Abstract
Effector T cells leave the lymph nodes armed with specialized functional attributes. Their antigenic targets may be located anywhere in the body, posing the ultimate challenge; how to efficiently identify the target tissue, navigate through a complex tissue matrix and ultimately locate the immunological insult. Recent advances in real time in situ imaging of effector T cell migratory behaviour have revealed a great degree of mechanistic plasticity that enables effector T cells to push and squeeze their way through inflamed tissues. This process is shaped by an array of ‘stop’ and ‘go’ guidance signals including target antigens, chemokines, integrin ligands, and the mechanical cues of the inflamed microenvironment. Effector T cells must sense and interpret these competing signals to correctly position themselves to mediate their effector functions for complete and durable responses in infectious disease and malignancy. Tuning T cell migration therapeutically will require a new understanding of this complex decision-making process.
Introduction
The rapid and targeted response of the immune system to tissue damage is reliant on the incredibly nimble movement of leukocytes between and within tissues. Tissue-specific mobilization depends on the ability of leukocytes to sense directional signals from specialized niches and to quickly respond and adapt to environmental cues and tissue landscapes remodeled by infection and inflammation. T cells surveying tissues move fast, but have the capacity to rapidly arrest upon antigen encounter. This ability for quick decision-making with respect to locomotion has captivated cell biologists, biophysicists and bioengineers alike who have used immune cells as a powerful tool to understand the mechanics of movement within 3D tissues. For immunologists, understanding the regulation of leukocyte migration is central to our ability to control immune function in health and disease.
T cell activation and function is dependent on the efficient scanning of inflamed tissues for the identification of cellular targets. Those targets are often innate immune cells capable of presenting cognate antigen to CD4+ and CD8+ T cells and non-haematopoietic targets in the form of infected, damaged or malignant cells. Optimal strategies to ‘find’ such targets will differ, depending on the location and distribution of targets as well as on the activation state of the T cell (that is, whether a T cell is naïve, effector or memory). Naïve T cells survey the extensive reticular cell network of the lymph node for rare ligand-bearing antigen presenting cells (APCs) during the initiation of an immune response. Meanwhile, effector T cells enter inflamed tissues many days after the initial insult and are met by a tissue microenvironment reshaped by inflammatory cytokines and innate cell recruitment. Within this context, effector T cells must integrate a plethora of extrinsic signals to optimize encounter with APCs for reactivation, and position themselves for precise delivery of effector function, while limiting collateral tissue damage. Since the discovery of lymphocyte recirculation by Gowans, significant progress has been made in understanding the migratory capacity of T cells between tissues and within the confines of the lymph node1,2. This Review focuses on the dynamic behaviour of effector T cells once they enter an inflamed site. We discuss the complexity of T cell movement within inflamed tissues and highlight the unique navigational challenges they face. Understanding spatiotemporal regulation of T cell migration will lead to novel therapies aimed at inhibiting the dysregulated autoimmune and allergic conditions as well as augmenting the host response to tumours, infectious agents and vaccines.
A matter of scale
The use of intravital imaging and new super-resolution imaging modalities has provided an unprecedented window into the leukocyte–tissue interface and has revealed both the requirement, and lack-thereof, of numerous migration cues including chemokines, integrin ligands, extracellular matrix (ECM) components and lipids. These often contradictory findings reflect context-dependent differences in the tissue and inflammatory milieu, and limitations imposed by the level of experimental analysis. Direct imaging of individual immune cell types in their native context provides the most accurate spatiotemporal assessment of immune effector functions, but the complexity of the milieu makes it challenging to parse out individual molecular contributions. Single-cell high-resolution imaging in controlled fabricated settings can define molecular dynamics of movement but such studies lack biological complexity, often testing one or two parameters at a time. Thus, there is an inherent inverse correlation between biological complexity and molecular resolution and a pressing need to bridge these levels to fully understand how T cells position themselves for action (Fig 1). Despite these divisions of scale, there is growing consensus that immune cell migration needs to remain plastic in the face of microanatomical changes in tissue terrain, cellular composition and ‘target’ distribution3–7
Figure 1. The study of T cell migration: a trade-off between molecular resolution and biological complexity.

The tool box for in situ analysis of T cell migration is growing rapidly thanks to innovations in resolution through tissue-clearing techniques and super-resolution imaging modalities. Increasing molecular resolution often results in loss of biological complexity. At one end of the spectrum, the use of photoactivated localization microscopy (PALM) and stochastic optical reconstruction microscopy (STORM) facilitates single molecule analysis in whole cells, enabling the determination of conformational changes in single integrin heterodimers in migrating cells. Use of total internal reflection fluorescence microscopy (TIRF) microscopy has enabled the study of the dynamics of force transmission close (within ~100nm) to the plasma membrane of migrating cells. Real-time 3D analysis of T cell surface topography is possible with light-sheet microscopy. These tools have provided critical insight into molecular mechanism, but cannot be utilized in complex 3D tissues of the living animal. At the other end of the spectrum, advances in 3D histology using tissue clearing techniques and multiplex confocal microscopy has provided an unprecedented look at antigen dispersal and immune cell position in 3D, but the dynamics of T cell migration are lost. In between, the use of intravital multiphoton microscopy provides the ability to visualize T cell migration in real-time in the context of tissue complexity (albeit for relatively short periods of time, hours). Development of photoactivation tools and force sensors to assess molecular details and manipulate signals in individual migrating cells in real time by multiphoton will help to bridge these gaps in scale.
Basic mechanics of 3D movement
Before we discuss context-specific requirements for T cell migration, we will briefly review the basic parameters for cell movement in 3D8,9. T cells migrating in 3D tissues move fast (10–15μm/min), often in a random fashion, and are confined by cellular networks embedded in ECM. Such locomotion requires cell polarization and the transmission of force between the cell and its environment carried out by adhesion molecule transmembrane force coupling (usually integrin-based) or friction with the surface, resulting in forward actin protrusions and actomyosin contraction (Box 1)10. The efficiency of migration is tuned by the degree of force generated and the extent of traction provided. Classically, modes of migration have been separated into two main forms; slow, high adhesive, movement (mesenchymal, highlighted by fibroblasts and cancer cells) and fast, low adhesive, movement (amoeboid, exemplified by immune cells). In practice, modes of migration are fluid, with a seemingly seamless transition between adhesion and non-adhesion-dependent modes11. Movement is therefore the sum of numerous input signals supplied by different signalling pathways in a spatiotemporal fashion (Fig 2).
Box 1. Actomyosin-based 3D migration.
At the molecular level, T cell migration involves applying internally generated force to generate traction against the extracellular environment for forward movement. Cellular polarization and actin retrograde flow is regulated in part by chemokines via GPCR signalling that drives an F-actin-rich leading edge and an acto-myosin rich uropod64. While chemokines can lead to quantitative increases in actin flow, chemokines alone are insufficient to drive forward locomotion, leaving the cells to ‘run in place’52. Tension with the extracellular environment is required for forward motion and can be achieved by active engagement of adhesion receptors such as integrins that act as a clutch to engage the actin motors (See Box 1 figure). Integrin binding to matrix ligands is coupled to the actin cytoskeleton via intracellular adaptor proteins such as kindlin and talin. Integrin engagement requires an activation event in which the integrin undergoes a conformational change from a closed, bent, form to an open, extended, form that has high binding activity51. Chemokine signalling serves to activate integrins by regulating the binding of talin to the integrin cytoplasmic tail and to actin itself. In reciprocal fashion, actin engagement creates tension or ‘pulling’ on the intregin β-chain to stabilize the active integrin conformational state165. For rapid T cell movement in 3D, T cells must balance force generation and adhesion to tune their response to the microenvironment. This appears to be achieved by a relative low integrin mediated force transmission by a seemingly dispersed network of integrins, possibly through highly dynamic micro-adhesive contacts that can provide sufficient friction to enable T cells to rapidly adapt to micro-anatomical terrains without losing momentum. More recently, non-integrin adhesion receptors or a non-receptor friction interface, created by confinement and surface texture or topography, has also been found to create sufficient shear forces to move the cell forwards31.
Figure 2. Mode of migration is shaped by input from multiple signals.

(A) A phase space diagram illustrating the cooperative behaviour of chemoattractants, adhesive ligands and the degree of confinement or tissue topography in promoting or inhibiting cell motility and tissue exploration. The upper diagram is based on a gaussian function with two variables (chemoattractant versus adhesive ligand). The presence of one parameter without the other is unable to support motility. High levels of adhesion or chemoattractant result in T cell arrest by being stuck in place or ‘spinning their wheels’, respectively. The shaded regions indicate migration modes occupied by T cells that favour chemokine or adhesion dependency. The lower diagrams introduce a third variable, confinement, and illustrate how the peak (optimal exploration) moves towards chemokine-based efficiency under high confinement associated with dense ECM (left) and towards adhesion-requirements under low confinement (right). (B) 4D landscape model of 3 variables (X, Y, Z) with a fourth functional dimension of exploration efficiency. In vivo, gaussian functions and simple relations between each variable are unlikely, rather the tuning of T cell exploration in the tissue will be determined by local microdomains resulting in a highly variable navigable landscape. Here x, y, z are hypothetical variables as there is insufficient data to map integrated responses to known guidance cues, but each variable would represent an individual chemokine, integrin ligand or specific mechanical parameter. (C) Intravital multiphoton microscope image of migrating Th1 cells (green) in a dermal collagen network (white, second harmonic generation (SHG) to illustrate the variability of just one visible parameter, physical confinement, in vivo. Although in reality, these cells are integrating signals from multiple ‘hidden’ factors such as chemokines and integrin ligands.
Unlike fibroblasts and tumour cells that use matrix metalloproteinases to breakdown matrix and forge their own path through tissues, most leukocytes do not utilize proteolysis for interstitial migration and therefore their migratory path is constrained by the size of natural channels and pores within the ECM scaffold12. Activated T cells moving in 3D collagen matrices make frequent directional changes in regions of narrow space13, which may explain the random migration patterns of effector T cells in infected tissues as diverse as the brain, lung, liver and skin7,14–17. High-resolution analysis in microfabricated 3D collagen gels suggests that when faced with a directional ‘choice’, T cells distinguish between differently sized pores in the matrix and preferentially migrate along the path of least resistance13,18. Rite of passage will be shaped by the interfibrillar space and the relative deformability of both the matrix fibres and the cell (Box 2)9. The degree of matrix resistance will depend on tissue-specific specialization of the matrix structure and context-dependent inflammatory mediators. T cells are extremely soft cells, with the rigidity of human T cells being lower than that of all myeloid cells tested19. Optimal pore size is regulated by the size and deformability of the cell’s nucleus, which is often 2 to 10 times stiffer than the cell body20,21. In fact, leukocytes migrate with the nucleus at the front of the cell where it appears to serve as a mechanical gauge, with the cell opting for a direction that best fits the ability of the nucleus to protrude into the space18. Neutrophils are particularly adept at quick passage through dense tissues aided by an unusually flexible nucleus that lacks lamin A/C intermediate filaments that usually form a rigid shell underneath the nuclear membrane22–24. Dendritic cells (DCs) optimize nucleus deformation by generating a dense and dynamic perinuclear actin network25. A recent study in T cells highlighted the role of formin-like 1 (FMNL1) in maneuvering of the nucleus through tight spaces26. Upregulation of FMNL1 has been linked to effector T cell trafficking in disease27,28 and may suggest that induced changes in the relative deformability of the nuclear can influence T cell movement within inflamed tissues.
Box 2. Physical constraints.
The interfibrillar space of the extracellular matrix (ECM) influences the direction and mechanisms used for 3D migration and can vary dramatically in different tissues (for example, from loose connective tissue in the spleen, intestinal lamina propria and lymph nodes to extremely dense basement membrane tissue) with spaces ranging between 2 – 30 μm (See Box 2 figure). While it is known that cell speed is a linear function of pore size, in vivo measurement of physical size constraints for 3D migration T cells is limited. For movement into the tissues, the minimum endothelial pore size during T cell transmigration was estimated at 5.1 – 5.4 μm38,166. Below a critical capillary size of approximately 4.5 μm, T cells appear to get trapped or can rupture19,167. * The apparent Young’s modulus Eapp represents a static estimation of the elastic behavior of the material. A human T cell elasticity, or ‘softness’, of ~85 Pa was shown to be considerably lower than that of monocytes (~520 Pa), dendritic cells (~440 Pa) and macrophages (~900 Pa)19.
In vivo, the size of the gaps in the matrix are dynamically regulated by the inflammatory milieu that can narrow or widen the spacing29. Migration in dense 3D tissue microenvironments has been shown to be largely adhesion (or integrin) independent30, (see exception below), utilizing frictional forces to ‘chimney’ between matrix fibres8. A recent in vitro study suggests that in tight confined spaces the topography of the microenvironment itself can support locomotion in the absence of receptor-mediated adhesion. When confined in 3D, T cells failed to migrate on a smooth surface in the absence of integrins, however provision of serrations to the surface was sufficient to support T cell locomotion31. The retrograde flow of actin followed the texture of the substrate and created sufficient shear forces to propel the cell forwards. A new in vivo study of tissue resident memory T (TRM) cells in the salivary glands supports the idea of integrin and chemokine-independent friction-based motility with macrophages providing the necessary topographical surface to support tissue surveillance32. This topographical model would allow T cells to autonomously move seamlessly through varying tissue landscapes regardless of the provision of extrinsic cues.
Guidance cues and their distribution
The 3D movement of leukocytes within inflamed tissues is controlled by the organization of the tissue ECM and the availability and distribution of chemotactic and adhesive signals. As illustrated in Figure 2, the balance between these guidance cues will dictate the most efficient mode of migration. Indeed, a recent study of effector T cell homing via afferent lymphatics nicely demonstrates the interplay between chemokines, integrins and mechanical constraints33. Individual guidance cues and their molecular mechanism of action have been reviewed in detail elsewhere29,34–37. Here we will discuss the ‘presentation’ of these cues within the tissue and how they may modulate effector T cell migration at multiple stages; from exiting the blood stream and traversing the inflamed tissue to identifying antigen-bearing or infected targets (Fig 3).
Figure 3. Regulation of effector T cell migration within inflamed tissues.

Successful tissue immunity requires efficient T cell migration within the inflamed tissue. The migratory path is shaped by multiple guidance cues spatiotemporal displayed in distinct microenvironments. At the tissue level, Effector T cells (green) must undergo extravasation or transendothelial migration from the blood into tissue, cross the basement membrane, ‘search’ the inflamed tissue for antigen-bearing target cells (antigen presenting cell, purple) for peripheral reactivation and exert effector function at foci of infection. This migratory path is influenced (promoted and inhibited) by: the density and composition of the matrix; the multivariate and dynamic display of chemoattractants; effector T cell intrinsic motility programming that pre-sets receptivity to guidance cues and optimizes the ability to ‘search’ for tissue targets; the density and distribution of antigen-bearing targets; pathogen-specific infection niches and the ability to retain T cells for rapid recall.
Chemokine presentation in tissues.
Chemokines play a central role in the determination of tissue-specificity and selectivity during T cell recruitment34. They direct migration by binding to G protein-coupled receptors (GPCRs) that initiate signalling cascades that can activate integrins and regulate remodelling of the actin cytoskeleton35. Initial T cell entry into inflamed tissues, or trans-endothelial migration (TEM), is guided by immobilized chemokines on surfaces of the endothelial cells and the glycocalyx36,38 . In addition, intra-endothelial chemokines stored in intracellular vesicles of the endothelial cells can guide TEM36,38. Immobilization is achieved by binding to sulphonated glycosaminoglycans (GAGs), such as heparan sulphate, that are covalently attached to endothelial cell-associated matrix. GAGs are negatively charged linear polysaccharides with incredible diversity due to differences in their length, composition and patterns of acetylation and sulphation39,40. Their ability to interact with other charged molecules such as chemokines makes them critical regulators of chemoattactant availability and stability that shapes the nature of the chemokine gradient. A steep gradient of heparan sulphate between the apical and basolateral sides of endothelial cells provides a mechanism for patterning of haptotactic gradients of the chemokines released by endothelial cells and pericytes, which can drive directional leukocyte migration during TEM41. It should be noted that immune cells can enter some visceral organs via a non-vascular route42,43 and will likely be regulated by distinct mechanisms of tissue-entry.
Within the tissue, there are a variety of GAGs that are mostly attached to core proteins of the ECM (heparin/ heparin sulphate, chondroitin sulphate/dermatan sulphate, keratan sulphate, and hyaluronic acid)44, although hyaluronan can also be found ‘free’ or non-ECM bound44. The levels of expression of these GAGs are dramatically increased in inflamed tissues, chronic infection and autoimmunity. Mutations in chemokines that reduce their capacity for GAG-binding abrogate leukocyte migration, highlighting the importance of chemokine–GAG interactions45,46. Chemokines likely exist in dynamic equilibrium between soluble and GAG-bound forms and will vary in their oligomerization states depending on the specific GAG interactions39,47. Growing evidence now suggests that chemokine signalling residues that mediate receptor interactions are also involved in GAG binding, indicating that GAG-bound chemokines may be unable to directly activate receptors of migrating leukocytes48. Instead, chemokine-GAG binding may function as a local depot in inflamed tissues, serving to preserve the microanatomical position and longevity of chemokines that would otherwise readily diffuse away from the induction site. How the release of chemokines from GAGs is regulated to inform cell migration is not well understood. During inflammation, GAGs can be released from their proteoglycan backbone by enzymes like heparanase, which may alter the ratio of bound to free chemokine. The newly released soluble chemokines may contribute to the local fine-tuning of chemotaxis to modulate cellular trafficking49, although this has yet to be demonstrated for leukocyte migration.
Spatially distinct integrin ligands and functionally distinct integrin expression.
Lymphocytes express numerous integrins that mediate cell–cell and cell–matrix interactions50. Integrins LFA1 (αLβ1) and VLA4 (α4β1) on effector T cells mediate TEM by binding to ICAM1 and VCAM1, respectively, that are upregulated on the vascular endothelium of inflamed tissues36. Once within the tissue, expression of matrix-binding integrins can facilitate interaction with the ECM and control migration and positioning at the inflamed site (see ECM section below). Specificity is determined by the α and β subunit pairings, and these integrins bind numerous ECM proteins such as laminins (α1β1, α2β1, α3β1, α5β1, αVβ1/β3, α6β1, α7β1), fibrillar collagens (α2β1, α3β1), Arg-Gly-Asp (RGD) motifs contained in many ECM proteins like fibronectin and laminin (α5β1, αVβ3, αVβ5, αIIbβ3) and proteins expressed on the tissue barriers like E-cadherin (αEβ7). Integrin binding avidity can be enhanced by extracellular cues such as chemokine-dependent conformational change and increased ligand-induced clustering51 (Box 1). Large integrin-based focal adhesions appear incompatible with the rapid amoeboid migration displayed by T cells, and is supported by studies showing integrin-independent leukocyte movement in 3D tissues30,31. It is worth noting here that, although possible to move in the absence of integrins, it is unlikely both in normal physiology and pathological conditions that actively migrating leukocytes are not constantly exposed to integrin ligands. How T cells tune their sensitivity to such integrin binding activity in the face of overwhelming ligand and chemokine activation cues is not well understood52. Actively migrating T cells likely form small, highly dynamic focal adhesions that provide just enough friction force to allow cells to rapidly shift back and forth between integrin mediated and non-integrin mediated modes of cell migration to optimize speed and tissue coverage11.
If not essential for interstitial migration per se, integrin ligands provide important localization cues. Crossing the basement membrane is a rate limiting step for leukocyte tissue entry53,54. The composition of the basement membrane varies along the length of post-capillary venules and can dictate tissue entry55, with sites of low laminin α5 being preferred sites of CD4+ T cell entry into the CNS, via T cell expression of α6β1 integrin56,57. The functional importance of T cell interactions (via α6β1 and αVβ1) with distinct laminins was recently highlighted in the mouse experimental autoimmune encephalomyelitis model of multiple sclerosis58. Loss of laminin 511 resulted in enhanced disease severity, that was in part explained by T cell high-affinity integrin interactions with laminin 511 that appears may limit TEM, and an interesting migration-independent role for laminin-511 in limiting the differentiation of pathogenic Th17 cells58. The ‘looser’ fibrillar network of the interstitial matrix is composed of fibrillar type I or III collagen which make up 90% of proteins in tissues, providing physical stability and a scaffold for other ECM proteins and proteoglycans carrying GAG chains29. Analogous to the differential expression of laminin isoforms, the collagen ‘backbone’ is associated with patchy expression of other ECM components, such as fibronectin and vitronectin, and their distribution likely creates preferred paths for interstitial migration and specific sites of tissue retention. For example, vitronectin is specifically upregulated in germinal centers (GCs) of the inflamed lymph node59 and may facilitate the retention of αVβ3+ follicular helper T (TFH) cells in GCs60. In the lung, α1β1 (also known as VLA1) and αEβ7 (also known as CD103) integrins play distinct roles in surveillance and epithelial positioning necessary for the retention of TRM cells61,62.
Physical and mechanical properties of ECM.
The ECM 3D ultrastructure provides a physical scaffold that facilitates or limits access to regions of the inflamed tissue and serves as a dynamic platform for the presentation of chemotactic cues (as discussed above) and growth factors29. Cytokines, such as tumour necrosis factor α (TNFα), interferon-γ (IFNγ) and transforming growth factor β (TGFβ), and proteases released in inflamed tissues modulate ECM density, composition and ‘stiffness’. Thus, migrating leukocytes are faced with a highly variable inflammatory landscape (Fig 2). The technical advances afforded by microfabrication of 3D collagen matrices have facilitated the testing of migrational potential of T cells and have shed light on some of the decisions made by T cells when faced with directional choices13,18. However, our practical knowledge of ECM guidance in vivo is hindered by the inability to visualize the complexity of matrix components in situ in real-time in relation to the migrating T cell. Even basic questions of whether T cells are interfacing directly with the matrix, move over cells attached to the matrix or move between cells are unresolved. Thus, we remain quite ignorant of both the micro- and macro-topography over which T cells migrate in vivo.
Intravital imaging within tissues has revealed that T cells often follow the fibrillar collagen structures illuminated by second harmonic generation (SHG) 17,63, suggesting T cells can use the ECM ultrastructure to guide movement through the tissue. Indeed, in the context of toxoplasma infection, inflammation results in the de-novo generation of fibrillar matrix structures in the brain parenchyma that appear to facilitate the positioning of effector CD8+ T cells at toxoplasma-infection foci63. Depending on the density of the fibre network, and hence the degree of cellular confinement, T cells may migrate between fibres in an adhesion-independent fashion or along fibres requiring adhesion8,64. Indeed, inflammation-induced loosening of the collagen fibres in the dermis necessitated the use of matrix-binding integrins αVβ1 and αVβ3 by effector T helper 1 (Th1) cells for interstitial migration17. While changes to the ECM can facilitate movement within inflamed tissues, matrix changes surrounding solid tumours can have the opposite effect and serve to limit access to the tumour65. Human lung tumours are a striking example, where tumour islets are surrounded by a dense network of fibronectin fibres that constrain migrating T cells to a futile ‘running track’ around the tumour preventing T cell movement into the tumour itself66.
Inflammation can also change the stiffness or elasticity of the tissue matrix impacting the mechanical forces applied to T cells as they migrate and the receptors used to sense such changes, in a process referred to as mechanosensing67. In general, cells tend to migrate toward stiffer environments due to increased traction forces moving from soft to stiff substrates68. The stiffness preferences for migrating leukocytes in 3D in vivo is unclear, in part due to the lack of tools to visualize individual matrix fibre rigidity. Studies using in vitro collagen matrices first showed that T cells can temporarily deform fibres coincident with their migration13 and a similar reversible deformation of fibronectin fibres by migrating T cells was recently observed in vivo69. Interestingly, the interaction between cell and matrix can be reciprocal12 with migrating cells inducing local matrix stiffening, opening up the possibility for guidance along self-fashioned stiffness gradients. Moreover, adhesion-based migration can leverage the elasticity of the matrix to enhance migration speed, the recoil from stretched fibres propelling rapid migration in a mode termed ‘sling-shot migration’70. Whether these alternative forms of mechanical migration are employed by T cells as they move through inflamed sites remains to be tested.
Antigen or chemokine-mediated arrest.
T cell migration in the inflamed tissue is shaped by the balance between positive migratory cues and negative arrest signals71,72; classically thought of as the balance between the strength of T cell receptor (TCR) (‘stop’) and chemokine (‘go’) signals. This competition was elegantly demonstrated in vitro where TCR-induced migratory arrest could be overridden by some, but not all, chemokine signals73. T cell arrest is dependent on LFA1–ICAM-1 mediated adhesion and TCR signalling that results in rapid actin cytoskeletal changes at the leading edge, reorientation of the microtubule organizing centre (MTOC), and the formation of a stable immunological synapse at the interface between the T cell and the APC74. At the molecular level, it remains unclear how stop and go signals are prioritized because both TCRs and chemokine receptors often share common downstream signalling intermediates. Differential sensitivity to negative regulators could be discriminatory, indeed the small GTPase RhoH appears to differentially regulate chemotaxis and TCR signalling, decreasing LFA1 adhesiveness for chemokine-mediated migration but enhancing prolonged contact with APCs75. Similarly, upregulation of negative modulators of T cell signalling such as cytotoxic T lymphocyte antigen 4 (CTLA4) and programmed cell death 1 (PD-1) can counteract TCR-mediated arrest and enhance T cell motility76 77. In contrast to the assignment of a ‘go’ function for chemokines, it is becoming clear that high concentrations of chemokines found near the source can restrict motility of cells by virtue of chemokine receptor desensitization71,78. Such chemokine-mediated arrest appears to be critical for promoting T cell retention at infection foci. Effector CD8+ T cells lacking CXC-chemokine receptor 3 (CXCR3) robustly migrated within the vaccinia virus-infected dermis but failed to stop at the CXCL9-enriched viral foci and showed reduced anti-viral function79.
At inflamed sites, cognate antigen–MHC complexes on the surface of APCs represent a powerful stop signal. Indeed, intravital multiphoton imaging studies often use changes in the arrest coefficient of T cells as a surrogate for T cell–APC interactions80,81. In an in vivo delayed type hypersensitivity (DTH) model, where presentation of antigen could be synchronized, antigen-specific and dose-dependent effector T cell arrest was observed within 1 minute of cognate peptide administration82. High affinity antigen induces T cell deceleration via a process dependent on Ca2+ signals and actin-related protein 2/3 (Arp2/3) activity83. In the absence of high affinity peptide–TCR interactions, T cells appear to adopt a migration mode that is more exploratory with a decrease in speed and frequent directional changes that may enable the integration of signals from multiple APCs or the search for alternative APCs displaying higher affinity ligands.
Lipids mediators and other guidance cues.
The control of T cell migration by bioactive lipids is best known in the context of lymph node (and tissue) exit via lymphatics, where a sphingosine-1-phospate gradient from the lymphatics is sensed by the GPCR sphingosine-1-phospate receptor (S1PR1) expressed on activated T cells84. Inflammation-induced leukotrienes, in particular leukotriene B4 (LTB4), are potent chemoattractants for leukocytes85. In parallel, the high affinity LTB4 receptor, BLT1, is upregulated during T cell differentiation86. These lipid chemoattractants can be produced within minutes and are highly diffusible, thus their availability and tissue coverage may be far more ubiquitous than the targeted production of chemokines in the milieu. A number of new players in immune guidance and positioning are being discovered through the study of lymph node GC dynamics where the positioning and retention of TFH cell and B cells within the GC is critical for successful development of high-affinity antibody secreting cells87. GC organization is guided in part by oxysterol ligands at the follicle border and expression of the GPCR EBI2 by TFH cells and B cells88. GC B cell expression of a cell-bound guidance cue, plexin B2, appears to recruit TFH cells expressing the guidance receptor semaphorin 4C into the GC89. In addition to chemoattractants, several chemorepulsive mechanisms have been identified in regulating TFH cell GC dwell time, including TFH cell upregulation of S1PR290 and GC B cell expression of Ephrin B191. Recently, B cell GC confinement was also shown to be shaped by the localized metabolism of a new glutathione ligand for the Gα13-coupled receptor P2RY8 on B cells that inhibits cell migration92,93. The role that these new guidance cues play in non-lymphoid tissues is unclear, but they may influence the way in which T cells interact with antigen-bearing APCs or how T cells are retained in specific micro-anatomical niches. Uncovering the importance of these alternative guidance cues for T cell retention at sites of infection and chronic inflammation will provide a wealth of new therapeutic targets.
Optimized T cell sensing
Optimum T cell guidance will depend on where and how the T cells mediate their effector functions in the inflamed tissue. CD8+ T cell-mediated cytotoxic function requires direct cell–cell contact, which necessitates precise localization to infected cells. In contrast, CD4+ T cells need to localize sufficiently close to the infection foci to be within the effective range of their secreted cytokines. For TH1 cell-secreted IFNγ, its ability to induce inducible nitric oxide synthase (iNOS) expression in macrophages was estimated to extend some 80 μm from the site of antigen presentation94. The effective range of other key effector molecules, such as Th2 cell-secreted IL-4 and Th17 cell-secreted IL-17, remains to be determined. Thus, distinct CD4+ effector T cell subsets may require individualized strategies to position themselves to best exert their effector function. While extrinsic guidance cues direct T cell migration, it is becoming clear that the motility of distinct T cell subsets is intrinsically tuned to optimize their ability to perform specific functions.
Intrinsic programming of T cell migration.
Early studies described fundamental differences in the way in which CD4+ and CD8+ T cells scan the lymph node for initial activation signals95 and how they migrate in the inflamed skin96. During activation and differentiation in the lymph node, subset-specific induction of chemokine receptors tailor responsiveness to chemokines expressed in particular effector niches within and between tissues97. TH1 cells and CD8+ T cells upregulate CXCR3 and this facilitates their repositioning to lymph node areas in which effector programming and lineage commitment processes are reinforced98–100. Upregulation of CXCR5 by TFH cell precursors similarly facilitates their positioning at the T–B border to receive further signals from B cells to ‘lock in’ the TFH cell programme101. For TH1, TH2 and TH17 cell subsets, expression of distinct chemokine receptors also provide homing biases to respective tissue inflammatory milieus102. These homing biases are imprinted during TH cell differentiation through specific STAT and linage-specific transcription factor driven epigenetic regulation103.
Effector T cells must balance the scope and intensity of exploration at inflamed sites to enable sufficient coverage of a tissue to detect rare targets, and sufficient time spent within each search region to promote productive cell–cell interactions72. Modified random walks, such as the Levy walk14, observed for effector T cells in a variety of tissues are predicted to facilitate this balance; combining intermittent thorough search of a small region (confined) with extensive search of a broad area (periods of straight relocation). Although not well understood at the molecular level, T cell meandering appears to be intrinsically regulated in part by expression of myosin 1G (MYO1G) motors that regulate T cell intrinsic speed and the propensity to turn104. These unconventional myosin motors link actin filaments with the cell membrane and may act as molecular force sensors105. Loss of MYO1G resulted in T cell ballistic motility (faster and straighter) and the covering of more ground, but they were less able to detect rare targets104. The Rho-associated protein kinase ROCK also shapes the T cell meandering pattern, by regulating track straightness106. These findings highlight the functional significance of T cell-intrinsic tuning of the random walk.
More recently, T cell functional programing was shown to also extend to induced expression of differential motility machinery. CD4+ T cell populations (such as TH1, TH2, TH17, and Treg cell subsets) express subset-specific combinations of motility-associated genes107. Functionally, these differences change the requirement for environmental cues and correlate with different ‘search’ patterns within the inflamed tissue. STAT6-dependent upregulation of αVβ3 expression by TH2 cells enables them to migrate through the inflamed dermis independent of GPCR signals. Conversely, TH1 cell differentiation led to lower levels of αVβ3 expression necessitating chemokine/GPCR signalling for interstitial migration107. Subset specific differences in navigation within the inflamed target tissue may reflect differences in the way in which theses subsets deliver effector function. Indeed, in an independent study, tissue-enhanced αVβ3 expression was linked to efficacy of TH17 (but not TH1) cell migration and pathogenicity in the CNS108. Although less well understood for effector CD8+ T cell subsets, central memory CD8+ T cells (TCM), but not effector memory CD8+ T cells (TEM), express glycosyltransferase enzymes that generate functional ligands for E- and P-selectin binding that may promote rapid TCM-specific entry into inflamed tissues109, although this may not be a general rule110. These examples suggest that effector T cells acquire distinct motility set-points during initial activation in the draining lymph node that shape the way in which they subsequently respond to environmental factors within the infected target tissue.
Sensitization and desensitization to extrinsic signals.
Our knowledge of how chemokines are ‘seen’ by T cells has lagged behind our ability to observe their migration in vivo due to the inability to visualize the gradients or measure the dynamics of receptor expression in real time, in situ. Standard genetic perturbation (knockdown, overexpression) is effective at identifying the receptors involved in a phenotype, but are less effective at extracting chemotactic mechanism. The steeply decaying gradients of GAG-associated chemokines only have an impact on directional migration over very short distances, in vivo just 50–75μm from the chemokine source itself78,111. Despite acting over small spatial distances, the directional sensing of this gradient has significant functional advantages for leukocyte positioning112. Increasing chemokine concentrations favour directional persistence and an increase in directional speed78,113–115 allowing for persistent movement towards the chemokine source. At high concentration, chemokines can drive cellular arrest, due to the loss of gradient sensing or ligand-induced receptor internalization71. In this way, chemokines act as both attractants and restrainers of leukocyte motility making them potent positional cues within the inflammatory milieu.
Sensitivity to guidance cues is dynamically tuned at both ligand and receptor levels. This level of migration control is often overlooked in vivo due to limitations in visualizing and manipulating such changes in real-time. Yet, there are many ways in which chemoattractant gradients and the response to those gradients can be actively edited71,116–118. Such dynamic editing falls into three categories: removal or sequestration, post-translation modification (PTM) and/or receptor desensitization (Box 3). Local chemokine availability can be modulated by proteolytic degradation and by a growing number of atypical chemokine receptors (ACKRs) that act as scavenger or decoy receptors119,120. These receptors, expressed by stroma, endothelial cells and immune cells, bind and sequester chemokines and thus play active roles in regulating chemokine bioavailability and shaping the chemokine gradient116. ACKR buffering of local chemokine concentrations may optimize the directional signal-to-noise ratio, ensuring robust cell migration in ‘noisy’ microenvironments121. This system has been co-opted by pathogens, with numerous pathogen-encoded chemokine decoy receptors that delay recruitment or disrupt positioning of critical immune effectors119. Adding to the complexity of chemokine sensing is an array of inflammation-induced PTMs including proteolytic truncation, nitration, citrullination and glycosylation117. These modifications dramatically alter the biological activity of the chemokine, promoting agonist or antagonist functions, modulating GAG-binding efficiency and changing receptor selectivity (Box 3). The impact of PTMs in vivo is as yet unclear, but therapeutic targeting may hold promise; small molecule blockade of CCL2 nitration in the tumour microenvironment augmented CD8+ effector T cell tumour invasion122. The ability to sense chemoattractants is also regulated at the receptor level, with strategies that enhance or desensitize receptivity. Enhanced chemoattractant sensitivity can be achieved by re-positioning or clustering of chemokine receptors to the leading edge or filopodia of migrating cells123, as seen in T cells with CXCR4 and active integrins being localized at the leading edge124. In contrast, both receptor internalization and desensitization of GPCR signalling118,125 play an important role in modulating chemokine sensing71, a process that is tightly regulated by GPCR kinases (GKRs) and by regulators of G protein signalling (RGS) proteins 114,125–127 (Box 3).
Box 3. Dynamic regulation of chemokine sensing.
1). Chemokine removal or sequestration.
The family of atypical chemokine receptors (ACKRs) include ACKR1 (also known as DARC), ACKR2 (also known as D6), ACKR3 (also known as CXCR7) and ACKR4 (also known as CCX-CKR)119. These receptors, which are often expressed by endothelium and immune cells, bind chemokines but do not couple to G proteins for GPCR signalling for cell migration. In part, ligand-dependent endocytosis targets the chemokine for degradation116. The scavenger receptor CXCR7 acts to buffer local chemokine concentrations by matching the attractant concentration to the receptor Kd to optimize the directional signal-to-noise ratio121. Models of ACKR genetic ablation result in overt T cell accumulation in peripheral tissues120.
2). Post-translational modifications (PTMs).
Inflammation-induced PTMs of chemoattractants include proteolytic truncation, nitration, citrullination and glycosylation. Truncation at the NH2 or COOH-terminus increases, decreases potency or changes receptor specificity. Examples include: CXCL8 and CXCL5 truncation by matrix metalloproteinases (MMPs), which increases potency of neutrophil chemotaxis168; CXCL10 and CXCL11 truncation by CD26 or CD13, which inactivates chemokine-mediated T cell chemotaxis and recruitment169–171, antagonizing CXCR3 and disrupting GAG binding; CCL5 truncation, which alters receptor specificity, abolishing CCR1 and CCR3 binding but maintaining CCR5 binding172. Nitration alters affinity for GPCRs and GAGs: CCL2 nitration reduces affinity for both the chemokine receptor and GAGs173 and blocks CD8+ T cell recruitment to tumours122. Citrullination alters the charge and interactions with GPCRs, GAGs and lipid: CXCL10 and CXCL11 citrullination reduces potency of CXCR3 signalling and reduces its capacity to bind GAGs, attenuating T cell migration174. Receptor modification also alter function: polysialylation of CCR7 regulates DC migration by enhancing CCL21 activity175 and TLR-induced upregulation of dihydroceramide leads to ceramide-dependent inversion of the topography of CCR5 in the ER of macrophages, preventing their migration towards CCL5176.
3). Receptor desensitization.
Ligand-induced receptor internalization is regulated by: clathrin/β-arrestin mediated internalization for degradation or recycling118; GPCR phosphorylation by GPCR kinases (GRKs) which can couple the GPCR to arrestins, resulting in steric inhibition of receptor interactions with G proteins114,125,126; and by regulators of G protein signaling (RGS) proteins that accelerate the activity of G protein GTPases127.
Signal prioritization.
As outlined in the preceding sections of this review, T cells entering an inflamed site are met with a dizzying array of physical and chemical cues that are dynamically regulated by the inflammatory milieu (Fig 4). Coupled with tremendous redundancy in both receptor and ligand systems (for both chemoattractants and integrins)128 our knowledge of how T cells prioritize spatiotemporal signals in complex environments in vivo remains understandably anecdotal. There is clearly a hierarchical response to competing chemokine gradients, physical guidance cues and arrest signals. At the molecular level, this can be established by competing downstream signaling129,130 and the rate of receptor desensitization131,132. Differential receptor desensitization can impose ligand biases133 that may also impose a spatial bias134. Live imaging of receptor internalization in vivo, revealed differential internalization of CXCR1 and CXCR2 by neutrophils in response to CXCL8 ligand that prevented overt neutrophil ‘congregation’ at the wound site134. A nascent understanding of crosstalk between receptors of different guidance cues is emerging with examples of chemokines regulating integrin ligand specificity135 and integrin expression levels modulating the dependency on chemokine signalling107. How physical cues are prioritized is not well understood, save for the physical and mechanical restrictions previously discussed, but there was an interesting study that showed that local hydraulic pressure (introduced by inflammation-driven oedema) could override chemotactic cues for directional migration136. The above mechanisms of dynamic tuning of sensitivity to environmental cues should enable T cells to dynamically regulate speed and directionality ‘on the fly’ and to integrate and prioritize such heterogeneous signals.
Figure 4. Spatiotemporal optimization of effector T cell positioning.

A summary of the control points that build an efficient effector T cell response. The complexity of external cues and T cell receptivity to these cues is increased by the inflammatory milieu and by T cell differentiation, but can be honed at the tissue site by spatiotemporal mechanisms that appear to amplify the target to optimize effector T cell-specific positioning. Receptivity is enhanced as T cells differentiate from naive to effector T cells (for example, Th1, Th2, Th17 cells) in the lymph node where intrinsic programming drives expression of receptor and signaling machinery that promotes biased receptivity to guidance cues at the inflamed tissue. Once the effector T cells enter the inflamed tissue, a sharp increase in complexity of external cues at the tissue site is driven by a host of competing guidance cues (indicated by the shaded gray boxes) that promote or restrict the ability of effector T cells to ‘search’ the inflamed tissue. This complexity is functionally simplified by non-random spatial clustering of antigen and guidance cues within the inflamed/infected tissue, that appears to amplify the target in an effector T cell-specific way, by reducing the scope of the tissue search and promoting local retention.
Amplifying guidance cues for T cell positioning
The ‘prize’ for successful integration of these overlapping guidance cues is the correct positioning of effector T cells for delivery of effector function and their timely dispersal for the resolution of inflammation. Key to success is the ability to optimize the detection of ‘targets’ (ligand-bearing APCs, infected or damaged cells) within a crowded cellular microenvironment. Lessons learnt from studies of patterns of foraging for food, suggest that the distribution of the targets within inflamed tissues will dictate successful strategies137. In ant models, the distribution of targets appears to dictate the necessity for ant-to-ant communication; patchy or clustered resources requiring cooperative signalling between foragers via pheromones that increases the ease and completeness of the search. While it has largely been assumed that antigen encounter is a relatively rare event at infection sites80,138, the observed clustering of T cells and APCs at sites of inflammation may serve to amplify the target for ease of search (Fig. 4).
Cellular collaboration.
For neutrophils, well-orchestrated spatial and temporal waves of attractants help guide precise positioning to sites of tissue damage. End-target attractants originating from damaged/dying cells or bacteria provide precise microanatomical coordinates, while secondary attractants produced by endothelial cells or other immune cells at the inflamed site promote recruitment and migration within the tissue116. Remarkably, the relay of these signals can be autonomously controlled, with the ‘hand off’ of signals between neutrophils occurring as they migrate by secretion of LTB4 at the rear of one migrating cell promoting the migration of the neutrophil next in line139,140. This relay system substantially increases the recruitment range of cells and supports directionality over long distances116. For T cells, the relay of signals requires collaboration between immune cell types, shaped by resident or recruited innate cells entering the tissue hours to days before the arrival of the T cells themselves. Indeed, neutrophils can leave long-lasting membranous trails containing CXCL12 that inform CD8+ T cell migration paths in the influenza-infected airway15. More recently, dying neutrophils appear to ‘hand off’ signals to infiltrating inflammatory monocytes that optimizes CD8+ T cell activation for anti-viral activity141.
Akin to end-target chemokines of neutrophils, it’s becoming increasingly clear that chemokine production by APCs, or co-clustering of antigen-bearing APC with chemokine-producing inflammatory monocytes or stroma, provides a powerful ‘end-target’ positioning cue for T cells at all functional stages. Production of CCL3 and CCL4 by antigen-specific CD4+ T cell-DC clusters in the lymph node makes for a more attractive target for initial CD8+ T cell activation, enhancing the CCR5+ CD8+ T cell-DC ‘hit rate’ and promoting CD8+ T cell memory113. TH cell differentiation is optimized by positioning of activated T cells at peri- or inter-follicular regions of the lymph node, dependent on CXCL9+ stroma and CXCL10+ DCs for TH1 cells100 and on CXCL13 and CXCL5+ DCs for TH2 cells142. In infected or inflamed tissues, the CXCR3 ligands CXCL9, CXCL10 and CXCL11 are produced by DCs, macrophages and nonhematopoietic cells and also guide sub-anatomical positioning to targets14,79,143,144. This critical protective mechanism can be amplified by an IFNγ-dependent positive feedback loop from strategically positioned effector T cells145 and is often actively subverted by pathogens to evade detection. Moreover, at the memory stage, rapid CXCR3-dependent relocation of central memory CD8+ T cells within the lymph node boosts early anti-viral recall responsiveness146,147.
Specialized tissue niches.
While the individual migratory paths of T cells are inherently random, recent findings suggest that encounters with antigen are far from arbitrary. Pre-positioned niches in the steady state optimize antigen encounter and ‘pop up’ activation niches in inflamed tissues provide hubs for signal integration. In the lymph node, strategic positioning of macrophages and DCs (in particular cDC2s) at the lymphatic sinus or subcapsular sinus floor facilitates the sampling of lymph-borne particulate materials coming from peripheral tissues and facilitates early activation of B cell and T cells, respectively148–150. Rapid redeployment of lymph node-resident DCs to these sites may also potentiate the activation niche151. More recently, naive CD4+ T cells were shown to have a similar steady-state spatial preference at the periphery of the lymph node paracortex regulated by the expression of the GPCR Ebi2152. This ‘anatomical platform’ appears to optimize naïve CD4+ T cell encounters with MHC class II+APCs for early activation. In many non-lymphoid tissues, a similar steady-state strategic positioning of resident lymphoid cells, such as innate lymphoid cells (ILCs), appears to facilitate early immune responses to damage or infection. ILC niches have been found in multiple organs including the barrier tissues of the skin, lung and gut and are composed of colocalized tissue stroma, peripheral nerves, resident innate immune cell types and regulatory T cells153. Crosstalk between cell types in the ILC niche potentiates or regulates initial immune activation154–156. The perivascular adventitial cuff has also emerged as an interesting microanatomical site where tissue-derived signals (TLR ligands, antigen, cytokines) carried in the draining interstitial fluid meet resident DCs and ILCs poised to respond157. Disrupting the adventitial stromal cells in this niche impaired helminth-induced ILC2 expansion and Th2 cell accumulation and function154. Whether these regional hubs designed for early innate immune sensing also provide a platform for the subsequent re-activation of incoming effector T cells is unclear at present. Nonetheless, these hubs, enriched in APCs and chemokines, are scattered throughout the tissue and could represent important tissue “outposts” for optimized presentation of antigens to newly recruited effector T cells157.
The widely observed clustering of effector T cells with APCs in inflamed peripheral tissues in human disease and mouse models suggests that spatial preferences for T cell activation also exist at inflamed and infected sites80,158–160. These activation clusters may simply mark the location of the pathogen itself, or represent induced hubs for peripheral re-activation. Indeed, the perivascular clustering of macrophages, DCs and effector CD8+ T cells was shown to be essential for peripheral activation of T cells in the inflamed skin during contact hypersensitivity161. These perivascular clusters appear to represent de novo generated activation platforms, induced by IL-1α-triggered macrophage production of CXCL12 that initiates DC clustering. Similarly, local CD8+ T cell expansion in the liver following acute viral infection was shown to occur in myeloid-cell aggregates induced by TNF162. The guidance cues required for effector T cells to accumulate within the perivascular clusters are unclear. T cells could use the clusters as sites of preferred tissue entry or be recruited to the site following entry elsewhere. Given their perivascular location, such specialized niches may reduce the scope of tissue needed to be searched by T cells early on in the immune response and could serve as initial local sorting houses, optimizing the peripheral expansion of ‘useful’ antigen-specific effector T cells. The spatial relationship between such activation clusters and subsequent delivery of effector function at infection foci remains to be determined. The functional advantage to non-random spatial distributions within tissues is perhaps best demonstrated by the strategically positioned TRM cells. At some mucosal tissues, TRM cells are maintained within tight clusters163,164, defined in part by chemokine-rich myeloid niches164. Due to their proximity to the mucosal surface, these memory hubs appear to serve as a local ‘immediate response centres’ for early detection of invading pathogens.
Closing remarks
Remarkable progress has been made in recent years on the mechanics of T cell migration at the single cell level and on defining the complexity of modulating factors in the inflammatory milieu that may shape migratory paths of effector T cells in target tissues. These studies have provided insight into the physiologic and pathologic migration patterns of T cells and their role in health and disease. Recent examples of strategic pre-positioning and induced micro-anatomical niches for immune activation at inflamed sites, provide a new conceptual framework for understanding how migration and positioning of effector T cells is optimized, and should help shape a more ‘surgical’ approach for site-specific intervention strategies.
Advanced imaging has enabled the sub-cellular analysis of molecular pathways as T cells migrate and facilitated the identification of sub-anatomical niches within whole tissues. Yet, the details of the molecular presentation patterns of T cell migration signals have not been clearly determined in intact tissues. How are micro- or macro-chemokine gradients established and maintained at sites of tissue infection? Does specific integrin ligand expression have any role to play in the ability of T cells to home and stay at the specific site of the tissue injury? How do effector T cells rapidly integrate the diverse and often redundant local mechanical signals with chemoattractant signals for migration within inflamed tissues? How does T cell intrinsic programming impact on this decision? Layered on top of these questions regarding basic mechanisms of migration in situ, are spatial and temporal considerations. We remain ill-informed regarding points of T cell tissue entry relative to target distribution and thus are unclear as to the range of tissue explored by incoming effector T cells. Furthermore, while the need for T cell peripheral reactivation by APCs is clear, the position of these reactivation events relative to the location at which effector function needs to be delivered is poorly understood. Current immunotherapies for the treatment of various cancers have exposed some of our ‘blind spots’ when it comes to understanding how effector T cells access and position themselves within inflamed tissues. The clinical efficacy of cancer immunotherapy strongly correlates with the number of T cells that infiltrate the tumour microenvironment, but the monitoring of chimeric antigen receptor (CAR) T cells after adoptive cell transfer has shown that only ~1% of the total transferred T cells initially migrate into the tumour. Therefore, it appears we have some ways to go before we can rationally exploit T cell migration strategies for clinical benefit.
In conclusion, our conventional understanding of T cell migration based on individual modules (chemoattractants, integrin activation and biophysical confinement) is far too simplistic. While there are numerous new and creative therapeutic strategies based on targeting T cell migration, success thus far has been modest (Box 4). T cell migration in vivo faces complex challenges from both cell-intrinsic factors that can directly modulate migration signals and cell-extrinsic cues arising from the inflammatory microenvironment. Specificity and flexibility of T cell migration is essential at multiple stages: successful pathogen clearance, prompt resolution of inflammation and the establishment of locally positioned memory potential, all while minimizing collateral tissue damage. A better understanding of the combinatorial biochemical decision making that occurs in support of T cell migration and of the local guidance cues that recruit and retain effector T cells in the correct sub-anatomical location will open up new therapeutic possibilities for inflammatory diseases and enhance the effectiveness of existing clinical approaches.
Box 4. Therapeutics targeting effector T cell migration.
1). Blocking tissue entry to mitigate inflammation.
Highly activated pro-inflammatory effector T cell migration is involved in a wide range of pathologic conditions, including vasculitis, atherosclerosis, stroke, rheumatoid arthritis, lupus, multiple sclerosis and Crohn’s disease177–179. Thus, T cell recruitment receptors (such as integrins and chemokines) have been key targets for novel therapies aimed at inhibiting the dysregulated immune response in autoimmune and allergic conditions.
Integrins.
Four integrins expressed exclusively in leukocytes (αLβ2, α4β1, α4β7, and αEβ7) are involved in the recruitment and retention of effector T cells at many tissue sites during inflammation. Natalizumab: a humanized antibody to α4 integrins, was shown to alleviate multiple sclerosis, but was withdrawn after seeing a few fatal cases of progressive multi-focal leukoencephalopathy (PML) likely due to an iatrogenic immune-defect by blocking α4 integrins that would be permissive for reactivation of a latent human polyoma virus 2 (also known as JC virus) infection180,181. Vedolizumab: selectively blocks α4β7 integrin and showed a significant clinical benefit in the treatment of inflammatory bowel diseases. Efalizumab: targets the integrin αL subunit and was originally developed for psoriasis, then was withdrawn in 2009 because of an association with PML. Etrolizumab: a humanized monoclonal antibody developed to selectively block the b7 integrins (a4b7, and aEb7). Etrolizumab met its primary endpoint of inducing remission versus placebo for patients with ulcerative colitis in a few clinical studies. A phase III study in Crohn’s disease is currently ongoing.
Chemokines.
The chemokine family has attracted great interest for treatment of inflammatory diseases, as it comprises major players in the determination of the tissue-specificity and selectivity during T cell recruitment. However, although G-protein-coupled receptors (GPCRs) represent one of the most successful targets for the drug discovery, intense efforts for several chemokine receptor inhibitors have failed to deliver fruitful drug candidates. Until now, chemokine receptor antagonists have only been clinically approved as antiviral agents (for example, CCR5 and CXCR4 antagonists for HIV). AMG487 is a potent CXCR3 antagonist developed to treat psoriasis. However, a Phase II efficacy and safety study with AMG487 later has been terminated due to the lack of efficacy.
2). Recruitment to tumours for T cell immunotherapy.
Many types of tumors can actively prevent T cell infiltration by modifying the expression of adhesion molecules in vascular endothelial cells182 or inducing posttranslational modification of local chemokine signals122. Several recent studies proposed novel molecular checkpoints that could be harnessed to improve homing of T cells to their target sites, thus, to increase the efficacy of T cell immunotherapy, while reducing the risks associated with non-specific cytotoxicity. For example, an oncolytic vaccinia virus engineered to produce chemokine CXCL11 increased the chemokine expression level within tumor sites, and successfully recruited T cells and augmented anti-tumor efficacy in a mouse preclinical study183. A CXCL10-loaded folate-modified chitosan nanoparticle showed anti-tumor T cell responses and reduced the growth of hepatocellular carcinoma in mice184. Targeted delivery of tumor necrosis factor superfamily member, LIGHT, eradicated established tumors in mice by activating lymphotoxin-β receptor and facilitating local production of CCL21 and CXCL13 that led CTLs into the tumor site (Tang Cancer Cell 2016). Epigenetic reprograming of the production of chemokines CXCL9 and CXCL10 has been attributed to poor T cell infiltration to the tumor microenvironment185. GSK126, a selective inhibitor of EZH2 methyltransferase activity, and DZNep, an all S-adenosyl-methionine-dependent enzymes inhibitor increased production of T cell trafficking chemokines and augmented the therapeutic effects of PD-L1 blockade. In a pancreatic cancer mouse model, AMD3100, a CXCR4 small-molecule inhibitor led to improved T cell migration to the tumor site and enhanced the anti-tumor effects of an anti-PD-L1186. Similarly, LY2510924, a potent and selective peptide antagonist of CXCR4, resulted in selective reduction of intratumor Treg cells and showed a synergistic effect with PD-1 inhibitors in a syngeneic squamous cell carcinoma model187. Based on these pre-clinical data, an open-label phase Ia study testing the safety of combination therapy with LY2510924 and the anti-PD-L1 antibody durvalumab in patients with advanced refractory solid tumors was recently completed188. Although the intratumoral delivery of a chemokine-encoding system has been proposed to enhance CTL homing to the tumor site in several pre-clinical studies, it is important note that chemokines directly contribute to tumor growth, metastasis, and angiogenesis189–191. Thus, it is critical to selectively and precisely control chemokine signals only in the tumor-targeting T cells to avoid potential adverse outcomes.
3). Boosting tissue resident T cells to promote vaccine efficacy.
Precise control of T cell migration and retention is a key to the development of effective vaccines as well as treatment of immune-mediated diseases. As we discussed above, direct applications of exogenous chemokine signals have been proposed to recruit a specific subtype of effector T cells into target tissues192. The functional importance of tissue resident memory T (TRM) cells has driven a novel immunization approach designed to ‘prime and pull’ T cells into tissues193. This technique combines two steps: conventional parenteral vaccination (prime) and topical administration of chemokines or adjuvants to recruit activated T cells to target tissues (pull)194,195. Recently, additional ‘prime and target’ approach utilizing an organ specific antigen-expressing system provided similar proof-of-principle results196, suggesting that protective TRM cells can be generated through this promising tissue-specific T cell recruitment strategy.
Supplementary Material
Acknowledgements
The authors thank members of the “Tissue Regulation of T cell Function” P01 for their input, Patrick Oakes for discussion and design of the graphics in Figure 2, and Ronen Alon for his careful review of the manuscript.
Footnotes
Competing interests
The authors declare no competing interests.
Peer review information
Nature Reviews Immunology thanks J. Schenkel and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.
REFERENCES
- 1.Masopust D & Schenkel JM The integration of T cell migration, differentiation and function. Nat Rev Immunol 13, 309–320, doi: 10.1038/nri3442 (2013). [DOI] [PubMed] [Google Scholar]
- 2.Schulz O, Hammerschmidt SI, Moschovakis GL & Forster R Chemokines and Chemokine Receptors in Lymphoid Tissue Dynamics. Annu Rev Immunol 34, 203–242, doi: 10.1146/annurev-immunol-041015-055649 (2016). [DOI] [PubMed] [Google Scholar]
- 3.Te Boekhorst V, Preziosi L & Friedl P Plasticity of Cell Migration In Vivo and In Silico. Annu Rev Cell Dev Biol 32, 491–526, doi: 10.1146/annurev-cellbio-111315-125201 (2016). [DOI] [PubMed] [Google Scholar]
- 4.Friedl P & Wolf K Plasticity of cell migration: a multiscale tuning model. J Cell Biol 188, 11–19, doi:jcb.200909003 [pii] 10.1083/jcb.200909003 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Vargas P, Barbier L, Saez PJ & Piel M Mechanisms for fast cell migration in complex environments. Curr Opin Cell Biol 48, 72–78, doi: 10.1016/j.ceb.2017.04.007 (2017). [DOI] [PubMed] [Google Scholar]
- 6.Lam PY & Huttenlocher A Interstitial leukocyte migration in vivo. Curr Opin Cell Biol 25, 650–658, doi: 10.1016/j.ceb.2013.05.007 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Lammermann T & Germain RN The multiple faces of leukocyte interstitial migration. Semin Immunopathol 36, 227–251, doi: 10.1007/s00281-014-0418-8 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Yamada KM & Sixt M Mechanisms of 3D cell migration. Nat Rev Mol Cell Biol 20, 738–752, doi: 10.1038/s41580-019-0172-9 (2019). [DOI] [PubMed] [Google Scholar]
- 9.Kameritsch P & Renkawitz J Principles of Leukocyte Migration Strategies. Trends Cell Biol, doi: 10.1016/j.tcb.2020.06.007 (2020). [DOI] [PubMed] [Google Scholar]
- 10.Huse M Mechanical forces in the immune system. Nat Rev Immunol 17, 679–690, doi: 10.1038/nri.2017.74 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Renkawitz J et al. Adaptive force transmission in amoeboid cell migration. Nat Cell Biol 11, 1438–1443, doi:ncb1992 [pii] 10.1038/ncb1992 (2009). [DOI] [PubMed] [Google Scholar]
- 12.van Helvert S, Storm C & Friedl P Mechanoreciprocity in cell migration. Nat Cell Biol 20, 8–20, doi: 10.1038/s41556-017-0012-0 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Wolf K, Muller R, Borgmann S, Brocker EB & Friedl P Amoeboid shape change and contact guidance: T-lymphocyte crawling through fibrillar collagen is independent of matrix remodeling by MMPs and other proteases. Blood 102, 3262–3269, doi: 10.1182/blood-2002-12-3791 (2003). [DOI] [PubMed] [Google Scholar]
- 14. Harris TH et al. Generalized Levy walks and the role of chemokines in migration of effector CD8+ T cells. Nature 486, 545–548, doi: 10.1038/nature11098 (2012). The first paper to suggest the random interstitial migration pattern of T cells resembles a Levy walk, a pattern thought to increase the chances of finding rare targets.
- 15. Lim K et al. Neutrophil trails guide influenza-specific CD8(+) T cells in the airways. Science 349, aaa4352, doi: 10.1126/science.aaa4352 (2015). This study demonstrated that neutrophils can leave chemokine-rich trails that guide virus-specific CD8+ T cell migration in the infected airway.
- 16.Egen JG et al. Macrophage and T cell dynamics during the development and disintegration of mycobacterial granulomas. Immunity 28, 271–284, doi:S1074–7613(08)00028–9 [pii] 10.1016/j.immuni.2007.12.010 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Overstreet MG et al. Inflammation-induced interstitial migration of effector CD4(+) T cells is dependent on integrin alphaV. Nat Immunol 14, 949–958, doi: 10.1038/ni.2682 (2013). This paper identifies the matrix-binding integrin αV as critical for CD4+ T cell interstital migration within inflammed skin.
- 18. Renkawitz J et al. Nuclear positioning facilitates amoeboid migration along the path of least resistance. Nature 568, 546–550, doi: 10.1038/s41586-019-1087-5 (2019). The study shows that the “nucleus-first configuration”, a distinctive feature of amoeboid cells, facilitates rapid navigation of DC along the path of least resistance.
- 19.Bufi N et al. Human Primary Immune Cells Exhibit Distinct Mechanical Properties that Are Modified by Inflammation. Biophys J 108, 2181–2190, doi: 10.1016/j.bpj.2015.03.047 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.McGregor AL, Hsia CR & Lammerding J Squish and squeeze-the nucleus as a physical barrier during migration in confined environments. Curr Opin Cell Biol 40, 32–40, doi: 10.1016/j.ceb.2016.01.011 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Davidson PM, Denais C, Bakshi MC & Lammerding J Nuclear deformability constitutes a rate-limiting step during cell migration in 3-D environments. Cell Mol Bioeng 7, 293–306, doi: 10.1007/s12195-014-0342-y (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Salvermoser M, Begandt D, Alon R & Walzog B Nuclear Deformation During Neutrophil Migration at Sites of Inflammation. Front Immunol 9, 2680, doi: 10.3389/fimmu.2018.02680 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Olins AL et al. The human granulocyte nucleus: Unusual nuclear envelope and heterochromatin composition. Eur J Cell Biol 87, 279–290, doi: 10.1016/j.ejcb.2008.02.007 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Salvermoser M et al. Myosin 1f is specifically required for neutrophil migration in 3D environments during acute inflammation. Blood 131, 1887–1898, doi: 10.1182/blood-2017-10-811851 (2018). [DOI] [PubMed] [Google Scholar]
- 25.Thiam HR et al. Perinuclear Arp2/3-driven actin polymerization enables nuclear deformation to facilitate cell migration through complex environments. Nat Commun 7, 10997, doi: 10.1038/ncomms10997 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Thompson SB et al. Formin-like 1 mediates effector T cell trafficking to inflammatory sites to enable T cell-mediated autoimmunity. Elife 9, doi: 10.7554/eLife.58046 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Degroote RL et al. Formin like 1 expression is increased on CD4+ T lymphocytes in spontaneous autoimmune uveitis. J Proteomics 154, 102–108, doi: 10.1016/j.jprot.2016.12.015 (2017). [DOI] [PubMed] [Google Scholar]
- 28.Odoardi F et al. T cells become licensed in the lung to enter the central nervous system. Nature 488, 675–679, doi: 10.1038/nature11337 (2012). [DOI] [PubMed] [Google Scholar]
- 29.Sorokin L The impact of the extracellular matrix on inflammation. Nat Rev Immunol 10, 712–723, doi:nri2852 [pii] 10.1038/nri2852 (2010). [DOI] [PubMed] [Google Scholar]
- 30.Lammermann T et al. Rapid leukocyte migration by integrin-independent flowing and squeezing. Nature 453, 51–55, doi:nature06887 [pii] 10.1038/nature06887 (2008). [DOI] [PubMed] [Google Scholar]
- 31. Reversat A et al. Cellular locomotion using environmental topography. Nature 582, 582–585, doi: 10.1038/s41586-020-2283-z (2020). The first demonstration that T cells can transmit forces by coupling the retrograde flow of actin to a geometrically irregular environment, in the complete absence of any adhesion.
- 32.Stolp B et al. Salivary gland macrophages and tissue-resident CD8(+) T cells cooperate for homeostatic organ surveillance. Sci Immunol 5, doi: 10.1126/sciimmunol.aaz4371 (2020). [DOI] [PubMed] [Google Scholar]
- 33.Martens R et al. Efficient homing of T cells via afferent lymphatics requires mechanical arrest and integrin-supported chemokine guidance. Nat Commun 11, 1114, doi: 10.1038/s41467-020-14921-w (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Griffith JW, Sokol CL & Luster AD Chemokines and chemokine receptors: positioning cells for host defense and immunity. Annu Rev Immunol 32, 659–702, doi: 10.1146/annurev-immunol-032713-120145 (2014). [DOI] [PubMed] [Google Scholar]
- 35.Lammermann T & Kastenmuller W Concepts of GPCR-controlled navigation in the immune system. Immunol Rev 289, 205–231, doi: 10.1111/imr.12752 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Nourshargh S & Alon R Leukocyte migration into inflamed tissues. Immunity 41, 694–707, doi: 10.1016/j.immuni.2014.10.008 (2014). [DOI] [PubMed] [Google Scholar]
- 37.Hogg N, Patzak I & Willenbrock F The insider’s guide to leukocyte integrin signalling and function. Nat Rev Immunol 11, 416–426, doi: 10.1038/nri2986 (2011). [DOI] [PubMed] [Google Scholar]
- 38.Shulman Z et al. Transendothelial migration of lymphocytes mediated by intraendothelial vesicle stores rather than by extracellular chemokine depots. Nat Immunol 13, 67–76, doi: 10.1038/ni.2173 (2011). [DOI] [PubMed] [Google Scholar]
- 39.Handel TM, Johnson Z, Crown SE, Lau EK & Proudfoot AE Regulation of protein function by glycosaminoglycans--as exemplified by chemokines. Annu Rev Biochem 74, 385–410, doi: 10.1146/annurev.biochem.72.121801.161747 (2005). [DOI] [PubMed] [Google Scholar]
- 40.Mihov D & Spiess M Glycosaminoglycans: Sorting determinants in intracellular protein traffic. Int J Biochem Cell Biol 68, 87–91, doi: 10.1016/j.biocel.2015.08.019 (2015). [DOI] [PubMed] [Google Scholar]
- 41.Stoler-Barak L et al. Blood vessels pattern heparan sulfate gradients between their apical and basolateral aspects. PLoS One 9, e85699, doi: 10.1371/journal.pone.0085699 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Wang J & Kubes P A Reservoir of Mature Cavity Macrophages that Can Rapidly Invade Visceral Organs to Affect Tissue Repair. Cell 165, 668–678, doi: 10.1016/j.cell.2016.03.009 (2016). [DOI] [PubMed] [Google Scholar]
- 43.Steinert EM et al. Cutting Edge: Evidence for Nonvascular Route of Visceral Organ Immunosurveillance by T Cells. J Immunol 201, 337–342, doi: 10.4049/jimmunol.1800279 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Thompson S et al. Regulation of Chemokine Function: The Roles of GAG-Binding and Post-Translational Nitration. Int J Mol Sci 18, doi: 10.3390/ijms18081692 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Proudfoot AE et al. Glycosaminoglycan binding and oligomerization are essential for the in vivo activity of certain chemokines. Proc Natl Acad Sci U S A 100, 1885–1890, doi: 10.1073/pnas.0334864100 (2003). This paper demonstrates that GAG binding mediates chemokine tethering and immobilization, which is a prerequisite of chemokine activation.
- 46.Gangavarapu P et al. The monomer-dimer equilibrium and glycosaminoglycan interactions of chemokine CXCL8 regulate tissue-specific neutrophil recruitment. J Leukoc Biol 91, 259–265, doi: 10.1189/jlb.0511239 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Salanga CL & Handel TM Chemokine oligomerization and interactions with receptors and glycosaminoglycans: the role of structural dynamics in function. Exp Cell Res 317, 590–601, doi: 10.1016/j.yexcr.2011.01.004 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Graham GJ, Handel TM & Proudfoot AEI Leukocyte Adhesion: Reconceptualizing Chemokine Presentation by Glycosaminoglycans. Trends Immunol 40, 472–481, doi: 10.1016/j.it.2019.03.009 (2019). [DOI] [PubMed] [Google Scholar]
- 49.Schumann K et al. Immobilized chemokine fields and soluble chemokine gradients cooperatively shape migration patterns of dendritic cells. Immunity 32, 703–713, doi:S1074–7613(10)00166–4 [pii] 10.1016/j.immuni.2010.04.017 (2010). [DOI] [PubMed] [Google Scholar]
- 50.Hynes RO Integrins: bidirectional, allosteric signaling machines. Cell 110, 673–687, doi:S0092867402009716 [pii] (2002). [DOI] [PubMed] [Google Scholar]
- 51.Sun Z, Costell M & Fassler R Integrin activation by talin, kindlin and mechanical forces. Nat Cell Biol 21, 25–31, doi: 10.1038/s41556-018-0234-9 (2019). [DOI] [PubMed] [Google Scholar]
- 52.Hons M et al. Chemokines and integrins independently tune actin flow and substrate friction during intranodal migration of T cells. Nat Immunol 19, 606–616, doi: 10.1038/s41590-018-0109-z (2018). [DOI] [PubMed] [Google Scholar]
- 53.Hallmann R et al. The regulation of immune cell trafficking by the extracellular matrix. Curr Opin Cell Biol 36, 54–61, doi: 10.1016/j.ceb.2015.06.006 (2015). [DOI] [PubMed] [Google Scholar]
- 54.Hyun YM et al. Uropod elongation is a common final step in leukocyte extravasation through inflamed vessels. J Exp Med 209, 1349–1362, doi:jem.20111426 [pii] 10.1084/jem.20111426 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Wang S et al. Venular basement membranes contain specific matrix protein low expression regions that act as exit points for emigrating neutrophils. J Exp Med 203, 1519–1532, doi: 10.1084/jem.20051210 (2006). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Wu C et al. Endothelial basement membrane laminin alpha5 selectively inhibits T lymphocyte extravasation into the brain. Nat Med 15, 519–527, doi: 10.1038/nm.1957 (2009). [DOI] [PubMed] [Google Scholar]
- 57.Sixt M et al. Endothelial cell laminin isoforms, laminins 8 and 10, play decisive roles in T cell recruitment across the blood-brain barrier in experimental autoimmune encephalomyelitis. J Cell Biol 153, 933–946, doi: 10.1083/jcb.153.5.933 (2001). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Zhang X et al. The endothelial basement membrane acts as a checkpoint for entry of pathogenic T cells into the brain. J Exp Med 217, doi: 10.1084/jem.20191339 (2020). This study highlights the functional importance of spatial patterning of ECM components, showing the expression of laminin at the interface between the endothelial monolayer and the underlying basement membrane is critical to control pathogenicity of infiltrating T cells.
- 59.Wang X, Rodda LB, Bannard O & Cyster JG Integrin-mediated interactions between B cells and follicular dendritic cells influence germinal center B cell fitness. J Immunol 192, 4601–4609, doi: 10.4049/jimmunol.1400090 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Schrock DC. et al. Pivotal role for alpha-V integrins in Tfh GC positioning and support of the germinal center response. In revision, PNAS. doi: 10.1073/pnas.1809329116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Ray SJ et al. The collagen binding alpha1beta1 integrin VLA-1 regulates CD8 T cell-mediated immune protection against heterologous influenza infection. Immunity 20, 167–179 (2004). [DOI] [PubMed] [Google Scholar]
- 62.Reilly EC et al. TRM integrins CD103 and CD49a differentially support adherence and motility after resolution of influenza virus infection. Proc Natl Acad Sci U S A 117, 12306–12314, doi: 10.1073/pnas.1915681117 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Wilson EH et al. Behavior of parasite-specific effector CD8+ T cells in the brain and visualization of a kinesis-associated system of reticular fibers. Immunity 30, 300–311, doi:S1074–7613(09)00060–0 [pii] 10.1016/j.immuni.2008.12.013 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Moreau HD, Piel M, Voituriez R & Lennon-Dumenil AM Integrating Physical and Molecular Insights on Immune Cell Migration. Trends Immunol 39, 632–643, doi: 10.1016/j.it.2018.04.007 (2018). [DOI] [PubMed] [Google Scholar]
- 65.Nicolas-Boluda A & Donnadieu E Obstacles to T cell migration in the tumor microenvironment. Comp Immunol Microbiol Infect Dis 63, 22–30, doi: 10.1016/j.cimid.2018.12.006 (2019). [DOI] [PubMed] [Google Scholar]
- 66. Salmon H et al. Matrix architecture defines the preferential localization and migration of T cells into the stroma of human lung tumors. J Clin Invest 122, 899–910, doi:45817 [pii] 10.1172/JCI45817 (2012). The study demonstrates that the density and orientation of the stromal extracellular matrix play key roles in controlling the migration of T cells.
- 67.Zhu C, Chen W, Lou J, Rittase W & Li K Mechanosensing through immunoreceptors. Nat Immunol 20, 1269–1278, doi: 10.1038/s41590-019-0491-1 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Trichet L et al. Evidence of a large-scale mechanosensing mechanism for cellular adaptation to substrate stiffness. Proc Natl Acad Sci U S A 109, 6933–6938, doi: 10.1073/pnas.1117810109 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Fernandes NRJ et al. CD4+ T cell interstital migration controlled by fibronectin in the inflamed skin. Frontiers in Immunology 11 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Wang WY, Davidson CD, Lin D & Baker BM Actomyosin contractility-dependent matrix stretch and recoil induces rapid cell migration. Nat Commun 10, 1186, doi: 10.1038/s41467-019-09121-0 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Sarris M & Sixt M Navigating in tissue mazes: chemoattractant interpretation in complex environments. Curr Opin Cell Biol 36, 93–102, doi: 10.1016/j.ceb.2015.08.001 (2015). [DOI] [PubMed] [Google Scholar]
- 72.Krummel MF, Bartumeus F & Gerard A T cell migration, search strategies and mechanisms. Nat Rev Immunol 16, 193–201, doi: 10.1038/nri.2015.16 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Bromley SK, Peterson DA, Gunn MD & Dustin ML Cutting edge: hierarchy of chemokine receptor and TCR signals regulating T cell migration and proliferation. J Immunol 165, 15–19 (2000). [DOI] [PubMed] [Google Scholar]
- 74.Dustin ML & Choudhuri K Signaling and Polarized Communication Across the T Cell Immunological Synapse. Annu Rev Cell Dev Biol 32, 303–325, doi: 10.1146/annurev-cellbio-100814-125330 (2016). [DOI] [PubMed] [Google Scholar]
- 75.Baker CM et al. Opposing roles for RhoH GTPase during T-cell migration and activation. Proceedings of the National Academy of Sciences 109, 10474–10479, doi: 10.1073/pnas.1114214109 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Brunner-Weinzierl MC & Rudd CE CTLA-4 and PD-1 Control of T-Cell Motility and Migration: Implications for Tumor Immunotherapy. Front Immunol 9, 2737, doi: 10.3389/fimmu.2018.02737 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Schneider H et al. Reversal of the TCR stop signal by CTLA-4. Science 313, 1972–1975, doi:1131078 [pii] 10.1126/science.1131078 (2006). [DOI] [PubMed] [Google Scholar]
- 78. Sarris M et al. Inflammatory chemokines direct and restrict leukocyte migration within live tissues as glycan-bound gradients. Curr Biol 22, 2375–2382, doi: 10.1016/j.cub.2012.11.018 (2012). The study highlights that, by interacting with extracellular heparan sulfate proteoglycans, chemokines can establish robust surface-bound gradients that mediate both recruitment and retention of leukocytes at sites of infection.
- 79. Hickman HD et al. CXCR3 chemokine receptor enables local CD8(+) T cell migration for the destruction of virus-infected cells. Immunity 42, 524–537, doi: 10.1016/j.immuni.2015.02.009 (2015). A powerful in vivo demonstration that chemokine signals guide the ability of CD8+ T cells to locate virus-infected cells within the infected tissue and thereby exert anti-viral effector functions.
- 80.Egen JG et al. Intravital imaging reveals limited antigen presentation and T cell effector function in mycobacterial granulomas. Immunity 34, 807–819, doi:S1074–7613(11)00183-X [pii] 10.1016/j.immuni.2011.03.022 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Shakhar G et al. Stable T cell-dendritic cell interactions precede the development of both tolerance and immunity in vivo. Nat Immunol 6, 707–714, doi:ni1210 [pii] 10.1038/ni1210 (2005). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. Honda T et al. Tuning of antigen sensitivity by T cell receptor-dependent negative feedback controls T cell effector function in inflamed tissues. Immunity 40, 235–247, doi: 10.1016/j.immuni.2013.11.017 (2014). Dynamic analysis of T cell interactions with APC in the inflamed tissue revealed that the pattern of transient arrest and rapid resumption of migration is controlled by a TCR-dependent negative feedback loop mediated by effector T cell upregulation of PD-1, that may protect the host from immunopathology.
- 83.Moreau HD et al. Dynamic in situ cytometry uncovers T cell receptor signaling during immunological synapses and kinapses in vivo. Immunity 37, 351–363, doi:S1074–7613(12)00227–0 [pii] 10.1016/j.immuni.2012.05.014 (2012). [DOI] [PubMed] [Google Scholar]
- 84.Cyster JG & Schwab SR Sphingosine-1-phosphate and lymphocyte egress from lymphoid organs. Annu Rev Immunol 30, 69–94, doi: 10.1146/annurev-immunol-020711-075011 (2012). [DOI] [PubMed] [Google Scholar]
- 85.Sadik CD & Luster AD Lipid-cytokine-chemokine cascades orchestrate leukocyte recruitment in inflammation. J Leukoc Biol 91, 207–215, doi: 10.1189/jlb.0811402 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Tager AM et al. Leukotriene B4 receptor BLT1 mediates early effector T cell recruitment. Nat Immunol 4, 982–990, doi: 10.1038/ni970 (2003). [DOI] [PubMed] [Google Scholar]
- 87.Qi H T follicular helper cells in space-time. Nat Rev Immunol 16, 612–625, doi: 10.1038/nri.2016.94 (2016). [DOI] [PubMed] [Google Scholar]
- 88.Cyster JG, Dang EV, Reboldi A & Yi T 25-Hydroxycholesterols in innate and adaptive immunity. Nat Rev Immunol 14, 731–743, doi: 10.1038/nri3755 (2014). [DOI] [PubMed] [Google Scholar]
- 89.Yan H et al. Plexin B2 and Semaphorin 4C Guide T Cell Recruitment and Function in the Germinal Center. Cell Rep 19, 995–1007, doi: 10.1016/j.celrep.2017.04.022 (2017). [DOI] [PubMed] [Google Scholar]
- 90.Moriyama S et al. Sphingosine-1-phosphate receptor 2 is critical for follicular helper T cell retention in germinal centers. J Exp Med 211, 1297–1305, doi: 10.1084/jem.20131666 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Lu P, Shih C & Qi H Ephrin B1-mediated repulsion and signaling control germinal center T cell territoriality and function. Science 356, doi: 10.1126/science.aai9264 (2017). [DOI] [PubMed] [Google Scholar]
- 92.Muppidi JR, Lu E & Cyster JG The G protein-coupled receptor P2RY8 and follicular dendritic cells promote germinal center confinement of B cells, whereas S1PR3 can contribute to their dissemination. J Exp Med 212, 2213–2222, doi: 10.1084/jem.20151250 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Lu E, Wolfreys FD, Muppidi JR, Xu Y & Cyster JG S-Geranylgeranyl-L-glutathione is a ligand for human B cell-confinement receptor P2RY8. Nature 567, 244–248, doi: 10.1038/s41586-019-1003-z (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Muller AJ et al. CD4+ T cells rely on a cytokine gradient to control intracellular pathogens beyond sites of antigen presentation. Immunity 37, 147–157, doi: 10.1016/j.immuni.2012.05.015 (2012). [DOI] [PubMed] [Google Scholar]
- 95.Mandl JN et al. Quantification of lymph node transit times reveals differences in antigen surveillance strategies of naive CD4+ and CD8+ T cells. Proc Natl Acad Sci U S A 109, 18036–18041, doi: 10.1073/pnas.1211717109 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Gebhardt T et al. Different patterns of peripheral migration by memory CD4+ and CD8+ T cells. Nature 477, 216–219, doi:nature10339 [pii] 10.1038/nature10339 (2011). [DOI] [PubMed] [Google Scholar]
- 97.Leon B & Lund FE Compartmentalization of dendritic cell and T-cell interactions in the lymph node: Anatomy of T-cell fate decisions. Immunol Rev 289, 84–100, doi: 10.1111/imr.12758 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Kurachi M et al. Chemokine receptor CXCR3 facilitates CD8(+) T cell differentiation into short-lived effector cells leading to memory degeneration. J Exp Med 208, 1605–1620, doi: 10.1084/jem.20102101 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Hu JK, Kagari T, Clingan JM & Matloubian M Expression of chemokine receptor CXCR3 on T cells affects the balance between effector and memory CD8 T-cell generation. Proc Natl Acad Sci U S A 108, E118–127, doi: 10.1073/pnas.1101881108 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. Groom JR et al. CXCR3 chemokine receptor-ligand interactions in the lymph node optimize CD4+ T helper 1 cell differentiation. Immunity 37, 1091–1103, doi: 10.1016/j.immuni.2012.08.016 (2012). This study, and Leon et al (reference 142), were the first studies to define a chemokine-controlled intranodal re-positioning event that was critical for the completion of Th differentiation in the lymph node.
- 101.Crotty S T follicular helper cell differentiation, function, and roles in disease. Immunity 41, 529–542, doi: 10.1016/j.immuni.2014.10.004 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Sallusto F & Lanzavecchia A Understanding dendritic cell and T-lymphocyte traffic through the analysis of chemokine receptor expression. Immunol Rev 177, 134–140 (2000). [DOI] [PubMed] [Google Scholar]
- 103.Tough DF, Rioja I, Modis LK & Prinjha RK Epigenetic Regulation of T Cell Memory: Recalling Therapeutic Implications. Trends Immunol 41, 29–45, doi: 10.1016/j.it.2019.11.008 (2020). [DOI] [PubMed] [Google Scholar]
- 104. Gerard A et al. Detection of rare antigen-presenting cells through T cell-intrinsic meandering motility, mediated by Myo1g. Cell 158, 492–505, doi: 10.1016/j.cell.2014.05.044 (2014). This study identified Myosin 1g (Myo1g) as a T cell-intrinsic regulator of the meandering search pattern of migrating T cells that appears to enhance the detection of rare cognate antigen-presenting cells.
- 105.Laakso JM, Lewis JH, Shuman H & Ostap EM Myosin I can act as a molecular force sensor. Science 321, 133–136, doi: 10.1126/science.1159419 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Mrass P et al. ROCK regulates the intermittent mode of interstitial T cell migration in inflamed lungs. Nat Commun 8, 1010, doi: 10.1038/s41467-017-01032-2 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107. Gaylo-Moynihan A et al. Programming of Distinct Chemokine-Dependent and -Independent Search Strategies for Th1 and Th2 Cells Optimizes Function at Inflamed Sites. Immunity 51, 298–309 e296, doi: 10.1016/j.immuni.2019.06.026 (2019). This study revealed that Th differentiation inparts distinct effector subset-specific migration patterns and particular dependencies on migrational cues: Th1 cells explored the inflamed tissue in a GPCR and chemokine-dependent fashion, while Th2 cells scanned the tissue independent of GPCR signals.
- 108.Du F et al. Inflammatory Th17 Cells Express Integrin alphavbeta3 for Pathogenic Function. Cell Rep 16, 1339–1351, doi: 10.1016/j.celrep.2016.06.065 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Osborn JF et al. Enzymatic synthesis of core 2 O-glycans governs the tissue-trafficking potential of memory CD8(+) T cells. Sci Immunol 2, doi: 10.1126/sciimmunol.aan6049 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Steinert EM et al. Quantifying Memory CD8 T Cells Reveals Regionalization of Immunosurveillance. Cell 161, 737–749, doi: 10.1016/j.cell.2015.03.031 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Weber M et al. Interstitial dendritic cell guidance by haptotactic chemokine gradients. Science 339, 328–332, doi: 10.1126/science.1228456 (2013). [DOI] [PubMed] [Google Scholar]
- 112.Ariotti S et al. Subtle CXCR3-Dependent Chemotaxis of CTLs within Infected Tissue Allows Efficient Target Localization. J Immunol 195, 5285–5295, doi: 10.4049/jimmunol.1500853 (2015). [DOI] [PubMed] [Google Scholar]
- 113.Castellino F et al. Chemokines enhance immunity by guiding naive CD8+ T cells to sites of CD4+ T cell-dendritic cell interaction. Nature 440, 890–895, doi:nature04651 [pii] 10.1038/nature04651 (2006). [DOI] [PubMed] [Google Scholar]
- 114.Petrie Aronin CE et al. Migrating Myeloid Cells Sense Temporal Dynamics of Chemoattractant Concentrations. Immunity 47, 862–874 e863, doi: 10.1016/j.immuni.2017.10.020 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Witt CM, Raychaudhuri S, Schaefer B, Chakraborty AK & Robey EA Directed migration of positively selected thymocytes visualized in real time. PLoS Biol 3, e160, doi: 10.1371/journal.pbio.0030160 (2005). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Majumdar R, Sixt M & Parent CA New paradigms in the establishment and maintenance of gradients during directed cell migration. Curr Opin Cell Biol 30, 33–40, doi: 10.1016/j.ceb.2014.05.010 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Vanheule V, Metzemaekers M, Janssens R, Struyf S & Proost P How post-translational modifications influence the biological activity of chemokines. Cytokine 109, 29–51, doi: 10.1016/j.cyto.2018.02.026 (2018). [DOI] [PubMed] [Google Scholar]
- 118.Borroni EM, Mantovani A, Locati M & Bonecchi R Chemokine receptors intracellular trafficking. Pharmacol Ther 127, 1–8, doi: 10.1016/j.pharmthera.2010.04.006 (2010). [DOI] [PubMed] [Google Scholar]
- 119.Mantovani A, Bonecchi R & Locati M Tuning inflammation and immunity by chemokine sequestration: decoys and more. Nat Rev Immunol 6, 907–918 (2006). [DOI] [PubMed] [Google Scholar]
- 120.Bonecchi R & Graham GJ Atypical Chemokine Receptors and Their Roles in the Resolution of the Inflammatory Response. Front Immunol 7, 224, doi: 10.3389/fimmu.2016.00224 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Lau S et al. A negative-feedback loop maintains optimal chemokine concentrations for directional cell migration. Nat Cell Biol 22, 266–273, doi: 10.1038/s41556-020-0465-4 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Molon B et al. Chemokine nitration prevents intratumoral infiltration of antigen-specific T cells. J Exp Med 208, 1949–1962, doi: 10.1084/jem.20101956 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Nieto M et al. Polarization of chemokine receptors to the leading edge during lymphocyte chemotaxis. J Exp Med 186, 153–158, doi: 10.1084/jem.186.1.153 (1997). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Hyun YM, Chung HL, McGrath JL, Waugh RE & Kim M Activated integrin VLA-4 localizes to the lamellipodia and mediates T cell migration on VCAM-1. J Immunol 183, 359–369, doi: 10.4049/jimmunol.0803388 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Vroon A, Heijnen CJ & Kavelaars A GRKs and arrestins: regulators of migration and inflammation. J Leukoc Biol 80, 1214–1221, doi: 10.1189/jlb.0606373 (2006). [DOI] [PubMed] [Google Scholar]
- 126.Schwarz J et al. Dendritic Cells Interpret Haptotactic Chemokine Gradients in a Manner Governed by Signal-to-Noise Ratio and Dependent on GRK6. Curr Biol 27, 1314–1325, doi: 10.1016/j.cub.2017.04.004 (2017). [DOI] [PubMed] [Google Scholar]
- 127.Kehrl JH The impact of RGS and other G-protein regulatory proteins on Galphai-mediated signaling in immunity. Biochem Pharmacol 114, 40–52, doi: 10.1016/j.bcp.2016.04.005 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Bromley SK, Mempel TR & Luster AD Orchestrating the orchestrators: chemokines in control of T cell traffic. Nat Immunol 9, 970–980 (2008). [DOI] [PubMed] [Google Scholar]
- 129.Campbell JJ, Foxman EF & Butcher EC Chemoattractant receptor cross talk as a regulatory mechanism in leukocyte adhesion and migration. Eur J Immunol 27, 2571–2578, doi: 10.1002/eji.1830271016 (1997). [DOI] [PubMed] [Google Scholar]
- 130.Heit B, Tavener S, Raharjo E & Kubes P An intracellular signaling hierarchy determines direction of migration in opposing chemotactic gradients. J Cell Biol 159, 91–102, doi: 10.1083/jcb.200202114 (2002). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Lin F & Butcher EC Modeling the role of homologous receptor desensitization in cell gradient sensing. J Immunol 181, 8335–8343, doi: 10.4049/jimmunol.181.12.8335 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Wu D & Lin F Modeling cell gradient sensing and migration in competing chemoattractant fields. PLoS One 6, e18805, doi: 10.1371/journal.pone.0018805 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Zidar DA, Violin JD, Whalen EJ & Lefkowitz RJ Selective engagement of G protein coupled receptor kinases (GRKs) encodes distinct functions of biased ligands. Proc Natl Acad Sci U S A 106, 9649–9654, doi: 10.1073/pnas.0904361106 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134. Coombs C et al. Chemokine receptor trafficking coordinates neutrophil clustering and dispersal at wounds in zebrafish. Nat Commun 10, 5166, doi: 10.1038/s41467-019-13107-3 (2019). This elegant study visualized the differential membrane trafficking of chemokine receptors in neutrophils and linked chemokine receptor availability to the accumulation and subsequent dispersal of neutrophils at a wound site, ensuring an effective and self-resolving response to tissue damage.
- 135.Sun H et al. Distinct chemokine signaling regulates integrin ligand specificity to dictate tissue-specific lymphocyte homing. Dev Cell 30, 61–70, doi: 10.1016/j.devcel.2014.05.002 (2014). [DOI] [PubMed] [Google Scholar]
- 136.Prentice-Mott HV et al. Biased migration of confined neutrophil-like cells in asymmetric hydraulic environments. Proc Natl Acad Sci U S A 110, 21006–21011, doi: 10.1073/pnas.1317441110 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Moses ME, Cannon JL, Gordon DM & Forrest S Distributed Adaptive Search in T Cells: Lessons From Ants. Front Immunol 10, 1357, doi: 10.3389/fimmu.2019.01357 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Torabi-Parizi P et al. Pathogen-related differences in the abundance of presented antigen are reflected in CD4+ T cell dynamic behavior and effector function in the lung. J Immunol 192, 1651–1660, doi: 10.4049/jimmunol.1301743 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Afonso PV et al. LTB4 is a signal-relay molecule during neutrophil chemotaxis. Dev Cell 22, 1079–1091, doi: 10.1016/j.devcel.2012.02.003 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140. Lammermann T et al. Neutrophil swarms require LTB4 and integrins at sites of cell death in vivo. Nature 498, 371–375, doi: 10.1038/nature12175 (2013). The study showed that neutrophil production of the LTB4 and cell surface integrins were required for long-distance neutrophil warming in the extravascular space of a damaged tissue.
- 141.Lim K et al. In situ neutrophil efferocytosis shapes T cell immunity to influenza infection. Nature Immunology (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Leon B et al. Regulation of T(H)2 development by CXCR5+ dendritic cells and lymphotoxin-expressing B cells. Nat Immunol 13, 681–690, doi: 10.1038/ni.2309 (2012). See reference 100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Goldberg MF et al. Salmonella Persist in Activated Macrophages in T Cell-Sparse Granulomas but Are Contained by Surrounding CXCR3 Ligand-Positioned Th1 Cells. Immunity 49, 1090–1102 e1097, doi: 10.1016/j.immuni.2018.10.009 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Richmond JM et al. Keratinocyte-Derived Chemokines Orchestrate T-Cell Positioning in the Epidermis during Vitiligo and May Serve as Biomarkers of Disease. J Invest Dermatol 137, 350–358, doi: 10.1016/j.jid.2016.09.016 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Groom JR & Luster AD CXCR3 in T cell function. Experimental Cell Research 317, 620–631, doi: 10.1016/j.yexcr.2010.12.017 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Sung JH et al. Chemokine guidance of central memory T cells is critical for antiviral recall responses in lymph nodes. Cell 150, 1249–1263, doi: 10.1016/j.cell.2012.08.015 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Kastenmuller W et al. Peripheral prepositioning and local CXCL9 chemokine-mediated guidance orchestrate rapid memory CD8+ T cell responses in the lymph node. Immunity 38, 502–513, doi: 10.1016/j.immuni.2012.11.012 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Carrasco YR & Batista FD B cells acquire particulate antigen in a macrophage-rich area at the boundary between the follicle and the subcapsular sinus of the lymph node. Immunity 27, 160–171, doi: 10.1016/j.immuni.2007.06.007 (2007). [DOI] [PubMed] [Google Scholar]
- 149.Junt T et al. Subcapsular sinus macrophages in lymph nodes clear lymph-borne viruses and present them to antiviral B cells. Nature 450, 110–114, doi: 10.1038/nature06287 (2007). [DOI] [PubMed] [Google Scholar]
- 150.Gerner MY, Torabi-Parizi P & Germain RN Strategically localized dendritic cells promote rapid T cell responses to lymph-borne particulate antigens. Immunity 42, 172–185, doi: 10.1016/j.immuni.2014.12.024 (2015). [DOI] [PubMed] [Google Scholar]
- 151.Woodruff MC et al. Trans-nodal migration of resident dendritic cells into medullary interfollicular regions initiates immunity to influenza vaccine. J Exp Med 211, 1611–1621, doi: 10.1084/jem.20132327 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Baptista AP et al. The Chemoattractant Receptor Ebi2 Drives Intranodal Naive CD4(+) T Cell Peripheralization to Promote Effective Adaptive Immunity. Immunity 50, 1188–1201 e1186, doi: 10.1016/j.immuni.2019.04.001 (2019). [DOI] [PubMed] [Google Scholar]
- 153.McFarland AP & Colonna M Sense and immuno-sensibility: innate lymphoid cell niches and circuits. Curr Opin Immunol 62, 9–14, doi: 10.1016/j.coi.2019.11.003 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154. Dahlgren MW et al. Adventitial Stromal Cells Define Group 2 Innate Lymphoid Cell Tissue Niches. Immunity 50, 707–722 e706, doi: 10.1016/j.immuni.2019.02.002 (2019). This study implicated adventitial stromal cells in the support of conserved niches for type 2 lymphocyte expansion and function in the lung.
- 155.Klose CSN et al. The neuropeptide neuromedin U stimulates innate lymphoid cells and type 2 inflammation. Nature 549, 282–286, doi: 10.1038/nature23676 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Moriyama S et al. beta2-adrenergic receptor-mediated negative regulation of group 2 innate lymphoid cell responses. Science 359, 1056–1061, doi: 10.1126/science.aan4829 (2018). [DOI] [PubMed] [Google Scholar]
- 157.Dahlgren MW & Molofsky AB Adventitial Cuffs: Regional Hubs for Tissue Immunity. Trends Immunol 40, 877–887, doi: 10.1016/j.it.2019.08.002 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Veres TZ et al. Allergen-Induced CD4+ T Cell Cytokine Production within Airway Mucosal Dendritic Cell-T Cell Clusters Drives the Local Recruitment of Myeloid Effector Cells. J Immunol 198, 895–907, doi: 10.4049/jimmunol.1601448 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Filipe-Santos O et al. A dynamic map of antigen recognition by CD4 T cells at the site of Leishmania major infection. Cell Host Microbe 6, 23–33, doi:S1931–3128(09)00188–7 [pii] 10.1016/j.chom.2009.04.014 (2009). [DOI] [PubMed] [Google Scholar]
- 160.Siracusa F et al. Nonfollicular reactivation of bone marrow resident memory CD4 T cells in immune clusters of the bone marrow. Proc Natl Acad Sci U S A 115, 1334–1339, doi: 10.1073/pnas.1715618115 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161. Natsuaki Y et al. Perivascular leukocyte clusters are essential for efficient activation of effector T cells in the skin. Nat Immunol 15, 1064–1069, doi: 10.1038/ni.2992 (2014). This work defined an induced perivascular niche in the skin where DCs form clusters with effector T cells to promote in situ T cell proliferation and activation.
- 162.Huang LR et al. Intrahepatic myeloid-cell aggregates enable local proliferation of CD8(+) T cells and successful immunotherapy against chronic viral liver infection. Nat Immunol 14, 574–583, doi: 10.1038/ni.2573 (2013). [DOI] [PubMed] [Google Scholar]
- 163.Wakim LM, Woodward-Davis A & Bevan MJ Memory T cells persisting within the brain after local infection show functional adaptations to their tissue of residence. Proc Natl Acad Sci U S A 107, 17872–17879, doi: 10.1073/pnas.1010201107 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164. Iijima N & Iwasaki A T cell memory. A local macrophage chemokine network sustains protective tissue-resident memory CD4 T cells. Science 346, 93–98, doi: 10.1126/science.1257530 (2014). One of the first studies to identify a TRM tissue niche, where chemokines secreted by local macrophages maintained vaginal CD4 TRM in memory lymphocyte clusters, independent of circulating memory T cells.
- 165.Nordenfelt P et al. Direction of actin flow dictates integrin LFA-1 orientation during leukocyte migration. Nat Commun 8, 2047, doi: 10.1038/s41467-017-01848-y (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Carman CV & Springer TA A transmigratory cup in leukocyte diapedesis both through individual vascular endothelial cells and between them. J Cell Biol 167, 377–388, doi: 10.1083/jcb.200404129 (2004). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Guillou L et al. T-lymphocyte passive deformation is controlled by unfolding of membrane surface reservoirs. Mol Biol Cell 27, 3574–3582, doi: 10.1091/mbc.E16-06-0414 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Song J, Wu C, Zhang X & Sorokin LM In vivo processing of CXCL5 (LIX) by matrix metalloproteinase (MMP)-2 and MMP-9 promotes early neutrophil recruitment in IL-1beta-induced peritonitis. J Immunol 190, 401–410, doi: 10.4049/jimmunol.1202286 (2013). [DOI] [PubMed] [Google Scholar]
- 169.Proost P et al. Proteolytic processing of CXCL11 by CD13/aminopeptidase N impairs CXCR3 and CXCR7 binding and signaling and reduces lymphocyte and endothelial cell migration. Blood 110, 37–44, doi: 10.1182/blood-2006-10-049072 (2007). [DOI] [PubMed] [Google Scholar]
- 170.Proost P et al. Amino-terminal truncation of CXCR3 agonists impairs receptor signaling and lymphocyte chemotaxis, while preserving antiangiogenic properties. Blood 98, 3554–3561, doi: 10.1182/blood.v98.13.3554 (2001). [DOI] [PubMed] [Google Scholar]
- 171.Ludwig A, Schiemann F, Mentlein R, Lindner B & Brandt E Dipeptidyl peptidase IV (CD26) on T cells cleaves the CXC chemokine CXCL11 (I-TAC) and abolishes the stimulating but not the desensitizing potential of the chemokine. J Leukoc Biol 72, 183–191 (2002). [PubMed] [Google Scholar]
- 172.Oravecz T et al. Regulation of the receptor specificity and function of the chemokine RANTES (regulated on activation, normal T cell expressed and secreted) by dipeptidyl peptidase IV (CD26)-mediated cleavage. J Exp Med 186, 1865–1872, doi: 10.1084/jem.186.11.1865 (1997). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Barker CE et al. CCL2 nitration is a negative regulator of chemokine-mediated inflammation. Sci Rep 7, 44384, doi: 10.1038/srep44384 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Loos T et al. Citrullination of CXCL10 and CXCL11 by peptidylarginine deiminase: a naturally occurring posttranslational modification of chemokines and new dimension of immunoregulation. Blood 112, 2648–2656, doi: 10.1182/blood-2008-04-149039 (2008). [DOI] [PubMed] [Google Scholar]
- 175.Kiermaier E et al. Polysialylation controls dendritic cell trafficking by regulating chemokine recognition. Science 351, 186–190, doi: 10.1126/science.aad0512 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Denard B, Han S, Kim J, Ross EM & Ye J Regulating G protein-coupled receptors by topological inversion. Elife 8, doi: 10.7554/eLife.40234 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Weber C, Zernecke A & Libby P The multifaceted contributions of leukocyte subsets to atherosclerosis: lessons from mouse models. Nat Rev Immunol 8, 802–815, doi: 10.1038/nri2415 (2008). [DOI] [PubMed] [Google Scholar]
- 178.Gewaltig J, Kummer M, Koella C, Cathomas G & Biedermann BC Requirements for CD8 T-cell migration into the human arterial wall. Hum Pathol 39, 1756–1762, doi: 10.1016/j.humpath.2008.04.018 (2008). [DOI] [PubMed] [Google Scholar]
- 179.Kyaw T et al. Cytotoxic and proinflammatory CD8+ T lymphocytes promote development of vulnerable atherosclerotic plaques in apoE-deficient mice. Circulation 127, 1028–1039, doi: 10.1161/CIRCULATIONAHA.112.001347 (2013). [DOI] [PubMed] [Google Scholar]
- 180.Kleinschmidt-DeMasters BK & Tyler KL Progressive multifocal leukoencephalopathy complicating treatment with natalizumab and interferon beta-1a for multiple sclerosis. The New England journal of medicine 353, 369–374 (2005). [DOI] [PubMed] [Google Scholar]
- 181.Van Assche G et al. Progressive multifocal leukoencephalopathy after natalizumab therapy for Crohn’s disease. The New England journal of medicine 353, 362–368 (2005). [DOI] [PubMed] [Google Scholar]
- 182.Fisher DT et al. IL-6 trans-signaling licenses mouse and human tumor microvascular gateways for trafficking of cytotoxic T cells. The Journal of clinical investigation 121, 3846–3859, doi: 10.1172/JCI44952 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Moon EK et al. Intra-tumoral delivery of CXCL11 via a vaccinia virus, but not by modified T cells, enhances the efficacy of adoptive T cell therapy and vaccines. Oncoimmunology 7, e1395997, doi: 10.1080/2162402X.2017.1395997 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184.Hu Z et al. Mouse IP-10 Gene Delivered by Folate-modified Chitosan Nanoparticles and Dendritic/tumor Cells Fusion Vaccine Effectively Inhibit the Growth of Hepatocellular Carcinoma in Mice. Theranostics 7, 1942–1952, doi: 10.7150/thno.16236 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185.Peng D et al. Epigenetic silencing of TH1-type chemokines shapes tumour immunity and immunotherapy. Nature 527, 249–253, doi: 10.1038/nature15520 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186.Feig C et al. Targeting CXCL12 from FAP-expressing carcinoma-associated fibroblasts synergizes with anti-PD-L1 immunotherapy in pancreatic cancer. Proc Natl Acad Sci U S A 110, 20212–20217, doi: 10.1073/pnas.1320318110 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187.Peng SB et al. Distinct mobilization of leukocytes and hematopoietic stem cells by CXCR4 peptide antagonist LY2510924 and monoclonal antibody LY2624587. Oncotarget 8, 94619–94634, doi: 10.18632/oncotarget.21816 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188.O’Hara MH et al. Safety and Pharmacokinetics of CXCR4 Peptide Antagonist, LY2510924, in Combination with Durvalumab in Advanced Refractory Solid Tumors. J Pancreat Cancer 6, 21–31, doi: 10.1089/pancan.2019.0018 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189.Weinstein EJ et al. VCC-1, a novel chemokine, promotes tumor growth. Biochem Biophys Res Commun 350, 74–81, doi: 10.1016/j.bbrc.2006.08.194 (2006). [DOI] [PubMed] [Google Scholar]
- 190.Kuroda T et al. Monocyte chemoattractant protein-1 transfection induces angiogenesis and tumorigenesis of gastric carcinoma in nude mice via macrophage recruitment. Clin Cancer Res 11, 7629–7636, doi: 10.1158/1078-0432.CCR-05-0798 (2005). [DOI] [PubMed] [Google Scholar]
- 191.Kang H, Mansel RE & Jiang WG Genetic manipulation of stromal cell-derived factor-1 attests the pivotal role of the autocrine SDF-1-CXCR4 pathway in the aggressiveness of breast cancer cells. Int J Oncol 26, 1429–1434 (2005). [PubMed] [Google Scholar]
- 192.Campanella GS, Medoff BD, Manice LA, Colvin RA & Luster AD Development of a novel chemokine-mediated in vivo T cell recruitment assay. J Immunol Methods 331, 127–139, doi: 10.1016/j.jim.2007.12.002 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193.Shin H & Iwasaki A A vaccine strategy that protects against genital herpes by establishing local memory T cells. Nature 491, 463–467, doi: 10.1038/nature11522 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194.Gopinath S, Lu P & Iwasaki A Cutting Edge: The Use of Topical Aminoglycosides as an Effective Pull in “Prime and Pull” Vaccine Strategy. J Immunol 204, 1703–1707, doi: 10.4049/jimmunol.1900462 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195.Mackay LK et al. Long-lived epithelial immunity by tissue-resident memory T (TRM) cells in the absence of persisting local antigen presentation. Proc Natl Acad Sci U S A 109, 7037–7042, doi:1202288109 [pii] 10.1073/pnas.1202288109 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196.Gola A et al. Prime and target immunization protects against liver-stage malaria in mice. Sci Transl Med 10, doi: 10.1126/scitranslmed.aap9128 (2018). [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
