Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2024 Mar 1.
Published in final edited form as: Immunol Rev. 2022 Oct 17;314(1):302–312. doi: 10.1111/imr.13146

Neutrophils as drivers of vascular injury in sickle cell disease

Lidiane S Torres 1,2, Andrés Hidalgo 3,4
PMCID: PMC10132504  NIHMSID: NIHMS1888788  PMID: 36251624

Summary

While neutrophils are the main effectors of protective innate immune responses, they are also key players in inflammatory pathologies. Sickle cell disease (SCD) is a genetic blood disorder in which red blood cells (RBCs) are constantly destroyed in the circulation which generates a highly inflammatory environment that culminates in vascular occlusions. Vaso-occlusion is the hallmark of SCD and a predictor of disease severity. Neutrophils initiate and propagate SCD-related vaso-occlusion through adhesive interactions with the activated and dysfunctional endothelium, sickle RBCs, and platelets, leading to acute and chronic complications that progress to irreversible organ damage and ultimately death. The use of SCD humanized mouse models, in combination with in vivo imaging techniques, has emerged as a fundamental tool to understand the dynamics of neutrophils under complex inflammatory contexts and their contribution to vascular injury in SCD. In this review, we discuss the various mechanisms by which circulating neutrophils sense and respond to the wide range of stimuli present in the blood of SCD patients and mice. We argue that the central role of neutrophils in SCD can be rationalized to develop targets for the management of clinical complications in SCD patients.

Keywords: endothelium, cell adhesion, inflammation, vaso-occlusion, hemolysis

1. INTRODUCTION

Neutrophils are key components of the innate immune system and the first responders to infection and inflammation 1. Approximately 1011 neutrophils are generated every day in the bone marrow compartment, where they undergo different stages of development from myeloid progenitor cells to mature neutrophils 2,3. The granulocyte colony stimulating factor (G-CSF) is the main regulator of granulopoiesis and release of mature neutrophils into the bloodstream 46. G-CSF induces the commitment of progenitor cells to the myeloid lineage, promotes proliferation of neutrophil precursors, and reduces the transit time through the bone marrow compartment 1,7. After their release from the bone marrow, neutrophils circulate for about half-day before migrating to tissues, such as liver, lung and spleen, or homing back to the bone marrow, with consequences in regulating the hematopoietic niches within 8.

During infections or upon inflammatory stimuli, granulopoiesis is increased in the bone marrow and a large number of neutrophils migrate to the site of infection or injury. In this process, their lifespan, originally short, is believed to be considerably extended 9. Acute inflammation is a highly coordinated process modulated by inflammatory mediators released by sentinel immune cells and the endothelium, followed by a resolution phase that reestablishes homeostasis 10. However, when the inflammatory stimulus is not removed or is constantly generated, this resolution phase fails and creates a chronic inflammatory state that drives organ pathology. This is the case of atherosclerosis, cancer, sterile injury, diabetes mellitus, autoimmune diseases and, as detailed below, sickle cell disease. In all of these disorders neutrophils have been demonstrated to play a prominent pathogenic role 11,12.

The endothelium is a physical barrier between blood cells and the tissue parenchyma that is composed by a highly dynamic monolayer of cells located in the inner face of the vessels. These endothelial cells continuously respond to the extracellular environment and play an extensive role in immune responses and maintenance of vascular hemostasis. The endothelium controls vessel wall permeability, regulates vascular pressure and blood flow, establishes the balance of pro- and anticoagulants, and regulates inflammation signaling 13. Constant activation of endothelial cells by excessive inflammatory stimuli, however, promotes endothelial damage and a gradual loss of function. While a healthy endothelium has vaso-dilation capacity and anti-thrombotic and anti-inflammatory functions preserved, a dysfunctional endothelium promotes vaso-constriction and presents a pro-inflammatory and pro-thrombotic state, which can irreversibly damage the vasculature and the closely irrigated organs 14.

Sickle cell disease (SCD), a genetic blood disorder, is a classic example of chronic (non-resolving) inflammatory condition with progressive multiorgan damage, in which neutrophil-endothelium interactions are key determinants of disease severity and mortality 1517. Over the past two decades, neutrophils have emerged as protagonists of SCD pathophysiology. The severely altered environment promoted by SCD creates a unique model of inflammatory disease accompanied by vasculopathy in which neutrophils may display a broad spectrum of phenotypes and functions. In this review, we explore neutrophil biology and elucidate some of the fascinating features of these cells under the prism of vascular occlusions in SCD.

2. SICKLE CELL DISEASE AS AN INFLAMMATORY DISEASE

The most common and severe form of SCD is caused by the homozygous inheritance of a point mutation in the beta-globin gene (HBB), affecting the beta-globin chains that form the hemoglobin tetramer 18. The normal adult hemoglobin (HbA) is formed by a tetramer of two alpha (α2) and two beta (β2) globin chains (α2β2), while the point mutation that leads to SCD results in the abnormal hemoglobin S (HbS, α2βS2). The pathophysiology of SCD is triggered by the high propensity of HbS to form polymers in red blood cells (RBCs) when deoxygenated, leading to severe cellular alterations in RBC, such as abnormal rheology, dehydration, expression of adhesion molecules, and the acquisition of a stiffed and sickled shape, the major cellular feature of mutation disease19. Abnormalities in RBCs caused by HbS polymerization manifest in a vicious cycle of hemolysis and vascular occlusions, the hallmarks of this disease. Shortening of sickle RBCs lifespan results in the destruction of 10% of total RBCs per day, leading to chronic hemolytic anemia. Approximately one-third of sickle-related hemolysis occurs intravascularly, with release of cell-free hemoglobin into the blood that directly damages the vasculature, generates ROS, and triggers various inflammatory pathways (Fig. 1A)20,21.

Figure 1. Current paradigm of vaso-occlusion in sickle cell disease.

Figure 1.

(A) Hemolysis leads to the release of damage associated molecular patterns (DAMPs) from red blood cells (RBCs) into the blood. Free hemoglobin (Hb) scavenges endothelial nitric oxide (NO), reducing its availability to smooth muscle cells and impairing blood vessel dilation. Hb oxidation forms methemoglobin (MetHb), a highly reactive molecule that generates reactive oxygen species (ROS) and releases free heme. (B) The endothelial damage is perpetuated by direct activation of endothelial cells by heme and ROS. The activated endothelium overexpresses adhesion molecules and releases inflammatory cytokines to the circulation, which in turn recruit and activate neutrophils. Neutrophils are also activated by heme and ROS. This cascade of reactions leads to inflammation and vaso-occlusion. (C) Neutrophils are the central effectors of vaso-occlusion due to adhesive interactions with RBCs, platelets and the endothelium, mediated by adhesion molecules as illustrated. PSGL-1, P-selectin glycoprotein ligand-1; ESL-1, E-selectin ligand-1; Mac-1, αMβ2 integrin (CD11b/CD18); LFA-1, αLβ2 integrin (CD11b/CD18); ICAM-1, intercellular adhesion molecule-1; VCAM-1, vascular cell adhesion molecule-1; GPIba, glycoprotein Ib alpha.

Extracellular oxygenated hemoglobin (Oxy-Hb-Fe2+) depletes endothelial nitric oxide (NO) in a rapid and irreversible reaction that forms methemoglobin (Hb-Fe3+) and nitrate (NO3−) 22,23. The compromised bioavailability of endothelial-derived NO to the adjacent smooth muscle cells impairs vascular homeostasis, leading to vaso-constriction and endothelial dysfunction, with subsequent leukocyte activation and adhesion, and increased platelet activity, adhesion and aggregation (Fig. 1B) 22. RBC lysis also releases arginase, which catabolizes arginine, the substrate for NO synthesis 24. Additionally, the oxidized hemoglobin (methemoglobin, Hb-Fe3+) releases free heme, the major erythrocyte-derived damage associated molecular pattern (DAMP) driving sterile inflammation in SCD 25. Cell-free heme into plasma generates ROS, propagating oxidative stress and endothelial activation, and stimulates the innate immune system through toll-like receptor 4 (TLR4) and inflammasome signaling 16,26,27.

The highly inflammatory environment in the vasculature of SCD patients is evidenced by high levels of proinflammatory cytokines in circulation, including tumor necrosis factor alpha (TNFα), interleukin-6 (IL-6), and interleukin-1 beta (IL-1β) 28,29, which promote and perpetuate endothelial and leukocyte activation. As discussed later, this proinflammatory milieu causes heterocellular adhesive interactions of sickle RBCs and activated leukocytes and platelets with the activated endothelium, which delay and obstruct the blood flow, promoting vascular occlusions (Fig. 1B). Ischemia-reperfusion episodes lead to acute tissue damage, and their recurrence drives progressive and irreversible organ damage, ultimately leading to multiple organ failure and, in some instances, death 16,30. Acute vaso-occlusive events accompanied by severe pain are the main cause of hospitalization in SCD patients 31 and often evolve to more severe complications, such as acute chest syndrome, a severe type of lung injury that represents the leading cause of death among SCD patients 32,33.

Hydroxyurea, also known as hydroxycarbamide, is the most widely used drug currently available to treat and prevent the acute complications of SCD. Hydroxyurea is a cytostatic drug that strongly induces an increase in fetal hemoglobin (HbF), which consequently dilutes the concentration of HbS into the RBCs, preventing HbS polymerization, RBC sickling, and the subsequent cascade of events that lead to vaso-occlusion 34,35. HbF is predominantly produced during fetal life, and is progressively replaced by HbA after birth, or by HbS in SCD patients 36,37. High levels of HbF in adult life of SCD patients ameliorate the clinical course by reducing the frequency of vaso-occlusive crisis, acute chest syndrome, blood transfusions, and hospitalization 3840. The beneficial effects of hydroxyurea, however, are not restricted to the induction of HbF, but also coincide with a reduction in leukocyte counts, particularly neutrophils, as supported by the observation that amelioration of clinical symptoms occurs even in the absence of raised HbF 38. Evidence from in vivo and in vitro studies point to hydroxyurea as a potent anti-inflammatory drug that promotes a decrease in neutrophil counts, prevents neutrophil activation and adhesion, and improves endothelial function 41,42. New drug therapies have been approved by the FDA within the last five years, including L-glutamine 43, an amino acid with anti-oxidant properties; crizanlizumab 44, a humanized antibody targeting P-selectin, which expressed by platelets and endothelial cells; and voxelotor 45, a modulator of hemoglobin-oxygen affinity. Besides targeting distinct pathways of SCD pathophysiology, they all directly or indirectly reduce leukocyte count, activation and adhesion, further supporting the prominent inflammatory aspect of SCD and the fundamental role of leukocytes to disease pathophysiology.

3. NEUTROPHILS IN SICKLE CELL DISEASE

Elevated leukocyte count is a feature of SCD 46,47 and a risk factor for disease severity 48,49. High baseline leukocyte counts have been associated with death at young age 50, increased susceptibility to acute chest syndrome 51, and high frequency of hemorrhagic stroke 52. Numerous clinical observations support the relevance of neutrophils to SCD pathophysiology. Evidence from G-CSF treatment used to mobilize hematopoietic stem cells in a SCD patient showed an increase in neutrophil count followed by severe painful vaso-occlusive crisis and acute chest syndrome, with resolution after stopping G-CSF and re-introducing hydroxyurea 53. Subsequent studies also reported acute multiorgan failure 54 and even death 55 after G-CSF administration to patients with SCD.

In vivo studies using transgenic murine models of SCD expressing human HbS revealed that neutrophils are crucial players in the vaso-occlusive process. Intravital microscopy of the microvasculature of the cremaster muscle in SCD mice demonstrated that neutrophils not only adhere to the activated endothelium, but additionally capture circulating sickle RBCs, thereby favoring cellular interactions that culminate in the obstruction of postcapillary venules 56,57. This may seem intuitive since neutrophils represent 50–70% of total circulating leukocytes in humans, while in mice they account for only 10–25% 58,59. The intravital imaging data revealed, however, that over 80% of total leukocytes participating in cellular interactions during vaso-occlusion in mice are neutrophils 57. Recent findings from in vivo lung imaging of pulmonary arterioles in SCD mice have shown the occlusion of pulmonary arterioles as a mechanism driven by neutrophil extracellular traps (NETs) and neutrophil-platelet aggregates 6062, supporting the involvement of neutrophils in causing vaso-occlusion of pulmonary vessels and in turn causing acute chest syndrome.

3.1. Central role of neutrophils in SCD-related vaso-occlusion

Vaso-occlusion in SCD is often initiated by inflammatory stimuli and erythrocyte-derived DAMPs released during hemolysis. Systemic vaso-occlusion preferentially occurs in small vessels, mainly postcapillary venules 63, while pulmonary arterioles are the primary site of occlusion in the lungs 60. Although RBC-endothelium interactions were initially described 64 and for many years considered the sole or main component of vascular occlusions in SCD, leukocytes were later identified as key players in triggering these events 65.

3.1.1. Neutrophils, RBCs and the endothelium

Neutrophil recruitment and interaction with the endothelium require adhesion molecules expressed by activated endothelial cells. The most important molecules for this step are the endothelial P- and E-selectins, which bind to glycosylated ligands on the surface of neutrophils (and other inflammatory leukocytes). P-selectin preferentially binds to P-selectin glycoprotein ligand-1 (PSGL-1), while E-selectin binds to E-selectin ligand-1 (ESL-1) and CD44 66. Such interactions are weak but favored by slowed blood flow during inflammation. They also activate the expression and activation of β2 integrins on neutrophils, most importantly lymphocyte function-associated antigen 1 (LFA-1, αLβ2, CD11a/CD18) and macrophage antigen 1 (Mac-1, αMβ2, CD11b/CD18), which bind to intercellular vascular adhesion molecules (ICAMs) expressed by endothelial cells and promote firmer interactions and crawling of rolling neutrophils with the vessel wall (Fig. 1C) 67.

Circulating endothelial cells from individuals with SCD are abnormally activated, with exacerbated expression of ICAM-1, vascular cell adhesion molecule-1 (VCAM-1), as well as E- and P-selectin as a result of the inflammatory milieu promoted by the disease 68,69. Consistently, in vitro culture of human umbilical vein endothelial cells (HUVECs) with TNFα, a potent inflammatory stimulus widely used to simulate SCD-related inflammation, enhances the expression of endothelial adhesion molecules and selectins 70,71. Cytokine-induced expression of endothelial adhesion molecules and selectins in vitro has shown to be repressed by NO, possibly through inhibition of NF-kB in endothelial cells 72, suggesting that reduced levels of NO in SCD patients exacerbates the inflammatory response. Soluble VCAM-1, ICAM-1 and E-selectin are elevated in patients with SCD and may function as important markers of endothelial dysfunction and disease severity, since they have been associated with pulmonary hypertension, liver disease, renal dysfunction and early mortality 73.

As indicated above, in vivo imaging of the microvasculature by intravital microscopy has become a fundamental tool for the understanding of the dynamics of neutrophils during inflammation. Intravital microscopy of the cremaster microcirculation in SCD mice lacking both P- and E-selectin showed a reduction in the rolling and adherence of leukocytes in postcapillary venules. These mice were also protected from vaso-occlusion induced by TNFα, compared with SCD mice with functional endothelial selectins 56. Adherent neutrophils, which are larger and more rigid than erythrocytes, potentially delay the blood flow by increasing the RBC transit time in circulation and favoring their sickling and entrapment in small vessels. Interestingly, adherent leukocytes have been observed to capture RBCs during vaso-occlusion, especially sickle-shaped RBCs, and the number of RBC interactions per adherent leukocyte was drastically reduced in the absence of P- and E-selectins 56. Further analysis of RBC-leukocyte adhesion in SCD mice showed that such interactions were mostly affected by E-selectin expression through upregulation of Mac-1 activity 74. E-selectin is expressed exclusively by endothelial cells and regulates the expression of Mac-1 by neutrophils (Fig. 2) 75. Mac-1 has been identified as the molecule responsible for the capture of adherent RBCs during vaso-occlusion in SCD mice (Fig. 1C), and adherent neutrophils from SCD mice display higher Mac-1 activity, which is notably reduced in the absence of E-selectin 74. Mac-1 is markedly upregulated also in neutrophils from SCD patients, compared with healthy individuals 76,77, and it has been demonstrated to mediate in vitro interactions of human neutrophils with endothelial cells 71 and matrix extracellular ligands (e.g. fibronectin) 78.

Figure 2. Effector mechanisms of neutrophils in sickle cell disease.

Figure 2.

Left: Neutrophil activation is characterized by increased αMβ2 integrin (Mac-1) activity, which is triggered by the contact of neutrophils with the activated endothelium and the consequent interaction between E-selectin and its ligand ESL-1. Mac-1 can also be activated by heme through toll-like receptor 4 (TLR4) signaling. Activation of TLR4 by heme leads to reactive oxygen species (ROS) generation and inflammasome formation in neutrophils. Further stimulus for neutrophil activation is provided by extracellular ROS and inflammatory mediators released by activated endothelial cells, platelets and leukocytes. Right: Damage associated molecular patterns (DAMPs), e.g. heme, ROS, and inflammatory cytokines, promote neutrophil extracellular traps (NETs) formation through ROS generation and gasdermin-D (GSDMD) activation. NETs are characterized by DNA, histones, and granule contents, such as myeloperoxidase (MPO) and elastase (NE). Entrapment of platelets by NETs promote thrombi formation and aggregation between platelets and neutrophils.

3.1.2. Neutrophil-platelet aggregates

While RBCs and neutrophils are the main drivers of systemic vaso-occlusion, the involvement of platelets in adhesive interactions with the endothelium has been described with great relevance for pulmonary vaso-occlusion. Platelets from SCD patients circulate in an activate state, with elevated expression of P-selectin 77. P-selectin expressed by platelets can bind to PSGL-1 expressed by adherent neutrophils 79 and interactions may additionally occur between Mac-1 on neutrophils and glycoprotein Ibα (GPIbα) on platelets (Fig. 1C) 74,80,81. Platelet activation leads to the release of granule contents, which is rich in inflammatory mediators, and microvesicles that activate endothelial cells and promote leukocyte recruitment and activation (Fig. 2). Platelets from SCD individuals are capable of promoting endothelial activation in vitro 82 and are enriched with IL-1β, an inflammasome-derived cytokine released through platelet extracellular vesicles and shown to further activate neutrophils, endothelial cells and platelets 61.

Neutrophil-platelet aggregates are abundant in the blood of SCD patients 77,83 and have been identified occluding pulmonary arterioles in SCD mice challenged with intravenous bacterial lipopolysaccharide (LPS), a TLR4 ligand, or Oxy-Hb, a precursor of heme 60,62. TLR4 activation by heme is known to prime inflammasome activation in leukocytes and to induce platelet NLRP3-inflammasome, with consequent generation of neutrophil-platelet aggregates (Fig. 2) 61. Heme-induced TLR4 signaling in endothelial cells leads to the activation of the NFkB pathway, thereby promoting endothelial activation and vaso-occlusion in SCD mice 84. Activation of NFkB by heme has been also reported in human neutrophils in vitro and resulted in an augmented adhesiveness of neutrophils to fibronectin and endothelial ICAM-1, increased ROS generation, and enhanced Mac-1 activity 78.

3.1.3. Neutrophil extracellular traps (NETs)

Another pathway of SCD vaso-occlusion lies on the potential of heme to trigger one of the most intriguing effector mechanisms used by neutrophils exposed to pathogens or sterile stimuli – the generation of NETs 85. NETs are defined as extracellular structures of DNA coated with histones and granule-derived proteins, such as neutrophil elastase (NE), cathepsin G, and myeloperoxidase (MPO) 85,86.

One of the mechanisms of NET generation is through induction of nuclear decondensation and release of cellular contents by gasdermin-D (GDSMD) (Fig. 2) 87. Briefly, activation of neutrophils triggers ROS generation by NADPH oxidase, leading to the activation of protein-arginine deiminase 4 (PAD4), which mediates histone hypercitrullination and induces chromatin unfolding 88. ROS-induced activity and release of MPO and NE from azurophilic granules promote degradation of cytoplasmic actin filaments and contribute to chromatin decondensation. Finally, the formation of pores in the plasma membrane and in the azurophilic granules by GSDMD leads to cell lysis and NET release 87,89.

NETs play an important role in controlling infections; however, they can be harmful to the endothelium and induce further inflammatory responses 90. Additionally, activated platelets or neutrophils-platelet aggregates can be entrapped into circulating NETs and promote thrombi formation 91. Growing evidence suggest that NETs favor the aggregation of neutrophils and platelets in the pulmonary arterioles, leading to pulmonary vaso-occlusion and acute lung injury 27,62. NETs and soluble NET components have been observed in the lungs and peripheral blood, respectively, of TNFα-stimulated SCD mice. In non-SCD control mice, in which hemolysis is absent, TNFα only induced NET formation when administered with heme. Consistently, treatment of SCD mice with hemopexin, a natural scavenger of heme, abrogated NET formation 27. Recent and intriguing findings showed that NETs found in the lungs of SCD mice were not locally originated, but instead originated remotely and traveled intravascularly from the liver to the lungs, where they finally promoted vaso-occlusion 62.

3.2. Neutrophils as a therapeutic target in SCD

The understanding of vaso-occlusion as a multicellular phenomenon, driven by different inflammatory pathways and blood cell types, expands the range of targets for the development of new therapeutic approaches to treat and prevent SCD complications. Such strategies may focus on the early instigating event by preventing HbS polymerization and subsequent events leading to vaso-occlusion, by inhibiting downstream pathways that directly promote vaso-occlusion or, indirectly, by attenuating the vicious cycle of HbS polymerization, hemolysis and inflammation. The identification of neutrophils at the initiation and propagation stages of vaso-occlusions has placed these cells in the spotlight.

Targeting pro- and anti-inflammatory pathways is an approach that has been tested in SCD mouse models and shown favorable outcomes. Neutralization of inflammasome cytokines by specific antibodies, such as anti-IL-1β and anti-IL-18, ameliorated endothelial activation, inflammation, and vaso-occlusion triggered by hypoxia or TNFα in SCD mice 92,93. Likewise, treatment with anti-inflammatory cytokines, for example transforming growth factor beta 1 (TGF-β1), a major immune regulator, protected SCD mice from TNFα-induced vaso-occlusion and reduced the adhesion of human neutrophils in vitro 70. Conversely, blockage of TGF-β1 signaling by an anti-TGFβ antibody aggravated vaso-occlusion in these mice 70. Although promising, modulation of cytokines may have undesirable effects in several pathways unrelated to vaso-occlusion due to their pleiotropic action. In this scenario, more specific approaches appear to be a better and safer choice.

Crizanlizumab, a humanized P-selectin antibody, is a prominent example of therapy with direct action in the adhesive interactions between blood cells and the endothelium. A clinical trial reported a 45% reduction in the frequency of vaso-occlusive crisis experienced by SCD patients receiving anti-P-selectin therapy compared with SCD patients receiving placebo 44. Likewise, P-selectin inhibition ameliorated lung vaso-occlusion in SCD mice by preventing the formation of large neutrophil-platelet aggregates in the pulmonary arterioles 60. The partial benefits achieved with anti-P-selectin treatment in both human and murine studies support the concept of multiple pathways involved in SCD vaso-occlusion. In line with these findings, a recent study reported a P-selectin-independent mechanism of lung vaso-occlusion in SCD mice. They showed that GSDMD, an effector protein of inflammasome signaling and NETs release, was overexpressed in SCD human neutrophils. Combination of both strategies, i.e., P-selectin blockage and GSDMD inhibition, potentialized the protection offered by the isolated approaches and resulted in complete abolition of lung vaso-occlusion in SCD mice 62.

3.3. Neutrophil heterogeneity and circadian considerations

A final (and poorly explored) question when considering how neutrophils trigger vascular disease in SCD is whether different populations exist that aggravate, or conversely alleviate, inflammatory disease in these patients. Indeed, although neutrophils were classically thought to be a homogeneous population, growing evidence of phenotypic and functional heterogeneity now exists that adds another layer of complexity to the participation of these cells in the pathophysiology of SCD and other inflammatory conditions. The concept of neutrophil heterogeneity arises from observations of distinct stages of maturation, function, activation state, and expression of surface proteins found in the total neutrophil population derived from the bone marrow, blood or other tissues 94. Whether such differences reflect a transient state, or the existence of pre-defined subpopulations is still a matter of discussion that are relevant when considering therapeutic avenues for neutrophil-driven diseases.

An interesting point of discussion is the diversity of neutrophils acquired while in the circulation (Fig. 3). These phenotypic variations of murine and human circulating neutrophils have been shown to follow circadian oscillations 95,96. The number of circulating neutrophils is determined by the release of “younger” (also referred to as fresh) neutrophils from the bone marrow and by the clearance of senescent neutrophils from the blood by the bone marrow and other organs such as spleen and liver. Physiological clearance of neutrophils occurs at the end of the animal’s resting period (daytime for mice; night-time for humans), when they migrate back to the bone marrow or other tissues, in a chemokine-dependent manner 8,95. The retention and migration of neutrophils in the bone marrow compartment is regulated by CXCL12, a chemokine produced by stromal cells within this organ that binds to its receptor CXCR4 on the surface of neutrophils. The expression of CXCR4 is low in fresh neutrophils but increases as they age in the circulation and instructs their homing back to the bone marrow 95,97. In contrast, freshly released neutrophils express high levels of L-selectin (CD62L), an adhesion molecule that regulates neutrophil recruitment to peripheral tissues and loss of L-selectin by neutrophils occurs gradually as they transit in circulation 95,98. The critical question is whether this phenomenon of circadian aging may underlie some of the pro-inflammatory activities of neutrophils in the context of SCD.

Figure 3. Sources of neutrophil heterogeneity in sickle cell disease.

Figure 3.

The release of freshly produced neutrophils from the bone marrow occurs at the beginning of the rest period, which is marked by the onset of light in mice and darkness in humans. As these neutrophils circulate in the blood, they downregulate the expression of L-selectin and upregulate the expression of chemokine receptor 4 (CXCR4) over time. Neutrophil aging is also accompanied by increased expression and activation of αMβ2 integrin (Mac-1). Clearance of aged neutrophils takes place in the bone marrow or other organs, such as liver and spleen, at the end of the resting period. Accumulation of circulating aged neutrophils in SCD mice is a result of the inflammatory milieu promoted by the disease and a response to signals generated by the gut microbiota. Microbial signals are under influence of external factors, such as antibiotics, or endogenous factors, including signals from the nervous system.

An augmented number of neutrophils expressing an aged phenotype, defined by low expression of CD62L and high expression of CXCR4, has been observed in the blood of SCD mice 99. These neutrophils upregulate Mac-1 activity, TLRs signaling, and NETs formation, and have been associated with acute vaso-occlusion and long-term organ damage. The accumulation of aged-like neutrophils in the blood of SCD mice was prevented by chronic treatment with broad-spectrum antibiotics, suggesting that signals from the gut microbiota controls the aging process of neutrophils (Fig. 3) 99. One possible mechanism connecting aging with the microbiome is the crosstalk between gut microbiota and the nervous system. Expansion of aged neutrophils has been observed in the blood of SCD mice subjected to a restraint protocol of psychological stress and resulted in worsening of inflammation and vaso-occlusion, which were prevented by gut microbiota depletion 100. Not only the intestinal barrier is compromised in SCD, but intestinal dysbiosis has been reported in both SCD patients and mice 101,102. Since the gut microbiota is essential for the regulation of the innate immune system, intrinsic or acquired perturbations in the composition of the gut microbiota can potentially affect circulating neutrophils in SCD.

The frequency of circulating neutrophils with high expression of CXCR4 is reportedly elevated in the blood of SCD patients 103. These cells demonstrate higher activity of Mac-1 and propensity to form neutrophil-platelet aggregates, with a greater number of platelets bound per neutrophil, when compared to neutrophils expressing low CXCR4. Platelets have been demonstrated to induce the expression of CXCR4 on neutrophils in vitro, through the release of serotonin, which is elevated in the plasma of SCD individuals. Incubation of healthy neutrophils with purified serotonin or with SCD plasma, in the absence of platelets, also induced the expression CXCR4. Corroborating the involvement of serotonin in the phenotypic modulation of neutrophils in SCD, the use of a non-selective inhibitor of serotine receptors prevented neutrophils from expressing CXCR4 in the presence of SCD plasma 103. Thus, an interplay between microbial or platelet derived metabolites may drive inflammatory episodes in SCD patients by modulating the activity of neutrophils.

The microenvironment in different tissues or under different stimuli can induce the acquisition of different functions by neutrophils and may underlie the acquisition of distinct phenotypes that differ in the expression of important activation markers 94. Early intravital studies reported that less than 20% of total adherent leukocytes capture sickle RBCs during TNFα-induced vaso-occlusion in SCD mice 56, an observation that suggests diversity in the activity of these cells during intravascular inflammation. Variations in the expression and activation of the RBC receptor Mac-1 by neutrophils in SCD, for example, has also been associated with changes in their physical density, which may in turn be related to maturation or degranulation 104. The concept of different density subsets of neutrophils derives from observations that during isolation of neutrophils by a Ficoll density gradient, a small fraction of low-density-neutrophils (LDN) is found in the mononuclear cell layer. These LDN represent a heterogeneous population composed by both mature and immature cells and have been extensively studied in the context of cancer due to their pro-tumoral properties 105,106. An elevated number of LDN is found in the blood of SCD patients and SCD mice and these cells feature higher expression and activity of Mac-1 and increased adhesion in vitro 104, suggesting that LDN may be potential promoters of vaso-occlusion in SCD.

While not discussed here, other sources of neutrophil diversity emanating from the unique conditions present in these patients may dictate the pathophysiology of SCD, not only inside vessels but also in peripheral organs. For example, the observation that neutrophils control angiogenesis in lungs and intestine during early life or upon genotoxic injury 107 suggests that this or other functions may also be affected in SCD patients, and this may be an attractive area of exploration in the future.

4. CONCLUDING REMARKS

The protagonist role of neutrophils in SCD pathology is now unquestionable. These cells promptly respond to inflammatory stimuli triggered by infection, hemolysis-derived DAMPs, endothelium-released mediators, and numerous factors secreted by other immune cells and injured tissues. Upregulation of adhesion molecules by activated neutrophils favors their cell-cell interactions and place these cells as central effectors and perpetuators of SCD-related vaso-occlusion. The wide range of functions displayed by neutrophils in SCD is mirrored by the identification of distinct phenotypes even in healthy individuals, which may in turn denote stages of a highly dynamic response to different stimuli and adaptation to different microenvironments 94. Because neutrophils are cells with a very short lifetime and need to be produced constantly, the observations made in the context of SCD suggests that, in addition to the activation of neutrophils in the bloodstream discussed above, alterations in the granulopoietic process may underlie the presence of abnormally primed neutrophils, and this in turn opens a very attractive area of investigation and intervention.

SCD also provides a highly dynamic scenario where a broad spectrum of stimuli may affect neutrophils in many particular ways. The use of single-cell technologies may now offer a more unbiased approach to study the rich and complex biology of neutrophils in this context, including single cell transcriptomics and multiparametric cytometric assays that may allow us to define alterations of the SCD neutrophil baseline, or during inflammatory responses and vaso-occlusive episodes. Further, behavioral screening of single adherent neutrophils in the cremaster microvasculature of mice subjected to TNFα-induced vaso-occlusion has been recently described and emerges as a potentially powerful tool to elucidate the dynamics and functional states of neutrophils SCD. Indeed, the use of over 70 morpho-kinetic parameters in intravascular adherent neutrophils within the inflamed vasculature of otherwise healthy mice revealed at least three well-defined behaviors, which differed in shape, size, motility or proximity to the vessel wall. Most notably, certain behavioral patterns associated with pathological inflammation thereby allowing the identification of disease-driving genes 108. These new technologies and other orthogonal approaches to investigating neutrophils may uncloud our understanding of how SCD modifies the inflammatory properties of neutrophils, and these in turn drive disease.

In sum, as we progressively increase our understanding of the biology of neutrophils and their alterations in SCD patients and animal models, we may soon develop strategies to effectively target the inflammatory complications seen in these patients, without compromising tissue homeostasis or anti-microbial defense. One step in this direction is the discovery that neutrophils are not phenotypically and functionally rigid but are instead highly dynamic and adaptable to different environments. We hope that harnessing such new concepts may provide real therapeutic options for millions of patients in the not-too-distant future.

Acknowledgements

We thank Keisuke Ito for continued support. We also thank our late mentor Paul Frenette, who instilled in us the passion for biomedical research. L.S.T. is supported by the National Institutes of Health (R01HL069438) and the Paul S. Frenette Scholar Awards Program of the Ruth L. and David S. Gottesman Institute for Stem Cell Research and Regenerative Medicine. A.H. is supported by RTI2018-095497- B-I00 from MCIN, HR17_00527 from Fundación La Caixa, the Transatlantic Network of Excellence (TNE-18CVD04) from the Leducq Foundation, and FET-OPEN (no. 861878) from the European Commission. The CNIC is supported by the MCIN and the Pro CNIC Foundation and is a Severo Ochoa Center of Excellence (CEX2020-001041-S).

References

  • 1.Kraus RF, Gruber MA. Neutrophils-From Bone Marrow to First-Line Defense of the Innate Immune System. Front Immunol 2021;12:767175. doi: 10.3389/fimmu.2021.767175 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Hidalgo A, Chilvers ER, Summers C, Koenderman L. The Neutrophil Life Cycle. Trends Immunol Jul 2019;40(7):584–597. doi: 10.1016/j.it.2019.04.013 [DOI] [PubMed] [Google Scholar]
  • 3.Neutrophils Borregaard N., from marrow to microbes. Immunity Nov 24 2010;33(5):657–70. doi: 10.1016/j.immuni.2010.11.011 [DOI] [PubMed] [Google Scholar]
  • 4.Richards MK, Liu F, Iwasaki H, Akashi K, Link DC. Pivotal role of granulocyte colony-stimulating factor in the development of progenitors in the common myeloid pathway. Blood Nov 15 2003;102(10):3562–8. doi: 10.1182/blood-2003-02-0593 [DOI] [PubMed] [Google Scholar]
  • 5.Lord BI, Bronchud MH, Owens S, et al. The kinetics of human granulopoiesis following treatment with granulocyte colony-stimulating factor in vivo. Proc Natl Acad Sci U S A Dec 1989;86(23):9499–503. doi: 10.1073/pnas.86.23.9499 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Lieschke GJ, Grail D, Hodgson G, et al. Mice lacking granulocyte colony-stimulating factor have chronic neutropenia, granulocyte and macrophage progenitor cell deficiency, and impaired neutrophil mobilization. Blood Sep 15 1994;84(6):1737–46. [PubMed] [Google Scholar]
  • 7.Liew PX, Kubes P. The Neutrophil’s Role During Health and Disease. Physiol Rev Apr 1 2019;99(2):1223–1248. doi: 10.1152/physrev.00012.2018 [DOI] [PubMed] [Google Scholar]
  • 8.Casanova-Acebes M, Nicolas-Avila JA, Li JL, et al. Neutrophils instruct homeostatic and pathological states in naive tissues. J Exp Med Nov 5 2018;215(11):2778–2795. doi: 10.1084/jem.20181468 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.McCracken JM, Allen LA. Regulation of human neutrophil apoptosis and lifespan in health and disease. J Cell Death 2014;7:15–23. doi: 10.4137/JCD.S11038 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Serhan CN, Gupta SK, Perretti M, et al. The Atlas of Inflammation Resolution (AIR). Mol Aspects Med Aug 2020;74:100894. doi: 10.1016/j.mam.2020.100894 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Herrero-Cervera A, Soehnlein O, Kenne E. Neutrophils in chronic inflammatory diseases. Cell Mol Immunol Feb 2022;19(2):177–191. doi: 10.1038/s41423-021-00832-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Quail DF, Amulic B, Aziz M, et al. Neutrophil phenotypes and functions in cancer: A consensus statement. J Exp Med Jun 6 2022;219(6)doi: 10.1084/jem.20220011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Feletou M The Endothelium: Part 1: Multiple Functions of the Endothelial Cells-Focus on Endothelium-Derived Vasoactive Mediators. 2011. Integrated Systems Physiology: from Molecule to Function to Disease [PubMed] [Google Scholar]
  • 14.Pober JS, Sessa WC. Evolving functions of endothelial cells in inflammation. Nat Rev Immunol Oct 2007;7(10):803–15. doi: 10.1038/nri2171 [DOI] [PubMed] [Google Scholar]
  • 15.Torres LS, Asada N, Weiss MJ, et al. Recent advances in “sickle and niche” research - Tribute to Dr. Paul S Frenette. Stem Cell Reports Jul 12 2022;17(7):1509–1535. doi: 10.1016/j.stemcr.2022.06.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Sundd P, Gladwin MT, Novelli EM. Pathophysiology of Sickle Cell Disease. Annu Rev Pathol Jan 24 2019;14:263–292. doi: 10.1146/annurev-pathmechdis-012418-012838 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Zhang D, Xu C, Manwani D, Frenette PS. Neutrophils, platelets, and inflammatory pathways at the nexus of sickle cell disease pathophysiology. Blood Feb 18 2016;127(7):801–9. doi: 10.1182/blood-2015-09-618538 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Ingram VM. Gene mutations in human haemoglobin: the chemical difference between normal and sickle cell haemoglobin. Nature Aug 17 1957;180(4581):326–8. doi: 10.1038/180326a0 [DOI] [PubMed] [Google Scholar]
  • 19.Bunn HF. Pathogenesis and treatment of sickle cell disease. N Engl J Med Sep 11 1997;337(11):762–9. doi: 10.1056/NEJM199709113371107 [DOI] [PubMed] [Google Scholar]
  • 20.Bensinger TA, Gillette PN. Hemolysis in sickle cell disease. Arch Intern Med Apr 1974;133(4):624–31. [PubMed] [Google Scholar]
  • 21.Hebbel RP. Reconstructing sickle cell disease: a data-based analysis of the “hyperhemolysis paradigm” for pulmonary hypertension from the perspective of evidence-based medicine. Am J Hematol Feb 2011;86(2):123–54. doi: 10.1002/ajh.21952 [DOI] [PubMed] [Google Scholar]
  • 22.Reiter CD, Wang X, Tanus-Santos JE, et al. Cell-free hemoglobin limits nitric oxide bioavailability in sickle-cell disease. Nat Med Dec 2002;8(12):1383–9. doi: 10.1038/nm1202-799 [DOI] [PubMed] [Google Scholar]
  • 23.Doherty DH, Doyle MP, Curry SR, et al. Rate of reaction with nitric oxide determines the hypertensive effect of cell-free hemoglobin. Nat Biotechnol Jul 1998;16(7):672–6. doi: 10.1038/nbt0798-672 [DOI] [PubMed] [Google Scholar]
  • 24.Morris CR, Kato GJ, Poljakovic M, et al. Dysregulated arginine metabolism, hemolysis-associated pulmonary hypertension, and mortality in sickle cell disease. JAMA Jul 6 2005;294(1):81–90. doi: 10.1001/jama.294.1.81 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Mendonca R, Silveira AA, Conran N. Red cell DAMPs and inflammation. Inflamm Res Sep 2016;65(9):665–78. doi: 10.1007/s00011-016-0955-9 [DOI] [PubMed] [Google Scholar]
  • 26.Kato GJ, Steinberg MH, Gladwin MT. Intravascular hemolysis and the pathophysiology of sickle cell disease. J Clin Invest Mar 1 2017;127(3):750–760. doi: 10.1172/JCI89741 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Chen G, Zhang D, Fuchs TA, Manwani D, Wagner DD, Frenette PS. Heme-induced neutrophil extracellular traps contribute to the pathogenesis of sickle cell disease. Blood Jun 12 2014;123(24):3818–27. doi: 10.1182/blood-2013-10-529982 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Lanaro C, Franco-Penteado CF, Albuqueque DM, Saad ST, Conran N, Costa FF. Altered levels of cytokines and inflammatory mediators in plasma and leukocytes of sickle cell anemia patients and effects of hydroxyurea therapy. J Leukoc Biol Feb 2009;85(2):235–42. doi: 10.1189/jlb.0708445 [DOI] [PubMed] [Google Scholar]
  • 29.Torres LS, Okumura JV, Silva DG, et al. Inflammation in Sickle Cell Disease: Differential and Down-Expressed Plasma Levels of Annexin A1 Protein. PLoS One 2016;11(11):e0165833. doi: 10.1371/journal.pone.0165833 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Kato GJ, Piel FB, Reid CD, et al. Sickle cell disease. Nat Rev Dis Primers Mar 15 2018;4:18010. doi: 10.1038/nrdp.2018.10 [DOI] [PubMed] [Google Scholar]
  • 31.Ballas SK, Lusardi M. Hospital readmission for adult acute sickle cell painful episodes: frequency, etiology, and prognostic significance. Am J Hematol May 2005;79(1):17–25. doi: 10.1002/ajh.20336 [DOI] [PubMed] [Google Scholar]
  • 32.Ballas SK, Gupta K, Adams-Graves P. Sickle cell pain: a critical reappraisal. Blood Nov 1 2012;120(18):3647–56. doi: 10.1182/blood-2012-04-383430 [DOI] [PubMed] [Google Scholar]
  • 33.Serjeant GR. The natural history of sickle cell disease. Cold Spring Harb Perspect Med Oct 1 2013;3(10):a011783. doi: 10.1101/cshperspect.a011783 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Charache S, Dover GJ, Moore RD, et al. Hydroxyurea: effects on hemoglobin F production in patients with sickle cell anemia. Blood May 15 1992;79(10):2555–65. [PubMed] [Google Scholar]
  • 35.McGann PT, Ware RE. Hydroxyurea therapy for sickle cell anemia. Expert Opin Drug Saf 2015;14(11):1749–58. doi: 10.1517/14740338.2015.1088827 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Stamatoyannopoulos G Control of globin gene expression during development and erythroid differentiation. Exp Hematol Mar 2005;33(3):259–71. doi: 10.1016/j.exphem.2004.11.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Lettre G, Bauer DE. Fetal haemoglobin in sickle-cell disease: from genetic epidemiology to new therapeutic strategies. Lancet Jun 18 2016;387(10037):2554–64. doi: 10.1016/S0140-6736(15)01341–0 [DOI] [PubMed] [Google Scholar]
  • 38.Charache S, Terrin ML, Moore RD, et al. Effect of hydroxyurea on the frequency of painful crises in sickle cell anemia. Investigators of the Multicenter Study of Hydroxyurea in Sickle Cell Anemia. N Engl J Med May 18 1995;332(20):1317–22. doi: 10.1056/NEJM199505183322001 [DOI] [PubMed] [Google Scholar]
  • 39.Steinberg MH. Fetal hemoglobin in sickle cell anemia. Blood Nov 19 2020;136(21):2392–2400. doi: 10.1182/blood.2020007645 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Noguchi CT, Rodgers GP, Serjeant G, Schechter AN. Levels of fetal hemoglobin necessary for treatment of sickle cell disease. N Engl J Med Jan 14 1988;318(2):96–9. doi: 10.1056/NEJM198801143180207 [DOI] [PubMed] [Google Scholar]
  • 41.Almeida CB, Scheiermann C, Jang JE, et al. Hydroxyurea and a cGMP-amplifying agent have immediate benefits on acute vaso-occlusive events in sickle cell disease mice. Blood Oct 4 2012;120(14):2879–88. doi: 10.1182/blood-2012-02-409524 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Ferreira WA, Jr., Chweih H, Lanaro C, et al. Beneficial Effects of Soluble Guanylyl Cyclase Stimulation and Activation in Sickle Cell Disease Are Amplified by Hydroxyurea: In Vitro and In Vivo Studies. J Pharmacol Exp Ther Sep 2020;374(3):469–478. doi: 10.1124/jpet.119.264606 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Niihara Y, Smith WR, Stark CW. A Phase 3 Trial of l-Glutamine in Sickle Cell Disease. N Engl J Med Nov 8 2018;379(19):1880. doi: 10.1056/NEJMc1811050 [DOI] [PubMed] [Google Scholar]
  • 44.Ataga KI, Kutlar A, Kanter J, et al. Crizanlizumab for the Prevention of Pain Crises in Sickle Cell Disease. N Engl J Med Feb 2 2017;376(5):429–439. doi: 10.1056/NEJMoa1611770 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Vichinsky E, Hoppe CC, Ataga KI, et al. A Phase 3 Randomized Trial of Voxelotor in Sickle Cell Disease. N Engl J Med Aug 8 2019;381(6):509–519. doi: 10.1056/NEJMoa1903212 [DOI] [PubMed] [Google Scholar]
  • 46.West MS, Wethers D, Smith J, Steinberg M. Laboratory profile of sickle cell disease: a cross-sectional analysis. The Cooperative Study of Sickle Cell Disease. J Clin Epidemiol Aug 1992;45(8):893–909. doi: 10.1016/0895-4356(92)90073-v [DOI] [PubMed] [Google Scholar]
  • 47.Platt OS. Sickle cell anemia as an inflammatory disease. J Clin Invest Aug 2000;106(3):337–8. doi: 10.1172/JCI10726 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Anyaegbu CC, Okpala IE, Akren’Ova YA, Salimonu LS. Peripheral blood neutrophil count and candidacidal activity correlate with the clinical severity of sickle cell anaemia (SCA). Eur J Haematol Apr 1998;60(4):267–8. doi: 10.1111/j.1600-0609.1998.tb01036.x [DOI] [PubMed] [Google Scholar]
  • 49.Miller ST, Sleeper LA, Pegelow CH, et al. Prediction of adverse outcomes in children with sickle cell disease. N Engl J Med Jan 13 2000;342(2):83–9. doi: 10.1056/NEJM200001133420203 [DOI] [PubMed] [Google Scholar]
  • 50.Platt OS, Brambilla DJ, Rosse WF, et al. Mortality in sickle cell disease. Life expectancy and risk factors for early death. N Engl J Med Jun 9 1994;330(23):1639–44. doi: 10.1056/NEJM199406093302303 [DOI] [PubMed] [Google Scholar]
  • 51.Castro O, Brambilla DJ, Thorington B, et al. The acute chest syndrome in sickle cell disease: incidence and risk factors. The Cooperative Study of Sickle Cell Disease. Blood Jul 15 1994;84(2):643–9. [PubMed] [Google Scholar]
  • 52.Ohene-Frempong K, Weiner SJ, Sleeper LA, et al. Cerebrovascular accidents in sickle cell disease: rates and risk factors. Blood Jan 1 1998;91(1):288–94. [PubMed] [Google Scholar]
  • 53.Abboud M, Laver J, Blau CA. Granulocytosis causing sickle-cell crisis. Lancet Mar 28 1998;351(9107):959. doi: 10.1016/S0140-6736(05)60614-9 [DOI] [PubMed] [Google Scholar]
  • 54.Grigg AP. Granulocyte colony-stimulating factor-induced sickle cell crisis and multiorgan dysfunction in a patient with compound heterozygous sickle cell/beta+ thalassemia. Blood Jun 15 2001;97(12):3998–9. doi: 10.1182/blood.v97.12.3998 [DOI] [PubMed] [Google Scholar]
  • 55.Adler BK, Salzman DE, Carabasi MH, Vaughan WP, Reddy VV, Prchal JT. Fatal sickle cell crisis after granulocyte colony-stimulating factor administration. Blood May 15 2001;97(10):3313–4. doi: 10.1182/blood.v97.10.3313 [DOI] [PubMed] [Google Scholar]
  • 56.Turhan A, Weiss LA, Mohandas N, Coller BS, Frenette PS. Primary role for adherent leukocytes in sickle cell vascular occlusion: a new paradigm. Proc Natl Acad Sci U S A Mar 5 2002;99(5):3047–51. doi: 10.1073/pnas.052522799 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Chiang EY, Hidalgo A, Chang J, Frenette PS. Imaging receptor microdomains on leukocyte subsets in live mice. Nat Methods Mar 2007;4(3):219–22. doi: 10.1038/nmeth1018 [DOI] [PubMed] [Google Scholar]
  • 58.Doeing DC, Borowicz JL, Crockett ET. Gender dimorphism in differential peripheral blood leukocyte counts in mice using cardiac, tail, foot, and saphenous vein puncture methods. BMC Clin Pathol Sep 12 2003;3(1):3. doi: 10.1186/1472-6890-3-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Mestas J, Hughes CC. Of mice and not men: differences between mouse and human immunology. J Immunol Mar 1 2004;172(5):2731–8. doi: 10.4049/jimmunol.172.5.2731 [DOI] [PubMed] [Google Scholar]
  • 60.Bennewitz MF, Jimenez MA, Vats R, et al. Lung vaso-occlusion in sickle cell disease mediated by arteriolar neutrophil-platelet microemboli. JCI Insight Jan 12 2017;2(1):e89761. doi: 10.1172/jci.insight.89761 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Vats R, Brzoska T, Bennewitz MF, et al. Platelet Extracellular Vesicles Drive Inflammasome-IL-1beta-Dependent Lung Injury in Sickle Cell Disease. Am J Respir Crit Care Med Jan 1 2020;201(1):33–46. doi: 10.1164/rccm.201807-1370OC [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Vats R, Kaminski TW, Brzoska T, et al. Liver to lung microembolic NETs promote Gasdermin-D-dependent inflammatory lung injury in Sickle Cell Disease. Blood Jun 23 2022;doi: 10.1182/blood.2021014552 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Kaul DK, Finnegan E, Barabino GA. Sickle red cell-endothelium interactions. Microcirculation Jan 2009;16(1):97–111. doi: 10.1080/10739680802279394 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Hebbel RP, Boogaerts MA, Koresawa S, Jacob HS, Eaton JW, Steinberg MH. Erytrocyte adherence to endothelium as a determinant of vasocclusive severity in sickle cell disease. Trans Assoc Am Physicians 1980;93:94–9. [PubMed] [Google Scholar]
  • 65.Frenette PS. Sickle cell vaso-occlusion: multistep and multicellular paradigm. Curr Opin Hematol Mar 2002;9(2):101–6. doi: 10.1097/00062752-200203000-00003 [DOI] [PubMed] [Google Scholar]
  • 66.Hidalgo A, Peired AJ, Wild M, Vestweber D, Frenette PS. Complete identification of E-selectin ligands on neutrophils reveals distinct functions of PSGL-1, ESL-1, and CD44. Immunity Apr 2007;26(4):477–489. doi: 10.1016/j.immuni.2007.03.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Phillipson M, Heit B, Colarusso P, Liu L, Ballantyne CM, Kubes P. Intraluminal crawling of neutrophils to emigration sites: a molecularly distinct process from adhesion in the recruitment cascade. J Exp Med Nov 27 2006;203(12):2569–75. doi: 10.1084/jem.20060925 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Solovey A, Lin Y, Browne P, Choong S, Wayner E, Hebbel RP. Circulating activated endothelial cells in sickle cell anemia. N Engl J Med Nov 27 1997;337(22):1584–90. doi: 10.1056/NEJM199711273372203 [DOI] [PubMed] [Google Scholar]
  • 69.Hebbel RP, Osarogiagbon R, Kaul D. The endothelial biology of sickle cell disease: inflammation and a chronic vasculopathy. Microcirculation Mar 2004;11(2):129–51. [PubMed] [Google Scholar]
  • 70.Torres LS, Chweih H, Fabris FCZ, et al. TGF-beta1 Reduces Neutrophil Adhesion and Prevents Acute Vaso-Occlusive Processes in Sickle Cell Disease Mice. Cells Apr 2 2022;11(7)doi: 10.3390/cells11071200 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Canalli AA, Proenca RF, Franco-Penteado CF, et al. Participation of Mac-1, LFA-1 and VLA-4 integrins in the in vitro adhesion of sickle cell disease neutrophils to endothelial layers, and reversal of adhesion by simvastatin. Haematologica Apr 2011;96(4):526–33. doi: 10.3324/haematol.2010.032912 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.De Caterina R, Libby P, Peng HB, et al. Nitric oxide decreases cytokine-induced endothelial activation. Nitric oxide selectively reduces endothelial expression of adhesion molecules and proinflammatory cytokines. J Clin Invest Jul 1995;96(1):60–8. doi: 10.1172/JCI118074 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Kato GJ, Martyr S, Blackwelder WC, et al. Levels of soluble endothelium-derived adhesion molecules in patients with sickle cell disease are associated with pulmonary hypertension, organ dysfunction, and mortality. Br J Haematol Sep 2005;130(6):943–53. doi: 10.1111/j.1365-2141.2005.05701.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Hidalgo A, Chang J, Jang JE, Peired AJ, Chiang EY, Frenette PS. Heterotypic interactions enabled by polarized neutrophil microdomains mediate thromboinflammatory injury. Nat Med Apr 2009;15(4):384–91. doi: 10.1038/nm.1939 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Lo SK, Lee S, Ramos RA, et al. Endothelial-leukocyte adhesion molecule 1 stimulates the adhesive activity of leukocyte integrin CR3 (CD11b/CD18, Mac-1, alpha m beta 2) on human neutrophils. J Exp Med Jun 1 1991;173(6):1493–500. doi: 10.1084/jem.173.6.1493 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Lum AF, Wun T, Staunton D, Simon SI. Inflammatory potential of neutrophils detected in sickle cell disease. Am J Hematol Jun 2004;76(2):126–33. doi: 10.1002/ajh.20059 [DOI] [PubMed] [Google Scholar]
  • 77.Polanowska-Grabowska R, Wallace K, Field JJ, et al. P-selectin-mediated platelet-neutrophil aggregate formation activates neutrophils in mouse and human sickle cell disease. Arterioscler Thromb Vasc Biol Dec 2010;30(12):2392–9. doi: 10.1161/ATVBAHA.110.211615 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Miguel LI, Leonardo FC, Torres LS, et al. Heme induces significant neutrophil adhesion in vitro via an NFkappaB and reactive oxygen species-dependent pathway. Mol Cell Biochem Nov 2021;476(11):3963–3974. doi: 10.1007/s11010-021-04210-5 [DOI] [PubMed] [Google Scholar]
  • 79.Sreeramkumar V, Adrover JM, Ballesteros I, et al. Neutrophils scan for activated platelets to initiate inflammation. Science Dec 5 2014;346(6214):1234–8. doi: 10.1126/science.1256478 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Simon DI, Chen Z, Xu H, et al. Platelet glycoprotein ibalpha is a counterreceptor for the leukocyte integrin Mac-1 (CD11b/CD18). J Exp Med Jul 17 2000;192(2):193–204. doi: 10.1084/jem.192.2.193 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Wang Y, Sakuma M, Chen Z, et al. Leukocyte engagement of platelet glycoprotein Ibalpha via the integrin Mac-1 is critical for the biological response to vascular injury. Circulation Nov 8 2005;112(19):2993–3000. doi: 10.1161/CIRCULATIONAHA.105.571315 [DOI] [PubMed] [Google Scholar]
  • 82.Proenca-Ferreira R, Brugnerotto AF, Garrido VT, et al. Endothelial activation by platelets from sickle cell anemia patients. PLoS One 2014;9(2):e89012. doi: 10.1371/journal.pone.0089012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Dominical VM, Samsel L, Nichols JS, et al. Prominent role of platelets in the formation of circulating neutrophil-red cell heterocellular aggregates in sickle cell anemia. Haematologica Nov 2014;99(11):e214–7. doi: 10.3324/haematol.2014.108555 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Belcher JD, Chen C, Nguyen J, et al. Heme triggers TLR4 signaling leading to endothelial cell activation and vaso-occlusion in murine sickle cell disease. Blood Jan 16 2014;123(3):377–90. doi: 10.1182/blood-2013-04-495887 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Brinkmann V, Reichard U, Goosmann C, et al. Neutrophil extracellular traps kill bacteria. Science Mar 5 2004;303(5663):1532–5. doi: 10.1126/science.1092385 [DOI] [PubMed] [Google Scholar]
  • 86.Castanheira FVS, Kubes P. Neutrophils and NETs in modulating acute and chronic inflammation. Blood May 16 2019;133(20):2178–2185. doi: 10.1182/blood-2018-11-844530 [DOI] [PubMed] [Google Scholar]
  • 87.Sollberger G, Choidas A, Burn GL, et al. Gasdermin D plays a vital role in the generation of neutrophil extracellular traps. Sci Immunol Aug 24 2018;3(26)doi: 10.1126/sciimmunol.aar6689 [DOI] [PubMed] [Google Scholar]
  • 88.Leshner M, Wang S, Lewis C, et al. PAD4 mediated histone hypercitrullination induces heterochromatin decondensation and chromatin unfolding to form neutrophil extracellular trap-like structures. Front Immunol 2012;3:307. doi: 10.3389/fimmu.2012.00307 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Sollberger G, Tilley DO, Zychlinsky A. Neutrophil Extracellular Traps: The Biology of Chromatin Externalization. Dev Cell Mar 12 2018;44(5):542–553. doi: 10.1016/j.devcel.2018.01.019 [DOI] [PubMed] [Google Scholar]
  • 90.Gomez-Moreno D, Adrover JM, Hidalgo A. Neutrophils as effectors of vascular inflammation. Eur J Clin Invest Nov 2018;48 Suppl 2:e12940. doi: 10.1111/eci.12940 [DOI] [PubMed] [Google Scholar]
  • 91.Fuchs TA, Brill A, Duerschmied D, et al. Extracellular DNA traps promote thrombosis. Proc Natl Acad Sci U S A Sep 7 2010;107(36):15880–5. doi: 10.1073/pnas.1005743107 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Torres LS, Gotardo EMF, Leonardo FC, et al. Neutralization of Inflammasome-Processed Cytokines Reduces Inflammatory Mechanisms and Leukocyte Recruitment in the Vasculature of TNF-α-Stimulated Sickle Cell Disease Mice. Blood 2021;138(Supplement 1):856–856. doi: 10.1182/blood-2021-147436 [DOI] [Google Scholar]
  • 93.Kaul DK, Thangaswamy S, Suzuka SM, Fabry ME, Wanderer AA, Gram H. Anti-Interleukin-1β Antibody-Based Therapy Ameliorates Endothelial Activation and Inflammation in Sickle Mice. Blood 2011;118(21):848–848. doi: 10.1182/blood.V118.21.848.848 [DOI] [Google Scholar]
  • 94.Ng LG, Ostuni R, Hidalgo A. Heterogeneity of neutrophils. Nat Rev Immunol Apr 2019;19(4):255–265. doi: 10.1038/s41577-019-0141-8 [DOI] [PubMed] [Google Scholar]
  • 95.Casanova-Acebes M, Pitaval C, Weiss LA, et al. Rhythmic modulation of the hematopoietic niche through neutrophil clearance. Cell May 23 2013;153(5):1025–35. doi: 10.1016/j.cell.2013.04.040 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Adrover JM, Del Fresno C, Crainiciuc G, et al. A Neutrophil Timer Coordinates Immune Defense and Vascular Protection. Immunity Feb 19 2019;50(2):390–402 e10. doi: 10.1016/j.immuni.2019.01.002 [DOI] [PubMed] [Google Scholar]
  • 97.Martin C, Burdon PC, Bridger G, Gutierrez-Ramos JC, Williams TJ, Rankin SM. Chemokines acting via CXCR2 and CXCR4 control the release of neutrophils from the bone marrow and their return following senescence. Immunity Oct 2003;19(4):583–93. doi: 10.1016/s1074-7613(03)00263-2 [DOI] [PubMed] [Google Scholar]
  • 98.van Eeden SF, Kitagawa Y, Klut ME, Lawrence E, Hogg JC. Polymorphonuclear leukocytes released from the bone marrow preferentially sequester in lung microvessels. Microcirculation Sep 1997;4(3):369–80. doi: 10.3109/10739689709146801 [DOI] [PubMed] [Google Scholar]
  • 99.Zhang D, Chen G, Manwani D, et al. Neutrophil ageing is regulated by the microbiome. Nature Sep 24 2015;525(7570):528–32. doi: 10.1038/nature15367 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Xu C, Lee SK, Zhang D, Frenette PS. The Gut Microbiome Regulates Psychological-Stress-Induced Inflammation. Immunity Aug 18 2020;53(2):417–428 e4. doi: 10.1016/j.immuni.2020.06.025 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Brim H, Taylor J, Abbas M, et al. The gut microbiome in sickle cell disease: Characterization and potential implications. PLoS One 2021;16(8):e0255956. doi: 10.1371/journal.pone.0255956 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Lim SH, Morris A, Li K, et al. Intestinal microbiome analysis revealed dysbiosis in sickle cell disease. Am J Hematol Aug 2018;93(4):E91–E93. doi: 10.1002/ajh.25019 [DOI] [PubMed] [Google Scholar]
  • 103.Garcia F, Mendonca R, Miguel LI, et al. CXCR4(hi) effector neutrophils in sickle cell anemia: potential role for elevated circulating serotonin (5-HT) in CXCR4(hi) neutrophil polarization. Sci Rep Aug 31 2020;10(1):14262. doi: 10.1038/s41598-020-71078-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Torres LS, Teles LIM, Shaul ME, et al. Accelerated low-density neutrophil transition in sickle cell anaemia may contribute to disease pathophysiology. Br J Haematol Apr 2022;197(2):232–235. doi: 10.1111/bjh.18009 [DOI] [PubMed] [Google Scholar]
  • 105.Sagiv JY, Voels S, Granot Z. Isolation and Characterization of Low- vs. High- Density Neutrophils in Cancer. Methods Mol Biol 2016;1458:179–93. doi: 10.1007/978-1-4939-3801-8_13 [DOI] [PubMed] [Google Scholar]
  • 106.Sionov RV, Assi S, Gershkovitz M, et al. Isolation and Characterization of Neutrophils with Anti-Tumor Properties. J Vis Exp Jun 19 2015;(100):e52933. doi: 10.3791/52933 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Ballesteros I, Rubio-Ponce A, Genua M, et al. Co-option of Neutrophil Fates by Tissue Environments. Cell Nov 25 2020;183(5):1282–1297 e18. doi: 10.1016/j.cell.2020.10.003 [DOI] [PubMed] [Google Scholar]
  • 108.Crainiciuc G, Palomino-Segura M, Molina-Moreno M, et al. Behavioural immune landscapes of inflammation. Nature Jan 2022;601(7893):415–421. doi: 10.1038/s41586-021-04263-y [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES