Elsevier

Current Opinion in Cell Biology

Volume 72, October 2021, Pages 124-130
Current Opinion in Cell Biology

Signal integration in forward and reverse neutrophil migration: Fundamentals and emerging mechanisms

https://doi.org/10.1016/j.ceb.2021.07.002Get rights and content

Highlights

  • Neutrophil reverse migration has emerged as a critical component of inflammation resolution.

  • In wounds, neutrophils integrate proinflammatory recruitment and retention signals and anti-inflammatory dispersal signals..

  • Emerging molecular players and pathways provide mechanistic insights into these complex signaling decisions.

  • A more holistic model of signal integration in migrating cells that encompasses forward and reverse migration is developing.

Abstract

Neutrophils migrate to sites of tissue damage, where they protect the host against pathogens. Often, the cost of these neutrophil defenses is collateral damage to healthy tissues. Thus, the immune system has evolved multiple mechanisms to regulate neutrophil migration. One of these mechanisms is reverse migration — the process whereby neutrophils leave the source of inflammation. In vivo, neutrophils arrive and depart the wound simultaneously — indicating that neutrophils dynamically integrate conflicting signals to engage in forward and reverse migration. This finding is seemingly at odds with the established chemoattractant hierarchy in vitro, which places wound-derived signals at the top. Here we will discuss recent work that has uncovered key players involved in retaining and dispersing neutrophils from wounds. These findings offer the opportunity to integrate established and emerging mechanisms into a holistic model for neutrophil migration in vivo.

Introduction

Neutrophils are often the first immune cells to arrive at sites of injury and infection. There, they mount a potent defense response that must be tightly regulated to limit collateral damage to the host [1]. Indeed, excessive or inappropriate neutrophilic inflammation can tip the balance from host protection to autoimmunity and tissue damage [1]. Neutrophils and other leukocytes exhibit complex decision-making within interstitial tissues in response to various cues to maintain tissue homeostasis and enable tissue repair.

An example of this complex decision-making is the neutrophil response to sterile tissue injury. Until recently, neutrophil recruitment to injury sites was considered unidirectional, with neutrophils undergoing apoptosis and subsequent clearance by macrophages at the wound [2]. However, it is increasingly evident that a subset of neutrophils leave inflamed tissues and re-enter the circulation [3, 4, 5, 6]. The reverse migration and reverse transmigration of neutrophils into the vasculature have now been described in zebrafish, mice, and humans, suggesting they play a critical role in local inflammation resolution [3, 4, 5, 6]. Indeed, in addition to clearing apoptotic neutrophils, macrophages have also been reported to repel wound-associated neutrophils [7,8] or ‘cloak’ sites of sterile damage, thereby limiting chemoattractant signaling and neutrophil inflammation [9•].

The mechanism underlying the prioritization between tissue damage cues and reverse migration is not fully understood. In part, this is due to the daunting complexity of the physical and chemical landscape neutrophils encounter in vivo [10,11]. Furthermore, the signals that regulate reverse migration are not well defined. Recent work in vivo and in vitro has just begun to uncover some of the molecular players and signaling pathways that regulate reverse migration. Here, we discuss how neutrophils prioritize chemoattractant signals in vitro and more complex in vivo contexts and discuss how these mechanisms may provide insight into the complex prioritization needed to reverse migrate and resolve a local response.

Section snippets

Reverse neutrophil migration conundrum: the end is not always the end

Neutrophils are inherently motile cells that migrate randomly (chemokinesis) and directionally (chemotaxis) in response to numerous chemical signals [12, 13, 14]. During episodes of inflammation, a torrent of overlapping directional cues guide neutrophils toward the afflicted tissue (Figure 1a–b) [10,11]. To effectively reach their target, they must integrate and prioritize these signals [15]. Indeed, there is a clear hierarchy of recruitment signals. Early studies in vitro revealed that

Attractant detection systems in neutrophils

Scientists first described neutrophils’ ability to migrate directionally toward chemical attractants in the late 19th century [19]. It would take almost 100 years before we began to understand the complexity and sophistication of their navigational system. In the 1970s, seminal work distinguished neutrophils’ attractant sensing mechanism from that of bacteria [12,20]. Bacteria employ temporal sensing in which chemoattractant concentration is compared over time. When higher concentrations of

Receptor-level regulation of motility in forward and reverse migration

In addition to inherent differences in sensing mechanisms, chemoattractant receptors are also subject to variations in receptor-level regulation. In neutrophils, the vast majority of chemoattractants are detected by G-protein-coupled receptors (GPCRs) [30, 31, 32]. Typically, GPCR phosphorylation results in desensitization and/or receptor endocytosis [14,30, 31, 32]. While most chemoattractant receptors undergo homologous desensitization, end-target signals also impose heterologous

Overcoming retention signals

In vivo, where the chemical and physical landscapes are very complex, it is likely that numerous signaling events and processes coordinate to regulate reverse migration and local resolution. In addition to recruitment signals, neutrophils must also overcome retention signals to leave the wound. Of course, both continued neutrophil recruitment and retention are important until host defenses have gained adequate control of an infection or injury. Thus, when functioning properly, many retention

Lipid signaling in reverse migration

Leukotriene B4 is a well-characterized chemoattractant that has evolved specialized roles in both forward and reverse neutrophil migration. LTB4 is one of many derivatives of arachidonic acid produced and secreted by neutrophils during inflammation [50]. Neutrophil release of LTB4 helps recruit other neutrophils by amplifying chemoattractant gradients and enhancing migration toward fMLF [50,53]. In mice, LTB4 is required for the formation of neutrophil swarms [54].

At the wound, neutrophils

Emerging mechanisms

While neutrophils’ attractant detection systems are sophisticated, there are boundaries within which they can successfully operate. For example, spatial sensing requires an adequately steep gradient (~2–5% difference in ligand concentration across the cell length), and temporal sensing fails when attractant concentrations are no longer rising [12,22,29]. Given the long distances they travel and the reliability with which neutrophils reach their targets in vivo, there must be mechanisms in place

Conclusion

There is a famous adage attributed to Aristotle: ‘The more you learn, the less you know’. Over a century of study on leukocyte migration has produced a wealth of knowledge on the fundamental principles of cellular navigation. Reductionist, in vitro approaches have allowed us to define the basic algorithms that govern signal integration in neutrophils. Through more complex studies, we have come to appreciate that neutrophil intrinsic mechanisms (e.g., dynamic regulation of attractant receptors

Conflict of interest statement

Nothing declared.

Acknowledgements

This work was supported by National Institutes of Health Grants R35 GM118027 to AH and Hematology T32 to HL07899 BRG.

References (68)

  • H.M. Isles et al.

    The CXCL12/CXCR4 signaling Axis retains neutrophils at inflammatory sites in zebrafish

    Front Immunol

    (2019)
  • A.L. Robertson et al.

    A zebrafish compound screen reveals modulation of neutrophil reverse migration as an anti-inflammatory mechanism

    Sci Transl Med

    (2014)
  • W.G. Kaelin et al.

    Oxygen sensing by metazoans: the central role of the HIF hydroxylase pathway

    Mol Cell

    (2008)
  • B.C. Subramanian et al.

    The role of the LTB4-BLT1 axis in chemotactic gradient sensing and directed leukocyte migration

    Semin Immunol

    (2017)
  • T. Lämmermann et al.

    Neutrophil swarms require LTB4 and integrins at sites of cell death in vivo

    Nature

    (2013)
  • B.D. Levy et al.

    Lipid mediator class switching during acute inflammation: signals in resolution

    Nat Immunol

    (2001)
  • C.A. Loynes 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)
  • S. Nourshargh et al.

    Leukocyte migration into inflamed tissues

    Immunity

    (2014)
  • L. Tweedy et al.

    Self-generated chemotactic gradients - cells steering themselves

    Curr Opin Cell Biol

    (2016)
  • L. Tweedy et al.

    Seeing around corners: cells solve mazes and respond at a distance using attractant breakdown

    Science

    (2020)
  • E. Kolaczkowska et al.

    Neutrophil recruitment and function in health and inflammation

    Nat Rev Immunol

    (2013)
  • J. Savill et al.

    Corpse clearance defines the meaning of cell death

    Nature

    (2000)
  • J.R. Mathias et al.

    Resolution of inflammation by retrograde chemotaxis of neutrophils in transgenic zebrafish

    J Leukoc Biol

    (2006)
  • A. Woodfin et al.

    The junctional adhesion molecule JAM-C regulates polarized transendothelial migration of neutrophils in vivo

    Nat Immunol

    (2011)
  • C.D. Buckley 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)
  • B. Hamza et al.

    Retrotaxis of human neutrophils during mechanical confinement inside microfluidic channels

    Integr Biol (United Kingdom)

    (2014)
  • S. Tauzin et al.

    Redox and Src family kinase signaling control leukocyte wound attraction and neutrophil reverse migration

    J Cell Biol

    (2014)
  • S. Uderhardt et al.

    Resident macrophages cloak tissue microlesions to prevent neutrophil-driven inflammatory damage

    Cell

    (2019)
  • M. Sarris et al.

    Navigating in tissue mazes: chemoattractant interpretation in complex environments

    Curr Opin Cell Biol

    (2015)
  • S.H. Zigmond

    Ability of polymorphonuclear leukocytes to orient in gradients of chemotactic factors

    J Cell Biol

    (1977)
  • A.J. Ridley et al.

    Cell migration: integrating signals from front to back

    Science

    (2003)
  • T. Lämmermann et al.

    Concepts of GPCR-controlled navigation in the immune system

    Immunol Rev

    (2019)
  • E.F. Foxman et al.

    Multistep navigation and the combinatorial control of leukocyte chemotaxis

    (1997)
  • B. Heit et al.

    An intracellular signaling hierarchy determines direction of migration in opposing chemotactic gradients

    J Cell Biol

    (2002)
  • View full text