granule proteins and chemokines [110, 111] (Table 1). Neutrophils also accelerate foam cell formation by oxidizing low-density lipoproteins via neutrophil-secreted MPO [112] and induce a pro-inflammatory macrophage profile [113] through the secretion of granule proteins [59]. Inhibiting PAD4, a key NET-forming enzyme, through pharmacological treatment [114] or using myeloid-deficient PAD4 knockout mice [115] reduced atherosclerosis. NETs containing DNA–cathelicidin-related antimicrobial peptide complexes enhance atherosclerosis formation through plasmacytoid dendritic cell–secreted IFN-α [116]. NETs can also act as mediators during cell communication, priming macrophages to produce pro-inflammatory IL-1β, and neutrophil serine proteases directly contribute to IL-1β generation by pro-IL-1β cleavage [117]. Neutrophils and NETs also play a role in the later stages of atherosclerosis, inducing plaque destabilization [118, 119] and acceleration of atherosclerosis [120]. Thus, NETosis and derived products have been associated with an increased risk of CVD in patients with CKD and ESKD [121].

Intriguingly, neutrophils also participate in tissue repair alongside their pro-inflammatory role in CVD. Studies in mice with myocardial infarction revealed that neutrophil gelatinase-associated lipocalin increased cardiac macrophages’ phagocytic capacity to eliminate apoptotic cells [56]. Similarly, neutrophil-borne Annexin A1 induces an angiogenic macrophage profile promoting cardiac repair after ischemia [122]. In ischemic hearts, neutrophil and macrophage-produced oncostatin M–induced dedifferentiated cardiomyocytes to release regenerating islet-derived protein 3β and regulate macrophage accumulation during myocardial healing [123]. Furthermore, neutrophil-derived cathelicidin promoted re-endothelialization and arterial healing in a mouse model of atherosclerosis, reducing in-stent stenosis [124]. In conclusion, while traditionally viewed as immune cells worsening CVDs, neutrophils have emerged as key players in modulating tissue repair and inflammation resolution.

Neutrophils in acute and chronic kidney disease

AKI is caused by many triggers including nephrotoxic drugs, ischemia and metabolites that crystallize inside the kidney tubules, for instance, in cholesterol crystal embolism, oxalosis and uric acid nephropathy is associated with tubular injury, necrosis and acute inflammation [125, 126]. Necrotic tubular epithelial cells release danger signals that activate TLRs and other pattern recognition receptors on resident immune cells in the kidney interstitium, which in turn triggers a massive neutrophil influx [127]. This recruitment process involves β2 integrin activation, arrest and crawling, and subsequent migration of neutrophils into the inflamed kidney [38], where neutrophils form NETs (Fig. 3). Such NETs contribute to necroinflammation and immunothrombosis [126] through the release of inflammatory mediators and receptor-interacting protein kinase (RIPK)1–RIPK3-mixed lineage kinase domain-like (MLKL)-dependent necroptosis, highlighting the importance of neutrophils in necroinflammation [126]. In a model of ischemia/reperfusion-induced AKI, PAD4 expression augmented kidney injury and inflammation by driving NET formation [128]. Whether other forms of neutrophil cell death such as pyroptosis contribute to IL-1β-mediated inflammation in AKI is currently unknown.

In patients with CKD, biomarkers of oxidative stress and inflammation are interrelated. Thus, uremia leads to chronic low-grade inflammation, in which permanent immune cell activation and secretion of inflammatory mediators, together with uremic solutes/metabolites and immunoregulatory proteins, contribute to neutrophil dysfunction [17]. A number of immunoregulatory proteins that inhibit selectin-induced slow rolling and neutrophil recruitment, and decrease neutrophil respiratory burst have been identified such as leptin, resistin, p-cresol and fibroblast growth factor 23 [17, 129]. In addition, renal NETosis was shown to be increased through lipid sphingosine-1-phosphate signaling in a mouse model of CKD [130]. This suggests that neutrophils from hemodialysis patients have augmented NET formation [131] which associates with clinical markers of kidney disease [130]. Due to a leaky gut and shift in the secretome of the intestinal microbiota in CKD patients, gut-derived metabolites including indoxyl sulfate, p-cresyl sulfate and short-chain fatty acids can also drive neutrophil dysfunction [17]. Recent evidence suggests that the metabolite soluble uric acid impairs β2 integrin activation and internalization/recycling and therefore alters the migratory capability of neutrophils in sterile inflammation [132]. Moreover, neutrophils from patients with ESKD are more likely to undergo apoptosis, which decreases the ability of neutrophils to form NETs and to phagocytose and kill pathogens, mechanisms that are associated with an impaired host defense in SIDKD.

In addition, kidney disease also affects granulopoiesis (potentially via G-CSF released from injured tubular epithelial cells [133]) similar to that observed in autoimmune diseases [134] and cancer [135]. Two subsets of LDGs, CD14lowCD16+CD15+ (mature neutrophil-like phenotype) and CD14CD16CD15+ (immature neutrophil-like phenotype) have been identified in CKD patients on dialysis [136]. These CD14CD16CD15+ immature neutrophils express the early granulopoiesis marker defensing 3 [136]. In general, low-density granulocytes are considered as highly pro-inflammatory cells with pathogenic features [137] as compared with normal-density neutrophils, and might therefore contribute to the chronic low-grade inflammation in CKD.

SUMMARY AND THERAPEUTIC PRINCIPLES

Taken together, neutrophils have recently been recognized as cells with multifaceted roles in various biological contexts. They participate in acute host defense and represent the first line of innate immunity in the context of infection, but also contribute to a variety of acute and chronic inflammatory diseases. In cancer, neutrophils display heterogeneous roles: while their abundance in the blood of cancer patients often correlates with poor prognosis, early-stage neutrophil presence can have immunostimulatory anti-tumor effects. Furthermore, they are also involved in the pathogenesis of a variety of autoimmune diseases, contributing to systemic inflammation, augmentation of adaptive immunity and subsequent tissue damage.

Given their emerging roles in a wide range of diseases, therapeutic targeting of neutrophil functions represents a promising treatment approach in the areas of kidney disease, oncology, immunology, infectiology and even cardiology. Indeed, simple removal of neutrophils from the circulation by monocyte- or granulocyte apheresis is able to ameliorate certain inflammatory diseases like psoriasis [138], although these relatively crude approaches have been abandoned in favor of more sophisticated therapies. Although few existing drugs are specifically aimed at neutrophils, several drugs already target neutrophils either directly or indirectly, including corticosteroids, dapsone, cytokine-inhibiting antibodies such as anti-TNF-α and anti-IL-17A antibodies, immunomodulatory treatments targeting type I IFNs, or neutrophil protease inhibitors (e.g. sivelestat, alvestat) [139]. Complement inhibitors including avacopan are clinically used for the management of patients with AAV to block C5a-mediated neutrophil activation [140], and are promising approaches also for patients with immunoglobulin A nephropathy [141] and other forms of kidney disease. Of note, so far specific therapies based on specifically inhibiting neutrophil β2 integrins have failed clinical trials [142]. Based on our current knowledge, more specific treatment approaches are warranted including AKI and CKD, for example drugs targeting NETosis (e.g. PAD4 inhibitor Cl-amidine, GSK199, necrostatin 1) [143] and NETs [144], neutrophilic ROS production, neutrophil adhesion and migration (e.g. 5-lipoxygenase inhibitors) as well as neutrophil survival pathways, to specifically address pathology mediated by neutrophils while preserving the function of other immune cells.

Contributor Information

Daniela Maier-Begandt, Institute of Cardiovascular Physiology and Pathophysiology, Walter Brendel Center for Experimental Medicine Biomedical Center (BMC), Ludwig-Maximilians-Universität München, Munich, Germany.

Noelia Alonso-Gonzalez, Institute of Immunology, Universität of Münster, Münster, Germany.

Luisa Klotz, Department of Neurology with Institute for Translational Neurology, University Hospital Münster, Münster, Germany.

Luise Erpenbeck, Department of Dermatology, University Hospital Münster, Münster, Germany.

Jadwiga Jablonska, Department of Otorhinolaryngology, University Hospital Essen, University Duisburg-Essen, Essen, Germany. German Cancer Consortium (DKTK) partner site Düsseldorf/Essen, Essen, Germany.

Roland Immler, Institute of Cardiovascular Physiology and Pathophysiology, Walter Brendel Center for Experimental Medicine Biomedical Center (BMC), Ludwig-Maximilians-Universität München, Munich, Germany.

Anja Hasenberg, Institute of Experimental Immunology and Imaging, University Hospital Essen, University Duisburg-Essen, Essen, Germany.

Tonina T Mueller, Department of Medicine I, Ludwig-Maximilians-University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany.

Andrea Herrero-Cervera, Institute for Experimental Pathology, Center for Molecular Biology of Inflammation, Universität of Münster, Münster, Germany.

Irene Aranda-Pardos, Institute of Immunology, Universität of Münster, Münster, Germany.

Kailey Flora, Renal Division, Department of Medicine IV, Ludwig-Maximilians-University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany.

Alexander Zarbock, Department of Anaesthesiology, Intensive Care and Pain Medicine, University Hospital Münster, Münster, Germany.

Sven Brandau, Department of Otorhinolaryngology, University Hospital Essen, University Duisburg-Essen, Essen, Germany.

Christian Schulz, Department of Medicine I, Ludwig-Maximilians-University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany.

Oliver Soehnlein, Institute for Experimental Pathology, Center for Molecular Biology of Inflammation, Universität of Münster, Münster, Germany.

Stefanie Steiger, Renal Division, Department of Medicine IV, Ludwig-Maximilians-University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany.

FUNDING

This work was supported by the Deutsche Forschungsgemeinschaft (DFG, SFB TRR332) to S.S and K.F. (TP A7 and STE2437/4-1), O.S. and A.H.-C. (TP A2), S.B. (TP A4), C.S. (TP A6), J.J. and A.H. (TP A5), L.K. (TP B2), A.Z. (TP C1), R.I. (TP C2 and TRR359 project B02), D.M.-B. (TP C3), N.A.-G. (TP C5), and L.E. (TP AP1), on behalf of the TRR332 consortium.

AUTHORS’ CONTRIBUTIONS

S.S. conceptualized the manuscript. S.S., O.S., S.B., C.S., J.J., A.H., L.K., A.Z., D.M.-B., N.A.-G., R.I., K.F., T.T.M., A.H.-C., I.A.-P. and L.E. contributed to the draft and its revisions and have approved the submitted version. S.S. and K.F. generated the figures with BioRender software. The authors have agreed to be personally accountable for their own contributions and will ensure that questions related to the accuracy or integrity of any part of the work, even ones in which the author was not personally involved, are appropriately investigated and resolved, and the resolution documented in the literature.

DATA AVAILABILITY STATEMENT

No data are reported with this review article.

CONFLICT OF INTEREST STATEMENT

The authors declare that there are no conflicts of interest related to this article.

REFERENCES

1. Deniset JF, Kubes P.. Neutrophil heterogeneity: bona fide subsets or polarization states?  J Leukoc Biol  2018;103:829–38. 10.1002/JLB.3RI0917-361R [Abstract] [CrossRef] [Google Scholar]
2. Maas SL, Soehnlein O, Viola JR.. Organ-specific mechanisms of transendothelial neutrophil migration in the lung, liver, kidney, and aorta. Front Immunol  2018;9:2739. 10.3389/fimmu.2018.02739 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
3. Nourshargh S, Renshaw SA, Imhof BA.. Reverse migration of neutrophils: where, when, how, and why?  Trends Immunol  2016;37:273–86. 10.1016/j.it.2016.03.006 [Abstract] [CrossRef] [Google Scholar]
4. Peiseler M, Kubes P.. More friend than foe: the emerging role of neutrophils in tissue repair. J Clin Invest  2019;129:2629–39. 10.1172/JCI124616 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
5. Ballesteros I, Rubio-Ponce A, Genua M  et al.  Co-option of neutrophil fates by tissue environments. Cell  2020;183:1282–97.e18. 10.1016/j.cell.2020.10.003 [Abstract] [CrossRef] [Google Scholar]
6. Zhu YP, Padgett L, Dinh HQ  et al.  Identification of an early unipotent neutrophil progenitor with pro-tumoral activity in mouse and human bone marrow. Cell Rep  2018;24:2329–41.e8. 10.1016/j.celrep.2018.07.097 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
7. Kwok I, Becht E, Xia Y  et al.  Combinatorial single-cell analyses of granulocyte-monocyte progenitor heterogeneity reveals an early uni-potent neutrophil progenitor. Immunity  2020;53:303–18.e5. 10.1016/j.immuni.2020.06.005 [Abstract] [CrossRef] [Google Scholar]
8. Silvestre-Roig C, Fridlender ZG, Glogauer M  et al.  Neutrophil diversity in health and disease. Trends Immunol  2019;40:565–83. 10.1016/j.it.2019.04.012 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
9. Lawrence SM, Corriden R, Nizet V.. The ontogeny of a neutrophil: mechanisms of granulopoiesis and homeostasis. Microbiol Mol Biol Rev  2018;82:e00057–17. 10.1128/MMBR.00057-17 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
10. Evrard M, Kwok IWH, Chong SZ  et al.  Developmental analysis of bone marrow neutrophils reveals populations specialized in expansion, trafficking, and effector functions. Immunity  2018;48:364–79.e8. 10.1016/j.immuni.2018.02.002 [Abstract] [CrossRef] [Google Scholar]
11. Adrover JM, Del Fresno C, Crainiciuc G  et al.  A neutrophil timer coordinates immune defense and vascular protection. Immunity  2019;50:390–402.e10. 10.1016/j.immuni.2019.01.002 [Abstract] [CrossRef] [Google Scholar]
12. 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;13:170–80. 10.1038/ni.2194 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
13. Oh SJ, Lee JK, Shin OS.. Aging and the immune system: the impact of immunosenescence on viral infection, immunity and vaccine immunogenicity. Immune Netw  2019;19:e37. 10.4110/in.2019.19.e37 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
14. Van Avondt K, Strecker JK, Tulotta C  et al.  Neutrophils in aging and aging-related pathologies. Immunol Rev  2023;314:357–75. 10.1111/imr.13153 [Abstract] [CrossRef] [Google Scholar]
15. Wenisch C, Patruta S, Daxbock F  et al.  Effect of age on human neutrophil function. J Leukoc Biol  2000;67:40–5. 10.1002/jlb.67.1.40 [Abstract] [CrossRef] [Google Scholar]
16. Sapey E, Greenwood H, Walton G  et al.  Phosphoinositide 3-kinase inhibition restores neutrophil accuracy in the elderly: toward targeted treatments for immunosenescence. Blood  2014;123:239–48. 10.1182/blood-2013-08-519520 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
17. Steiger S, Rossaint J, Zarbock A  et al.  Secondary Immunodeficiency related to kidney disease (SIDKD)-definition, unmet need, and mechanisms. J Am Soc Nephrol  2022;33:259–78. 10.1681/ASN.2021091257 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
18. Xu J, Zhou L, Liu Y.. Cellular senescence in kidney fibrosis: pathologic significance and therapeutic strategies. Front Pharmacol  2020;11:601325. 10.3389/fphar.2020.601325 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
19. Margraf A, Lowell CA, Zarbock A.. Neutrophils in acute inflammation: current concepts and translational implications. Blood  2022;139:2130–44. 10.1182/blood.2021012295 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
20. Immler R, Simon SI, Sperandio M.. Calcium signalling and related ion channels in neutrophil recruitment and function. Eur J Clin Invest  2018;48  Suppl 2:e12964. 10.1111/eci.12964 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
21. Nemeth T, Sperandio M, Mocsai A.. Neutrophils as emerging therapeutic targets. Nat Rev Drug Discov  2020;19:253–75. 10.1038/s41573-019-0054-z [Abstract] [CrossRef] [Google Scholar]
22. Chung KJ, Mitroulis I, Wiessner JR  et al.  A novel pathway of rapid TLR-triggered activation of integrin-dependent leukocyte adhesion that requires Rap1 GTPase. Mol Biol Cell  2014;25:2948–55. 10.1091/mbc.e14-04-0867 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
23. Yago T, Zhang N, Zhao L  et al.  Selectins and chemokines use shared and distinct signals to activate beta2 integrins in neutrophils. Blood Adv  2018;2:731–44. 10.1182/bloodadvances.2017015602 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
24. Pruenster M, Kurz AR, Chung KJ  et al.  Extracellular MRP8/14 is a regulator of beta2 integrin-dependent neutrophil slow rolling and adhesion. Nat Commun  2015;6:6915. 10.1038/ncomms7915 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
25. Pruenster M, Immler R, Roth J  et al.  E-selectin-mediated rapid NLRP3 inflammasome activation regulates S100A8/S100A9 release from neutrophils via transient gasdermin D pore formation. Nat Immunol  2023;24:2021–31. 10.1038/s41590-023-01656-1 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
26. Wen L, Moser M, Ley K.. Molecular mechanisms of leukocyte beta2 integrin activation. Blood  2022;139:3480–92. 10.1182/blood.2021013500 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
27. Pick R, Begandt D, Stocker TJ  et al.  Coronin 1A, a novel player in integrin biology, controls neutrophil trafficking in innate immunity. Blood  2017;130:847–58. 10.1182/blood-2016-11-749622 [Abstract] [CrossRef] [Google Scholar]
28. Bader A, Winkelmann M, Forne I  et al.  Decoding the signaling profile of hematopoietic progenitor kinase 1 (HPK1) in innate immunity: a proteomic approach. Eur J Immunol  2022;52:760–9. 10.1002/eji.202149283 [Abstract] [CrossRef] [Google Scholar]
29. Margraf A, Germena G, Drexler HCA  et al.  The integrin-linked kinase is required for chemokine-triggered high-affinity conformation of the neutrophil beta2-integrin LFA-1. Blood  2020;136:2200–5. 10.1182/blood.2020004948 [Abstract] [CrossRef] [Google Scholar]
30. Immler R, Nadolni W, Bertsch A  et al.  The voltage-gated potassium channel KV1.3 regulates neutrophil recruitment during inflammation. Cardiovasc Res  2022;118:1289–302. 10.1093/cvr/cvab133 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
31. Zehrer A, Pick R, Salvermoser M  et al.  A fundamental role of Myh9 for neutrophil migration in innate immunity. J Immunol  2018;201:1748–64. 10.4049/jimmunol.1701400 [Abstract] [CrossRef] [Google Scholar]
32. Kurz AR, Pruenster M, Rohwedder I  et al.  MST1-dependent vesicle trafficking regulates neutrophil transmigration through the vascular basement membrane. J Clin Invest  2016;126:4125–39. 10.1172/JCI87043 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
33. Salvermoser M, Pick R, Weckbach LT  et al.  Myosin 1f is specifically required for neutrophil migration in 3D environments during acute inflammation. Blood  2018;131:1887–98. 10.1182/blood-2017-10-811851 [Abstract] [CrossRef] [Google Scholar]
34. Rohwedder I, Kurz ARM, Pruenster M  et al.  Src family kinase-mediated vesicle trafficking is critical for neutrophil basement membrane penetration. Haematologica  2020;105:1845–56. 10.3324/haematol.2019.225722 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
35. Volmering S, Block H, Boras M  et al.  The neutrophil Btk signalosome regulates integrin activation during sterile inflammation. Immunity  2016;44:73–87. 10.1016/j.immuni.2015.11.011 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
36. Crainiciuc G, Palomino-Segura M, Molina-Moreno M  et al.  Behavioural immune landscapes of inflammation. Nature  2022;601:415–21. 10.1038/s41586-021-04263-y [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
37. Kuligowski MP, Kitching AR, Hickey MJ.. Leukocyte recruitment to the inflamed glomerulus: a critical role for platelet-derived P-selectin in the absence of rolling. J Immunol  2006;176:6991–9. 10.4049/jimmunol.176.11.6991 [Abstract] [CrossRef] [Google Scholar]
38. Block H, Herter JM, Rossaint J  et al.  Crucial role of SLP-76 and ADAP for neutrophil recruitment in mouse kidney ischemia-reperfusion injury. J Exp Med  2012;209:407–21. 10.1084/jem.20111493 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
39. Liew  PX. Mired in the glomeruli: witnessing live neutrophil recruitment in the kidney. Am J Physiol Cell Physiol  2021;321:C384–93. 10.1152/ajpcell.00429.2020 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
40. Wu D, Zeng Y, Fan Y  et al.  Reverse-migrated neutrophils regulated by JAM-C are involved in acute pancreatitis-associated lung injury. Sci Rep  2016;6:20545. 10.1038/srep20545 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
41. Elks PM, van Eeden FJ, Dixon G  et al.  Activation of hypoxia-inducible factor-1alpha (Hif-1alpha) delays inflammation resolution by reducing neutrophil apoptosis and reverse migration in a zebrafish inflammation model. Blood  2011;118:712–22. 10.1182/blood-2010-12-324186 [Abstract] [CrossRef] [Google Scholar]
42. 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;12:761–9. 10.1038/ni.2062 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
43. 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  2005;79:303–11. 10.1189/jlb.0905496 [Abstract] [CrossRef] [Google Scholar]
44. Barkaway A, Rolas L, Joulia R  et al.  Age-related changes in the local milieu of inflamed tissues cause aberrant neutrophil trafficking and subsequent remote organ damage. Immunity  2021;54:1494–510.e7. 10.1016/j.immuni.2021.04.025 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
45. Owen-Woods C, Joulia R, Barkaway A  et al.  Local microvascular leakage promotes trafficking of activated neutrophils to remote organs. J Clin Invest  2020;130:2301–18. 10.1172/JCI133661 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
46. Shi C, Kim T, Steiger S  et al.  Crystal clots as therapeutic target in cholesterol crystal embolism. Circ Res  2020;126:e37–52. 10.1161/CIRCRESAHA.119.315625 [Abstract] [CrossRef] [Google Scholar]
47. Rossaint J, Thomas K, Mersmann S  et al.  Platelets orchestrate the resolution of pulmonary inflammation in mice by T reg cell repositioning and macrophage education. J Exp Med  2021;218:e20201353. 10.1084/jem.20201353 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
48. Hidalgo A, Chang J, Jang JE  et al.  Heterotypic interactions enabled by polarized neutrophil microdomains mediate thromboinflammatory injury. Nat Med  2009;15:384–91. 10.1038/nm.1939 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
49. Megyeri P, Sadowska J, Issekutz TB  et al.  Endotoxin-stimulated human macrophages produce a factor that induces polymorphonuclear leucocyte infiltration and is distinct from interleukin-1, tumour necrosis factor alpha and chemotactic factors. Immunology  1990;69:155–61. [Abstract] [Google Scholar]
50. Chow A, Lucas D, Hidalgo A  et al.  Bone marrow CD169+ macrophages promote the retention of hematopoietic stem and progenitor cells in the mesenchymal stem cell niche. J Exp Med  2011;208:261–71. 10.1084/jem.20101688 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
51. Schulz C, Petzold T, Ishikawa-Ankerhold H.. Macrophage regulation of granulopoiesis and neutrophil functions. Antioxid Redox Signal  2021;35:182–91. 10.1089/ars.2020.8203 [Abstract] [CrossRef] [Google Scholar]
52. Wang G, Huang W, Wang S  et al.  Macrophagic extracellular vesicle CXCL2 recruits and activates the neutrophil CXCR2/PKC/NOX4 axis in sepsis. J Immunol  2021;207:2118–28. 10.4049/jimmunol.2100229 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
53. De Filippo K, Dudeck A, Hasenberg M  et al.  Mast cell and macrophage chemokines CXCL1/CXCL2 control the early stage of neutrophil recruitment during tissue inflammation. Blood  2013;121:4930–7. 10.1182/blood-2013-02-486217 [Abstract] [CrossRef] [Google Scholar]
54. Impellizzieri D, Ridder F, Raeber ME  et al.  IL-4 receptor engagement in human neutrophils impairs their migration and extracellular trap formation. J Allergy Clin Immunol  2019;144:267–79.e4. 10.1016/j.jaci.2019.01.042 [Abstract] [CrossRef] [Google Scholar]
55. Soehnlein O, Zernecke A, Eriksson EE  et al.  Neutrophil secretion products pave the way for inflammatory monocytes. Blood  2008;112:1461–71. 10.1182/blood-2008-02-139634 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
56. Horckmans M, Ring L, Duchene J  et al.  Neutrophils orchestrate post-myocardial infarction healing by polarizing macrophages towards a reparative phenotype. Eur Heart J  2017;38:187–97. [Abstract] [Google Scholar]
57. Yang W, Tao Y, Wu Y  et al.  Neutrophils promote the development of reparative macrophages mediated by ROS to orchestrate liver repair. Nat Commun  2019;10:1076. 10.1038/s41467-019-09046-8 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
58. Monteith AJ, Miller JM, Maxwell CN  et al.  Neutrophil extracellular traps enhance macrophage killing of bacterial pathogens. Sci Adv  2021;7:eabj2101. 10.1126/sciadv.abj2101 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
59. Soehnlein O, Kai-Larsen Y, Frithiof R  et al.  Neutrophil primary granule proteins HBP and HNP1-3 boost bacterial phagocytosis by human and murine macrophages. J Clin Invest  2008;118:3491–502. 10.1172/JCI35740 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
60. Costa S, Bevilacqua D, Cassatella MA  et al.  Recent advances on the crosstalk between neutrophils and B or T lymphocytes. Immunology  2019;156:23–32. 10.1111/imm.13005 [Abstract] [CrossRef] [Google Scholar]
61. Herrero-Cervera A, Soehnlein O, Kenne E.. Neutrophils in chronic inflammatory diseases. Cell Mol Immunol  2022;19:177–91. 10.1038/s41423-021-00832-3 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
62. Chorny A, Casas-Recasens S, Sintes J  et al.  The soluble pattern recognition receptor PTX3 links humoral innate and adaptive immune responses by helping marginal zone B cells. J Exp Med  2016;213:2167–85. 10.1084/jem.20150282 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
63. Beauvillain C, Delneste Y, Scotet M  et al.  Neutrophils efficiently cross-prime naive T cells in vivo. Blood  2007;110:2965–73. 10.1182/blood-2006-12-063826 [Abstract] [CrossRef] [Google Scholar]
64. Vono M, Lin A, Norrby-Teglund A  et al.  Neutrophils acquire the capacity for antigen presentation to memory CD4(+) T cells in vitro and ex vivo. Blood  2017;129:1991–2001. 10.1182/blood-2016-10-744441 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
65. Carmona-Rivera C, Carlucci PM, Moore E  et al.  Synovial fibroblast-neutrophil interactions promote pathogenic adaptive immunity in rheumatoid arthritis. Sci Immunol  2017;2:eaag3358. 10.1126/sciimmunol.aag3358 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
66. Nakazawa D, Kumar SV, Marschner J  et al.  Histones and neutrophil extracellular traps enhance tubular necrosis and remote organ injury in ischemic AKI. J Am Soc Nephrol  2017;28:1753–68. 10.1681/ASN.2016080925 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
67. Mulay SR, Linkermann A, Anders HJ.. Necroinflammation in kidney disease. J Am Soc Nephrol  2016;27:27–39. 10.1681/ASN.2015040405 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
68. McKenna E, Wubben R, Isaza-Correa JM  et al.  Neutrophils in COVID-19: not innocent bystanders. Front Immunol  2022;13:864387. 10.3389/fimmu.2022.864387 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
69. Belizna CC, Hamidou MA, Levesque H  et al.  Infection and vasculitis. Rheumatology (Oxford)  2009;48:475–82. 10.1093/rheumatology/kep026 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
70. Miyabe C, Miyabe Y, Miyata R  et al.  Pathogens in vasculitis: is it really idiopathic?  JMA J  2021;4:216–24. [Europe PMC free article] [Abstract] [Google Scholar]
71. Paudel S, Ghimire L, Jin L  et al.  Regulation of emergency granulopoiesis during infection. Front Immunol  2022;13:961601. 10.3389/fimmu.2022.961601 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
72. Massberg S, Schaerli P, Knezevic-Maramica I  et al.  Immunosurveillance by hematopoietic progenitor cells trafficking through blood, lymph, and peripheral tissues. Cell  2007;131:994–1008. 10.1016/j.cell.2007.09.047 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
73. Barouni RM, Ostuni R.. Charting granulopoietic disturbances in sepsis. Nat Immunol  2023;24:746–8. 10.1038/s41590-023-01495-0 [Abstract] [CrossRef] [Google Scholar]
74. Chevrier S, Zurbuchen Y, Cervia C  et al.  A distinct innate immune signature marks progression from mild to severe COVID-19. Cell Rep Med  2021;2:100166. 10.1016/j.xcrm.2020.100166 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
75. Song Z, Bhattacharya S, Clemens RA  et al.  Molecular regulation of neutrophil swarming in health and disease: lessons from the phagocyte oxidase. iScience  2023;26:108034. 10.1016/j.isci.2023.108034 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
76. Kienle K, Lammermann T.. Neutrophil swarming: an essential process of the neutrophil tissue response. Immunol Rev  2016;273:76–93. 10.1111/imr.12458 [Abstract] [CrossRef] [Google Scholar]
77. Song Z, Huang G, Chiquetto Paracatu L  et al.  NADPH oxidase controls pulmonary neutrophil infiltration in the response to fungal cell walls by limiting LTB4. Blood  2020;135:891–903. 10.1182/blood.2019003525 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
78. Hopke A, Scherer A, Kreuzburg S  et al.  Neutrophil swarming delays the growth of clusters of pathogenic fungi. Nat Commun  2020;11:2031. 10.1038/s41467-020-15834-4 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
79. Dallenga T, Schaible UE.. Neutrophils in tuberculosis—first line of defence or booster of disease and targets for host-directed therapy?  Pathog Dis  2016;74:ftw012. 10.1093/femspd/ftw012 [Abstract] [CrossRef] [Google Scholar]
80. Rossaint J, Spelten O, Kassens N  et al.  Acute loss of renal function attenuates slow leukocyte rolling and transmigration by interfering with intracellular signaling. Kidney Int  2011;80:493–503. 10.1038/ki.2011.125 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
81. Zarbock A, Schmolke M, Spieker T  et al.  Acute uremia but not renal inflammation attenuates aseptic acute lung injury: a critical role for uremic neutrophils. J Am Soc Nephrol  2006;17:3124–31. 10.1681/ASN.2006040358 [Abstract] [CrossRef] [Google Scholar]
82. Chonchol  M. Neutrophil dysfunction and infection risk in end-stage renal disease. Semin Dial  2006;19:291–6. 10.1111/j.1525-139X.2006.00175.x [Abstract] [CrossRef] [Google Scholar]
83. Coffelt SB, Wellenstein MD, de Visser KE.. Neutrophils in cancer: neutral no more. Nat Rev Cancer  2016;16:431–46. 10.1038/nrc.2016.52 [Abstract] [CrossRef] [Google Scholar]
84. Moses K, Brandau S.. Human neutrophils: their role in cancer and relation to myeloid-derived suppressor cells. Semin Immunol  2016;28:187–96. 10.1016/j.smim.2016.03.018 [Abstract] [CrossRef] [Google Scholar]
85. Trellakis S, Bruderek K, Dumitru CA  et al.  Polymorphonuclear granulocytes in human head and neck cancer: enhanced inflammatory activity, modulation by cancer cells and expansion in advanced disease. Int J Cancer  2011;129:2183–93. 10.1002/ijc.25892 [Abstract] [CrossRef] [Google Scholar]
86. Mjaess G, Chebel R, Karam A  et al.  Prognostic role of neutrophil-to-lymphocyte ratio (NLR) in urological tumors: an umbrella review of evidence from systematic reviews and meta-analyses. Acta Oncol  2021;60:704–13. 10.1080/0284186X.2021.1886323 [Abstract] [CrossRef] [Google Scholar]
87. Eruslanov EB, Bhojnagarwala PS, Quatromoni JG  et al.  Tumor-associated neutrophils stimulate T cell responses in early-stage human lung cancer. J Clin Invest  2014;124:5466–80. 10.1172/JCI77053 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
88. Pylaeva E, Korschunow G, Spyra I  et al.  During early stages of cancer, neutrophils initiate anti-tumor immune responses in tumor-draining lymph nodes. Cell Rep  2022;40:111171. 10.1016/j.celrep.2022.111171 [Abstract] [CrossRef] [Google Scholar]
89. Fridlender ZG, Sun J, Kim S  et al.  Polarization of tumor-associated neutrophil phenotype by TGF-beta: “N1” versus “N2” TAN. Cancer Cell  2009;16:183–94. 10.1016/j.ccr.2009.06.017 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
90. Maas RR, Soukup K, Fournier N  et al.  The local microenvironment drives activation of neutrophils in human brain tumors. Cell  2023;186:4546–66.e27. 10.1016/j.cell.2023.08.043 [Abstract] [CrossRef] [Google Scholar]
91. Jablonska J, Leschner S, Westphal K  et al.  Neutrophils responsive to endogenous IFN-beta regulate tumor angiogenesis and growth in a mouse tumor model. J Clin Invest  2010;120:1151–64. 10.1172/JCI37223 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
92. 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;138:1982–93. 10.1002/ijc.29945 [Abstract] [CrossRef] [Google Scholar]
93. Kalafati L, Kourtzelis I, Schulte-Schrepping J  et al.  Innate immune training of granulopoiesis promotes anti-tumor activity. Cell  2020;183:771–85.e12. 10.1016/j.cell.2020.09.058 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
94. Jensen HK, Donskov F, Marcussen N  et al.  Presence of intratumoral neutrophils is an independent prognostic factor in localized renal cell carcinoma. J Clin Oncol  2009;27:4709–17. 10.1200/JCO.2008.18.9498 [Abstract] [CrossRef] [Google Scholar]
95. Li R, Jiang X, Wang P  et al.  Prognostic value of neutrophil extracellular trap signature in clear cell renal cell carcinoma. Front Oncol  2023;13:1205713. 10.3389/fonc.2023.1205713 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
96. 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;69:1553–60. 10.1158/0008-5472.CAN-08-1921 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
97. Hollstein MM, Manzke V, Scheidmann SEF  et al.  Targeting neutrophil extracellular traps (NETs) ameliorates inflammation in murine psoriasiform dermatitis. J Dermatol Sci  2023;109:143–6. 10.1016/j.jdermsci.2023.02.006 [Abstract] [CrossRef] [Google Scholar]
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;3:73ra19. 10.1126/scitranslmed.3001180 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
99. Singhal A, Kumar S.. Neutrophil and remnant clearance in immunity and inflammation. Immunology  2022;165:22–43. 10.1111/imm.13423 [Abstract] [CrossRef] [Google Scholar]
100. Nakazawa D, Masuda S, Tomaru U  et al.  Pathogenesis and therapeutic interventions for ANCA-associated vasculitis. Nat Rev Rheumatol  2019;15:91–101. 10.1038/s41584-018-0145-y [Abstract] [CrossRef] [Google Scholar]
101. Jankowski J, Floege J, Fliser D  et al.  Cardiovascular disease in chronic kidney disease: pathophysiological insights and therapeutic options. Circulation  2021;143:1157–72. 10.1161/CIRCULATIONAHA.120.050686 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
102. Angkananard T, Anothaisintawee T, McEvoy M  et al.  Neutrophil lymphocyte ratio and cardiovascular disease risk: a systematic review and meta-analysis. Biomed Res Int  2018;2018:1. 10.1155/2018/2703518 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
103. Drechsler M, Megens RT, van Zandvoort M  et al.  Hyperlipidemia-triggered neutrophilia promotes early atherosclerosis. Circulation  2010;122:1837–45. 10.1161/CIRCULATIONAHA.110.961714 [Abstract] [CrossRef] [Google Scholar]
104. Luo J, Thomassen JQ, Nordestgaard BG  et al.  Neutrophil counts and cardiovascular disease. Eur Heart J  2023;44:4953–64. 10.1093/eurheartj/ehad649 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
105. Sud M, Tangri N, Pintilie M  et al.  Risk of end-stage renal disease and death after cardiovascular events in chronic kidney disease. Circulation  2014;130:458–65. 10.1161/CIRCULATIONAHA.113.007106 [Abstract] [CrossRef] [Google Scholar]
106. Rickli C, Borato DCK, Silva A  et al.  Serum myeloperoxidase, C-reactive protein and alpha1-acid glycoprotein: insights about cardiovascular risk in end-stage renal disease. Exp Clin Endocrinol Diabetes  2020;128:731–6. [Abstract] [Google Scholar]
107. Kim JK, Lee HW, Joo N  et al.  Prognostic role of circulating neutrophil extracellular traps levels for long-term mortality in new end-stage renal disease patients. Clin Immunol  2020;210:108263. 10.1016/j.clim.2019.108263 [Abstract] [CrossRef] [Google Scholar]
108. Jeong JC, Kim JE, Gu JY  et al.  Significance of the DNA-histone complex level as a predictor of major adverse cardiovascular events in hemodialysis patients: the effect of uremic toxin on DNA-histone complex formation. Blood Purif  2016;41:64–71. 10.1159/000440974 [Abstract] [CrossRef] [Google Scholar]
109. Gautam N, Olofsson AM, Herwald H  et al.  Heparin-binding protein (HBP/CAP37): a missing link in neutrophil-evoked alteration of vascular permeability. Nat Med  2001;7:1123–7. 10.1038/nm1001-1123 [Abstract] [CrossRef] [Google Scholar]
110. Winter C, Silvestre-Roig C, Ortega-Gomez A  et al.  Chrono-pharmacological targeting of the CCL2-CCR2 axis ameliorates atherosclerosis. Cell Metab  2018;28:175–82.e5. 10.1016/j.cmet.2018.05.002 [Abstract] [CrossRef] [Google Scholar]
111. Doring Y, Drechsler M, Wantha S  et al.  Lack of neutrophil-derived CRAMP reduces atherosclerosis in mice. Circ Res  2012;110:1052–6. 10.1161/CIRCRESAHA.112.265868 [Abstract] [CrossRef] [Google Scholar]
112. Delporte C, Boudjeltia KZ, Noyon C  et al.  Impact of myeloperoxidase-LDL interactions on enzyme activity and subsequent posttranslational oxidative modifications of apoB-100. J Lipid Res  2014;55:747–57. 10.1194/jlr.M047449 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
113. Warnatsch A, Ioannou M, Wang Q  et al.  Inflammation. Neutrophil extracellular traps license macrophages for cytokine production in atherosclerosis. Science  2015;349:316–20. 10.1126/science.aaa8064 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
114. Knight JS, Luo W, O'Dell AA  et al.  Peptidylarginine deiminase inhibition reduces vascular damage and modulates innate immune responses in murine models of atherosclerosis. Circ Res  2014;114:947–56. 10.1161/CIRCRESAHA.114.303312 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
115. Liu Y, Carmona-Rivera C, Moore E  et al.  Myeloid-specific deletion of peptidylarginine deiminase 4 mitigates atherosclerosis. Front Immunol  2018;9:1680. 10.3389/fimmu.2018.01680 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
116. Doring Y, Manthey HD, Drechsler M  et al.  Auto-antigenic protein-DNA complexes stimulate plasmacytoid dendritic cells to promote atherosclerosis. Circulation  2012;125:1673–83. 10.1161/CIRCULATIONAHA.111.046755 [Abstract] [CrossRef] [Google Scholar]
117. Soehnlein O, Ortega-Gomez A, Doring Y  et al.  Neutrophil-macrophage interplay in atherosclerosis: protease-mediated cytokine processing versus NET release. Thromb Haemost  2015;114:866–7. [Abstract] [Google Scholar]
118. Silvestre-Roig C, Braster Q, Wichapong K  et al.  Externalized histone H4 orchestrates chronic inflammation by inducing lytic cell death. Nature  2019;569:236–40. 10.1038/s41586-019-1167-6 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
119. Geng S, Zhang Y, Lee C  et al.  Novel reprogramming of neutrophils modulates inflammation resolution during atherosclerosis. Sci Adv  2019;5:eaav2309. 10.1126/sciadv.aav2309 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
120. Schumski A, Ortega-Gomez A, Wichapong K  et al.  Endotoxinemia accelerates atherosclerosis through electrostatic charge-mediated monocyte adhesion. Circulation  2021;143:254–66. 10.1161/CIRCULATIONAHA.120.046677 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
121. Thakur M, Junho CVC, Bernhard SM  et al.  NETs-induced thrombosis impacts on cardiovascular and chronic kidney disease. Circ Res  2023;132:933–49. 10.1161/CIRCRESAHA.123.321750 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
122. Ferraro B, Leoni G, Hinkel R  et al.  Pro-angiogenic macrophage phenotype to promote myocardial repair. J Am Coll Cardiol  2019;73:2990–3002. 10.1016/j.jacc.2019.03.503 [Abstract] [CrossRef] [Google Scholar]
123. Lorchner H, Poling J, Gajawada P  et al.  Myocardial healing requires Reg3beta-dependent accumulation of macrophages in the ischemic heart. Nat Med  2015;21:353–62. 10.1038/nm.3816 [Abstract] [CrossRef] [Google Scholar]
124. Soehnlein O, Wantha S, Simsekyilmaz S  et al.  Neutrophil-derived cathelicidin protects from neointimal hyperplasia. Sci Transl Med  2011;3:103ra98. 10.1126/scitranslmed.3002531 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
125. Kellum JA, Romagnani P, Ashuntantang G  et al.  Acute kidney injury. Nat Rev Dis Primers  2021;7:52. 10.1038/s41572-021-00284-z [Abstract] [CrossRef] [Google Scholar]
126. Mulay SR, Steiger S, Shi C  et al.  A guide to crystal-related and nano- or microparticle-related tissue responses. FEBS J  2020;287:818–32. 10.1111/febs.15174 [Abstract] [CrossRef] [Google Scholar]
127. Immler R, Lange-Sperandio B, Steffen T  et al.  Extratubular polymerized uromodulin induces leukocyte recruitment and inflammation in vivo. Front Immunol  2020;11:588245. 10.3389/fimmu.2020.588245 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
128. Raup-Konsavage WM, Wang Y, Wang WW  et al.  Neutrophil peptidyl arginine deiminase-4 has a pivotal role in ischemia/reperfusion-induced acute kidney injury. Kidney Int  2018;93:365–74. 10.1016/j.kint.2017.08.014 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
129. Rossaint J, Oehmichen J, Van Aken H  et al.  FGF23 signaling impairs neutrophil recruitment and host defense during CKD. J Clin Invest  2016;126:962–74. 10.1172/JCI83470 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
130. Jones BA, Myakala K, Guha M  et al.  Farnesoid X receptor prevents neutrophil extracellular traps via reduced sphingosine-1-phosphate in chronic kidney disease. Am J Physiol Renal Physiol  2023;325:F792–810. 10.1152/ajprenal.00292.2023 [Abstract] [CrossRef] [Google Scholar]
131. Kim JK, Park MJ, Lee HW  et al.  The relationship between autophagy, increased neutrophil extracellular traps formation and endothelial dysfunction in chronic kidney disease. Clin Immunol  2018;197:189–97. 10.1016/j.clim.2018.10.003 [Abstract] [CrossRef] [Google Scholar]
132. Ma Q, Immler R, Pruenster M  et al.  Soluble uric acid inhibits beta2 integrin-mediated neutrophil recruitment in innate immunity. Blood  2022;139:3402–17. 10.1182/blood.2021011234 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
133. Volkmann J, Schmitz J, Nordlohne J  et al.  Kidney injury enhances renal G-CSF expression and modulates granulopoiesis and human neutrophil CD177 in vivo. Clin Exp Immunol  2019;199:97–108. 10.1111/cei.13372 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
134. Singh N, Traisak P, Martin KA  et al.  Genomic alterations in abnormal neutrophils isolated from adult patients with systemic lupus erythematosus. Arthritis Res Ther  2014;16:R165. 10.1186/ar4681 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
135. Sagiv JY, Michaeli J, Assi S  et al.  Phenotypic diversity and plasticity in circulating neutrophil subpopulations in cancer. Cell Rep  2015;10:562–73. 10.1016/j.celrep.2014.12.039 [Abstract] [CrossRef] [Google Scholar]
136. Rodriguez-Carrio J, Carrillo-Lopez N, Ulloa C  et al.  A subset of low density granulocytes is associated with vascular calcification in chronic kidney disease patients. Sci Rep  2019;9:13230. 10.1038/s41598-019-49429-x [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
137. 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;184:3284–97. 10.4049/jimmunol.0902199 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
138. Kanekura T, Kawabata H, Maruyama I  et al.  Treatment of psoriatic arthritis with granulocyte and monocyte adsorption apheresis. J Am Acad Dermatol  2004;50:242–6. 10.1016/S0190-9622(03)02474-5 [Abstract] [CrossRef] [Google Scholar]
139. Bronze-da-Rocha E, Santos-Silva A.. Neutrophil elastase inhibitors and chronic kidney disease. Int J Biol Sci  2018;14:1343–60. 10.7150/ijbs.26111 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
140. Jayne DRW, Merkel PA, Schall TJ  et al.  Avacopan for the treatment of ANCA-associated vasculitis. N Engl J Med  2021;384:599–609. 10.1056/NEJMoa2023386 [Abstract] [CrossRef] [Google Scholar]
141. Bruchfeld A, Magin H, Nachman P  et al.  C5a receptor inhibitor avacopan in immunoglobulin A nephropathy-an open-label pilot study. Clin Kidney J  2022;15:922–8. 10.1093/ckj/sfab294 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
142. Dove  A. CD18 trials disappoint again. Nat Biotechnol  2000;18:817–8. 10.1038/78412 [Abstract] [CrossRef] [Google Scholar]
143. Cecchi I, Arias de la Rosa I, Menegatti E  et al.  Neutrophils: novel key players in rheumatoid arthritis. Current and future therapeutic targets. Autoimmun Rev  2018;17:1138–49. 10.1016/j.autrev.2018.06.006 [Abstract] [CrossRef] [Google Scholar]
144. Chirivi RGS, van Rosmalen JWG, van der Linden M  et al.  Therapeutic ACPA inhibits NET formation: a potential therapy for neutrophil-mediated inflammatory diseases. Cell Mol Immunol  2021;18:1528–44. 10.1038/s41423-020-0381-3 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
145. Ella K, Csepanyi-Komi R, Kaldi K.. Circadian regulation of human peripheral neutrophils. Brain Behav Immun  2016;57:209–21. 10.1016/j.bbi.2016.04.016 [Abstract] [CrossRef] [Google Scholar]
146. Hellebrekers P, Vrisekoop N, Koenderman L.. Neutrophil phenotypes in health and disease. Eur J Clin Invest  2018;48:e12943. 10.1111/eci.12943 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
147. 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:e69575. 10.1371/journal.pone.0069575 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
148. Puellmann K, Kaminski WE, Vogel M  et al.  A variable immunoreceptor in a subpopulation of human neutrophils. P Natl Acad Sci USA  2006;103:14441–6. 10.1073/pnas.0603406103 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
149. Massena S, Christoffersson G, Vagesjo E  et al.  Identification and characterization of VEGF-A-responsive neutrophils expressing CD49d, VEGFR1, and CXCR4 in mice and humans. Blood  2015;126:2016–26. 10.1182/blood-2015-03-631572 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
150. Rasmuson J, Kenne E, Wahlgren M  et al.  Heparinoid sevuparin inhibits Streptococcus-induced vascular leak through neutralizing neutrophil-derived proteins. FASEB J  2019;33:10443–52. 10.1096/fj.201900627R [Abstract] [CrossRef] [Google Scholar]
151. Tay SH, Celhar T, Fairhurst AM.. Low-density neutrophils in systemic lupus erythematosus. Arthritis Rheumatol  2020;72:1587–95. 10.1002/art.41395 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
152. Miao N, Wang Z, Wang Q  et al.  Oxidized mitochondrial DNA induces gasdermin D oligomerization in systemic lupus erythematosus. Nat Commun  2023;14:872. 10.1038/s41467-023-36522-z [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
153. Soehnlein O, Lindbom L, Weber C.. Mechanisms underlying neutrophil-mediated monocyte recruitment. Blood  2009;114:4613–23. 10.1182/blood-2009-06-221630 [Abstract] [CrossRef] [Google Scholar]
154. Ortega-Gomez A, Salvermoser M, Rossaint J  et al.  Cathepsin G controls arterial but not venular myeloid cell recruitment. Circulation  2016;134:1176–88. 10.1161/CIRCULATIONAHA.116.024790 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
155. Alard JE, Ortega-Gomez A, Wichapong K  et al.  Recruitment of classical monocytes can be inhibited by disturbing heteromers of neutrophil HNP1 and platelet CCL5. Sci Transl Med  2015;7:317ra196. 10.1126/scitranslmed.aad5330 [Abstract] [CrossRef] [Google Scholar]
156. Kolaczkowska E, Kubes P.. Neutrophil recruitment and function in health and inflammation. Nat Rev Immunol  2013;13:159–75. 10.1038/nri3399 [Abstract] [CrossRef] [Google Scholar]

Articles from Nephrology Dialysis Transplantation are provided here courtesy of Oxford University Press

Citations & impact 


This article has not been cited yet.

Impact metrics

Alternative metrics

Altmetric item for https://www.altmetric.com/details/157631760
Altmetric
Discover the attention surrounding your research
https://www.altmetric.com/details/157631760

Similar Articles 


To arrive at the top five similar articles we use a word-weighted algorithm to compare words from the Title and Abstract of each citation.

Funding 


Funders who supported this work.

Deutsche Forschungsgemeinschaft (3)