Neutrophil in Reverse Migration: Role in Sepsis

Jingjing Ji, Jie Fan, Jingjing Ji, Jie Fan

Abstract

Sepsis is life-threatening organ dysfunction caused by a dysregulated host response to infection. During the development and progression of sepsis, polymorphonuclear neutrophils (PMNs) are the most abundantly recruited innate immune cells at sites of infection, playing critical roles in the elimination of local infection and healing of the injury. PMN reverse migration (rM) describes the phenomenon in which PMNs migrate away from the inflammatory site back into the vasculature following the initial PMN infiltration. The functional role of PMN rM within inflammatory scenarios requires further exploration. Current evidence suggests that depending on the context, PMN rM can be both a protective response, by facilitating an efficient resolution to innate immune reaction, and also a tissue-damaging event. In this review, we provide an overview of current advancements in understanding the mechanism and roles of PMN rM in inflammation and sepsis. A comprehensive understanding of PMN rM may allow for the development of novel prophylactic and therapeutic strategies for sepsis.

Keywords: PMN; infection; inflammation; rerverse migration; sepsis.

Conflict of interest statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Copyright © 2021 Ji and Fan.

Figures

Figure 1
Figure 1
Mechanisms of PMN rM. 1. Breach of endothelium results in leakage of chemokine, i.e., CXCL1, from the inflammatory site into the vasculature, therefore driving PMN to reenter the circulation; 2. LTB4 induces PMN release of neutrophil elastase (NE), which in turn cleaves endothelial JAM-C and subsequent damage of endothelial junction and promotes PMN rM. The binding between PMN CD11b/CD18 and endothelial ICAM-1 retains the PMN on the surface of endothelial cells and secures the cleavage of JAM-C by NE; 3. Activation of HIF1α suppresses PMN rM; and 4. Many factors, including cold-inducible RNA-binding protein (CIRP), Lipoxin A4, PEG2, and cathepsin C, can promote PMN rM. The phenotype of reverse migrated PMN (rM-ed PMN) is ICAM1high CXCR1low.

References

    1. Amulic B, Cazalet C, Hayes GL, Metzler KD, Zychlinsky A. Neutrophil function: from mechanisms to disease. Annu Rev Immunol. (2012) 30:459–89. 10.1146/annurev-immunol-020711-074942
    1. Langereis JD. Neutrophil integrin affinity regulation in adhesion, migration, and bacterial clearance. Cell Adh Migr. (2013) 7:476–81. 10.4161/cam.27293
    1. Sadik CD, Kim ND, Luster AD. Neutrophils cascading their way to inflammation. Trends Immunol. (2011) 32:452–60. 10.1016/j.it.2011.06.008
    1. Kolaczkowska E, Kubes P. Neutrophil recruitment and function in health and inflammation. Nat Rev Immunol. (2013) 13:159–75. 10.1038/nri3399
    1. Summers C, Rankin SM, Condliffe AM, Singh N, Peters AM, Chilvers ER. Neutrophil kinetics in health and disease. Trends Immunol. (2010) 31:318–24. 10.1016/j.it.2010.05.006
    1. Injarabian L, Devin A, Ransac S, Marteyn BS. Neutrophil metabolic shift during their lifecycle: impact on their survival and activation. Int J Mol Sci. (2019) 21:287. 10.3390/ijms21010287
    1. Pittman K, Kubes P. Damage-associated molecular patterns control neutrophil recruitment. J Innate Immun. (2013) 5:315–23. 10.1159/000347132
    1. de Oliveira S, Rosowski EE, Huttenlocher A. Neutrophil migration in infection and wound repair: going forward in reverse. Nat Rev Immunol. (2016) 16:378–91. 10.1038/nri.2016.49
    1. Ley K, Laudanna C, Cybulsky MI, Nourshargh S. Getting to the site of inflammation: the leukocyte adhesion cascade updated. Nat Rev Immunol. (2007) 7:678–89. 10.1038/nri2156
    1. Hughes J, Johnson RJ, Mooney A, Hugo C, Gordon K, Savill J. Neutrophil fate in experimental glomerular capillary injury in the rat. Emigration exceeds in situ clearance by apoptosis. Am J Pathol. (1997) 150:223–34.
    1. Nourshargh S, Renshaw SA, Imhof BA. Reverse migration of neutrophils: where, when, how, and why? Trends Immunol. (2016) 37:273–86. 10.1016/j.it.2016.03.006
    1. Starnes TW, Huttenlocher A. Neutrophil reverse migration becomes transparent with zebrafish. Adv Hematol. (2012) 2012:398640. 10.1155/2012/398640
    1. Mathias JR, Perrin BJ, Liu T-X, Kanki J, Look AT, Huttenlocher A. Resolution of inflammation by retrograde chemotaxis of neutrophils in transgenic zebrafish. J Leukoc Biol. (2006) 80:1281–8. 10.1189/jlb.0506346
    1. Buckley CD, Ross EA, McGettrick HM, Osborne CE, Haworth O, Schmutz C, et al. . Identification of a phenotypically and functionally distinct population of long-lived neutrophils in a model of reverse endothelial migration. J Leukoc Biol. (2006) 79:303–11. 10.1189/jlb.0905496
    1. Silvestre-Roig C, Hidalgo A, Soehnlein O. Neutrophil heterogeneity: implications for homeostasis and pathogenesis. Blood. (2016) 127:2173–81. 10.1182/blood-2016-01-688887
    1. Powell DR, Huttenlocher A. Neutrophils in the tumor microenvironment. Trends Immunol. (2016) 37:41–52. 10.1016/j.it.2015.11.008
    1. Wang J, Hossain M, Thanabalasuriar A, Gunzer M, Meininger C, Kubes P. Visualizing the function and fate of neutrophils in sterile injury and repair. Science. (2017) 358:111–6. 10.1126/science.aam9690
    1. Yoo SK, Huttenlocher A. Spatiotemporal photolabeling of neutrophil trafficking during inflammation in live zebrafish. J Leukoc Biol. (2011) 89:661–7. 10.1189/jlb.1010567
    1. Hall C, Flores MV, Chien A, Davidson A, Crosier K, Crosier P. Transgenic zebrafish reporter lines reveal conserved Toll-like receptor signaling potential in embryonic myeloid leukocytes and adult immune cell lineages. J Leukoc Biol. (2009) 85:751–65. 10.1189/jlb.0708405
    1. Burn T, Alvarez JI. Reverse transendothelial cell migration in inflammation: to help or to hinder? Cell Mol Life Sci. (2017) 74:1871–81. 10.1007/s00018-016-2444-2
    1. Wu D, Zeng Y, Fan Y, Wu J, Mulatibieke T, Ni J, et al. . Reverse-migrated neutrophils regulated by JAM-C are involved in acute pancreatitis-associated lung injury. Sci Rep. (2016) 6:20545. 10.1038/srep20545
    1. Hind LE, Huttenlocher A. Neutrophil reverse migration and a chemokinetic resolution. Dev Cell. (2018) 47:404–5. 10.1016/j.devcel.2018.11.004
    1. Zhang Q, Raoof M, Chen Y, Sumi Y, Sursal T, Junger W, et al. . Circulating mitochondrial DAMPs cause inflammatory responses to injury. Nature. (2010) 464:104–7. 10.1038/nature08780
    1. Li L, Chen K, Xiang Y, Yoshimura T, Su S, Zhu J, et al. . New development in studies of formyl-peptide receptors: critical roles in host defense. J Leukoc Biol. (2016) 99:425–35. 10.1189/jlb.2RI0815-354RR
    1. Finger EB, Puri KD, Alon R, Lawrence MB, von Andrian UH, Springer TA. Adhesion through L-selectin requires a threshold hydrodynamic shear. Nature. (1996) 379:266–9. 10.1038/379266a0
    1. Lawrence MB, Kansas GS, Kunkel EJ, Ley K. Threshold levels of fluid shear promote leukocyte adhesion through selectins (CD62L,P,E). J Cell Biol. (1997) 136:717–27. 10.1083/jcb.136.3.717
    1. Vestweber D. How leukocytes cross the vascular endothelium. Nat Rev Immunol. (2015) 15:692–704. 10.1038/nri3908
    1. Weninger W, Biro M, Jain R. Leukocyte migration in the interstitial space of non-lymphoid organs. Nat Rev Immunol. (2014) 14:232–46. 10.1038/nri3641
    1. Owen-Woods C, Joulia R, Barkaway A, Rolas L, Ma B, Nottebaum AF, et al. . Local microvascular leakage promotes trafficking of activated neutrophils to remote organs. J Clin Invest. (2020) 130:2301–18. 10.1172/JCI133661
    1. Huang AJ, Manning JE, Bandak TM, Ratau MC, Hanser KR, Silverstein SC. Endothelial cell cytosolic free calcium regulates neutrophil migration across monolayers of endothelial cells. J Cell Biol. (1993) 120:1371–80. 10.1083/jcb.120.6.1371
    1. Marki A, Ley K. Leaking chemokines confuse neutrophils. J Clin Invest. (2020) 130:2177–9. 10.1172/JCI136259
    1. Colom B, Bodkin JV, Beyrau M, Woodfin A, Ody C, Rourke C, et al. . Leukotriene B4-neutrophil elastase axis drives neutrophil reverse transendothelial cell migration in vivo. Immunity. (2015) 42:1075–86. 10.1016/j.immuni.2015.05.010
    1. Li B, Han X, Ye X, Ni J, Wu J, Dai J, et al. . Substance P-regulated leukotriene B4 production promotes acute pancreatitis-associated lung injury through neutrophil reverse migration. Int Immunopharmacol. (2018) 57:147–56. 10.1016/j.intimp.2018.02.017
    1. Hirano Y, Ode Y, Ochani M, Wang P, Aziz M. Targeting junctional adhesion molecule-C ameliorates sepsis-induced acute lung injury by decreasing CXCR4+ aged neutrophils. J Leukoc Biol. (2018) 104:1159–71. 10.1002/JLB.3A0218-050R
    1. Powell D, Tauzin S, Hind LE, Deng Q, Beebe DJ, Huttenlocher A. Chemokine signaling and the regulation of bidirectional leukocyte migration in interstitial tissues. Cell Rep. (2017) 19:1572–85. 10.1016/j.celrep.2017.04.078
    1. Elks PM, van Eeden FJ, Dixon G, Wang X, Reyes-Aldasoro CC, Ingham PW, et al. . Activation of hypoxia-inducible factor-1α (Hif-1α) delays inflammation resolution by reducing neutrophil apoptosis and reverse migration in a zebrafish inflammation model. Blood. (2011) 118:712–22. 10.1182/blood-2010-12-324186
    1. Tharp WG, Yadav R, Irimia D, Upadhyaya A, Samadani A, Hurtado O, et al. . Neutrophil chemorepulsion in defined interleukin-8 gradients in vitro and in vivo. J Leukoc Biol. (2006) 79:539–54. 10.1189/jlb.0905516
    1. Loynes CA, Lee JA, Robertson AL, Steel MJ, Ellett F, Feng Y, et al. . PGE2 production at sites of tissue injury promotes an anti-inflammatory neutrophil phenotype and determines the outcome of inflammation resolution in vivo. Sci Adv. (2018) 4:eaar8320. 10.1126/sciadv.aar8320
    1. Levy BD, Clish CB, Schmidt B, Gronert K, Serhan CN. Lipid mediator class switching during acute inflammation: signals in resolution. Nat Immunol. (2001) 2:612–9. 10.1038/89759
    1. Serhan CN, Chiang N, Dalli J, Levy BD. Lipid mediators in the resolution of inflammation. Cold Spring Harb Perspect Biol. (2014) 7:a016311. 10.1101/cshperspect.a016311
    1. Hamza B, Wong E, Patel S, Cho H, Martel J, Irimia D. Retrotaxis of human neutrophils during mechanical confinement inside microfluidic channels. Integr Biol (Camb). (2014) 6:175–83. 10.1039/C3IB40175H
    1. Bystrom J, Evans I, Newson J, Stables M, Toor I, van Rooijen N, et al. . Resolution-phase macrophages possess a unique inflammatory phenotype that is controlled by cAMP. Blood. (2008) 112:4117–27. 10.1182/blood-2007-12-129767
    1. Tauzin S, Starnes TW, Becker FB, Lam P, Huttenlocher A. Redox and Src family kinase signaling control leukocyte wound attraction and neutrophil reverse migration. J Cell Biol. (2014) 207:589–98. 10.1083/jcb.201408090
    1. Serhan CN, Savill J. Resolution of inflammation: the beginning programs the end. Nat Immunol. (2005) 6:1191–7. 10.1038/ni1276
    1. Wright HL, Moots RJ, Edwards SW. The multifactorial role of neutrophils in rheumatoid arthritis. Nat Rev Rheumatol. (2014) 10:593–601. 10.1038/nrrheum.2014.80
    1. Cantin AM, Hartl D, Konstan MW, Chmiel JF. Inflammation in cystic fibrosis lung disease: pathogenesis and therapy. J Cyst Fibros. (2015) 14:419–30. 10.1016/j.jcf.2015.03.003
    1. Woodfin A, Voisin M-B, Beyrau M, Colom B, Caille D, Diapouli F-M, et al. . The junctional adhesion molecule JAM-C regulates polarized transendothelial migration of neutrophils in vivo. Nat Immunol. (2011) 12:761–9. 10.1038/ni.2062
    1. Robertson AL, Holmes GR, Bojarczuk AN, Burgon J, Loynes CA, Chimen M, et al. . A zebrafish compound screen reveals modulation of neutrophil reverse migration as an anti-inflammatory mechanism. Sci Transl Med. (2014) 6:225ra29. 10.1126/scitranslmed.3007672
    1. Thompson AAR, Elks PM, Marriott HM, Eamsamarng S, Higgins KR, Lewis A, et al. . Hypoxia-inducible factor 2α regulates key neutrophil functions in humans, mice, and zebrafish. Blood. (2014) 123:366–76. 10.1182/blood-2013-05-500207
    1. Jin H, Aziz M, Ode Y, Wang P. CIRP induces neutrophil reverse transendothelial migration in sepsis. Shock. (2019) 51:548–56. 10.1097/SHK.0000000000001257
    1. Hirano Y, Aziz M, Wang P. Role of reverse transendothelial migration of neutrophils in inflammation. Biol Chem. (2016) 397:497–506. 10.1515/hsz-2015-0309
    1. Singer M, Deutschman CS, Seymour CW, Shankar-Hari M, Annane D, Bauer M, et al. . The third international consensus definitions for sepsis and septic shock (Sepsis-3). JAMA. (2016) 315:801–10. 10.1001/jama.2016.0287
    1. Seymour CW, Rosengart MR. Septic shock: advances in diagnosis and treatment. JAMA. (2015) 314:708–17. 10.1001/jama.2015.7885
    1. Shen X-F, Cao K, Jiang J-P, Guan W-X, Du J-F. Neutrophil dysregulation during sepsis: an overview and update. J Cell Mol Med. (2017) 21:1687–97. 10.1111/jcmm.13112
    1. Zhang F, Liu A-L, Gao S, Ma S, Guo S-B. Neutrophil dysfunction in sepsis. Chin Med J. (2016) 129:2741–4. 10.4103/0366-6999.193447
    1. Delano MJ, Ward PA. Sepsis-induced immune dysfunction: can immune therapies reduce mortality? J Clin Invest. (2016) 126:23–31. 10.1172/JCI82224
    1. Gotts JE, Matthay MA. Sepsis: pathophysiology and clinical management. BMJ. (2016) 353:i1585. 10.1136/bmj.i1585
    1. Lämmermann T. In the eye of the neutrophil swarm-navigation signals that bring neutrophils together in inflamed and infected tissues. J Leukoc Biol. (2016) 100:55–63. 10.1189/jlb.1MR0915-403
    1. Sadik CD, Luster AD. Lipid-cytokine-chemokine cascades orchestrate leukocyte recruitment in inflammation. J Leukoc Biol. (2012) 91:207–15. 10.1189/jlb.0811402
    1. Ode Y, Aziz M, Wang P. CIRP increases ICAM-1+ phenotype of neutrophils exhibiting elevated iNOS and NETs in sepsis. J Leukoc Biol. (2018) 103:693–707. 10.1002/JLB.3A0817-327RR
    1. Hesse A-K, Dörger M, Kupatt C, Krombach F. Proinflammatory role of inducible nitric oxide synthase in acute hyperoxic lung injury. Respir Res. (2004) 5:11. 10.1186/1465-9921-5-11
    1. Zhang D, Chen G, Manwani D, Mortha A, Xu C, Faith JJ, et al. . Neutrophil ageing is regulated by the microbiome. Nature. (2015) 525:528–32. 10.1038/nature15367
    1. Ng LG, Ostuni R, Hidalgo A. Heterogeneity of neutrophils. Nat Rev Immunol. (2019) 19:255–65. 10.1038/s41577-019-0141-8
    1. Woodfin A, Beyrau M, Voisin M-B, Ma B, Whiteford JR, Hordijk PL, et al. . ICAM-1-expressing neutrophils exhibit enhanced effector functions in murine models of endotoxemia. Blood. (2016) 127:898–907. 10.1182/blood-2015-08-664995

Source: PubMed

3
Suscribir