You are on page 1of 7

Effects of sepsis on neutrophil chemotaxis

Raju C. Reddy and Theodore J. Standiford


Department of Internal Medicine, Division of Pulmonary Purpose of review
and Critical Care Medicine, University of Michigan
Medical Center, Ann Arbor, Michigan, USA
Neutrophil recruitment to sites of infection is a critical element of the innate immune
response. In patients with sepsis, this response is dysregulated, with exuberant
Correspondence to Raju C. Reddy, MD, University of
Michigan Medical Center, Division of Pulmonary and inflammation being followed by a state of profound immune suppression, including
Critical Care Medicine, 109 Zina Pitcher Place, 4062 inhibition of neutrophil recruitment. This review examines mechanisms underlying
BSRB, Ann Arbor, MI 48109-2200, USA
Tel: +1 734 615 2871; fax: +1 734 615 2111; suppression of neutrophil migration during sepsis.
e-mail: rajuc@umich.edu Recent findings
Current Opinion in Hematology 2010,
Mechanisms governing neutrophil chemotactic function in sepsis are complex. Bacterial
17:1824 products, cytokines, and chemokines can modulate neutrophil migratory responses
during sepsis via induction of cytoskeletal changes, inhibition of polymorphonuclear
leukocyte (PMN)endothelial cell interactions, and alterations in G protein-coupled
receptor expression or signaling. Impaired chemotactic responses can occur as a result
of dysregulated PMN Toll-like receptor signaling. Other recently identified inhibitory
mechanisms include exposure to elevated temperatures, activation of the anti-
inflammatory nuclear transcription factor peroxisome proliferator-activated receptor-g,
and suppression of PMNendothelial interactions due to nitric oxide and its
metabolites. Finally, circulating microparticles released in sepsis exert important
immunomodulatory effects on PMN adherence and transmigration.
Summary
Neutrophil recruitment is a coordinated process that is altered at multiple stages during
sepsis, culminating in defective innate immunity and increased risk of infection in these
patients. Defining mechanisms involved and strategies to interrupt these deleterious
responses requires further investigation.

Keywords
immunosuppression, nitric oxide, peroxisome proliferator-activated receptor-g,
systemic inflammatory response syndrome, Toll-like receptors

Curr Opin Hematol 17:1824


2010 Wolters Kluwer Health | Lippincott Williams & Wilkins
1065-6251

ance, as an increase in mortality is observed in severe


Introduction sepsis patients who display impaired PMN chemotactic
Sepsis is a progressive, injurious inflammatory response to responses [5,6]. This review will cover recent advances in
infection [1]. Burns, trauma, and major surgery can our knowledge of how sepsis modulates neutrophil
promote analogous clinical responses, referred to as the migration and the molecular mechanisms involved.
systemic inflammatory response syndrome (SIRS) [2].
Growing evidence indicates that the failure to maintain
the necessary balance between excessive and inadequate Overview of sepsis
inflammation is a fundamental pathologic feature of Host immune responses in sepsis can be conceptualized
sepsis [3,4]. Neutrophils are key cells in the innate as occurring in distinct, but overlapping, phases. The
response, releasing important regulatory cytokines and initial response in sepsis, referred to as SIRS, is charac-
chemokines, engulfing invading microbes, and releasing terized by the release of a number of proinflammatory
antimicrobial proteins and oxidants required for microbial mediators, including early response cytokines such as
killing. Neutrophil migration in severe sepsis is impaired, tumor necrosis factor-a (TNFa), interleukin (IL)-1b,
resulting in an inadequate recruitment of polymorpho- leukocyte-active chemokines, leukotrienes, adhesion
nuclear leukocytes (PMNs) to sites of microbial invasion molecules, reactive oxygen species, and nitric oxide
[5,6,7,8]. Sepsis is also characterized by deleterious [4,9]. SIRS is counter-regulated by the release of inhibi-
accumulation of neutrophils in vital organs, culminating tory molecules, including anti-inflammatory cytokines
in PMN-mediated organ failure [3]. Impaired recruit- [e.g. IL-10 and transforming growth factor-b (TGFb)],
ment of PMNs in sepsis is of considerable clinical import- suppressors of pathogen recognition signaling cascades,
1065-6251 2010 Wolters Kluwer Health | Lippincott Williams & Wilkins DOI:10.1097/MOH.0b013e32833338f3

Copyright Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
Effects of sepsis on neutrophil chemotaxis Reddy and Standiford 19

Figure 1 Schematic depicting pathophysiological phases in sepsis, including mediators involved, biological effects, and clinical
manifestations

CARS, compensatory anti-inflammatory response syndrome; G-CSF, granulocyte colony-stimulating factor; GM-CSF, granulocyte-macrophage
colony-stimulating factor; IL, interleukin; NO, nitric oxide; PPARg, peroxisome proliferator-activated receptor-g; SIRS, systemic inflammatory response
syndrome; TLR, Toll-like receptor; TNF, tumor necrosis factor. Adapted with permission from [11].

immunomodulatory eicosanoids, and hormones. This Neutrophil origin and mobilization


counter-regulatory phase is referred to as the compensa- Neutrophils originate from bone marrow stem cells under
tory anti-inflammatory response syndrome (CARS) [9,10]. the influence of granulocyte colony-stimulating factor
Molecules released during CARS are believed to limit (G-CSF) [12]. G-CSF also stimulates release of mature
systemic inflammation, and the expression of these neutrophils from the marrow under baseline conditions,
mediators is induced by both microbial-derived and with release being further stimulated by inflammation-
host-derived signals. Historically, therapeutic interven- associated cytokines. In the absence of inflammation,
tions in sepsis have targeted selected mediators of SIRS. neutrophils circulate for a brief period (6 h) before
However, many randomized, controlled trials directed becoming senescent and being cleared by liver, spleen,
against these mediators have failed to improve outcomes and bone marrow [13]. Once neutrophils have reached the
in patients with sepsis, and in some cases have resulted in site of infection, however, their otherwise short lifetime is
detrimental effects. The failure of these trials has brought extended by inflammatory mediators such as granulocyte-
the functional importance of CARS in sepsis to light [10]. macrophage colony-stimulating factor (GM-CSF), chemo-
Molecules expressed during SIRS and CARS are depicted kines, and other chemoattractant molecules [14].
in Fig. 1 [11].
Margination and rolling
Movement of neutrophils to the vessel wall allows low-
Overview of neutrophil recruitment and affinity interactions between neutrophils and the endo-
migration thelial layer [15]. The primary molecular effectors of these
Recruitment of neutrophils to sites of inflammatory or interactions are L-selectin, constitutively expressed on
infectious insult is one of the initial and most critical arms circulating leukocytes, and E-selectin and P-selectin,
of the innate immune response. Four distinct phases of expressed on endothelial cells after activation by chemo-
neutrophil recruitment have been described: mobiliz- kines and other inflammatory mediators.
ation, margination (movement to the vessel periphery)
and rolling, adherence, and transmigration through the Adhesion
vessel wall along chemotactic gradients. All phases of The combination of low-affinity contact between neu-
PMN migration are altered during sepsis [7,8]. trophils and endothelial cells and chemokine activation

Copyright Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
20 Myeloid biology

induces molecules that mediate high-affinity adhesion. migrating in the direction of highest receptor occupancy
The relevant adhesion molecules on neutrophils are the [17].
integrins, two-chain heterodimers with a number of vari-
ants for each subunit. The aLb2-integrin, also known as
CD11a/CD18, is expressed on the surface of most leuko- Mechanisms influencing neutrophil migration
cytes, whereas the aMb2-integrin, also known as CD11b/ in sepsis
CD18, is mobilized to the cell surface of monocytes and Multiple mechanisms contribute to sepsis-induced sup-
neutrophils following activation by microbial components, pression of neutrophil recruitment, although our under-
inflammatory cytokines, or chemokines. These integrins standing of this complex process remains incomplete.
interact with adhesion molecules on the surface of the Key events, including recent findings, are summarized in
endothelial cell to provide the firm anchorage that charac- this review and illustrated schematically in Fig. 2.
terizes adhesion, with the most important endothelial
adhesion molecules being intercellular adhesion mol- Alterations in neutrophil rigidity and sequestration
ecule-1 (ICAM-1) and vascular cell adhesion molecule-1 Factors released during sepsis result in marked increase in
(VCAM-1). the rigidity of neutrophil cell membranes. As a result of
altered deformability, neutrophils sequester in capillary
Transmigration and chemotaxis beds, especially those of the lung. Sequestered neutrophils
The final stage in neutrophil recruitment is passage across neither migrate through endothelium nor complete their
the endothelium through tight junctions into infected passage through the capillary. The occlusion of vasculature
tissues. This process is guided by concentration gradients by PMN limits blood flow, resulting in tissue ischemia,
of chemoattractants, which include both microbial com- which contributes to multiple organ failure [18,19]. Rigid-
ponents, such as formyl-methionyl-leucyl-phenylalanine ity increases with sepsis severity [19], whereas a progress-
(fMLP), and host-derived chemoattractants, including ive decrease in rigidity signals recovery [18]. This
complement components (e.g. C5a), eicosanoids (e.g. increased rigidity, which can be induced in vitro by
leukotriene B4 and platelet-activating factor), and che- exposure to fMLP or TNFa [18], is associated with
mokines [16]. Many of these chemoattractants act accumulation of F-actin immediately below the cell mem-
through G protein-coupled receptors (GPCRs), with cells brane but not with cytoskeletal rearrangement [20]. A

Figure 2 Schematic depicting stages of polymorphonuclear leukocyte recruitment and how these various stages are altered by
mediators expressed in sepsis

CXCR2, CXC chemokine receptor 2; G-CSF, granulocyte colony-stimulating factor; GM-CSF, granulocyte-macrophage colony-stimulating factor;
GPCR, G protein-coupled receptor; HO-1, heme oxygenase-1; ICAM, intercellular adhesion molecule; Ig, immunoglobulin; NO, nitric oxide; PMN,
polymorphonuclear leukocyte; PPARg, peroxisome proliferator-activated receptor-g; TLR, Toll-like receptor; TNF, tumor necrosis factor; VCAM,
vascular cell adhesion molecule. , selectins; , Ig superfamily members; , integrins.

Copyright Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
Effects of sepsis on neutrophil chemotaxis Reddy and Standiford 21

causal connection between submembrane F-actin and In addition, decreased CXCR2 expression appears to be
sequestration is demonstrated by the finding that only partially mediated by activation of TLR2, as treatment of
neutrophils with submembrane F-actin rims are lost during PMNs with the TLR2 agonist lipoteichoic acid reduced
passage through the lungs of rats with bacterial pneumonia CXCR2 expression and chemotactic responses in vitro,
[21]. whereas neutrophil CXCR2 expression is well maintained
in septic mice deficient in TLR2 [29].
Alterations in Toll-like receptor expression and
signaling Receptor desensitization can also occur as a consequence of
Toll-like receptors (TLRs) are a family of cell surface and differential ligand binding to high and low-affinity recep-
intracellular pathogen recognition receptors that are tors. For instance, Herrmann et al. [30] have recently
required for the generation of immune responses to micro- reported that stimulation of PMNs with fMLP at low
bial pathogens. PMNs express several TLRs, including concentration (10 7 mol/l) results in binding to its high-
TLR2, which is activated by pathogen-associated molecu- affinity receptor, leading to chemotactic responses without
lar patterns (PAMPs) expressed by Gram-positive bacteria, release of oxidants. Conversely, exposure to this chemoat-
and TLR4, which is activated by lipopolysaccharide tractant at higher concentrations (10 5 mol/l) activates its
released from Gram-negative bacteria [22]. Stimulation low-affinity receptor, resulting in stimulation of oxidative
of either TLR2 or TLR4 enhances random motility of burst while blocking high-affinity receptor-mediated che-
neutrophils via the extracellular-signal regulated kinase motactic responses, including fMLP-induced calcium flux.
mitogen-activated protein kinase (ERKMAPK) signaling
pathway, but directed motility (chemotaxis) requires A final means by which the presence of GPCR ligands in
further stimulation with a chemoattractant such as fMLP high concentrations can mitigate chemotactic responses
[23]. Neutrophils can also be activated in a TLR-depen- is via phosphorylation of agonist-occupied GPCR by
dent fashion by host-derived inflammatory mediators GPCR kinases (GRKs) [31]. An increase in the expres-
released in sepsis, referred to as danger-associated molecu- sion of selected GRKs (GRK2 and GRK5) is found in
lar patterns (DAMPs) or alarmins. Observations made in PMNs isolated from sepsis patients [32]. Induction of
animal models of sepsis and in neutrophils isolated from PMN GRKs can be reproduced in vitro by treatment with
patients with septic shock indicate that neutrophil expres- inflammatory stimuli released in the setting of sepsis.
sion of TLR2 and TLR4 is diminished [24]. Moreover,
microarray analysis of PMNs isolated from septic patients A number of inhibitors of GPCR-mediated signaling have
within 24 h of onset reveals a global suppression of immune been described. Recent studies have shown that the
regulation and inflammatory response gene clusters, receptor for activated C kinase 1 (RACK1) modulates
particularly genes regulated in a TLR or nuclear factor chemotaxis by competitively binding to G protein bg and
(NF)-kB-dependent fashion [25]. Conversely, the expres- thereby blocking binding to the G-protein targets such as
sion of selected suppressive genes is enhanced, including phosphatidylinositol 3-kinase g and phospholipase C
the NF-kB inhibitor alpha, NFkBIA. Sepsis also induces [33]. The role of this GPCR inhibitor as a mediator of
the upregulation of cytokines or proteins that inhibit down- impaired PMN chemotactic responses has not been
stream TLR signaling cascades. In particular, IL-10 pro- described but is worthy of further investigation.
duced by macrophages and other cells can substantially
diminish TLR activation and PMN responses to chemoat- Another novel mechanism for inhibition of chemoattrac-
tractants in a paracrine fashion [26], whereas we have found tant-mediated signaling is exposure to elevated tempera-
that the expression of the IL-1 receptor-associated kinase tures similar to those associated with infection [34]. In a
IRAK-M is upregulated in PMNs isolated from patients murine model of skin infection, raising the body tempera-
with sepsis (unpublished observations). ture from baseline to 408C strongly reduced subsequent
neutrophil infiltration. Neutrophil chemotaxis toward
Alteration of G protein-coupled receptor expression and GM-CSF or IL-8, but not adhesion and spreading, was
downstream signaling events in sepsis likewise inhibited by elevated temperatures in vitro,
A paramount feature of the septic response is the exuber- which was associated with a reduction in chemoattrac-
ant release of leukocyte chemoattractants that function tant-induced downstream activation of NF-kB. These
through activation of the GPCR. Exposure to high levels of results suggest that sepsis-induced fever may be one of
ligand can result in functional desensitization of receptor the mechanisms contributing to impaired chemotactic
responsiveness, which can occur as a result of downregula- responses in this syndrome.
tion of GPCR cell surface expression [27]. Decreased
expression of the CXC chemokine receptor 2 (CXCR2) Nitric oxide as a mediator of sepsis-induced impairment
on neutrophils isolated from sepsis patients has been in neutrophil chemotaxis
observed [28]. This rapid downregulation is due, in part, Sepsis results in a dramatic increase in the elaboration of
to internalization and recycling of occupied receptors [27]. nitric oxide, which is largely attributed to inflammatory

Copyright Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
22 Myeloid biology

cytokine and endotoxin-mediated upregulation of indu- isolated from septic mice was reversed by treatment with
cible nitric oxide synthase (iNOS or NOS-2) [35]. Nitric the PPARg antagonist GW9662.
oxide has been identified as an important modulator of
neutrophil migration, although data exist to support both Napimoga et al. [47] studied the influence of PPARg on
stimulatory and inhibitory effects of nitric oxide on PMN PMN adhesion responses in mesenteric postcapillary
migration [36,37]. This molecule directly inhibits leuko- venules using a carrageenan-induced peritonitis model
cyteendothelial cell interactions, primarily by inhibiting in vivo [48]. Importantly, treatment with 15d-PGJ2
leukocyte b2-integrins and selectins, as well as endo- inhibited both PMN rolling and tight adherence to endo-
thelial cell ICAM-1 and VCAM-1 [38,39]. Mestriner et al. thelium, which was attributable, in part, to suppression of
[40] demonstrated that acute-phase protein a-1 dose- mesenteric ICAM-1 expression. A role for nitric oxide in
dependently inhibited neutrophil migration into the rat mediating these effects was indicated by the inability of
peritoneal cavity following carrageenan injection or cecal 15d-PGJ2 to block migration when nitric oxide synthase
ligation and puncture (CLP), and that this effect could be was inhibited or iNOS was genetically absent. In-vitro
eliminated either by pharmacological inhibition of iNOS neutrophil chemotaxis was likewise inhibited by 15d-
or by genetic deficiency of this enzyme. PGJ2 or rosiglitazone, an effect that was again blocked by
specific inhibitors of either PPARg or by a specific
Nitric oxide can also interact with other molecules gener- inhibitor of iNOS. It is uncertain whether nitric oxide
ated during the septic response including reactive oxygen is acting intracellularly or as an extracellular autocrine
species. For instance, nitric oxide reacts with the super- mediator in this setting.
oxide anion to form peroxynitrite, which has been shown
to inhibit PMN migration by blocking leukocyteendo- Role of microparticles in regulating polymorphonuclear
thelial cell interactions in a P-selectin-dependent fashion leukocyte chemotactic responses in sepsis
and by inhibiting actin polymerization required for neu- Circulating microparticles are membrane-coated particles
trophil locomotion [4143]. Moreover, nitric oxide released from activated or apoptotic cells, including leu-
induces the expression of the microsomal enzyme heme kocytes, platelets, erythrocytes, and endothelial cells
oxygenase-1 (HO-1). Metabolites of HO-1 disrupt PMN [50]. Microparticle release is enhanced in patients with
rolling and adhesion [4446]. Importantly, inhibition of sepsis and can exert both beneficial and detrimental
HO-1 during experimental sepsis (CLP) restores PMN effects on inflammatory responses in sepsis, including
responses in septic animals. neutrophil recruitment [51,52]. Microparticles released
from neutrophils enhance PMN chemotaxis to IL-8,
Peroxisome proliferator-activated receptor-g activation which is mediated by L-selectin and P-selectin glyco-
suppresses neutrophil chemotaxis protein ligand-1 present on the surface of these micro-
Two recent studies [47,48] highlight the importance of particles [53]. Interestingly, treatment of PMNs with
peroxisome proliferator-activated receptor-g (PPARg) as the NOS inhibitor NG-nitro-L-arginine methyl ester
a negative regulator of neutrophil migration. PPARs are a (L-NAME) stimulated the release of microparticles from
family of nuclear hormone receptors that play a promi- these cells, indicating that nitric oxide tonically sup-
nent role in regulating fundamental aspects of cellular presses neutrophil microparticle formation and release.
activation, proliferation, and differentiation. The ligand- Conversely, it has recently been shown that neutrophil-
activated nuclear transcription factor PPARg not only derived microparticles can also inhibit, rather than facili-
stimulates transcription of certain target genes involved tate, neutrophil chemotaxis [54]. This inhibitory effect
in adipocyte differentiation and glucose metabolism but appears to be mediated by the action of the anti-inflam-
also downregulates activity of key proinflammatory trans- matory protein annexin 1, which is bound to the surface of
cription factors, including NF-kB and signal transducer microparticles. Collectively, the aforementioned studies
and activator of transcription 6 (STAT6), by competing indicate that microparticles can exert varied effects on
for common coactivators [49]. We have shown that PMN migratory responses depending upon the cell of
PPARg expression is increased in neutrophils isolated microparticle origin and the nature of the molecules
either from septic mice or from humans with sepsis [48]. present on the surface of these structures.
Induction of neutrophil PPARg expression could be repli-
cated in vitro by exposure to either TNFa or IL-4, cyto-
kines elaborated in sepsis. Treatment with either the Conclusion
synthetic PPARg agonist troglitazone or the natural ago- Sepsis represents a severe derangement of the immune
nist 15-deoxy-D12,14-prostaglandin J2 (15d-PGJ2) inhibited response to infection, resulting in end-organ injury and
neutrophil chemotaxis in response to IL-8 or fMLP, which a profound state of immune suppression. Recruitment
occurred in association with reduced fMLP-induced phos- of neutrophils to sites of infection, a crucial component
phorylation of ERK-1/2 and alterations in F-actin assem- of the innate immune response, is substantially impaired
bly. Moreover, the reduced ex-vivo chemotaxis of PMNs in sepsis. Recently explored mechanisms include

Copyright Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
Effects of sepsis on neutrophil chemotaxis Reddy and Standiford 23

downregulation or inhibition of cell surface GPCR and 19 Skoutelis AT, Kaleridis V, Athanassiou GM, et al. Neutrophil deformability in
patients with sepsis, septic shock, and adult respiratory distress syndrome.
TLRs that activate neutrophils. Factors that mediate Crit Care Med 2000; 28:23552359.
these deleterious responses include anti-inflammatory 20 Saito H, Lai J, Rogers R, et al. Mechanical properties of rat bone marrow and
cytokines, activation of the nuclear transcription factor circulating neutrophils and their responses to inflammatory mediators. Blood
2002; 99:22072213.
PPARg, and nitric oxide and its metabolites. Strategies to
21 Yoshida K, Kondo R, Wang Q, et al. Neutrophil cytoskeletal rearrangements
interrupt dysregulated PMN migratory response in sepsis during capillary sequestration in bacterial pneumonia in rats. Am J Respir Crit
are desperately needed. Care Med 2006; 174:689698.
22 Akira S, Hemmi H. Recognition of pathogen-associated molecular patterns by
TLR family. Immunol Lett 2003; 85:8595.
23 Aomatsu K, Kato T, Fujita H, et al. Toll-like receptor agonists stimulate human
Acknowledgements  neutrophil migration via activation of mitogen-activated protein kinases. Im-
This study is supported by National Institutes of Health grants HL093196 munology 2008; 123:171180.
(R.C.R.), HL25243, and P50 HL074024 (T.J.S.). The authors would like This article addresses the intracellular signaling mechanisms by which TLRs
induce chemotaxis.
to thank Robin Kunkel for her assistance in preparation of the schematics
included in this review. 24 Salomao R, Brunialti MK, Gomes NE, et al. Toll-like receptor pathway
signaling is differently regulated in neutrophils and peripheral mononuclear
cells of patients with sepsis, severe sepsis, and septic shock. Crit Care Med
2009; 37:132139.
References and recommended reading 25 Tang BMP, McLean AS, Dawes IW, et al. The use of gene-expression profiling
Papers of particular interest, published within the annual period of review, have to identify candidate genes in human sepsis. Am J Respir Crit Care Med
been highlighted as: 2007; 176:676684.
 of special interest 26 Mashimo H, Obguro N, Nomura S, et al. Neutrophil chemotaxis and local
 of outstanding interest expression of interleukin-10 in the tolerance of endotoxin-induced uveitis.
Additional references related to this topic can also be found in the Current Invest Opthalmol Vis Sci 2008; 49:54505457.
World Literature section in this issue (p. 68).
27 Baggiolini M. Chemokines and leukocyte traffic. Nature 1998; 392:565568.
1 American College of Chest Physicians/Society of Critical Care Medicine 28 Cummings CJ, Martin TR, Frevert CW, et al. Expression and function of the
Consensus Conference: definitions for sepsis and organ failure and guide- chemokine receptors CXCR1 and CXCR2 in sepsis. J Immunol 1999;
lines for the use of innovative therapies in sepsis. Crit Care Med 1992; 162:23412346.
20:864874.
29 Alves-Filho JC, Freitas A, Souto FO, et al. Regulation of chemokine receptor
2 Brun-Buisson C. The epidemiology of the systemic inflammatory response.  by toll-like receptor 2 is critical to neutrophil migration and resistance to
Intensive Care Med 2000; 26 (Suppl 1):S64S74. polymicrobial sepsis. Proc Natl Acad Sci U S A 2009; 106:40184023.
3 Cohen J. The immunopathogenesis of sepsis. Nature 2002; 420:885891. This article defines the contribution of TLR2 to shedding of CXCR2 in murine
sepsis.
4 Dinarello CA. Proinflammatory and anti-inflammatory cytokines as mediators
in the pathogenesis of septic shock. Chest 1997; 112:321S329S. 30 Herrmann JM, Bernardo J, Long HJ, et al. Sequential chemotactic and
phagocytic activation of human polymorphonuclear neutrophils. Infect Immun
5 Muller Kobold AC, Tulleken JE, Zijlstra JG, et al. Leukocyte activation in sepsis: 2007; 75:39893998.
correlations with disease state and mortality. Intensive Care Med 2000;
26:883892. 31 Penela P, Ribas C, Mayor F Jr. Mechanisms of regulation of the expression
and function of G protein-coupled receptor kinases. Cell Signal 2003;
6 Tavares-Murta BM, Zaparoli M, Ferreira RB, et al. Failure of neutrophil 15:973981.
chemotactic function in septic patients. Crit Care Med 2002; 30:1056
1061. 32 Arraes SM, Freitas MS, da Silva SV, et al. Impaired neutrophil chemotaxis in
sepsis associates with GRK expression and inhibition of actin assembly and
7 Pinheiro da Silva F, Soriano FG. Neutrophils recruitment during sepsis: critical tyrosine phosphorylation. Blood 2006; 108:29062913.
 points and crossroads. Front Biosci 2009; 14:44644476.
A detailed review focused on molecular mechanisms of neutrophil migration and 33 Chen S, Lin F, Shin ME, et al. RACK1 regulates directional cell migration by
tissue damage in sepsis. acting on G betagamma at the interface with its effectors PLC beta and PI3K
gamma. Mol Biol Cell 2008; 19:39093922.
8 Alves-Filho JC, de Freitas A, Spiller F, et al. The role of neutrophils in severe
 sepsis. Shock 2008; 30 (Suppl 1):39. 34 Choi M, Salanova B, Rolle S, et al. Short-term heat exposure inhibits
A comprehensive review addressing mechanisms underlying impaired neutrophil  inflammation by abrogating recruitment of and nuclear factor-kB activation
migration in sepsis. in neutrophils exposed to chemotactic cytokines. Am J Pathol 2008; 172:
367777.
9 van der Poll T, van Deventer SJ. Cytokines and anticytokines in the pathogen- This study describes the novel effect of fever on neutrophil migratory responses in
esis of sepsis. Infect Dis Clin North Am 1999; 13:413426. sepsis.
10 Bone RC, Grodzin CJ, Balk RA. Sepsis: a new hypothesis for pathogenesis of 35 Alderton WK, Cooper CE, Knowles RG. Nitric oxide synthases: structure,
the disease process. Chest 1997; 112:235243. function and inhibition. Biochem J 2001; 357:593615.
11 Lyn-Kew K, Standiford TJ. Immunosuppression in sepsis. Curr Pharm Des 36 Benjamim CF, Ferreira SH, Cunha FQ. Role of nitric oxide in the failure of
2008; 14:18701881. neutrophil migration in sepsis. J Infect Dis 2000; 182:214223.
12 Furze RC, Rankin SM. Neutrophil mobilization and clearance in the bone 37 Moilanen E, Vuorinen P, Kankaanranta H, et al. Inhibition by nitric oxide-donors
marrow. Immunology 2008; 125:281288. of human polymorphonuclear leucocyte functions. Br J Pharmacol 1993;
13 Furze RC, Rankin SM. The role of the bone marrow in neutrophil clearance 109:852858.
under homeostatic conditions in the mouse. FASEB J 2008; 22:31113119. 38 Lefer DJ, Jones SP, Girod WG, et al. Leukocyte-endothelial cell interactions in
14 Hofman P. Molecular regulation of neutrophil apoptosis and potential targets nitric oxide synthase-deficient mice. Am J Physiol 1999; 276:H1943H1950.
for therapeutic strategy against the inflammatory process. Curr Drug Targets 39 Ialenti A, Ianaro A, Maffia P, et al. Nitric oxide inhibits leucocyte migration in
Inflamm Allergy 2004; 3:19. carrageenin-induced rat pleurisy. Inflamm Res 2000; 49:411417.
15 Muller WA. Leukocyte-endothelial-cell interactions in leukocyte transmigra- 40 Mestriner FL, Spiller F, Laure HJ, et al. Acute-phase protein alpha-1-acid
tion and the inflammatory response. Trends Immunol 2003; 24:327334. glycoprotein mediates neutrophil migration failure in sepsis by a nitric oxide-
16 Bokoch GM. Chemoattractant signaling and leukocyte activation. Blood dependent mechanism. Proc Natl Acad Sci U S A 2007; 104:1959519600.
1995; 86:16491660. 41 Lefer DJ, Scalia R, Campbell B, et al. Peroxynitrite inhibits leukocyte-
17 Katanaev VL. Signal transduction in neutrophil chemotaxis. Biochemistry endothelial cell interactions and protects against ischemia-reperfusion injury
(Mosc) 2001; 66:351368. in rats. J Clin Invest 1997; 99:684691.
18 Drost EM, Kassabian G, Meiselman HJ, et al. Increased rigidity and priming of 42 Torres-Duenas D, Celes MR, Freitas A, et al. Peroxynitrite mediates the failure
polymorphonuclear leukocytes in sepsis. Am J Respir Crit Care Med 1999; of neutrophil migration in severe polymicrobial sepsis in mice. Br J Pharmacol
159:16961702. 2007; 152:341352.

Copyright Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
24 Myeloid biology

43 Clements MK, Siemsen DW, Swain SD, et al. Inhibition of actin polymerization 49 Becker J, Delayre-Orthez C, Frossard N, et al. Regulation of inflammation by
by peroxynitrite modulates neutrophil functional responses. J Leukoc Biol PPARs: a future approach to treat lung inflammatory diseases? Fund Clin
2003; 73:344355. Pharmacol 2006; 20:429447.
44 Vicente AM, Guillen MI, Alcaraz MJ. Modulation of haem oxygenase-1 ex- 50 Gasser O, Hess C, Miot S, et al. Characterisation and properties of ecto-
pression by nitric oxide and leukotrienes in zymosan-activated macrophages. somes released by human polymorphonuclear neutrophils. Exp Cell Res
Br J Pharmacol 2001; 133:920926. 2003; 285:243257.
45 Freitas A, Alves-Filho JC, Secco DD, et al. Heme oxygenase/carbon monoxide- 51 Walenta KL, Link A, Friedrich EB, Bohm M. Circulating microparticles in septic
biliverdin pathway down regulates neutrophil rolling, adhesion and migration in  shock. Am J Respir Crit Care Med 2009; 180:100108.
acute inflammation. Br J Pharmacol 2006; 149:345354. One of the initial descriptions of the presence and function of microparticles in
patients with sepsis.
46 Zegdi R, Perrin D, Burdin M, et al. Increased endogenous carbon monoxide
production in severe sepsis. Intensive Care Med 2002; 28:793796. 52 Mortaza S, Martinez MC, Baron-Menguy C, et al. Detrimental hemodynamic
and inflammatory effects of microparticles originating from septic rats. Crit
47 Napimoga MH, Vieira SM, Dal-Secco D, et al. Peroxisome proliferator-
Care Med 2009; 37:20452050.
 activated receptor-gamma ligand, 15-deoxy-delta12,14-prostaglandin J2, re-
duces neutrophil migration via a nitric oxide pathway. J Immunol 2008; 53 Nolan S, Dixon R, Norman K, et al. Nitric oxide regulates neutrophil migration
180:609617.  through microparticle formation. Am J Pathol 2008; 172:265273.
The authors show that activation of PPARg inhibits neutrophil chemotaxis, and that This study describes the novel finding that nitric oxide can inhibit the release of
generation of nitric oxide is essential for this effect. microparticles from PMNs.
48 Reddy RC, Narala VR, Keshamouni VG, et al. Sepsis-induced inhibition of 54 Dalli J, Norling LV, Renshaw D, et al. Annexin 1 mediates the rapid anti-
 neutrophil chemotaxis is mediated by activation of peroxisome proliferator-  inflammatory effects of neutrophil-derived microparticles. Blood 2008;
activated receptor-g. Blood 2008; 112:42504258. 112:25122519.
This study demonstrates that activation of PPARg inhibits neutrophil chemotaxis, in This article shows that microparticles inhibit neutrophil chemotaxis as a result of
part, by blocking the ERK-1/2 intracellular signaling pathway. bound annexin.

Copyright Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.

You might also like